You are on page 1of 767

C O M M O N A B B R E V I AT I O N S

USED IN THE TEXT

2D two-dimensional DTI diffusion tensor imaging


3D three-dimensional DTPA diethylenetriamine pentaacetic acid
4D four-dimensional DUS Doppler ultrasound
A2C apical two chamber DWI diffusion weighted imaging
A4C apical four chamber EAM electroanatomic map
AAR area at risk ECG electrocardiogram
ABI ankle-brachial index ECV extracellular volume
ACC American College of Cardiology ED end diastole
ACEI angiotensin-converting enzyme inhibitor EDV(I) end-diastolic volume (index)
ACR American College of Radiology EF ejection fraction
ACS acute coronary syndrome EGE early gadolinium enhancement
ADP adenosine diphosphate eGFR estimated glomerular filtration rate
AF atrial fibrillation EMB endomyocardial biopsy
AHA American Heart Association EPI echo planar imaging
AMI acute myocardial infarction ESC European Society of Cardiology
APMHR age-predicted maximal heart rate ESV(I) end-systolic volume (index)
ARVC arrhythmogenic right ventricular cardiomyopathy FDA US Food and Drug Administration
AS aortic stenosis FDG fluorodeoxyglucose
ASD atrial septal defect FFR fractional flow reserve
ASL arterial spin labeling FHS Framingham Heart Study
ATP adenosine triphosphate FID free induction decay
AUC appropriate use criteria; area under the curve FLASH fast low angle shot
BMI body mass index fMRI functional magnetic resonance imaging
BOLD blood-oxygen-level–dependent FOV field of view
BP blood pressure FPP first-pass perfusion
BSA body surface area FSE fast spin echo
bSSFP balanced steady-state free precession FT feature tracking; Fourier transform
C carbon FWHM full width at half maximum
CABG coronary artery bypass graft GBCA gadolinium-based contrast agent
CAD coronary artery disease Gd gadolinium
CAV coronary allograft vasculopathy GRAPPA generalized autocalibrating partially parallel acquisition
CE contrast enhanced GRE gradient recalled echo
CFR coronary flow reserve HARP harmonic phase
CHD congenital heart disease HASTE half-Fourier single-shot turbo spin echo
CI confidence interval HCM hypertrophic cardiomyopathy
CIED cardiac implantable electronic device HDL high-density lipoprotein
CK creatine kinase HF heart failure
CKD chronic kidney disease HFpEF heart failure with preserved ejection fraction
CMR cardiovascular magnetic resonance HFrEF heart failure with reduced ejection fraction
CMRS cardiovascular magnetic resonance spectroscopy HIP high-intensity plaque
CNR contrast-to-noise ratio HLA horizontal long axis
CoV coefficient of variation HOCM hypertrophic obstructive cardiomyopathy
Cr creatinine HR heart rate
CRT cardiac resynchronization therapy ICC intraclass correlation coefficient
CS compressed sensing ICD implantable cardioverter-defibrillator
CSA cross-sectional area ICM ischemic cardiomyopathy
CSPAMM complementary spatial modulation of magnetization IHD ischemic heart disease
CT computed tomography IPH intraplaque hemorrhage
CTO chronic total occlusion IQR interquartile range
CVA cerebrovascular attack IR inversion recovery
DBP diastolic blood pressure IVUS intravascular ultrasound
DCM dilated cardiomyopathy LA left atrial; left atrium
DCMR dobutamine stress cardiovascular magnetic resonance LAD left anterior descending (coronary artery)
DENSE displacement encoding with stimulated echoes LCX left circumflex (coronary artery)
DIR double inversion recovery LDL low-density lipoprotein
DSA digital subtraction angiography LGE late gadolinium enhancement
DSE dobutamine stress echocardiography LV left ventricle; left ventricular

IFC3
IFC4 COMMON ABBREVIATIONS USED IN THE TEXT

LVEDVP left ventricular end-diastolic pressure Qs systemic flow


LVEF left ventricular ejection fraction RA right atrial; right atrium
LVNC left ventricular noncompaction RARE rapid acquisition with relaxation enhancement
LVOT left ventricular outflow tract RCA right coronary artery
MACE major adverse cardiac event RF radiofrequency
MAP mean arterial pressure ROC receiver operator characteristic; receiver operator curve
MAPSE mitral annular plane systolic excursion ROI region of interest
MBF myocardial blood flow RPP rate pressure product
MBV myocardial blood volume RV right ventricle; right ventricular
MDCT multidetector computed tomography RVED(I) right ventricular end-diastolic volume (index)
MESA Multiethnic Study of Atherosclerosis RVEF right ventricular ejection fraction
MI myocardial infarction RVOT right ventricular outflow tract
MIP maximal intensity projection RVSP right ventricular systolic pressure
MMP metalloproteinases SAR specific absorption rate
Mn manganese SAX short axis
MOCO motion corrected SBP systolic blood pressure
MOLLI modified Look-Locker inversion recovery SCMR Society for Cardiovascular Magnetic Resonance
MPO myeloperoxidase SD standard deviation
MPR multiplanar reconstruction/reformatting; myocardial SEE standard error of the estimate
perfusion reserve SENSE sensitivity encoding
MR magnetic resonance ShMOLLI shortened modified Look-Locker inversion recovery
MRA magnetic resonance angiography SI signal intensity
MRI magnetic resonance imaging SNR signal-to-noise ratio
MRS magnetic resonance spectroscopy SPAMM spatial modulation of magnetization
MT magnetization transfer SPECT single-photon emission computed tomography
MTC magnetization transfer contrast SSFP steady-state free precession
MVD microvascular disease STEAM stimulated echo acquisition mode
MVO microvascular obstruction STEMI ST elevation myocardial infarction
MVO2 myocardial oxygen consumption STIR short-inversion time inversion recovery
MVP mitral valve prolapse STS Surgical Thoracic Society
MVR mass/volume ratio SVC superior vena cava
NHLBI National Heart, Lung, and Blood Institute SV(I) stroke volume (index)
NSF nephrogenic systemic fibrosis SVR systemic vascular resistance
NSTEMI non–ST elevation myocardial infarction T Tesla
NYHA New York Heart Association T1W T1 weighted
OR odds ratio T2W T2 weighted
P phosphorus TD delay time
PAD peripheral arterial disease TE echo time
PCA phase contrast angiography; principal component analysis TEE transesophageal echocardiography
PCI percutaneous coronary intervention TFE turbo field echo
PCMR phase contrast magnetic resonance TGA transposition of the great arteries
PCr phosphocreatine TI inversion time
PDA patent ductus arteriosus; posterior descend­ing artery TIA transient ischemic attack
PDW proton-density weighted TOF tetralogy of Fallot
PET positron emission tomography TR repetition time
PLAX parasternal long axis TSE turbo spin echo
PMR plaque:myocardial signal ratio TTC triphenyltetrazolium chloride
POC point of care TTE transthoracic echocardiography
PPM permanent pacemaker USPIO ultrasmall superparamagnetic particles of iron oxide
PSAX parasternal short axis VCAM vascular cell adhesion molecule
PSIR phase-sensitive inversion recovery VENC velocity encoded
PWV pulse wave velocity VF ventricular fibrillation
QALY quality-adjusted life year VSD ventricular septal defect
QCA quantitative coronary angiography VT ventricular tachycardia
Qp pulmonary flow XMR combined x-ray cardiac magnetic resonance laboratories
CARDIOVASCULAR
MAGNETIC
RESONANCE
CARDIOVASCULAR
MAGNETIC
RESONANCE
A COMPANION TO BRAUNWALD’S HEART DISEASE

THIRD EDITION

WARREN J. MANNING, MD, FACC, FACP, FAHA, FISMRM, FSCMR


Professor of Medicine and Radiology
Harvard Medical School
Section Chief, Non-invasive Cardiac Imaging and Testing
Cardiovascular Division
Beth Israel Deaconess Medical Center
Boston, Massachusetts

DUDLEY J. PENNELL, MD, FRCP, FACC, FESC, FAHA, FRCR, FMedSci, FSCMR
Professor of Cardiology
National Heart and Lung Institute
Imperial College London;
Director, Cardiovascular Magnetic Resonance Unit
Royal Brompton Hospital
London, United Kingdom
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

CARDIOVASCULAR MAGNETIC RESONANCE: A COMPANION TO


BRAUNWALD’S HEART DISEASE, THIRD EDITION ISBN: 978-0-323-41561-3
Copyright © 2019 by Elsevier, Inc. All rights reserved.

Chapter 47: Interventional Cardiovascular Magnetic Resonance by Toby Rogers and Robert Lederman is in
public domain.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience broaden
our understanding, changes in research methods, professional practices, or medical treatment may become
necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and
using any information, methods, compounds, or experiments described herein. In using such information
or methods they should be mindful of their own safety and the safety of others, including parties for
whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the most
current information provided (i) on procedures featured or (ii) by the manufacturer of each product to be
administered, to verify the recommended dose or formula, the method and duration of administration,
and contraindications. It is the responsibility of practitioners, relying on their own experience and
knowledge of their patients, to make diagnoses, to determine dosages and the best treatment for each
individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume
any liability for any injury and/or damage to persons or property as a matter of products liability,
negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.

Previous editions copyrighted 2010 and 2002.

Library of Congress Cataloging-in-Publication Data

Names: Manning, Warren J., 1957- editor. | Pennell, Dudley J., 1958- editor.
Title: Cardiovascular magnetic resonance : a companion to Braunwald’s heart disease / [edited by] Warren J.
Manning, Dudley J. Pennell.
Other titles: Cardiovascular magnetic resonance (Manning) | Complemented by (expression): Braunwald’s
heart disease. 11th edition.
Description: Third edition. | Philadelphia, PA : Elsevier, [2019] | Complemented by: Braunwald’s heart disease /
edited by Douglas P. Zipes, Peter Libby, Robert O. Bonow, Douglas L. Mann, and Gordon F. Tomaselli. 11th
ed. 2018. | Includes bibliographical references and index.
Identifiers: LCCN 2018006374 | ISBN 9780323415613 (hardcover : alk. paper)
Subjects: | MESH: Cardiovascular Diseases–diagnosis | Magnetic Resonance Imaging–methods | Diagnostic
Techniques, Cardiovascular
Classification: LCC RC683.5.M35 | NLM WG 141.5.M2 | DDC 616.1/207548–dc23 LC record available at
https://lccn.loc.gov/2018006374

Executive Content Strategist: Robin Carter


Senior Content Development Specialist: Jennifer Ehlers
Publishing Services Manager: Catherine Jackson
Book Production Specialist: Kristine Feeherty
Design Direction: Renee Duenow

Printed in China

Last digit is the print number: 9 8 7 6 5 4 3 2 1


To the joys, inspirations, and blessings of my life—
mom and dad, Susan Gail, Anya and Elie, Sara
and Jeremy, Isaac, Raziel, and JJ.

—WJM

To the memory of my parents, Terence and Joan;


the love of my wife, Elisabeth;
and the joy of my daughter, Indigo.

—DJP
CONTRIBUTORS
Mehmet Akçakaya, PhD Craig Ronald Butler, MD, MSc
Department of Electrical and Computer Engineering Associate Professor of Medicine
Center for Magnetic Resonance Research Division of Cardiology
University of Minnesota University of Alberta
Minneapolis, Minnesota Edmonton, Alberta, Canada

Francisco Alpendurada, MD, PhD Peter Caravan, PhD


Consultant A.A. Martinos Center for Biomedical Imaging
CMR Unit, Royal Brompton Hospital Massachusetts General Hospital
London, United Kingdom Boston, Massachusetts

Evan Appelbaum, MD Csilla Celeng, MD, PhD


Department of Medicine Department of Radiology
Cardiovascular Division University Medical Center Utrecht
Beth Israel Deaconess Medical Center Utrecht, The Netherlands
Harvard Medical School
Boston, Massachusetts Michael L. Chuang, MD, ScM
Cardiovascular Imaging Core Laboratory
Andrew Arai, MD Beth Israel Deaconess Medical Center
Chief, Advanced Cardiovascular Imaging Laboratory Boston, Massachusetts
National Institutes of Health
National Heart, Lung, and Blood Institute Albert de Roos, MD, PhD
Bethesda, Maryland Professor of Radiology
Department of Radiology
Dominique Auger, MD, PhD Leiden University Medical Center
Adjunct Professor of Medicine Leiden, The Netherlands
Cardiology
Centre Hospitalier de l’Université de Montréal; Victoria Delgado, MD, PhD
Associate Professor of Medicine Consultant Cardiologist
McGill University Health Center Department of Cardiology
Montreal, Quebec, Canada Leiden University Medical Center
Leiden, The Netherlands
Robert S. Balaban, PhD
Scientific Director, NHLBI Rohan Dharmakumar, PhD
Laboratory of Cardiac Energetics Cedars-Sinai Medical Center
National Heart, Lung, and Blood Institute University of California
National Institutes of Health Los Angeles, California
Bethesda, Maryland
Marc R. Dweck, MD, PhD
Jeroen J. Bax, MD, PhD Translational and Molecular Imaging Institute
Professor of Cardiology Zena and Michael A. Wiener Cardiovascular Institute
Department of Cardiology Icahn School of Medicine at Mount Sinai
Leiden University Medical Center New York, New York;
Leiden, The Netherlands Centre for Cardiovascular Science
University of Edinburgh
Nicholas G. Bellenger, MD Edinburgh, United Kingdom
Consultant Cardiologist
Royal Devon and Exeter Hospital Afshin Farzaneh-Far, MD, PhD
Exeter, United Kingdom Section of Cardiology
Department of Medicine
David A. Bluemke, MD, PhD University of Illinois at Chicago
Department of Radiology Chicago, Illinois;
School of Medicine and Public Health Department of Medicine
University of Wisconsin-Madison Division of Cardiology
Madison, Wisconsin Duke University
Durham, North Carolina
René M. Botnar, PhD
Division of Imaging Sciences and Biomedical Engineering
King’s College London
London, United Kingdom

vii
viii CONTRIBUTORS

Zahi A. Fayad, PhD Brian P. Halliday, MBChB (Hons), MRCP


Translational and Molecular Imaging Institute BHF Clinical Research Fellow
Zena and Michael A. Wiener Cardiovascular Institute National Heart and Lung Institute
Icahn School of Medicine at Mount Sinai Imperial College London;
New York, New York Clinical Research Fellow
Cardiovascular Magnetic Resonance Unit and Cardiovascular
David Firmin, PhD Research Centre
Director of Physics Royal Brompton and Harefield NHS Trust
CMR Unit London, United Kingdom
Royal Brompton Hospital
Professor of Biomedical Imaging Michael Hansen, PhD
National Heart and Lung Institute Investigator
Imperial College London National Heart, Lung, and Blood Institute
London, United Kingdom National Institutes of Health
Bethesda, Maryland
Mark A. Fogel, MD, FACC, FAHA, FAAP, FNASCI
Professor of Pediatrics (Cardiology) and Radiology Thomas H. Hauser, MD, MMSc
The Perelman School of Medicine at the University of Pennsylvania Director of Nuclear Cardiology
Director of Cardiac Magnetic Resonance Beth Israel Deaconess Medical Center
The Children’s Hospital of Philadelphia Assistant Professor of Medicine
Philadelphia, Pennsylvania Harvard Medical School
Boston, Massachusetts
Herbert Frank, MD
Professor of Internal Medicine Susie N. Hong, MD, MSc
Director, Department of Internal Medicine Assistant Professor
University Hospital Tulln and Medical University of Vienna Department of Medicine
Tulln, Austria Division of Cardiovascular Medicine
Assistant Professor
Matthias G. Friedrich, MD Department of Diagnostic Radiology and Nuclear Medicine
Professor of Medicine and Diagnostic Radiology University of Maryland Medical Center
McGill University; Baltimore, Maryland
Adjunct Professor of Radiology
Université de Montréal Till Huelnhagen, Dipl.-Ing.
Montreal, Quebec, Canada; Berlin Ultrahigh Field Facility (B.U.F.F.)
Professor of Medicine Max-Delbrueck Center for Molecular Medicine in the Helmholtz
Heidelberg University Association
Heidelberg, Germany; Berlin, Germany
Adjunct Professor
Cardiac Sciences and Radiology W. Gregory Hundley, MD
University of Calgary Department of Internal Medicine (Cardiology Section)
Calgary, Alberta, Canada Department of Radiology
Wake Forest University School of Medicine
Eric M. Gale, PhD Winston-Salem, North Carolina
Assistant Professor in Radiology
A.A. Martinos Center for Biomedical Imaging El-Sayed H. Ibrahim, PhD
Massachusetts General Hospital Medical College of Wisconsin
Boston, Massachusetts Milwaukee, Wisconsin

Tal Geva, MD Esra Gucuk Ipek, MD


Cardiologist-in-Chief Department of Medicine
Department of Cardiology Section for Cardiac Electrophysiology
Boston Children’s Hospital Johns Hopkins University School of Medicine
Professor of Pediatrics Baltimore, Maryland
Harvard Medical School
Boston, Massachusetts Michael Jerosch-Herold, PhD
Department of Radiology
John D. Grizzard, MD Brigham and Women’s Hospital and Harvard Medical School
Department of Radiology Boston, Massachusetts
VCU Health Systems
Richmond, Virginia Robert M. Judd, PhD
Duke Cardiovascular Magnetic Resonance Center
Duke University Medical Center
Durham, North Carolina
CONTRIBUTORS ix

Jennifer Keegan, PhD Eric V. Krieger, MD


Principal Physicist Associate Professor of Medicine
Cardiovascular Research Centre Department of Medicine
Royal Brompton Hospital Division of Cardiology
Adjunct Reader University of Washington School of Medicine
National Heart and Lung Institute University of Washington Medical Center
Imperial College London Seattle Children’s Hospital
London, United Kingdom Seattle, Washington

Peter Kellman, PhD Raymond Y. Kwong, MD, MPH


Staff Scientist Department of Medicine
National Heart, Lung, and Blood Institute Cardiovascular Division
National Institutes of Health Brigham and Women’s Hospital
Bethesda, Maryland Harvard Medical School
Boston, Massachusetts
Muhammad Shahzeb Khan, MD
Department of Internal Medicine Jay S. Leb, MD
John H. Stroger Jr. Hospital of Cook County Assistant Professor
Chicago, Illinois Department of Radiology
Columbia University Medical Center
Faisal Khosa, MD, MBA, FFRRCSI, FRCPC, DABR New York, New York
Assistant Professor
Department of Radiology Robert Lederman, MD
Vancouver General Hospital Cardiovascular Branch
Vancouver, British Columbia, Canada Division of Intramural Research
National Heart, Lung, and Blood Institute
Kiran Khurshid, MBBS, MD National Institutes of Health
Research Fellow Bethesda, Maryland
Department of Radiology
Vancouver General Hospital Tim Leiner, MD, PhD, EBCR
Vancouver, British Columbia, Canada Professor of Radiology
Department of Radiology
Philip J. Kilner, MD University Medical Center Utrecht
Consultant, CMR Unit Utrecht, The Netherlands
Royal Brompton Hospital
London, United Kingdom Debiao Li, PhD
Cedars-Sinai Medical Center
Daniel H. Kim, MD University of California
Professor of Medicine Los Angeles, California
Department of Medicine
Division of Cardiology David Lopez, MD
University of Alberta Heart and Vascular Institute
Edmonton, Alberta, Canada Cleveland Clinic
Weston, Florida
Raymond J. Kim, MD
Duke Cardiovascular Magnetic Resonance Center Alicia M. Maceira, MD, PhD, FESC
Duke University Medical Center Director, Cardiovascular Imaging Unit
Durham, North Carolina ERESA Medical Center
Valencia, Spain
W. Yong Kim, PhD
Department of Cardiology Heiko Mahrholdt, MD
Aarhus University Hospital Department of Cardiology
Aarhus, Denmark Robert-Bosch-Krankenhaus
Stuttgart, Germany
Christopher M. Kramer, MD
Ruth C. Heede Professor of Cardiology Marcus R. Makowski, MD
Professor of Radiology Department of Radiology
University of Virginia Health System The Charité
Charlottesville, Virginia Berlin, Germany
x CONTRIBUTORS

Warren J. Manning, MD, FACC, FACP, FAHA, FISMRM Stefan Neubauer, MD, FRCP, FACC, FMedSci
Professor of Medicine and Radiology Professor of Cardiovascular Medicine
Harvard Medical School Head, Division of Cardiovascular Medicine
Section Chief, Non-invasive Cardiac Imaging and Testing Director, Oxford Centre for Clinical Magnetic Resonance Research
Cardiovascular Division Division of Cardiovascular Medicine
Beth Israel Deaconess Medical Center Radcliffe Department of Medicine
Boston, Massachusetts University of Oxford
John Radcliffe Hospital
Constantin B. Marcu, MD Oxford, United Kingdom
Associate Professor of Cardiology
Director of Advanced Cardiac Imaging and Imaging Education Reza Nezafat, PhD
Cardiovascular Sciences Department of Medicine
Brody School of Medicine at East Carolina Cardiovascular Division
Greenville, North Carolina Beth Israel Deaconess Medical Center
Harvard Medical School
Martin S. Maron, MD Boston, Massachusetts
Hypertrophic Cardiomyopathy Center
Division of Cardiology Christoph A. Nienaber, MD, PhD
Tufts Medical Center Royal Brompton & Harefield Hospital NHS Foundation Trust/
Boston, Massachusetts; Imperial College
Chanin T. Mast Center for Hypertrophic Cardiomyopathy Cardiology and Aortic Centre
Morristown Medical Center London, United Kingdom
Morristown, New Jersey
Thoralf Niendorf, PhD
Raad H. Mohiaddin, MD Berlin Ultrahigh Field Facility (B.U.F.F.)
Professor of Cardiovascular Imaging Max Delbrueck Center for Molecular Medicine in the Helmholtz
Royal Brompton Hospital and Harefield NHS Trust Association
National Heart and Lung Institute DZHK (German Centre for Cardiovascular Research) Partner Site
Imperial College London MRI.TOOLS GmbH
London, United Kingdom Berlin, Germany

James C. Moon, MD, MRCP Sara L. Partington, MD


Professor Assistant Professor of Clinical Medicine
Institute of Cardiovascular Science Philadelphia Adult Congenital Heart Disease Center
University College London The Hospital of the University of Pennsylvania and the Children’s
Advanced Cardiac Imaging Hospital of Philadelphia
Barts Heart Centre Perelman Center for Advanced Medicine
London, United Kingdom Philadelphia, Pennsylvania

Manish Motwani, MB, ChB, PhD Ian Paterson, MD


Consultant Cardiologist Department of Medicine
Manchester Heart Centre Division of Cardiology
Manchester University NHS Foundation Trust University of Alberta
Manchester, United Kingdom Edmonton, Alberta, Canada

Shiro Nakamori, MD, PhD Katharina Paul, PhD


Postdoctoral Research Fellow Berlin Ultrahigh Field Facility (B.U.F.F.)
Harvard Medical School Max-Delbrueck Center for Molecular Medicine in the Helmholtz
Department of Medicine Association
Cardiovascular Division Berlin, Germany
Beth Israel Deaconess Medical Center
Boston, Massachusetts Dudley J. Pennell, MD, FRCP, FACC, FESC, FAHA, FRCR, FMedSci
Professor of Cardiology
Saman Nazarian, MD, PhD National Heart and Lung Institute
Associate Professor of Medicine Imperial College London;
Cardiac Electrophysiology Director, Cardiovascular Magnetic Resonance Unit
University of Pennsylvania Perelman School of Medicine Royal Brompton Hospital
Philadelphia, Pennsylvania London, United Kingdom

Felix Nensa, MD Ronald M. Peshock, MD


Department of Diagnostic and Interventional Radiology and Professor of Internal Medicine and Radiology
Neuroradiology Vice Chair of Information Technology
University Hospital Essen University of Texas Southwestern Medical Center
Essen, Germany Dallas, Texas
CONTRIBUTORS xi

Dana C. Peters, PhD Reza S. Razavi, MD


Magnetic Resonance Research Center Professor, Paediatric Cardiovascular Science
Associate Professor of Radiology and Biomedical Imaging School of Biomedical Engineering and Imaging Sciences
Yale University King’s College London;
New Haven, Connecticut Consultant Paediatric Cardiologist
Evelina London Children’s Hospital
R. Nils Planken, MD, PhD, EBCR London, United Kingdom
Department of Radiology and Nuclear Medicine
Academic Medical Center Wolfgang G. Rehwald, PhD
Amsterdam, The Netherlands Duke Cardiovascular Magnetic Resonance Center
Siemens Healthineers
Sven Plein, MD, PhD Durham, North Carolina
Professor of Cardiology
British Heart Foundation Professor of Cardiovascular Imaging Philip M. Robson, PhD
Honorary Consultant Cardiologist Translational and Molecular Imaging Institute
Division of Biomedical Imaging Zena and Michael A. Wiener Cardiovascular Institute
Leeds Institute of Cardiovascular and Metabolic Medicine Icahn School of Medicine at Mount Sinai
(LICAMM) New York, New York
University of Leeds
Leeds, United Kingdom Christopher T. Rodgers, MChem, DPhil
Associate Professor of Biomedical Imaging
Andrew J. Powell, MD Division of Cardiovascular Medicine
Chief, Cardiac Imaging Division Radcliffe Department of Medicine
Department of Cardiology University of Oxford
Boston Children’s Hospital John Radcliffe Hospital
Associate Professor of Pediatrics Oxford, United Kingdom
Harvard Medical School
Boston, Massachusetts Toby Rogers, BM BCh, PhD
Cardiovascular Branch
Sanjay Prasad, MD, FRCP, FESC Division of Intramural Research
Consultant Cardiologist National Heart, Lung, and Blood Institute
CMR Unit National Institutes of Health
Royal Brompton Hospital Bethesda, Maryland
Honorary Senior Lecturer
Cardiovascular Biomedical Research Unit Ethan J. Rowin, MD
Royal Brompton Hospital and Imperial College London Hypertrophic Cardiomyopathy Center
London, United Kingdom Division of Cardiology
Tufts Medical Center
Claudia Prieto, PhD Boston, Massachusetts;
School of Biomedical Engineering and Imaging Sciences Chanin T. Mast Center for Hypertrophic Cardiomyopathy
King’s College London Morristown Medical Center
St Thomas’ Hospital Morristown, New Jersey
London, United Kingdom
James H.F. Rudd, MD, PhD
Kuberan Pushparajah, BMBS, BMedSci, MD(Res) Division of Cardiovascular Medicine
Consultant Paediatric Cardiologist University of Cambridge
Paediatric Cardiology Cambridge, United Kingdom
Evelina London Children’s Hospital;
Clinical Senior Lecturer Hajime Sakuma, MD, PhD
Division of Imaging Sciences and Biomedical Engineering Professor and Chairman of Radiology
King’s College London Mie University Hospital
London, United Kingdom Mie University Graduate School/Faculty of Medicine
Mie, Japan
Imran Rashid, MBBS, PhD
School of Biomedical Engineering and Imaging Sciences Michael Salerno, MD, PhD
King’s College London Associate Professor
St Thomas’ Hospital Departments of Medicine and Radiology
London, United Kingdom University of Virginia Health System
Charlottesville, Virginia
xii CONTRIBUTORS

Thomas Schlosser, MD Anne Marie Valente, MD


Professor Associate Professor, Pediatrics and Internal Medicine
Institute of Diagnostic and Interventional Radiology and Harvard Medical School;
Neuroradiology Staff Cardiologist, Pediatric Cardiology
University Hospital Essen Children’s Hospital Boston;
Essen, Germany Staff Cardiologist, Internal Medicine
Division of Cardiology
Juerg Schwitter, MD Brigham and Women’s Hospital
Professor Boston, Massachusetts
Cardiovascular Department
Division of Cardiology Harrie van den Bosch, MD
Director, Cardiac MR Center of the CHUV Department of Radiology
University Hospital Lausanne, CHUV Catharina Hospital
Lausanne, Switzerland Eindhoven, The Netherlands

Udo P. Sechtem, MD Pieter van der Bijl, MB, ChB, MMed


Chairman, Department of Cardiology Research Fellow
Robert-Bosch-Krankenhaus Department of Cardiology
Stuttgart, Germany; Leiden University Medical Center
Associate Professor of Medicine and Cardiology Leiden, The Netherlands
University of Tubingen
Tubingen, Germany Albert C. van Rossum, MD, PhD
Chair, Department of Cardiology
R. Brandon Stacey, MD, MS VU University Medical Center
Department of Internal Medicine (Cardiology Section) Amsterdam, The Netherlands
Wake Forest University School of Medicine
Winston-Salem, North Carolina David C. Wendell, PhD
Duke Cardiovascular Magnetic Resonance Center
Matthias Stuber, PhD Duke University Medical Center
University of Lausanne Durham, North Carolina
Lausanne, Switzerland
Jos J.M. Westenberg, PhD
Teresa Sykora, MD Associate Professor
Consultant Department of Radiology
Department of Internal Medicine Leiden University Medical Center
University Hospital Tulln and Medical University of Vienna Leiden, The Netherlands
Tulln, Austria
Mark A. Westwood, MA, MD, FRCP
Upasana Tayal, BMBCh, MRCP Consultant Cardiologist
MRC Clinical Research Fellow Department of Cardiology
National Heart and Lung Institute Barts Heart Centre
Imperial College London London, United Kingdom
London, United Kingdom
Norbert Wilke, MD
Anneline S.J.M. te Riele, MD, PhD Department of Radiology
Department of Medicine Krankenhaus St. Anna
Division of Cardiology Höchstadt an der Aisch, Germany
University Medical Center Utrecht
Netherlands Heart Institute Lukas Winter, R.rer.nat, Dipl.-Ing
Utrecht, The Netherlands Berlin Ultrahigh Field Facility (B.U.F.F.)
Max-Delbrueck Center for Molecular Medicine in the Helmholtz
Thomas A. Treibel, PhD Association
Professor Berlin, Germany
Institute of Cardiovascular Science
University College London Hsin-Jung Yang, PhD
Advanced Cardiac Imaging Cedars-Sinai Medical Center
Barts Heart Centre University of California
London, United Kingdom Los Angeles, California

Sotirios A. Tsaftaris, PhD


University of Edinburgh
Edinburgh, United Kingdom
F O R E WO R D

In 1900 the first Nobel Prize in Physics was awarded to Dr. Wilhelm does not require ionizing radiation, can reduce the need for coronary
Roentgen. His monumental discovery provided, for the first time, images angiography, now performed on more than 1.5 million patients each
of the heart in intact patients. Roentgenology, named in honor of its year in the United States alone.
discoverer, ranks high among the greatest advances of modern medi- Just as it became necessary for the pioneer cardiologists of the early
cine. The two specialties of Imaging Sciences and Cardiology developed 20th century to become familiar with cardiac radiology, it is now equally
hand-in-hand during the first half of the twentieth century. Imaging important for their successors to do the same with CMR. Warren J.
progressed with a variety of technologies that included angiography, Manning and Dudley J. Pennell, two distinguished leaders of this impor-
echocardiography, radionuclide imaging, computed tomography, pos- tant technology, and their talented contributors to Cardiovascular Mag-
itron emission tomography, and cardiovascular magnetic resonance netic Resonance have taken an important step toward enabling clinicians
(CMR). and clinical investigators to accomplish this. They deserve the apprecia-
Of these, CMR offers a veritable treasure trove of information about tion of cardiovascular specialists, radiologists, and trainees for producing
the heart and great vessels. It not only provides excellent three-dimen- this detailed and authoritative yet eminently readable book. We are
sional displays of the structure and function of the cardiac chambers very pleased to welcome this third edition of Cardiovascular Magnetic
and valves, but it is also useful in the assessment of the normal and Resonance into the family of companions to Heart Disease: A Textbook
diseased aorta. The lumina, walls, and obstructive lesions in the major of Cardiovascular Medicine.
coronary arteries can be visualized, and myocardial perfusion assessed.
Myocardial ischemia, necrosis, infiltration, and fibrosis can be detected Eugene Braunwald, MD
and quantified. When combined with the ability to determine myocardial Peter Libby, MD
viability and to measure cardiac high-energy phosphate stores nonin- Robert O. Bonow, MD, MS
vasively by CMR spectroscopy, the assessment of patients with ischemic Douglas L. Mann, MD
heart disease is greatly enhanced. This noninvasive technique, which Gordon F. Tomaselli, MD

xiii
P R E FA C E

Cardiovascular magnetic resonance (CMR) is a medical imaging field growing field in mainstream cardiology. Our aim is to continue to
that excites great interest because it combines superb image quality provide instruction in the current clinical practice of CMR, while also
with new techniques for probing the cardiovascular system in novel highlighting areas of clinical potential, which are presently in varying
ways. What surprises is the versatility of the technology: blood flow, stages of development. If we succeed in drawing new investigators and
angiography, assessment of atherosclerosis, myocardial perfusion, focal clinicians to enter the field or in illuminating new areas for those already
necrosis, tissue characterization, oxygen saturation, spectroscopy, tech- involved, then we will have achieved our objective—the healthy growth
nology, and chemical composition are among the measurements that of competent and motivated practitioners in CMR for the benefit of
are being refined for clinical use, in addition to the well-known “gold clinical science, patient care, and the advancement of the field.
standard” capabilities of CMR in defining anatomy, fibrosis, ventricular The reader should be forewarned that CMR is constantly developing,
function, and blood flow. Such potential comes at a price, however, as and no printed text can include all of the most recent developments;
this technology is not quickly learned, and high-quality clinical practice however, the foundations provided in these pages will serve the reader
needs experience. Professional didactic and clinical training is required for many years to come. The future of CMR remains very bright. Join
for all newcomers to the field and to maintain cutting-edge competency. us on the journey!
In the third edition of this textbook, we are excited to have partnered
with the exceptional Braunwald companion series. This brings Cardio- Warren J. Manning, MD
vascular Magnetic Resonance to a larger audience and cements our Dudley J. Pennell, MD

xv
AC K N OW L E D G M E N T S

It takes a village to create a book. Like any large endeavor, this text is a success because of the efforts of the
many individuals to whom we owe great thanks. These include the outstanding contributions of our primary
authors and their collaborators; Robin Carter, our editor at Elsevier; Kristine Feeherty, our project manager;
our office assistants, Lillian Robles at the Beth Israel Deaconess Medical Center, Boston, and Fei Wang at
the Royal Brompton Hospital, London; the multitude of CMR mentors, trainees, and colleagues who have
stimulated and educated us over the past three decades; and most of all our families for allowing us the
time to pursue this endeavor among the myriad other activities that occupy our daily lives. To all, we express
our thanks and appreciation.

Warren J. Manning, MD
Dudley J. Pennell, MD

xvii
SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

1 
Basic Principles of Cardiovascular
Magnetic Resonance
Robert S. Balaban and Dana C. Peters

property of the spins in a magnetic field and is defined by the Larmor


INTRODUCTION
equation:
This introduction to the basic principles of cardiovascular magnetic
γ
resonance (CMR) describes the concepts of magnetization, T1, T2, ν= B Eq. 1.1

T2*, and image formation, and describes some common CMR pulse
sequences and parameters. These basic ideas will require some time where B is the magnetic field strength experienced by the spin, which
and rereading to fully appreciate, but each new concept will add to the is approximately B0, but includes any variations as described below,
understanding of CMR. v is the spin’s precessional frequency in cycles/s or hertz (Hz), and
The human body is composed of mostly water, and a lot of fat. γ (gamma) is the gyromagnetic ratio, which is a constant related to
The water (H2O) and fat contain many hydrogen atoms. Hydrogen the mass and charge of the water proton, γ/2π = 42.58 × 106 Hz Tesla
atoms in turn are made up of a proton (1H, the hydrogen nucleus) for water protons. The precessional frequency for water protons at
and an electron. Hydrogen protons are in very high concentration in 1.5 Tesla (T; a common CMR field strength) is ν = 63.87 × 106 Hz,
the body, roughly ~100 molar.a These hydrogen protons within the or roughly 64 MHz (about the frequency of an FM radio station and
body can be imaged through magnetic resonance imaging (MRI). one of the reasons the CMR environment must be shielded from FM
MRI is possible because hydrogen protons have “spin” and all nuclei radio waves). Thus, at 3 T, the precessional frequency is twice this,
with spin interact with magnetic fields. Spin is called the “intrinsic ν = 127.74 × 106 Hz (Eq. 1.1).
angular momentum,” and it is a fundamental property of protons. The Larmor equation (see Eq. 1.1) looks simple, but it is the basis of
MRI physics is often called “spin physics” because it describes how CMR imaging. To determine the location of different magnetic spins,
the proton spins are used to image the body. In this chapter we the magnetic field (B) is made to vary (linearly) with position using
will use the term spin to indicate the hydrogen proton’s spin. MRI specialized coils of wire (called “gradient coils”) inside the magnet.
images these spins, and in this way it images the tissues in which they This results in a precessional frequency (v) that depends on a spin’s
are embedded. location in the scanner. By measuring the number of spins precessing
In the absence of a magnetic field, the spins are randomly oriented at each location (i.e., frequency), a magnetic resonance (MR) image is
(Fig. 1.1A). But if placed in a large magnetic field (called B0; i.e., created1 as will be discussed below.
the CMR scanner), the water spins partly align in the same orienta-
tion of this applied magnetic field much like iron filings (Fig. 1.1B), DETECTION OF THE MRI SIGNAL
with larger magnetic fields causing greater alignment of the spins.
In fact, the bulk magnetization, Mz, is proportional to the strength Alignment With the Main Magnetic Field
of the applied field. Unlike iron filings, however, the interaction How can the MRI signal from water spins be detected in order to image
of the spins with the B0 field results in the spins rotating around the sample? Remember, when a sample (i.e., a patient) is placed in the
the axis of the magnetic field (Fig. 1.1C). This is why the hydro- main magnetic field (i.e., inside the bore of the magnet), the hydrogen
gen nuclei in MRI are also referred to as spins: they spin (precess) spins align with that field (see Fig. 1.1B) and begin to precess at the
around the B0 field. The frequency of precession (v) is a very important Larmor frequency. We call the direction of the main magnetic field,
B0, the Z-axis direction or the longitudinal direction. Since the spins
partially align with the B0 field, they create a net magnetic field along
the Z axis (oriented parallel with B0). However, the spins precess or
rotate around the Z axis in a random, incoherent fashion, resulting
a
The molarity of 1H can be estimated as ~(2 moles hydrogen/mole in no net magnetization in the X-Y plane (also called the transverse
H2O) × (1 mole H2O/18 g of tissue) × 1000 g/L (density of the body) plane) (see Fig. 1.1C). Magnetization by the B0 field (i.e., Mz) is not
~ 100 mole/L. enough to generate an MR signal. The MR signal is measured as the

1
2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

No net Net magnetization Random phase of


Z
magnetization Z individual spins with
net 1H magnetic field

X
X
X
Y
Y
Y
B0 = 0 B0 B0
A B C

FIG. 1.1  Orientation of molecular spins. (A) With no magnetic field, the spins are oriented randomly, produc-
ing no net magnetization. (B) The spins orient with an applied magnetic field (B0), producing a net magnetic
field aligned along B0. (C) The magnetized spins rotate around the B0 field (the Z axis) in a random way,
resulting in no net magnetization in the X-Y plane.

Now coherent
spins rotate
around Z (B0)
Z B0

Final
magnetization

Z B0 Receiver
Initial magnetization X
(“longitudinal” coil
or MZ) Initial
magnetization
Pulsed Y
B1 field
oscillating
at Larmor
frequency 90°

B1 X Voltage
Final
Free induction decay
magnetization
(“transverse or Mxy) (FID) − oscillating at
Y Time Larmor frequency
A RF excitation coil B
FIG. 1.2  Effects of a perpendicular B1 magnetic field. (A) The B1 field oscillating at the Larmor frequency of
the spins is absorbed by the spins and causes a rotation around the B1 field axis (Y axis). A 90-degree RF
pulse is shown in which longitudinal magnetization (Mz) becomes transverse magnetization (Mxy). (B) Once
the B1 field is removed, the spins continue to rotate around the Z axis at the Larmor frequency, but the
signal decays (in the X-Y plane) as equilibrium is reestablished. The result is a coherent oscillating magnetic
field that is detected with the coil as the free induction decay (FID). The FID is a decaying sinusoidal voltage
signal with a frequency equal to the Larmor frequency of the spins. RF, Radiofrequency.

magnetization on the X-Y plane (called Mxy). The MR signal is detected powerful) magnetic field, with a strength that oscillates at the Larmor
by placing a receiver coil (loops of wire tuned to receive signals oscil- frequency is applied perpendicular to the main magnetic field (Fig.
lating at the Larmor frequency) near the subject to detect any spin 1.2A). This process is called radiofrequency (RF) excitation, and the
precession in the X-Y plane. A receiver coil, placed to detect the preces- field is called an RF pulse or a B1 field, since it uses a magnetic (B1)
sion of spins in the X-Y plane, will not detect any signal if Mxy is zero field rotating at a high (radio wave) frequency. The frequency of this
(see Fig. 1.1C). perpendicular B1 field has to precisely match the Larmor frequency of
the water to excite the spins in the presence of the stronger B0 field.
Radiofrequency Excitation (This concept is analogous to exciting a piano string with a tuning fork.
To create an MR signal, the water spins must be precessing in a coher- Only the string with the same resonant frequency as the tuning fork
ent manner in the X-Y plane. To accomplish this task, another (less will efficiently absorb the energy from the fork and resonate. In fact,
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 3

magnetic resonance imaging gets its name from the resonant frequency myocardial infarction and inflammatory diseases such as myocarditis. T1
used for RF excitation.) The excitation field (B1) has a strength of only a is also longer in amyloid cardiomyopathy3 and Anderson-Fabry disease.
few gauss, compared to the B0 field, which has a strength of 15,000 gauss In siderotic disease (iron overload), native myocardial T1 is shorter.4
at 1.5 T (1 T = 10,000 gauss). In addition, the B1 field is only applied
transiently (for milliseconds), long enough to temporarily deflect the T2* and T2 Relaxation: The Effects of Spin Phase
spins into a plane perpendicular to B0. This effect on the net magnetic The other form of relaxation, T2* (T2 star) relaxation, is that of the
field is illustrated in Fig. 1.2A, which shows a B1 pulse that drives the randomization of the phase of the spins. After RF excitation, all of the
magnetization completely into the transverse plane (X-Y plane). This spins precess about the Z axis, with slight differences in the precessional
is called a 90-degree pulse (or saturation pulse), referring to the angle rate, and this eventually results in “T2* decay.” T2* decay comes from
through which the initial magnetization (Mz) moves relative to the Z differences in the phase of each spin, so that the net X-Y magnetization,
axis. However, in practice, the B1 pulse can be used to flip the mag- Mxy, decays to 0. The phase indicates the direction of the spin in the
netization by any angle from 0 to 180 degrees. A small B1 pulse (<90 transverse plane. The phase (ϕ) of a spin in the transverse plane depends
degrees) is called an alpha pulse, while a 180-degree pulse is called an on its initial phase, ϕ0, the precessional frequency of that spin, and the
inversion pulse. A 180-degree flip is achieved by applying the B1 field time, t, that it has spent in the transverse plane (assuming a constant
for twice as long or with twice the strength as the 90-degree pulse. The frequency).
RF pulse is usually directed along the X or Y axis and oscillates at the φ = φ0 + 2πνt Eq. 1.3
Larmor frequency, with an amplitude and shape that determine the true
flip angle. Once the B1 field is turned off, the only field affecting the Since this precessional frequency depends on the magnetic field (see
magnetization is the B0 field. Now the net magnetization of the water Eq. 1.1), which changes slightly with location, time, and molecular
proton spin is in the transverse plane and can be detected as an oscil- environment, each spin on the transverse plane has a different frequency,
lating signal in the receiver coil (Fig. 1.2B), precessing at the Larmor and thus phase, and this phase difference generally increases with time
frequency. This is the signal measured in MRI. (Eq. 1.3). Fig. 1.3A defines the phase (ϕ) of the spin magnetic field
After the RF excitation, the water spins will rotate around the B0 vector as the direction of the transverse magnetization relative to the
field on the X-Y plane. Fig. 1.2B shows the signal detected by the receiver X axis.
coil as the spins precess in the X-Y plane. The signal oscillates at the In Fig. 1.3B a phase diagram is used to show the changing phase
Larmor frequency and decays in amplitude with time; it is called a free relationships for three spins in a sample that are precessing at different
induction decay (FID). The decay in net magnitude in the transverse frequencies. The magnetic field always varies because of imperfections
plane is known as magnetic relaxation. The decay occurs because the in the main magnetic field and changes in the molecular environment.
spins, having absorbed energy from the transient B1 field, which is now In this schematic, the three spins experience slightly different magnetic
turned off, return to their original state in equilibrium with the B0 field, fields, one higher than B0, one exactly B0, and one lower. Therefore,
with spins partially aligned along the Z axis (see Fig. 1.1C). This occurs after some time on the X-Y plane, each spin experiences phase accrual,
via two extremely important processes called T1 and T2 relaxation. until the net magnetization (the sum of all of the spins magnetization)
Nature abhors order and seeks to minimize the energy in the system decreases to 0.
by returning to equilibrium with the main magnetic field, B0, through Within a single pixel in a CMR image, there are a quintillion water
T1 and T2 relaxation. T1 and T2 relaxation are the foundations of MRI spins, each with a slightly different frequency. Therefore, while all of
because they are responsible for MRI’s excellent contrast. the spins are precessing at about the Larmor frequency, at any time one
spin might be precessing a little faster and another might be precessing
T1 Relaxation a little slower.
T1 relaxation, also known as spin-lattice relaxation, is the release of energy T2* relaxation can be understood as the characteristic time in which
to the environment, or lattice, that results in the reestablishment of the the spins in the (X-Y) plane become so out of phase that the signal
magnetization along the Z axis. Thus T1 is the time constant by which decays (dies out). The signal decays because the relative phase of the
the longitudinal (or Z) magnetization relaxes to its equilibrium value, spins in a single voxel are different. After the initial excitation using a
M0. After a 90-degree RF pulse, the Z magnetization, Mz(t), is initially B1 pulse, all of the spins on the transverse plane have identical phase
0 but regrows with a relaxation time T1 to its equilibrium value (M0). but then begin to dephase quickly. This dephasing is described by a
time constant T2*. The dephasing causes the bulk transverse magnetiza-
M z (t ) = M 0 (1− e−t T1) Eq. 1.2
tion, Mxy, to decay and approach 0, due to incoherent phase.
where t is the time after the 90-degree pulse. Each tissue type has a
M xy (t ) = M xy 0 e−t T 2*
Eq. 1.4
unique T1, which changes with field strength and in disease. Table 1.1
gives values for the normal T1 of many cardiac tissues, although refer- Thus the CMR signal (Mxy) decays in a simple exponential way, with
ence values vary depending on the method of T1 quantification. Studies2 a time constant of T2*.
using T1 mapping show that native T1 is longer than normal in acute
T2 Versus T2*
We need to further understand T2* and how it differs from T2. Both
are related to dephasing due to variability in the main magnetic field.
TABLE 1.1  T1 and T2 Values In general, two distinct processes contribute to the dephasing of spins
Field Strength 1.5 T 3 T in a voxel. The first mechanism is the true T2 decay that is unavoidable,
Tissue T1 (ms) T2 (ms) T1 (ms) T2 (ms) irreversible, and dependent on the molecular interactions within the
Myocardium 95071 5572 115073 4074 sample. T2 relaxation is also called spin-spin relaxation. It is called this
Arterial blood 155071 250 165075 200 because the mechanism of the relaxation process is through the inter-
Fat 260 110 320 70 action of spins in the sample with each other at or below the Larmor
Skeletal muscle 100076 4576 140076 5076 frequency, making this process dependent on the microscopic motions
within the sample. The second process is T2* decay and includes T2
4 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Low B field Spin 2


Spin 1
Center B field
Spin 3
X-Y plane phase diagram High B Field
Y
y
90°
Spin 3
Spin 3
Mxy M x y =0
Mxy
X Y X
Spin 2
φ X
Spin 2

φ Spin 1 Spin 1
180° 0°
Immediately
after 90° pulse, t = 0. Time t later Time 2t later
Z All spins in phase. Spins are out of
phase.
270° B Time
A
FIG. 1.3  The phase diagram. (A) A phase diagram is a view of the magnetization vectors projected into the
X-Y plane. (B) The magnetic field of a sample varies as shown. Spins located in the lower field (spin 1),
central field (spin 2), and higher field (spin 3) regions are followed in time after a 90-degree pulse using the
phase diagram. The phase diagram presents the phases of the spins relative to spin 2, which is always
shown with 0-degree phase. Note that spin 1 lags behind spin 2, while spin 3 is ahead, because it has a
higher phase velocity. This difference is due to the higher frequency of the spins in the higher magnetic
field. This process is called dephasing.

relaxation and also relaxation due to static inhomogeneity of the main the process of imaging—localizing the T1- or T2-weighted signal to a
magnetic field (B0) within the sample. These B0 field imperfections region—will be explained.
result from hardware imperfections and susceptibility-induced B0 varia-
tions due to the influence of the body itself. In the heart, both the lung
cavity and the deoxygenated blood in the right heart contribute to
SPATIAL LOCALIZATION
B0 inhomogeneity. B0 field imperfections also arise from any metallic To create a CMR image, the MR signal intensity from the sample (e.g.,
objects (sternal wires, devices, etc.) near the heart. As illustrated in patient) must be determined in three dimensions (X, Y, and Z). Thus,
Fig. 1.3B, if the B0 field is not homogeneous throughout the sample for a single image, it is necessary to collect information on X, Y, and
then the frequency of the spins in different regions will vary (see Eq. Z positions and signal amplitude. This signal amplitude can be T1,
1.1). These spins are called “off-resonance” spins. Both T2 and T2* T2, or T2* weighted, as desired. This localization is performed using
processes result in a randomization of the spin phases as they precess multiple measurements or repetition times (TRs). To localize signal
at different frequencies, and a decrease in the net transverse magnetiza- to Nx × Ny voxels (e.g., a 128 × 128 matrix), approximately Nx • Ny
tion. However, while T2 relaxation is irreversible, T2* relaxation can signals (and Ny TRs) are needed. This is actually a limitation of CMR,
be partly reversed since some of it is due to static off-resonance: T2* resulting in a relatively slow image acquisition rate compared to many
includes the irreversible molecular spin-spin interactions (T2) and static other modalities.
off-resonance. Myocardial T2 relaxation rate is ~55 ms at 1.5 T (see The simplest imaging experiment can be divided into four stages
Table 1.1),5,6 while myocardial T2* is ~30 ms at 1.5 T.7 T2* is always as shown in the block diagram in Fig. 1.4: (1) RF excitation with slice
less than T2 for all tissues, because it includes effects of T2 relaxation, selection (localization in Z); (2) phase encoding (localization in Y);
as well as off-resonance effects. Later we will describe how to obtain (3) gradient and RF rephasing/refocusing; and (4) frequency encoding
T2 contrast instead of T2* contrast. (localization in X). Each stage is used to encode the MR data with infor-
These basic relaxation processes, T1, T2, and T2* are key in the mation on the position and the amplitude of the water proton signal.
generation of image contrast as well as determining the optimal image By convention, the Z-position information is encoded with the slice-
sequence for gathering information on cardiovascular anatomy, func- selection step, the X position is determined in the frequency-encoding/
tion, and physiology. What is important is that T1, T2, and T2* vary readout step, and the Y position is obtained with the phase-encoding
for different tissues, and in disease, thereby providing “contrast” in step. In practice the frequency-encoding (X), phase-encoding (Y), and
the CMR image (see Table 1.1). For example, the T1 and T2 values slice-selection (Z) directions can be rotated in any appropriate direction.
are prolonged with increasing water content, as with edema, which MR is tomographic and can create an image of the body along any plane.
is present in different disease states.8,9 Overall, myocardial remodel- Indeed, oblique imaging, slicing through a tissue at 45 degrees, or any
ing, or myocyte replacement with connective tissue, also changes the other angle, is possible and common. In this way CMR differs from
water relaxation properties since the nature of the macromolecules in other modalities (e.g., computed tomography [CT] or two-dimensional
contact with water are critical for these relaxation processes. Finally, [2D] echocardiography), because CMR can acquire images in any plane.
most exogenous, intravenously injected CMR contrast agents act by
shortening T110–12 and T2/T2*13,14 of the water spins. By appropriately Magnetic Gradients
modifying the imaging sequences, these changes in relaxation properties Spatial encoding in CMR is performed using magnetic field gradients
due to pathology or exogenous contrast agent can be highlighted or applied to the main magnetic field: slice-select gradients, phase-encoding
even mapped (measured on a pixel-by-pixel basis) in the heart. First, gradients, and frequency-encoding gradients. Gradient coils are special
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 5

ϕ(x, y, z, t) of the spins also varies with position (see Eqs. 1.1 and 1.3,
MRI BLOCK DIAGRAM and Fig. 1.5).
γ γ
“Z” v= B(x , y , z ) = (B0 + Gx ⋅ x + Gy ⋅ y + Gz ⋅ z ) Eq. 1.6
Time Slice select 2π 2π
The first term, (γ/2π) • Β0, is just the Larmor frequency. In CMR,
“Y” we usually describe Mxy magnetization in the “rotating frame.” Therefore
Phase encode it is as if we are rotating with the spins at the Larmor frequency, and
only differences between the spin’s frequency and the Larmor frequency
are important. In that frame, ν = γ/2π(Gx • x + Gy • y + Gz • z). Also
Refocus note that at any time, gradients on X, Y, and Z axes may be applied
gradient/RF
together or alone and the amplitudes can be slewed up and down or
set to 0.
Frequency encode The phase ϕ(x, y, z, t) of the spins also varies with position. For a
(or readout) “X” constant gradient, ϕ = γΒ • t = γ(Gx • x + Gy • y + Gz • z) • t, and is related
to the gradient strengths, the time (t) that the gradient is on, and the
FIG. 1.4  General imaging scheme for collecting a simple cardiovascular
spin’s location. Note that phase accumulation due to precession at the
magnetic resonance image (MRI). RF, Radiofrequency. Larmor frequency (γ • Β0 • t) is omitted, because the phase is described
in the rotating frame. For nonconstant gradients (e.g., ramped gradi-
ents), the phase is proportional to the area of the gradient-time curve
(gradient × time):

Z φ(x , y , z , t ) = γ [area(Gx , t ) ⋅ x + area(Gy , t ) ⋅ y + area(Gz , t ) ⋅ z ]


Eq. 1.7
B0 field with
Gx The idea of gradient “area” is very important. The gradient area is
the product of the gradient amplitude, G, and the duration, t, of the
gradient. The phase of a spin at any given time is proportional to the
total gradient area and its location. Gradients cause the frequency and
X
phase of the spins to depend on location, so that if frequency of a
certain spin can be measured, its position is determined using slice
selection, frequency encoding, and phase encoding.

Y Slice-Selective Excitation: Position in Z


During the slice-select stage, the RF excitation is performed to rotate
Spin frequency varies with position the spins into the X-Y plane. During excitation, one of the spatial coor-
dinates (z) is determined by only flipping (with the B1) magnetization
in a selected slice of the sample. The magnetization remains unchanged
FIG. 1.5  A magnetic field gradient. A gradient is applied along the X (i.e., aligned with B0) except in that slice. The slice-selection process is
axis, causing the B field to vary from less than to greater than B0 along
illustrated in Fig. 1.6. A linear magnetic field gradient, Gz, is applied to
X. This leads to a linear increase in spin frequency as a function of
position along the X direction. the sample to vary the B0 field along the slice direction (the Z axis).
The magnetic field gradient causes the spins along this axis to have
slightly different frequencies as defined by the Larmor equation:

coils within the magnet that modify the main magnetic field (B), causing ν (z ) = (Gz ⋅ z ) ⋅ (γ 2π )
its strength (but not direction) to change slightly in space.
The B1 field is used to excite the spins, but only within a range of
B (x , y , z ) = B0 + Gx ⋅ x + Gy ⋅ y + Gz ⋅ z Eq. 1.5 precessional frequencies. If only a selected band of frequencies are excited
(Δν), then a slice of spins with precessional frequencies in that range
where Gx, Gy, and Gz are called “gradient strengths,” and x, y, and z are (Δz) is placed into the transverse plane, not affecting the rest of the
the spatial coordinates with x = 0, y = 0, z = 0 at the center of the magnet sample. The “slice thickness” can be freely chosen by adjusting the Z
bore. Eq. (1.5) describes how the main magnetic field strength—which gradient. Typical slices range from thin (e.g., 2 mm) slices to thick (e.g.,
always is directed along the Z axis—varies spatially with x, y, and z. 30 cm) slabs. The slice position can also be freely chosen. Furthermore,
Fig. 1.5 demonstrates the variation of the B field in the presence of an if no Z gradient is applied during the slice selection, this is equivalent
x gradient. In the same way, gradients on the Y or Z axis vary the B to an infinite slab thickness, so that all of the spins in the body will be
field in y or z. Using gradients, the main magnetic field, with which the tipped into the transverse plane. This is called a “nonselective” (NS)
spins align, and around which they precess, varies with spatial position RF pulse and is commonly used in CMR for preparation pulses.
inside the scanner. Remember, this magnetic field points in the Z direc- How can the B1 field, which oscillates at about the Larmor frequency,
tion. The gradients do not change this direction but slightly increase be used to excite only spins precessing within a range of frequencies?
or decrease its strength spatially. These spatially varying magnetic fields To excite a bandwidth of frequencies, the Z gradient is turned on, and
are called gradients because they create a spatially varying magnetic the B1 strength is modulated in time:
field (a magnetic field gradient). Of course, since the magnetic field
varies with position, the precessional frequency ν(x, y, z) and the phase B1(t ) = B10 ⋅ sin(π ⋅ t T ) (π ⋅ t T ) Eq. 1.8
6 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Sinc(t)
Frequency selective B1
pulse
Sinc function RF excitation
Flat-top Equal areas
Ramp
B1
Dn = 1/T Z slice-select
amplitude
Slice-
FT select Slice rephaser
gradient
T Y phase-encode
Frequency
Time Phase- Phase rewinder
encode
Gz “blip” Frequency encoding
Equal areas
x X frequency-
z encode (or readout)
Dz
The FT of a frequency square wave is approximated Dephasing
gradient Echo
by a sin(t)/(t) function. Excitation of a frequency Signal
band generates a slice of excited spins.
Time
FIG. 1.6  The slice-selective B1 pulse. The shape of the slice-selective
B1 pulse in time is sin(t)/t. This is called a “sinc” function. It has the FIG. 1.7  Pulse sequence diagram. In a pulse sequence diagram a time
property that it excites a distinct band of frequencies. This is true because line is shown for the B1 pulse, the gradients on each axis, and the signal
the Fourier transform (FT) of the sinc B1 field is a square wave in fre- received. This convention is used in all pulse sequence diagrams. Here
quency space. The excitation of a band of frequencies by the B1, used the slice-selection process is shown (see top portion of figure), in which
in conjunction with the gradient Gz (similar to Fig. 1.5, but along the Z a Z gradient is used with a sinc B1 pulse. The frequency encoding is
axis), causes a slice of spins to be placed on the transverse plane. shown on the X axis. The frequency encoding shows a dephasing gradi-
ent, and then a flat top, during which time the signal is acquired, as
indicated on the last line (signal). The phase-encoding gradient “blip”
is often applied just prior to frequency encoding. Often after the fre-
This B1 shape, sin(t)/t, is called a “sinc” function and is plotted in quency encoding, the phase-encoding gradient is rephased. During each
repeated pulse sequence, only the phase-encoding strength changes.
Fig. 1.6. This B1(t) excites a well-defined band of frequencies of width,
RF, Radiofrequency.
Δν = 1/T, centered around the Larmor frequency. Here, a Fourier trans-
form (FT) is used to show how the spins interpret the sin(t)/t pulse in
terms of frequency. If a Z gradient is applied during the sinc RF excita-
tion, then the band of frequencies excited is a “slice” of spins at a certain applied, and only the predefined slice within the sample will be placed
Z location, with a slice thickness Δz. Modifications to the sinc pulse onto the transverse plane for further modification to create an image.
are possible to optimize the performance and slice definition of this
approach.15,16 Frequency Encoding: Position in X
The slice-selection process is summarized in the imaging “pulse While the Z position is now known, X and Y positions remain unknown.
sequence diagram” shown in Fig. 1.7. A pulse sequence diagram presents If a gradient Gx is applied after the RF excitation in the X direction
all of the gradients, the B1 field excitation, and also marks the timing (called the “frequency-encoding direction” in MRI), recall that the fre-
of data collection (i.e., when the receiver coil records the MR signal) quency of the spins will reflect the locations of the spins in the X
on a time line. In Fig. 1.7 (boxed region), the slice-selection gradient direction:
is first increased to a constant level by a ramp. The gradient is main-
tained while the frequency selective (i.e., slice-selective) B1 field is applied; ν (x ) = (Gx ⋅ x ) ⋅ (γ 2π )
note its sinc shape. Subsequently, the slice gradient is ramped down
and the rephasing gradient is applied to remove the phase introduced (in the rotating frame). This is illustrated in Fig. 1.8 for three spins at
by the slice-select gradient. After performing the slice-select excitation, different X locations. The spins are located along the X axis, and a
only a slice of the sample has been placed in the transverse plane and magnetic field gradient in the X is turned on. One spin is located at
all of the spins are in phase. x = 0, where the magnetic field remains B0, but the others experience
The rephasing Z gradient shown in Fig. 1.7 is performed so that all slightly higher fields. The signal received from each spin depends on
of the spins are “in phase” after the slice-selection process. During the its precessional frequency, as shown in Fig. 1.8, with each spin’s signal
90-degree pulse the spins will dephase, depending on where they are oscillating at a different frequency. The combined signal from all three
in the slice (similar to that seen in Fig. 1.3B) since a magnetic field spins is measured (while Gx is “on”) by the receiver coil as the MR
gradient is applied. Thus another magnetic field gradient of opposite signal. The location of each spin can then be determined using an FT,
magnitude must be applied to reverse the dephasing caused by the which converts time oscillating total signal into its frequency (i.e., loca-
slice-selection process (shaded regions in Fig. 1.7). The gradient area tion) components.
to rephase the spins is half the slice-select area, since the spins reach
the transverse plane exactly coincident with the peak of the RF sinc FT (signal) = ∫ signal(t ) ⋅ e−2⋅π ⋅ivt dt Eq. 1.9
pulse. By applying the gradient in the opposite direction, the spins are
forced back to the same phase as they had before application of the where ν is related to the X location, as before. Using a frequency-
slice-select gradient. This small gradient is called a slice-rephasing encoding gradient, the MR signal provides a measure of the number
gradient. of spins precessing at each frequency.
In summary, to selectively excite a slice, a linear magnetic field gra- As shown in Fig. 1.7 (bottom two rows), the gradient is applied
dient Gz is placed along the axis to be sliced. A sinc B1 field pulse is for a few milliseconds, during which the MR signal is measured. The
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 7

to its area (gradient × time, Eq. 1.7), and to the spin’s Y position. The
phase-encoding gradient is applied prior to the frequency encoding. A
+32 kHz rephaser is commonly applied after frequency encoding, to remove any
frequency
B field or

phase accumulations in Y. However, many phase-encoding gradients


64 MHz
of progressively increasing areas are required in order to localize a spin
-32 kHz X axis in the Y direction, depending on the object size (called an FOV), and
x=0 the desired spatial resolution (Δy). The number of phase encodings
(Ny) is given by the relationship FOV/Ny = Δy.
+ + = Recall that to determine the position of a spin within the sample,
a gradient is applied, and the resulting frequency reports its position.
(Signal) Frequency is simply a measure of how fast the phase is changing in
FT (signal)

time (see Eq. 1.3). Phase encoding is a method for measuring the loca-
tions of the spins, by measuring their response to different strengths
of Y gradient, and deducing their precessional frequency (and therefore
location) from these measurements. Unlike frequency encoding, which
-32 kHz ν = 64 MHz +32 kHz
provides the locations of spins in X with a single measurement, phase
Frequency encoding is performed slowly, over Ny measurements. In successive
measurements, the amplitude of the phase-encoding gradient is pro-
FIG. 1.8  Frequency encoding of position. A frequency-encoding (also
gressively increased, and the signal is measured. Each measurement is
called readout) gradient is applied on the X axis, causing the B field to
vary from less than to greater than the main magnetic field. Three spins called a repetition, and the time to acquire a single measurement is
are shown at different X locations, with different precessional frequen- called the repetition time (TR).
cies. The signal from the spins during the application of this gradient is Fig. 1.9 describes the phase-encoding technique. Consider three
acquired, and its frequency depends on location. The total signal is then spins, located at three Y positions, but at any position in X. The Y
Fourier transformed (FT) to provide a measurement of the number of gradient is “blipped” on, increased for a time T, to a maximum value
spins precessing at each frequency, thereby indicating the X location Gy, and then ramped down to 0. The area of the gradient (Gy • T) and
of each spin. the spin’s Y location determines the phase accumulation (see Eq. 1.7).
With no Gy gradient on, all spins precess at the Larmor frequency and
have the same phase. If the signal is measured from these spins, it is
the bulk signal. The application of a gradient Gy for a short time T,
gradient must remain on for a specific time during frequency encoding, results in a phase that depends on the spin’s Y location. If a small gradi-
in order to sufficiently record the oscillating signals with high fidelity. ent is used, the phase changes gradually with Y position. The sphere at
Note that a dephasing gradient is applied with an area equal to half y = 0 accumulates no phase, but the other spheres accumulate phase,
that of the readout gradient. The readout gradient is then ramped up depending on their Y location. A larger phase-encoding gradient causes
and held constant for a period of time, and then the gradient is ramped even more phase accumulation at y ≠ 0. Now two of the spins are
back down to zero. This time (when Gx is constant) is the time during completely out of phase. The signal measured during each phase-encoding
which the signal is acquired—the data acquisition. During this period blip is dependent on each spins’ phase, which in turn is determined by
the signal is continuously sampled, providing Nx time samples, where each spins’ Y location. With three phase encodings, it is not yet possible
the spatial resolution (Δx) is related to Nx, by FOV/Nx = Δx. The field- to locate the spins in Y, but after collecting the signal from multiple
of-view (FOV) is the size of the sample (the patient) in the imaging (32 to 512) phase-encoding steps, the signals can be used to determine
field. Remember that this signal is already localized in Z, and using Y locations, using the FT. It is important to realize that for each phase-
slice-selective RF excitation, and is now localized in X using frequency encoding, an entire frequency encoding must be collected. When the readout
encoding. The signal measured contains this X and Z information. signal is collected during phase encoding, each of the spins’ phases is
However, it does not yet contain Y-position information. influenced by their position in Y and X. In this way, X and Y spatial
Fig. 1.7 shows a pulse sequence called a gradient recalled echo (GRE) information are encoded together. Thus this slow phase-encoding process
or gradient echo, since the readout gradient is used to refocus the dephas- provides the last piece of information, Y axis position, needed to create
ing gradient applied before it. A rephaser is sometimes applied also, the MR image.
which is similar to a dephaser but is applied after a gradient. A very
thoughtful reader may wonder, why are the gradients always “ramped” Raw k-Space Data and the Fast Fourier Transform
up and down? The ramps reflect the reality that the magnetic field Fig. 1.7 shows all of the steps of spatial encoding. A separate echo is
cannot be changed instantaneously, but rather the change is constrained collected for each phase-encode step. The phase-encode step is most
by the scanner hardware. With current hardware, the gradient coils can commonly applied immediately after the slice-select process, frequently
be “slewed” to reach their maximum value (currently 4–8 gauss/cm) at the same time as the dephasing gradient for readout occurs (as shown
within about 100–200 ms. Slew rate is currently constrained by safety in Fig. 1.7). This is done to minimize the scan time. All of the frequency
concerns related to peripheral nerve stimulation. and phase-encoded raw data are combined to create k-space (Fig. 1.10).
The k-space of Fig. 1.10 represents the raw data collected at each phase-
Phase Encoding: Position in Y encoding step. The phase- and frequency-encoding directions are labeled.
Y spatial information is determined through a process called phase- MR imaging can be understood as acquiring k-space data. Each frequency
encoding. Just like frequency encoding, phase encoding occurs after the encoding acquires a signal for a full line of k-space (kx) at a single ky
RF pulse. The spins can be phase encoded by transiently applying a value. Each phase-encoding step moves up along the phase-encoding axis
magnetic field gradient (Gy) along the phase-encoding axis (Y axis by (ky) to acquire another frequency-encoding line. Once enough k-space
convention) as shown in Fig. 1.7 (labeled a “phase-encoding blip”). signal is acquired, this k-space signal is converted into an image using
This transient gradient or blip imparts a phase to each spin proportional a 2D FT. Ideally, the raw k-space data contains enough information,
8 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

FOV with 3 spins


No gradient 2Gy
Gy = 0
TI
Gy

Time T Time T Time

A A A
Phase
of
B B B
spins
C C C

FIG. 1.9  Phase encoding. A Y gradient is transiently applied for a time T using larger and larger gradient
strengths, Gy. Three spins, A, B, and C, located at three different Y locations, respond differently to the
phase-encoding gradient. The gradient does not affect the spin at y = 0 (spin B) because it still precesses
at the Larmor frequency. But the gradient causes spin A to precess more slowly and spin C to precess more
quickly, resulting in phase differences proportional to the gradient strength (Gy) and the time t. These phase
differences allow the locations of the spins to be determined. FOV, Field of view.

Raw data collected:


k-space Image
y
+½(1/∆y)
1/FOVy {
+FOVy

∆y {
{

{
1/FOVx ∆x
Phase encodings (ky)

FT
0

-½(1/∆y)
0

-½(1/∆x) 0 +½(1/∆x)
0 FOVx
Frequency encodings (kx)

FIG. 1.10  The frequency-encoding data collected during each phase-encoding step is called the k-space
signal. The k-space signal is collected on a grid. The k-space signal does not look like an image, but is trans-
formed into an image using the Fourier transform. The spacing in k-space (Δkx, Δky) determines the field of
view (FOVx, FOVy) of the image. The width of k-space is related to the pixel size of the image, as shown.
Collecting “more” k-space reduces the pixel size.

so that after the 2D FT an image of infinite spatial resolution results. PULSE SEQUENCES AND CONTRAST
In reality, there are limits on the amount of data that can be collected.
The Y resolution of the CMR image is directly related to the number of Spin Echo Imaging
phase-encoded echoes that are collected (Δy = FOVy/Ny), and therefore Among the very first pulse sequences used in CMR was the spin echo
infinite spatial resolution would require infinite time to collect. The X sequence and, even today, it is routinely used in its accelerated form (fast
resolution is related to the number of frequency-encoding data points spin echo). As described earlier, in tissue, T2* causes rapid magnetic
acquired (Δx = FOVx/Nx). It is easier to acquire very high resolution in relaxation of water protons, compared with T2 relaxation. Thus the decay
the frequency-encoding direction, and often in CMR spatial resolution rate of the Mxy signal after an RF pulse excitation is actually a measure
in the frequency-encoding direction is higher. In CMR, time is limited of T2*. Since T2* decay is a rapid process, it limits the time that we
due to respiratory and cardiac motions, so the tradeoff between spatial can detect the MR signal. As noted above, it’s possible to circumvent
resolution required to observe the structure of interest and scan time and reverse some of the T2* dephasing, because some of it is caused by
is critically important. Schemes to increase the efficiency of collecting fixed B0 field inhomogeneity. This reversal is accomplished using a classic
k-space data (e.g., spiral, radial) will be discussed throughout this book. and important MR method called spin echo imaging. This method uses
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 9

Dephasing Rephasing Spin echo

90°
180°
Fast spins

X X X X

Y Y Y Y
Slow spins

t=τ t=τ
t=0 t = 2τ
(before 180°) (after 180°)

Time

FIG. 1.11  Spin echo: effects of a 180-degree B1 pulse after a 90-degree B1 pulse. The spins are first excited
by a 90-degree pulse, as in Fig. 1.2. After some time in which dephasing occurs (as in Fig. 1.3B), a 180-
degree pulse is applied. The 180-degree pulse, by flipping the spins 180 degrees about the X axis, exchanges
the phase position of the slower and faster spins, so that the faster spins are now lagging the slower spins.
The faster spins begin to catch up, and after a time τ equal to the time of dephasing, the spins are all aligned
in the same direction (have the same phase). This process is called “rephasing” or “refocusing” to form a
spin echo.

two B1 pulses to generate an “echo” (a signal), as outlined in Fig. 1.11.


TR and TE in a Spin Echo Sequence
First, a 90-degree pulse is applied to create Mxy magnetization. Due
to magnetic field imperfections, the magnetization begins to dephase. TR
Next a 180-degree B1 pulse is applied to flip the magnetization by 180 90° 180°
90° 180° (NS)
Excitation
degrees around the X axis. This 180-degree flip is important, because
now any slow spins (precessing slower than the Larmor frequency) are Slice select
“ahead” of fast spins, so that the spins now drift back into phase (i.e., Phase encoding Phase
“rephase”) taking the same amount of time to rephase that they were strength changes encoding

allowed to dephase. This is analogous to two people standing back to Frequency-


encode
back in a field and then walking apart (90-degree pulse) at some fixed
rate (but different rates for each person). At some time later, they reverse Signal
(180-degree pulse) their direction and now walk toward each other at
their same “individual” pace. The time it takes them to reach each other
will be the same amount of time as they walked apart. This is the nature TE
of the echo that effectively recaptures all of the coherent transverse FIG. 1.12  The pulse sequence diagram for a spin echo pulse sequence.
magnetization that had been destroyed by the B0 field inhomogene- A 90-degree slice-selective excitation is followed by a 180-degree refo-
ity in the sample. Note that the echo has both a rephasing as well as cusing pulse, as described in Fig. 1.11. Then phase encoding on Y and
a dephasing period. The spins continue to dephase after reaching the frequency encoding on the X axis are performed. The repetition time
coherent echo due to ongoing field inhomogeneity; just as in the field (TR) and echo time (TE) are defined. The diagram shows two TRs. During
they would walk past each other and continue to “dephase.” The signal each repeated TR, everything is the same, except that the phase-encoding
at the time of the spin echo only decays by T2, not T2*. strength changes, as shown.
The spin echo sequence is shown in Fig. 1.12. Note that it is almost
identical to the GRE sequence (see Fig. 1.7), except for the 180-degree
refocusing pulse. Even in this simple sequence, several factors of its slice-selective pulses, the spins’ Z magnetization will not fully regrow
design will change the contrast of the MR image, based on T1 and T2 between pulses, and this leads to a reduction in the MR signal. This
relaxation. Since a 180-degree refocusing pulse is used in this sequence, reduction in signal is dependent on the TR and the T1 of the sample.
the total time that the spins stay in the transverse plane, before the Therefore T1 weighting can be achieved using a short TR, combined
signal is measured, determines the amount of T2 relaxation that will with a short TE, so that T2 weighting is minimized. T2 weighting is
occur (see Eq. 1.3). This time is called the time to echo or the echo time achieved with a long TE and long TR. “Proton density” weighting (i.e.,
(TE) and is measured from the center of the slice-select sinc pulse to how many spins are in each location) can be achieved with a short TE
the center of the refocused echo during the readout or data acquisition and a long TR.
(see Fig. 1.12). Generally, the longer the TE, the more T2 contrast or As an example of the importance of TE, the MR water signals from
T2 weighting is generated in the image (see Eq. 1.4). normal (T2 ~55 ms) and acutely infarcted (T2 ~70 ms)17 regions of
Another sequence timing parameter that influences the signal ampli- the myocardium are shown in Fig. 1.13A. Note that the signal of both
tude is based on T1 relaxation. The time between each slice excitation tissues decreases with increasing TE, but that the infarcted tissue signal
pulse is the critical factor in this sequence and is called the time to decays more slowly, because of its longer T2. This results in a contrast,
repetition or the TR (see Fig. 1.12). If a shorter TR is used between or increased difference, in signal between the normal and the infarcted
10 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TE and TR Effects on Myocardial


Signal Amplitude
100 100

Normal
80 80
Infarct Infarct
Relative signal

60 60

40 40
Normal

20 20
Difference Difference

0 0
0 0.02 0.04 0.06 0.08 0.10 0.12 0.14 0 1 2 3 4 5 6 7
A TE (s) B TR (s)

FIG. 1.13  Echo time (TE) and repetition time (TR) effects on myocardial signal amplitude. Effect of TE
(A) and TR (B) on cardiovascular magnetic resonance signal amplitude, for normal and infarcted myocardium.
Because the T1 and T2 of acutely infarcted myocardium differ from normal myocardium, contrast between
these tissues can be created.

tissue at prolonged TEs. Inspecting the difference between the signal specific time within the echo train. This time is called the effective TE.
of the two tissue curves (dotted line), a TE of about 50 ms could be The longer the time between the initial 90 degrees, and the acquisition
selected to optimize the contrast between these two tissue types. Thus, of central k-space (i.e., the longer the effective TE), the more T2 weight-
by simply adjusting the imaging parameters, fundamental information ing will occur. The FSE technique is very commonly used in CMR to
on the heart structure can be obtained. Conversely, pathology may be visualize anatomy and measure sizes of cardiac chambers (Fig. 1.14
obscured if the imaging parameters are not ideal. shows an FSE image). It is usually used with a black-blood preparation
The impact of TR on signal amplitude is shown in Fig. 1.13B for pulse described below. T2-weighted black-blood FSE has been advocated
a spin echo sequence. The effect of TR is illustrated for normal myo- for identifying edematous tissue associated with acute infarct,19 and for
cardium with a T1 of 950 ms and abnormal myocardium with a T1 of identifying the region at risk.20 In Chapter 2, methods for quantifying
1200 ms. Note that the shorter the T1 the more rapidly the pulses can myocardial T2 will be described, which are currently in greater use.
be applied and still maintain the MR signal. Also apparent in Fig. 1.13B
is that by varying the TR, the contrast or difference between tissues, can Gradient Echo Imaging
be altered. Thus the TR can be used to vary the image contrast based on GRE imaging, like spin echo, is a fundamental CMR method and enjoys
T1. Note that a TR of 1 s would be optimal in generating the largest dif- greater use than any other method. For imaging the beating heart, and
ference (or contrast; “dotted” line) between the normal and the infarcted other highly dynamic applications, gradient echo imaging (also known
tissue. Current CMR techniques perform T1 mapping or T1-weighted as GRE, FLASH, or TFE)21 with very short TRs is one of the most com-
imaging using inversion recovery, as described below and in Chapter 2, monly used methods in CMR. This sequence is shown (again) in Fig.
to differentiate infarction and normal myocardium based on “native” T1. 1.15, displaying multiple TRs. GRE imaging uses short TRs and lower flip
angles. Furthermore, in GRE imaging the 180-degree refocusing pulse is
Fast Spin Echo Imaging eliminated. The signal on the transverse plane will then decay with T2*
The relatively long TRs required for full T1 relaxation is the major instead of T2, but this is acceptable if the TE is very short (TE < 3 ms).
reason that the spin echo method is very rarely used: Scan time (TR • As the TR is shortened, the flip angle must be reduced to provide the
Ny) is too long. To circumvent this problem, one highly useful approach optimal SNR per unit time. The flip angle provides maximum signal
is to use multiple 180-degree refocusing pulses and to collect many at steady state (i.e., after many slice-selective pulses) and is determined
echoes during each slice-selective excitation (see Fig. 1.13). This method, by a tradeoff. A larger flip angle provides more spins on the transverse
called fast spin echo (FSE) or turbo spin echo (TSE), is an important plane (more signal), but less Z magnetization is preserved, and therefore
technique in CMR.18 A phase-encoding gradient is applied between each the following echoes have less signal. A smaller flip angle provides less
180-degree pulse, thereby providing multiple phase-encoded steps from signal on the transverse plane, but more Z magnetization is preserved
a single 90-degree pulse. For CMR, typically 16 to 64 echoes are col- for subsequent echoes. The maximum signal at steady state is achieved
lected for each slice-selective pulse, thereby reducing the time to collect using the Ernst angle for a given TR and T1:
a spin echo image by 16 to 64 times, respectively. Since large blocks of
time are required to collect all of these echoes, this method is usually α = cos−1 (e− TR T1 ) Eq. 1.10
restricted to relatively motion free phases of the cardiac cycle, such as
diastole. The inherent high signal-to-noise ratio (SNR) of these FSE This low flip angle RF pulse is also called an “alpha pulse” and is
approaches supports very high spatial resolution images of the heart. suitable for short TRs. Short TRs are needed for dynamic imaging. For
In addition, true T2 contrast can be generated by acquiring the central example, the collection of 32 phase-encode lines in k-space, each with
phase-encoding data (which largely controls the image contrast) at a a 5-ms TR, requires 160 ms (5 ms × 32 phase-encode lines). This time
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 11

Fast Spin Echo Sequence


Repeated
90° 180° 180° 180° “N ” times

Excitation

Slice
select

Phase-
encode

Frequency
encoding

Signal

TE TE TE

FIG. 1.14  Pulse sequence diagram for fast spin echo imaging. TE, Echo time.

Gradient Recalled Echo


TR

α α α α α Excitation

Slice select

Equal area Phase-


encode

Crusher Frequency-
encode

Signal

Repeated
“N ” times

TE

FIG. 1.15  Pulse sequence diagrams for gradient recalled echo imaging showing multiple repetition times
(TRs). TRs and echo times (TE) are indicated.

is sufficiently short that 32 TRs can be acquired in the quiescent diastolic GRE sequence can be used to quantify myocardial T2* (by imaging at
period. Thus data can be collected rather rapidly using a small excita- multiple TE times) which may help detect diseases with myocardial
tion pulse rather than the 90-degree pulse. The GRE imaging method iron deposition such as thalassemia23–25 with good reproducibility.
uses short TRs, low flip angles (alpha pulses), and no 180-degree refo-
cusing pulse. Note that in GRE imaging (see Fig. 1.15), to keep the Three-Dimensional Fast Gradient Echo:
phase information intact, rewinding gradients are applied after each MR Angiography
acquisition equal to and opposite to the phase-encoding gradient. Fur- The fast GRE pulse sequence is also used for contrast-enhanced MR
thermore, the readout gradient is also rewound, so that on all axes, the angiography, which is the imaging of arteries and veins during the first
spins are in phase after each TR. Usually an extra gradient is applied pass of an exogenous contrast agent26,27 or in steady state.28 During the
at the end of each TR on the X or Z axis, called a “crusher,” “killer,” or first pass of a gadolinium contrast agent, the blood T1 is reduced to
“spoiler” gradient (Fig. 1.16), which dephases any remaining transverse ~30 ms. The Ernst angle equation (see Eq. 1.9) identifies an optimal
magnetization so that it does not contribute signal during the next angle of ~30 degrees for a TR of 5 ms. Because this method is three
phase-encode step (TR). These crushers are effective, but it is recognized dimensional (3D), “phase-encoding” in Z (now called “slice encoding”)
that some residual magnetization remains during the approach to steady is performed in the slice direction also. Therefore the scan time is Ny •
state, possibly affecting image contrast.22 Although the fast GRE images Nz • TR, where Nz is the number of slice-encodings (slices). The acquisi-
have T2* dephasing, this does not usually reduce image quality, since tion of a 3D fast GRE volume provides high-quality images of vessels
very short TEs are achievable, unless a serious source of magnetic field (see Chapters 43–46). However, without a contrast agent, the SNR of
inhomogeneity (e.g., a metallic implant) is present. Furthermore, the fast GRE imaging can be low, because the time between TRs, during
12 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TR
+60° –60° +60° –60° +60°
Excitation

Slice select

Phase-
encode

Frequency-
Net area encode
=0

TE Signal

Repeated
“N ” times

FIG. 1.16  Balanced steady-state free precession (SSFP) imaging. The sequence is identical to gradient
recalled echo, except that the flip angle for SSFP is high and its sign is alternated in each repetition time
(TR). Also, the gradients in the SSFP sequence are fully rephased (see shaded areas), so that the net gradi-
ent area (summed over the TR) is zero, for each gradient axis. TE, Echo time.

which the longitudinal magnetization can regrow, is short. This limita-


20°
tion is partly overcome using balanced SSFP. RF Excitation

Balanced Steady-State Free Precession Z Slice-select


Directly related to the GRE sequence is the balanced SSFP method (see
Fig. 1.16). Today, almost all CMR imaging of ventricular function at Y Phase-
1.5 T employs this method, and it also is used for anatomical localiza- encode
tion, valve visualization, and coronary artery imaging.29 This very old Readouts
technique30 was later revived31,32 because better scanner hardware allows X Frequency-
for short TRs (e.g., 2–4 ms on 1.5 T) required for balanced SSFP. Bal- encode
anced SSFP uses a pulse sequence identical to GRE (see Fig. 1.16),
except for two distinctions. First, after each TR, the spins are rephased Signal
on each gradient axis (X, Y, and Z) to zero phase. No crushers, killers,
or spoilers are used. This keeps the spins completely in phase (except FIG. 1.17  Echo planar imaging (also called multiecho imaging), in which
for dephasing due to magnetic field inhomogeneities) so that the trans- a portion of the frequency encodings are collected after each B1 pulse.
verse magnetization can be reused in the next TR. For this reason the Here eight frequency encodings are collected after a single B1 excitation
sequence is called “balanced.” Second, a large flip angle (e.g., 60 degrees) pulse (here with a 20-degree flip angle). All of the CMR data can be
is used, flipping spins around the positive and negative Y axis, in alter- acquired after a single excitation, or only a portion. The phase-encoding
gradient is blipped with a constant area, which leads to increasing phase
nate TRs. This flip angle alternation scheme reuses the remaining trans-
encoding, and readouts are acquired using both positive and negative
verse magnetization, and mixes it with longitudinal magnetization, for lobes of the frequency-encoding gradient.
increased signal in each TR.
Balanced SSFP provides high SNR of blood and a unique T2/T1
weighting.33 As stated above, balanced SSFP is highly sensitive to off- phase-encodings values are acquired. To reduce time between echoes,
resonance, requires short TRs and large flip angles, and is challenging the readout gradient is rapidly reversed with every phase-encode step,
at 3 T, where off-resonance creates artifacts,34 and RF heating concerns so that MR signals are collected during both the positive and negative
(specific absorption rate) limits the flip angle. lobes of the readout gradient (arrows). This approach is challenging in
the heart, where the T2* can be short36 compared to the brain, but has
Echo Planar Imaging, Spiral and Radial found applications in cardiac perfusion37 and the cardiac diffusion
Analogous to reducing scan time, by replacing a spin echo acquisition weighted imaging.38 Because of the acquisition of many phase-encode
by a fast spin echo acquisition, the GRE sequence can be accelerated steps after one RF excitation, the EPI method is fast, but results in poor
by acquiring a train of gradient recalled echoes after a single RF pulse. SNR. EPI (along with spiral and radial described below) has k-space
This approach is called multiecho or echo planar imaging (EPI) and “trajectory” errors that need correction,39–42 and image distortions due
was among the first CMR imaging methods described.35 For anatomy to off-resonance. However, these limitations can be overcome by only
with long T2* tissues (e.g., the head), EPI can result in a complete collecting a few (e.g., 3–9) phase-encode steps per slice-select RF excita-
image collection with a single slice-select pulse and a train of gradient tion (instead all phase-encode steps) and repeating this process until
echoes. EPI is the basis for the functional MRI (fMRI) technique widely all of the data have been collected.
used in brain mapping. Fig. 1.17 shows an EPI sequence, in which after Spiral imaging is similar to EPI, obtaining more image data after a
a single RF excitation, frequency-encoding information at multiple single RF pulse, compared to conventional imaging, by continuously
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 13

GRE or SSFP or EPI

Repeat “N ”
A times to obtain
EKG
single image
Prep Imaging

T2prep
1
B C
Spoiler 180° 180°
0.5
90°

Mz
90° or 90° (y)
-90° (y) (tip up)
180° TI 0

T1 = 300 ms
Saturation or -0.5
T1 = 400 ms
inversion recovery -1
180° TE
0 200 400 600 800
TI
1000

D E 90° (selective) 180° F


Diffusion
Spoiler
TI
180°
90°
TI
G Diff G Diff
Fat-only excitation
Black-blood

FIG. 1.18  Preparation pulses. (A) A preparation pulse precedes the acquisition of a segment of data (e.g.,
multiple repetition times [TR] with gradient recalled echo [GRE] or steady-state free precession [SSFP]),
usually as part of an electrocardiogram (ECG)-gated acquisition. All preparations affect the longitudinal mag-
netization (Mz). (B) A nonselective 180-degree or 90-degree followed by an inversion time (TI) pulse generates
T1 contrast, because Mz depends on the T1 and the TI time chosen. (C) T2prep uses the method of fast
spin echo to refocus magnetization, which is then stored on the Mz axis. This results in Z magnetization,
which depends on T2 of the imaged tissue. (D) Fat saturation is performed by exciting fat with a 90-degree
pulse and then crushing the resulting fat Mxy. Using a short TI, the Mz of fat is 0 after the saturation pulse,
and the fat signal is suppressed. (E) Black-blood preparation inverts all blood, and images at a TI when blood
Mz magnetization is close to 0. The myocardial signal is unaffected, because a slice of interest is reinverted
immediately after the 180 degrees. (F) Diffusion-encoding preparation. EPI, Echo planar imaging; TE, echo
time.

sampling k-space in a spiral pattern,43–45 and has found applications in cycles, another preparation pulse is applied and another segment of
flow imaging,46 coronary imaging, and 3D late gadolinium enhance- data is acquired. Fig. 1.18B–F shows a few important preparation pulses.
ment,47 where SNR is important. Radial imaging is another trajectory Fig. 1.18B shows nonselective RF pulses, both a 90-degree “saturation
that acquires k-space data as radial spokes, analogous to CT. It has recovery” pulse and a 180-degree “inversion recovery” pulse. These pulses
been applied to the heart, especially using undersampling of k-space generate T1 weighting, determined by the inversion time (TI) choice.
for fast imaging.48–51 Radial imaging was the first imaging method,1 A spoiler gradient destroys any Mxy magnetization, created by the RF
and its clinical utility has been facilitated by trajectory improvements pulse. These are commonly used for perfusion and late gadolinium
and recent developments in reconstruction of undersampled MR enhancement.57 They are nonselective, so that all spins experience these
data52–55 using sparsity constraints. Spiral and radial also need trajec- pulses. Adiabatic RF pulses are often used, which are less susceptible
tory correction. MR fingerprinting is a way of randomly sampling to B0 inhomogeneity.58 Eq. (1.2) describes the T1 weighting achieved
k-space with variable T1/T2 contrast.56 In combination with advanced for a 90-degree saturation pulse.
reconstruction methods, T1, T2, and proton density images can be Fig. 1.18C shows a T2prep pulse.59,60 This is also nonselective, affect-
generated. ing all of the spins in the magnetic field. A 90-degree excitation is
followed by multiple 180-degree refocusing pulses. The signal decays
Preparation Pulses by T2, just like fast spin echo imaging. However, the signal is tipped
In CMR imaging, preparation pulses are often used to generate contrast, back to the Z axis with a 90-degree pulse, generating Mz magnetization
by generating Mz magnetization, which is T1 or T2 weighted. Fig. 1.18A that is weighted by the T2 of the tissue, with higher Mz for long T2
shows how preparation pulses are used in CMR. First, a preparation tissues. Thus this preparation pulse is called T2prep. Very importantly,
pulse is applied, and then a segment (16–64 lines or more of k-space) Fig. 1.18D shows how fat saturation can be achieved using a 90-degree
of the MR data is acquired, at a certain time within the cardiac cycle. excitation of fat only, which is possible because the Larmor frequency
Then, either in the next cardiac cycle or after skipping one or more of fat is different from that of water (~210 Hz difference at 1.5 T). After
14 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

the saturation of fat, a spoiler kills the fat Mxy. Imaging then begins, refocusing pulse and an identical gradient that should rephrase any
with the fat signal suppressed. Fig. 1.18E shows a double inversion phase accumulation not due to spin motion. Cardiac diffusion imaging
recovery (DIR) black-blood preparation.61 This preparation consists of was proposed early on62,63 but is only now gaining acceptance38 due to
two inversion (180-degree) pulses, which immediately follow each other. lessening concerns about the influences of cardiac and respiratory motion.
The first inversion is a nonselective inversion pulse, which inverts all Many other preparation pulses, such as magnetization transfer (MT),64
magnetization. The second inversion is a slice-selective inversion pulse, are available for generating contrast.
which reinverts a slice of interest, which is centered on the imaging
slice but is slightly larger (by a factor of 1.5–2). This preparation is
timed so that at the time of acquisition, all blood magnetization has
NEW HARDWARE ADVANCES
regrown from −Mz to 0 (i.e., is black). Any blood that was reinverted, For CMR, a B0 of 1.5 T is most commonly used; however, cardiac imaging
because it was within the slice of interest, moves out of the imaging at 3 T65 provides equivalent or improved quality for coronary artery
slice during systole. Any blood that has flowed into the slice will con- imaging,66,67 late gadolinium enhancement, and perfusion,68 and func-
tribute no signal because it is nulled (see Fig. 1.10 for a black-blood tion,34 and there is progress in CMR at 7 T.69 The number of receiver
image). Fig. 1.18F shows a “diffusion weighting” preparation. In this coil channels has increased from a single surface coil or body coil to
technique, the microstructure of the myocardium can be studied. The 16–32 channels routinely on new systems, up to 128 channels.70 Higher
diffusion of water in the myocardium depends on the myocyte’s ori- gradient strengths of 80 mT/m are now available, which might hold
entation and therefore fiber structure. The diffusion is measured using promise for cardiac diffusion imaging.
preparation pulses: a 90-degree RF saturation pulse, to excite the spins,
followed by the application of a maximal strength and rather long
gradient (~10 ms, on any or multiple axes) to sensitize the signal to
REFERENCES
microscopic motions (diffusion). This is followed by a 180-degree A full reference list is available online at ExpertConsult.com
CHAPTER 1  Basic Principles of Cardiovascular Magnetic Resonance 14.e1

REFERENCES 26. Prince MR, et al. Dynamic gadolinium-enhanced three-dimensional


abdominal MR arteriography. J Magn Reson Imaging. 1993;3(6):877–881.
1. Lauterbur P. Image formation by induced local interactions: examples 27. Zhang H, Maki JH, Prince MR. 3D contrast-enhanced MR angiography. J
employing nuclear magnetic resonance. Nature. 1973;190–191. Magn Reson Imaging. 2007;25(1):13–25.
2. Schelbert EB, Messroghli DR. State of the art: clinical applications of 28. Grist TM, et al. Steady-state and dynamic MR angiography with MS-325:
cardiac T1 mapping. Radiology. 2016;278(3):658–676. initial experience in humans. Radiology. 1998;207(2):539–544.
3. Karamitsos TD, et al. Noncontrast T1 mapping for the diagnosis of 29. Weber OM, Martin AJ, Higgins CB. Whole-heart steady-state free
cardiac amyloidosis. JACC Cardiovasc Imaging. 2013;6(4):488–497. precession coronary artery magnetic resonance angiography. Magn Reson
4. Alam MH, et al. T1 at 1.5T and 3T compared with conventional T2* at Med. 2003;50(6):1223–1228.
1.5T for cardiac siderosis. J Cardiovasc Magn Reson. 2015;17:102. 30. Oppelt A, et al. FISP- a new fast MRI sequence. Electromedica.
5. Botnar RM, et al. Improved coronary artery definition with T2-weighted, 1986;54:15–18.
free-breathing, three-dimensional coronary MRA. Circulation. 1999; 31. Duerk JL, et al. Remember true FISP? A high SNR, near 1-second
99(24):3139–3148. imaging method for T2-like contrast in interventional MRI at .2 T. J
6. Foltz WD, et al. T2 fluctuations in ischemic and post-ischemic viable Magn Reson Imaging. 1998;8(1):203–208.
porcine myocardium in vivo. J Cardiovasc Magn Reson. 2006;8(3):469–474. 32. Heid O. True FISP cardiac fluoroscopy. In: Fifth Annual Proceedings of
7. Reeder SB, et al. In vivo measurement of T*2 and field inhomogeneity International Society of Magnetic Resonance in Medicine; 1997;
maps in the human heart at 1.5 T. Magn Reson Med. 1998;39(6):988–998. Vancouver, BC, Canada.
8. Croisille P, Revel D, Saeed M. Contrast agents and cardiac MR imaging of 33. Scheffler K, Lehnhardt S. Principles and applications of balanced SSFP
myocardial ischemia: from bench to bedside. Eur Radiol. 2006;16(9): techniques. Eur Radiol. 2003;13(11):2409–2418.
1951–1963. 34. Schar M, et al. Cardiac SSFP imaging at 3 Tesla. Magn Reson Med.
9. Scholz TD, Martins JB, Skorton DJ. NMR relaxation times in acute 2004;51(4):799–806.
myocardial infarction: relative influence of changes in tissue water and fat 35. Mansfield P, Pykett I. Biological and medical imaging by NMR. J Magn
content. Magn Reson Med. 1992;23(1):89–95. Reson. 1978;29:355–373.
10. Goldfarb JW, Mathew ST, Reichek N. Quantitative breath-hold 36. Reeder SB, et al. In vivo measurement of T2* and field inhomogeneity
monitoring of myocardial gadolinium enhancement using inversion maps in the human heart at 1.5 T. Magn Reson Med. 1998;39(6):988–998.
recovery TrueFISP. Magn Reson Med. 2005;53(2):367–371. 37. Epstein FH, Wolff SD, Arai AE. Segmented k-space fast cardiac imaging
11. Klein C, et al. The influence of myocardial blood flow and volume of using an echo-train readout. Magn Reson Med. 1999;41(3):609–613.
distribution on late Gd-DTPA kinetics in ischemic heart failure. J Magn 38. Nielles-Vallespin S, et al. In vivo diffusion tensor MRI of the human
Reson Imaging. 2004;20(4):588–593. heart: reproducibility of breath-hold and navigator-based approaches.
12. Parker DL, et al. Optimized visualization of vessels in contrast enhanced Magn Reson Med. 2013;70(2):454–465.
intracranial MR angiography. Magn Reson Med. 1998;40(6):873–882. 39. Peters DC, Derbyshire JA, McVeigh ER. Centering the projection
13. Mathur-De Vre R, Lemort M. Invited review: biophysical properties and reconstruction trajectory: reducing gradient delay errors. Magn Reson
clinical applications of magnetic resonance imaging contrast agents. Br J Med. 2003;50(1):1–6.
Radiol. 1995;68(807):225–247. 40. Reeder SB, et al. Referenceless interleaved echo-planar imaging. Magn
14. Weissleder R, et al. Ultrasmall superparamagnetic iron oxide: Reson Med. 1999;41(1):87–94.
characterization of a new class of contrast agents for MR imaging. 41. Duyn JH, et al. Simple correction method for k-space trajectory
Radiology. 1990;175(2):489–493. deviations in MRI. J Magn Reson. 1998;132(1):150–153.
15. Conolly S, et al. A reduced power selective adiabatic spin-echo pulse 42. Campbell-Washburn AE, et al. Real-time distortion correction of spiral
sequence. Magn Reson Med. 1991;18(1):28–38. and echo planar images using the gradient system impulse response
16. Meyer CH, et al. Simultaneous spatial and spectral selective excitation. function. Magn Reson Med. 2015.
Magn Reson Med. 1990;15(2):287–304. 43. Nayak KS, et al. Real-time interactive coronary MRA. Magn Reson Med.
17. Ugander M, et al. Myocardial edema as detected by pre-contrast T1 and 2001;46(3):430–435.
T2 CMR delineates area at risk associated with acute myocardial 44. Yang PC, et al. Spiral magnetic resonance coronary angiography with
infarction. JACC Cardiovasc Imaging. 2012;5(6):596–603. rapid real-time localization. J Am Coll Cardiol. 2003;41(7):1134–1141.
18. Hennig J, Nauerth A, Friedburg H. RARE imaging: a fast imaging method 45. Hardy CJ, et al. Coronary MR angiography: respiratory motion
for clinical MR. Magn Reson Med. 1986;3(6):823–833. correction with BACSPIN. J Magn Reson Imaging. 2003;17(2):170–176.
19. Abdel-Aty H, et al. Delayed enhancement and T2-weighted cardiovascular 46. Kowalik GT, et al. Rapid breath-hold assessment of myocardial velocities
magnetic resonance imaging differentiate acute from chronic myocardial using spiral UNFOLD-ed SENSE tissue phase mapping. J Magn Reson
infarction. Circulation. 2004;109(20):2411–2416. Imaging. 2016;44:1003.
20. Aletras AH, et al. Retrospective determination of the area at risk for 47. Knowles BR, et al. Three-dimensional late gadolinium-enhanced MR
reperfused acute myocardial infarction with T2-weighted cardiac imaging of the left atrium: a comparison of spiral versus Cartesian
magnetic resonance imaging: histopathological and displacement k-space trajectories. J Magn Reson Imaging. 2014;39(1):211–216.
encoding with stimulated echoes (DENSE) functional validations. 48. Larson AC, et al. Self-gated cardiac cine MRI. Magn Reson Med. 2004;
Circulation. 2006;113(15):1865–1870. 51(1):93–102.
21. Haase A, et al. Dynamic digital subtraction imaging using fast low-angle 49. Peters DC, et al. Undersampled projection reconstruction applied to MR
shot MR movie sequence. Radiology. 1986;160(2):537–541. angiography. Magn Reson Med. 2000;43(1):91–101.
22. Roeloffs V, Voit D, Frahm J. Spoiling without additional gradients: radial 50. Bi X, et al. Contrast-enhanced 4D radial coronary artery imaging at 3.0 T
FLASH MRI with randomized radiofrequency phases. Magn Reson Med. within a single breath-hold. Magn Reson Med. 2005;54(2):470–475.
2015. 51. Barger AV, et al. Single breath-hold 3D contrast-enhanced method
23. Pennell DJ, et al. Cardiovascular function and treatment in beta- for assessment of cardiac function. Magn Reson Med. 2000;44(6):
thalassemia major: a consensus statement from the American Heart 821–824.
Association. Circulation. 2013;128(3):281–308. 52. Mistretta CA. Undersampled radial MR acquisition and highly
24. Anderson LJ, et al. Cardiovascular T2-star (T2*) magnetic resonance for constrained back projection (HYPR) reconstruction: potential medical
the early diagnosis of myocardial iron overload. Eur Heart J. 2001;22(23): imaging applications in the post-Nyquist era. J Magn Reson Imaging.
2171–2179. 2009;29(3):501–516.
25. Westwood M, et al. A single breath-hold multiecho T2* cardiovascular 53. Lustig M, Donoho D, Pauly JM. Sparse MRI: the application of
magnetic resonance technique for diagnosis of myocardial iron overload. compressed sensing for rapid MR imaging. Magn Reson Med.
J Magn Reson Imaging. 2003;18(1):33–39. 2007;58(6):1182–1195.
14.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

54. Block KT, Uecker M, Frahm J. Undersampled radial MRI with multiple 67. Nezafat R, et al. B1-insensitive T2 preparation for improved coronary
coils. Iterative image reconstruction using a total variation constraint. magnetic resonance angiography at 3 T. Magn Reson Med.
Magn Reson Med. 2007;57(6):1086–1098. 2006;55(4):858–864.
55. Wundrak S, et al. Golden ratio sparse MRI using tiny golden angles. 68. Gebker R, et al. Diagnostic performance of myocardial perfusion MR at
Magn Reson Med. 2016;75(6):2372–2378. 3 T in patients with coronary artery disease. Radiology.
56. Ma D, et al. Magnetic resonance fingerprinting. Nature. 2013;495(7440): 2008;247(1):57–63.
187–192. 69. Niendorf T, et al. W(h)ither human cardiac and body magnetic resonance
57. Simonetti OP, et al. An improved MR imaging technique for the at ultrahigh fields? technical advances, practical considerations,
visualization of myocardial infarction. Radiology. 2001;218(1):215–223. applications, and clinical opportunities. NMR Biomed.
58. Rashid S, et al. Improved late gadolinium enhancement MR imaging for 2016;29(9):1173–1197.
patients with implanted cardiac devices. Radiology. 2014;270(1):269–274. 70. Schmitt M, et al. A 128-channel receive-only cardiac coil for highly
59. Brittain JH, et al. Three-dimensional flow-independent peripheral accelerated cardiac MRI at 3 Tesla. Magn Reson Med.
angiography. Magn Reson Med. 1997;38(3):343–354. 2008;59(6):1431–1439.
60. Giri S, et al. T2 quantification for improved detection of myocardial 71. Dabir D, et al. Reference values for healthy human myocardium using a
edema. J Cardiovasc Magn Reson. 2009;11:56. T1 mapping methodology: results from the International T1 Multicenter
61. Simonetti OP, et al. “Black blood” T2-weighted inversion-recovery MR cardiovascular magnetic resonance study. J Cardiovasc Magn Reson.
imaging of the heart. Radiology. 1996;199(1):49–57. 2014;16:69.
62. Tseng WY, et al. Imaging myocardial fiber disarray and intramural strain 72. Bonner F, et al. Myocardial T2 mapping reveals age- and sex-related
hypokinesis in hypertrophic cardiomyopathy with MRI. J Magn Reson differences in volunteers. J Cardiovasc Magn Reson. 2015;17(1):9.
Imaging. 2006;23(1):1–8. 73. Rauhalammi SM, et al. Native myocardial longitudinal (T) relaxation
63. Reese TG, et al. Imaging myocardial fiber architecture in vivo with time: regional, age, and sex associations in the healthy adult heart. J Magn
magnetic resonance. Magn Reson Med. 1995;34(6):786–791. Reson Imaging. 2016;44:541.
64. Wolff SD, Balaban RS. Magnetization transfer imaging: practical aspects 74. Ding H, et al. Three-dimensional whole-heart T2 mapping at 3T. Magn
and clinical applications. Radiology. 1994;192(3):593–599. Reson Med. 2015;74(3):803–816.
65. Gharib AM, Elagha A, Pettigrew RI. Cardiac magnetic resonance at high 75. Zhang X, et al. In vivo blood T(1) measurements at 1.5 T, 3 T, and 7 T.
field: promises and problems. Curr Probl Diagn Radiol. 2008;37(2):49–56. Magn Reson Med. 2013;70(4):1082–1086.
66. Stuber M, et al. Preliminary report on in vivo coronary MRA at 3 Tesla in 76. Stanisz GJ, et al. T1, T2 relaxation and magnetization transfer in tissue at
humans. Magn Reson Med. 2002;48(3):425–429. 3T. Magn Reson Med. 2005;54(3):507–512.
B R AU N WA L D ’ S H E A R T D I S E A S E
F A M I LY O F B O O K S

BRAUNWALD’S HEART DISEASE COMPANIONS

BHATT BALLANTYNE KORMOS AND MILLER


Cardiovascular Intervention Clinical Lipidology Mechanical Circulatory Support

ANTMAN AND SABATINE MCGUIRE AND MARX MORROW


Cardiovascular Therapeutics Diabetes in Cardiovascular Disease Myocardial Infarction

DE LEMOS AND OMLAND MANN AND FELKER BLUMENTHAL, FOODY, AND WONG
Chronic Coronary Artery Disease Heart Failure Preventive Cardiology

ISSA, MILLER, AND ZIPES BAKRIS AND SORRENTINO OTTO AND BONOW
Clinical Arrhythmology and Electrophysiology Hypertension Valvular Heart Disease

xxi
xxii BRAUNWALD’S HEART DISEASE FAMILY OF BOOKS

CREAGER, BECKMAN, AND LOSCALZO SOLOMON, WU, AND GILLAM


Vascular Medicine Essential Echocardiography

BRAUNWALD’S HEART DISEASE REVIEW AND ASSESSMENT

LILLY
Braunwald’s Heart Disease Review and Assessment

BRAUNWALD’S HEART DISEASE IMAGING COMPANIONS

TAYLOR KRAMER AND HUNDLEY ISKANDRIAN AND GARCIA


Atlas of Cardiovascular Computer Atlas of Cardiovascular Magnetic Atlas of Nuclear Cardiology
Tomography Resonance Imaging
2 
Techniques for T1, T2, and Extracellular
Volume Mapping
Peter Kellman and Michael Hansen

Many disease processes alter the local molecular environment of the parametric mapping is a powerful tool in the assessment of diffuse
myocardium, and consequently the longitudinal (T1) and trans- myocardial disease.
verse (T2) relaxation times can change. While such changes may be
observed directly as changes in image contrast,1–6 the tissue processes
may be global and diffuse, hampering reliable detection of the disease.
T1 AND EXTRACELLULAR VOLUME MAPPING
Quantitative methods for characterizing myocardial tissue based on Changes in both native T1 and T1 following the administration of
parametric mapping of T1 and T2 have been proposed as an objec- gadolinium (Gd) contrast agents are considered important biomarkers,
tive way of detecting and quantifying both focal and global changes and multiple methods have been suggested for quantifying myocardial
in tissue. In combination with extracellular contrast agent injection, T1 in vivo.21 In general, methods for measuring myocardial T1 consist
T1 mapping can also provide an estimate of the extracellular volume of three components: (1) a perturbation of the longitudinal magnetiza-
(ECV) fraction. tion (i.e., an inversion or saturation), (2) an experiment to sample the
Native T1 mapping, as well as ECV mapping, is currently being relaxation curve as the longitudinal magnetization returns to its original
explored as a diagnostic tool for a wide range of cardiomyopathies, and level, and (3) a model used to fit the sampled curve and extract the
native T1 changes are detectable in both acute and chronic myocardial myocardial T1. This chapter focuses on the technical aspects of key
infarction (MI)7,8 and may be used to characterize the edematous area methods and imaging protocols and describes their limitations and the
at risk.4,9,10 Elevated native T1 has also been reported in a number of factors that influence their accuracy, precision, and overall reproduc-
diseases with cardiac involvement (e.g., myocarditis,11 amyloidosis,12 ibility. The accuracy and precision of these measurements affect the
lupus,13 and system capillary leakage syndrome14), and decreases in detection and quantification of abnormal myocardial tissue.
native T1 have been associated with Anderson-Fabry disease15 and high Late gadolinium enhancement (LGE) is currently the primary tool
iron content.16,17 Native T2 mapping is used to detect edema in acute for tissue characterization in CMR because it provides excellent depic-
MI and myocarditis18–20 and to identify the area at risk4 in acute MI. tion of MI and focal scar and has become an accepted standard for
Application of tissue characterization using parametric mapping for assessing myocardial viability.22 LGE is also useful for detecting and
detection of disease is discussed elsewhere in this book. characterizing fibrosis that is “patchy” in appearance, for example, as
This chapter describes state-of-the-art methods for measuring T1, T2, seen in nonischemic cardiomyopathies such as hypertrophic cardiomy-
and ECV in the myocardium. Because the purpose of such techniques opathy (HCM).23 Diffuse myocardial fibrosis is, however, more difficult
is a quantitative (objective) evaluation of biomarkers and disease, the to distinguish using LGE, since the myocardial signal intensity may be
discussion of the available techniques is framed in terms of accuracy, nearly isointense and may be globally “nulled,” thus appearing to be
precision, and general reliability. In parametric mapping it is important normal tissue.24 Alternatively, quantitative measurement of myocardial
to understand errors in quantification and other artifact mechanisms, T1 following the administration of an extracellular Gd contrast agent
which may be less familiar than conventional cardiovascular magnetic has been shown to be sensitive to increased ECV associated with diffuse
resonance (CMR) artifact mechanisms. Method imperfections may myocardial fibrosis. However, a single postcontrast T1 measurement
manifest themselves as subtle changes in measured tissue parameters, has limitations because of a variety of confounding factors such as
and artifactual changes may well be correlated with specific patient Gd clearance rate, time of measurement, injected dose, body com-
groups—that is, they are potential confounders. In particular, the esti- position, and hematocrit.25,26 These factors cause a significant varia-
mated parameter values such as T1 may be affected by other variables tion in postcontrast T1, making it difficult to distinguish diseased and
such as myocardial wall thickness, T2, protocol settings, or scanner normal tissue based on absolute T1 values alone. Precontrast T1 varies
adjustments. Although this is not new to magnetic resonance imaging with water content and may be elevated in cases of diffuse myocardial
(MRI), where T1-weighted contrast may have some T2 contrast, these fibrosis. Precontrast T1 also varies significantly with field strength.27
confounding effects may influence the interpretation of parametric Direct measurement of ECV was initially developed for quantifying
maps. Normal values for parameters may depend on the field strength the myocardial extracellular fractional distribution volume28 and has
and the specific measurement technique or imaging protocol. There- been proposed as a means for detection and quantification of diffuse
fore well-controlled and optimized protocols are key to reproducibility, myocardial fibrosis.24,29–34 This approach is based on the change in T1
which is particularly important in applications aimed at the detection following administration of an extracellular contrast agent and cir-
of subtle fibrosis and preclinical disease, and normal values need to be cumvents the limitation of a single postcontrast T1 measurement in
established for specific protocols. Limited spatial resolution will lead detecting a global change in T1. Myocardial ECV is measured as the
to errors caused by partial volume effects, particularly between myo- percent of tissue composed of extracellular space, which is a physiologi-
cardium and adjacent blood pool or fat tissue. Despite limitations, cally intuitive unit of measurement and is independent of field strength.

15
16 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

ECV has been shown to correlate with collagen volume fraction in


TABLE 2.1  Widely Used Inversion and
some diseases.29,31
Saturation Recovery Methods for T1
Brief History of Methods for T1 Mapping in the Heart Mapping in the Heart
Methods for measuring myocardial T1 were initially based on region of Inversion recovery (IR) Multiple breath-hold FLASH35
interest (ROI) analysis rather than pixelwise parametric maps. Inversion MOLLI37
recovery (IR) images at different inversion times were acquired with ShMOLLI38
multiple breath-holds35 or IR cine protocols were used as a means of Modified MOLLI protocols32,33,44,45
acquiring data in a single breath-hold.36 These early methods were ROI- Saturation recovery (SR) SAP-T146
based schemes and were not suitable for pixelwise mapping. Pixelwise SASHA47,51,52
T1 mapping was introduced with the modified Look-Locker inversion SASHA VFA53
recovery (MOLLI) imaging strategy,37 which propelled the use of T1 SMART154
mapping in CMR and inspired many new methods. MOLLI is widely used Combined IR/SR SAPPHIRE48
today with some protocol optimization and other adaptations. A short-
FLASH, Fast low angle shot; MOLLI, modified Look-Locker inversion;
ened breath-hold adaptation with conditional curve fitting (ShMOLLI)38
SAP, short acquisition period; SASHA, saturation recovery single-shot
was proposed as a means of mitigating heart rate dependence as well
acquisition; SAPPHIRE, saturation pulse prepared heart rate
as shortening the breath-hold. Further protocol optimization has been independent inversion-recovery; ShMOLLI, shortened breath-hold
aimed at shortening the breath-hold and optimizing precision.32,39 Motion modified Look-Locker inversion.
correction was developed to mitigate respiratory motion for subjects
with poor breath-holding,40 and phase-sensitive IR reconstruction with
motion correction further improved image quality.41 A number of pub- the same initial condition. The T1-map precision is related to the number
lications analyzed the accuracy of T1 measurements,37,38,42–45 leading to and position of samples along the IR curve, and accuracy of the signal
a better understanding of the influence of various protocol parameters model is also affected by the sampling strategy due to the influence of
on T1 measurement errors. Saturation recovery (SR) methods devel- the readout on the apparent recovery.
oped initially for T1 measurements during first-pass contrast-enhanced The MOLLI method uses an SSFP readout. The readout drives
perfusion (short acquisition period-T1 [SAP-T1])46 have been recently the IR to recover more quickly and reaches a steady state that is less
adapted for T1 mapping using SR single-shot acquisition (SASHA),47 than the equilibrium magnetization (M0). The effect of the readout
with steady-state free precession (SSFP) readout as a means of mitigat- (Fig. 2.2) is an apparent recovery time referred to as T1*, which is
ing the T1 underestimation in MOLLI and reducing the influence of less than the actual longitudinal recovery time, T1, which is the
confounding factors such as T2, magnetization transfer (MT), heart desired tissue parameter. As a result of the influence of the readout,
rate, and off-resonance. Even more recently, hybrid schemes have been the IR curve follows a three-parameter exponential signal model,
proposed that incorporate both inversion and SR methods (saturation S(t) = A − B exp(−t/T1*), where t represents the inversion time and T1* is
pulse prepared heart rate independent inversion-recovery [SAPPHIRE]).48 the apparent T1. The measured values may be fit to the three-parameter
ECV measurements were initially introduced using ROI-based mea- model to estimate A, B, and T1*, which may be used to approximate
surement,28–30 and pixelwise ECV mapping was later introduced.33,39 T1 ≈ T1* (B/A − 1). The derivation for the so-called Look-Locker
Improvements to T1 and ECV mapping are continuously introduced, correction factor (B/A − 1) is based on a continuous readout using
such as navigated methods for higher-resolution three-dimensional fast low angle shot (FLASH).55 Despite the fact that the MOLLI uses
(3D)49 or two-dimensional (2D) multislice.50 a gated SSFP readout, the signal model behaves as a three-parameter
This chapter focuses on the basic concepts behind the widely used model where the Look-Locker correction is reasonably effective at low
MOLLI and SASHA acquisition strategies for T1 mapping, followed readout excitation flip angles (FAs).
by a review of factors influencing accuracy and precision. Discussion The analytic relationship between T1* and T1 for SSFP has been
includes a description of other limitations and a summary of the pros derived for continuous SSFP under somewhat idealized conditions such
and cons of various protocols. as ideal slice profile.56 Useful analytic derivations for gated SSFP with
realistic slice profiles have not been developed due to complexity. Bloch
T1-Mapping Methods simulations may be used to calculate the error inherent in this approxi-
The currently used protocols for T1 mapping in the heart (Table 2.1) mation43 and to gain insight into the sensitivity of various protocol
are based on IR or SR. Images are acquired at multiple time points on parameters and design variables. The influence of various parameters
the recovery curve, and pixelwise curve fitting is performed to estimate such as T2, heart rate, off-resonance, and actual FA are referred to as
the relaxation time parameter to produce a pixel map of T1. Images confounders and are discussed later.
are generally acquired at the same cardiac phase and respiratory posi- A number of modifications (Table 2.2) to the original proposed
tion to eliminate tissue motion. Although initial implementation involved MOLLI protocol have been proposed to shorten the acquisition dura-
multiple breath-holds, current methods generally use single breath-hold tion or to improve the accuracy or precision.32,38,39,44,45 A shorthand
protocols with single-shot 2D imaging. To achieve higher spatial resolu- nomenclature is used to label these protocols. The notation captures
tion and/or 3D imaging, segmentation may be required. how many inversions (or saturations) are included in the experiment,
MOLLI, the original scheme, was proposed by Messroghli et al.37 how many images are acquired after each inversion, and how long the
and is illustrated in Fig. 2.1. For each inversion, the MOLLI method waiting period is between inversions. For example, a 3(3)3(3)5 protocol
samples the IR curve at multiple inversion times using single-shot would indicate that there are a total of three inversions; three images
imaging spaced at heartbeat intervals. Multiple inversions are used with are acquired (over three RR intervals) after the first inversion; this is
different trigger delays to acquire measurements at different inversion followed by a waiting period of three RR intervals, and then three
times to sample the IR curve more finely. Recovery periods are needed images are acquired, followed by another three RR waiting period; finally,
between the inversions to ensure that samples from the different inver- there is a third inversion, after which five images are acquired. In an
sions are from the same recovery curve; that is, each inversion starts at extension of this nomenclature, an “s” can be added to the intervals to
CHAPTER 2  Techniques for T1, T2, and Extracellular Volume Mapping 17

LL1 LL2 LL3

Recovery Original
period MOLLI
3(3)3(3)5
protocol
Time between inversions
Raw magnitude images sorted by inversion time

Mag IR signal

Pixelwise fit
0
0 TI (ms) T1 map
FIG. 2.1  Modified Look-Locker inversion (MOLLI) recovery scheme for T1-mapping in the heart.37 The original
protocol employed 3 inversions with 3, 3, and 5 images acquired in the beats following inversions, and 3
heartbeat recovery periods between inversions, referred to here as 3(3)3(3)5. All images are acquired at the
same delay from the R-wave trigger for mid-diastolic imaging. Curve fitting is performed on a pixelwise basis
using the actual measured inversion times. IR, Inversion recovery. (From Kellman P, Hansen MS. T1-mapping
in the heart: accuracy and precision. J Cardiovasc Magn Reson. 2014;16:2.)

1 indicate that images are acquired for a certain number of seconds and
Ideal
the waiting period is in seconds: that is, 5s(3s)3s would indicate two
0.5 inversions with acquisition of images for at least 5 s, followed by a
Amplitude

Influence of recovery of at least 3 s, and a second inversion with images acquired
0 for at least 3 s. Because the number of electrocardiogram (ECG)-triggered
readout
images must be a whole number, the acquisition and recovery periods
-0.5 are rounded to the nearest multiple of the RR period to ensure an
adequate duration. To avoid acquiring too few images for low heart
-1 rates (<60 beats per minute), the sequence never acquires fewer than
0 2000 4000 6000
Time (ms) the specified number of images: that is, 5 + 3 = 8 in this example. The
recovery period is never less than the specified number of seconds.
FIG. 2.2  The apparent inversion recovery (T1*) is influenced by the
steady-state free precession readout. The effective inversion recovery
Acquiring and recovering with fixed minimum time periods helps gain
is fit using a three-parameter model, and the T1 is estimated using the independence of heart rate.
so-called Look-Locker correction. (From Kellman P, Hansen MS. T1-map- SR is an alternative to IR that has gained renewed attention. Despite
ping in the heart: accuracy and precision. J Cardiovasc Magn Reson. having a reduced dynamic range, SR has potential for improved accu-
2014;16:2.) racy. SR methods that use a saturation preparation for each measure-
ment have the benefit that each measurement becomes independent
of the others. By starting the recovery from a saturated state, the prior
history is erased. Recovery periods between successive measurements
TABLE 2.2  Reported Schemes for are not required unless longer SR times are needed for fitting. Shown
Modified Look-Locker Inversion Sampling in Fig. 2.3, the SASHA method47 uses SSFP readout very similar to the
earlier SAP-T1 method,46 which used a spoiled gradient recalled echo
MOLLI 3(3)3(3)5 Messroghli et al.37 (original publication)
(GRE) readout.
3(3)5 Ugander et al.33 and Salerno et al.44
The SASHA method acquires multiple time points on the SR curve
5(3)3 Kellman et al.39
and does a pixelwise curve fit. To acquire a fully recovered image, an
4(1)3(1)2 Schelbert et al.32
image is initially acquired before any saturation preparation: that is,
2(2)2(2)4 Salerno et al.44
starting from the equilibrium magnetization. Images are acquired on
5(3s)3 Kellman et al.57,58
successive heartbeats using SR preparations with varying trigger delays.
4(1s)3(1s)2 Kellman et al.57
In the original proposed SASHA protocol, there are 10 images acquired
5s(3s)3s Kellman and Hansen45
at saturation delays uniformly spaced over the RR interval plus the
4s(1s)3s(1s)2s Kellman and Hansen45
initial fully recovered image, which serves as an important anchor point
ShMOLLI 5(1)1(1)1(with Piechnik et al.38
for the curve fit. The order in which the various delays are acquired is
conditional fitting)
not significant for fitting, assuming ideal saturation. Importantly, the
MOLLI, Modified Look-Locker inversion; ShMOLLI, shortened SR curve recovers as T1 and is not influenced by the readout so that it
breath-hold modified Look-Locker inversion. is not shortened to an apparent T1* < T1 as in the case of MOLLI.
18 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TS1 TS2 TS3 TS4 TS5 TS6 TS7 TS8 TS9 TS10
TS

TD

SR signal

Pixelwise fit
S = A (1- exp(-T1/T1) 2-parameter model
S = A – B exp (-T1/T1) 3-parameter model

TS (ms) T1 map

FIG. 2.3  Saturation recovery single-shot acquisition (SASHA) scheme for T1 mapping in the heart.47 A single
image is acquired without saturation and used as the fully recovered measurement followed by a series of
saturation recovery images at different saturation recovery times (TSi). All images are acquired at the same
delay from the R-wave trigger for mid-diastolic imaging. Curve fitting is performed on a pixelwise basis.
(From Kellman P, Hansen MS. T1-mapping in the heart: accuracy and precision. J Cardiovasc Magn Reson.
2014;16:2.)

Therefore no correction is necessary, which eliminates the source of gaining the main benefits of SR. A combined IR/SR approach known as
many inaccuracies of the IR-based MOLLI scheme. Because the readout SAPPHIRE48 gains many of the benefits of IR and SR but still retains some
does not lead to an apparent T1*, a higher FA readout is possible, which of the problems associated with IR. Each method has its strengths and
makes up for some of the lost dynamic range in using SR. The higher weaknesses in terms of accuracy, precision, and overall reproducibility.
FA readout using SSFP with linear phase-encode ordering does slightly
alter the shape of the recovery curve, causing an apparent bias: that is, ECV Mapping Methods
the curve does not start at 0 for 0 delay. Thus the otherwise two-parameter The ECV in the myocardium may be estimated from the concentration
signal model S(t) = A(1 − exp[−t/T1]) for SR assuming ideal saturation of extracellular contrast agent in the myocardium relative to the blood
becomes a three-parameter model S(t) = A − B exp(−t/T1). The three- in a dynamic steady state.28,29,33 The ECV may be calculated as:
parameter model also absorbs any imperfection in the saturation effi-
ciency attributed to the RF saturation pulse. However, the cost of  1 1 

estimating the additional parameter is loss of precision. Therefore there ∆R1myo  T1myo post T1myo pre 
is a tradeoff between accuracy and precision in considering whether ECV = (1− hct) = (1− hct) Eq. 2.1
∆R1blood  1 1 
to use two- or three-parameter fitting. Although a center-out phase-  T1 −
blood post T1blood pre 
encode order in which the center of k-space is acquired first has the
potential to completely remove the influence of the readout, the use of
center-out ordering with SSFP is problematic because of artifacts, and where hct is the hematocrit, and the change in relaxation rate ΔR1
the use of center-out FLASH is associated with a significant loss of (where R1 = 1/T1) between precontrast and postcontrast is directly
signal-to-noise ratio (SNR). An improved version of SASHA53 that uses proportional to the Gd concentration, ΔR1 = γ[Gd] (γ = 4.5 L/mmol
variable flip angle (VFA) for SSFP readout minimizes the small bias per second for Gd-DTPA). A dynamic steady state exists for tissues
attributed to a linear phase-encode order and therefore makes two- that have a contrast exchange rate with the blood, which is faster than
parameter fitting practical with negligible loss in accuracy. The SASHA the net clearance of contrast from the blood.28 A dynamic steady state
VFA also has the benefit of improved transition to steady state, which between the plasma and interstitium may be achieved by slow intrave-
greatly reduces ghost artifacts, particularly as a result of off-resonant nous infusion24,29 or by imaging 15 minutes following an intravenous
fat, which was an issue with the original implementation. bolus administration32,33 for normally perfused myocardium, although
The SASHA sampling scheme may be altered to acquire longer satu- 15 minutes may not be adequate for recently infarcted myocardium.60
ration delay measurements by allowing one or more heartbeat recovery The bolus method is more generally used because it fits well with clinical
periods between saturations. Measurements strategies that use recovery workflow and permits conventional late enhancement imaging at the
periods such as SMART154 may improve precision for specific ranges desired dose. The ECV formula (Eq. 2.1) implies that our myocardial
of T1 and heart rate but reduce the overall SNR efficiency somewhat. ECV measurements include both the intravascular and extravascular
Optimized sampling strategies for SASHA are described for both two-51 space. The factor (1 − hct), which varies between individuals, repre-
and three-parameter52 fittings. Schemes that simply use a MOLLI strat- sents the blood volume of distribution (blood ECV) and converts the
egy replaced with SR59 incur the problems of an apparent T1* without equation from a partition coefficient calculation to a myocardial ECV.
CHAPTER 2  Techniques for T1, T2, and Extracellular Volume Mapping 19

Precontrast and postcontrast image series are acquired in separate


breath-holds, which are typically 15 to 30 minutes apart. Even small Reproducibility: Accuracy, Precision, and
differences in respiratory position or changes in patient position attrib- Confounding Factors
uted to movement will cause significant misregistration of the images. The sensitivity for detecting abnormal elevation of T1 and ECV derived
Care must be taken that the patient does not move, and coregistration from measurements of T1 is fundamentally limited by the reproduc-
of the precontrast and postcontrast images must be performed using ibility of T1 estimates. The reproducibility is affected by a number of
a nonrigid image registration to mitigate in-plane motion.40,41 The factors that include measurement precision and accuracy as well as
complete processing workflow may be fully automated39 to include biological variability. It is important to establish normal baseline values
precontrast and postcontrast T1 mapping with respiratory motion cor- and ranges21 for the specific technique and protocol.
rection and blood pool segmentation. A prototype ECV mapping tool The precision refers to the effect of random noise on the measure-
has been integrated onto a clinical scanner.61 Inline ECV mapping pre- ment and is a function of the number and timing of measurements
sumes that the hematocrit has been measured before the scan using along the IR or SR curve, the SNR, the tissue T1, and the method of
point-of-care devices. For instances for which the hematocrit has not fitting. The precision of T1 methods has been well characterized,45
been measured, it is possible to estimate an approximate hematocrit and pixelwise maps of standard deviation (SD) attributed to thermal
from the value of blood T1 and use this derived hct for a synthetic noise may be generated as a quality map.57 Example T1 and corre-
ECV.62 In this way, the ECV map may be available immediately follow- sponding SD maps are shown (Fig. 2.5) for a normal subject using
ing the completion of the postcontrast T1 map, regardless of whether four different mapping protocols. This example illustrates several
or not hct was measured. points: (1) normal T1 values vary considerably by method and spe-
Although detecting global changes that are difficult to see in non- cific imaging protocol, (2) the SD varies across the heart as the SNR
quantitative imaging has been a driver for quantitation, there is also varies because of surface coil drop off with distance, and (3) different
potential value in cases of focal abnormalities in determination of methods of T1 mapping have different precisions, leading to differing
whether remote tissue is in fact normal and in measurement of border sensitivities.
zones. Given adequate precision, the strength of pixelwise mapping of Other factors contributing to measurement error that are not random
T1 is the ability to detect small abnormalities and discriminate spatial further limit the reproducibility and are highly dependent on the tech-
structures. Fig. 2.4 illustrates T1 and ECV maps in cases ranging from nique and specific protocol. Some of these factors may depend on the
focal scar to patchy to diffuse. scanner adjustments such as off-resonance errors58 due to center fre-
quency adjustment error, inability to shim the volume perfectly, or
because of variation in the actual FA attributed to transmitter field
2000 ms inhomogeneity or FA adjustment error. Techniques must be designed
to be robust in the presence of such variations because these variables
Pre-T1

are generally not well controlled. Variations may also relate to the actual
tissue composition. For instance, IR methods using SSFP readout are
dependent on the tissue T243,45,47 and MT resulting from the macro-
0 molecular content of proteins.63 Still other variables relating to the
700 ms patient such as heart rate, heart rate variability, and cardiac or respira-
tory motion may affect estimates of T1. The accuracy for a given tech-
Post-T1

nique may also be influenced by specific protocol parameters such as


FA, matrix size, repetition rate (TR), and sampling strategy. The sensitivity
of MOLLI (Fig. 2.6) and SASHA (Fig. 2.7) techniques has been calcu-
300 lated based on Bloch simulations for a common protocol with matrix
size 256 × 144, partial Fourier factor 7/8, factor 2 acceleration, bandwidth
1085 Hz/pixel, TR = 2.8 ms, excitation FA (FA) 35 degrees, minimum
LGE

TI 100 ms, and TI increment 80 ms. These calculations illustrate the


dependencies on T2, HR, FA, and off-resonance leading to an under-
estimation of T1. The MOLLI-based methods have the largest sensitivity
100%
to adjustments and protocol parameters. For a 25% variation in transmit
FA often seen in practice, the variation in apparent native T1 is 2%.
For 100 Hz off-resonance, the variation in apparent native T1 can be
ECV

as high as 4%. For heart variation from 60 to 120 beats per minute,
there is a 2.5% variation in apparent native T1. Over the range of
normal to highly edematous T2 (45 to 90 ms), the native T1 will vary
0% approximately 4%. The SR-based method of SASHA is much less sensi-
(A) Chronic MI (B) HCM (C) Cardiac
tive to these variables. However, the IR methods such as MOLLI achieve
amyloidosis a higher precision.45
FIG. 2.4  Examples illustrating mapping in cases ranging from focal and An example of how edematous myocardium may confound the
patchy to diffuse disease. Native (precontrast) T1 maps (first row), post- measurement of native T1 (Fig. 2.8) is presented for a subject with
contrast T1 maps (second row), late gadolinium enhancement (LGE)
acute MI. The edematous region of MI has an elevated value of T2 that
(third row), and extracellular volume fraction (ECV) maps (fourth row)
for patients with (A) chronic myocardial infarction (MI), (B) hypertrophic is 15 ms greater than remote. The increase in T1 for this MI region is
cardiomyopathy (HCM), and (C) cardiac amyloidosis. (From Kellman P, 227 ms above remote measured using MOLLI, and elevated 199 ms
Wilson JR, Xue H, Ugander M, Arai AE. Extracellular volume fraction measured using SASHA. The difference in T1 elevation 227 − 199 =
mapping in the myocardium, part 1: evaluation of an automated method. 28 ms is explained in part due to the 15 ms elevation of T2 attributed
J Cardiovasc Magn Reson. 2012;14:63.) to edema as seen in plot of MOLLI sensitivity to T2 (see Fig. 2.6) and
20 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

SASHA SASHA
MOLLI ShMOLLI 1-10 1-10
5s(3s)3s 5 (1)1(1)1 2-param fit 3-param fit

2000

1500

1000 T1 (ms)
500

0
Mean = 1017 ms Mean = 954 Mean = 1201 Mean = 1209

100

75

50 SD (ms)

25

0
SD = 31 ms/pixel SD = 51 SD = 47 SD = 76

FIG. 2.5  Examples of in vivo T1 maps and corresponding pixelwise standard deviation (SD) maps acquired
using modified Look-Locker inversion (MOLLI) 5s(3s)3s, shortened breath-hold MOLLI, and saturation recovery
single-shot acquisition (SASHA) protocols with two- and three-parameter fitting. Variation in SD across the
heart is apparent because of variation in signal-to-noise ratio from surface coil sensitivity roll-off. MOLLI has
the best precision but underestimates T1 as a result of the approximate nature of the Look-Locker correction
and because of magnetization transfer. Note that SASHA with two-parameter fitting has a small T1 under-
estimation; three-parameter fitting is more accurate but has significant loss of precision. (From Kellman P,
Hansen MS. T1-mapping in the heart: accuracy and precision. J Cardiovasc Magn Reson. 2014;16:2.)

Sensitivity to T2 Sensitivity to heart rate


0 0

-20 -20
T1 error (ms)

T1 error (ms)

-40 200 -40


400 200
400
600
-60 800 -60 600
1000 800
1000
-80 1200 -80
1200

-100 -100
45 50 55 60 65 70 75 80 85 90 60 70 80 90 100 110 120
T2 (ms) Heart rate (beats/min)

Sensitivity to flip angle Sensitivity to off-resonance


0 0

200
-20 -20
400
T1 error (ms)

T1 error (ms)

600
-40 -40 800
200 1000
400 1200
-60 600 -60
800
-80 1000 -80
1200
-100 -100
0 5 10 15 20 25 30 35 -100 -50 0 50 100
Filp angle (deg) Off-resonance frequency (Hz)
FIG. 2.6  Sensitivity of T1 estimates using the modified Look-Locker inversion method to T2, heart rate, flip
angle, and off-resonance frequency using a 5s(3s)3s sampling scheme.45

because of the different degree of MT between the MI and remote Limitations and Potential Pitfalls
region. The MT will be less in the edematous region because of increased The use of T1 to characterize myocardial tissue is a simplification or
water content; thus the T1 underestimation of MOLLI attributed to first-order model. The myocardium consists of several compartments
MT will be less, corresponding to an increased T1. The SASHA method such as myocytes, interstitium, and capillaries, each with different con-
is relatively insensitive to both T2 and MT. stituent molecular makeup to include free water and macromolecules.
CHAPTER 2  Techniques for T1, T2, and Extracellular Volume Mapping 21

0
Sensitivity to T2 Sensitivity to heart rate
0

-20 -20

T1 error (ms)

T1 error (ms)
-40 -40
200 200
400 400
-60 -60
600 600
800 800
-80 1000 -80 1000
1200 1200
-100 -100
45 50 55 60 65 70 75 80 85 90 60 70 80 90 100 110 120
T2 (ms) Heart rate (beats/min)

Sensitivity to flip angle Sensitivity to off-resonance


0 0

-20 -20
T1 error (ms)

T1 error (ms)
-40 -40
200 200
-60 400 400
-60
600 600
800 800
-80 1000 -80 1000
1200 1200
-100 -100
0 10 20 30 40 50 60 70 -100 -50 0 50 100
FA (degrees) Off-resonance frequency (Hz)

FIG. 2.7  Sensitivity of T1 estimates using the saturation recovery single-shot acquisition variable flip angle
two-parameter method to T2, heart rate, flip angle, and off-resonance frequency using an NS + [(0)1]9 sam-
pling scheme.51

T1 maps

T2 map MOLLI SASHA SASHA

Same window level Level adjusted

Edema Remote Difference


T1 (MOLLI) 1231 1004 227
T1 (SASHA) 1382 1183 199
T2 59 44 15

FIG. 2.8  Example of T1 and T2 maps for subject with edematous acute myocardial infarction. The edematous
region is elevated 15 ms above remote, and T1 is elevated 227 ms (using the modified Look-Locker inversion
[MOLLI]) and 199 ms (using saturation recovery single-shot acquisition [SASHA]). The greater increase in
apparent T1 for MOLLI edematous is because of the confounding effects of T2 and magnetization transfer.
Note that the baseline values for SASHA are greater than for MOLLI.

The interactions between the proton spins contributing to the observed factors described earlier, wherein estimates of T1 may be affected by
MRI signal are highly complex. Despite the fact that current T1 mapping tissue T2, MT, patient heart rate, off-resonance (B0), or actual FA (trans-
largely ignores these complexities in measuring a single macroscopic mitted B1), there are other artifact mechanism that may affect measure-
T1, the T1 measurements have been demonstrated to have potential in ment. One of the most important limitations is the partial volume
clinical use. contamination of the myocardium by the adjacent blood pool signal,
The potential for quantification of myocardial tissue is to detect which is generally greater than the myocardium. The blood pool con-
small changes as a result of disease processes or treatment. The desire tamination is caused by both limited in-plane spatial resolution as well
to measure subtle changes places stringent requirements on reliability as through-plane. A voxel in a myocardial region may contain a mixture
of the measurement. For this reason there is great emphasis on accuracy, of myocardium and blood and thus be biased. The degree of bias as a
precision, and general reproducibility. In addition to the confounding result of this partial volume between myocardium and blood will depend
22 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

1100 ms 650 ms

198 ms 153 ms

1.4 x 1.9 mm2 1.9 x 2.3 mm2

1600 1600
1500

T1 (ms)
T1 (ms)

1400 ≈ 10 mm 1400 ≈ 7 mm
1300
1200 1200

1000 1100
1000
−5 −4 −3 −2 −1 0 1 2 3 4 5
−6 −4 −2 0 2 4 6 Pixels
Pixels
A B
FIG. 2.9  Example of T1 maps in two subjects. (A) Subject with heart rate of 58 beats per minute acquired
using a modified Look-Locker inversion protocol with 256 × 144 matrix. (B) Subject with heart rate of approxi-
mately 90 beats per minute using a 192 × 120 matrix. Although the interpolated maps are of good quality,
the subject with higher heart rate and thinner wall has only about 3.5 pixels across the septum, leading to
a degree of partial volume error in region of interest measurements. (From Kellman P, Hansen MS. T1-mapping
in the heart: accuracy and precision. J Cardiovasc Magn Reson. 2014;16:2.)

on proximity to the blood pool as well as effective spatial resolution. In addition to limited spatial resolution, there may be additional
With in-plane image resolution of 1.5 to 2 mm and thin myocardial blurring as a result of cardiac motion in cases of higher heart rates,
walls (<1 cm), there may be only a few (2–5) pixel widths across the variations in RR interval, or imaging during a period of cardiac cycle in
myocardial wall. Gibb’s ringing can lead to fluctuations of ≥5% in T1 which there is wall motion. The temporal duration of image acquisition
values even mid-wall, and errors can be much larger close to the blood can be reduced to mitigate blurring by using higher parallel imaging
myocardial boundary. For slices for which there is through plane angu- acceleration, although this may come at the expense of loss in SNR. The
lation of the wall, particularly at the apex, there can be additional partial partial volume bias attributed to potential blurring at high heart rates
volume effects. Caution must be exercised in making and interpreting is important to consider and is distinct from the previously described
measurements for thin walls, particularly when comparing populations heart rate dependence of the MOLLI-type sequences. Finally, imper-
with different wall thickness. fect respiratory motion correction may lead to an additional source
Fig. 2.9 illustrates the effect of spatial resolution on native T1 of blurring.
maps for two subjects with different wall thickness and heart rates. There are partial volume errors for voxels with a mixture of myo-
Subject 1 (A) has a myocardial wall thickness of approximately 10 mm cardium and fat.64 This may occur at tissue boundaries or within the
(approximately 6 pixels) in the septum and heart rate approximately myocardium in the case of lipomatous metaplasia of replacement fibrosis,
55 beats per minute. A profile plot of T1 across the septal wall shows which is commonly seen in chronic MI or other scarring. The presence
a ringing of approximately 50 ms (5%) attributed to Gibb’s effect and of fat leads to a bias in T1 measurement, which may be either a positive
a smooth transition between the myocardium and adjacent blood pool or negative bias depending on the specific protocol and off-resonance
of approximately 2 pixels. Subject 2 (B) has a thinner myocardial wall frequency. T1 biases will be additive or subtractive depending on whether
(approximately 7 mm) and a higher heart rate. At the higher heart rate the center frequency corresponds to the myocardium and fat is in-phase
(approximately 92 beats per minute), a slightly reduced matrix size was or out-of-phase, respectively. Lipomatous metaplasia is prevalent and
used to mitigate temporal motion blur, resulting in a myocardial wall often difficult to detect at low fat fractions. The influence on T1 at
thickness corresponding to 3 to 4 pixels. In this case, the measured these low-fat fractions is significant and is readily misinterpreted.
T1 (see profile plot) is contaminated by adjacent blood for all but Although this may be mitigated to some extent by means of chemical
the mid-wall, thereby compromising the accuracy of measurements. shift fat suppression (i.e., fat saturation), the fat saturation may also
Most clinical image viewers use interpolation to display images when influence the measurement of normal myocardial T1 if there is any
zoomed, thus hiding the true resolution limits that would be appar- significant off-resonance error caused by imperfect shim.
ent when displaying the pixelated image. Depending on the displayed Quality maps such as goodness of fit or standard deviation maps57
range (window level), the zoomed and interpolated maps often appear may be used for quality control as a means of detecting cases for which
to have distinct boundaries, even in situations with significant partial there is increased error because of cardiac or respiratory motion in
volume contamination. cases where the motion is fairly extreme. However, quality maps do not
CHAPTER 2  Techniques for T1, T2, and Extracellular Volume Mapping 23

account for all partial volume errors. Other potentially useful quality
T2 MAPPING
metrics are off-resonance field maps and FA maps. When ECV is ele-
vated, it may not be clear whether this is because of fibrosis, edema, The transverse relaxation time (T2) of myocardial tissue may be altered
or both, which may be either diffuse or focal. In such instances, pre- by disease. Edematous tissue with increased water content has elevated
contrast T1 or T2 maps, in addition to patient history and contextual T218,19 and may be a by-product of an acute disease process caused by
imaging clues like signs of heart failure, may help to differentiate these inflammation. Presence or absence of edema is used to differentiate
mechanisms. acute and chronic MI,65 to identify the area at risk,4 and in assessment
One of the attractive aspects of ECV measurement is that the value of nonischemic disease such as myocarditis.66 Increased iron concentra-
represents a quantitative measurement. However, biases in the measure- tion leads to a reduction in both T2 and T2* as seen in iron overload
ment of precontrast and postcontrast T1 do not cancel out, and methods (e.g., due to thalassemia).17 Although T2-weighted imaging may be
with different baseline normal values of T1 such as MOLLI and SASHA used to detect focal abnormalities, it is more challenging to detect global
will yield different baseline normal values of ECV. changes in T2 caused by diffuse disease where there generally is not a
The ability to distinguish partial volume border pixels and correctly remote healthy point of reference. T2 mapping provides a means of
determine whether pixels are contaminated by partial volume effects, automatic, quantitative measurement of T2 on a pixelwise basis that
or are true pathophysiology such as subendocardial or subepicardial may be used in diagnosis of disease across a spectrum of heterogeneity
fibrosis, is a residual and important issue as it was shown in the T1 from diffuse to focal.
mapping example. This issue is not only important in terms of the
visual readout but also may introduce biases into quantitative measure- Methods
ments, which become particularly significant in the context of more T2 mapping is performed by acquiring multiple images (N ≥ 2) with
subtle diseases. One approach to reducing the bias in measurements is different T2 weightings and estimating the T2 at each pixel by per-
to restrict the measurement to the mid-wall region, although one must forming a two-parameter curve fit to the measured signal for each
exercise caution for subjects with thin-walled myocardium or patients pixel Sn(x, y). A mono-exponential model Sn = PD exp(−TEn/T2) is
with thin rims of subendocardial fibrosis. used, where T2 is the unknown to be estimated, PD is the unknown
proton density, and TEn (known) is the effective echo time of the indi-
Summary vidual T2-weighted image for the nth acquisition (n = 1, 2, …, N).
There are a number of methods and imaging protocols for T1 mapping T2-weighted images may be acquired by a number of methods.
in the heart, and new methods are emerging. A comparison of several Methods for cardiac MRI T2 mapping include (1) T2-prepared SSFP
of the widely used protocols is shown in Table 2.3. This assessment of (T2p-SSFP),67 (2) multiecho spin echo (MESE),68 and (3) a hybrid
pros and cons, albeit subjective, is meant to capture the key issues that of MESE and either turbo spin echo (TSE)69,70 or gradient spin echo
relate to overall reproducibility and affect the ability to detect subtle (GraSE).71,72
changes in T1 from normal values. The inherent difficulty of tightly The most widely used method at this time is the T2p-SSFP. An
controlling the shim and uniformity of transmit FA in clinical scanners example of pixelwise T2 mapping for a subject with acute MI is shown
reduces the overall reproducibility of the MOLLI-based methods using in Fig. 2.10 for T2p-SSFP. In this example, three T2-weighted images
IR and SSFP readout as compared with the SR approach. Limitations are acquired with echo times of TE = 0, 25, and 55 ms, where the
such as partial volume effects between myocardium and blood are TE = 0 is acquired without any T2 preparation. The anterior MI is
common to all of these protocols. In this table, falsely elevated T1 due edematous, with elevated T2 approximately 72 ms, and the normal
to partial volume with fat is considered an artifact. myocardium remote from the MI has a T2 of approximately 48 ms.
ECV mapping appears promising to complement LGE imaging in The single-shot T2-prepared readout approach acquires each
cases of more homogenously diffuse myocardial disease states, which T2-weighted image in a single beat, which has the benefit of being
affect the myocardial extracellular space. The ability to display ECV robust to cardiac and respiratory motion. ECG gating is used with
maps in quantitative units that may be interpreted on an absolute scale acquisition in mid-diastole to minimize cardiac motion; respiratory
offers the potential for simplified detection and measurement of the motion may be mitigated by breath-holding or navigated acquisition.
extent of abnormalities affecting the myocardial ECV. There is a con- Residual in-plane respiratory motion is typically corrected using non-
tinuous spectrum of spatial heterogeneity between diffuse and focal rigid image registration. Either SSFP or FLASH readout may be used.67
fibrosis. The use of pixelwise mapping offers a new tool to better assess SSFP readout is most commonly used to maximize SNR, particularly
the heterogeneity of the myocardial tissue and to measure ECV of the at 1.5 T. Single-shot SSFP readout uses a linear phase-encode order
diseased regions. to minimize artifacts caused by eddy currents. It is possible to use

TABLE 2.3  Summary of Pros and Cons of Various Reported T1-Mapping Protocols
MOLLI MOLLI MOLLI SASHA SASHA SASHA VFA
3(3)3(3)5 5s(3s)3s 4s(1s)3s(1s)2s ShMOLLI 2p-fit 3p-fit 2p-fit
Short breath-hold − + + + + + +
HR insensitivity − + + + ++ ++ ++
Absolute accuracy − − − − + ++ +
Precision ++ ++ ++ + + − +
Few image artifacts + + + + − − +
Reproducibility − + + + − − ++

HR, Heart rate; MOLLI, Modified Look-Locker inversion; SASHA, saturation recovery single-shot acquisition; ShMOLLI, shortened breath-hold
modified Look-Locker inversion; VFA, variable flip angle.
24 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TE = 0 TE = 25 TE = 55

A 100
1 90
80
Signal intensity (a.u.)

0.8
70
0.6 60
50 ms
0.4
40

0.2 30
20
0 10
0 10 20 30 40 50 60
TE (ms) 0
B C
FIG. 2.10  Illustration of T2 mapping approach. T2 mapping approach using T2p-steady-state free precession:
(A) acquiring images at three echo times, (B) performing a monoexponential curve fit to signal intensities at
each pixel to estimate T2 to (C) produce a pixelwise T2 map.

centric phase encode ordering with a single-shot FLASH readout. By The MESE approach acquires the data in a segmented fashion
using centric ordering, the center of k-space, which determines the with multiple echoes, with different TEs acquired at each beat for a
principal contrast, is acquired immediately following the T2 prepara- given phase encode using a spin echo train. This requires a lengthy
tion. However, by using linear ordering, there is some delay follow- breath-hold or navigated scan and is included here for complete-
ing the T2 preparation and the center of k-space during which there ness of discussion. The MESE is acquired in conjunction with a dark
is T1 regrowth of the magnetization that leads to a T1-dependent blood preparation to avoid blood flow artifacts, typically double
bias = PD(1−exp[−TS/T1]), where TS is the time from end of the prepa- inversion recovery (DIR) applied immediately following the R-wave
ration to the center of k-space. ECG trigger.
With the T2p-SSFP approach, it is necessary to have several heart- The acquisition of MESE may be accelerated by acquiring mul-
beats (typically 3 or 4 seconds) between image acquisitions to allow tiple phase encodes at each echo time using a hybrid approach such
for full magnetization recovery (dependent on T1). If there is insufficient as GraSE,77,78 which employs a short echo train gradient recalled echo
signal recovery, the images will be affected by the preceding image, thus planar imaging (EPI) readout between 180-degree refocusing pulses.
altering the estimated value of T2 and introducing a dependence on In this way, 3 to 7 echoes may be acquired per heartbeat, accelerating
T1. Alternatively, it has been proposed to use a saturation preparation the image acquisition for T2 mapping.71,72 This scheme is also used
to reset the initial condition for each heartbeat to a fixed value.73 In in conjunction with a DIR dark blood preparation and may be either
this way, the number of images acquired may be increased because no breath-held or navigated. EPI readout is prone to artifacts caused by
recovery beats are required, but there is a significant SNR loss because off-resonance or fat chemical shift and is often used in conjunction
of the incomplete SR. with chemical shift fat saturation. To mitigate ghosting artifacts, a linear
The T2 preparation is implemented by tipping the magnetization interleaved phase-encode order is typically used with echo time shift-
vector down on the transverse plane for a prescribed duration (TE), ing to ensure a smooth phase transition across k-space for different
followed by tipping the vector back, and crushing any residual trans- EPI echoes.
verse magnetization. Refocusing pulses are used between the tip-down Spin echo methods are highly motion sensitive. DIR dark blood
and tip-up pulses to avoid dephasing caused by myocardial motion or preparations rely on imaging at the same cardiac phase as the DIR
blood flow, which can result in artifacts. T2 preparations have been preparation, which is difficult to achieve at higher heart rates or in
designed that are robust to variations in both B0 (off-resonance) and patients with motion during the imaging interval in diastole. Further-
B1 (effective transmitted FA). These are based on either composite more, the spin echo readout itself is sensitive to motion. It is common
pulses20,74 or adiabatic RF designs.75,76 The minimum TE for the T2 for myocardial wall motion to cause signal loss or complete signal
preparation is dependent on the duration and number of refocusing drop out.
pulses. For designs that use several refocusing pulses, a short TE is
not feasible20 and in this case a readout without any T2 preparation Reproducibility: Accuracy, Precision, and
is used for an effective TE = 0. A significant error in FA because of Confounding Factors
in homogeneity in transmitted B1+ or calibration error resulting in Similar to the discussion of T1 and ECV mapping, many of the same
a reduced signal for the prepared measurements and not the TE = 0 factors and considerations affect reproducibility. These include blood
measurement may lead to an underestimate in T2. This may be mitigated pool contamination caused by partial volume effects, dependence
using T2 preparation for all measurements and shortening the overall on off-resonance and transmitted FA, confounding influence of T1,
refocusing duration. and SNR.
CHAPTER 2  Techniques for T1, T2, and Extracellular Volume Mapping 25

2p fit 3p fit 3p fit


(0, 25, 55) (0, 25, 55) (0, 25, 55, ∞)
120
100
80
T2 map 60 ms
40
20
0
50
40
SD map 30 ms
20
10
0

FIG. 2.11  T2 (top row) and corresponding standard deviation (SD) maps (bottom row) using T2p- steady-state
free precession method with two-parameter fitting (left column), three-parameter fitting including a constant
bias term to account for T1-dependent magnetization regrowth (center column), and three-parameter fit with
bias term and additional measurement without T2 preparation (TE = ∞) (right column). The two-parameter
fit overestimates the T2 because of the bias term. The bias is mitigated using three-parameter fitting but
greatly increases the SD unless an additional measurement at TE = ∞ is included.

With the T2p-SSFP approach, there is a T1 bias due to the mag- 19


25
netization regrowth following the T2 preparation and the acquisition 18 40
of the center of k-space. This signal bias can lead to an overestima- FA
50
tion of T2 by 5 to 10 ms. Although in theory a three-parameter fit 17
70
(S = PD exp[−TE/T2] + b) may be used to eliminate this confounding
16
bias variable, b, three-parameter fit with only a few relatively short TE
measurements is very noisy. It has been proposed to directly measure 15
T2 error (ms)

the bias term79 by acquisition of an image using an SR preparation


14
without any T2 preparation, which corresponds to the TE = ∞ image.
In this way, a three-parameter fit may be used without incurring a large 13
SNR penalty, thus effectively eliminating this T1-confounder. This is
illustrated in Fig. 2.11, which shows the reduced benefit of a three- 12

parameter fit in reducing the T2 bias, and the improvement in SD of 11


the three-parameter fit using the additional TE = ∞ measurement. The
SSFP readout itself has image contrast dependent on T1 and T2. To 10

a large extent the image contrast is shared for all of the T2-weighted
9
images and does not affect the fit. However, single-shot imaging is -150 -100 -50 0 50 100 150
acquired on the approach to steady state, and the contrast depends Off-resonance frequency (Hz)
on the initial conditions, which in turn depends on the TE of the T2 FIG. 2.12  T2 estimates using T2p–steady-state free precession method
preparation. Thus there is some residual T1 contrast dependent on the are sensitive to off-resonance and flip angle (FA), with increasing errors
readout that may alter the T2 estimate. at higher FA. Significant errors are observed for off-resonance > ± 50 Hz.
The T2-SSFP measurement is sensitive to off-resonance. The SSFP
readout has an off-resonance response that is different for the different
T2 preparation echo times (TEs). This leads to an overall dependence
of T2 measurement on the off-resonance frequency. The T2 error has pathways, which may also lead to a T1 dependence in the contrast.
been characterized experimentally80 for a specific imaging protocol and This bias is not well characterized, and there are no methods for
found to vary significantly for off-resonance > ±150 Hz, and errors eliminating it.
of approximately 5% at ±100 Hz. The frequency dependence of the The precision of the T2 measurement is determined by the SNR,
T2 preparation also contributes to the response. The off-resonance number of measurements, the actual value of T2, and specific echo
sensitivity is reduced by using a lower excitation FA. The T2 error is times used. The SD of the T2 estimate attributed to random noise (Fig.
shown in Fig. 2.12, which illustrates off-resonance sensitivity and the FA 2.13) was calculated analytically for a myocardial T2 of 45 ms at various
dependence, as well as on-resonance bias described above. The composite SNR for three echo time measurements (solid lines) and four echo time
hard pulse design is wideband, but the adiabatic designs used for greater measurements (dotted lines). Measurements were equally spaced at 0,
independence of FA generally have a narrower bandwidth. Thus care TEmax/2, TEmax for three TEs and 0, TEmax/3, 2TEmax/3, and TEmax for
must be taken to ensure the volume is properly shimmed. The T2p- four TEs. These agree well with numerical simulation performed for a
SSFP approach will not work well in the presence of device implants. specific T2-SSFP protocol80 with three measurements constrained to
Using the GraSE approach, there is also a significant bias.71 Using be 0, TE, and 2TE, where the best TE was found for normal myocardium
a TSE approach, it is difficult to completely eliminate stimulated echo at T2 = 45 ms to be in the range of 30 to 40 ms: that is, echo times (0,
26 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

T2 = 45 (ms) at 3 T where the SNR of FLASH may be adequate. It is also impor-


tant to avoid T2 mapping after administering Gd contrast to avoid
4 30
50
short T1.
100 Current imaging protocols have been designed for myocardial T2
3.5
200 measurement and are not suitable for accurate measurement of blood
3
T2. The echo times are much shorter than the T2 of the blood, leading
to poorly conditioned fitting and sensitivity to blood signal loss due to
dephasing at longer echo times.
T2 SD (ms)

2.5

2 Summary
T2 mapping in the heart using 2D imaging is feasible in a clinical
1.5 environment and has sufficient accuracy and precision to be of value
to answer a number of clinical questions. Absolute baseline values will
1
depend on the field strength, and to some extent on the method. T2p-
0.5
SSFP is the most widely used method and is fairly robust in terms of
image quality.
0
30 40 50 60 70 80 90 100 110 120
Maximum TE (ms) CONCLUSION
FIG. 2.13  Variation of standard deviation (SD) for T2 estimates at Future developments to address the current limitations in quantita-
SNRs = 30, 50, 100, and 200. Equally spaced echo times are between 0
tive measurement are focused on reduction in partial volume effects,
and the maximum echo time (TEmax) using three measurements (solid
lines) and four measurements (dotted lines). Baseline normal myocardium differentiation between edema and fibrosis, establishment of base-
T2 = 45 ms is used in this calculation, and SD is fairly insensitive to line normal values, standardization, and quality control. Quantita-
TEmax over a wide range (50–100 ms). tive methods of T1, T2, and ECV mapping have been successful in
population-based studies. Addressing the remaining issues is key to the
reliable application of quantitative methods to the diagnosis of disease in
individual patients.
T2 SD A better understanding of confounding factors such as the effect of
100 20
T2 on T1 measurement is important for interpretation of measure-
80 ments. Understanding how scanner adjustments such as off-resonance
15
or FA may affect quantitative measurements is important for the reliable
60
10 ms
detection of subtle disease. Multiparametric measurements may help
40 differentiate between edema and fibrosis and may also play a role in
5 disentangling the coupling in measurements of T1 and T2.
20
Partial volume contamination by adjacent blood pool or by intra-
0 0 myocardial fat is an important issue in current protocols. Use of higher
FIG. 2.14  Examples of T2 and standard deviation (SD) maps. T2p-steady- spatial resolution or 3D imaging helps mitigate partial volume to some
state free precession is used for a subject with acute myocardial infarc- extent. Suppression of blood or fat may be required to further reduce
tion. SD is increased in the edematous region because of elevated T2. this contamination; however, these methods may introduce other
side effects.
Standardization encompasses methods for acquiring quantitative
maps, displays, ROI measurements, and reporting. Visualization of para-
30, 60 ms) to (0, 40, 80 ms). The SD may be calculated on a pixelwise metric maps is most commonly done by color display. However, there is
fashion in a similar manner as for T157 or T2*.81 Examples of T2 and no current consensus on the scale or colormap. The visual display has
SD maps for the case shown in Fig. 2.10 are shown in Fig. 2.14. The a significant effect on classification of normal versus abnormal tissue
pixelwise noise SD for this case is approximately 2.1 ms in a remote or on the assessment on quality of the map. Use of quality control
region, with T2 = 44 ms and an SD approximately 3.7 ms in the edema- phantoms may also play a role in standardization.82
tous region with elevated T2 = 68 ms. Quantitative mapping has the potential for increasing the objec-
tiveness of CMR and improving the sensitivity for detecting subtle
Limitations and Potential Pitfalls changes in the global state due to disease processes. Mapping has come
The dark blood preparation used in spin echo-based techniques such a long way in the past decade, and technical developments continue to
as GraSE helps to eliminate contamination of myocardial signal caused improve methods and find new and interesting clinical and research
by partial volume effects. However, spin echo readouts and DIR dark applications.
blood preparations are highly motion sensitive and prone to signal loss,
particularly at higher heart rates. Off-resonance leads to bias errors in
SSFP methods and to artifacts in EPI methods; therefore it is essential
ACKNOWLEDGMENTS
to ensure the volume is adequately shimmed. Portions of text and figures have been excerpted and modified from the
T1-weighted contrast may confound the T2 measurement caused Journal of Cardiovascular Magnetic Resonance Open Access publication.39,45
by magnetization regrowth in the case of T2p-SSFP and stimulated
echoes in SE methods. Methods have been proposed to mitigate bias
in T2p-SSFP79 but may not be widely available. This may be avoided
REFERENCES
by using T2p single-shot FLASH with centric readout, particularly A full reference list is available online at ExpertConsult.com
CHAPTER 2  Techniques for T1, T2, and Extracellular Volume Mapping 26.e1

REFERENCES 21. Moon JC, Messroghli DR, Kellman P, et al. Myocardial T1 mapping and
extracellular volume quantification: a Society for Cardiovascular
1. Abdel-Aty H, Zagrosek A, Schulz-Menger J, et al. Delayed enhancement Magnetic Resonance (SCMR) and CMR Working Group of the European
and T2-weighted cardiovascular magnetic resonance imaging differentiate Society of Cardiology consensus statement. J Cardiovasc Magn Reson.
acute from chronic myocardial infarction. Circulation. 2004;109:2411–2416. 2013;15:92.
2. Bohl S, Wassmuth R, Abdel-Aty H, et al. Delayed enhancement cardiac 22. Pennell DJ, Sechtem UP, Higgins CB, et al. Clinical indications for
magnetic resonance imaging reveals typical patterns of myocardial injury cardiovascular magnetic resonance (CMR): Consensus Panel report. Eur
in patients with various forms of non-ischemic heart disease. Int J Heart J. 2004;25:1940–1965.
Cardiovasc Imaging. 2008;24:597–607. 23. Hunold P, Schlosser T, Vogt FM, et al. Myocardial late enhancement in
3. Friedrich MG, Kim HW, Kim RJ. T2-weighted imaging to assess contrast-enhanced cardiac MRI: distinction between infarction and
post-infarct myocardium at risk. JACC Cardiovasc Imaging. non-infarction-related disease. AJR Am J Roentgenol. 2005;184:1420–1426.
2011;4:1014–1021. 24. Sado DM, Flett AS, Moon JC. Novel imaging techniques for diffuse
4. Ugander M, Bagi PS, Oki AJ, et al. Myocardial edema as detected by myocardial fibrosis. Future Cardiol. 2011;7:643–650.
pre-contrast T1 and T2 CMR delineates area at risk associated with acute 25. Gai N, Turkbey EB, Nazarian S, et al. T1 mapping of the gadolinium-
myocardial infarction. JACC Cardiovasc Imaging. 2012;5:596–603. enhanced myocardium: adjustment for factors affecting interpatient
5. Zagrosek A, Abdel-Aty H, Boyé P, et al. Cardiac magnetic resonance comparison. Magn Reson Med. 2011;65:1407–1415.
monitors reversible and irreversible myocardial injury in myocarditis. 26. Mewton N, Liu CY, Croisille P, Bluemke D, Lima JA. Assessment of
JACC Cardiovasc Imaging. 2009;2:131–138. myocardial fibrosis with cardiovascular magnetic resonance. J Am Coll
6. Kim R, Wu E, Rafael A, et al. The use of contrast-enhanced magnetic Cardiol. 2011;57:891–903.
resonance imaging to identify reversible myocardial dysfunction. N Engl J 27. Lee JJ, Liu S, Nacif MS, et al. Myocardial T1 and extracellular volume
Med. 2000;343:1445–1453. fraction mapping at 3 tesla. J Cardiovasc Magn Reson. 2011;13:75.
7. Messroghli DR, Walters K, Plein S, et al. Myocardial T1 mapping: 28. Arheden H, Saeed M, Higgins CB, et al. Measurement of the
application to patients with acute and chronic myocardial infarction. gadopentetate dimeglumine at echo-planar MR imaging to quantify
Magn Reson Med. 2007;58:34–40. myocardial infarction: comparison with 99m Tc-DTPA autoradiography
8. Messroghli DR, Niendorf T, Schulz-Menger J, Dietz R, Friedrich MG. T1 in rats. Radiology. 1999;311:698–708.
mapping in patients with acute myocardial infarction. J Cardiovasc Magn 29. Flett AS, Hayward MP, Ashworth MT, et al. Equilibrium contrast
Reson. 2003;5:353–359. cardiovascular magnetic resonance for the measurement of diffuse
9. Dall’armellina E, Piechnik SK, Ferreira VM, et al. Cardiovascular myocardial fibrosis: preliminary validation in humans. Circulation.
magnetic resonance by non contrast T1 mapping allows assessment of 2010;122:138–144.
severity of injury in acute myocardial infarction. J Cardiovasc Magn 30. Jerosch-Herold M, Sheridan DC, Kushner JD, et al. Cardiac magnetic
Reson. 2012;14:15. resonance imaging of myocardial contrast uptake and blood flow in
10. Ferreira VM, Piechnik SK, Armellina ED, et al. Non-contrast T1-mapping patients affected with idiopathic or familial dilated cardiomyopathy. Am J
detects acute myocardial edema with high diagnostic accuracy: a Physiol Heart Circ Physiol. 2008;295:H1234–H1242.
comparison to T2-weighted cardiovascular magnetic resonance. J 31. Kehr E, Sono M, Chugh SS, Jerosch-Herold M. Gadolinium-enhanced
Cardiovasc Magn Reson. 2012;14:1–12. magnetic resonance imaging for detection and quantification of fibrosis
11. Ferreira VM, Piechnik SK, Dall’Armellina E, et al. T(1) mapping for the in human myocardium in vitro. Int J Cardiovasc Imaging. 2008;24:61–68.
diagnosis of acute myocarditis using CMR: comparison to T2-weighted 32. Schelbert EB, Testa SM, Meier CG, et al. Myocardial extravascular
and late gadolinium enhanced imaging. JACC Cardiovasc Imaging. extracellular volume fraction measurement by gadolinium cardiovascular
2013;6:1048–1058. magnetic resonance in humans: slow infusion versus bolus. J Cardiovasc
12. Karamitsos TD, Piechnik SK, Banypersad SM, et al. Noncontrast T1 Magn Reson. 2011;13:16.
mapping for the diagnosis of cardiac amyloidosis. JACC Cardiovasc 33. Ugander M, Oki AJ, Hsu L-Y, et al. Extracellular volume imaging by
Imaging. 2013;6:488–497. magnetic resonance imaging provides insights into overt and sub-clinical
13. Puntmann VO, D’Cruz D, Smith Z, et al. Native myocardial T1 mapping myocardial pathology. Eur Heart J. 2012;1–11:im.
by cardiovascular magnetic resonance imaging in subclinical 34. Broberg CS, Chugh SS, Conklin C, Sahn DJ, Jerosch-Herold M.
cardiomyopathy in patients with systemic lupus erythematosus. Circ Quantification of diffuse myocardial fibrosis and its association with
Cardiovasc Imaging. 2013;6:295–301. myocardial dysfunction in congenital heart disease. Circ Cardiovasc
14. Ertel A, Pratt D, Kellman P, et al. Increased myocardial extracellular Imaging. 2010;3:727–734.
volume in active idiopathic systemic capillary leak syndrome. J Cardiovasc 35. Iles L, Pfluger H, Phrommintikul A, et al. Evaluation of diffuse
Magn Reson. 2015;17:76. myocardial fibrosis in heart failure with cardiac magnetic resonance
15. Sado DM, White SK, Piechnik SK, et al. Identification and assessment of contrast-enhanced T1 mapping. J Am Coll Cardiol. 2008;52:1574–1580.
Anderson-Fabry disease by cardiovascular magnetic resonance 36. Flacke SJ, Fischer SE, Lorenz CH. Cardiac imaging abbreviations:
noncontrast myocardial T1 mapping. Circ Cardiovasc Imaging. gadopentetate dimeglumine partition coefficient in human myocardium
2013;6:392–398. in vivo: normal distribution and elevation in acute and chronic infarction
16. Pedersen SF, Thrysøe SA, Robich MP, et al. Assessment of intramyocardial 1. Radiology. 2001;218:703–710.
hemorrhage by T1-weighted cardiovascular magnetic resonance in 37. Messroghli DR, Radjenovic A, Kozerke S, Higgins DM, Sivananthan MU,
reperfused acute myocardial infarction. J Cardiovasc Magn Reson. Ridgway JP. Modified Look-Locker inversion recovery (MOLLI) for
2012;1–8. high-resolution T1 mapping of the heart. Magn Reson Med. 2004;52:
17. Feng Y, He T, Carpenter J-P, et al. In vivo comparison of myocardial T1 141–146.
with T2 and T2* in thalassaemia major. J Magn Reson Imaging. 38. Piechnik SK, Ferreira VM, Dall’Armellina E, et al. Shortened modified
2013;38:588–593. Look-Locker inversion recovery (ShMOLLI) for clinical myocardial
18. Thavendiranathan P, Walls M, Giri S, et al. Improved detection of T1-mapping at 1.5 and 3 T within a 9 heartbeat breathhold. J Cardiovasc
myocardial involvement in acute inflammatory cardiomyopathies using Magn Reson. 2010;12:69.
T2 mapping. Circ Cardiovasc Imaging. 2012;5:102–110. 39. Kellman P, Wilson JR, Xue H, Ugander M, Arai AE. Extracellular volume
19. Verhaert D, Thavendiranathan P, Giri S, et al. Direct T2 quantification of fraction mapping in the myocardium, part 1: evaluation of an automated
myocardial edema in acute ischemic injury. JACC Cardiovasc Imaging. method. J Cardiovasc Magn Reson. 2012;14:63.
2011;4:269–278. 40. Xue H, Shah S, Greiser A, et al. Motion correction for myocardial T1
20. Giri S, Chung Y-C, Merchant A, et al. T2 quantification for improved mapping using image registration with synthetic image estimation. Magn
detection of myocardial edema. J Cardiovasc Magn Reson. 2009;11:56. Reson Med. 2012;67:1644–1655.
26.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

41. Xue H, Greiser A, Zuehlsdorff S, et al. Phase-sensitive inversion recovery without hematocrit sampling. JACC Cardiovasc Imaging. 2016;9:
for myocardial T1 mapping with motion correction and parametric 54–63.
fitting. Magn Reson Med. 2013;69:1408–1420. 63. Robson MD, Piechnik SK, Tunnicliffe EM, Neubauer S. T1 measurements
42. Messroghli DR, Greiser A, Fröhlich M, Dietz R, Schulz-Menger J. in the human myocardium: the effects of magnetization transfer on the
Optimization and validation of a fully-integrated pulse sequence for SASHA and MOLLI sequences. Magn Reson Med. 2013;670:664–670.
modified look-locker inversion-recovery (MOLLI) T1 mapping of the 64. Kellman P, Bandettini WP, Mancini C, Hammer-Hansen S, Hansen MS,
heart. J Magn Reson Imaging. 2007;26:1081–1086. Arai AE. Characterization of myocardial T1-mapping bias caused by
43. Gai ND, Stehning C, Nacif M, Bluemke DA. Modified Look-Locker T(1) intramyocardial fat in inversion recovery and saturation recovery
evaluation using Bloch simulations: human and phantom validation. techniques. J Cardiovasc Magn Reson. 2015;17:33.
Magn Reson Med. 2013;69:329–336. 65. Abdel-Aty H, Zagrosek A, Schulz-Menger J, et al. Delayed enhancement
44. Salerno M, Janardhanan R, Jiji RS, et al. Comparison of methods for and T2-weighted cardiovascular magnetic resonance imaging differentiate
determining the partition coefficient of gadolinium in the myocardium acute from chronic myocardial infarction. Circulation. 2004;109:
using T1 mapping. J Magn Reson Imaging. 2013;38(1):217–224. 2411–2416.
45. Kellman P, Hansen MS. T1-mapping in the heart: accuracy and precision. 66. Bohnen S, Radunski UK, Lund GK, et al. Performance of t1 and t2
J Cardiovasc Magn Reson. 2014;16:2. mapping cardiovascular magnetic resonance to detect active myocarditis
46. Higgins DM, Ridgway JP, Radjenovic A, Sivananthan UM, Smith MA. T1 in patients with recent-onset heart failure. Circ Cardiovasc Imaging.
measurement using a short acquisition period for quantitative cardiac 2015;8.
applications. Med Phys. 2005;32:1738. 67. Giri S, Shah S, Xue H, et al. Myocardial T2 mapping with respiratory
47. Chow K, Flewitt JA, Green JD, Pagano JJ, Friedrich MG, Thompson RB. navigator and automatic nonrigid motion correction. Magn Reson Med.
Saturation recovery single-shot acquisition (SASHA) for myocardial T(1) 2012;68:1570–1578.
mapping. Magn Reson Med. 2014;71:2082–2095. 68. Baeßler B, Schaarschmidt F, Stehning C, Schnackenburg B, Maintz D,
48. Weingärtner S, Akçakaya M, Basha T, et al. Combined saturation/ Bunck AC. A systematic evaluation of three different cardiac T2-mapping
inversion recovery sequences for improved evaluation of scar and diffuse sequences at 1.5 and 3T in healthy volunteers. Eur J Radiol.
fibrosis in patients with arrhythmia or heart rate variability. Magn Reson 2015;84:2161–2170.
Med. 2014;71:1024–1034. 69. He T, Gatehouse PD, Anderson LJ, et al. Development of a novel
49. Mehta BB, Chen X, Bilchick KC, Salerno M, Epstein FH. Accelerated and optimized breathhold technique for myocardial T2 measurement in
navigator-gated look-locker imaging for cardiac T1 estimation (ANGIE): thalassemia. J Magn Reson Imaging. 2006;24:580–585.
development and application to T1 mapping of the right ventricle. Magn 70. Krumm P, Martirosian P, Rath D, et al. Signal decay mapping of
Reson Med. 2015;73:150–160. myocardial edema using dual-contrast fast spin-echo MRI. J Magn Reson
50. Weingärtner S, Roujol S, Akçakaya M, Basha TA, Nezafat R. Free- Imaging. 2015;1–8.
breathing multislice native myocardial T1 mapping using the slice- 71. Baeßler B, Schaarschmidt F, Stehning C, Schnackenburg B, Maintz D,
interleaved T1 (STONE) sequence. Magn Reson Med. 2014;74(1): Bunck AC. Cardiac T2-mapping using a fast gradient echo spin echo
115–124. sequence—first in vitro and in vivo experience. J Cardiovasc Magn Reson.
51. Kellman P, Xue H, Chow K, Spottiswoode BS, Arai AE, Thompson RB. 2015;17:67.
Optimized saturation recovery protocols for T1-mapping in the heart: 72. Fernández-Jiménez R, Sánchez-González J, Aguero J, et al. Fast T2
influence of sampling strategies on precision. J Cardiovasc Magn Reson. gradient-spin-echo (T2-GraSE) mapping for myocardial edema
2014;16:55. quantification: first in vivo validation in a porcine model of ischemia/
52. Akçakaya M, Weingärtner S, Roujol S, Nezafat R. On the selection of reperfusion. J Cardiovasc Magn Reson. 2015;17:92.
sampling points for myocardial T1 mapping. Magn Reson Med. 73. Ding H, Fernandez-de-Manuel L, Schär M, et al. Three-dimensional
2015;73:1741–1753. whole-heart T2 mapping at 3T. Magn Reson Med. 2015;74:803–816.
53. Chow K, Spottiswoode BS, Pagano JJ, Thompson RB. Improved precision 74. Brittain JH, Hu BS, Wright GA, Meyer CH, Macovski A, Nishimura DG.
in SASHA T1 mapping with a variable flip angle readout. J Cardiovasc Coronary angiography with magnetization-prepared T2 contrast. Magn
Magn Reson. 2014;16(suppl 1):M9. Reson Med. 1995;33:689–696.
54. Stainsby JA, Slavin GS. Comparing the accuracy and precision of 75. Jenista ER, Rehwald WG, Chen E-L, et al. Motion and flow insensitive
SMART1Map, SASHA and MOLLI. J Cardiovasc Magn Reson. adiabatic T2-preparation module for cardiac MR imaging at 3 Tesla.
2014;16(suppl 1):P11. Magn Reson Med. 2013;70:1360–1368.
55. Deichmann R, Haase A. Quantification of T1 values by SNAPSHOT- 76. Nezafat R, Stuber M, Ouwerkerk R, Gharib AM, Desai MY, Pettigrew RI.
FLASH NMR Imaging. J Magn Reson. 1992;612:608–612. B1-insensitive T2 preparation for improved coronary magnetic resonance
56. Schmitt P, Griswold MA, Jakob PM, et al. Inversion recovery TrueFISP: angiography at 3 T. Magn Reson Med. 2006;55:858–864.
quantification of T(1), T(2), and spin density. Magn Reson Med. 77. Oshio K, Feinberg A. GRASE (gradient- and spin-echo) imaging: a novel
2004;51:661–667. fast MRI technique. Magn Reson Med. 1991;20:344–349.
57. Kellman P, Arai AE, Xue H. T1 and extracellular volume mapping in the 78. Oshio K, Feinberg DA. Single-Shot GRASE Imaging without fast
heart: estimation of error maps and the influence of noise on precision. J gradients. Magn Reson Med. 1992;26:355–360.
Cardiovasc Magn Reson. 2013;15:56. 79. Akçakaya M, Basha TA, Weingärtner S, Roujol S, Berg S, Nezafat R.
58. Kellman P, Herzka DA, Arai AE, Hansen MS. Influence of off-resonance Improved quantitative myocardial T2 mapping: impact of the fitting
in myocardial T1-mapping using SSFP based MOLLI method. J model. Magn Reson Med. 2015;74(1):93–105.
Cardiovasc Magn Reson. 2013;15:63. 80. Bano W, Feliciano H, Coristine AJ, Stuber M, van Heeswijk RB. On the
59. Song T, Stainsby JA, Ho VB, Hood MN, Slavin GS. Flexible cardiac T1 accuracy and precision of cardiac magnetic resonance T2 mapping: a
mapping using a modified Look-Locker acquisition with saturation high-resolution radial study using adiabatic T2 preparation at 3 T. Magn
recovery. Magn Reson Med. 2012;67:622–627. Reson Med. 2017;77:159–169.
60. Klein C, Schmal TR, Nekolla SG, Schnackenburg B, Fleck E, Nagel E. 81. Sandino CM, Kellman P, Arai AE, Hansen MS, Xue H. Myocardial T2*
Mechanism of late gadolinium enhancement in patients with acute mapping: influence of noise on accuracy and precision. J Cardiovasc
myocardial infarction. J Cardiovasc Magn Reson. 2007;9:653–658. Magn Reson. 2015;17:7.
61. Spottiswoode BS, Ugander M, Kellman P. Automated inline extracellular 82. Captur G, Gatehouse P, Kellman P, et al. A T1 and ECV phantom for
volume (ECV) mapping. J Cardiovasc Magn Reson. 2015;17(suppl 1):W6. global T1 mapping quality assurance: the T1 mapping and ECV
62. Treibel TA, Fontana M, Maestrini V, et al. Automatic measurement of the standardisation in CMR (T1MES) program. J Cardiovasc Magn Reson.
myocardial interstitium: synthetic extracellular volume quantification 2016;18:571–572.
3 
Cardiovascular Magnetic Resonance
Contrast Agents
Eric M. Gale and Peter Caravan

Although currently not approved by the US Food and Drug Adminis- result of endogenous transverse relaxation in tissue. With most pulse
tration (FDA) for cardiac imaging, the vast majority of cardiovascular sequences, this dominant T1-lowering effect gives rise to increases in
magnetic resonance (CMR) studies use a gadolinium-based contrast signal intensity, and thus these agents are referred to as “positive” con-
agent.1 The CMR contrast agent typically makes diseased tissue appear trast agents. The T2 agents largely increase the 1/T2 of tissue selectively
brighter (or in some cases darker) than the surrounding tissue. Car- and cause a reduction in signal intensity, and thus they are known as
diovascular applications, such as magnetic resonance angiography (MRA), “negative” contrast agents. Paramagnetic gadolinium-based contrast
functional imaging of myocardial perfusion, and viability with late agents are examples of T1 agents, whereas ferromagnetic large iron
gadolinium enhancement (LGE) imaging of fibrosis represent the bulk oxide particles are examples of T2 agents.
of CMR scans that use a contrast agent. The first magnetic resonance There are many mechanisms by which contrast agents shorten T1
(MR) approved contrast agent, gadopentetate (Gd-DTPA), appeared and T2. Considerable chemistry and biophysics can be applied to under-
in 1988, and several other compounds followed. These first contrast stand or predict these mechanisms. However, in many cases, the effect
agents were extracellular fluid (ECF) agents. Although frequently used of these mechanisms can be reduced to a single constant, called “relax-
in CMR and an essential component of CMR perfusion and LGE assess- ivity.” Fig. 3.1 shows the effect and definition of relaxivity.
ment of fibrosis, none of these agents is currently approved by the FDA Fig. 3.1A shows the effect of a typical contrast agent on the
for cardiac applications. Thus, the use of these agents in CMR is con- relaxation time of two hypothetical tissues, one with T1 = 1200 ms
sidered “off-label.” There are now also an approved hepatobiliary contrast (similar to heart muscle at 1.5 T) and one with T1 = 400 ms. At low
agent and an intravascular agent designed specifically to enhance contrast- concentration (left side of the graph), it appears that the contrast
enhanced (CE) MRA. At the preclinical stage, there are exciting advance- agent has a larger effect (change in T1) on the tissue with the longer
ments in molecular imaging agents, including compounds that detect T1. At higher contrast agent concentrations (right side of Fig. 3.1A),
pH changes, enzymatic activity, specific biomolecules such as fibrin or both tissues approach approximately the same T1. A simple way to
collagen, and magnetically labeled cells. quantify this effect is to consider the rate of relaxation, 1/T1 (some-
This chapter focuses first on the general underlying chemistry and times denoted “R1”). In most cases in medical imaging, the contrast
biophysics of contrast agents in clinical CMR. The mechanism of action agent increases the relaxation rate proportional to the amount of
of different classes of contrast agents is described, with examples drawn contrast agent:
from CMR applications. Finally, there is a brief survey of novel contrast
agents potentially useful for cardiovascular indications that are currently 1 1
= + r1 [CA] Eq. 3.1
in clinical or preclinical development. T1 T10
The vast majority of CMR agents in clinical use are small molecules
based on chelated gadolinium (Gd). The bulk of this chapter focuses where T1 is the observed T1 with contrast agent in the tissue, T10 is
on gadolinium complexes, including their chemistry, biophysics, and the T1 before addition of the contrast agent, [CA] is the concentra-
applications. Other exogenous compounds have been used to change tion of contrast agent, and r1 is the longitudinal relaxivity, often just
signal properties in MRI (e.g., iron particles, hyperpolarized nuclei), and “relaxivity.” The conventional units for r1 are mM−1s−1 (per millimolar
these will be discussed as appropriate to CMR. This chapter assumes per second, sometimes L•mol−1s−1). Thus, the slope of 1/T1 as a function
that the reader has a basic understanding of CMR vocabulary, and the of contrast agent concentration (Fig. 3.1B) shows the relaxivity, in this
reader is referred to Chapter 1 for further clarification. case, 4 mM−1s−1. Fig. 3.1B shows that the effect of the contrast agent on
the relaxation rate is independent of the initial T1 of the tissue: that
is, in terms of relaxation rate, the contrast agent has the same effect,
INTRODUCTION TO THE BIOPHYSICS OF regardless of initial T1. Transverse, or T2, relaxivity is defined in an
analogous way:
MAGNETIC RESONANCE IMAGING
All contrast agents shorten both T1 and T2 relaxation times. However, 1 1
= + r2 [CA] Eq. 3.2
it is useful to classify CMR contrast agents into two broad groups based T1 T20
on whether the substance increases the transverse relaxation rate (1/
T2) by roughly the same amount that it increases the longitudinal For all contrast agents, r2 is larger than r1. Relaxivity is dependent
relaxation rate (1/T1) or whether 1/T2 is altered to a much greater on magnetic field strength, on temperature, and in some instances
extent. The first category is referred to as “T1 agents” because, on a can depend on protein binding, pH, or even the presence of
percentage basis, these agents alter 1/T1 of tissue more than 1/T2 as a enzymes.

27
28 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

20
1200

1000 15
800

1/T1 (s−1)
T1 (ms)
10
600

400
5
200

0 0
0 1 2 3 4 0 1 2 3 4
A Contrast agent (mM) B Contrast agent (mM)
FIG. 3.1  Change in (A) longitudinal relaxation time (T1) and (B) longitudinal relaxation rate (1/T1) for typical
myocardial tissue (solid line, T1 = 1200 ms at 1.5 T) and short T1 tissue (dashed line, T1 = 400 ms).

Contrast agent behavior in vivo is seldom as simple as the pure contrast agent is becoming nonlinear, but the signal is still increasing
linear effect relaxation rate shown in Fig. 3.1. Even in the simple case with increasing contrast agent concentration.
of pure linear relaxation, the effect of the contrast agent on the CMR
image is generally nonlinear. In traditional spin echo sequences, non- Commercial Contrast Agents and Those in
linearity can be a result of T1 saturation or T2 signal loss. Once the Clinical Development
contrast agent reduces T1 < repetition time (TR)/2, increasing contrast The addition of paramagnetic materials to reduce relaxation times goes
agent concentration will have little effect on increasing the available back to the earliest days of MR. In the 1940s, Bloch and colleagues used
longitudinal magnetization because the tissue will have nearly fully ferric nitrate to enhance the relaxation rate of water.2 Exogenous contrast
recovered the magnetization before the next radiofrequency (RF) pulse. was applied to MRI in 1978 when Lauterbur and associates reported
Furthermore, because contrast agents affect both T1 and T2 relaxation, using manganese dichloride to differentiate normal from infarcted
at high enough concentration, the contrast agent will reduce T2 to the myocardium in dogs.3 Carr and colleagues reported the first use of a
order of the echo time (TE), and will then decrease MR image intensity. gadolinium complex, gadopentetate dimeglumine (Gd-DTPA; Magnev-
These effects are seen in Fig. 3.2, where signal intensity is plotted versus ist, Bayer, Berlin, Germany), in patients with brain tumors in 1984.4 By
contrast agent concentration for T1- and T2-weighted spin echo 1988, Gd-DTPA was FDA approved for clinical use.
sequences. Fig. 3.2 was generated assuming a contrast agent relaxivity
of 4 mM−1s−1, typical of most commercial ECF gadolinium agents, and Extracellular Agents
tissue relaxation times typical of myocardium at 1.5 T (T1 = 1200 ms, The most common contrast agents used clinically are ECF agents.
T2 = 50 ms). For the T1-weighted sequence (TE/TR = 15/600), Fig. Although several are approved for clinical use, as mentioned previously,
3.2A, signal intensity begins to level out at a contrast agent concentra- none is specifically FDA approved for cardiac applications. These all
tion between 0.5 and 1.0 mM. From Fig. 3.1A, this is the range at which behave in a very similar manner, and are typically referred to as “gado-
the T1 of the myocardium decreased to approximately 300 ms, or TR/2. linium” or “gado” agents. Fig. 3.3 shows the chemical structures of several
At concentrations >1 mM, the T1 effect is saturated, and the only imaging approved ECF agents. Chemically, these compounds exhibit three similar
effect of the contrast agent is to make T2 shorter and cause signal loss, features: they all contain Gd, they all contain an 8-coordinate ligand
even on this T1-weighted sequence. Signal is lost because even a binding to Gd, and they all contain a single water molecule coordination
T1-weighted sequence has a finite TE, and T2 effects can enter when site to Gd. Nomenclature for contrast agents can be confusing: there is
T2 is short enough. a generic name (e.g., gadopentetate dimeglumine), a trade name (e.g.,
The signal intensity plateau on the T2-weighted scan (TE/TR = Magnevist), and usually a chemical code name or abbreviation (e.g.,
80/3000), Fig. 3.2B, occurs at much lower contrast agent concentration. Gd-diethylenetriaminepentaacetic acid or Gd-DTPA). Any of these three
Because TR is so long, the only real effect of the contrast agent is to names may be used in the scientific literature.
reduce (rather than increase) signal intensity on this T2-weighted scan. The multidentate ligand is required for safety.5 The ligand encap-
However, this negative contrast can also be medically useful, and certain sulates the Gd, resulting in a high thermodynamic stability and kinetic
contrast agents (notably, the iron oxide particles) create negative contrast inertness with respect to metal loss. This enables the contrast agent to
exactly by providing enhanced T2 relaxation, and thus darker images be excreted intact, an important property because these contrast agents
on T2-weighted scans. tend to be much less toxic than their substituents. For example, the
Increasing the relaxivity (r1 or r2) will have the effect of pushing DTPA ligand and gadolinium chloride both have an LD50 of 0.5 mmol/
the simulated curves in Fig. 3.2A to the left, which means that peak kg in rats (LD50 = dose that causes death in 50% of the animals),
signal and subsequent signal loss will occur at lower contrast agent whereas the Gd-DTPA complex has a safety margin that is higher by
concentrations. A more linear response of signal to contrast agent can nearly a factor of 20, with an LD50 of 8 mmol/kg for the Gd-DTPA
be achieved with a fast three-dimensional (3D) spoiled gradient recalled complex.6
echo (GRE) sequence. This is seen in Fig. 3.2C, where signal intensity The Gd ion and coordinated water molecule are essential to provid-
is plotted versus contrast agent concentration using the same tissue ing contrast. The Gd (III) ion has a high magnetic moment and a rela-
relaxation times and relaxivities as in Fig. 3.2A and B for a typical fast tively slow electronic relaxation rate, properties that make it an excellent
3D spoiled GRE sequence, TE/TR/flip = 2.2/9.0/40 degrees. The short relaxer of water protons. The proximity of the coordinated water molecule
TR and very short TE ensure that signal intensity increases across the leads to efficient relaxation. The coordinated water molecule is in rapid
entire concentration range. At high concentration, the effect of the chemical exchange (106 exchanges/s) with solvating water molecules.7
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 29

0.7 This rapid exchange leads to a catalytic effect whereby the Gd complex
effectively shortens the relaxation times of the bulk solution.
0.6 The ECF agents have very similar properties, and these are sum-
marized in Table 3.1. They are all very hydrophilic complexes with
0.5 similar relaxivities and excellent safety profiles. In addition, they can
be formulated at high concentrations. On injection, these ECF agents
0.4 quickly and freely distribute to the extracellular space. Administration
of any of these agents (with rare exceptions)8 yields similar diagnostic
0.3 information.
There are some differences among the physical properties. The
0.2 diamide complexes gadodiamide and gadoversetamide have consider-
Spin echo ably lower thermodynamic stability (log K ~ 17 vs. log K > 21 for other
0.1 TR/TE/flip = 600/15/90o Gd complexes).9,10 The nonionic (neutral) compounds gadodiamide,
gadoteridol, gadoversetamide, and gadobutrol were designed to minimize
0 the osmolality of the formulation. This was prompted by the distinct
0 1 2 3 4 reduction in toxicity and side effects brought on by the development
A Contrast agent (mM) of nonionic x-ray contrast media. However, for CMR, the injection
0.7 volumes are much smaller than the volumes used for x-ray angiography
or computed tomography (CT) angiography. Thus the overall increase
0.6 Spin echo in osmolality after injection of a CMR contrast agent is minimal. Unlike
TR/TE/flip = 3000/80/90o with x-ray contrast, there is no documented safety benefit in using
0.5 nonionic CMR contrast agents. One benefit of the nonionic compounds
is the ability to formulate them at high concentration (1 M)11 without
0.4 drastically increasing osmolality or viscosity (see Table 3.1). These high-
concentration formulations may be useful in fast dynamic studies, such
0.3 as dynamic MRA or myocardial perfusion. These ECF agents, as with
iodinated preparations used for x-ray and CT, are excreted by the kidneys.
0.2 As a result, clearance is impaired in patients with impaired/reduced
renal function (discussed later).
0.1
Blood Pool Agents
0 Because CMR contrast agents are administered intravenously, they are
0 1 2 3 4 all potentially capable of imaging the blood lumen. The ECF agents
B Contrast agent (mM) described earlier are used routinely for CE-MRA (see Chapters 25, 26,
44, 45, and 46). One potential drawback of using ECF agents for MRA
0.7 is their pharmacokinetics. ECF agents rapidly leak out of the vascular
space into all the interstitial spaces of the body. Angiography with ECF
0.6 3D GRE
TR/TE/flip = 9.0/2.2/40o agents is thus typically limited to dynamic/first-pass arterial studies.
There has been considerable effort made toward designing specific blood
0.5
pool agents that would be tailored for vascular imaging. The ideal blood
pool agent should remain in the vascular compartment and not leak
0.4
out into the extracellular space. It should be capable of being given as
a bolus such that a dynamic arterial image can be obtained. At the same
0.3
time, it should have sufficient relaxivity and blood half-life to allow
high-resolution steady-state images to be obtained. MS-325 (gadofos-
0.2
veset, initially marketed as Vasovist; and more recently as Ablavar; in
the United States by Lantheus Medical Imaging, Billerica, MA; Fig. 3.4)
0.1
is approved, and there are several other blood pool agents that have
reached various stages of clinical development. However, as this chapter
0
was being written, Lantheus announced that it would no longer produce
0 1 2 3 4
gadofosveset. Three approaches have been taken to design blood pool
C Contrast agent (mM) agents: protein binding, increased size, and ultrasmall iron oxide par-
FIG. 3.2  Effect of contrast agent on myocardial image intensity on ticles. These are discussed later.
T1-weighted and T2-weighted spin echo scans. (A) T1-weighted spin MS-325 (gadofosveset)12,13 is a Gd-based compound that binds revers-
echo (repetition time [TR] = 600 ms) assumes a patient with 100 beats ibly to serum albumin. Albumin is the most abundant protein in plasma,
per minute heart rate and shows a linear increase of signal only for and its concentration is high enough (600–700 µM) to reversibly bind
contrast agent concentration <0.5 mM. (B) T2-weighted spin echo images
most of the MS-325 after injection. Reversible albumin binding serves
(TR = 3000 ms) show only T2 signal loss effects as a result of contrast
agent with no T1 enhancement because of the long TR. (C) Typical
four purposes: (1) the albumin slows the leakage of the contrast agent
short-TR fast spoiled gradient recalled echo (GRE) sequence. The very out of the intravascular space; (2) the reversible binding still allows a
short echo time (TE) and short TR give monotonically increased image path for excretion—the unbound fraction can be filtered through the
intensity across the entire range of contrast agent concentrations typi- kidneys or taken up by hepatocytes; (3) the bound fraction is “hidden”
cally found in clinical scans. from the liver and kidneys, leading to an extended plasma half-life; and
30 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

N N
N N N N
O O NH NH
Gd O Gd O
O O O O
O O O O O O
O O O O
O O
H H H H
Gd-DTPA (Magnevist) Gd-DTPA-BMA (Omniscan)
Gadopentetate Gadodiamide

O
OH
O
OH
N N OH
Gd OH2 N
O N N
N N NH NH
Gd O
O O O
O O O O O O
O O
O
O H H
Gd-DO3A-butrol (Gadovist) Gd-DTPA-BMEA (OptiMARK)
Gadobutrol Gadoversetamide

O O
O O O
N N O N N OH
Gd Gd
OH2 OH2
O N N O N N
O O O O
O O
Gd-DOTA (Dotarem) Gd-HPDO3A (ProHance)
Gadoterate Gadoteridol
FIG. 3.3  Chemical structures of commercial (United States or Europe) extracellular fluid contrast agents.

TABLE 3.1  Approved (December 2016) Magnetic Resonance Imaging Contrast Agents:
Relaxivity,150 Osmolality, and Viscosity
Chemical r1, 0.47 T r2, 0.47 T Osmolalitya Viscositya
Generic Name Product Name Abbreviation 40°C 40°C (Osmol/kg) (cP)
Gadopentetate Magnevist (Bayer Gd-DTPA 3.4 4.0 1.96151 2.9151
HealthCare
Pharmaceuticals)
Gadoterate Dotarem (Guerbet Gd-DOTA 3.4 4.1 1.35152 2.0152
Group) (4.02)152
Gadodiamide Omniscan (GE Gd-DTPA-BMA 3.5 3.8 0.79151 3.9152
Healthcare) (1.90)152
Gadoteridol ProHance (Bracco Gd-HPDO3A 3.1 3.7 0.63152 1.3152
Diagnostics, Inc.) 1.91152 (3.9)152
Gadobutrol Gadovist (Bayer Gd-DO3A-butrol 3.7 5.1 0.57153 1.4153
HealthCare, Inc.) (1.39)151 (3.7)151
Gadoversetamide OptiMARK GD-DTPA-BMEA 4.2 5.2 1.11154 2.0154
(Mallinckrodt, Inc.)
Gadobenate Multihance (Bracco Gd-BOPTA 4.2 4.8 1.10155 5.3mPas155
Diagnostics, Inc.)
Gadofosveset Ablavar (Lantheus MS-325 5.8 6.7 0.83156 3.0156
Medical, Inc.)
Gadoxetic acid Eovist (Bayer Gd-EOB-DTPA 5.3 6.2 0.69157 1.2157
HealthCare
Pharmaceuticals)
a
All concentrations 0.5 M except those in parentheses, which are 1 M.
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 31

(4) the relaxivity of the contrast agent is increased 4-fold to 10-fold albumin.5 The large size of these compounds restricted diffusion out
on binding to albumin (discussed later). Gd-BOPTA (gadobenate; see of the vascular space and led to very good vascular imaging properties.
Fig. 3.4) has weak affinity14,15 for albumin (~10% bound), which leads However, these compounds cleared very slowly in preclinical studies
to a modest increase in relaxivity relative to the other ECF agents. and there were concerns about a potential immunologic response. An
The binding and relaxivity features of Gd-based blood pool agents alternative approach is to use a coated iron oxide nanoparticle. So-called
are listed in Table 3.2. Because binding affinity is moderate to weak for ultrasmall superparamagnetic particles of iron oxide (USPIO) are very
these compounds, the fraction bound to albumin will depend on the large (10–30 nm) but still small enough to evade substantial capture by
concentrations of albumin and the contrast agent.13,16 Immediately after the reticuloendothelial system.18 Such particles also have strong r1 relaxiv-
injection, when the concentration of the contrast agent is high relative ity, especially at lower field strengths. Unlike the Gd-based compounds,
to albumin, there will be a greater free fraction. As the concentration which will extravasate to some extent, these iron oxide particles are true
of the contrast agent begins to stabilize (at ~0.5 mM) the fraction bound intravascular blood pool agents. Currently there are no USPIO commer-
will become constant. The observed relaxivity will depend on the frac- cially available that are approved for MRI, but ferumoxytol (Feraheme)
tion bound; unlike ECF agents, the plasma T1 change is not linearly is a carboxymethyldextran-coated USPIO that is approved as an intra-
related to contrast agent concentration.16,17 venous iron replacement therapy.19 Ferumoxytol has been used off-label
Early work on blood pool compounds involved Gd covalently linked as a blood pool agent and has seen some use in pediatric CMR owing
to macromolecules, such as polylysine, dextran, or modified bovine serum to the long circulating blood half-life which facilitates high-resolution
imaging and obviates the need for timing of boluses.20,21 Ferumoxytol
can also be used to image monocytes and macrophages on delayed phase
imaging (next day) as accumulation in the reticuloendothelial system is
the ultimate fate of these particles.22,23 The iron oxide particles are not
excreted; the iron is eventually resorbed into the body.
The Guerbet company is developing a new extracellular fluid agent
O with high relaxivity. The compound P03277 is reported to have relaxivi-
N ties of 12.8 mM−1s−1 at 1.5 T and 11.6 mM−1s−1 at 3 T.24 This Gd chelate
N N
O O contains two water molecules bound to the metal ion, which leads to
Gd O
O O the high observed relaxivity. P03277 is a low molecular weight (970
O O O
O O Da) contrast agent that distributes in the extracellular space similarly
O
H H to other ECF agents and has blood elimination kinetics comparable to
gadobutrol in animal models. At the time of writing (2017), P03277 is
Gd-BOPTA (MultiHance)
Gadobenate currently undergoing Phase 2 clinical trials.

RELAXIVITY
Because their effect is indirect, CMR contrast agents differ from other
diagnostic imaging agents. Water and fat are imaged, and it is the action
of the contrast agent on the relaxation properties of the water hydrogen
nuclei that generates contrast; in x-ray contrast media and nuclear
O O
imaging agents, the effect observed is more direct. Because water is
O P
O present at a very high concentration (55,000 mM) and the contrast
agent is typically at a much lower concentration (0.1–1 mM), the con-
N trast agent must act catalytically to relax the water protons for there to
N N
O O be a measurable effect. Relaxivity, r1 and r2, thus describes this catalytic
Gd O
O O efficiency. Some compounds are better magnetic catalysts than others
O O O
O O (have higher relaxivity). Moreover, relaxivity is dependent on the external
O
H H magnetic field, B0. This section explains these differences and the molecu-
MS-325 (Vasovist; Ablavar) lar basis for them. For discrete ions, such as Gd(III) and manganese
Gadofosveset (Mn[II]), the factors influencing relaxivity are the same; for iron oxide
FIG. 3.4  Chemical structures of other commercial contrast agents with particles, the relaxation mechanism is different and will be treated
weak (Gd-BOPTA) or strong (MS-325) serum albumin binding. separately.

TABLE 3.2  Albumin Binding and Observed Relaxivities (20 MHz) of MS-325, B22956,
Gd-BOPTA, Gadomer-17, at 37°C
MS-325 Gadofosveset Gd-BOPTA Gadobenate Gadomer-17a
Agent type Strong protein binding Weak protein binding Increased size
r1 buffer (mM−1s−1) 6.612 4.4158 16.5150
r1 plasma (mM−1s−1) 5012 9.7159 19.0150
% bound plasma 9112
a
Relaxivity per gadolinium.
32 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Relaxivity can be factored into inner- and outer-sphere terms. “Inner imaging fields is much slower. For example, at 1.5 T, the Larmor fre-
sphere” quency is approximately 65 MHz, so relaxation is not as efficient as it
could be. Larger molecules, such as proteins, tumble much more slowly.
∆(1 T1) IS OS When contrast agents are made to tumble more slowly, relaxivity is
r1 = = r1 + r1 Eq. 3.3
[Gd] increased. Newer contrast agents take advantage of this phenomenon.
Lauffer25 pointed out that if small contrast agents could be made to
refers to the relaxation enhancement because of the exchangeable waters bind noncovalently to protein targets, then their relaxivity would be
in the inner sphere. “Outer sphere” refers to relaxivity resulting from increased on binding because the contrast agent would take on the
water in the second and outer-coordination spheres. This separation rotational characteristics of the protein (receptor-induced magnetiza-
is often used because the inner-sphere component is easier to understand tion enhancement). MS-325 is an example of a contrast agent designed
from a theoretical framework and the effect of changing specific molecular to exploit the receptor-induced magnetization enhancement effect.13
parameters on relaxivity can be tested. For ECF agents, the inner-sphere MS-325 targets the blood protein serum albumin. In the absence of
and outer-sphere contribution to relaxivity is about the same, whereas albumin, the relaxivity of MS-325 is approximately 50% greater than
for newer, high-relaxivity contrast agents, the inner-sphere component that of Gd-DTPA, but in the presence of albumin, the relaxivity is
accounts for the majority of overall relaxivity. approximately 600% greater than that of Gd-DTPA at 1.5 T. This is
seen later in Fig. 3.6, where the magnetic field dependence on relaxivity
q [H2O] is plotted for MS-325 and Gd-DTPA in either serum albumin solution
r1IS = Eq. 3.4
T1m + τ m (filled symbols) or in buffered saline.
Increasing the molecular size is another way to increase relaxivity.
Inner-sphere relaxivity is given by Eq. 3.4, where q represents the Gadomer is a dendrimer that was formerly in clinical development and
number of water molecules bound to the metal ion (typically q = 1), contains 24 Gd with a molecular weight of 17,500 (Fig. 3.5). The increased
[H2O] is the water concentration in millimolars, T1m is the relaxation size results in slower motion and higher relaxivity. However, the rota-
time of the water that is bound to the metal ion, and τm is the lifetime tional effect is defined by more than just molecular weight. Linear
of the water molecule in the inner sphere. Eq. 3.4 shows that the more polymers containing Gd have lower relaxivities26,27 than may first be
sites available for water to bind (q), the more efficient the process. expected because of fast rotation around one axis of the molecule.
Likewise, the T1 of the bound water (T1m) should be very short. This Electrons undergoing relaxation in a paramagnetic ion also generate
is indicative of how effectively the metal ion can relax the coordinated a fluctuating field that can cause nuclear relaxation. It is the faster of
water. The lifetime τm of the bound water is the inverse of the water the two processes (rotation or electronic relaxation) that defines how
exchange rate (kex = 1/τm). If turnover is slow (τm is long), then it will efficiently the hydrogen is relaxed. For Gd and Mn complexes, electronic
not matter how efficiently the bound water is relaxed if this relaxation relaxation depends on the applied field and it gets slower as the magnetic
cannot be transmitted to the bulk water. field is increased. At low fields (<0.5 T), electronic relaxation governs
Paramagnetic relaxation (1/T1m) occurs via a dipolar mechanism. hydrogen relaxation, whereas at higher fields, rotational diffusion is the
At a magnetic field encountered in medical imaging, 1/T1m is shown in dominant mechanism. Gd3+ and Mn2+ have symmetrical electronic
Eq. 3.5, where C is a constant,5 configurations (half-filled f and d shells, respectively), and as a result,
electronic relaxation is relatively slow. Other ions, such as dysprosium
1 S (S + 1)  3τ c  (Dy3+), have a higher magnetic moment, but are rather poor relaxors
=C 6 Eq. 3.5
T1m r MH 1+ ωH2τ c2  because electronic relaxation is so fast.
Eq. 3.5 also indicates that at higher magnetic fields, the T1 relaxation
S is the spin quantum number, rMH is the metal ion-to-proton distance, mechanism becomes less efficient as ωH2τc2 > 1. The magnetic field at
τc is the correlation time, and ωH is the hydrogen Larmor frequency. which this condition is met is dependent on the magnitude of the cor-
The product S(S+1) is proportional to the magnetic moment. All other relation time. This is seen in Fig. 3.6, where the relaxivity of MS-325
factors being equal, the higher the magnetic moment, the more efficient in serum albumin solution begins to decrease after a peak at approxi-
the relaxation. This is why Gd3+ (S = 7/2) is preferred to copper (Cu2+, mately 0.7 T. Fig. 3.7 further illustrates the effect of correlation time
S = 1/2). The dipolar effect depends on the distance between the ion and field strength on relaxivity. In Fig. 3.7, r1 and r2 are simulated over
and the hydrogen nucleus, rMH, to the inverse sixth power. The inner- a range of fields encountered in CMR for correlation times of 0.1 ns
sphere water is critical; it has the shortest metal-to-hydrogen distance (typical of ECF agents), 1 ns (intermediate motion), and 10 ns (typical
of water hydrating the metal complex. of albumin-bound agents). Fig. 3.7 shows that the benefits of very slow
The correlation time, τc, is the time constant that governs the inter- rotation are seen at lower field strengths. Note also that r1 does not go
action between the electron spin of the ion and the nuclear spin of the to zero because there is also an outer-sphere component to relaxivity12
hydrogen. Depending on the ion and the field strength, the correlation and the correlation times that govern outer-sphere relaxivity are quite
time is either the time constant for rotational diffusion of the molecule, short. Transverse relaxivity is defined by equations similar to Eqs. 3.4
τR, or the electronic T1 of the metal ion, T1e. In the MR experiment, and 3.5, with the exception that there are other mechanisms causing
nuclei are excited by applying RF energy at the Larmor frequency. To r2 to increase at high fields.5,28 The r2 is always greater than r1, and for
relax these nuclei, there needs to be a resonant source for energy transfer. very slow-tumbling systems, the r2/r1 ratio becomes large at high fields.
A paramagnetic molecule tumbling in solution creates a fluctuating Electronic and nuclear relaxation is described in greater detail in various
magnetic field. Molecules tumble (undergo rotational diffusion) because reviews and books.5,28
of thermal energy; but because they also collide with each other, there Fig. 3.7 suggests that slow-tumbling T1 agents become less effective
is a distribution of rotational diffusion rates, with a mean rate charac- at high fields, but relaxation times for tissue are longer at high fields
terized by 1/τR. The closer this tumbling rate is to the Larmor frequency, and the signal-to-noise ratio (SNR) increases with increased field strength
the more efficient the relaxation by the contrast agent. (B0). These factors and the choice of sequence mean that a contrast
Small molecules, such as Gd-DTPA, tumble very fast, in the gigahertz agent with a lower relaxivity at 3 T than at 1.5 T may still provide
range (1 GHz = 1000 MHz), but the Larmor frequency for protons at greater contrast at 3 T than at 1.5 T.
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 33

LGd GdL
Lys Lys
HO2C O Lys GdL
LGd Lys
Lys Lys Lys
O
N LGd LGd GdL
H GdL = N O Lys Lys
Lys
HO Gd
OH2 LGd Lys N O Lys Lys GdL
O N N
NH Lys Lys Lys Lys Lys
O O O LGd Lys GdL GdL
Lys Lys
O
LGd O N GdL
N Lys Lys
N N HN
O O O LGd Lys N LGd O Lys GdL
Gd O Lys Lys Lys Lys Lys
O O Lys =
O O O
O O Lys Lys GdL
O N Lys
H H Lys Lys Lys Lys Lys Lys
H
B22956 GdL GdL Lys GdL GdL
(Gadocoletic acid) Lys Lys
GdL GdL
Gadomer (aka Gadomer-17)
FIG. 3.5  Chemical structures of some investigational gadolinium-based blood pool agents.

NJ] has a crystal diameter of 4.3–4.8 nm and a global particle diam-


40 eter of ~200 nm).29 USPIOs and SPIOs are small enough to form a
stable suspension and can be administered intravenously. The differ-
ence in particle size determines their pharmacokinetic behavior. There
30
are no inner-sphere water molecules in iron particles, and relaxation
r1 (mM−1 s−1)

of water arises from the water molecules diffusing near the particle.
20 However, the mechanism of outer-sphere relaxation differs from that
described earlier. One feature is that the crystals have a net magne-
tization, and as the external field is increased, this magnetization is
10
increased (as is true for Gd, but the effect is much smaller). The modu-
lation of this net magnetization can cause proton relaxation and have
0 field dependence.29
There are some generalities about relaxivity in these particles. For
0.5 1.0 1.5 2.0 2.5 3.0 the USPIOs, longitudinal relaxivity (r1) can be quite high and these can
B0 (T) function as effective T1 agents. The r2/r1 ratio for USPIOs is significantly
FIG. 3.6  Magnetic field dependence on relaxivity for MS-325 (gadofos- larger than for Gd complexes, and r2 increases with increasing magnetic
veset; circles) and gadopentetate (Gd-DTPA; squares) in either serum field. When there is aggregation of crystals, which is the case in SPIOs,
albumin solution (filled symbols) or buffered saline (open symbols) at r1 tends to decrease and r2 increases. Thus, for the particles themselves
37°C. as well as for aggregates of particles, the ratio of r2/r1 typically increases
as the size of the particles or aggregates increases, although the T2
relaxivity as a function of particle size can be quite complicated. The
The iron oxide-based contrast agents are not discrete molecules, effect of aggregation of crystals is that the aggregate itself can be con-
but crystals of iron oxide (Fe3O4) surrounded by a coating. The indi- sidered a large magnetized sphere whose magnetic moment increases
vidual spins of each iron cooperatively via quantum mechanical inter- with increasing field strength. This gives rise to susceptibility effects
actions build up to give the crystal a very large total spin, and thus and the SPIOs can act as T2* relaxation agents. This has important
relaxivity will be a function of the number of spins. Such a material is consequences when considering the effects of contrast agent compart-
called “ferromagnetic,” and its magnetism persists outside the external mentalization on imaging (discussed later).
magnetic field. A weaker form of this is superparamagnetism, in which Table 3.3 gives r1 and r2 values in plasma at 1.5 T and 3 T for a range
small particles of iron oxides with aligned spins occur in a magnetic of contrast media. The ECF agents have slightly lower r1 at 3 T and low
field. Because the particles are small (submicron), the magnetic sus- r2/r1 ratios. The weak albumin binders have higher relaxivities, and the
ceptibility effect is smaller than for large crystals of ferrites. Superpara- slow-tumbling Gd compounds, such as gadomer or albumin-bound
magnets are no longer magnetic outside of the external field. MS-325, have several-fold higher relaxivities than the ECF agents. As
The iron oxide particles consist of a core of one or more magnetic Fig. 3.7 suggests, the slow-tumbling compounds also have significantly
crystals of Fe3O4 embedded in a coating. Because these are materi- lower r1 at 3 T than at 1.5 T and the r2/r1 ratio is increased at 3 T. Three
als rather than discrete molecular entities, there is a distribution of iron particle formulations are listed. The SPIOs Feridex and Resovist
sizes. USPIOs have a single crystal core and a submicron diameter. (Bayer Healthcare, Berlin, Germany) have a large r2 and a very large
SPIOs have cores containing more than one crystal of Fe3O4 and are r2/r1 ratio, and this ratio is increased at 3.0 T. The USPIO SHU555C
larger than USPIOs, but are still submicron in size (e.g., ferumoxide (Bayer Healthcare, Berlin, Germany) has good T1 relaxivity at 1.5 T,
[Endorem, Guerbet, Roissy, France or Feridex, Bayer Healthcare, Wayne, but at 3 T, the transverse relaxivity dominates.
34 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

100 100
90 90
80 80
70 70
60 60
50 50
40 40
30 30
20 20
10 10
0 0
0 0.5 1 1.5 2 2.5 3 0 0.5 1 1.5 2 2.5 3
A B0 (T) B B0 (T)
FIG. 3.7  Effect of rotational correlation time on longitudinal (r1, 7a) and transverse (r2, 7b) relaxivities as a
function of field strength. Long correlation time (τR = 10 ns, solid line), typical of albumin binding, gives high
r1 that decreases with increasing field and high r2; intermediate correlation time (τR = 1 ns, short dashed line)
shows relaxivity maximum pushed out to a higher field; short correlation time (τR = 0.1 ns, long dashed line),
typical of extracellular fluid agents, shows low, roughly field-independent r1, r2. Simulations with other param-
eters typical of gadolinium-based agents.12

TABLE 3.3 Relaxivities150 of Selected Contrast Media (0.25 mM) in Plasma at 1.5 T and 3.0 T
at 37°C
COMPOUND 1.5 T 3 T
−1 −1 −1 −1 −1 −1
Chemical/Code Generic Name r1 (mM s ) r2 (mM s ) r1 (mM s ) r2 (mM−1s−1)
Gd-DTPA Gadopentetate 4.1 4.6 3.7 5.2
Gd-DTPA-BMA Gadodiamide 4.3 5.2 4.0 5.6
Gd-HPDO3A Gadoteridol 4.1 5.0 3.7 5.7
Gd-BOPTA Gadobenate 6.3 8.7 5.2 11.0
MS-325a Gadofosveset 27.7 72.6 9.9 73
a
Data from Eldredge HB, Spiller M, Chasse JM, et al. Species dependence on plasma protein binding and relaxivity of the gadolinium-based MRI
contrast agent MS-325. Invest Radiol. 2006;41:229–243.

effects on a CMR pulse sequence with a single tissue relaxivity can be


CONTRAST-ENHANCED TISSUE RELAXATION
misleading. Care must be taken when trying to estimate the concentra-
The extent to which a metal complex influences tissue relaxation rates tion of a contrast agent from signal intensity changes.
depends on three factors. The actual relaxivity within a compartment might be affected by
1. The chemical environment encountered by the metal complex. the biologic milieu. For some compounds with strong or weak albumin
Binding of the agent to macromolecules can cause significant relax- binding (e.g., MS-325 [gadofosveset], Gd-BOPTA [gadobenate]), local
ivity enhancement. An example of this is shown in Fig. 3.6, comparing variation in the albumin concentration will affect the amount of contrast
the relaxivity of MS-325 in buffer solution and in serum albumin agent bound to albumin and thus affect the overall relaxivity. For example,
solution. in the plasma space, albumin concentration is typically high (0.6 to
2. Compartmentalization of the metal complex in tissue. Generally, 0.7 mM) compared with the extracellular space in the normal heart
tissue water is compartmentalized into intravascular, interstitial (fluid (0.2 to 0.3 mM).
space between cells and capillaries), and intracellular space, consti- The ECF agents interact very weakly if at all with plasma proteins
tuting roughly 5%, 15%, and 80% of total water, respectively. Cellular or membrane structures. Tweedle, Wedeking, and colleagues showed
organelles further subdivide the intracellular component. If water that the relaxivity in blood and soft tissue of Gd-DTPA (gadopentetate)
exchange between any of these compartments is slow relative to the and Gd-DOTA (gadoterate) is the same as the relaxivity in aqueous
relaxation rate in the compartment with the longest T1, multiexpo- solution.30,31 In extreme settings, the actual relaxivity could vary.32,33
nential relaxation may result. This can decrease the effective tissue Except for pathologic situations, most CMR contrast agents in use today
relaxivity of an agent because not all of the tissue water is encoun- are excluded from intact cells. Thus, the contrast agent will be localized
tering the paramagnetic center. to the extracellular space. As a result, the simple relaxivity equations
3. The magnetic susceptibility of the contrast agent. The contrast agent do not necessarily hold. For a Gd-based ECF agent in a test tube, it
causes a microscopic field inhomogeneity on a biologic scale of 10 takes approximately 3 µs for water to diffuse between Gd molecules34;
to 1000 nm rather than on the chemical scale of 0.1 to 1 nm. This in the time of a typical imaging TR, a given water molecule may interact
results in a reduction in apparent T2. with thousands or millions of Gd molecules, and all water molecules
The result of the first two effects is that the simple relaxivity equation will interact with approximately the same number of Gd ions. However,
(Eq. 3.1) often is not valid in a biologic setting; likewise, describing the if that same ECF agent is compartmentalized solely within the cardiac
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 35

microvasculature, it takes between 2 and 20 seconds for most of the


NOVEL CONTRAST AGENTS IN DEVELOPMENT
water in the tissue (85% of it is extravascular) to physically diffuse into
the microvasculature; most of the water in the tissue does not have the Molecular imaging has been defined as “the in vivo characterization and
opportunity to be relaxed by the Gd within TR of an imaging acquisi- measurement of biological processes at the cellular and molecular level.”41
tion, resulting in a lower signal enhancement than that predicted by Combining the high-resolution anatomic and functional imaging ability
Eq. 3.1 and assuming a uniform distribution of contrast agent throughout of CMR with the specificity of molecular targeting is very appealing,
the tissue. but somewhat limited by the inherent sensitivity limitations of CMR.
To deal with compartmentalization, the concepts of “water exchange” Examples of molecular CMR contrast agents used for cardiovascular
and “exchange time,” τ, between compartments are often used.35,36 The applications are numerous. The vast majority of these agents have only
water exchange rate and the size of the compartments will determine been studied in animal models. A few examples are described here.
the effect of the contrast agent on CMR signal. To illustrate this phe- EP-2104R and EP-1873 were Gd-based agents that target fibrin,42,43
nomenon, the two limiting causes of exchange will be described.34,35 In an abundant component of thrombus. These molecules contain a peptide
one extreme, water moves so quickly between the biologic compartments sequence specific for fibrin that has been linked to several Gd chelates.
that the net effect is as if the contrast agent were uniformly spread This class of compounds has been shown to positively enhance thrombus
throughout the entire tissue. This situation, called “fast exchange,” occurs visualization in animal models of atherosclerotic plaque rupture44 and
whenever the exchange rate, 1/τ, between the compartments is much carotid artery thrombus,45 and in embolic models of thrombus in the
faster than the difference in relaxation rates between the compartments.37 coronary arteries,46,47 left atrium,48 and lung.49 EP-2104R uptake was
This occurs in blood, where red cells have a short water exchange time, shown to correlate with fibrin content in a murine model of venous
on the order of 5 to 10 ms.38 The intact red blood cell prevents most thrombosis.50 This contrast agent has been shown to visualize thrombus
CMR contrast agents from entering the cell, but as long as the plasma in human subjects, one of the first examples of clinical translation of
T1 is longer than 20 ms, the two compartments of the blood (plasma a molecular MR probe.51
+ red cells) are in fast exchange and blood behaves for CMR purposes The Barnes Hospital–Washington University group developed a plat-
as if the contrast agent were spread uniformly through the blood. In form technology based on large particles prepared from a perfluorocarbon
this case, in general, the effective relaxation rate will be the weighted emulsion.52,53 The particles can be functionalized by adding molecules
average of the relaxation in the two compartments. That is, if for com- with lipophilic components that are noncovalently incorporated into the
partment i the volume fraction is fi, the initial T1 is T1i and the con- particle. Signal generation arises from thousands of lipophilic gadolinium
centration of agent is Ci (which could be zero), the entire tissue together chelates incorporated on the surface. Targeting vectors can similarly be
will behave as shown: incorporated on the surface to guide the particle’s distribution. This
group used antibodies to target the particles to either fibrin54–56 or tissue
1  1  factor.57 Alternatively, small molecules that bind to the αvβ3 integrin
= f1  + rC + f Eq. 3.6
T1  T11 1 1 2 were incorporated and the particle was used to detect overexpression
of this integrin that occurs during angiogenesis.58–60 Use of the αvβ3-
In “slow exchange,” the water exchange rate is much slower than targeted particle to deliver fumagillin to inhibit angiogenesis has also
the difference in relaxation rates between the compartments. In this been described while monitoring the process by molecular MRI.61
case, a single relaxation time, and thus a single relaxivity, is meaning- Naresh and colleagues used large (0.5 ± 0.25 µm diameter) Gd-
less, because the two microscopic compartments will relax with their loaded liposomes to monitor the kinetics of monocyte infiltration and
own relaxation times. Very few biologic compartments show true slow clearance in a mouse model of myocardial infarction.62 Large particles
exchange, whereas the intermediate case, when exchange is neither exhibit a short blood half-life and are rapidly accumulated within the
slow nor fast (“intermediate exchange”) occurs very commonly. With reticuloendothelial system. The biodistribution profile of the Gd-loaded
intermediate exchange, relaxation behavior appears biexponential, liposomes provides a mechanism for long-term monocyte tracking with
although both the apparent compartment size and the effective T1 positive MRI contrast.
of the two compartments will vary from their true biologic size and Imaging and characterization of atherosclerotic plaque has been an
T1. It is possible to model the signal intensity behavior as a function active area of investigation (see Chapters 26 and 27). There are some
of contrast agent concentration to estimate water exchange times in new contrast agents that have shown efficacy in animal models of ath-
vivo. Although characterizing human tissue as having only one or two erosclerosis. One interesting class of compounds is the “gadofluorines.”63–65
compartments is an oversimplification, these types of models have These amphiphilic small molecules consist of a Gd chelate linked to a
proved useful for explaining the effects of biologic water mobility on perfluorocarbon chain and a hydrophilic group, such as a sugar. Per-
contrast-enhanced scans.39 fluorocarbons do not associate with lipids and impart interesting bio-
Biologic compartmentalization also results in susceptibility contrast. distribution properties. Gadofluorine M has been shown to localize in
The contrast agent causes microscopic field inhomogeneities, sometimes plaques induced in Watanabe heritable hyperlipidemic rabbits, but not
called “mesoscopic” inhomogeneities.40 Water diffusion causes the protons in the vessel wall of normal New Zealand white rabbits.65
to dephase from one another because of the different magnetic fields The Mt. Sinai group has taken many approaches to imaging plaque
that they experience. Even in the absence of water diffusion, the field that target various plaque components. One approach was to use high-
inhomogeneity causes intravoxel dephasing and thus signal loss on density lipoprotein as a carrier for a lipophilic contrast agent.66 The
GRE images because of the different microscopic magnetic fields within high-density lipoprotein is believed to facilitate transport of the contrast
the voxel. The strength of the perturbing magnetic field is directly agent to the plaque. They used mixed micelle platforms to nonspecifi-
proportional to contrast agent concentration and its molar magnetic cally target plaques67 and also incorporated antibodies to macrophage-
susceptibility (χ). The actual magnitude and even the direction of the scavenging receptor to home the mixed micelles to macrophage in the
magnetic field shifts depend strongly on the size and the shape of the plaque.68,69 They also incorporated collagen-seeking peptides (see later)
biologic compartment in which the contrast agent resides40; the size of to image collagen content in plaques.70 In collaboration with the Guerbet
the susceptibility contrast effect depends on how the water diffuses group, they reported a Gd-based agent targeting matrix metalloproteases
through the tissue. in the plaque.71
36 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A Gd-based contrast agent specific to type I collagen was described,72 content in injury or healing cardiovascular tissue is a sufficiently abun-
and this probe was used to show in vivo molecular imaging of fibrosis dant biomarker for monitoring tissue remodeling. The elastin specific
in a mouse model of healed postinfarction myocardial scarring.73 Because magnetic resonance contrast agent Gd-ESMA targets elastin with high
the extracellular matrix of the heart is very rich in collagen, this com- affinity and specificity. Fig. 3.8 shows increased Gd-ESMA retention
pound was used to assess myocardial perfusion in a pig model of coronary and contrast enhancement in the vessel wall of an aneurysm-bearing
artery stenosis. Collagen binding served to retain the probe in the myo- artery versus a normal artery. In this study, Gd-ESMA was used to
cardial extracellular matrix for extended periods such that a delayed, visualize rupture of aortal elastic lamina and monitor the compensa-
high-resolution steady-state image of heart could be obtained that tory increase in elastin content of the dilated vessel over the course of
reflected the initial perfusion of the contrast agent.74 This probe has weeks in a mouse model of aortic abdominal aneurysm.78 Gd-ESMA
been also used to stage liver fibrosis in rodents and could be useful in has also been used to visualize the vascular remodeling process at the
tracking the progress of cardiovascular pathologies with elevated col- site of disrupted plaques in rabbit model of atherosclerosis.79 Chang-
lagen levels.75–77 ing elastin content postmyocardial infarction in both the infarcted and
An agent that targets elastin has also been developed. Elastin levels remote myocardium has serially monitored Gd-ESMA out to 3 weeks
are largely increased following injury to load-bearing tissues. Elastin in a mouse model.80 In another study, Gd-ESMA was used to quantify

Precontrast Gd-DTPA Gd-ESMA

Fusion with TOF


Sham

B C D E

TOF
Precontrast Gd-DTPA Gd-ESMA

Fusion with TOF


4 Weeks Angiotensin

G H I J

6
aorta by Gd-ESMA (mm2)

F 5
Cross-sectional area

4
TOF
3
2
1 y = 1.64x + 0.22
R2 = 0.95, P < .05
0
0 1 2 3 4 5 6
Cross-sectional area aorta by EvG (mm2)
FIG. 3.8  Magnetic resonance images (MRI) of the suprarenal abdominal aorta in sham-treated mice (A–E)
and mice bearing a pharmacologically induced abdominal aortic aneurysm (F–J). Time-of-flight (TOF) angio-
grams are shown in (A) and (F), the red lines mark the axial cross sections of the vessels depicted in images
(B–E) and (G–J). Images (B–D) and (G–I) were acquired using the same T1-weighted protocol. (B) and (G)
were acquired without contrast enhancement, (C) and (H) were contrast-enhanced using Gd-DTPA, (D) and
(I) were contrast-enhanced using the elastin-seeking MRI probe Gd-ESMA. The TOF and Gd-ESMA enhanced
imaging data are fused in (E) and (J). The aneurysm-bearing vessel is strongly enhanced compared with the
vessel in the negative control animal. An increase in elastin content accompanies the compensatory remodel-
ing of vessel wall at the site of the dilation. (K) The cross-sectional area of the aneurysm-bearing arterial wall
measured using Gd-ESMA-enhanced MRI correlates closely with cross-sectional area determined by ex vivo
histology (Elastica van Gieson staining). (Courtesy Dr. R. Botnar, King’s College, London, UK.)
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 37

A B C D 50

25

CNR
+/+
*
+/- -/-
MPO +/+ +/- -/-
FIG. 3.9  Magnetic resonance image of myeloperoxidase (MPO) secreted by infiltrating neutrophils in a
mouse model of myocardial infarction using the myeloperoxidase-sensing probe Gd-MPO. Images were
acquired 2 hours after intravenous injection of Gd-MPO. (A–C) Short-axis view of T1-weighted images of
wild-type (+/+), heterozygous (+/-), and homozygous (-/-) MPO knockout mice, respectively. The infarct in the
left ventricle wall is denoted with yellow arrows. (D) Infarct zone versus remote myocardium contrast-to-noise
ratio (CNR) correlates with MPO activity. (Courtesy Dr. D. Sosnovik, Massachusetts General Hospital/ Harvard
Medical School, Boston, USA.)

intrascar elastin out to 3 weeks following myocardial infarction in a USPIO will void the MRI signal, regardless of cell viability. A recent
mouse model.81 study using MRI and histology to track the fate of USPIO-labeled mes-
The Massachusetts General Hospital group developed a gadolinium- enchymal stem cells implanted into infarcted myocardium provides a
based agent (MPO-Gd) sensitive to myeloperoxidase (MPO) activity. cautionary example. Four weeks after implantation of the labeled cells,
Ischemic injury of the myocardium causes timed recruitment of neu- the injection site remained hypointense but histologic analysis revealed
trophils and monocytes/macrophages, which produce substantial amounts very few viable stem cells. The USPIOs had accumulated in macrophages
of local MPO. MPO forms reactive chlorinating species capable of that infiltrated the injection site.95
inflicting oxidative stress and altering protein function by covalent The Massachusetts General Hospital group has also shown that the
modification. MPO-Gd is first radicalized by MPO and then either iron oxide coating material can be chemically modified to introduce new
spontaneously oligomerizes or binds to matrix proteins, all leading to targeting vectors.96–98 They termed this contrast agent platform “cross-
enhanced spin-lattice relaxivity and delayed washout kinetics. MPO-Gd linked iron oxide (CLIO).” For example, annexin V can be grafted onto a
was used to noninvasively measure inflammatory responses after myo- cross-linked iron oxide99 to detect apoptotic cardiomyocytes in a mouse
cardial ischemia or stroke in murine models,82,83 and vascular inflam- model of transient left anterior descending artery occlusion.100 Another
mation in a murine modes of Kawasaki disease and a rabbit model of CLIO-based probe is targeted to vascular cell adhesion molecule-1 using
aneurysm.82–85 Fig. 3.9 shows differing degrees of contrast enhancement a peptide specific to this molecule.101 This probe was used to identify
at the site of myocardial infarction in wild-type, heterozygous, and inflammatory activation of cells in a mouse model of atherosclerosis.
homozygous MPO knockout mice. There is a clear correlation between The fluorine 19F signal of perfluorocarbon nanoemulsions has been
infarct enhancement and local MPO activity.82 used to image macrophage infiltration during inflammation. 19F reso-
The Massachusetts General Hospital group also developed a Gd- nances are absent from tissues and thus fluorinated molecular imaging
based contrast agent (Gd-TO) to detect extracellular DNA, which is a probes can report with a high degree of specificity. Intravenously injected
useful biomarker of acute necrosis.86 Gd-TO binds DNA with high perfluorocarbon nanoemulsions can be taken up by circulating mono-
specificity and affinity. Gd-TO was used to monitor the clearance of cytes, and monocyte accumulation at the site of inflammation results
intracellular debris from acutely necrotic cardiac tissue in a mouse in a strong, localized 19F signal. These 19F probes have been used to
model of myocardial infarction.86 visualize monocyte infiltration into the site of myocardial infarction
Manganese (Mn)-based complexes have also been explored as con- in explanted porcine hearts 4 days following the ischemic injury, and
trast agents. The Massachusetts General Hospital group is developing in vivo in a mouse model of myocarditis.102,103 Combined 19F MR spec-
an Mn-based complex agent as a Gd-free alternative.87 A lead candidate troscopy, T2-weighted MRI, T1-weighted LGE imaging, and cine imaging
agent (Mn-PyC3A) provides contrast equivalent to the commercial were applied to monitor changes in monocyte flux, edema, infarct size,
Gd-based agents. Preliminary biodistribution and clearance studies and cardiac function over a period of 4 weeks in a mouse model of
performed in a mouse model indicate >99% clearance of Mn-PyC3A myocardial infarction.104 Perfluorocarbon nanoemulsions that seek
within 24 hours by mixed renal and hepatobiliary paths. molecular targets have also been developed. For example, nanoemul-
There is a rich literature describing the use of iron oxide particles sions decorated with polyethylene glycol to avoid macrophage uptake
as targeted contrast agents. Commercial USPIOs passively accumulate and with α2-antiplasmin peptide fragments have been shown to selec-
in macrophages. This property has been exploited to provide negative tively accumulate at sites of active thrombosis.105
contrast in macrophage-rich atherosclerotic plaques. There appears to The lack of sensitivity in CMR stems from the very small degree of
be a positive correlation between macrophage uptake and plasma polarization among the nuclear spins. In a magnetic field there is a net
half-life.88,89 magnetization, but this is small, and approximately 0.0006% of the
Iron oxides have also been used to label cells, and then the cells are spins are polarized. A technique called “spin exchange” uses a high-
tracked in vivo using CMR.90,91 Visualization of mesenchymal stem cells powered laser (also called “optical pumping”) to increase the polarization
engrafted into the myocardium of a pig has been demonstrated at 1.5 by four to five orders of magnitude (hyperpolarization).106 Isotopes
T.92,93 Injection sites containing >105 cells could be detected in vivo.92 with long T1 values can be hyperpolarized and used as contrast agents.
In a mouse model of myocardial infarction, a therapeutic intervention The long T1 is necessary to maintain the contrast medium in the hyper-
of mouse embryonic stem cells could be followed by visualizing the polarized state long enough to obtain an image before the spins relax
stem cells and following their effect on left ventricular (LV) function back to the equilibrium value.
over a period of 4 weeks.94 Care must be taken in interpreting MRI Gases often have long T1 values, and isotopes of the noble gases
data to track USPIO-labeled stem cells because the local presence of helium (He-3) and xenon (Xe-129) have been used for imaging. Contrast
38 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

agents with hyperpolarized carbon C-13 have been reported using a chelator (ligand) with the metal. Stability constants for approved agents
C-13-enriched water-soluble compound107 with long relaxation times have the order Gd-DOTA (gadoterate) > Gd-HPDO3A (gadoteridol) ~
(in vitro, T1 = approx. 82 s, T2 = approx. 18 s; in vivo, T1 = approx. Gd-DO3A-butrol (gadobutrol) ~ MS-325 (gadofosveset) ~ Gd-BOPTA
38 s, T2 = approx. 1.3 s). This could be formulated at a C-13 concentra- (gadobenate) ~ Gd-DTPA (gadopentetate) > Gd-DTPA-BMA (gado-
tion of 200 mM and hyperpolarized to 15%. The authors used this diamide) ~ Gd-DTPA-BMEA (gadoversatamide).5 The compounds
material for CE-MRA in rats and swine.108 with lowest thermodynamic stability are the neutral complexes with
Hyperpolarized C-13 also offers the potential for investigating meta- acyclic chelators: Gd-DTPA-BMA (gadodiamide) and Gd-DTPA-BMEA
bolic pathways. For instance, Merritt and colleagues studied the metabo- (gadoversetamide).
lism of [1-13C]-pyruvate in a perfused rat heart.109 [1-13C]-pyruvate Stability constants describe equilibrium values, but do not indicate
metabolism has also been used to serially monitor the metabolic changes how quickly equilibrium is reached. In the biologic milieu, protons and
that accompany cardiac dysfunction in a rat model of myocardial infarc- other metal ions compete to bind the chelator ligand, whereas there
tion.110 Another study using rats temporally tracked [1-13C]-pyruvate are ions such as phosphate and carbonate that have a high affinity for
metabolism across the entire myocardium throughout and following Gd. Under conditions that favor dissociation of the Gd (e.g., low pH),
the course of ischemia/ reperfusion.111 Imaging frameworks have also two complexes with comparable stability may have very different rates
been optimized for in vivo cardiac imaging of hyperpolarized 13C- labeled of dissociation. For example, the macrocyclic complex Gd-HPDO3A
metabolites such as acetate and acetylcarnitine.112 (gadoteridol) is an example of a compound more kinetically inert to
A major benefit of hyperpolarized contrast media is the excellent Gd loss than Gd-DTPA (gadopentetate), which has a similar stability
sensitivity with no background (high SNR). Challenges include the constant. The rate of acid-assisted Gd dissociation is 20 times faster
distribution and availability of the hyperpolarization equipment and for Gd-DTPA (gadopentetate) than for Gd-HPDO3A (gadoteridol).134
imaging hardware compatibility for imaging nonhydrogen nuclei (not Laurent and colleagues measured the rate of Gd loss in the presence
available on all clinical scanners). However, there is now a commercial of zinc and phosphate under a standard set of conditions for various
polarizer available. approved agents.135,136 The macrocyclic complexes Gd-DOTA (gadoter-
ate), Gd-HPDO3A (gadoteridol), and Gd-DO3A-butrol (gadobutrol)
were the most inert with respect to Gd loss. The compound that released
SAFETY Gd the most rapidly was Gd-DTPA-BMA (gadodiamide). These differ-
The safety of CMR contrast agents, which themselves offer no direct ences in kinetics and thermodynamic stabilities may well translate into
therapeutic benefit, is always a question of appropriate medical concern. safety with regard to patients with chronic kidney disease. For instance,
The Gd-based chelates had initially been considered among the safest there is a much lower incidence of NSF among patients who received
injectable compounds in medical use, with a specific reputation for Gd-HPDO3A (gadoteridol),137 and this may be a result of its combina-
superior safety in patients with renal dysfunction (compared with iodin- tion of stability and inertness.
ated preparations used in fluoroscopy and CT). This view has changed A series of studies published since 2013 have raised concerns over
with the apparent link between Gd contrast and nephrogenic systemic long-term Gd retention in the brain, bone, and skin of patients receiv-
fibrosis (NSF). ing multiple contrast-agent-enhanced examinations, including patients
A very rare, but potentially devastating condition affecting patients with normal renal function. The observation that the dentate nucleus
with acute or chronic kidney disease, NSF has an estimated prevalence and globus pallidus appear hyperintense in contrast-free T1-weighted
of >10% among patients receiving Gd who are on hemodialysis.113 scans of patients having received multiple prior contrast-enhanced scans
Although presenting primarily with skin thickening, tethering, hyper- suggested residual gadolinium accumulation in those regions of the
pigmentation, and disabling joint flexion contractures, patients with brain.138–139b Elemental analysis of autopsy specimens collected from
NSF can also have multiorgan/systemic fibrosis, leading to organ dys- patients receiving numerous injections of Gd-based contrast agents
function and even death.114,115 The universal feature of NSF is Gd admin- confirmed that there was measurable gadolinium in the brain, albeit
istration and acute or chronic kidney disease, most commonly with an at low levels.140 The amount of central nervous system (CNS) Gd
estimated glomerular filtration rate of <30 mL/min/1.73 m2.115–118 deposition may depend on the type of contrast agent received. CNS
However, NSF-like symptoms have also been reported in patients with tissue enhancement by residual Gd appears to be greater after receiv-
apparently normal renal function.119,120 Gd has been shown to be present ing multiple injections of Gd contrast with acyclic chelators.141 This
in skin biopsy specimens of patients with NSF,121–126 and also in internal is evidenced by consistently hyperintense dentate nuclei in patients
organs.127 The prevailing hypothesis is that acute or chronic kidney receiving multiple doses of the Gd complexes with acyclic chelators,
disease results in prolonged exposure to the contrast agent, providing whereas the effect is not observed in patients exclusively receiving
opportunity for the Gd to dissociate from its chelator, and this dissoci- Gd complexes with macrocyclic chelators. Nonetheless, postmortem
ated Gd is believed to be responsible for the toxic response.128 The least elemental analysis detects non-negligible Gd levels in CNS tissue of
stable contrast agent, Gd-DTPA-BMA (gadodiamide), is the agent that patients receiving only macrocyclic agents, including healthy patients
has been most frequently associated with NSF,129–131 but it is now estab- receiving a single dose in their lifetime,142 and imaging studies demon-
lished that other Gd compounds also pose a risk.115,132 strate retained Gd after high doses of macrocytic agents.139b The same
Although ECF agents are similar in terms of their imaging efficacy, study found highly elevated Gd levels in the bone of all patients receiv-
they differ in terms of how well they bind Gd. This may prove to be ing both acyclic and macrocyclic agents; Gd retention in the skin was
an important risk factor for NSF because both the metal (Gd) and also observed.
the chelate have potential toxic effects6 and have shown acute toxicity The speciation of Gd retained in the skin, bone, and CNS remains
in animal studies at high enough doses. These various animal studies largely underexplored (chelated vs. unchelated), as do the mechanisms
have been reviewed,133 and a high-dose study in rats showed NSF-like by which Gd is accumulated and the corresponding health risks. A
lesions.131 Metal complexes such as Gd contrast agents are character- recent study used chromatography coupled with elemental spectroscopy
ized in terms of their thermodynamic stability and kinetic inertness. to evaluate Gd speciation in the skin of a patient with healthy kidney
Stability is a measure of the affinity of the chelator for the metal ion function who received 61 contrast-enhanced examination over the course
and is expressed as an equilibrium constant for the association of the of 11 years.143 The patient received multiple Gd-based agents including
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 39

Gd-DTPA, Gd-BOPTA, Gd-DTPA-BMA, and Gd-HPDO3A. Speciation appear to interfere with the reagent used in the clinical chemistry test
analysis revealed skin accumulation of intact Gd-DTPA and Gd-BOPTA for calcium.149
as well as still uncharacterized Gd-containing species that likely exist The safety profile for current CMR contrast agents of course will
in particulate form or are protein bound. In 2015 the FDA announced not necessarily be the same for future compounds. For all contrast
a formal investigation into the safety risks of Gd retained in the CNS144 agents, the package insert should always be consulted for the latest
and subsequently recommended against the use of linear agents and safety information.
have issued a new class warning.144a
Regarding other side effects, Kirchin and Runge145 reviewed the
safety record of clinically approved contrast agents as of 2003. The
CONCLUSION
seven approved Gd agents (Gd-DTPA [gadopentetate], Gd-DOTA A large number of contrast agents have been approved for MRI in the
[gadoterate], Gd-BOPTA [gadobenate], Gd-DTPA-BMA [gadodiamide], last 26 years. A significant number of new agents, including tissue-
Gd-DTPA-BMEA [gadoversatamide], Gd-DO3A-butrol [gadobutrol], specific agents that are potentially relevant for CMR, are currently in
and Gd-BOPTA [gadobenate]) appeared indistinguishable with respect development. The behavior of these agents, which often is summarized
to their safety profile. The most common adverse events were headache, by a single effectiveness parameter, r1, is actually quite complex, in terms
nausea, taste perversion, and urticaria (hives). Nearly all adverse events of both the underlying chemistry and the biophysics in vivo. Although
with these agents were transient, mild, and self-limiting. Nevertheless, many elements of that complexity remain active areas of research, both
there are reports of serious adverse reactions, including life-threatening for understanding existing agents as well as for creating new agents and
anaphylactoid reactions and death. The best estimate puts the rate of new uses for those agents, the relative safety and ease of use of these
these events at between 1 in 200,000 and 1 in 400,000 patient admin- agents has brought them into routine use in many medical applications.
istrations.146 The Gd-based ECF contrast agents have been studied, and As clinical CMR expands, no doubt the use of these contrast agents
most are approved for pediatric use in patients older than 2 years, will expand as well, but CMR practitioners need to be cognizant of
although there are differences in their approval wording. Gd-DO3A- potential moderate-term complications including NSF in patients with
butrol (gadobutrol) is the only Gd-based contrast agent approved for renal dysfunction. The long-term impact of Gd retention in the CNS
use in patients younger than 2 years of age in the United States.147 and elsewhere remains uncertain.
One distinguishing clinical indicator among the Gd-based agents is
that patients receiving Gd-DTPA-BMA (gadodiamide) or Gd-DTPA-
BMEA (gadoversatamide) often show spuriously low serum calcium
REFERENCES
levels.148 These two contrast agents (but not the other Gd-based agents) A full reference list is available online at ExpertConsult.com
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 39.e1

REFERENCES 22. Khurana A, Nejadnik H, Gawande R, et al. Intravenous ferumoxytol


allows noninvasive MR imaging monitoring of macrophage migration
1. Bruder O, Wagner A, Lombardi M, et al. European cardiovascular into stem cell transplants. Radiology. 2012;264:803–811.
magnetic resonance (EuroCMR) registry—multinational results from 57 23. Hasan DM, Mahaney KB, Magnotta VA, et al. Macrophage imaging
centers in 15 countries. J Cardiovasc Magn Reson. 2013;15:9. within human cerebral aneurysms wall using ferumoxytol-enhanced
2. Bloch F, Hansen WW, Packard M. The nuclear induction experiment. MRI: a pilot study. Arterioscler Thromb Vasc Biol. 2012;32:1032–1038.
Phys Rev. 1948;70:474–485. 24. Fries P, Müller A, Seidel R, et al. P03277—a new approach to achieve
3. Lauterbur PC, Mendonca Dias MH, Rudin AM. Augmentation of high-contrast enhancement: initial results of an experimental
tissue water proton spin-lattice relaxation rates by in vivo addition extracellular gadolinium-based magnetic resonance contrast agent.
of paramagnetic ions. In: Dutton PO, Leigh J, Scarpa A, eds. Invest Radiol. 2015;50:835–842.
Frontiers of Biological Energetics. New York: Academic Press; 1978: 25. Lauffer RB. Targeted relaxation enhancement agents for MRI. Magn
752–759. Reson Med. 1991;22:339.
4. Carr DH, Brown J, Bydder GM, et al. Intravenous chelated gadolinium 26. Toth E, Van Uffelen I, Helm L, et al. Gadolinium-based linear polymer
as a contrast agent in NMR imaging of cerebral tumors. Lancet. with temperature-independent proton relaxivities: a unique interplay
1984;1:484–486. between the water exchange and rotational contributions. Magn Reson
5. Caravan P, Ellison JJ, McMurry TJ, Lauffer RB. Gadolinium(III) Chelates Chem. 1998;36:S125–S134.
as MRI contrast agents: structure, dynamics, and applications. Chem 27. Bertini I, Luchinat C, Parigi G. Solution NMR of Paramagnetic Molecules.
Rev. 1999;99:2293–2352. Amsterdam: Elsevier; 2001.
6. Gries H. Extracellular MRI contrast agents based on gadolinium. Top 28. Muller RN, Roch A, Colet J-M, Ouakssim A, Gillis P. Particulate
Curr Chem. 2002;221:1–24. magnetic contrast agents. In: Merbach AE, Toth E, eds. Chemistry of
7. Powell DH, Ni Dhubhghaill OM, Pubanz D, et al. High-pressure NMR Contrast Agents in Medical Magnetic Resonance Imaging. New York:
kinetics. Part 74. Structural and dynamic parameters obtained from 17O Wiley; 2001:417–435.
NMR, EPR, and NMRD studies of monomeric and dimeric Gd3+ 29. Jung CW, Jacobs P. Physical and chemical properties of
complexes of interest in magnetic resonance imaging: an integrated and superparamagnetic iron oxide MR contrast agents: ferumoxides,
theoretically self-consistent approach. J Am Chem Soc. ferumoxtran, ferumoxsil. Magn Reson Imaging. 1995;13:661–674.
1996;118:9333–9346. 30. Tweedle MF, Wedeking P, Telser J, et al. Dependence of MR signal
8. Gillis A, Gray M, Burstein D. Relaxivity and diffusion of gadolinium intensity on Gd tissue concentration over a broad dose range. Magn
agents in cartilage. Magn Reson Med. 2002;48:1068–1071. Reson Med. 1991;22:191–194.
9. Kumar K, Tweedle MF. Ligand basicity and rigidity control formation of 31. Wedeking P, Sotak CH, Telser J, Kumar K, Chang CA, Tweedle MF.
macrocyclic polyamino carboxylate complexes of gadolinium(III). Inorg Quantitative dependence of MR signal intensity on tissue concentration
Chem. 1993;32:4193–4199. of Gd(HP-DO3A) in the nephrectomized rat. Magn Reson Imaging.
10. White DH, DeLearie LA, Moore DA, et al. The thermodynamics of 1992;10:97–108.
complexation of lanthanide(III) DTPA-bisamide complexes and their 32. Wang Y, Spiller M, Caravan P. Evidence for weak protein binding of
implication for stability and solution structure. Invest Radiol. commerical extracellular gadolinium contrast agents. Magn Reson Med.
1991;26:S226–S228. 2010;63:609–616.
11. Tombach B, Heindel W. Value of 1.0- M gadolinium chelates: review of 33. Stanisz GJ, Henkelman RM. Gd-DTPA relaxivity depends on
preclinical and clinical data on gadobutrol. Eur Radiol. macromolecular content. Magn Reson Med. 2000;44:665–667.
2002;12:1550–1556. 34. Donahue KM, Weisskoff RM, Burstein D. Water diffusion and exchange
12. Caravan P, Cluotier NJ, Greenfield MT, et al. The interaction of MS-325 as they influence contrast enhancement. J Magn Reson Imaging.
with human serum albumin and its effect on proton relaxation rates. J 1997;7:102–110.
Am Chem Soc. 2002;124:3152–3162. 35. Li X, Rooney WD, Springer CS Jr. A unified magnetic resonance imaging
13. Lauffer RB, Parmelee DJ, Dunham SU, et al. MS-325: albumin-targeted pharmacokinetic theory: intravascular and extracellular contrast
contrast agent for MR angiography. Radiology. 1998;207:529–538. reagents. Magn Reson Med. 2005;54:1351–1359.
14. de Haen C, La Ferla R, Maggioni F. Gadobenate dimeglumine 0.5 M 36. Hazlewood CF, Chang DC, Nichols BL, Woessner DE. Nuclear magnetic
solution for injection (MultiHance) as contrast agent for magnetic resonance transverse relaxation times of water protons in skeletal
resonance imaging of the liver: mechanistic studies in animals. J Comput muscle. Biophys J. 1974;14:583–606.
Assist Tomogr. 1999;23:S169–S179. 37. McLaughlin AC, Leigh JS. Relaxation times in systems with chemical
15. Vander Elst L, Maton F, Laurent S, Seghi F, Chapelle F, Muller RN. A exchange. J Magn Reson. 1973;9:296–304.
multinuclear MR study of Gd-EOB-DTPA: comprehensive preclinical 38. Wright GA, Hu BS, Macovski A. Estimating oxygen saturation of blood in
characterization of an organ specific MRI contrast agent. Magn Reson vivo with MR imaging at 1.5 T. J Magn Reson Imaging. 1991;1:275–283.
Med. 1997;38:604–614. 39. Donahue KM, Weisskoff RM, Chesler DA, et al. Improving MR
16. Eldredge HB, Spiller M, Chasse JM, Greenwood MT, Caravan P. Species quantification of regional blood volume with intravascular T1 contrast
dependence on plasma protein binding and relaxivity of the agents: accuracy, precision, and water exchange. Magn Reson Med.
gadolinium-based MRI contrast agent MS-325. Invest Radiol. 1996;36:858–867.
2006;41:229–243. 40. Sukstanskii AL, Yablonskiy DA. Gaussian approximation in the theory of
17. Port M, Corot C, Violas X, Robert P, Raynal I, Gagneur G. How to MR signal formation in the presence of structure-specific magnetic field
compare the efficiency of albumin-bound and nonalbumin-bound inhomogeneities. Effects of impermeable susceptibility inclusions. J
contrast agents in vivo: the concept of dynamic relaxivity. Invest Radiol. Magn Reson. 2004;167:56–67.
2005;40:565–573. 41. Weissleder R, Mahmood U. Molecular imaging. Radiology. 2001;219:
18. Li S-D, Huang L. Pharmacokinetics and biodistribution of nanoparticles. 316–333.
Mol Pharm. 2008;5:496–504. 42. Caravan P, Kolodziej AF, Greenwood JM, et al. EP-1242: a fibrin targeted
19. AMAG Pharmaceuticals I. Feraheme Package Insert. Waltham, MA: contrast agent for thrombus imaging. 10th ISMRM Scientific Sessions.
AMAG Pharmaceuticals, Inc. 2002:217.
20. Hope MD, Hope TA, Zhu C, et al. Vascular imaging with ferumoxytol as 43. Overoye-Chan K, Koerner S, Looby RJ, et al. EP-2104R: a fibrin-specific
a contrast agent. AJR Am J Roentgenol. 2015;205:W366–W374. gadolinium-based MRI contrast agent for detection of thrombus. J Am
21. Ruangwattanapaisarn N, Hsiao A, Vasanawala SS. Ferumoxytol as an Chem Soc. 2008;130:6025–6039.
off-label contrast agent in body 3T MR angiography: a pilot study in 44. Botnar RM, Perez AS, Witte S, et al. In vivo molecular imaging of acute
children. Pediatr Radiol. 2015;45:831–839. and subacute thrombosis using a fibrin-binding magnetic resonance
39.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

imaging contrast agent. Circulation. 2004;109:2023–2029. [Epub 2004 65. Barkhausen J, Ebert W, Heyer C, Debatin JF, Weinmann HJ. Detection of
Apr 5]. atherosclerotic plaque with Gadofluorine-enhanced magnetic resonance
45. Sirol M, Aguinaldo JG, Graham PB, et al. Fibrin-targeted contrast agent imaging. Circulation. 2003;108:605–609. [Epub 2003 Jun 30].
for improvement of in vivo acute thrombus detection with magnetic 66. Frias JC, Williams KJ, Fisher EA, Fayad ZA. Recombinant HDL-like
resonance imaging. Atherosclerosis. 2005;182:79–85. nanoparticles: a specific contrast agent for MRI of atherosclerotic
46. Spuentrup E, Buecker A, Katoh M, et al. Molecular magnetic resonance plaques. J Am Chem Soc. 2004;126:16316–16317.
imaging of coronary thrombosis and pulmonary emboli with a novel 67. Briley-Saebo KC, Amirbekian V, Mani V, et al. Gadolinium mixed-
fibrin-targeted contrast agent. Circulation. 2005;111:1377–1382. micelles: effect of the amphiphile on in vitro and in vivo efficacy in
47. Botnar RM, Buecker A, Wiethoff AJ, et al. In vivo magnetic resonance apolipoprotein E knockout mouse models of atherosclerosis. Magn
imaging of coronary thrombosis using a fibrin-binding molecular Reson Med. 2006;56:1336–1346.
magnetic resonance contrast agent. Circulation. 2004;110:1463–1466. 68. Mulder WJ, Strijkers GJ, Briley-Saebo KC, et al. Molecular imaging of
[Epub 2004 Jul 6]. macrophages in atherosclerotic plaques using bimodal PEG-micelles.
48. Spuentrup E, Katoh M, Wiethoff AJ, et al. Molecular magnetic resonance Magn Reson Med. 2007;58:1164–1170.
imaging of pulmonary emboli with a fibrin-specific contrast agent. Am J 69. Amirbekian V, Lipinski MJ, Briley-Saebo KC, et al. Detecting and
Respir Crit Care Med. 2005;172:494–500. [Epub 2005 Jun 3]. assessing macrophages in vivo to evaluate atherosclerosis noninvasively
49. Spuentrup E, Katoh M, Buecker A, et al. Molecular MR imaging of using molecular MRI. Proc Natl Acad Sci USA. 2007;104:961–966.
human thrombi in a swine model of pulmonary embolism using a 70. Chen WC, Carmode DP, Vengrenyuk Y, et al. Collagen-specific peptide
fibrin-specific contrast agent. Invest Radiol. 2007;42:586–595. conjugated HDL nanoparticles as MRI contrast agent to evaluate
50. Andia ME, Saha P, Jenkins J, et al. Fibrin-targeted magnetic resonance compositional changes in atherosclerotic plaque regression. J Am Coll
imaging allows in vivo quantification of thrombus fibrin content and Cardiol. 2013;6:373–384.
identifies thrombi amenable for thrombolysis. Arterioscler Thromb Vasc 71. Lancelot E, Amirbekian V, Brigger I, et al. Evaluation of matrix
Biol. 2014;34:1193–1198. metalloproteinases in atherosclerosis using a novel noninvasive imaging
51. Spuentrup E, Botnar RM, Wiethoff AJ, et al. MR imaging of thrombi approach. Arterioscler Thromb Vasc Biol. 2008;28:425–432.
using EP-2104R, a fibrin-specific contrast agent: initial results in 72. Caravan P, Das B, Dumas S, et al. Collagen-targeted MRI constrast agent for
patients. Eur Radiol. 2008;18:1995–2005. molecular imaging of fibrosis. Angew Chem Int Ed Engl. 2007;46:8171–8173.
52. Cyrus T, Winter PM, Caruthers SD, Wickline SA, Lanza GM. Magnetic 73. Helm PA, Caravan P, French BA, et al. Postinfarction myocardial
resonance nanoparticles for cardiovascular molecular imaging and scarring in mice: molecular MR imaging with use of a collagen-targeting
therapy. Expert Rev Cardiovasc Ther. 2005;3:705–715. contrast agent. Radiology. 2008;3:788–796.
53. Lanza GM, Winter P, Caruthers S, et al. Novel paramagnetic contrast 74. Spuentrup E, Ruhl KM, Botnar RM, et al. Molecular magnetic resonance
agents for molecular imaging and targeted drug delivery. Curr Pharm imaging of myocardial perfusion with EP-3600, a collagen-specific
Biotechnol. 2004;5:495–507. contrast agent. Circulation. 2009;119:1768–1775.
54. Winter PM, Caruthers SD, Yu X, et al. Improved molecular imaging 75. Polasek M, Fuchs BC, Uppal R, et al. Molecular MR imaging of liver
contrast agent for detection of human thrombus. Magn Reson Med. fibrosis: a feasibility study using rat and mouse models. J Hepatol.
2003;50:411–416. 2012;57:549–555.
55. Flacke S, Fischer S, Scott MJ, et al. Novel MRI contrast agent for 76. Fuchs BC, Wang H, Yang Y, et al. Molecular MRI of collagen to diagnose
molecular imaging of fibrin: implications for detecting vulnerable and stage liver fibrosis. J Hepatol. 2013;59:992–998.
plaques. Circulation. 2001;104:1280–1285. 77. Caravan P, Yang Y, Zachariah R, et al. Molecular magnetic resonance
56. Yu X, Song SK, Chen J, et al. High-resolution MRI characterization of imaging of pulmonary fibrosis in mice. Am J Respir Cell Mol Biol.
human thrombus using a novel fibrin-targeted paramagnetic 2013;49:1120–1126.
nanoparticle contrast agent. Magn Reson Med. 2000;44:867–872. 78. Botnar RM, Wiethoff AJ, Ebersberger U, et al. In vivo assessment of
57. Morawski AM, Winter PM, Crowder KC, et al. Targeted nanoparticles aortic aneurysm wall integrity using elastin-specific molecular magnetic
for quantitative imaging of sparse molecular epitopes with MRI. Magn resonance imaging. Circ Cardiovasc Imaging. 2015;7:679–689.
Reson Med. 2004;51:480–486. 79. Phinikaridou A, Andia ME, Indermuehle A, et al. Vascular remodeling
58. Schmieder AH, Winter PM, Caruthers SD, et al. Molecular MR imaging and plaque vulnerability in a rabbit model of atherosclerosis:
of melanoma angiogenesis with alphanubeta3-targeted paramagnetic comparison of delayed enhancement MR imaging with an elastin-
nanoparticles. Magn Reson Med. 2005;53:621–627. specific contrast agent and unenhanced black-blood MR imaging.
59. Winter PM, Morawski AM, Caruthers SD, et al. Molecular imaging of Radiology. 2014;271:390–399.
angiogenesis in early-stage atherosclerosis with alpha(v)beta3-integrin- 80. Protti A, Lavin B, Dong X, et al. Assessment of myocardial remodeling
targeted nanoparticles. Circulation. 2003;108:2270–2274. [Epub 2003 using an elastin/tropoelastin specific agent with high field magnetic
Oct 13]. resonance imaging (MRI). J Am Heart Assoc. 2015;4:e001851.
60. Winter PM, Caruthers SD, Kassner A, et al. Molecular imaging of 81. Wildgruber M, Bielicki I, Aichler M, et al. Assessment of myocardial
angiogenesis in nascent Vx-2 rabbit tumors using a novel alpha(nu) infarction and postinfarction scar remodeling with an elastin-specific
beta3-targeted nanoparticle and 1.5 tesla magnetic resonance imaging. magnetic resonance agent. Circ Cardiovasc Imaging. 2014;7(2):321.
Cancer Res. 2003;63:5838–5843. 82. Nahrendorf M, Sosnovik D, Chen JW, et al. Activatable magnetic
61. Winter PM, Neubauer AM, Caruthers SD, et al. Endothelial alpha(v) resonance imaging agent reports myeloperoxidase activity in healing
beta3 integrin-targeted fumagillin nanoparticles inhibit angiogenesis infarcts and noninvasively detects the antiinflammatory effects of
in atherosclerosis. Arterioscler Thromb Vasc Biol. 2006;26:2103– atorvastatin on ischemia-reperfusion injury. Circulation.
2109. 2008;117:1153–1169.
62. Naresh NK, Xu Y, Klibanov AL, et al. Monocyte and/or macrophage 83. Breckwoldt MO, Chen JW, Stangenberg L, et al. Tracking the
infiltration of heart after myocardial infarction: MR imaging by using inflammatory response in stroke in vivo by sensing the enzyme
T1-shortening liposomes1. Radiology. 2012;2:428–435. myeloperoxidase. Proc Natl Acad Sci USA. 2008;105:18584–18589.
63. Meding J, Urich M, Licha K, et al. Magnetic resonance imaging of 84. DeLeo MJ III, Gounis MJ, Hong B, Ford JC, Wakhloo AK, Bogdanov AA
atherosclerosis by targeting extracellular matrix deposition with Jr. Carotid artery brain aneurysm model: in vivo molecular
Gadofluorine M. Contrast Media Mol Imaging. 2007;2: enzymespecific MR imaging of active inflammation in a pilot study.
120–129. Radiology. 2009;252:696–703.
64. Sirol M, Itskovich VV, Mani V, et al. Lipid-rich atherosclerotic plaques 85. Su HS, Nahrendorf M, Panizzi P, et al. Vasculitis: molecular imaging by
detected by gadofluorine-enhanced in vivo magnetic resonance imaging. targeting the inflammatory enzyme myeloperoxidase. Radiology.
Circulation. 2004;109:2890–2896. 2012;262:181–190.
CHAPTER 3  Cardiovascular Magnetic Resonance Contrast Agents 39.e3

86. Huang S, Chen HH, Yuan H, et al. Molecular MRI of actue necrosis 109. Merritt ME, Harrison C, Storey C, Jeffrey FM, Sherry AD, Malloy CR.
with a novel DNA-binding gadolinium chelate: kinetics of cell death and Hyperpolarized 13C allows a direct measure of flux through a single
clearance in infarcted myocardium. Circ Cardiovasc Imaging. 2011;4: enzyme-catalyzed step by NMR. Proc Natl Acad Sci USA.
729–737. 2007;104:19773–19777.
87. Gale EM, Atanasova I, Blasi F, Ay I, Caravan P. A manganese alternative 110. Dodd MS, Atherton HJ, Carr CA, et al. Impaired in vivo mitochondrial
to gadolinium for MRI contrast. J Am Chem Soc. 2015;137:15548–15557. krebs cycle activity after myocardial infarction assessed using
88. Corot C, Petry KG, Trivedi R, et al. Macrophage imaging in central hyperpolarized magnetic resonance spectroscopy. Circ Cardiovasc
nervous system and in carotid atherosclerotic plaque using ultrasmall Imaging. 2014;7:895–904.
superparamagnetic iron oxide in magnetic resonance imaging. Invest 111. Oh-Ici D, Wespi P, Busch J, et al. Hyperpolarized metabolic MR imaging
Radiol. 2004;39:619–625. of acute myocardial changes and recovery after ischemia-reperfusion in
89. Yancy AD, Olzinski AR, Hu TC, et al. Differential uptake of a small-animal model. Radiology. 2016;278:742–751.
ferumoxtran-10 and ferumoxytol, ultrasmall superparamagnetic iron 112. Koellisch U, Gringeri CV, Rancan G, et al. Metabolic imaging of
oxide contrast agents in rabbit: critical determinants of atherosclerotic hyperpolarized [1-13C]acetate and [1-13C]acetylcarnitine—
plaque labeling. J Magn Reson Imaging. 2005;21:432–442. investigation of the influence of dobutamine induced stress. Magn Reson
90. Modo M, Hoehn M, Bulte JW. Cellular MR imaging. Mol Imaging. Med. 2015;74:1011–1018.
2005;4:143–164. 113. Todd DJ, Kagan A, Chibnik LB, Kay J. Cutaneous changes of nephrogenic
91. Bulte JW, Kraitchman DL. Monitoring cell therapy using iron oxide MR systemic fibrosis: predictor of early mortality and association with
contrast agents. Curr Pharm Biotechnol. 2004;5:567–584. gadolinium exposure. Arthritis Rheum. 2007;56:3433–3441.
92. Hill JM, Dick AJ, Raman VK, et al. Serial cardiac magnetic resonance 114. Cowper SE. Nephrogenic systemic fibrosis: a review and exploration of
imaging of injected mesenchymal stem cells. Circulation. 2003;108:1009– the role of gadolinium. Adv Dermatol. 2007;23:131–154.
1014. [Epub 2003 Aug 11]. 115. Cowper SE, Kuo PH, Bucala R. Nephrogenic systemic fibrosis and
93. Kraitchman DL, Heldman AW, Atalar E, et al. In vivo magnetic gadolinium exposure. Association and lessons for idiopathic fibrosing
resonance imaging of mesenchymal stem cells in myocardial infarction. disorders. Arthritis Rheum. 2007;56:3173–3175.
Circulation. 2003;107:2290–2293. 116. Grobner T. Gadolinium—a specific trigger for the development of
94. Arai T, Kofidis T, Bulte JW, et al. Dual in vivo magnetic resonance nephrogenic fibrosing dermopathy and nephrogenic systemic fibrosis?
evaluation of magnetically labeled mouse embryonic stem cells and Nephrol Dial Transplant. 2006;21:1104–1108.
cardiac function at 1.5 t. Magn Reson Med. 2006;55:203–209. 117. Grobner T, Prischl FC. Gadolinium and nephrogenic systemic fibrosis.
95. Ma N, Cheng H, Lu M, et al. Magnetic resonance imaging with Kidney Int. 2007;72:260–264.
superparamagnetic iron oxide fails to track the long-term fate of 118. Thomsen HS. Nephrogenic systemic fibrosis: a serious late adverse
mesenchymal stem cells transplanted into heart. Sci Rep. 2015;5:9058. reaction to gadodiamide. Eur Radiol. 2006;16:2619–2621.
96. Wunderbaldinger P, Josephson L, Weissleder R. Crosslinked iron oxides 119. Burke LMB, Ramalho M, AlObaidy M, Chang E, Jay M, Semelka RC.
(CLIO): a new platform for the development of targeted MR contrast Self-reported gadolinium toxicity: a survey of patients with chronic
agents. Acad Radiol. 2002;9:S304–S306. symptoms. Magn Reson Imaging. 2016;34:1078–1080.
97. Moore A, Sun PZ, Cory D, Hogemann D, Weissleder R, Lipes MA. MRI 120. Semelka RC, Commander C, Jay M, Burke LMB, Ramalho M. Presumed
of insulitis in autoimmune diabetes. Magn Reson Med. 2002;47: gadolinium toxicity in subjects with normal renal function: a report of 4
751–758. cases. Invest Radiol. 2016;51:661–665.
98. Hogemann D, Josephson L, Weissleder R, Basilion JP. Improvement of 121. Boyd AS, Zic JA, Abraham JL. Gadolinium deposition in nephrogenic
MRI probes to allow efficient detection of gene expression. Bioconjug fibrosing dermopathy. J Am Acad Dermatol. 2007;56:27–30.
Chem. 2000;11:941–946. 122. High WA, Ayers RA, Chandler J, Zito G, Cowper SE. Gadolinium is
99. Schellenberger EA, Bogdanov A Jr, Hogemann D, Tait J, Weissleder R, detectable within the tissue of patients with nephrogenic systemic
Josephson L. Annexin V-CLIO: a nanoparticle for detecting apoptosis by fibrosis. J Am Acad Dermatol. 2007;56:21–26.
MRI. Mol Imaging. 2002;1:102–107. 123. High WA, Ayers RA, Cowper SE. Gadolinium is quantifiable within the
100. Sosnovik DE, Schellenberger EA, Nahrendorf M, et al. Magnetic tissue of patients with nephrogenic systemic fibrosis. J Am Acad
resonance imaging of cardiomyocyte apoptosis with a novel magneto- Dermatol. 2007;54:710–712.
optical nanoparticle. Magn Reson Med. 2005;54:718–724. 124. Saussereau E, Lacroix C, Cattaneo A, Mahieu L, Goulle JP. Hair and
101. Nahrendorf M, Jaffer FA, Kelly KA, et al. Noninvasive vascular cell fingernail gadolinium ICP-MS contents in an overdose case associated
adhesion molecule-1 imaging identifies inflammatory activation of cells with nephrogenic systemic fibrosis. Forensic Sci Int. 2008;176:54–57.
in atherosclerosis. Circulation. 2006;114:1504–1511. 125. Abraham JL, Thakral C, Skov L, Rossen K, Marckmann P. Dermal
102. Jacoby C, Borg N, Heusch P, et al. Visualization of immune cell inorganic gadolinium concentrations: evidence for in vivo
infiltration in experimental viral myocarditis by 19F MRI in vivo. Magn transmetallation and long-term persistence in nephrogenic systemic
Reson Mater Phys. 2014;27:101–106. fibrosis. Br J Dermatol. 2008;158:273–280.
103. Bönner F, Merx MW, Klingel K, et al. Monocyte imaging aftermyocardial 126. Thakral C, Alhariri J, Abraham JL. Long-term retention of gadolinium
infarction with 19F MRI at 3 T: a pilot study in explanted porcine in tissues from nephrogenic systemic fibrosis patient after multiple
hearts. Eur Heart J Cardiovasc Imaging. 2015;16:612–620. gadolinium-enhanced MRI scans: case report and implications. Contrast
104. Bönner F, Jacoby C, Temme S, et al. Multifunctional MR monitoring of Media Mol Imaging. 2007;2:199–205.
the healing process after myocardial infarction. Basic Res Cardiol. 127. Kay J, Bazari H, Avery LL, Koreishi AF. Case records of the
2014;109:430. Massachusetts General Hospital. Case 6-2008: a 46-year-old woman with
105. Temme S, Grapentin C, Quast C, et al. Noninvasive imaging of early renal failure and stiffness of the joints and skin. N Engl J Med.
venous thrombosis by 19F magnetic resonance imaging with targeted 2008;358:827–838.
perfluorocarbon nanoemulsions. Circulation. 2015;131:1405–1414. 128. Kurtkoti J, Snow T, Hiremagalur B. Gadolinium and nephrogenic systemic
106. Moller HE, Chen XJ, Saam B, et al. MRI of the lungs using fibrosis: association of causation. Nephrology (Carlton). 2008;13:235–241.
hyperpolarized noble gases. Magn Reson Med. 2002;47:1029–1051. 129. Richmond H, Zwerner J, Kim Y, Fiorentino D. Nephrogenic systemic
107. Svensson J, Mansson S, Johansson E, Petersson JS, Olsson LE. fibrosis: relationship to gadolinium and response to photopheresis. Arch
Hyperpolarized 13C MR angiography using true FISP. Magn Reson Med. Dermatol. 2007;143:1025–1030.
2003;50:256–262. 130. Rydahl C, Thomsen HS, Marckmann P. High prevalence of nephrogenic
108. Olsson LE, Chai CM, Axelsson O, Karlsson M, Golman K, Petersson JS. systemic fibrosis in chronic renal failure patients exposed to
MR coronary angiography in pigs with intraarterial injections of a gadodiamide, a gadolinium-containing magnetic resonance contrast
hyperpolarized 13C substance. Magn Reson Med. 2006;55:731–737. agent. Invest Radiol. 2008;43:141–144.
39.e4 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

131. Sieber MA, Pietsch H, Walter J, Haider W, Frenzel T, Weinmann HJ. A 144. US Food and Drug Administration. FDA Drug Safety Communication:
preclinical study to investigate the development of nephrogenic systemic FDA evaluating the risk of brain deposits with repeated use of
fibrosis: a possible role for gadolinium-based contrast media. Invest gadolinium-based contrast agents for magnetic resonance imaging
Radiol. 2008;43:65–75. (MRI); 2015. http://www.fda.gov/Drugs/DrugSafety/ucm455386.htm.
132. Leiner T, Herborn CU, Goyen M. Nephrogenic systemic fibrosis is not 144a.  U.S. Food & Drug Administration. Gadolinium-based contrast agents
exclusively associated with gadodiamide. Eur Radiol. 2007;17:1921–1923. (GBCAs): drug safety communication – retained in body; new
133. Caravan P, Lauffer RB. Contrast agents: basic principles. In: Edelman class warning; 2017. https://www.fda.gov/Safety/MedWatch/
RR, Hesselink JR, Zlatkin MB, Crues JV, eds. Clinical Magnetic Resonance SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm589580
Imaging. Vol. 1. 3rd ed. Philadelphia: Saunders; 2005:357–375. .htm. Accessed February 20, 2018.
134. Kumar K, Chang CA, Tweedle MF. Equilibrium and kinetic studies of 145. Kirchin MA, Runge VM. Contrast agents for magnetic resonance
lanthanide complexes of macrocyclic polyamino carboxylates. Inorg imaging: safety update. Top Magn Reson Imaging. 2003;14:
Chem. 1993;32:587–593. 426–435.
135. Laurent S, Elst LV, Muller RN. Comparative study of the 146. Carr JJ. Magnetic resonance contrast agents for neuroimaging. Safety
physicochemical properties of six clinical low molecular weight issues. Neuroimaging Clin N Am. 1994;4:43–54.
gadolinium contrast agents. Contrast Media Mol Imaging. 147. Kunze CW, Mentel H-J, Krishnamurthy R, et al. Pharmacokinetics and
2006;1:128–137. safety of macrocyclic gadobutrol in children aged younger than 2 years
136. Laurent S, Vander Elst L, Copoix F, Muller RN. Stability of MRI including term newborns in comparison to older populations. Invest
paramagnetic contrast media: a proton relaxometric protocol for Radiol. 2016;51:50–57.
transmetallation assessment. Invest Radiol. 2001;36:115–122. 148. Prince MR, Erel HE, Lent RW, et al. Gadodiamide administration causes
137. Reilly RF. Risk for nephrogenic systemic fibrosis with gadoteridol spurious hypocalcemia. Radiology. 2003;227:639–646.
(prohance) in patients who are on long-term hemodialysis. Clin J Am 149. Emerson J, Kost G. Spurious hypocalcemia after Omniscan- or
Soc Nephrol. 2008;3:747–751. OptiMARK-enhanced magnetic resonance imaging: an algorithm for
138. Kanda T, Kawaguchi H. Hyperintense dentate nucleas and globus minimizing a false-positive laboratory value. Arch Pathol Lab Med.
pallidus on unenhanced T1-weighted MR images are associated with 2004;128:1151–1156.
gadolinium-based contrast media. Neuroradiology. 2013;55:1268–1269. 150. Rohrer M, Bauer H, Mintorovitch J, Requardt M, Weinmann HJ.
139. Kanda T, Ishii K, Kawaguchi H, Kitajima K, Takenaka D. High signal Comparison of magnetic properties of MRI contrast media solutions at
intensity in the dentate nucleus and globus pallidus on unenhanced different magnetic field strengths. Invest Radiol. 2005;40:715–724.
T1-weighted MR images: relationship with increasing cumulative dose 151. Tweedle MF, Hagan JJ, Kumar K, Mantha S, Chang CA. Reaction of
of a gadolinium-based contrast material. Radiology. 2014;270:834–841. gadolinium chelates with endogenously available ions. Magn Reson
139a.  Kanal E. Gadolinium-based contrast agents: the plot thickens. Imaging. 1991;9:409–415.
Radiology. 2017;285:340–342. 152. Tweedle MF. Physicochemical properties of gadoteridol and other
139b.  Bjornerud A, Vatnehol SAS, Larsson C, et al. Signal enhancement of magnetic resonance contrast agents. Invest Radiol. 1992;27:2–6.
the dentate nucleus at unenhanced MR imaging after very high 153. Vogler H, Platzek J, Schuhmann-Giampieri G, et al. Pre-clinical
cumulative doses of the macrocytic gadolinium-based contrast agent evaluation of gadobutrol: a new, neutral, extracellular contrast agent for
gadobutrol: an observational study. Radiology. 2017;285:434–444. magnetic resonance imaging. Eur J Radiol. 1995;21:1–10.
140. Kanda T, Fukusato T, Matsuda M, et al. Gadolinium-based contrast 154. OptiMARK Package Insert. St. Louis: Mallinckrodt, Inc.
agent accumulates in the brain even in subjects without severe renal 155. MultiHance Package Insert. Monroe, NJ: Bracco Diagnostics Inc.
dysfunction: evaluation of autopsy brain specimens with inductively 156. Ablavar Package Insert. North Billerica, MA: Lantheus Medical Imaging,
coupled plasma mass spectroscopy. Radiology. 2015;276:228–232. Inc.
141. Kanda T, Osawa M, Oba H, Toyoda K, Kotoku J, Haruyama T. High 157. Eovist Package Insert. Whippany, NJ: Bayer HealthCare Pharmaceuticals,
signal intensity in dentate nucleus on unenhanced T1-weighted MR Inc.
images: association with linear versus macrocyclic gadolinium chelate 158. Uggeri F, Aime S, Anelli PL, et al. Novel contrast agents for magnetic
administration. Radiology. 2015;275:803–809. resonance imaging. Synthesis and characterization of the ligand BOPTA
142. Murata N, Gonzalez-Cuyar LF, Murata K, et al. Macrocyclic and other and Its Ln(III) complexes (Ln = Gd, La, Lu). X-ray structure of
non–group 1 gadolinium contrast agents deposit low levels of disodium (TPS-9-145337286-C-S)-[4-carboxy-5,8,11-
gadolinium in brain and bone tissue preliminary results from 9 patients tris(carboxymethyl)-1-phenyl-2-oxa- 5,8,11-triazatridecan-13-oato(5-)]
with normal renal function. Invest Radiol. 2016;51:447–453. gadolinate(2-) in a mixture with its enantiomer. Inorg Chem.
143. Roberts DR, Lindhorst SM, Welsh CT, et al. High levels of gadolinium 1995;34:633–642.
deposition in the skin of a patient with normal renal function. Invest 159. de Haën C, Gozzini L. Soluble-type hepatobiliary contrast agents for
Radiol. 2016;51:280–289. MR imaging. J Magn Reson Imaging. 1993;3:1993.
4 
Myocardial Perfusion Imaging Theory
Michael Jerosch-Herold and Norbert Wilke

the territory of a normal coronary artery are significantly amplified


INTRODUCTION
with maximal vasodilation.
The concept of injecting a tracer into the blood stream and detecting Myocardial perfusion imaging during pharmacologic vasodilation
its transit and distribution in the heart muscle for the assessment of (e.g., with adenosine, dipyridamole, or regadenoson) rests on the physi-
myocardial perfusion is well established in nuclear cardiology and car- ologic observation that the hemodynamic significance of an epicardial
diovascular magnetic resonance (CMR). Both exogenous, injected contrast lesion is most apparent during maximal vasodilation.3,4 A related measure
agents and endogenous contrast mechanisms have been used to assess can be obtained in the catheterization laboratory with an intravascular
myocardial perfusion with CMR. The use of a gadolinium (Gd)-based Doppler flow probe by measuring the coronary flow reserve, to assess
contrast agents for the assessment of myocardial perfusion with CMR lesion severity,6 or by measuring the fractional flow reserve,7 defined
has been extensively validated and successfully applied in patient studies as mean distal coronary artery pressure divided by the aortic pressure
(see Chapter 18). Recent developments, in particular the introduction during maximal vasodilatation. These functional tests of coronary resis-
of parallel imaging, and sparse sampling methods (see Chapter 5), have tance and MBF overcome the known limitations of vessel lumen diameter
made it possible to combine the requirements for spatial and temporal measurements by x-ray angiography for determining the hemodynamic
resolution for myocardial perfusion imaging during the first pass, with significance of epicardial lesions.
multi-slice coverage of the heart. The need for a quantitative, largely Myocardial perfusion imaging can also be used to assess functional
observer-independent analysis of perfusion studies has also received impairments in the coronary microcirculation, when there are no epi-
increasing support.1 Several approaches have been developed that allow cardial lesions. Both myocardial perfusion reserve, and coronary flow
quantification of myocardial blood flow (MBF) with CMR. Advances reserve were found to be abnormally low in women with microvascular
in rapid imaging techniques for CMR perfusion imaging (covered in dysfunction, and without significant epicardial stenoses.8,9 There is also
detail in Chapter 5) and the development of methods to obtain repro- evidence from epidemiologic studies that it is feasible with CMR perfu-
ducible, quantitative measures of myocardial perfusion have established sion imaging to detect subclinical disease and silent ischemia in persons
CMR perfusion imaging as an attractive alternative to nuclear perfusion without a history or symptoms of atherosclerotic disease. The myocardial
imaging.2 perfusion reserve in response to adenosine was found to be associated
with coronary artery disease risk factors in patients with a low likeli-
THE PHYSIOLOGIC BASIS FOR MEASURING hood of significant epicardial disease.10 In asymptomatic patients a
lower myocardial perfusion reserve is associated with decreased regional
MYOCARDIAL PERFUSION left ventricular (LV) function11 and also with lower regional myocardial
Coronary circulation blood flow resistance is under normal conditions strain.12
determined primarily by the myocardial microcirculation, vessels with With the development of CMR perfusion imaging, it has become
a diameter less than 300 µm. The adequate supply of oxygen and metabo- possible to probe with sufficient spatial resolution for more subtle
lites to the myocytes is tightly coupled to MBF. The assessment of indicators of myocardial ischemia. Blood flow across the myocardial
microcirculatory function has therefore been used as a surrogate marker wall is not uniform but instead favors the subendocardium to accom-
for the detection of myocardial ischemia, although ischemic conditions modate the higher workload and higher rate of oxygen consumption
are also critically dependent on the workload and metabolic function. in the subendocardial layer.13,14 Under normal resting conditions the
For example, under conditions of myocardial hibernation, the degree ratio of endocardial to epicardial blood flow is on the order of 1.15 : 1.15
of ischemia is reduced by a corresponding reduction of the cardiac With a coronary artery stenosis, blood flow is first diverted away from
workload. Adequate and near-constant blood flow is maintained through the subendocardial layer and the endocardial-to-epicardial blood flow
autoregulation of coronary tone. Compensatory vasodilation can main- ratio is often <1 : 1, in particular when under stress.15,16 With myocar-
tain adequate myocardial blood under resting conditions for up to 80% dial ischemia, the subendocardial layer is accordingly most susceptible
diameter epicardial coronary artery narrowing.3,4 With more severe to necrosis. The transmural gradient of MBF can only be sufficiently
narrowing in an epicardial vessel, and in the absence of significant appreciated if the imaging modality provides spatial resolution on the
collateral flow, the distal perfusion bed is fully vasodilated even under order of ≤2 mm. With CMR it has become feasible to detect flow impair-
resting conditions, and no augmentation of blood flow is feasible during ments limited or most accentuated in the subendocardial layer.17,18 The
periods of increased demand (e.g., if the patient exercises or undergoes spatial resolution of conventional imaging modalities such as positron
pharmacologically induced stress). In healthy subjects, MBF can increase emission tomography (PET) and single photon emission computed
3- to 4-fold with maximal vasodilation.5 This means that differences tomography (SPECT) was insufficient to detect MBF deficits limited to
in MBF between a region subtended by a stenosed coronary artery and the subendocardial layer. More specifically, the sensitivity of a myocardial

40
CHAPTER 4  Myocardial Perfusion Imaging Theory 41

perfusion imaging technique is directly related to its spatial resolution, resolution of 2 to 3 mm. Because of the short TE, the signal is relatively
and the ability to discern transmural variations of flow.17,19 insensitive to flow and magnetic susceptibility variations (e.g., between
CMR offers the unique possibility of quantitatively assessing both tissue and blood pool during the first pass). With a linear ordering
perfusion, viability, and function with high accuracy, to distinguish, for the phase-encoding steps, the image acquisition only needs to be
stunned, hibernating, and infarcted myocardium. Bolli et al. showed delayed by 10 to 20  ms after a saturation recovery preparation. Depend-
that even small differences in blood flow during ischemia result in large ing on the effective time after saturation (TS) or time after inversion
differences in postischemic function, suggesting that the ability to quan- (TI, if an inversion pulse is used), the signal increases approximately
tify flow in the low-flow range is of importance to predict the probability linearly with contrast agent concentration, or as a function of the relax-
of postischemic recovery.20 The extent and incidence of microvascular ation rate constant. (Note that the relaxivity of the contrast reagent is
obstruction observed with CMR were associated with the duration of not appreciably different between blood and tissue.) Eventually the
ischemia before coronary intervention.21 signal ceases to increase because of the opposite effect of T2* on the
signal at higher contrast concentrations, and even in the case of neg-
FIRST-PASS IMAGING WITH ligible TEs (e.g., with image readouts with ultra-short TE) the signal
intensity from a proton density–weighted image represents an effec-
EXOGENOUS TRACERS tive upper limit for the signal intensity dynamic range. Fig. 4.1 shows
The success of applying CMR to detect the first pass of an injected an example of a CMR perfusion study with a spoiled gradient echo
contrast agent and detect perfusion abnormalities starts with an under- technique.
standing of the contrast mechanisms that allow contrast agent detection. The signal-to-noise (SNR) and the contrast-to-noise ratio (CNR)
The first-pass transit of a contrast agent through the vasculature and with gradient echo perfusion imaging are relatively low, when short
tissue leads to changes in the T1 and T2* relaxation time constants for TRs, and wide receiver bandwidths are used. The maximum signal
the detected 1H signal: the contrast reagent is not detected directly, but intensity is reached with relatively low flip angles. (The flip angle cor-
rather through its effects on the signal from 1H nuclei in tissue. The responding to maximum signal intensity is referred to as “Ernst angle.”)
chelates of paramagnetic Gd ions used as contrast reagents have a rela- To overcome this limitation, one can use a bSSFP image readout, which
tively high magnetic susceptibility, which causes on a microscopic scale maximizes the preservation of phase coherence of the transverse mag-
magnetic field inhomogeneities and shortens the transverse (T2*) relax- netization between repetition periods (TRs) in the pulse sequence, and
ation rate of water (see Chapter 3). The contrast reagent also shortens efficiently converts magnetization between transverse and longitudinal
the longitudinal (T1) magnetization recovery after a radiofrequency orientations. With bSSFP, the Ernst angle can approach 90 degrees—that
pulse has disturbed the equilibrium state. is, one can reach the maximum theoretical signal amplitude after each
CMR methods for perfusion imaging can be subdivided into T1- radiofrequency (RF) excitation, while still repeating the excitations with
and T2*-weighted techniques: T1-weighted techniques produce signal very short TRs. In fact, the TR should be as short as feasible, because
enhancement during the transit of the contrast agent, whereas T2* any off-resonance phase shifts increase signal loss, and phase shifts
techniques cause signal loss. For contrast agents confined to the vascular increase with TR, and also with flow.22 Off-resonance frequency shifts
bed, and in the absence of significant organ motion, T2*-weighted result in dark bands at locations where the frequency shift (in radians/s)
perfusion imaging gives rise to relatively larger signal changes than times TR (in seconds) equals an odd multiple of π/2.
T1-weighted techniques, because the magnetic susceptibility effects of The passage of a contrast bolus in the LV cavity can produce a
the contrast agents extend beyond the capillaries. But T2*-weighted frequency shift as large as approximately 100 Hz at 1.5 T and larger at
imaging techniques have an inherent sensitivity to motion, leading to higher field strengths. As the off-resonance shift varies with changing
signal loss in the presence of motion. For myocardial perfusion imaging, contrast loading of the blood pool during the first pass, it can induce
T1-weighted techniques such as gradient echo imaging with short echo signal variations over the myocardium, which can mimic perfusion
times, and a magnetization preparation for optimal T1 weighting have defects (not to be mistaken with dark-rim artifacts, which are discussed
therefore steadily gained preference in CMR. We provide here a brief later and which have a different appearance). At field strengths of ≥3 T,
overview of three major sequence techniques for T1-weighted perfusion bSSFP image readouts during first-pass imaging can give rise to quite
imaging of the heart. In historical order, they are spoiled gradient echo severe image artifacts, with dark bands appearing over the myocar-
imaging (e.g., “TurboFLASH” or “turbo field echo” [TFE]); gradient dium. The use of bSSFP readouts has therefore been mostly limited
echo imaging with balanced steady-state free precession (bSSFP); and for cardiac perfusion imaging to field strengths of ≤1.5 T. Although
multi-shot T1-weighted echo planar imaging. the bSSFP readouts can produce appealing image quality,23 its use has
Single-shot gradient echo imaging with a magnetization prepara- to be approached with caution as the artifacts on bSSFP images can
tion (e.g., TurboFLASH or TFE) is well suited for T1-weighted, quanti- be deceptive.
tative myocardial perfusion imaging. A magnetization preparation for Echo planer imaging (EPI) eliminates the need for RF excitation
T1 weighting can take the form of an inversion pulse or a saturation before each phase-encoding step, and, as a result, it is one of the fastest
pulse; either of them is generally applied in a non-slice-selective mode imaging methods for freezing heart motion (50–100 ms for single-
so that blood flowing into the image plane during the subsequent shot acquisitions). In an EPI pulse sequence, an initial RF excitation
image acquisition has been subjected to the same T1 preparation as creates a coherent precession of transverse magnetization, and a train
myocardium. In a sequential multi-slice imaging sequence, a satura- of gradient echoes is then generated by applying a rapidly oscillating
tion preparation can be repeated for each slice, and this will result in magnetic field gradient along the readout direction. Each of these gradi-
the same degree of T1 weighting for each slice. After the magnetiza- ent echoes is preceded by a short, blipped gradient pulse, applied along
tion preparation, images for one or more slices are rapidly read out a direction perpendicular to the oscillating magnetic field gradient. The
within approximately <200 ms per image, or even less depending on blipped phase-encoding gradients between echoes advance the trajec-
the imaging acceleration technique that is being used. Typical sequence tory in the frequency acquisition space (k-space) perpendicular to the
parameters for such rapid readouts are a repetition time per phase- readout direction. Instead of just a single line, one acquires after each
encoding step (TR) of ≤2.5 ms, an echo time (TE) of ≤1 ms, a receiver RF excitation a set of parallel lines in frequency space (“k-space”). The
bandwidth on the order of 400 to 700 Hz/pixel, and an in-plane spatial T2*-related decay of the gradient echo amplitudes in the echo train
42 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

300
LV
blood
pool 250

200

Signal intensity (a.u.)


150

100
LV
RV 50
Tissue
0

0 10 20 30 40 50 60 70 80 90
(IV contrast injection) Time (s)
FIG. 4.1  The gradient echo cardiovascular magnetic resonance (CMR) images depict the first pass of an
extracellular contrast agent (0.03 mmol/kg of gadodiamide/Omniscan, GE Healthcare) in a healthy subject,
with appearance of the contrast first in the right ventricle (RV), followed by the left ventricle (LV), and finally
leading to myocardial contrast enhancement. The contrast agent was injected after acquisition of approximately
four precontrast images. Images were acquired in the short-axis view at the level of the papillary muscle.
The arrows show the correspondence between images and the time course of changes in signal intensity
for a region of interest in the LV (circles) and a myocardial segment in the inferolateral wall (diamonds).
Characteristics of the signal curve for a tissue region such as its increased dispersion, lower amplitude and
reduced rate of signal enhancement compared with the ventricle result from transit through the coronary
microcirculation, where the volume of distribution is limited to either the extracellular or the intravascular space.

results in an increasing sensitivity of later echoes to T2* and motion


COMPARISON OF LV CURVES WITH
and causes blurring. The effective TE can be an order of magnitude 0.1 mmol/kg BOLUS OF Gd-DTPA
longer for EPI sequences compared with fast gradient echo sequences
(e.g., 30–40 ms vs. 1.2–2.0 ms for fast gradient echo imaging). Unfor- 1000
tunately, a longer effective TE gives rise to magnetic susceptibility and
flow artifacts in the LV cavity. Furthermore, flow- and susceptibility- 800 T1-FARM
related artifacts in the LV make it difficult to measure the arterial Fast gradient echo
600
transit of the contrast agent. To partially capture the speed advantage
Signal (a.u.)

of EPI, and to minimize T2*- and flow-related artifacts, one reduces the
400
length of the echo trains, and the images are read out with several RF
excitations/echo trains.24 This results in a 30% to 40% speed advantage
200
compared with conventional gradient echo sequences, and is particularly
useful at field strengths of ≤1.5 T. (At ≥3 T, any increase of echo time
0
beyond the minimum allowed by the gradient system nearly inevitably
gives rise to susceptibility artifacts, in particular during passage of a –200
contrast bolus.) 0 10 20 30 40 50
Ultra-fast T1 measurements (Fig. 4.2) would provide the most accu-
Time (s)
rate estimates of contrast agent concentration, and, with the advent of
FIG. 4.2  Comparison of signal time curves for left ventricular regions
parallel imaging, this approach may become practical. Chen et al. devel-
of interest obtained in a canine with quantitative T1 imaging (T1-FARM)
oped such a T1 Fast Acquisition Relaxation Mapping (T1-FARM) method and T1-weighted, saturation recovery prepared TurboFLASH (TR/TE
to obtain single-slice T1 maps of the heart with 1 s resolution.25,26 With = 2.4/1.2 ms; flip angle = 18 degrees) and a Gd-DTPA dosage of
a direct T1 measurement, the problems associated with the saturation 0.075 mmol/kg at 1.5 T. The curves were normalized so the Turbo-
of the signal with increasing contrast agent dosage can be avoided. This FLASH and T1-FARM recirculation peaks were equal. (From Z. Chen,
should allow for a more accurate quantification of blood flow using C.A. McKenzie, and F.S. Prato, St. Joseph’s Hospital, London, Ontario.)
CHAPTER 4  Myocardial Perfusion Imaging Theory 43

higher contrast agent dosages. A T1-based estimation of the contrast ENDOGENOUS CONTRAST FOR THE ASSESSMENT
concentration is particularly advantageous for the blood pool, as satura-
OF MYOCARDIAL PERFUSION
tion of the contrast enhancement is of foremost concern for the arterial
input. If instead one measures T1 in the blood pool during the first Approaches have been developed for an assessment of myocardial perfu-
pass of the contrast agent, the arterial input can be reconstructed from sion that are not based on the use of an exogenous MR contrast agent
these T1 data rather than the signal intensity because the latter suffers but instead exploit endogenous contrast mechanisms related to blood
from saturation effects. Chen et al. developed a slice-interleaved radial flow and blood oxygenation. These approaches fall under the categories
imaging technique,27 that runs continuously and can be used for recon- of spin labeling,28 blood-oxygen-level-dependent (BOLD) contrast,29,30
struction of cardiac phase-resolved images, and beat-by-beat estimation and magnetization transfer (MT) contrast.31,32 These techniques have
of T1 for arterial input sampling. to be considered technically more challenging than measurements with
In its current state, all CMR techniques offer higher spatial resolu- exogenous contrast agents, or offer only an indirect measure of blood flow.
tion than is feasible with nuclear imaging techniques. An illustration of With spin labeling, the spins are either inverted or saturated in a
this is shown in Fig. 4.3 from studies in an experimental animal model. slab that is generally located upstream from the imaged slice.33–35 The
flow-dependent change of signal intensity because of the inflow of
ADVANCED TECHNIQUES FOR PERFUSION saturated or inverted spins into the image slice provides a measure of
tissue perfusion. The spin-labeling method generally relies on the
IMAGING ACCELERATION assumption that the net arterial blood flow in the myocardium follows
Myocardial perfusion CMR has been at the forefront of developments the direction from base to apex. Cardiac motion complicates the inter-
in image acceleration techniques aimed at increasing coverage of the pretation of signal changes in a spin-labeling experiment because the
LV. Increased coverage can translate into a larger number of image labeled spins can be transported into the imaged slice either through
slices, or, by switching to three-dimensional image acquisitions, increase blood flow or by through-plane motion of the heart.
spatial coverage of the myocardium. The increased LV coverage should BOLD offers a measure of hemoglobin saturation reflecting regional
not come at the cost of lower temporal resolution—for example, by oxygen supply and demand. Deoxyhemoglobin is paramagnetic, whereas
increasing the repetition time for each slice location from one to two oxyhemoglobin is only diamagnetic, which means that deoxyhemoglobin
heart beats—because a lower temporal resolution will affect the accuracy causes a considerably larger reduction of T2* than oxyhemoglobin and
for distinguishing gradual changes in MBF, in particular during stress therefore a larger signal intensity attenuation in gradient echo images.
conditions. Chapter 5 of this book is dedicated to advanced techniques Deoxyhemoglobin can be used as an endogenous intravascular tracer
for CMR perfusion imaging that provide increased coverage of the because of the tight coupling between oxygen demand and blood flow.
LV, possibly higher in-plane spatial resolution, and suffer from fewer BOLD contrast changes have been observed in the heart after admin-
image artifacts (e.g., “dark-rim” artifact). This chapter will instead istration of dipyridamole and dobutamine.29,30 The link between BOLD
focus on the underlying theory and principles of CMR myocardial contrast and blood flow depends on the balance between blood flow
perfusion imaging. and oxygen metabolism—that is, between oxygen supply and demand.36

RV ANT
RV

CMR (1st pass) FDG-PET TTC-stained


FIG. 4.3  Images from cardiovascular magnetic resonance (CMR), positron emission tomography (PET),
and triphenyltetrazolium chloride (TTC) staining in a porcine model in which obtuse branches of the left
circumflex coronary artery had been ligated 4 weeks before combined CMR and PET studies. From CMR
images acquired during the first pass of an intravascular iron oxide contrast agent (NC100150 Injection,
Nycomed), the one shown at left corresponds to the highest peak signal enhancement in tissue and indicates
a subendocardial perfusion defect in the posterior segment, in agreement with TTC staining shown at right
13
NH2 PET (middle image) was carried out 3 hours before the CMR study and shows a fixed defect in the
inferolateral segment, suggesting a transmural infarct. FDG-PET images (not shown) also indicated irrevers-
ible damage in the posterior segment, in disagreement with the findings from CMR and TTC staining. ANT,
Anterior wall; RV, right ventricle.
44 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Spin labeling, BOLD, and MT contrast CMR have an advantage in With a fast T1-weighted gradient echo sequence and low contrast
that they can be carried out in the steady state, meaning there is no agent dosages (e.g., 0.03 mmol/kg of Gd-DTPA for an intravenous bolus
need to capture with ultra-fast imaging the transient signal changes injection into an antecubital vein) the myocardial contrast enhancement
observable in the myocardium after injection of an exogenous tracer. is approximately proportional to the local contrast agent concentration.
Ultimately these techniques may allow an assessment of myocardial Under these conditions the signal curves can be interpreted as, or trans-
perfusion with very high spatial resolution (~1 mm or less). formed into, contrast agent residue curves. Absolute units do not matter
here as much as a faithful reproduction of the relation in contrast
QUANTITATIVE EVALUATION OF concentrations between tissue and blood pool—enhancement in the
tissue is interpreted relative to the enhancement in the blood pool. In
MYOCARDIAL PERFUSION fact, the tissue is often, for the purpose of quantifying perfusion, viewed
The images acquired in a “first-pass” study of myocardial perfusion— as a linear, stationary system, where the enhancement in the tissue can
meaning rapid dynamic imaging during the first pass of an injected be modeled as a linear response to arterial input of contrast.
contrast bolus—can be qualitatively evaluated for differential signal Investigators using PET have over the years presented compelling
enhancement in the myocardium. With a T1-weighted imaging tech- arguments for a quantitative approach to myocardial perfusion imaging,
nique, myocardial segments showing reduced signal intensity enhance- including absolute quantification of MBF.37 With CMR, a quantitative
ment during the first pass, relative to other myocardial segments, are analysis of contrast enhancement is based on signal intensity curves,
interpreted as hypoperfused. This type of qualitative judgment of signal which can be generated for user-defined regions of interest (ROI), or
enhancement differences can suffer from substantial observer bias and at the pixel level. CMR perfusion studies require a careful approach for
small reductions in perfusion may be missed. Although higher dosages image segmentation and registration. CMR perfusion studies are not
of contrast agent would increase the dynamic contrast enhancement, motion-averaged, the endocardial borders are relatively well defined
and thereby render myocardial regions with reduced contrast enhance- during transit of contrast through the ventricular cavity, and the large
ment more apparent, the images are also more likely to be contaminated signal enhancement in the ventricular cavity requires accurate image
by artifacts near the endocardial border. Furthermore, global reductions segmentation to avoid spill-over effects. Fig. 4.4 shows an example of
of blood flow because of diffuse microvascular ischemia will be missed a CMR perfusion study in a patient for absolute quantification based
by this approach. It then becomes advantageous to evaluate the contrast on model-independent analysis of signal intensity curves.38
state during stress or vasodilation, relative to a baseline or resting state In the vein of Zierler’s39–41 original work on the central volume
to uncover global impairments of perfusion. principle, we consider here a tissue ROI with a single arterial input and

1.19 mL/g/min (lateral-posterior)


100
Myocardial SI (a.u.)

LAT

50

0.70 mL/g/min (anteriorseptal)


0
RV

A 10 20 30 40 50 60
Time (s)

600
LV SI (a.u.)

400
RV
200

0
B
10 20 30 40 50 60
C Time (s)
FIG. 4.4  A 71-year-old male patient had presented with a silent non-Q-wave anterior myocardial infarction.
(A) Image from a series of 60 dynamic images taken at rest shows the initial contrast enhancement in the
myocardium, with reduced contrast uptake in the anterior and anterior-septal segments because of an
eccentric 90% stenosis with a lesion length of 10 mm in the proximal left anterior descending (LAD) coronary
artery. (B) Late gadolinium enhancement was observed in anterior and anterior-septal segments (highlighted
by white arrow) from mid level to apex, but not at a more basal level including the level of the slice in shown
in A. (C) Myocardial blood flow was quantified by model-independent deconvolution of the myocardial signal
intensity (SI) curves, using an arterial input measured in the left ventricular (LV) blood pool. Blood flow at
rest in an anterior-septal segment (highlighted with arrow in A) was 0.7 mL/g/min, compared with 1.2 mL/g/
min in the inferolateral wall. The anterior and anterior-septal segments appeared hypokinetic on cine cardio-
vascular magnetic resonance (CMR). After CMR study, the patient was referred for coronary angiography.
During percutaneous coronary intervention, a drug-eluting stent was deployed in the proximal LAD. LAT,
Lateral; RV, right ventricle.
CHAPTER 4  Myocardial Perfusion Imaging Theory 45

a single venous output. The injection of a tracer is described by the extracellular Gd-based contrast agent is used. The Fermi model for
variation of tracer concentration at the (arterial) input cin(t). The amount the quantification of myocardial perfusion and the perfusion reserve
of tracer that remains in the tissue region at any time t, q(t), represents was validated by comparison to blood flow measurements with labeled
the difference between the amount supplied to the tissue region up to microspheres45,46 and also by comparison to invasive coronary flow reserve
time t, and the total amount of tracer that has exited the region of measurements.44
interest up to the time t: The Fermi model, although used in numerous studies, has some
well-known limitations. It was initially proposed by Axel43 for decon-
t
volution analysis of tissue contrast enhancement from an intravascular
q( t ) = F ∫ [cin ( s ) − cout ( s ) ] ⋅ dt Eq. 4.1
0
contrast agent. The shape of the Fermi function cannot reproduce
the delayed wash-out of contrast observed with an extracellular con-
where F represents the flow blood, for example, in units of mL/min/g trast agent, which results from leakage of contrast across the capillary
of tissue, and cout is the concentration at the output of the region of barrier into the interstitial space, and the subsequent relatively slow
interest. (We consider here a single arterial input and venous output clearance of contrast from the interstitial space—the interstitial space
to the ROI, but this overly restrictive model is assumed here only for volume in myocardial tissue is two to three times larger than the
the sake of simplifying the presentation. It can be shown that one can vascular space volume. In a CMR first-pass perfusion study this slow
generalize the theory to allow for multiple inputs and outputs.42) clearance of contrast from myocardial tissue manifests itself by the fact
Zierler showed that if the transport within a tissue ROI is assumed that after the first pass of the contrast in the arterial input, the signal
to be linear and stationary, then the response to an arbitrary arterial intensity in the myocardium shows little or a relatively small decline
input is given by the convolution of this input with the tissue impulse over the approximately 1- to 2-minute time frame that images are typi-
response41: cally collected in a cardiac perfusion study. To overcome this shortcom-
t ing of the Fermi function model for analyzing perfusion studies where
q( t ) = ∫ cin ( t − τ ) ⋅ R( τ )dτ Eq. 4.2 extracellular contrast agents are used, one can add a constant offset
0 term to the Fermi function that accounts for the persistent contrast
enhancement. This amounts to assuming that the contrast leaks into
We denote the impulse response by R(t). The response in a tissue region the interstitial space, but wash out is negligible.
to an impulse input, the tissue impulse response, has the useful property A more general approach has been developed that is limited to
that its initial amplitude equals the MBF through that region. This can imposing conditions on the impulse response that are consistent with
be seen from Eq. 4.1 by considering the special case when the input is the physiologic basis of the contrast enhancement:
a Dirac delta impulse, cin(t) = δ(t), and requiring that cout(t = 0) = 0, 1 The impulse response has to be a monotonically decaying function
that is, the tracer cannot instantaneously reach the output after injec- of time because, by definition of the impulse response, no additional
tion. This property of the impulse response is independent of the vascular tracer enters the tissue region after the initial contrast input impulse—
and compartmental structure inside the tissue ROI, or the properties after this initial impulse input of contrast, the concentration can
of barriers such as their permeability. For example, leakage of a contrast only remain the same or decline. This constraint by itself can still
agent from the capillaries into the interstitial space produces a redis- lead to impulse responses that show sudden step-size changes, which
tribution of the contrast agent within the ROI, but does not contribute is inconsistent in that the transport of contrast within the region
to flow in and out of the ROI, as long as transport by diffusion and of interest proceeds at steady and finite rates.
convection is much slower than by blood flow. For a CMR contrast 2 As a second condition, one can require that the impulse response
agent such as Gd-DTPA these assumptions hold up well, but for freely shape changes smoothly—that is, the aforementioned sudden changes
diffusible tracers they may not always apply. in shape are not consistent with the underlying steady-state transport
The central volume principle indicates that the blood flow can be processes in tissue. Smoothness of the impulse response follows to
determined from the impulse response amplitude. To obtain the impulse the degree that the magnitudes of flow, diffusion, and permeation
response one has to perform a deconvolution of the measured tissue in the heart impose constraints that exclude abrupt signal intensity
residue curve with the arterial input curve—that is, perform the inverse changes in myocardial tissue after contrast agent injection. To impose
of the convolution operation shown in Eq. 4.2. The deconvolution such smoothness constraints, one can represent the impulse response
analysis is quite sensitive to noise in the data, and one therefore needs as a sum of smooth cubic spline basis functions. The degree of
to constrain the deconvolution operation. A Fermi function has been smoothness is then determined by the number of spline components.
used as an empirical model for the impulse response to fit the first-pass The number of spline basis functions needs to be empirically deter-
portion of the signal curves.43,44 The Fermi model of the impulse response mined. For this purpose, it is useful to use simulated data (using
has the following functional form: the types of models discussed in the next section).38 One can then
verify whether with a certain number of spline basis function com-
A ponents one can estimate without a significant bias the myocardial
R( t ) = Eq. 4.3
1+ exp[−( t − w ) τ ] blood flow over a value range that covers the physiologic range of
blood flow in the myocardium. In this context it is particularly the
where A, w, and t are model parameters, with no direct physiologic maximum flow that is important because it is associated with the
meaning. From an empirical standpoint, the shape of the Fermi func- most rapid changes of contrast enhancement.
tion provides a reasonable approximation to the shape of the impulse The deconvolution algorithms that use these physiologic constraints
response of an intravascular tracer. The portion of the signal intensity without assuming a specific shape of the impulse response have been
curves up to the peak has higher sensitivity to flow changes than later termed “model-independent” deconvolution38 to distinguish them from
phases of the contrast enhancement, and the early enhancement is the approach where a parameterized representation of the impulse
relatively insensitive to capillary leakage of contrast agent. This part response (e.g., Fermi model) is used. The advantage of this more general
of the signal intensity curve can be approximated well by the Fermi approach is that it is far less sensitive to the choice of data window that
function model to assess blood flow, even when an extravascular, is chosen for deconvolution analysis.
46 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

As an alternative to the deconvolution analysis, one can build a dispersion of the contrast bolus will broaden the arterial input before
mathematical model of tracer transport through tissue. For an extracel- it enters a tissue region of interest. This is schematically illustrated in
lular contrast agent, this involves exchange of tracer between vascular Fig. 4.7. The degree of bolus dispersion during the transit through the
and interstitial spaces. For a compartmental model it is assumed that epicardial vessels can only be guessed or estimated from computational
the tracer is well mixed within each compartment, meaning that the models. Neglecting bolus dispersion will result in an underestimate of
contrast agent concentration equilibrates instantaneously within each the MBF, because the measured arterial input is narrower and has a
compartment. The rapid injection of a tracer will create a tracer con- higher peak value than the true input. The true input therefore produces
centration gradient from the arterial to the venous side. Spatially dis- less contrast enhancement in a myocardial segment than would be
tributed models are described by partial differential equations that expected for a certain tissue blood flow level, based on the measured
account for the variation of tracer concentrations within each tissue arterial input. Based on comparisons of estimates of MBF obtained
region as a function of time and at least one spatial variable.47,48 The with the CMR first-pass technique and estimates of blood flow from
number of parameters in these models tends to be rather large. Sensi- injected microspheres, one can arguably conclude that the neglect of
tivity analysis is needed to determine which (hopefully few) model the bolus dispersion does not appear to create a major bias to under-
parameters need to be adjusted for a best fit of the measured residue estimate MBF under normal conditions, unless both methods would
curves. The number of degrees of freedom can be reduced by using an suffer from systematic bias in the same direction. In patients with by-pass
intravascular contrast agent.48,49 The realism of spatially distributed grafts, any dispersion of the arterial input in the by-pass graft appears
models of blood tissue exchange is of advantage for the identification to be small enough that at least under resting conditions it does not
and quantification of physiologic changes observed indirectly with lead to significant bias of the MBF estimate.52 With a significant epi-
tomographic imaging techniques such as CMR. An example of the use cardial stenosis it is likely that the passage of the bolus through the
of a tracer kinetic model for analysis of a CMR perfusion study is shown vessel narrowing could lead to significant bolus dispersion. The uncer-
in Fig. 4.5, including an illustration of the central volume principle, tainties surrounding the true shape of the arterial input to a tissue ROI
based on model simulations. therefore continue to represent a significant limitation of “first-pass”
perfusion imaging. The increasing capabilities for imaging of the epi-
cardial vessels create an opportunity to combine this information about
ARTERIAL INPUT FUNCTION the vessel geometry with computational fluid-dynamics modeling to
Quantification of MBF generally requires the measurement of an arte- estimate the changes in the arterial input between LV or aorta, and
rial input function, which serves as reference for analysis of the myo- myocardial tissue ROIs. Arterial spin labeling, which represents a method
cardial contrast enhancement. Under ideal conditions the input function where the arterial input of blood can be “shaped” by the pulse sequence,
would be measured immediately before the blood enters the region in probably represents the conceptually most elegant approach to avoid
the myocardium that is being analyzed to determine the tissue blood uncertainties in the estimate of tissue blood flow that would result from
flow. With CMR it is not feasible to reliably measure the arterial input a poorly characterized arterial input.
even in the epicardial vessels. One therefore has to settle for a measure- With contrast-enhanced (CE) CMR one faces a further constraint
ment at a more upstream location, either near the aortic root, or in the in the limited dynamic range of contrast enhancement, which results
left ventricle. in signal saturation at higher contrast concentrations, as illustrated in
One consequence of measuring the arterial input at a relatively large Fig. 4.8A. Nevertheless, the dynamic range can be tuned to the con-
distance from the tissue region of interest is that the true arterial input centration range with the sequence parameters, and it is possible to
is delayed compared with the arterial input measured upstream (e.g., achieve approximate linearity between signal change (above precontrast
in left ventricular blood pool) from the myocardial region of interest. signal) and tracer concentration by using very short TI down to 5 to
This issue can be dealt with relatively easily by including a time-shift 10 ms using low-resolution imaging with centric k-space ordering of
operator, applied to the measured arterial input, in the analysis. The the phase encodings. However, the same tuning to short TI values is
value of the time-shift for the arterial input can be determined by not optimal for capturing the myocardial contrast enhancement, because
least-squares minimization—that is, by choosing the value for the time the tracer concentrations are lower than in the blood pool and the short
shift that gives the best agreement between the measured and calculated TI values decrease the sensitivity to small R1 changes. Fig. 4.8 shows
tissue curves. (In the case of the deconvolution analysis, this would be the effects of the delay time after saturation preparation on the measured
the tissue curve calculated by convolution of the estimated impulse contrast enhancement. There are essentially three solutions to the problem
response with the measured arterial input.) This arterial input delay of signal saturation:
parameter was found to be particularly noticeable in myocardial regions 1. Dual bolus technique: one performs two injections with a low and
that depend on collateral blood supply. In fact, an abnormally delayed a relatively higher contrast dosage, each optimized to measure con-
start of contrast enhancement in a myocardial segment, as shown in trast enhancement in the myocardium, and the blood pool respec-
the example from a patient study in Fig. 4.6, has been used to identify tively, but with otherwise identical injection protocols, and under
myocardial regions that depend significantly on blood supply through stable hemodynamic conditions.45 The signal curves for a blood
collateral vessels. In collateral-dependent segments the MBF under pool region of interest are scaled according to the ratio of contrast
resting conditions is often still within the normal range, and the delayed dosages used in the dual bolus protocol, and then further quantita-
arrival of contrast may provide the only indication of collaterization.50 tive analysis can be performed on curves for tissue and blood pool
The presence and extent of collateral circulation in the heart are associ- with optimized CNR.
ated with lower mortality in patients with chronic coronary artery 2. As a second alternative one can include in the pulse sequence a rapid
disease.51 Therefore the assessment of the collateral circulation by CMR low-resolution image acquisition with very short inversion time
perfusion imaging may find some clinical utility. to measure the arterial input with minimal signal saturation.53,54
A more significant challenge for obtaining an accurate measure of Such “dual contrast” pulse sequence techniques with arterial input
the arterial input is the fact that during the transit of the arterial input scout impose a small time penalty, but otherwise show promise
from the measurement location (e.g., the center of the LV blood pool of keeping the acquisition protocol simple and not requiring dual
at the basal level) through the epicardial vessels one can expect that bolus injections.
CHAPTER 4  Myocardial Perfusion Imaging Theory 47

BLOOD-TISSUE EXCHANGE UNIT

Axial direction (x) Blood plasma flow (Fp)

Plasma volume = Vp
PScap

Interstitial space

Parenchymal cell
A

50
LV input
40
Signal intensity (a.u.)

30
4 mL/g/min
20

10
1 mL/g/min
0

0 10 20 30 40 50 60 70
B Time (s)

200

5 mL/g/min
Signal intensity (a.u.)

Blood flow (F) ~ impulse amplitude


150
4 mL/g/min

100
2.7 mL/g/min
2 mL/g/min
50
1 mL/g/min
0
0 10 20 30 40 50 60 70
C Time (s)
FIG. 4.5  (A) Conceptual diagram of the spatially distributed, two-region model that was implemented in
JSim simulation environment (http://www.physiome.org/jsim/) for analysis of tracer residue curves and quan-
tification of myocardial blood flow. The following model parameters are optimized for fitting to experimental
data: plasma flow (Fp), plasma volume (Vp), and capillary permeability surface area product (PScap). (B) A tissue
contrast residue curve measured in a 56-year-old healthy woman during vasodilation with adenosine (circles)
was analyzed with the model described in A, and the best fit corresponding to a tissue blood flow of 2.8 mL/g/
min is shown as a solid line. Additional model curves for flows of 1, 2, and 4 mL/g/min, and otherwise
unchanged model parameters are shown as dashed lines, to illustrate the sensitivity of the initial contrast
enhancement to blood flow. The rate of the initial contrast enhancement (“upslope”) changes as the flow
is varied, whereas the curves cluster together after the first pass, when sensitivity to flow changes is low.
Also note that for equal increments of blood flow, the peak contrast enhancement changes less as the
absolute magnitude of flow increases. (C) The measured arterial input was replaced by an impulse input,
with approximately the same area under the curve. Now the amplitude of the simulated tissue response to
the impulse input increases in proportion to the blood flow (F ~ impulse amplitude), as predicted by Zierler’s
central volume principle. LV, Left ventricular.
48 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

3. Finally, it is often feasible to construct calibration curves of signal echogenic bubbles that can be destroyed by ultrasound pulses. For CMR,
versus R1, or tracer concentration, to correct for signal saturation,55 hyperpolarized contrast media56 could be one possible approach because
as long as the calibration curves are monotonically increasing as a it is relatively easy to selectively quench the hyperpolarized signal.
function of R1. An example of calibration-based saturation correc-
tion appears in Fig. 4.8C. PRACTICAL ASPECTS OF MAGNETIC RESONANCE
It may be possible in the future to control the arterial input of an
injected contrast agent in a manner similar to the approach used by
PERFUSION IMAGING
arterial spin labeling, or in myocardial contrast echocardiography with This chapter has mostly focused on theoretical aspects of myocardial
perfusion imaging, and the presentation was in part motivated by a
30 desire to provide useful insights for optimizing myocardial perfusion
Delay through collaterals studies, and for avoiding some pitfalls. We now summarize below some
practical rules of thumb for myocardial perfusion imaging protocols
20 Anterior segment that have proven to be of relevance in practice.
1. Contrast agent injection. Because of the dependence of myocardial
Signal (a.u.)

contrast enhancement on the arterial input of contrast, it is important


10 to inject the tracer sufficiently fast so that different levels of tissue
blood flow can be sensitively discriminated. The use of a power
injector with an injection of rate of approximately >3 mL/s, especially
0
Posterior- for studies during coronary hyperemia, is important.
lateral
2. Temporal resolution of measurements. The temporal resolution
−10 should allow updating of images from every slice at a rate equal to
0 10 20 30 40 50 the heart rate. This rule of thumb automatically compensates for
Time (s) the higher temporal resolution requirements during stress, compared
FIG. 4.6  A male patient with an occluded right coronary artery on with at rest.
catheter-based angiography and a right-dominant coronary distribution. 3. Spatial resolution. Mild reductions of MBF because of an epicardial
A cardiovascular magnetic resonance first-pass perfusion study during lesion manifest themselves first in the endocardial layer. A spatial
resting conditions showed a significantly delayed contrast enhancement
resolution of ≤2.5 mm should be used to resolve transmural varia-
in the inferolateral and inferior segments (blue circles), compared with
contrast enhancement in the anterior segment (red squares) supplied
tions in blood flow. For rest studies the temporal resolution can be
by the left coronary artery. A quantitative analysis showed that resting reduced from 1 R-to-R interval to 2 R-to-R intervals, to accommodate
blood flow was similar in these segments, suggesting that during resting the requirements for spatial resolution, but for hyperemia the 1
conditions the delay in the arrival of the arterial input of contrast was R-to-R interval tends to be closer to the critical limit for temporal
an important marker for identifying collateral-dependent segments. resolution.

AIF dispersion
0.14

0.12

0.10
SI (a.u.)

0.08

0.06

0.04

0.02
AIF sampling 0.00
in LV 0 10 20 30 40 50 60
Time (s)
LV or Ao
BTEX Venule
Arteriole Vein
Artery BTEX Venule
Arteriole
Tissue ROI
FIG. 4.7  Conceptual illustration of the change in the shape of the arterial input between the location where
it is routinely measured in cardiovascular magnetic resonance perfusion studies (e.g., at basal level in the
center of the left ventricular [LV] blood pool) and the tissue region of interest (ROI) where perfusion is quanti-
fied with the measured arterial input. Ideally one would use the true immediate arterial input to the ROI,
which, because of dispersion, is broader and has a lower peak value than the arterial input measured in the
LV blood pool. Neglect of the described dispersion of the arterial input can lead to a systematic underestimate
of myocardial blood flow. Ao, Aorta; AIF, arterial input function; BTEX, blood-tissue exchange unit.
CHAPTER 4  Myocardial Perfusion Imaging Theory 49

MODEL-BASED CALIBRATION CURVE

1200 3000

Expected CE (%)
TR/TE = 2.3/1.0 ms
Flip angle = 18o 2000

Contrast enhancement (CE) (%)


1000
TD = 10
1000
800
0
600 TD = 100 0 200 400 600 800 1000 1200 1400
B Observed CE (%)
400 TD = 300 ms
1000
200
500

CE (%)
0
0 Measured LV CE
0 2 4 6 8 10 Corrected LV CE
A R1 (s) 500
0 10 20 30 40 50
C Image #
FIG. 4.8  (A) Model-based calculations based on the Bloch equations were used to calculate the contrast
enhancement in the left ventricular (LV) blood pool for a range of relaxation rate constants (R1), and assum-
ing a precontrast T1 value of 1100 ms at 1.5 T. The delay time (TD) between saturation preparation and
gradient echo image readout was varied between 10 and 300 ms and had a marked effect on the nonlinearity
of the contrast enhancement curves. Short TD values are most appropriate for measuring contrast enhance-
ment for a wide range of R1 values, as encountered during the first pass of a contrast agent in the blood
pool. Longer TD values result in better contrast to noise. The blue line corresponds to the ideal case where
contrast enhancement is linearly proportional to R1, and contrast agent concentration. (B) Model-based cali-
bration curves can be used to convert the observed contrast enhancement to the ideal case where contrast
enhancement is linearly proportional to contrast concentration. Such calibration curves can be generated
from the simulations shown in A, using the sequence parameters and precontrast T1 values of an actual
study. (C) With the model-based calibration curve in B, a measured curve of LV contrast enhancement was
corrected for saturation effects. The LV contrast enhancement was measured with a bolus of 0.05 mmol/
kg of Gd-DTPA (gadopentetate dimeglumine/Magnevist, Bayer Healthcare, Whippany, NJ). With this dosage,
the peak signal measured in the LV shows approximately 30% saturation. The contrast concentration change
in the blood pool would be underestimated by 30% if the measured signal were taken at face value and not
corrected for saturation.

4. CMR sequence for contrast residue detection. T1-weighted imaging factors on the order of 2 are recommended, although higher factors
can be rendered relatively insensitive to the effects of water exchange may work well when SSFP techniques are used.
by use of higher flip angles (above the Ernst angle) and this simpli- 7. Analysis and modeling. The rate of contrast enhancement in a myocar-
fies the conversion of signal time curves into residue curves. There dial tissue region (“upslope”) has empirically been proven to provide
is some evidence that avoidance of high contrast dosages (e.g., a relatively good surrogate measure of blood flow, but it should be
>0.1 mmol/kg for Gd chelates) and high spatial resolution may be appropriately normalized by a measure of contrast enhancement at
effective measures against artifacts, and the appearance of a transi- the arterial input.57 The resulting perfusion index can be determined
tional dark rim at the endocardial border. The rationale for these for rest and stress, and the ratio can provide an approximate measure
measures is that (a) the susceptibility difference between blood loaded of the perfusion reserve. Several CMR equipment vendors have now
with contrast and tissue gives rise to signal loss at the endocardial included this upslope method in their cardiac perfusion analysis
border, which is less likely to be observed with lower contrast dosages packages. Other parameters, such as the maximum amplitude of
and (b) Gibbs ringing, which may be one source of the dark rim contrast enhancement, are less sensitive than the upslope parameter
artifact, is reduced by increasing the number of sampling points, to changes in tissue blood flow. Absolute measures of MBF can
that is, the spatial resolution in this case. be determined if contrast enhancement is linearly proportional to
5. Measurement of arterial input. For a semiquantitative or quantita- contrast concentration in blood. Blood flow estimates are obtained
tive analysis, contrast dosages need to be kept below approximately by deconvolution of the tissue curves with the arterial input, either
0.05 mmol/kg body weight for Gd chelates such as gadopentetate by using a model-independent approach or by modeling. Currently
dimeglumine/Magnevist, or gadodiamide/Omniscan, and this may these latter approaches for blood flow quantification are used only
also be recommended on the basis of recent safety concerns. in a research setting, and commercial software is not yet available.
6. Parallel imaging. For near-complete coverage of the heart, the use of
parallel imaging techniques has become a blessing and necessity. Nev-
ertheless, myocardial perfusion studies are relatively signal-intensity
CONCLUSIONS
“starved,” and for this reason acceleration factors have to be used In the context of assessing blood flow in the myocardial microcirculation,
conservatively to avoid excessive noise and blurring. Acceleration the CMR first-pass imaging technique can address several clinical issues,
50 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

if the appropriate conditions for spatial and temporal resolution can be


ACKNOWLEDGMENT
met. Valuable, quantitative, and largely observer-independent informa-
tion related to the pathophysiology of ischemic heart disease becomes The authors would like to express their deep gratitude to the late Keith
available with myocardial perfusion imaging. CMR perfusion reserve Kroll, PhD, and James Bassingthwaighte, MD, PhD, at the National
measurements can be applied to assess the degree of atherosclerosis, Simulation Resource, University of Washington, Seattle for many enrich-
the functional significance of coronary artery lesions, and to evaluate ing and instructive discussions, and their interest and support of our
indirectly the degree of ischemia. In milder forms of ischemic heart endeavors in quantitative CMR perfusion imaging.
disease and ischemic cardiomyopathies, CMR at rest and during stress is
a good test to probe for subtle perfusion limitations that may be limited
to subendocardium or that have only resulted in moderate blood flow
reductions. For these applications, quantitative CMR methods have been
REFERENCES
successfully developed and validated, and provide a true measure of MBF. A full reference list is available online at ExpertConsult.com
CHAPTER 4  Myocardial Perfusion Imaging Theory 50.e1

REFERENCES after primary angioplasty: a study performed with contrast-enhanced


magnetic resonance. J Am Coll Cardiol. 2005;46(7):1229–1235.
1. Stollfuss JC, Kosa I, Ziegler S, Schwaiger M. Quantitative assessment of 22. Li W, Storey P, Chen Q, Li BS, Prasad PV, Edelman RR. Dark flow
myocardial perfusion: is it of clinical relevance? Q J Nucl Med. artifacts with steady-state free precession cine MR technique: causes and
1996;40(1):76–84. implications for cardiac MR imaging. Radiology. 2004;230(2):569–575.
2. Schwitter J, Wacker CM, Wilke N, Al-Saadi N, Hoebel N, Simor T. 23. Hunold P, Maderwald S, Eggebrecht H, Vogt FM, Barkhausen J.
Magnetic resonance Imaging for Myocardial Perfusion Assessment in Steady-state free precession sequences in myocardial first-pass perfusion
Coronary artery disease Trial (MR-IMPACT II): a phase III multicenter, MR imaging: comparison with TurboFLASH imaging. Eur Radiol.
multivendor trial comparing perfusion cardiac magnetic resonance versus 2004;14(3):409–416.
single photon emission computed tomography for the detection of 24. Epstein FH, Wolff SD, Arai AE. Segmented k-space fast cardiac imaging
coronary artery disease. Circulation. 2006;114(18):II-806. using an echo-train readout. Magn Reson Med. 1999;41(3):609–613.
3. Gould KL, Kirkeeide RL, Buchi M. Coronary flow reserve as a physiologic 25. Chen Z, Prato FS, McKenzie CA. T1 Fast Acquisition Relaxation Mapping
measure of stenosis severity. J Am Coll Cardiol. 1990;15(2):459–474. (T1-FARM): An Optimised Reconstruction. IEEE Trans Med Imag.
4. Gould KL, Lipscomb K. Effects of coronary stenosis on coronary flow 1998;17:155–160.
reserve and resistance. Am J Cardiol. 1974;34:48–55. 26. Bellamy DD, Pereira RS, McKenzie CA, Prato FS, Drost DJ, Sykes J.
5. Wilson RF, Wyche K, Christensen BV, Zimmer S, Laxson DD. Effects of Wisenberg G. Gd-DTPA bolus tracking in the myocardium using T1 fast
adenosine on human coronary arterial circulation. Circulation. 1990; acquisition relaxation mapping (T1 FARM). Magn Reson Med.
82(5):1595–1606. 2001;46(3):555–564.
6. Wilson RF. Assessment of the human coronary circulation using a 27. Chen D, Sharif B, Bi X, et al. Quantification of myocardial blood flow
Doppler catheter. Am J Cardiol. 1991;67(14):44D–56D. using non-electrocardiogram-triggered MRI with three-slice coverage.
7. Pijls NH, De Bruyne B, Peels K, et al. Measurement of fractional flow Magn Reson Med. 2016;75(5):2112–2120.
reserve to assess the functional severity of coronary-artery stenoses [see 28. Fidler F, Wacker CM, Dueren C, et al. Myocardial perfusion
comments]. NEJM. 1996;334(26):1703–1708. measurements by spin-labeling under different vasodynamic states. J
8. Panting JR, Gatehouse PD, Yang GZ, et al. Abnormal subendocardial Cardiovasc Magn Reson. 2004;6(2):509–516.
perfusion in cardiac syndrome X detected by cardiovascular magnetic 29. Fieno DS, Shea SM, Li Y, Harris KR, Finn JP, Li D. Myocardial perfusion
resonance imaging. N Engl J Med. 2002;346(25):1948–1953. imaging based on the blood oxygen level-dependent effect using
9. Wilke N, Jerosch-Herold M, Wang Y, et al. Myocardial perfusion reserve: T2-prepared steady-state free-precession magnetic resonance imaging.
assessment with multisection, quantitative, first-pass MR imaging. Circulation. 2004;110(10):1284–1290.
Radiology. 1997;204(2):373–384. 30. Klocke FJ, Li D. Testing coronary flow reserve without a provocative
10. Wang L, Jerosch-Herold M, Jacobs DR Jr, Shahar E, Folsom AR. Coronary stress. A “BOLD” approach. J Am Coll Cardiol. 2003;41(5):841–842.
risk factors and myocardial perfusion in asymptomatic adults: the 31. Tsekos NV, Wilke N, Xu Y, Merkle H, Ugurbil K. Magnetization transfer
Multi-Ethnic Study of Atherosclerosis (MESA). J Am Coll Cardiol. contrast in assesment of myocardial perfusion. Radiology. 1993;189(P):
2006;47(3):565–572. 166.
11. Rosen BD, Lima JA, Nasir K, et al. Lower myocardial perfusion reserve is 32. Wolff SD, Balaban RS. Magnetization transfer contrast (MTC) and
associated with decreased regional left ventricular function in tissue water proton relaxation in vivo. Magn Reson Med. 1989;10:
asymptomatic participants of the multi-ethnic study of atherosclerosis. 135–144.
Circulation. 2006;114(4):289–297. 33. Detre JA, Zhang W, Roberts DA, et al. Tissue specific perfusion imaging
12. Rosen BD, Edvardsen T, Lai S, et al. Left ventricular concentric remodeling using arterial spin labeling. NMR Biomed. 1994;7(1–2):75–82.
is associated with decreased global and regional systolic function: the 34. Wacker CM, Fidler F, Dueren C, et al. Quantitative assessment of
Multi-Ethnic Study of Atherosclerosis. Circulation. 2005;112(7):984–991. myocardial perfusion with a spin-labeling technique: preliminary results
13. Decking UK, Schrader J. Spatial heterogeneity of myocardial perfusion in patients with coronary artery disease. J Magn Reson Imaging.
and metabolism. Basic Res Cardiol. 1998;93(6):439–445. 2003;18(5):555–560.
14. Arai AE, Grauer SE, Anselone CG, Pantely GA, Bristow JD. Metabolic 35. Zhang H, Shea SM, Park V, et al. Accurate myocardial T1 measurements:
adaptation to a gradual reduction in myocardial blood flow. Circulation. toward quantification of myocardial blood flow with arterial spin
1995;92(2):244–252. labeling. Magn Reson Med. 2005;53(5):1135–1142.
15. Bache RJ, Schwartz JS. Effect of perfusion pressure distal to a coronary 36. Bauer WR, Nadler W, Bock M, et al. The relationship between the
stenosis on transmural myocardial blood flow. Circulation. BOLD-induced T(2) and T(2)(*): a theoretical approach for the
1982;65(5):928–935. vasculature of myocardium. Magn Reson Med. 1999;42(6):1004–1010.
16. Canty IJ, Piotrowski B, Klocke F. Relative reductions in subendocardial 37. Schwaiger M. Myocardial perfusion imaging with PET. J Nucl Med.
and full-thickness flow during pharmacologic vasodilation as an index of 1994;35(4):693–698.
functional stenosis seventy. J Am Coll Cardiol. 1990;15:182A. 38. Jerosch-Herold M, Swingen C, Seethamraju RT. Myocardial blood flow
17. Schwitter J, Nanz D, Kneifel S, et al. Assessment of myocardial perfusion quantification with MRI by model-independent deconvolution. Med
in coronary artery disease by magnetic resonance: a comparison with Phys. 2002;29(5):886–897.
positron emission tomography and coronary angiography. Circulation. 39. Zierler K. Indicator dilution methods for measuring blood flow, volume,
2001;103(18):2230–2235. and other properties of biological systems: a brief history and memoir.
18. Muehling OM, Panse P, Jerosch-Herold M, et al. Cardiac magnetic Ann Biomed Eng. 2000;28(8):836–848.
resonance perfusion imaging identifies transplant arteriopathy by a 40. Zierler KL. Theoretical basis of indicator-dilution methods for measuring
reduced endomyocardial resting perfusion. J Heart Lung Transplant. flow and volume. Circ Res. 1962;10:393–407.
2005;24(8):1122–1123. 41. Zierler KL. Equations for measuring blood flow by external monitoring
19. Wagner A, Mahrholdt H, Holly TA, et al. Contrast-enhanced MRI and of radioisotopes. Circ Res. 1965;16:309–321.
routine single photon emission computed tomography (SPECT) 42. Keeling SL, Bammer R, Stollberger R. Revision of the theory of tracer
perfusion imaging for detection of subendocardial myocardial infarcts: an transport and the convolution model of dynamic contrast enhanced
imaging study. Lancet. 2003;361(9355):374–379. magnetic resonance imaging. J Math Biol. 2007;55(3):389–411.
20. Bolli R, Zhu W-X, Thornby JI, O’Neill PG, Roberts R. Time course and 43. Axel L. Tissue mean transit time from dynamic computed tomography by
determinants of recovery of function after reversible ischemia in a simple deconvolution technique. Invest Radiol. 1983;18(1):94–99.
conscious dogs. Am J Physiol. 1988;254:H102–H114. 44. Jerosch-Herold M, Wilke N, Stillman AE, Wilson RF. MR quantification
21. Tarantini G, Cacciavillani L, Corbetti F, et al. Duration of ischemia is a of the myocardial perfusion reserve with a Fermi function model for
major determinant of transmurality and severe microvascular obstruction constrained deconvolution. Med Phys. 1998;25(1):73–84.
50.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

45. Christian TF, Rettmann DW, Aletras AH, et al. Absolute myocardial 52. Arnold JR, Francis JM, Karamitsos TD, et al. Myocardial perfusion
perfusion in canines measured by using dual-bolus first-pass MR imaging after coronary artery bypass surgery using cardiovascular
imaging. Radiology. 2004;232(3):677–684. magnetic resonance: a validation study. Circ Cardiovasc Imaging.
46. Mühling OM, Wang Y, Panse P, et al. Transmyocardial laser 2011;4(3):312–318.
revascularization preserves regional myocardial perfusion: an MRI first 53. Köstler H, Ritter C, Baunach D, et al Determination of the Arterial Input
pass perfusion study. Cardiovasc Res. 2003;57(1):63–70. Function in High Dose Radial Myocardial Perfusion Imaging. Paper
47. Bassingthwaigthe JB, Raymond GR, Chan JIS. Principles of tracer presented at: Scientific Sessions of the International Society of Magnetic
kinetics. In: Zaret BL, Beller GA, eds. Nuclear Cardiology: State of the Art Resonance in Medicine, 2005; Miami Beach, FL.
and Future Directions. St. Louis: Mosby-Year Book; 1993:3–23. 54. Gatehouse PD, Elkington AG, Ablitt NA, Yang GZ, Pennell DJ, Firmin
48. Kroll K, Wilke N, Jerosch-Herold M, et al. Accuracy of modeling of DN. Accurate assessment of the arterial input function during high-dose
regional myocardial flows from residue functions of an intravascular myocardial perfusion cardiovascular magnetic resonance. J Magn Reson
indicator. Am J Physiol. 1996;40:H1643–H1655. Imaging. 2004;20(1):39–45.
49. Jerosch-Herold M, Wilke N, Wang Y, et al. Direct comparison of an 55. Cernicanu A, Axel L. Theory-based signal calibration with single-point
intravascular and an extracellular contrast agent for quantification of T1 measurements for first-pass quantitative perfusion MRI studies. Acad
myocardial perfusion. Int J Card Imaging. 1999;15(6):453–464. Radiol. 2006;13(6):686–693.
50. Muehling OM, Huber A, Cyran C, et al. The delay of contrast arrival in 56. Johansson E, Mansson S, Wirestam R, et al. Cerebral perfusion
magnetic resonance first-pass perfusion imaging: a novel non-invasive assessment by bolus tracking using hyperpolarized 13C. Magn Reson Med.
parameter detecting collateral-dependent myocardium. Heart. 2004;51(3):464–472.
2007;93(7):842–847. 57. Al-Saadi N, Nagel E, Gross M, et al. Noninvasive detection of myocardial
51. Seiler C, Engler R, Berner L, et al. Prognostic relevance of coronary ischemia from perfusion reserve based on cardiovascular magnetic
collateral function: confounded or causal relationship? Heart. resonance. Circulation. 2000;101(12):1379–1383.
2013;99(19):1408–1414.
5 
Myocardial Perfusion Cardiovascular
Magnetic Resonance: Advanced Techniques
Manish Motwani and Sven Plein

The physiologic basis for and the general principles of myocardial per- methods that are needed for high-resolution and 3D perfusion CMR
fusion cardiovascular magnetic resonance (CMR) are covered in detail also have specific limitations and are not necessarily available outside
in other chapters. The key challenge of first-pass dynamic contrast- of research institutions. This chapter gives a brief overview of the prin-
enhanced acquisition is to capture data with high temporal and spatial ciples of advanced acceleration in perfusion CMR, and summarizes
resolution during the myocardial contrast passage. The recommended the current evidence for high spatial resolution and 3D whole-heart
minimal requirements for standard perfusion CMR are to acquire data acquisitions.
in three short-axis sections of the heart according to criteria defined
by the American Heart Association (AHA) at every R-R interval and
with an in-plane spatial resolution of at least 3 × 3 mm. These require-
ACCELERATION METHODS
ments are most commonly met with saturation recovery fast gradient Most of the studies reporting high-resolution or 3D perfusion CMR
echo pulse sequences combined with spatial undersampling methods have used techniques based on “prior-knowledge” to derive the required
or echo planar imaging to accelerate data acquisition by 2- to 3-fold. acceleration factors for data acquisition. Prior-knowledge methods are
The standardized CMR protocols 2013 update by the Society for Car- based on the observation that image datasets exhibit considerable cor-
diovascular Magnetic Resonance (SCMR) recommends a basic technique relation in space and time.2,4 Perfusion CMR datasets contain a high
for perfusion CMR1 (Table 5.1). degree of temporal redundancy, because data are acquired at a single
time point in the cardiac cycle using electrocardiogram (ECG)-gating
and during breath-holding—this means that most of the image is
ADVANCED TECHNIQUES static and the predominant change between neighboring time frames
The need for higher spatial and temporal resolution in dynamic CMR is related to the passage of contrast. This image redundancy can be
has prompted the development of several dedicated imaging speed-up taken advantage of by undersampling data in the time (t) domain as
techniques. Effects on signal-to-noise ratio (SNR) and increasing artifacts well as the more conventional undersampling in the spatial (k-space)
limit the practically achievable acceleration to approximately 2- to 3-fold domain.2–4 With some of the acceleration schemes discussed later, data
using standard acceleration techniques such as echo planar imaging or acquisition can be accelerated up to a factor of 10 times. This level of
parallel imaging. However, data acquisition can be further accelerated acceleration has been used to improve spatial resolution (high-resolution
with methods that exploit both temporal and spatial correlations capi- perfusion CMR), increase spatial coverage within a single acquisi-
talizing on significant spatiotemporal redundancy within imaging data, tion shot (3D whole-heart perfusion CMR) or to achieve a mixture
which is particularly relevant to first-pass perfusion imaging.2–5 Several of both.6,8,11,12,14,15
of these approaches have been used to improve in-plane spatial resolu-
tion of myocardial perfusion CMR with reductions in dark rim artifact, k-t Undersampling Techniques
better detection of subendocardial ischemia, and possible improvement The k-t broad linear acquisition speed-up technique (k-t BLAST) is
in diagnostic accuracy versus standard resolution acquisition (Figs. 5.1 a prior-knowledge spatiotemporal (k-t) undersampling technique
and 5.2).6–11 Others have used the speed-up to increase spatial coverage that has gained common use. In k-t BLAST, undersampling is applied
through acquisition of more than 3 slices or even whole-heart three- along k-space and time, while a low spatial resolution image (“train-
dimensional (3D) acquisition, which allows more reliable calculation ing data”) is obtained in an interleaved fashion during the acquisi-
of the total myocardial ischemic burden (MIB), akin to nuclear myo- tion.17 A non­aliased, full image series is then reconstructed using prior
cardial perfusion imaging (MPI) (Fig. 5.3).5,12–15 An additional benefit knowledge derived from the training data (Fig. 5.4). The signal alias-
of 3D perfusion CMR is that it allows the acquisition of all slices at the ing is resolved by an adaptive filtering process in which the aliased
same time point in the cardiac cycle—for example, in mid-diastole or signal is distributed according to a low-resolution estimate of the
end-systole—so that motion artifacts can be reduced and registration signal covariance, as derived from the training images. A particular
between slices improved.16 advantage of this approach is that it allows some overlapping of the
However, although both high-resolution and 3D whole-heart perfu- aliased object, thereby permitting higher acceleration factors. However,
sion CMR methods have been shown to be feasible and to have high the inherent temporal filtering reduces temporal fidelity, which can
diagnostic accuracy, most clinical studies to date have had small sample lead to reconstruction errors or limit the reduction in data acquisition
sizes and were conducted in single centers. Furthermore, direct com- actually achieved.
parison between standard and advanced methods are largely lacking, A further limitation of k-t BLAST is that the reconstruction problem
so that it remains unclear whether their theoretical benefits translate is intrinsically underdetermined (i.e., there are fewer equations than
into altered patient management. The more advanced acceleration unknowns), which is a reflection of its dependence on the estimated

51
52 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

signal covariance from training data. One method of improving this is to reduces highly dimensional datasets to lower dimensionality by extract-
incorporate sensitivity encoding information into the reconstruction— ing and exploiting relevant correlations within the data. The extension
this is the k-t SENSE method.18 A further solution, which allows the use of k-t BLAST or k-t SENSE methods using PCA in the reconstruction
of higher acceleration factors, is to constrain the reconstruction using is referred to as k-t PCA.19
a standard data compression technique, called principal component In the k-t PCA method, the adaptive filter used to remove aliasing
analysis (PCA)—this is a commonly used mathematical algorithm that is improved by applying PCA to the training data from which the filter
is derived. Effectively, PCA is used to transform the images into a new
mathematical domain of temporal “basis function” more suitable for
TABLE 5.1  Basic Perfusion CMR Technique reconstruction. The advantage of this new mathematical domain is that
it is sparser, even in cases of nonperiodic motion such as respiration
Pulse sequence Saturation recovery imaging with
or misgating, and therefore the core perfusion data is contained within
readout:
a few principal components and the rest can be discarded. This math-
• Spoiled GRE
ematical transformation process facilitates the easier separation of
• GRE-echo planar hybrid
overlapping signals before they are converted back into images and
• Steady-state free precession
accounts for the greater temporal fidelity and relative robustness of k-t
Acceleration method Parallel imaging, if available
PCA to motion.
Temporal resolution Every heartbeat (for ischemia detection)
Readout temporal resolution ~100–125 ms or shorter as available Non-Cartesian Techniques
(= acquisition shot duration)
Alternative approaches to acceleration involve data acquisition along
No. of dynamics Image for 40–50 heartbeats, by which
non-Cartesian patterns such as radial trajectories through the center
time contrast has passed through the
of k-space (Fig. 5.5). Spatial and temporal redundancy are exploited
LV myocardium
serially and the altered geometry of k-space coverage facilitates greater
No. of slices At least 3 short-axis slices
efficiency compared with conventional Cartesian techniques by collect-
In-plane spatial resolution <3 mm
ing more data with each RF excitation.2,18,20 Non-Cartesian techniques
Slice thickness 8 mm
can also be combined with parallel imaging techniques and hold potential
Contrast agent dose and 0.05–0.1 mmol/kg (3–7 mL/s) followed by
for large gains in spatial–temporal resolution.
administration regime at least 30 mL saline flush (3–7 mL/s)
In the highly constrained back-projection reconstruction (HYPR)
Breath-holding Breath-hold starts during early phases of
method and other radial acquisition variants, k-space data is acquired
contrast infusion before contrast
with undersampled radial projections and overall rotation of the under-
reaches the LV cavity
sampling pattern at different time points.20–22 In image reconstruction,
CMR, Cardiovascular magnetic resonance; GRE, gradient echo; LV, left a fully sampled composite image is formed by populating missing data
ventricular. from neighboring time frames. This very low temporal resolution

A Standard-resolution B C

D High-resolution E F
FIG. 5.1  High-resolution perfusion cardiovascular magnetic resonance (CMR). Standard (A–C) and high-
resolution (D–F) stress perfusion CMR in a patient with three-vessel coronary artery disease. Standard
resolution shows perfusion defects (arrows) in the basal-inferior (A), mid-inferior, mid-inferoseptal (B), apical-
anterior and apical-inferior segments (C). High resolution shows a similar distribution of perfusion defects
but demonstrates additional ischemia in the basal-lateral (D), mid-anterior and mid-anterolateral segments
(E) with a circumferential defect in the apical slice (F). Perfusion defects are also better delineated at high
resolution and the transmural extent of ischemia more clearly seen. (From Motwani M, Maredia N, Fairbairn
TA, et al. Assessment of ischaemic burden in angiographic three-vessel coronary artery disease with high-
resolution myocardial perfusion cardiovascular magnetic resonance imaging. Eur Heart J Cardiovasc Imaging.
2014;15:701–708.)
DRA occurs early ~1 voxel width

2.6mm

Frame 0 Frame 1 Frame 3

Lasts <8 frames

FIG. 5.2  Dark rim artifact. (A) Dark rim artifact (DRA) (arrows) is a frequent
finding in standard-resolution perfusion cardiovascular magnetic resonance
(CMR) and relates to several factors, including cardiac motion, Gibb’s ringing,
susceptibility and partial volume cancellation between the myocardium and
A blood pool. Although DRAs may mimic perfusion defects, they can be
Frame 5 Frame 6 Frame 7 distinguished by characteristic features: they occur at the arrival of contrast
in the left ventricular cavity and before its arrival in the myocardium (Frame
1); they tend to disappear within 8 to 10 frames (Frame 7); their location
is usually typical for a particular pulse sequence; and their width roughly
equates to the in-plane spatial resolution, which was 2.5 mm in this example
(Frame 3). (B) A 60-year-old man with suspected angina underwent stress
perfusion CMR at 1.5 T with both standard-resolution (2.5 mm in-plane)
and high-resolution (1.5 mm in-plane) acquisition (8 × k-t BLAST). There
was no perfusion defect seen with either acquisition but there was signifi-
B cant DRA on the standard-resolution images (arrows). X-ray angiography
Standard-resolution High-resolution confirmed normal coronary arteries.

Base

Mid

A Apex

FIG. 5.3  Three-dimensional (3D) perfusion car-


Base diovascular magnetic resonance (CMR). (A) Con-
secutive slices of a 3D perfusion CMR scan
facilitated by k-t principal component analysis during
adenosine stress in a patient with significant ste-
noses in the proximal right coronary artery and
distal left anterior descending coronary artery. (B)
Identical images illustrating volumetry of myocardial
Mid hypoenhancement using a signal intensity threshold
of 2 standard deviations below remote myocardium
(red areas). The volume of myocardial hypoen-
hancement was 30.4% of total myocardium. (From
Motwani M, Jogiya R, Kozerke S, et al. Advanced
cardiovascular magnetic resonance myocardial
perfusion imaging: high-spatial resolution versus
B 3-dimensional whole-heart coverage. Clin Cardio-
Apex vasc Imaging. 2013;6:339–348.)
54 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Image
reconstruction

A Aliased image B Training data C


FIG. 5.4  k-t undersampling acceleration techniques. (A) In k-t broad linear speed-up technique (BLAST) and
k-t sensitivity encoding (SENSE), data acquisition is accelerated by undersampling along the spatial encoding
(k) and time (t) axes, which leads to an aliased image. (B) Low-resolution “training data” is also obtained to
determine data correlations: that is, “prior-knowledge.” (C) Finally, a nonaliased full image series is recon-
structed using a statistical model derived from the training data. (From Motwani M, Jogiya R, Kozerke S,
et al. Advanced cardiovascular magnetic resonance myocardial perfusion imaging: high-spatial resolution
versus 3-dimensional whole-heart coverage. Clin Cardiovasc Imaging. 2013;6:339–348.)
Partition-encoding

ng
direction

di
n co
ct en
re e-
di has
io

A Readout direction B C
P

FIG. 5.5  Non-Cartesian trajectories. Examples of three potential alternate trajectories discussed in the text:
(A) echo planar imaging demonstrated with an echo train length of 4; (B) a spiral trajectory with 4 interleaves;
and (C) a radial projection design. Partition encoding direction in (B) and (C) is the same as for (A). (From Fair
MJ, Gatehouse PD, DiBella EV, et al. A review of 3D first-pass, whole-heart, myocardial perfusion cardio-
vascular magnetic resonance. J Cardiovasc Magn Reson. 2015;17:68.)

composite is then used to constrain back projection of the undersampled Compressed Sensing
data acquired for each individual time frame.20,22 Non-Cartesian trajectories have reduced cardiac motion-induced dark rim
To date, perhaps because other approaches are already well estab- artifact compared with conventional Cartesian acquisition strategies.5,28
lished, radial imaging has not been widely applied to two-dimensional In addition, the acquisition efficiency and benign aliasing artifacts enable
(2D) perfusion CMR, other than for specific research purposes such acquisition of higher-resolution images for a given acquisition time.
as multiple samples through the center of k-space to calculate the arte- However, both radial and spiral techniques require accurate scanner
rial input function (AIF) or to capitalize on its inherent motion robust- calibration and special image reconstruction techniques. An alterna-
ness for developing free-breathing techniques.23,24 Radial sampling does, tive method for reconstruction of undersampled data is compressed
however, lend itself to combination with compressed sensing methods sensing (CS), which is based upon the principle that an image with a
(see later) and may therefore see greater future application in 3D perfu- sparse representation in a known transform domain can be recovered
sion CMR techniques requiring particularly high acceleration factors.5 from randomly undersampled k-space data using a nonlinear recon-
Spiral imaging is another non-Cartesian technique in which data struction (Fig. 5.7).29,30 Sparsity in this context describes a matrix of
is acquired spiraling outward from the central raw data through pixels with predominantly zero values in a single image or series of
k-space.25–28 Spiral pulse sequences have some inherent efficiency and images. CS is therefore readily applicable to perfusion CMR because
SNR advantages over the radial technique, but are also more sensitive the signal is sparse in the combined temporal and spatial domain with
to off-resonance effects. However, careful selection of readout duration, only portions of the field of view requiring full temporal bandwidth
flip angle strategy, and other sequence characteristics have been shown and the other regions having only static information or at low tem-
to compensate for spiral-related artifacts to produce high-quality high- poral frequencies. The additional CS requirement of incoherence (i.e.,
resolution perfusion images.25 Most commonly, for 3D data acquisition, that undersampling artifacts look like additive noise) can be achieved
a stack of spiral planes with Fourier encoding in the third direction in perfusion CMR by randomly omitting phase-encoding lines (ky)
is used to achieve whole-heart coverage in a cylindrical distribution with a different pattern for each time point (t), generating a random
(Fig. 5.6).5,26 undersampling pattern in ky-t space. Compared with ky-only random
A

4 4
ANT ANT
Signal Intensity (a.u.)

Signal Intensity (a.u.)


ANT SEP ANT SEP
3 INF SEP
3 INF SEP
INF INF
INF LAT INF LAT
ANT LAT ANT LAT
2 2

1 1

5 10 15 20 25 30 35 40 5 10 15 20 25 30 35 40
B Cardiac cycles C Cardiac cycles
FIG. 5.6  (A) Stack-of-spirals three-dimensional perfusion cardiovascular magnetic resonance. Illustrative images
acquired through stack of spirals during right ventricle blood pool (top), LV blood pool (middle), and LV myo-
cardial (bottom) enhancement. Dotted lines indicate the middle slice, which was used for comparison with
the corresponding single-slice two-dimensional (2D) Cartesian acquisition (not shown). (B–C) The correspond-
ing myocardial signal–time curves for this and its corresponding 2D slice are shown, demonstrating good
agreement, except for k-t parallel imaging artifacts in the early and late frames. (From Shin T, Nayak KS,
Santos JM, et al. Three-dimensional first-pass myocardial perfusion MRI using a stack-of-spirals acquisition.
Magn Reson Med. 2013;69:839–844.)

B C
FIG. 5.7  Compressed sensing. Conceptual illustration of the compressed sensing technique for cardiac
perfusion magnetic resonance imaging. (A) Sparsity: the fully sampled ky-t data is sparse in y-f space.
(B) Incoherence: ky-t pseudo-random undersampling preserves the original sparse representation in y-f space
and presents artifacts that look like additive noise (pseudo-noise). (C) Nonlinear reconstruction: the sparse
coefficients in y-f space can be recovered with a nonlinear reconstruction that minimizes the number of
nonzero coefficients (minimum L1-norm). F, Fourier transform. (From Otazo R, Kim D, Axel L, et al. Combina-
tion of compressed sensing and parallel imaging for highly accelerated first-pass cardiac perfusion MRI. Magn
Reson Med. 2010;64:767–776.)
56 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

undersampling, ky-t random undersampling increases the incoherence, 1.5 mm.17 The study reported similar overall image quality as standard-
because the undersampling artifacts are incoherently distributed along resolution (in-plane spatial resolution: 2.6 mm) SENSE-accelerated
two dimensions rather than one. acquisition, but there was a significant reduction in the extent of dark
Unlike k-t BLAST and k-t SENSE, CS does not require training data, rim artifact (mean thickness: 1.7 vs. 2.4 mm; P < .01) and SNR corrected
and, as a result, it may be less sensitive to inconsistencies between train- for pixel size was higher for the k-t method. In a subsequent small study
ing and imaging data. Application of CS to dynamic CMR has been by Maredia et al.,11 different tradeoffs between spatial resolution and
presented in methods such as k-t SPARSE and k-t FOCUSS, using the shot duration were explored and a reduction in dark rim artifact with
temporal fast Fourier transform (FFT) as the sparsifying transform, the highest in-plane spatial resolution was confirmed.
and ky-t random undersampling.29,31–33 Maximum acceleration rate in Following the first feasibility studies in volunteers, high-resolution
CS is determined by image sparsity, and in practice, the number of perfusion CMR has been validated in several small patient studies (Table
required samples is approximately three to five times the number of 5.2).6–8,11 Plein et al. used an identical sequence to their previous vol-
nonzero coefficients in the image.29,32 Similar to the other advanced unteer study (1.5 T, 5-fold k-t SENSE, in-plane resolution 1.4 mm) in
acceleration methods discussed, CS can be combined with parallel 51 patients with known or suspected coronary artery disease (CAD).6
imaging techniques when multiple receiver coils are employed to obtain High-resolution acquisition was found to have high image quality and
higher accelerations (Fig. 5.8). a high diagnostic accuracy (area under the curve, AUC: 0.85) compared
with quantitative coronary angiography (QCA) with similar performance
HIGH-RESOLUTION PERFUSION CARDIOVASCULAR in single-vessel and multivessel disease (AUC: 0.87 vs. 0.82). Manka
et al.8 used k-t SENSE with 8-fold acceleration at 3 T to achieve an
MAGNETIC RESONANCE in-plane spatial resolution of 1.1 mm, reporting an AUC of 0.94 in 20
There is no clear definition of “high-resolution perfusion CMR,” but an patients with suspected CAD against QCA. Lockie et al.9 validated high-
in-plane spatial resolution better than 2 mm, which is comparable with resolution perfusion CMR (5-fold k-t BLAST, in-plane resolution 1.2 mm)
that of other common CMR methods such as late gadolinium enhance- against invasive fractional flow reserve (FFR) and in 42 patients reported
ment (LGE) or cine imaging, is generally considered high. The rationale a high diagnostic accuracy (AUC: 0.92) for the detection of hemody-
for pursuing high-resolution CMR is to facilitate comparison with cine namically significant lesions (FFR < 0.75). In a study by Salerno et al.,28
and LGE images, to improve the detection of subendocardial ischemia a saturation recovery interleaved variable-density spiral pulse sequence
and transmural ischemic gradients, and to reduce dark rim artifacts. achieving 2 × 2 mm in-plane spatial resolution for three short-axis
These artifacts are a common finding in conventional perfusion CMR slices was used in 41 patients listed for QCA. The sensitivity, specificity,
and are thought to be caused by magnetic susceptibility effects, Gibbs and accuracy were 89%, 85%, and 88%, respectively, to detect coronary
ringing, and cardiac motion during acquisition.34 Because these artifacts stenosis >50%. Finally, a variant of the HYPR method known as sliding-
are directly proportional to voxel size, high-resolution perfusion CMR window conjugate gradient HYPR (6 contiguous slices, 1.6 mm in-plane
leads to a reduction of dark rim artifact (see Fig. 5.2).6,10,11 As discussed, spatial resolution), has been shown to be clinically feasible in a single-
non-Cartesian sampling techniques such as radial or spiral imaging have center study (n = 50) and demonstrated high diagnostic accuracy (sen-
shown particular benefits in minimizing dark rim artifact reduction sitivity 96%, specificity 82%).22 Radial perfusion CMR has also been
because of their robustness to motion and Gibbs ringing effects.34,35 shown to be effective in eliminating the need for ECG gating and has
the potential to achieve high diagnostic accuracy in arrhythmic patients
Methods and Clinical Validation (Fig. 5.9).36
High-resolution perfusion CMR was first described in a feasibility study Although these clinical validation studies show good diagnostic
using 5-fold k-t SENSE to achieve an in-plane spatial resolution of performance of high-resolution perfusion CMR against anatomical

FIG. 5.8  Compressed sensing three-dimensional perfusion cardiovascular magnetic resonance. An example
of breath-held, 8-fold accelerated whole-heart perfusion acquisition (10 short-axis slices) is achieved by a
combination of compressed sensing and parallel imaging. In this example, compressed sensing and parallel
imaging are combined by merging the k-t SPARSE technique with SENSE reconstruction to substantially
increase the acceleration rate for perfusion imaging. These k-t SPARSE-SENSE reconstructed images exhibited
good image quality. Image acquisition matrix = 128 × 128, spatial resolution = 3.2 × 3.2 mm2, and temporal
resolution = 38.4 ms. The reconstruction time per slice was approximately 15 minutes, which represented
a total reconstruction time of approximately 2.5 hours for the 10 slices using Matlab (The MathWorks, Natick,
MA) on a 64-bit quad core workstation. (From Otazo R, Kim D, Axel L, et al. Combination of compressed
sensing and parallel imaging for highly accelerated first-pass cardiac perfusion MRI. Magn Reson Med.
2010;64:767–776.)
CHAPTER 5  Myocardial Perfusion Cardiovascular Magnetic Resonance: Advanced Techniques 57

TABLE 5.2  Clinical Validation of Advanced Accelerated Perfusion Cardiovascular Magnetic


Resonance
Advanced
Acceleration In-Plane Spatial Reference Diagnostic
No. Method Resolution Coverage Standard Accuracy
High Resolution
Plein et al. 20086 51 1.5 T 5 × k-t-SENSE 1.4 mm 4 slices (NC)b QCA > 50% 0.85c
Plein et al. 20087 33 1.5 T 5 × k-t SENSE 1.5 mm 4 slices (NC)b QCA > 50% 0.80c
Plein et al. 20087 33 3.0 T 5 × k-t SENSE 1.3 mm 4 slices (NC)b QCA > 50% 0.89c
Manka et al. 20108 20 3.0 T 8 × k-t SENSE 1.1 mm 3 slices (NC) QCA > 50% 0.94c
Lockie et al. 20119 42 3.0 T 5 × k-t BLAST 1.2 mm 3 slices (NC) FFR < 0.75 0.92c
Lockie et al. 20119 42 3.0 T 5 × k-t BLAST 1.2 mm 3 slices (NC) FFR < 0.75 0.89c [MPR]a
Motwani et al. 201210 100 1.5 T 8 × k-t BLAST 1.6 mm 3 slices (NC) QCA ≥ 50% 0.93c
Ma et al. 201222 50 3.0 T SW-CG-HYPR 1.6 mm 6 slices (WH) QCA ≥ 50% 0.90d
Chirbiri et al. 201340 67 3.0 T 5 × k-t SENSE 1.2 mm 3 slices (NC) FFR < 0.80 0.86c [TPG]a
Salerno et al. 201428 41 1.5 T 8 × VD Spiral 2.0 mm 3 slices (NC) QCA > 50% 0.88d

3D
Manka et al. 201113 146 3.0 T 6 × k-t SENSE 2.3 mm 16 slices (WH) QCA ≥50% 0.83d
Manka et al. 201214 120 1.5 T 10 × k-t PCA 2.0 mm 16 slices (WH) FFR < 0.75 0.87d
Jogiya et al. 201215 53 3.0 T 10 × k-t PCA 2.3 mm 12 slices (WH) FFR < 0.75 0.91d
Jogiya et al. 201443 33 3.0 T 10 × k-t PCA 2.3 mm 12 slices (WH) QCA ≥ 70% 0.88d
Manka et al. 201549 150 3.0 T 10 × k-t PCA 2.3 mm 16 slices (WH) FFR < 0.80 0.87c
Motwani et al. 201446 20 3.0 T 10 × k-t PCA 2.3 mm 12 slices (WH) QCA ≥ 70% 0.95b [sMBF]a
a
These data refer to quantitative analysis rather than visual analysis.
b
In these studies, temporal resolution was 2 R-R intervals.
c
In these studies, diagnostic accuracy was calculated by receiver-operating characteristic analysis.
d
In these studies, diagnostic accuracy was expressed as the proportion of correctly classified subjects.
3D, Three-dimensional; BLAST, broad linear speed-up technique; FFR, fractional flow reserve; HYPR, highly constrained back-projection
reconstruction; MPR, myocardial perfusion reserve; NC, noncontiguous; PCA, principal component analysis; QCA, quantitative coronary
angiography; SENSE, sensitivity encoding; sMBF, stress myocardial blood flow; SW-CG-HYPR, sliding-window conjugate-gradient HYPR; TPG,
transmural perfusion gradient; VD, variable density; WH, whole-heart.

FIG. 5.9  Radial acquisition all-systolic nongated high-resolution perfusion cardiovascular magnetic resonance
(CMR). Non-Cartesian perfusion CMR performed using a non–electrocardiogram (ECG) gated continuous
radial sampling enables systolic imaging of all slices at 3 T at stress (top) and rest (bottom) at high-resolution
(1.4 × 1.4 mm2) in a patient with a history of coronary artery disease and prior revascularization. A transmural
perfusion gradient with hypoperfused subendocardium in the inferior wall is consistent with the severe
disease throughout the right coronary artery by angiography (arrows, right panel). Continuous radial acquisi-
tion can achieve high-resolution systolic imaging without ECG gating, which is free of dark rim artifact. (From
Sharif B, Arsanjani R, Dharmakumar R, et al. All-systolic non-ECG-gated myocardial perfusion MRI: feasibility
of multi-slice continuous first-pass imaging. Magn Reson Med. 2015;74:1661–1674.)
58 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

and functional endpoints, the key question is whether high-resolution feasibility of quantitative transmural perfusion gradients, derived
CMR offers incremental value over standard perfusion methods. Only from high-resolution myocardial perfusion CMR data, to detect CAD
one study has so far addressed this question. In a direct comparison with high diagnostic accuracy compared with an FFR reference stan-
between high-resolution and standard-perfusion CMR, high-resolution dard.39,40 More recent work using radial undersampling to facilitate
perfusion CMR (8-fold k-t BLAST, in-plane spatial resolution 1.6 mm) high-resolution perfusion CMR without ECG gating and without
had significantly greater overall diagnostic accuracy in 100 patients saturation-recovery magnetization preparation enables reconstruc-
with suspected CAD than standard-resolution imaging (AUC: 0.93 vs. tion of images in any cardiac phase.41,42 This approach has demon-
0.83, respectively; P < .001). This improved diagnostic performance strated particularly accurate visualization of transmural perfusion
was attributed to better and more confident detection of subendo- gradients enabled by the option of selecting the end-systolic phase to
cardial perfusion defects, consistent with the higher susceptibility of be chosen for all of the three myocardial slices (“all systolic” imaging)
the endocardial layer to ischemia.10,37 Another theoretical advantage (see Fig. 5.9).42
of high-resolution perfusion imaging is the more confident detection
of multivessel CAD (Figs. 5.1 and 5.10). In a small study of 35 patients THREE-DIMENSIONAL WHOLE-HEART PERFUSION
with angiographically confirmed three-vessel CAD, 24 of 35 patients had
an MIB >10%, with the standard-resolution perfusion CMR method
CARDIOVASCULAR MAGNETIC RESONANCE
compared with 33 patients by the high-resolution technique, suggest- The most obvious advantage of 3D over conventional 3-slice acquisi-
ing more accurate detection of ischemic burden with the higher spatial tion is that contiguous spatial coverage allows the true extent of perfu-
resolution.38 sion defects to be visualized.5,12,14,43 This would be expected to increase
High-resolution CMR also allows more confident analysis of trans- the diagnostic confidence but also allow more accurate estimation of
mural perfusion differences, as a hallmark of myocardial ischemia the MIB. Additionally, 3D perfusion CMR with whole-heart coverage
in both epicardial coronary disease as well as microvascular disease. includes the true apex of the heart, which is not conventionally imaged
Two studies employing a k-t SENSE sequence have demonstrated the with 2D methods.

A Stress B Rest

C Left Coronary Artery D Right Coronary Artery


FIG. 5.10  High-resolution perfusion cardiovascular magnetic resonance (CMR) in multivessel disease.
A patient with suspected angina underwent stress perfusion CMR at 1.5 T using a high-resolution technique
(1.4 mm in-plane) facilitated by 5-fold k-t broad linear speed-up technique. Stress-induced perfusion defects
were seen in all three territories (arrows, A, B) and are clearly demarcated as subendocardial. Subsequent
invasive coronary angiography revealed significant three-vessel disease (arrows, C, D). This case highlights
the ability of high-resolution acquisition to overcome the potential effects of balanced ischemia in multivessel
disease by detecting transmural perfusion gradients and subendocardial ischemia. (From Motwani M, Jogiya
R, Kozerke S, et al. Advanced cardiovascular magnetic resonance myocardial perfusion imaging: high-spatial
resolution versus 3-dimensional whole-heart coverage. Clin Cardiovasc Imaging. 2013;6:339–348.)
CHAPTER 5  Myocardial Perfusion Cardiovascular Magnetic Resonance: Advanced Techniques 59

is an advantage over conventional 2D imaging, in which the acquisi-


Methods and Clinical Validation tion timepoint cannot be optimized for all slices individually and may
Three-dimensional myocardial perfusion CMR has been achieved using therefore coincide with rapid cardiac motion in some slices, it means
a range of acceleration methods, including k-t BLAST, k-t SENSE, and that an upfront choice must be made as to the preferred cardiac phase
k-t PCA, as well as TSENSE, TWIST, and CS with mostly Cartesian, but for acquisition, and at present there is no uniform consensus. Both
also radial and spiral readout.5,13,15 Typically 7- to 10-fold acceleration end systole and mid-diastole have been used and both have specific
is required to acquire a 3D stack with whole-heart coverage, an in- benefits.16,44,45 In mid-diastole, the heart is usually at its most stationary,
plane spatial resolution of 2 to 3 mm, and an acquisition shot lasting but the relatively thin myocardium can compound the effect of dark
less than 250 ms.13,15 rim artifact and limit the assessment of the transmurality of perfusion
One important aspect of 3D data acquisition to consider is that defects.16,46 End systole, on the other hand, has a shorter quiescent period
the entire stack is acquired at the same part of the cardiac cycle (Fig. but the thicker myocardium reduces the effect of dark rim artifact and
5.11), and this specific timepoint is open to selection. Although this facilitates the grading of defect transmurality.16,46 Systolic acquisition

100−200 ms

Basal Mid Apex

Early systole Mid-systole Early diastole

A
TSR

Undersampled
3D readout

Undersampled k-space

ky Fully sampled training data

kx

Reconstructed
3D perfusion images.
All slices are at
the same timepoint
in cardiac cycle.
B
FIG. 5.11  Pulse sequence diagrams for two-dimensional (2D) and three-dimensional (3D) perfusion cardio-
vascular magnetic resonance (CMR). (A) In 2D perfusion CMR, 3 to 4 noncontiguous slices are acquired in
different phases of the cardiac cycle to maximize spatial coverage. Each slice is acquired after a saturation
prepulse (black bar). (B) In 3D-perfusion CMR, a single saturation prepulse is followed by a saturation recovery
time (TSR) and undersampled 3D perfusion data readout. The use of advanced spatiotemporal undersampling
allows sufficient data acquisition to reconstruct 16 contiguous slices (whole-heart coverage). The “training
data” is acquired with the undersampled data in an interleaved fashion. Because all perfusion data in the 3D
technique are acquired at the same point in the cardiac cycle, the reconstructed slices all appear in the same
cardiac phase. (From Motwani M, Jogiya R, Kozerke S, et al. Advanced cardiovascular magnetic resonance
myocardial perfusion imaging: high-spatial resolution versus 3-dimensional whole-heart coverage. Clin Car-
diovasc Imaging. 2013;6:339–348.)
60 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

is also less sensitive to R-R variability, which is particularly relevant Whether or not 3D whole-heart assessment is superior to 2D per-
to patients with atrial fibrillation.44 The choice of cardiac phase may fusion CMR has only been addressed directly in one small study: 27
also affect quantitative measures of myocardial blood flow and recent patients with stable angina underwent both 2D high-resolution and
studies have shown higher flow values in data acquired in systole than whole-heart 3D perfusion CMR. The mean MIB between the two
in diastole.16,46–48 methods was similar (4.3 ± 5.2% vs. 4.1 ± 4.9%; P = .81) with no sys-
Several small-scale single-center studies and one multicenter study tematic bias between techniques (mean bias = −0.17%, 95% CI: −1.7
have evaluated the clinical feasibility and diagnostic performance of to −1.3%). However, when used to categorize MIB as >10% or <10%,
3D myocardial perfusion CMR to detect significant CAD (see Table there was only fair agreement between the two techniques (κ = 0.29,
5.2).12–15,43 In the first clinical validation study, 146 patients with suspected 95% CI: −0.12 to 0.70). Larger comparative studies are needed to decide
CAD underwent 3D perfusion CMR facilitated by k-t SENSE (6.3-fold whether 3D perfusion CMR is more accurate than 2D acquisition to
nominal acceleration).13 Sensitivity, specificity, and diagnostic accuracy estimate MIB.
of 3D perfusion CMR to detect ≥50% stenosis on QCA were 92%, 74%, Because the majority of clinical studies evaluating 3D perfusion
and 83%, respectively. Two subsequent studies compared 3D perfusion CMR are from related centers, parameters are fairly similar and all
CMR against the invasive functional endpoint of FFR. Both studies used have used Cartesian-based methods.5 In-plane spatial resolution is 2.3
k-t PCA acceleration to achieve whole-heart 3D acquisition in systole. × 2.3 mm in all cases, with through-plane resolution ranging from 5
Manka et al.14 studied 120 patients at 1.5 T and reported sensitivity, to 10 mm. Because of this limited variation in implementation and
specificity, and diagnostic accuracy of 3D perfusion against FFR of reconstruction methods in the clinical studies to date, the extrapolation
90%, 82%, and 87%, respectively. Jogiya et al.15 used a very similar 3D of results to different CMR systems and nonspecialist CMR centers is
method in 53 patients at 3 T and reported values of 91%, 90%, and not yet possible. Nonetheless, these early results are encouraging and
91%, respectively. Most recently, a multicenter study of 150 patients validate many of the proposed benefits of 3D whole-heart acquisition.
from five European centers established a sensitivity and specificity of Technical development of 3D perfusion CMR sequences using non-
85% and 91%, relative to FFR at 3.0 T.49 Cartesian approaches is also already at an advanced stage. Recently,
These studies demonstrate that 3D myocardial perfusion CMR is radial trajectories have been used for 3D perfusion sequences, com-
feasible in typical patient populations and achieves high diagnostic bined with CS.5,27,50 CS is particularly suited to 3D data acquisition
performance for the detection of CAD at both 1.5 and 3.0 T. The reported because the extra dimension and spatial coverage allow greater com-
levels of diagnostic accuracy are comparable with standard- and high- pressibility of the data. Despite this theoretical advantage, the issues
resolution 2D myocardial perfusion CMR. However, because a sizeable of respiratory motion/misgating mentioned earlier have resulted in
direct comparison of 3D and 2D acquisition has not been performed, relatively limited application of CS to 3D perfusion CMR. However, a
any potential incremental value of 3D whole-heart imaging has not yet CS algorithm using localized spatiotemporal constraints has recently
been established. been used to demonstrate the feasibility of free-breathing 3D perfu-
The separate issue of quantifying ischemic burden with whole- sion CMR.51 In work optimizing the first spiral 3D perfusion sequence,
heart perfusion CMR has also been addressed in most of the studies the higher efficiency of this k-space traversal achieved acquisition
discussed previously. In the first 3D perfusion CMR study by Manka windows closer to those seen using Cartesian approaches, at the
et al.,13 48 of their 146 patients had repeat CMR after revascularization. smallest undersampling factor for any combined CS/k-t technique
The authors measured MIB with a volumetric thresholding method. (see Fig. 5.6).26
Myocardium with a signal level <2 SD below remote at the time of Finally, there is potential for novel admixture of the benefits of many
peak contrast enhancement was considered as ischemic. The authors of the previously discussed approaches. For example, a spiral twist could
showed a relative reduction in the ischemic burden of 79% ± 25% be added to the ends of radial projections, known as twisting radial
between baseline and follow-up CMR, highlighting the potential role lines (TWIRL) or a Cartesian grid acquisition used for each projection
of 3D perfusion CMR to serially monitor the response to antiischemic angle, known as periodically rotated overlapping parallel lines with
therapies (see Fig. 5.3). In their second study, Manka et al.14 reported enhanced reconstruction (PROPELLER).52,53 Although these concepts
that the quantified MIB had a high diagnostic accuracy itself (AUC: have not yet even been applied to 2D perfusion CMR, they are possible
0.90) for identifying FFR-defined CAD—with an optimal cutoff value candidates for the highly optimized sequences required for 3D perfu-
of >4.4%. Jogiya et al. repeated a strong correlation (r = 0.82; 95% sion acquisitions.5
confidence interval [CI]: 0.70–0.89; P < .0001) between the MIB on 3D
perfusion CMR and the Duke Jeopardy score (a validated angiographic
index of ischemic burden based on lesion severity and location).15 A LIMITATIONS OF HIGHLY ACCELERATED
separate study by Jogiya et al.43 found strong correlation (r = 0.70, P < PERFUSION CARDIOVASCULAR MAGNETIC
.001) between estimates of MIB from 3D perfusion CMR and single
photon emission computed tomography (SPECT) in 38 patients with
RESONANCE
confirmed perfusion defects on both modalities. These data suggest that In general, advanced acceleration techniques add complexity to the
3D perfusion CMR may fulfill the expectation of providing accurate acquisition of perfusion CMR, often require research software, and are
assessment of MIB. largely confined to academic institutions. Reconstruction processes can
Although the latter study found no systematic bias between MIB be demanding on computing hardware and are therefore often lengthy
estimates from the two techniques, and therefore suggested potential and performed off-line. Methods that include temporal undersampling
interchangeability, the 95% limits of agreement were relatively wide can be sensitive to respiratory motion and cardiac arrhythmias. In many
(−14.3% to 13.1%). Considering that a 10% MIB threshold is often of the published studies, breathing-related artifacts were minimized by
used to direct clinical management, this caution is particularly relevant acquiring fewer than standard dynamics (in many studies only 30
at the lower end of the MIB spectrum given the clinical impact of even dynamics), which requires very precise breath-holding and contrast
a 5% to 10% absolute difference. Nonetheless, disagreement between injection timing to ensure that the main contrast bolus is imaged.6
3D perfusion CMR and SPECT estimates of MIB at the 10% threshold Myocardial SNR is a particular concern for high-resolution and 3D
only occurred in 3 out of 38 cases.43 perfusion CMR techniques because of the theoretical potential for SNR
CHAPTER 5  Myocardial Perfusion Cardiovascular Magnetic Resonance: Advanced Techniques 61

loss caused by many of the acceleration methods. However, although quantitative analysis of perfusion CMR, but there is intention for this
there is lower SNR associated with the smaller voxel size acquired in to be addressed by an international standardization task force.1,67 Sec-
high-resolution techniques, this can be compensated for by constraining ondly, there is also no widely available and validated analysis software
the reconstruction with low-resolution, high-SNR training data. Fur- for quantitative analysis of perfusion CMR data, and research groups
thermore, in 3D sequences the raw data (k-space) are acquired with generally use in-house solutions. Analysis can be time-consuming, pre-
additional repeated acquisitions of phase-encoding used to collect the cluding routine clinical application. Finally, the incremental value of
third dimension (slice or partition encoding)—which delivers a fun- quantitative analysis of myocardial perfusion CMR analysis still needs
damental increase in SNR as well as facilitating contiguous coverage. to be shown in large clinical studies.
Therefore, if all other factors are kept the same, a 3D acquisition has
inherently higher SNR than a comparable 2D sequence.
OPTIMAL USE OF ACCELERATION
As shown, high spatial resolution and whole-heart 3D perfusion CMR
QUANTITATIVE ANALYSIS are both feasible using advanced data acceleration methods. Both methods
Over the last decade, several animal, normal volunteer, and patient have distinct potential advantages over standard-resolution 2D methods;
studies have validated the use of conventional perfusion CMR for abso- higher spatial resolution improves the detection of subendocardial
lute myocardial blood flow (MBF) quantification against microsphere ischemia and reduces dark rim artifact, whereas 3D whole-heart imaging
and invasive coronary flow reserve measurements.54–56 Furthermore, promises more reliable ischemia quantification. In the published litera-
several clinical CMR studies have demonstrated the high diagnostic ture, the overall diagnostic performance to detect CAD between the
accuracy of absolute MBF and myocardial perfusion reserve (MPR) two approaches has been similar, but large-scale direct comparisons
estimates for CAD detection compared with both QCA and FFR.9,16,57–59 between the methods and in comparison with standard 2D acquisition
However, the spatiotemporal undersampling methods required for are lacking.8,10,14,15,49,68
high-resolution or 3D data acquisition are sensitive to respiratory motion, Pending more conclusive evidence, where all methods are available,
cardiac arrhythmia, and low-pass temporal filtering, all of which pose clinicians may already choose one of the available methods according
additional challenges to applying quantitative assessment to these to the clinical context. For example, in a patient first investigated for
advanced perfusion sequences. In particular, low-pass temporal filtering the presence of CAD, diagnostic performance may be the clinical prior-
may lead to significant underestimation of MBF. Nonetheless, quantita- ity. On current evidence, standard, high spatial resolution or 3D perfu-
tive methods for the estimation of MBF have been successfully applied sion CMR would appear suitable and comparable. In a patient with
to both high-resolution and 3D perfusion CMR acquisitions.9,46,60 Lockie known or suspected multivessel disease, high-resolution perfusion CMR
et al. found that applying standard quantitative analysis methods to a may be beneficial to detect the presence of balanced ischemia (see Figs.
k-t SENSE high-resolution perfusion sequence for MPR estimation 5.1 and 5.10). In patients with a history of myocardial infarction and
yielded a very high diagnostic accuracy (AUC: 0.89) for ischemia detec- suspected peri-infarct ischemia, high-resolution perfusion CMR may
tion compared with invasive FFR. Similarly, Motwani et al.46 found high be most useful to detect epicardial peri-infarct ischemia, whereas 3D
diagnostic accuracy for stress MBF and MPR estimates (AUC: 0.95 for perfusion is most suitable to match perfusion and LGE images to quantify
both) derived from a k-t PCA 3D perfusion sequence using standard MI and MIB. Figs. 5.12 through 5.14 show examples of patients under-
quantitative analysis methods. Notably, the use of the constrained k-t going both acquisitions and the relative merits (Table 5.3) of both
PCA framework for image reconstruction has been shown to improve techniques in each case.
temporal fidelity, permitting robust measurements of MBF at very high
acceleration factors.19 As discussed earlier, k-t PCA is also less prone to
respiratory artifact as temporal basis functions are derived based on
CONCLUSION
the low-resolution training data acquired in every heartbeat.61 The development of advanced acceleration techniques allows improve-
A widely acknowledged limitation of quantitative perfusion CMR, ments of perfusion CMR methods compared with standard techniques.
in general, is that the predominant models used for estimating MBF In the published literature, data acceleration has mostly been invested
assume a linear relation between signal and contrast agent concentra- in improving the in-plane spatial resolution in 2D acquisition or to
tion, that is, ignoring saturation effects in the LV blood pool, which
can lead to underestimation of MBF.62 This assumption is particularly
relevant to high-resolution and 3D perfusion techniques because rela-
tively high contrast agent doses are often used to optimize visual image TABLE 5.3  High-Resolution Versus 3D
quality. Proposed solutions include the dual-sequence method in which Whole-Heart Perfusion CMR
two interleaved imaging sequences are used to acquire the AIF and
High-Resolution 3D Whole-Heart
myocardial tissue first-pass curves concurrently from the same contrast
Perfusion CMR Perfusion CMR
bolus; or the dual-bolus method which employs a low-dose bolus for
the AIF measurement, followed by a higher dose for the myocardium. Greater detection of Assesses true extent of perfusion
Another sequence-based approach is the use of radial trajectories and subendocardial ischemia defects
reconstruction of separate images for blood pool and myocardial enhance- Benefit in multivessel disease Benefit in apical ischemia
ment from different amounts of projections in the same dataset. Although Less dark rim artifact Inherently high SNR
such methods add further complexity to data acquisition and/or post- Similar spatial resolution to Single selected phase for all
processing, they have also been successfully applied to high-resolution LGE imaging slices
and 3D whole-heart acquisitions.23,63–66 Nonetheless, there still remain Quantitative analysis of TPG Quantitative assessment of MIB
a number of other limitations that hold back the wider clinical adop- 3D, Three-dimensional; CMR, cardiovascular magnetic resonance;
tion of quantitative perfusion CMR. Arguably, the current lack of stan- LGE, late gadolinium enhancement; MIB, myocardial ischemic
dardization in image acquisition, contrast dosing protocols, postprocessing, burden; SNR, signal-to-noise ratio; TPG, transmural perfusion
mathematical modeling, and interpretation is the major inhibitor for gradient.
62 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

3D stress perfusion

Base

Mid

Apex

LGE imaging

Base Base Mid Mid

Apex Apex Apical cap Apical cap

High-resolution stress perfusion

Base Mid Apex


FIG. 5.12  Case example 1. A 62-year-old man with a history of prior bypass surgery presented with angina
recurrence. The top panel dimensional shows three-dimensional (3D) perfusion cardiovascular magnetic
resonance (CMR) at stress (2.5 mm in-plane resolution, 12 slices); the middle panel shows late-gadolinium
enhancement (LGE) imaging (1.5 mm in-plane resolution), and the bottom panel shows high-resolution stress
perfusion CMR (1.1 mm in-plane resolution, 3 slices), all performed on the same patient at 3.0 T. Both 3D
perfusion and high-resolution techniques show inferior perfusion defects from base to apex. The benefit of
whole-heart coverage with the 3D technique is demonstrated in this case because hypoperfusion is seen
to extend beyond the scar into the apical cap (top panel, arrows) which is not covered by the 3-slice high-
resolution technique. On the other hand, the perfusion defects and their transmural extent are better delin-
eated with the high-resolution technique, particularly at the mid-ventricular level. By virtue of their similar
in-plane spatial resolution, it is easier to correlate LGE images with high-resolution perfusion CMR on a per
slice basis, compared with the 3D technique. (From Motwani M, Jogiya R, Kozerke S, et al. Advanced car-
diovascular magnetic resonance myocardial perfusion imaging: high-spatial resolution versus 3-dimensional
whole-heart coverage. Clin Cardiovasc Imaging. 2013;6:339–348.)
CHAPTER 5  Myocardial Perfusion Cardiovascular Magnetic Resonance: Advanced Techniques 63

3D stress perfusion

Base

Mid

Apex

LGE imaging

Base Mid Apex VLA

High-resolution stress perfusion

Base Mid Apex X-ray angiography


FIG. 5.13  Case example 2. A 45-year-old man with prior percutaneous coronary intervention (PCI) to the
left anterior descending (LAD) presented with significant angina. The top panel shows 3D perfusion cardio-
vascular magnetic resonance (CMR) (12 slices) at stress; the middle panel shows late-gadolinium enhance-
ment (LGE) imaging, and the bottom panel shows high-resolution (1.1 mm in-plane) stress perfusion CMR,
all performed at 3.0 T. Three-dimensional (3D) perfusion CMR shows stress-induced hypoperfusion throughout
the anterior wall from base to apex—that is, well beyond the area of scar seen in the mid-anterior wall on
LGE imaging (arrows). This example shows the benefit of whole-heart coverage with the 3D acquisition, as
the 3-slice high-resolution techniques did not demonstrate any significant ischemia beyond the established
scar in the mid-ventricle. Invasive coronary angiography confirmed a subtotal occlusion of a large diagonal
branch, accounting for the anterior ischemia (black arrow). VLA, Vertical long axis. (From Motwani M, Jogiya
R, Kozerke S, et al. Advanced cardiovascular magnetic resonance myocardial perfusion imaging: high-spatial
resolution versus 3-dimensional whole-heart coverage. Clin Cardiovasc Imaging. 2013;6:339–348.)
64 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

3D stress perfusion

Base

Mid

Apex

LGE imaging

Base Base Mid Mid

Mid Apex Apex VLA

High-resolution stress perfusion

Base Mid Apex X-ray angiography


FIG. 5.14  Case example 3. The top panel dimensional shows stress three-dimensional (3D) perfusion car-
diovascular magnetic resonance (CMR) (12 slices); the middle panel shows late-gadolinium enhancement
(LGE) imaging; and the bottom panel shows high-resolution (1.1 mm in-plane) stress perfusion CMR, all
performed at 3.0 T. The 3D technique demonstrated significant stress-induced hypoperfusion in the inferior
wall from base to apex and extending into the basal inferolateral segments; but there was also significant
dark rim artifact in the septum (endocardial border opposite dashed lines). X-ray angiography confirmed total
occlusion of the mid left circumflex artery (arrow). Because of the sparsity of coverage, the 3-slice high-
resolution technique only detected a significant perfusion defect at the mid-ventricular level and therefore
significantly underestimated the ischemic burden in this case, compared with the 3D technique. Additionally,
interpretation of the apical high-resolution slice is difficult as it is significantly more diastolic than the other
slices, which is a disadvantage of all two-dimensional acquisitions which employ a single-shot technique. By
comparison, with 3D perfusion CMR, all slices are acquired at the same point in the cardiac cycle, which
makes it easier to determine the extent of perfusion defects across different myocardial sections. VLA, Verti-
cal long axis. (From Motwani M, Jogiya R, Kozerke S, et al. Advanced cardiovascular magnetic resonance
myocardial perfusion imaging: high-spatial resolution versus 3-dimensional whole-heart coverage. Clin Car-
diovasc Imaging. 2013;6:339–348.)
CHAPTER 5  Myocardial Perfusion Cardiovascular Magnetic Resonance: Advanced Techniques 65

permit whole-heart 3D imaging methods. Some studies have proposed and 3D perfusion CMR already gives the opportunity to tailor the
compromise solutions which combine high spatial resolution with acquisition to an individual patient to best answer the specific clinical
the acquisition of more than the standard three 2D short-axis slices. question.
Current evidence suggests that both high-resolution and 3D perfusion
CMR have at least comparable diagnostic performance to standard
2D acquisition, but large direct comparisons are lacking. However,
REFERENCES
where expertise in both exists, the choice between high-resolution A full reference list is available online at ExpertConsult.com
CHAPTER 5  Myocardial Perfusion Cardiovascular Magnetic Resonance: Advanced Techniques 65.e1

REFERENCES 21. Mistretta CA, Wieben O, Velikina J, et al. Highly constrained


backprojection for time-resolved MRI. Magn Reson Med. 2006;55:30–40.
1. Kramer CM, Barkhausen J, Flamm SD, Kim RJ, Nagel E. Standardized 22. Ma H, Yang J, Liu J, et al. Myocardial perfusion magnetic resonance
cardiovascular magnetic resonance (CMR) protocols 2013 update. J imaging using sliding-window conjugate-gradient highly constrained
Cardiovasc Magn Reson. 2013;15:91. back-projection reconstruction for detection of coronary artery disease.
2. Kozerke S, Plein S. Accelerated CMR using zonal, parallel and prior Am J Cardiol. 2012;109:1137–1141.
knowledge driven imaging methods. J Cardiovasc Magn Reson. 23. Kholmovski EG, DiBella EVR. Perfusion MRI with radial acquisition for
2008;18:1–18. arterial input function assessment. Magn Reson Med. 2007;57:821–827.
3. Motwani M, Lockie T, Greenwood JP, Plein S. Accelerated, high spatial 24. Ge L, Kino A, Griswold M, Carr JC, Li D. Free-breathing myocardial
resolution cardiovascular magnetic resonance myocardial perfusion perfusion MRI using SW-CG-HYPR and motion correction. Magn Reson
imaging. J Nucl Cardiol. 2011;18:952–958. Med. 2010;64:1148–1154.
4. Tsao J, Kozerke S. MRI temporal acceleration techniques. J Magn Reson 25. Salerno M, Sica CT, Kramer CM, Meyer CH. Optimization of spiral-
Imaging. 2012;36:543–560. based pulse sequences for first-pass myocardial perfusion imaging. Magn
5. Fair MJ, Gatehouse PD, DiBella EVR, Firmin DN. A review of 3D Reson Med. 2011;65:1602–1610.
first-pass, whole-heart, myocardial perfusion cardiovascular magnetic 26. Shin T, Nayak KS, Santos JM, Nishimura DG, Hu BS, McConnell MV.
resonance. J Cardiovasc Magn Reson. 2015;17:68. Three-dimensional first-pass myocardial perfusion MRI using a
6. Plein S, Kozerke S, Suerder D, et al. High spatial resolution myocardial stack-of-spirals acquisition. Magn Reson Med. 2013;69:839–844.
perfusion cardiac magnetic resonance for the detection of coronary 27. Chen L, Adluru G, Schabel MC, McGann CJ, Dibella EV. Myocardial
artery disease. Eur Heart J. 2008;29:2148–2155. perfusion MRI with an undersampled 3D stack-of-stars sequence. Med
7. Plein S, Schwitter J, Suerder D, Greenwood JP, Boesiger P, Kozerke S. k-Space Phys. 2012;39:5204–5211.
and time sensitivity encoding–accelerated myocardial perfusion MR imaging 28. Salerno M, Taylor A, Yang Y, et al. Adenosine stress cardiovascular
at 3.0 T: comparison with 1.5 T1. Radiology. 2008;249:493–500. magnetic resonance with variable-density spiral pulse sequences
8. Manka R, Vitanis V, Boesiger P, Flammer AJ, Plein S, Kozerke S. Clinical accurately detects coronary artery disease initial clinical evaluation. Clin
feasibility of accelerated, high spatial resolution myocardial perfusion Cardiovasc Imaging. 2014;7:639–646.
imaging. JACC Cardiovasc Imaging. 2010;3:710–717. 29. Otazo R, Kim D, Axel L, Sodickson D. Combination of compressed
9. Lockie T, Ishida M, Perera D, et al. High-resolution magnetic resonance sensing and parallel imaging for highly accelerated first-pass cardiac
myocardial perfusion imaging at 3.0-Tesla to detect hemodynamically perfusion MRI. Magn Reson Med. 2010;64:767–776.
significant coronary stenoses as determined by fractional flow reserve. J 30. Lustig M, Donoho D, Pauly JM. Sparse MRI: the application of
Am Coll Cardiol. 2011;57:70–75. compressed sensing for rapid MR imaging. Magn Reson Med. 2007;58:
10. Motwani M, Maredia N, Fairbairn TA, et al. High-resolution versus 1182–1195.
standard-resolution cardiovascular MR myocardial perfusion imaging for 31. Feng L, Srichai MB, Lim RP, et al. Highly accelerated real-time cardiac
the detection of coronary artery disease. Clin Cardiovasc Imaging. cine MRI using k-t SPARSE-SENSE. Magn Reson Med. 2013;70:64–74.
2012;5:306–313. 32. Gamper U, Boesiger P, Kozerke S. Compressed sensing in dynamic MRI.
11. Maredia N, Radjenovic A, Kozerke S, Larghat A, Greenwood JP, Plein S. Magn Reson Med. 2008;59:365–373.
Effect of improving spatial or temporal resolution on image quality and 33. Jung H, Sung K, Nayak KS, Kim EY, Ye JC. k-t FOCUSS: a general
quantitative perfusion assessment with k-t SENSE acceleration in first-pass compressed sensing framework for high resolution dynamic MRI. Magn
CMR myocardial perfusion imaging. Magn Reson Med. 2010;64:1616–1624. Reson Med. 2009;61:103–116.
12. Shin T, Hu HH, Pohost GM, Nayak KS. Three dimensional first-pass 34. Di Bella EVR, Parker DL, Sinusas AJ. On the dark rim artifact in dynamic
myocardial perfusion imaging at 3T: feasibility study. J Cardiovasc Magn contrast-enhanced MRI myocardial perfusion studies. Magn Reson Med.
Reson. 2008;10:57. 2005;54:1295–1299.
13. Manka R, Jahnke C, Kozerke S, et al. Dynamic 3-dimensional stress 35. Sharif B, Dharmakumar R, Labounty T, et al. Towards elimination of the
cardiac magnetic resonance perfusion imaging: detection of coronary dark-rim artifact in first-pass myocardial perfusion MRI: removing gibbs
artery disease and volumetry of myocardial hypoenhancement before and ringing effects using optimized radial imaging. Magn Reson Med. 2014;72:
after coronary stenting. J Am Coll Cardiol. 2011;57:437–444. 124–136.
14. Manka R, Paetsch I, Kozerke S, et al. Whole-heart dynamic three- 36. Harrison A, Adluru G, Damal K, et al. Rapid ungated myocardial
dimensional magnetic resonance perfusion imaging for the detection of perfusion cardiovascular magnetic resonance: preliminary diagnostic
coronary artery disease defined by fractional flow reserve: determination accuracy. J Cardiovasc Magn Reson. 2013;15:26.
of volumetric myocardial ischaemic burden and coronary lesion location. 37. Bache RJ, Schwartz JS. Effect of perfusion pressure distal to a coronary
Eur Heart J. 2012;33:2016–2024. stenosis on transmural myocardial blood flow. Circulation. 1982;65:928–935.
15. Jogiya R, Kozerke S, Morton G, et al. Validation of dynamic 38. Motwani M, Maredia N, Fairbairn TA, Kozerke S, Greenwood JP, Plein S.
3-dimensional whole heart magnetic resonance myocardial perfusion Assessment of ischaemic burden in angiographic three-vessel coronary
imaging against fractional flow reserve for the detection of significant artery disease with high-resolution myocardial perfusion cardiovascular
coronary artery disease. J Am Coll Cardiol. 2012;68:756–765. magnetic resonance imaging. Eur Heart J Cardiovasc Imaging. 2014;15:
16. Motwani M, Fairbairn TA, Larghat A, Mather AN, Biglands JD. Systolic 701–708.
versus diastolic acquisition in myocardial perfusion MR imaging. 39. Hautvast GLTF, Chiribiri A, Lockie T, Breeuwer M, Nagel E, Plein S.
Radiology. 2012;262:816–823. Quantitative analysis of transmural gradients in myocardial perfusion
17. Plein S, Ryf S, Schwitter J, Radjenovic A, Boesiger P, Kozerke S. Dynamic magnetic resonance images. Magn Reson Med. 2011;1487:1477–1487.
contrast-enhanced myocardial perfusion MRI accelerated with k-t sense. 40. Chiribiri A, Hautvast GLTF, Lockie T, et al. Assessment of coronary artery
Magn Reson Med. 2007;58:777–785. stenosis severity and location: quantitative analysis of transmural
18. Tsao J, Boesiger P, Pruessmann KP. k-t BLAST and k-t SENSE: dynamic perfusion gradients by high-resolution MRI versus FFR. JACC Cardiovasc
MRI with high frame rate exploiting spatiotemporal correlations. Magn Imaging. 2013;6:600–609.
Reson Med. 2003;50:1031–1042. 41. Sharif B, Dharmakumar R, Arsanjani R, et al. Non-ECG-gated myocardial
19. Pedersen H, Kozerke S, Ringgaard S, Nehrke K, Kim WY. k t PCA: perfusion MRI using continuous magnetization-driven radial sampling.
temporally constrained k t BLAST reconstruction using principal Magn Reson Med. 2014;72:1620–1628.
component analysis. Magn Reson Med. 2009;62:706–716. 42. Sharif B, Arsanjani R, Dharmakumar R, Bairey Merz CN, Berman DS, Li
20. Ge L, Kino A, Griswold M, Mistretta C, Carr JC, Li D. Myocardial D. All-systolic non-ECG-gated myocardial perfusion MRI: feasibility of
perfusion MRI with sliding-window conjugate-gradient HYPR. Magn multi-slice continuous first-pass imaging. Magn Reson Med. 2015;74:
Reson Med. 2009;62:835–839. 1661–1674.
65.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

43. Jogiya R, Morton G, De Silva K, et al. Ischemic burden by three- 56. Wilke N, Jerosch-Herold M, Wang Y, et al. Myocardial perfusion reserve:
dimensional myocardial perfusion cardiovascular magnetic resonance: assessment with multisection, quantitative, first-pass MR imaging.
comparison with myocardial perfusion scintigraphy. Clin Cardiovasc Radiology. 1997;204:373–384.
Imaging. 2014;7:647–654. 57. Costa MA, Shoemaker S, Futamatsu H, et al. Quantitative magnetic
44. Shin T, Pohost GM, Nayak KS. Systolic 3D first-pass myocardial perfusion resonance perfusion imaging detects anatomic and physiologic coronary
MRI: comparison with diastolic imaging in healthy subjects. Magn Reson artery disease as measured by coronary angiography and fractional flow
Med. 2010;63:858–864. reserve. J Am Coll Cardiol. 2007;50:514–522.
45. Motwani M, Kidambi A, Fairbairn T, et al. Quantitative whole-heart 58. Patel AR, Antkowiak PF, Nandalur KR, et al. Assessment of advanced
three-dimensional magnetic resonance myocardial perfusion imaging in coronary artery disease advantages of quantitative cardiac magnetic
systole and diastole at 3.0T. J Cardiovasc Magn Reson. 2013;15:P206. resonance perfusion analysis. J Am Coll Cardiol. 2010;56:561–569.
46. Motwani M, Kidambi A, Sourbron S, et al. Quantitative three- 59. Futamatsu H, Wilke N, Klassen C, et al. Evaluation of cardiac magnetic
dimensional cardiovascular magnetic resonance myocardial perfusion resonance imaging parameters to detect anatomically and
imaging in systole and diastole. J Cardiovasc Magn Reson. 2014; hemodynamically significant coronary artery disease. Am Heart J.
16:19. 2007;154:298–305.
47. Radjenovic A, Biglands JD, Larghat A, et al. Estimates of systolic and 60. Jerosch-Herold M, Wilke N, Stillman AE, Wilson RF. Magnetic resonance
diastolic myocardial blood flow by dynamic contrast-enhanced MRI. quantification of the myocardial perfusion reserve with a Fermi function
Magn Reson Med. 2010;1703:1696–1703. model for constrained deconvolution. Med Phys. 1998;25:73–84.
48. Motwani M, Kidambi A, Uddin A, Sourbron S, Greenwood JP, Plein S. 61. Vitanis V, Manka R, Giese D, et al. High resolution three-dimensional
Quantification of myocardial blood flow with cardiovascular magnetic cardiac perfusion imaging using compartment-based k-t principal
resonance throughout the cardiac cycle. J Cardiovasc Magn Reson. component analysis. Magn Reson Med. 2011;65:575–587.
2015;17:4. 62. Sourbron S. Technical aspects of MR perfusion. Eur J Radiol.
49. Manka R, Wissmann L, Gebker R, et al. Multicenter evaluation of 2010;76:304–313.
dynamic three-dimensional magnetic resonance myocardial perfusion 63. Wissmann L, Niemann M, Gotschy A, Manka R, Kozerke S. Quantitative
imaging for the detection of coronary artery disease defined by fractional three-dimensional myocardial perfusion cardiovascular magnetic
flow reserve. Clin Cardiovasc Imaging. 2015;8:1–8. resonance with accurate two-dimensional arterial input function
50. Wang H, Bangerter NK, Park DJ, et al. Comparison of centric and assessment. J Cardiovasc Magn Reson. 2015;17:1.
reverse-centric trajectories for highly accelerated three-dimensional 64. Sammut E, Zarinabad N, Wesolowski R, et al. Feasibility of high-
saturation recovery cardiac perfusion imaging. Magn Reson Med. resolution quantitative perfusion analysis in patients with heart failure. J
2015;74:1070–1076. Cardiovasc Magn Reson. 2015;17:1.
51. Akçakaya M, Basha TA, Pflugi S, et al. Localized spatio-temporal 65. Kim TH, Pack NA, Chen L, DiBella EVR. Quantification of myocardial
constraints for accelerated CMR perfusion. Magn Reson Med. 2014; perfusion using CMR with a radial data acquisition: comparison with a
72:629–639. dual-bolus method. J Cardiovasc Magn Reson. 2010;12:1.
52. Jackson JI, Nishimura DG, Macovski A. Twisting radial lines with 66. Chen D, Sharif B, Dharmakumar R, et al. Quantification of myocardial
application to robust magnetic resonance imaging of irregular flow. Magn blood flow using non–ECG-triggered MR imaging. Magn Reson Med.
Reson Med. 1992;25:128–139. 2015;74:765–771.
53. Pipe JG. Motion correction with PROPELLER MRI: application to head 67. Schulz-Menger J, Bluemke DA, Bremerich J, et al. Standardized image
motion and free-breathing cardiac imaging. Magn Reson Med. 1999;42: interpretation and post processing in cardiovascular magnetic resonance:
963–969. Society for Cardiovascular Magnetic Resonance (SCMR) Board of
54. Christian TF, Rettmann DW, Aletras AH, et al. Absolute myocardial Trustees Task Force on Standardized Post Processing. J Cardiovasc Magn
perfusion in canines measured by using dual-bolus first-pass MR Reson. 2013;15:35.
imaging. Radiology. 2004;232:677–684. 68. McDiarmid AK, Ripley DP, Mohee K, et al. Three-dimensional whole-
55. Jerosch-Herold M, Swingen C, Seethamraju RT. Myocardial blood flow heart vs. two-dimensional high-resolution perfusion-CMR: a pilot study
quantification with MRI by model-independent deconvolution. Med Phys comparing myocardial ischaemic burden. Eur Hear J Cardiovasc Imaging.
AAPM. 2002;29:886–897. 2016;17:900–908.
6 
Blood Flow Velocity Assessment
David Firmin

The idea of mapping measurements of blood flow onto a magnetic Methods have also been described in which the TOF flow movement
resonance (MR) image was first discussed in an article by Singer in can be visualized directly on an image.12,13 The methods involved the
1978.1 The methods that followed could generally be categorized into application of slice selection and frequency encoding in the same axis.
time-of-flight (TOF) or phase shift types and were based on the tech- In this way, material that had moved in this axis between selection and
niques that had previously been described for nonimaging MR flow readout would be displaced relative to the stationary material. These
studies.2 A number of review articles have covered the subject and techniques were therefore making use of signal misregistration, an effect
described the variety of methods3–5 that have been used and validated that is often seen as a problem in other methods of flow imaging.
both in vitro and in vivo. The interest in flow in MR imaging has not Another TOF approach is to saturate a band of tissue, for example, in
been solely directed toward the goal of quantitative flow measurement. a transverse plane, and then to follow the progress of this dark band
A large amount of effort has also been devoted to understanding the in the coronal or sagittal plane.14 The major limitation of these satura-
appearance of a flowing fluid on an image because this can often be tion methods is that they are limited by the T1 of the various tissues
indicative of the type of flow present and therefore can give important being saturated. The contrast of the saturated blood will decrease with
information on the diagnosis of a particular disorder. Also, the develop- time, eventually making it difficult to measure accurately the distances
ment of MR angiography techniques has required a full understanding traveled. Also, motion during the sampling gradients results in signal
of these effects. and thus image distortion.15 In addition, for arterial flow measurements
In 1984, soon after the development of the first clinical MR scanners, in which cardiac gating is required, only two-dimensional (2D) images
there was an increase in interest in the search for an MR method of can be acquired in a reasonable time so that only limited details of the
imaging flow. Review articles were published and a number of techniques flow profile can be studied.
described.6–10 This chapter provides a description and brief historical
overview of the methods that have been used to measure blood flow
in the heart and great vessels.
PHASE FLOW IMAGING METHODS
Considerable knowledge had been gained on the measurement of flow
from the phase of the cardiac magnetic resonance signal from the non-
TIME-OF-FLIGHT METHODS imaging studies after an original suggestion by Hahn in 1960 of a method
There are two categories of TOF techniques. The first, often known as of measuring the slow flow of currents in the sea.16 In 1984, the first
wash-in/wash-out, or flow enhancement, normally rely on the satura- attempts of measuring blood flow were described by van Dijk8 and
tion or partial saturation of material in a selected slice or volume being Bryant and associates,9 using methods based on the theory suggested
replaced by fully magnetized “high signal” spins as a result of flow (Fig. by Moran 2 years earlier.17 The imaging methods that followed fell
6.1A). The second involves some form of tagging and then imaging to broadly into two categories:
follow the motion of the tagged material (Fig. 6.1B). 1. Phase contrast velocity mapping methods that mapped the phase
Singer and Crooks11 adopted the first approach in an attempt to of the signal directly to measure the flow.
measure flow in the internal jugular veins, although quantification 2. Fourier flow imaging methods that phase encoded flow velocity to
was questionable because of other factors affecting the flow signal. produce an image after Fourier transformation with velocity resolved
The first report to describe a tagged TOF approach was by Feinberg on one image axis.
and colleagues.10 Their method involved a variation on a dual echo Both of these methods rely on the same principles that cause flowing
spin echo sequence; the first 180-degree selected slice was displaced by material to attain a phase shift that is related to its motion. Fig. 6.2
3 mm from the initial excitation slice, and the second was displaced by shows these principles for a fluid flowing down a tube surrounded by
9 mm. The first 180-degree selection overlapped sufficiently with the stationary material. A bipolar gradient pulse is applied, consisting of
90-degree selection to produce a good anatomic image. The second a positive magnetic field gradient, followed a certain time later by an
180-degree pulse selection did not overlap with the 90-degree or the equal but opposite negative magnetic field gradient in the direction
first 180-degree pulse selection and therefore produced no anatomic of the flow. During the period of the positive gradient, flowing and
image but gave high signal from the blood that had experienced all of stationary materials in a particular location will take up a frequency
the preceding radiofrequency (RF) pulses (i.e., that had passed between shift that depends on their position in the direction of the field gra-
the different selected planes). The technique was used to identify flow dient. When the gradient is turned off, the phase of the flowing and
in the carotid and vertebral arteries of a volunteer’s neck, although flow stationary materials can be considered equal. In the period between the
velocities had to be within a specific range and could not be defined positive and negative gradients, the flowing material moves away from
accurately. its stationary neighbor. During the period of the negative gradient,

66
CHAPTER 6  Blood Flow Velocity Assessment 67

where Ag is the area of one gradient pulse (amplitude × duration), Δ


is the time between the centers of the two gradient pulses, v is the
velocity, and γ is the gyromagnetic ratio. Ag, Δ, and γ are all constants
for a particular imaging sequence so that a quantitative measure of
velocity can be determined if the phase shift can be measured.
The two principal approaches of using the phase shift to produce
a quantitative flow image, phase contrast velocity mapping and Fourier
Signal intensity

flow imaging, are discussed in this chapter.

Phase Contrast Velocity Mapping


The early phase contrast velocity mapping methods8,9 used a spin echo
sequence, which was not ideal because of problems in repeating the
sequence rapidly, and signal loss as a result of shear and other more
complex flows. These problems were reduced and the methods were
A Flow rate
made clinically more useful, partly by the use of a gradient echo
sequence18,19 and, more importantly, by the introduction of velocity-
compensated gradient waveforms.20,21
Normally, two images are acquired with different gradient waveforms
Time 1 Time 2
in the direction of desired flow measurement. The difference in the
B (tagging) (detection)
waveforms is calculated to produce a well-defined velocity-related phase
FIG. 6.1  Time-of-flight approaches to magnetic resonance flow mea- difference between the two images. A phase reconstruction is produced
surement. In the saturation wash-in method (A), the signal is enhanced
for each of the images, which are then subtracted pixel by pixel to
by an amount related to the in-flow of fully magnetized blood. In the
true time-of-flight method (B), there is a time of spatial labeling or tagging,
produce the final velocity map. This process of subtraction removes
followed after a defined period by detection of the movement of the any phase variations that are not related to flow. The velocity phase
labeled blood. sensitivity of the final image is normally set such that the expected
velocity-related phase shifts are within the range of ± π radians. If a
larger range of velocities is present, then aliasing or wrap-around will
occur, resulting in measurement of ambiguous velocities. This problem
can be avoided by reducing the velocity sensitivity or potentially can
be corrected by a process known as phase unwrapping (described later).
Gradient
waveform Fig. 6.3A shows the phase velocity images of a series of time frames
Time Time
from a slice just above the heart. Flow can be seen in the ascending and
descending aortas, pulmonary artery, and superior vena cava. Flow
Magnetic versus time curves throughout the cardiac cycle are shown in Fig. 6.3B.
field Position Position
The stroke volume can be measured by integrating under the aortic or
pulmonary flow curve. The technique has been validated both in vitro
Flow Flow and in vivo22 and is now routinely used to provide useful measurements
in clinical and physiologic flow studies.23
Signal
phase
Fourier Flow Imaging
Stationary Flowing Stationary Flowing Fourier flow imaging normally involves the addition of a bipolar velocity
material material material material
phase-encoding gradient that is stepped through a range of defined
Time 1 Time 2
amplitudes (Fig. 6.4). Because this increases the scan time by a multiple
FIG. 6.2  Principles of phase velocity encoding. At time 1, a positive of the number of steps, it is often applied as a replacement of one of
magnetic field gradient is applied that results in an equal frequency and the spatial phase-encoding gradients. The image is normally recon-
associated phase shift for neighboring stationary and flowing spins. At
structed and displayed with velocity information in one dimension.
time 2, an equal but opposite magnetic field is applied. By this time,
the spins in the flowing blood have separated from their original neigh-
Stationary material is positioned in the center of the image, with faster
bors to be in a different strength of magnetic field during the gradient velocities toward the edge.
application. The result is that although the phase of the stationary spins The method was first described by Redpath and colleagues24 in 1984;
will be returned to zero, the flowing spins will accumulate a phase shift in this case, eight velocity phase-encoding steps were added to a 2D
proportional to the distance moved and hence the velocity. imaging sequence, to image velocities in a circle of fluid-filled tubing
that rotated in the image plane. Different segments of the circle, each
corresponding to different velocity ranges, were seen on the eight resul-
tant images. A year later, Feinberg and coworkers25 applied the method,
the stationary material takes up an equal but opposite frequency shift both in vitro and in vivo, and increased the velocity range and resolution
and returns to the phase it had before the first gradient. However, the by increasing the number of flow phase-encoding steps. In this case,
flowing fluid takes up a different frequency shift that is dependent on however, to maintain a tolerable scan time, only one spatial phase-
the distance it has moved; its final phase therefore also depends on this encoding direction was used so that there was only spatial resolution
distance and hence its velocity. in one direction. The accuracy of the method was shown with a phantom,
The relationship between the phase of the signal and flow velocity is: whereas the in vivo study, which showed the flow in the descending
aorta, highlighted the problem of very high zero velocity signal from
φ = γ v∆Ag Eq. 6.1 the large amount of stationary tissue imaged. In 1988, Hennig and
68 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

45
150 msec 250 msec
AA 40
MPA AA
35 MPA
30 DA
SVC
SVC

Flow (L/min)
25
DA
20
15
300 msec 350 msec 400 msec
10
5
0
–5
0 200 400 600 800
A B
Time (msec)
FIG. 6.3  (A) Flow velocity images of a transverse slice at the level of the right pulmonary artery showing
head/foot flow velocities at six times in the cardiac cycle. (B) Plot of measured volume flow versus time for
the ascending aorta (AA), descending aorta (DA), main pulmonary artery (MPA), and superior vena cava (SVC).

SPATIAL PHASE ENCODING IS REPLACED shift greater than 2π that will result in aliasing that can be corrected
BY VELOCITY PHASE ENCODING by use of a phase unwrapping algorithm (Fig. 6.5).31
Another approach to improving accuracy has been suggested by
Bittoun and associates.32 The method is a combination of phase contrast
velocity mapping and Fourier velocity imaging with a small number of
velocity phase-encoding steps. The final phase contrast velocity map
is calculated from the best fit through the Fourier velocity encoded
result. One potential problem with this method that can also affect
Spatial phase- Velocity phase- conventional phase contrast velocity imaging occurs if there is any
encoding gradient encoding gradient beat-to-beat variation in flow velocity. As a result of the high-velocity
waveform waveform
phase sensitivity used with this method, significant phase variations can
FIG. 6.4  Gradient waveforms required for spatial and velocity phase occur, with resulting ghosting artifacts and loss of flow information.
encoding.
Another method that was originally described to give a measure of
velocity and flow quantification was phase contrast angiography. This
technique again involves acquiring two image datasets with sequences
having opposite phase velocity sensitivities, although in this case, the
colleagues26 described a development of this method in which the signal raw data are subtracted before reconstruction. This technique has the
from stationary tissue was saturated and the sequence was repeated advantage of subtracting out signal from stationary tissue, which removes
much more rapidly. The issue of time precluding the use of spatial errors caused by partial volumes where voxels contained a mixture of
phase encoding remains a problem, although 2D RF pulses have been flowing and stationary tissue. The method is, however, generally less
used successfully to locate signals within a column.27,28 More recently, accurate because the signal and hence the velocity measurement can be
Luk Pat and associates29 showed a method of real-time Fourier velocity affected by factors such as in-flow enhancement and intravoxel dephas-
imaging that used an excited column to localize the signals. In vivo ing (signal loss). In the mid-1990s, Polzin and colleagues33 suggested
aortic flow waveforms were presented with a temporal resolution of combining this method with phase contrast velocity mapping, which
only 33 ms. they showed to be more accurate in a number of phantom studies. The
methods are yet to be fully validated in vivo, however, and are likely to
IMPROVING THE ACCURACY OF PHASE CONTRAST be affected by problems of signal loss and motion, particularly when
imaging is performed on small mobile vessels, such as the coronary
VELOCITY MEASUREMENTS arteries.34
The vast majority of MR flow imaging applications have used the method One of the most significant factors affecting the accuracy of the
of phase contrast velocity mapping. The accuracy of this method is flow measurement methods is flow-related signal loss. This is normally
highly dependent on such factors as flow pulsatility, velocity, and size and the result of loss of phase coherence within a voxel, and eventually it
tortuosity of the vessel. One simple approach to improving the overall results in an inability to detect the encoded phase of the flow signal
accuracy of the method is to adjust the velocity sensitivity of the sequence above the random phase of the background noise. Even if a velocity-
so that the velocity-related phase shift is close to 2π for the maximum compensated imaging sequence is used, the acceleration and even the
expected velocity. Buonocore30 extended this approach by varying the higher orders of motion present in complex flows can result in loss of
velocity sensitivity during the cardiac cycle, based on the knowledge phase coherence. Fig. 6.6 shows an example of a long-axis image of a
that the arterial flow velocity is high in systole but low in diastole. The patient with a mitral valve stenosis in which the valve is also regurgitant.
accuracy can be improved further by allowing a velocity-related phase In this case, a region of blood signal is lost from the ventricle during
CHAPTER 6  Blood Flow Velocity Assessment 69

A B

C D
FIG. 6.5  Method of phase unwrapping. (A) Systolic image in which high velocities result in aliasing in both
the positive and negative directions. (B and C) Adjustment of the velocity window to remove aliasing in the
positive and negative directions, respectively. (D) The same image data after processing by the antialiasing
algorithm.

LV

LA

FIG. 6.6  Systolic and diastolic long-axis frames from cine datasets acquired in a patient with a stenotic and
regurgitant mitral valve. Signal loss can be seen in the left atrium (LA) during systole and the left ventricle
(LV) during diastole.

diastole as a result of the stenosis generating complex flows and from appropriate gradient profile design. A good way to reduce signal loss
the atrium during systole because of a regurgitant jet of flow through is to use a symmetrical gradient waveform that nullifies phase shifts
the valve. Partial signal loss, however, does not greatly affect the accuracy caused by all of the odd-order derivatives of position and then to shorten
of the phase contrast velocity mapping measurement unless it is accom- the sequence as much as possible to reduce the effects of the even-order
panied by partial volume errors. When signal loss is the result of a derivatives.36 Signal loss of the type described is much less of a problem
spread of phase within a voxel, the mean phase will be detected, although with the Fourier flow imaging method. In this case, the Fourier trans-
this will be affected by differential saturation effects.35 The phase contrast form is used to separate out constituent velocities.
velocity mapping techniques are most susceptible to signal loss of one Errors can be caused by phase differences for reasons other than
form or another, although this can normally be minimized by the velocity encoding pulses, resulting in the phase map velocity values
70 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

being offset from zero, even for stationary tissues.37 This background
phase offset generally varies gradually with position across the image, Rapid Phase Flow Imaging Methods
and it also varies with image plane orientation and other sequence With the very rapid scanning hardware available today, it is possible to
parameters that affect the gradient waveforms, such as Venc. Distortion repeat a phase contrast velocity sequence so fast that low-resolution
of the requested magnetic field gradients is unavoidable because of images can be acquired in 100 ms or high-resolution images can be
the fundamental laws of electromagnetism, and these are known in acquired in a breath-hold. The major problem is for pulsatile flow
MR imaging as Maxwell, or concomitant, gradients. These background where the accuracy of the measurements and the temporal resolution
phase shifts become more significant when high-amplitude gradients can be limited if the acquisition period per cardiac cycle is too long.
are used and also when imaging is performed at lower main magnetic Also, if high spatial resolution is required, the cardiac motion of struc-
field strengths. However, with the latest gradient systems, Maxwell tures, such as coronary arteries, can cause blurring, with subsequent
gradient effects are certainly a factor for phase velocity mapping at errors in flow measurement.49 For this reason, it is likely that more
1.5 T. However, these velocity map offsets can be corrected precisely efficient k-space coverage methods, such as interleaved spirals, will be
and automatically in software, with no user intervention required.38 important.
A second common reason for background phase shifts is the pres- Ultra-fast flow imaging techniques have also been developed, either
ence of small uncorrected side effects of the gradient pulses in the by combining a phase-mapping type approach with imaging methods,
magnet, known as eddy currents. These phase shifts became more of such as single-shot echo planar and spiral imaging,50,51 or by imaging
a problem with the advent of higher-performance gradients, although only one spatial dimension.52 A compromise generally has to be made
more recently the problem appears to have been reduced. Software is in temporal or spatial resolution and probably also in the signal-to-
sometimes provided that allows the user to place markers identifying noise ratio. However, taking into account these constraints, the methods
stationary tissues so that this background phase error can be calculated have generally been shown to be accurate. One complication with the
and removed from the entire image. Sometimes, however, if the phase echo planar sequence is flow signal loss because of its inherent phase
shifts are nonlinear or if there is little signal from stationary tissue, sensitivity, even when additional flow compensation is applied. However,
then the only way to correct for them is to acquire an additional set of this has been used to advantage for more qualitative flow imaging
images of a large static phantom using the same sequence parameters showing flow disturbances, for example.53
as those used in vivo, and then to subtract out the phase errors on a The one-dimensional rapid acquisition mode, real-time acquisition
pixel-by-pixel basis.39 and velocity evaluation (RACE),52 can be used to measure flow per-
Pixels without any signal in velocity maps have a random phase or pendicular to the slice. The technique can be repeated rapidly throughout
show the phase of a weak ghost that may not be visible on the magnitude the cardiac cycle to give near-real-time flow information. One problem
image with normal brightness settings. Particularly for poststenotic with this type of approach is that data are acquired from a projection
jet images, it is important to check that the magnitude image pixels through the patient; this means that any signal overlapping with the
of the jet are not affected by signal loss. Avoiding the inclusion of flow signal will combine and introduce errors to flow measurement.
noise pixels can be problematic in regions of interest around the great Several strategies have been suggested for localizing the signal to avoid
vessels, and ideally, the image analysis software enables the user to set this: they include spatial presaturation, projection dephasing (applying
the velocity to zero for pixels whose magnitude is below a user-defined a gradient to suppress stationary tissue), and collecting a cylinder of
threshold. data and multiple oblique measurements.
Provided that the sequence parameters are carefully chosen such Yang and colleagues used a 2D RF excitation scheme to excite a
that the potential errors and artifacts discussed earlier can be minimized narrow rectangular X-section column and used only 16 echoes to spa-
or avoided, phase velocity mapping has been shown to be accurate and tially resolve the other dimension in high resolution.54 This approach
reproducible. Validation has been reported in phantoms by comparison allowed real-time flow measurements to be acquired. The authors used
with true measured flow and with Doppler36,40,41 in animal models by the method to show the effect of controlled breathing on flow in the
comparison with in vivo flow meter measurements.42 Validation has ascending aorta and superior vena cava (Fig. 6.7).
also been reported in humans by comparison with methods such as
Doppler ultrasound or catheterization.43–45 Perhaps the most convincing
forms of validation were those performed initially by comparing the
aortic flow with the left ventricular stroke volume and later flow in Magnitude
both the aorta and pulmonary artery with left and right ventricular
Velocity
stroke volumes in normal subjects.22,46,47 For the latter, the four mea-
surements should be the same except for small differences caused by
coronary and bronchial flow, and it can be calculated that flow mea- 600 Aortic flow
surements in large vessels are accurate to within 6%. 20
The effect of breath-holding on flow measurement is another factor Expiration
400 Diaphragm position 10
(mm)/sec

that could affect more recent studies. Sakuma and associates showed a
(mm)

significant change in both pulmonary and aortic cardiac output during 0


a large lung volume breath-hold.48 Conversely, flows measured during 200 –10
a small volume breath-hold were found to be similar to those measured SVC flow Inspiration
–20
during normal breathing.
There are other potential sources of error that have been reported. 0 –30
These include misalignment of the vessel with the direction of velocity 1 4 7 10 13 16 19 22
encoding and misregistration of flow signal caused by flow between Cardiac cycles
excitation and readout. Because of these and the other sources of errors FIG. 6.7  Real-time magnitude and flow images from the excited region
discussed earlier, care is required when setting up the scan parameters, containing the aorta and the superior vena cava (SVC). The plot shows
to minimize their effect. the variation in flow during a period of respiratory maneuver.
CHAPTER 6  Blood Flow Velocity Assessment 71

shown previously in Fig. 6.3A. When flow is measured in more than


Visualizing Flow and Flow Parameters one direction, more sophisticated methods of display can be used. Fig.
The method used to visualize MR flow data has depended on the method 6.9 shows a vector map of flow in the root of the aorta of a patient
used for acquisition. For Fourier velocity measurement, each voxel may with an atherosclerotic aneurysm. This systolic image, shown alongside
contain a range of measured velocities, and the Fourier velocity image a pressure map (described later), shows high-velocity flow impinging
normally takes the form of a plot of velocity versus time or velocity on the wall of the aneurysm. An alternative type of representation
versus position in one direction. Fig. 6.8 shows an example in which would be to use the cine velocity images to calculate the path of a seed
velocity images were acquired from a column of excited tissue, including over time.56
the descending aorta.55 The front edge of the aortic pulse wave can be The ability to study flow in such detail and at any site in the body
seen on successive frames as it travels down the vessel. is unique to MR imaging. For this reason, a large amount of interest
Phase contrast velocity images contain only one velocity measure is being generated from those who wish to understand the physiology
per image voxel. Historically, these have been shown with a gray scale of blood flow and its interaction with blood vessels and the cardiac
such that flow in one direction tends toward white, flow in the other chambers. Despite the relatively poor spatial resolution, a number of
direction tends toward black, and stationary material is mid-gray, as groups have studied methods of extracting a measure of the wall shear
stress from the MR images. Both Oshinski and colleagues and Oyre
and associates developed fitting methods to derive the velocity profile
at subpixel distances from the vessel wall.57,58 Both groups presented
expected values of stress, although it is difficult to suggest a method
ECG Gate Delay
of validating the accuracy of these measurements. Frayne and Rutt
suggested an alternative approach that potentially gave more infor-
100 msec 105 msec 115 msec 125 msec 135 msec 145 msec 155 msec 165 msec 175 msec
mation about the flow within a voxel that straddled the vessel wall.59
Their method used Fourier velocity encoding to distinguish the dis-
tribution of flow velocities, so that only the spatial location had to be
considered.
There has also been considerable interest in the possibility of deriv-
250 mm FOV

ing pressure measurements from MR images. Urchuk and coworkers


considered vessel compliance and the flow pulse wave to calculate the
pressure waveform and showed a good correlation with catheter pres-
sure measurements made in a pig model.60 In contrast, Yang and col-
leagues derived flow pressure maps from the cine phase contrast velocity
maps using the Navier-Stokes equations.61 Fig. 6.10 shows an example
of the changing flow pressure around the aortic arch during the first
half of the cardiac cycle. Fig. 6.11 shows an interesting example of a
flow pressure map, showing the descending aorta in a patient who
0 168 cm/sec underwent polyester fiber (Dacron, Dupont, Wilmington, DE) graft
FIG. 6.8  Series of nine Fourier velocity images at 10-ms intervals showing repair of aortic coarctation. In contrast to the rest of the aorta, no
the velocity pulse wave propagating down the descending aorta. ECG, pressure gradient can be seen in the repaired region, possibly because
Electrocardiogram; FOV, field of view. of the reduced compliance.

Flow Pattern Pressure

FIG. 6.9  Flow vector map showing the systolic flow pattern in the aortic root of a patient with an athero-
sclerotic aneurysm. The associated image shows the corresponding pressure distribution.
72 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

66 (msec) 162 (msec) 258 (msec) 354 (msec) 450 (msec)

A B C D E
FIG. 6.10  Selected frames from a cine series of calculated flow pressure maps showing the variation in
pressure at different times in the cardiac cycle (A to E). Each gray-scale band from white to black represents
a pressure gradient of 1 mm Hg. A positive pressure gradient during systole (A) reverses during the decel-
eration phase of diastole (D).

FIG. 6.11  Systolic pressure map of the aortic arch in patient with a polyester fiber (Dacron, Dupont, Wil­
mington, DE) repair. No pressure gradient is seen in the region of the repair.
CHAPTER 6  Blood Flow Velocity Assessment 73

Pathlines emitted from the mitral valve at approx. time of peak A-wave (left panel) and traced to early systole (right panel)
TECG = 0.66 sec TECG = 0.70 sec TECG = 0.77 sec TECG = 0.80 sec

AAo
LA

LV

A
Instantaneous streamlines at E-wave Instantaneous streamlines at A-wave Instantaneous streamlines at peak systole

B C D
FIG. 6.12  Illustrative examples of four-dimensional flow cardiovascular magnetic resonance visualization
techniques in the normal heart. In these examples, flow visualization is overlaid onto a two-dimensional
bSSFP acquisition in a three-chamber view. Pathlines, the trajectories that massless fluid particles would
follow, allow the study of the path of pulsatile blood flow over time. (A) The transit of blood through the left
ventricle (LV) is shown by pathlines emitted from the mitral valve at the timepoint of the peak of the A-wave
and traced to the time point of early systole. AAo, Ascending aorta; LA, left atrium. (B–D) Streamlines gener-
ated in a long-axis plane show parts of the intracardiac velocity field at the timepoints of peak early filling
(E-wave), peak late filling (A-wave), and peak systole.86 (From Dyverfeldt P, Bissell M, Barker AJ, et al. 4D
flow cardiovascular magnetic resonance consensus statement. J Cardiovasc Magn Reson. 2015;17:72.)

Four-Dimensional Phase Contrast Flow Although the clinical importance of the four-dimensional (4D)
Velocity Mapping approach is still not entirely clear, a number of studies have shown that
To fully understand, visualize, and measure flow parameters in blood similar quantitative measures of volume flow are made when compared
vessels, a method of acquisition is required that measures velocity in with 2D.66–74 4D flow measurements share the same problems as 2D
three dimensions and three directions over time, with high spatial and with regard to background phase errors;75,76 however, there are a number
temporal resolution. This has presented a significant problem in terms of advantages with regard to volume flow measurement. One important
of acquisition and data handling, but methods have been developed advantage is that several volume flow measures from the same or dif-
and are now used routinely in clinical research applications. Following ferent vessels can be made from the same acquisition dataset, and this
early implementation by Firmin et al.62 in 1993 and then improvements enables the employment of the principle of conservation of mass to
by Wigstrom et al. in 1996,63 particularly more recently, there has been check the internal consistency of the data.67,73,77–85 Another advantage
considerable work to speed up the acquisition by a variety of accelera- is that analysis planes can be retrospectively placed and located at will
tion techniques.64,65 during the time of analyzing the flow data and this can be particularly
74 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Velocity [m/s]
1.00

0.75

0.50

0.25

0.00

A tECG = 133 ms D tECG = 255 ms

B tECG = 173 ms C tECG = 204 ms E tECG = 296 ms

FIG. 6.13  (A–E) Visualization of multidirectional flow structures in a normal thoracic aorta illustrating the
temporal evolution of blood flow at five different time points in systole. Pathlines were repetitively emitted
at successive instants and originate from two emitter planes, one in the ascending aorta and one in the
proximal descending aorta. Color coding reflects the local absolute velocity.90 (From Markl M, Kilner PJ,
Ebbers T. Comprehensive 4D velocity mapping of the heart and great vessels by cardiovascular magnetic
resonance. J Cardiovasc Magn Reson. 2011;13:7.)

advantageous when a large number of flow planes are required.71 Unlike different cardiovascular diseases.56,87–90 Studies have reported applica-
with 2D flow measurement, where great care and expertise can be tions in the heart chambers and connecting valves3,79,91–106 (Fig. 6.12),
required during the scan for the placement of the flow imaging planes, the great vessels77,106–124 (Fig. 6.13), carotid and intracranial arteries and
the three-dimensional volume of the 4D dataset can be more simply veins,125–136 hepatic arterial and portal venous systems of the liver,82,122,137–139
positioned. An additional potential advantage is that with the improved renal arteries,140,141 and peripheral arteries.142 Additionally, in complex
coverage available through the third spatial dimension, the chances of congenital heart disease there are also promising applications.74,122,143,144
capturing the peak velocity through a valve jet should be increased.72
However, contradictory studies have shown lower velocities measured
by the 4D approach,70 a finding that might be explained by the relatively
REFERENCES
low temporal resolution (50–55 ms).86 A full reference list is available online at ExpertConsult.com
The most striking feature of 4D flow imaging is in its use to intuitively
visualize multidirectional flow structures and how these are affected by
CHAPTER 6  Blood Flow Velocity Assessment 74.e1

REFERENCES In: Proceedings of the Third Annual Meeting of the International Society of
Magnetic Resonance. 1995;318.
1. Singer JR. NMR diffusion and flow measurement and an introduction 28. Hardy CJ, Bolster BD Jr, McVeigh ER, et al. Pencil excitation with
to spin phase graphing. J Phys E Sci Instrum. 1978;11:281–291. interleaved Fourier velocity encoding: NMR measurement of aortic
2. Jones DW, Child TF. NMR in flowing systems. Adv Magn Reson. 1976;8: distensibility. Magn Reson Med. 1996;35:814–819.
123–148. 29. Luk Pat GT, Pauly JM, Hu BS, Nishimura DG. One-shot spatially
3. Bradley WG. Flow phenomenon in MR imaging. AJR Am J Roentgenol. resolved velocity imaging. Magn Reson Med. 1998;40:603–613.
1988;150:983–994. 30. Buonocore MH. Blood flow measurement using variable velocity
4. Alfidi RJ, Masaryk TJ, Haacke EM, et al. MR angiography of peripheral, encoding in the RR interval. Magn Reson Med. 1993;29:790–795.
carotid, and coronary arteries. AJR Am J Roentgenol. 1987;149:1097. 31. Yang GZ, Burger P, Kilner PJ, et al. Dynamic range extension of cine
5. Firmin DN, Dumoulin C, Mohiaddin RH. Quantiative MR flow velocity measurements using motion registered spatio-temporal phase
measurement. In: Haacke EM, Potchen EJ, Gottschalk A, Siebert JE, eds. unwrapping. J Magn Reson Imag. 1996;6:495–502.
Magnetic Resonance Angiography: Concepts and Applications. St Louis: 32. Bittoun J, Bourroul E, Jolivet O, et al. High-precision MR velocity
Mosby; 1993:187–219. mapping by 3D-Fourier phase encoding with a small number of
6. Crooks LE, Kaufman L. NMR imaging of blood flow. Br Med Bull. encoding steps. Magn Reson Med. 1993;29:674–680.
1984;40:167–169. 33. Polzin JA, Alley MT, Korosec FR, et al. A complex-difference phase-
7. Axel L. Blood flow effects in magnetic resonance imaging. AJR Am J contrast technique for measurement of volume flow rates. J Magn Reson
Roentgenol. 1984;143:1157–1166. Imaging. 1995;5:129–137.
8. van Dijk P. Direct cardiac NMR imaging of heart wall and blood flow 34. Frayne R, Polzin JA, Mazaheri Y, Grist TM, Mistretta CA. Effect of and
velocity. J Comput Assist Tomogr. 1984;8:429–436. correction for in-plane myocardial motion on estimates of coronary
9. Bryant DJ, Payne JA, Firmin DN, Longmore DB. Measurement of flow volume flow rates. J Magn Reson Imaging. 1997;7:815–828.
with NMR imaging using a gradient pulse and phase difference 35. Polzin JA, Korosec FR, Wedding KL, et al. Effects of through-plane
technique. J Comput Assist Tomogr. 1984;8:588–593. myocardial motion on phase-difference and complex-difference
10. Feinberg DA, Crooks LE, Hoenninger J, et al. Pulsatile blood velocity in measurements of absolute coronary artery flow. J Magn Reson Imaging.
human arteries displayed by magnetic resonance imaging. Radiology. 1996;6:113–123.
1984;153:177–180. 36. Firmin DN, Nayler GL, Kilner PJ, Longmore DB. The application of
11. Singer JR, Crooks LE. Nuclear magnetic resonance blood flow phase shifts in NMR for flow measurement. Magn Reson Med.
measurements in the human brain. Science. 1983;221:654–656. 1990;14:230–241.
12. Shimizu K, Matsuda T, Sakurai T, et al. Visualisation of moving fluid: 37. Kilner PJ, Gatehouse PD, Firmin DN. Flow measurement by magnetic
quantitative analysis of blood flow velocity using MR imaging. resonance: a unique asset worth optimising. J Cardiovasc Magn Reson.
Radiology. 1986;159:195–199. 2007;9:723–728.
13. Axel L, Shimakawa A, MacFall J. A time-of-flight method of measuring 38. Bernstein MA, Zhou XJ, Polzin JA, et al. Concomitant gradient terms in
flow velocity by magnetic resonance imaging. Magn Reson Imaging. phase contrast MR: analysis and correction. Magn Reson Med. 1998;39:
1986;4:199–205. 300–308.
14. Edelman RR, Mattle HP, Kleefield J, Silver MS. Quantification of blood 39. Chernobelsky A, Shubayev O, Comeau CR, Wolff SD. Baseline
flow with dynamic MR imaging and presaturation bolus tracking. correction of phase contrast images improves quantification of blood
Radiology. 1989;171:551–556. flow in the great vessels. J Cardiovasc Magn Reson. 2007;9:681–685.
15. Izen SH, Haacke EM. Measuring non-constant flow in magnetic 40. Kilner PJ, Firmin DN, Mohiaddin RH, Underwood SR, Rees RSO,
resonance imaging. IEEE Trans Med Imaging. 1990;9:450–460. Longmore DB. Valve and great vessel stenosis: assessment with MR jet
16. Hahn EL. Detection of sea-water motion by nuclear precession. Geophys velocity mapping. Radiology. 1991;178:229–235.
Res. 1960;65:776–777. 41. Meier D, Maier S, Boesiger P. Quantitative flow measurements on
17. Moran PR. A flow zeugmatographic interlace for NMR imaging in phantoms and on blood vessels with MR. Magn Reson Med. 1988;8:
humans. Magn Reson Imaging. 1982;1:197–203. 25–34.
18. Young IR, Bydder GM, Payne JA. Flow measurement by the development 42. Pettigrew RI, Dannels W, Galloway JR, et al. Quantitative phase-flow
of phase differences during slice formation in MR imaging. Magn Reson imaging in dogs by using standard sequences: comparison with in
Med. 1986;3:175–179. vivo-flow meter measurements. AJR Am J Roentgenol. 1987;148:
19. Ridgway JP, Smith MA. A technique for velocity imaging using magnetic 411–414.
resonance imaging. Br J Radiol. 1986;59:603–607. 43. Kilner PJ, Manzara CC, Mohiaddin RH, et al. Magnetic resonance jet
20. Nayler GL, Firmin DN, Longmore DB. Blood flow imaging by cine velocity mapping in mitral and aortic valve stenosis. Circulation.
magnetic resonance. J Comput Assist Tomogr. 1986;10:715–722. 1993;87:1239–1248.
21. Haacke EM, Lenz GW. Improving MR image quality in the presence of 44. Maier SE, Meier D, Boesiger P, Moser UT, Vieli A. Human abdominal
motion by using rephasing gradients. AJR Am J Roentgenol. aorta: comparative measurements of blood flow with MR imaging and
1987;148:1251–1258. multigated Doppler US. Radiology. 1989;171:487–492.
22. Firmin DN, Nayler GL, Klipstein RH, et al. In vivo validation of MR 45. Van Rossum A, Sprenger KH, Peels FC, et al. In vivo validation of
velocity imaging. J Comput Assist Tomogr. 1987;11:751–756. quantitative flow imaging in arteries and veins using magnetic resonance
23. Mohiaddin RH, Pennell DJ. MR Blood flow measurement: clinical phase shift techniques. Eur Heart J. 1991;12:117–126.
application in the heart and circulation. Cardiol Clin. 1998;16: 46. Bogren HG, Klipstein RH, Firmin DN, et al. Quantitation of antegrade
161–187. and retrograde blood flow in the human aorta by magnetic resonance.
24. Redpath TW, Norris DG, Jones RA, Hutchinson MS. A new method of Am Heart J. 1989;117:1214–1222.
NMR flow imaging. Phys Med Biol. 1984;29:891–895. 47. Bogren HG, Klipstein RH, Mohiaddin RH, et al. Pulmonary artery
25. Feinberg DA, Crooks LE, Sheldon P, et al. Magnetic resonance imaging distensibility and blood flow patterns: a magnetic resonance study of
and velocity vector components of fluid flow. Magn Reson Med. 1985;2: normal subjects and of patients with pulmonary arterial hypertension.
555–566. Am Heart J. 1989;118:990–999.
26. Hennig J, Mueri M, Brunner P, Friedburg H. Quantitative flow 48. Sakuma H, Kawada N, Kubo H, et al. Effect of breath holding on blood
measurement with the fast Fourier flow technique. Radiology. 1988; flow measurement using fast velocity encoded cine MRI. Magn Reson
166:237–240. Med. 2001;45:346–348.
27. Gatehouse PD, Link K, Bebbington MWP, et al. Pulse-wave and stenosis 49. Hofman MB, van Rossum AC, Sprenger M, Westerhof N. Assessment of
studies by cylinder excitation with Fourier velocity encoding [Abstract]. flow in the right human coronary artery by magnetic resonance phase
74.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

contrast velocity measurement: effects of cardiac and respiratory kt BLAST: head to head comparison and validation at 1.5 T and 3 T. J
motion. Magn Reson Med. 1996;35:521–531. Cardiovasc Magn Reson. 2011;13:55.
50. Firmin DN, Klipstein RH, Hounsfield GL, et al. Echo-planar high- 71. Valverde I, Nordmeyer S, Uribe S, et al. Systemic-to-pulmonary
resolution flow velocity mapping. Magn Reson Med. 1989;12:316–327. collateral flow in patients with palliated univentricular heart physiology:
51. Gatehouse PD, Firmin DN, Collins S, Longmore DB. Real time blood measurement using cardiovascular magnetic resonance 4D velocity
flow imaging by spiral scan phase velocity mapping. Magn Reson Med. acquisition. J Cardiovasc Magn Reson. 2012;14:25.
1994;31:504. 72. Nordmeyer S, Riesenkampff E, Messroghli D, et al. Four-dimensional
52. Mueller E, Laub G, Grauman R, Loeffler W. RACE—real time velocity-encoded magnetic resonance imaging improves blood flow
acquisition and evaluation of pulsatile blood flow on a whole body MRI quantification in patients with complex accelerated flow. J Magn Reson
unit [Abstract]. In: Proceedings of the Seventh Annual Meeting of the Imaging. 2013;37:208–216.
International Society of Magnetic Resonance in Medicine. 1988;729. 73. Hsiao A, Tariq U, Alley MT, Lustig M, Vasanawala SS. Inlet and outlet
53. Kose K. One shot velocity mapping using multiple spin-echo EPI and its valve flow and regurgitant volume may be directly and reliably
application to turbulent flow. J Magn Reson. 1991;92:631–635. quantified with accelerated, volumetric phase-contrast MRI. J Magn
54. Yang GZ, Gatehouse PD, Mohiaddin RH, et al. Zonal echo-planar flow Reson Imaging. 2014;41:376–385.
imaging with respiratory monitoring [Abstract]. In: Proceedings of the 74. Giese D, Wong J, Greil GF, Buehrer M, Schaeffter T, Kozerke S. Towards
Fifth Annual Meeting of the International Society of Magnetic Resonance highly accelerated Cartesian time-resolved 3D flow cardiovascular
in Medicine. 1997;1885. magnetic resonance in the clinical setting. J Cardiovasc Magn Reson.
55. Gatehouse PD, Link K, Bebbington MWP, et al. Pulse-wave and stenosis 2014;16:42.
studies by cylinder excitation with Fourier velocity encoding [Abstract]. 75. Gatehouse PD, Rolf MP, Graves MJ, et al. Flow measurement by
In: Proceedings of the Third Annual Meeting of the International Society of cardiovascular magnetic resonance: a multi-centre multi-vendor study
Magnetic Resonance and the Twelfth Annual Meeting of the European of background phase offset errors that can compromise the accuracy of
Society for Magnetic Resonance in Medicine and Biology. 1995;318. derived regurgitant or shunt flow measurements. J Cardiovasc Magn
56. Napel S, Lee DH, Frayne R, Rutt BK. Visualizing three-dimensional flow Reson. 2010;12:5.
with simulated streamlines and three-dimensional phase-contrast MR 76. Rolf MP, Hofman MB, Gatehouse PD, et al. Sequence optimization to
imaging. J Magn Reson Imaging. 1992;2:143–153. reduce velocity offsets in cardiovascular magnetic resonance volume
57. Oshinski JN, Ku DN, Mukundan S Jr, et al. Determination of wall shear flow quantification—a multi-vendor study. J Cardiovasc Magn Reson.
stress in the aorta with the use of MR phase velocity mapping. J Magn 2011;13:18.
Reson Imaging. 1995;5:640–647. 77. Wentland AL, Grist TM, Wieben O. Repeatability and internal
58. Oyre S, Ringgaard S, Kozerke S, et al. Accurate noninvasive quantitation consistency of abdominal 2D and 4D phase contrast MR flow
of blood flow, cross-sectional lumen vessel area and wall shear stress by measurements. Acad Radiol. 2013;20:699–704.
three-dimensional paraboloid modeling of magnetic resonance imaging 78. Hanneman K, Sivagnanam M, Nguyen ET, et al. Magnetic resonance
velocity data. J Am Coll Cardiol. 1998;32:128–134. assessment of pulmonary (Qp) to systemic (Qs) flows using 4D
59. Frayne R, Rutt BK. Measurement of fluid-shear rate by Fourier-encoded phase-contrast imaging: pilot study comparison with standard
velocity imaging. Magn Reson Med. 1995;34:378–387. through-plane 2D phase-contrast imaging. Acad Radiol.
60. Urchuk SN, Fremes SE, Plewes DB. In vivo validation of MR pulse 2014;21:1002–1008.
pressure measurement in an aortic flow model: preliminary results. 79. Westenberg JJ, Roes SD, Ajmone Marsan N, et al. Mitral valve and
Magn Reson Med. 1997;38:215–223. tricuspid valve blood flow: accurate quantification with 3D velocity-
61. Yang GZ, Kilner PJ, Wood NB, Underwood SR, Firmin DN. encoded MR imaging with retrospective valve tracking. Radiology.
Computation of flow pressure fields from magnetic resonance velocity 2008;249:792–800.
mapping. Magn Reson Med. 1996;36:520–526. 80. Roes SD, Hammer S, van der Geest RJ, et al. Flow assessment through
62. Firmin DN, Gatehouse PD, Konrad JP, Yang GZ, Kilner PJ, Longmore four heart valves simultaneously using 3-dimensional 3-directional
DB. Rapid 7-dimensional imaging of pulsatile flow. Comput Cardiol velocity-encoded magnetic resonance imaging with retrospective valve
IEEE Comput Soc. 1993;14:353–356. tracking in healthy volunteers and patients with valvular regurgitation.
63. Wigstrom L, Sjoqvist L, Wranne B. Temporally resolved 3D phase- Invest Radiol. 2009;44:669–675.
contrast imaging. Magn Reson Med. 1996;36:800–803. 81. Nett EJ, Johnson KM, Frydrychowicz A, et al. Four-dimensional phase
64. Dyvorne H, Knight-Greenfield A, Jajamovich G, et al. Abdominal 4D contrast MRI with accelerated dual velocity encoding. J Magn Reson
flow MR imaging in a breath hold: combination of spiral sampling and Imaging. 2012;35:1462–1471.
dynamic compressed sensing for highly accelerated acquisition. 82. Roldan-Alzate A, Frydrychowicz A, Niespodzany E, et al. In vivo
Radiology. 2015;275:245–254. validation of 4D flow MRI for assessing the hemodynamics of portal
65. Santelli C, Loecher M, Busch J, Wieben O, Schaeffter T, Kozerke S. hypertension. J Magn Reson Imaging. 2013;37:1100–1108.
Accelerating 4D flow MRI by exploiting vector field divergence 83. Frydrychowicz A, Wieben O, Niespodzany E, Reeder SB, Johnson KM,
regularization. Magn Reson Med. 2016;75:115–125. Francois CJ. Quantification of thoracic blood flow using volumetric
66. Brix L, Ringgaard S, Rasmusson A, Sorensen TS, Kim WY. Three magnetic resonance imaging with radial velocity encoding: in vivo
dimensional three component whole heart cardiovascular magnetic validation. Invest Radiol. 2013;48:819–825.
resonance velocity mapping: comparison of flow measurements from 84. Barker AJ, Roldán-Alzate A, Entezari P, et al. Four-dimensional flow
3D and 2D acquisitions. J Cardiovasc Magn Reson. 2009;11:3. assessment of pulmonary artery flow and wall shear stress in adult
67. Uribe S, Beerbaum P, Sorensen TS, Rasmusson A, Razavi R, Schaeffter T. pulmonary arterial hypertension: results from two institutions. Magn
Four-dimensional (4D) flow of the whole heart and great vessels using Reson Med. 2014;73:1904–1913.
real-time respiratory self-gating. Magn Reson Med. 2009;62:984–992. 85. Tariq U, Hsiao A, Alley M, Zhang T, Lustig M, Vasanawala SS. Venous
68. Hope MD, Meadows AK, Hope TA, et al. Clinical evaluation of aortic and arterial flow quantification are equally accurate and precise with
coarctation with 4D flow MR imaging. J Magn Reson Imaging. parallel imaging compressed sensing 4D phase contrast MRI. J Magn
2010;31:711–718. Reson Imaging. 2013;37:1419–1426.
69. Nordmeyer S, Riesenkampff E, Crelier G, et al. Flow-sensitive four- 86. Dyverfeldt P, Bissell M, Barker AJ, et al. 4D flow cardiovascular magnetic
dimensional cine magnetic resonance imaging for offline blood flow resonance consensus statement. J Cardiovasc Magn Reson. 2015;17:72.
quantification in multiple vessels: a validation study. J Magn Reson 87. Buonocore MH. Visualizing blood flow patterns using streamlines,
Imaging. 2010;32:677–683. arrows, and particle paths. Magn Reson Med. 1998;40:210–226.
70. Carlsson M, Toger J, Kanski M, et al. Quantification and visualization of 88. Markl M, Draney MT, Hope MD, et al. Time-resolved 3-dimensional
cardiovascular 4D velocity mapping accelerated with parallel imaging or velocity mapping in the thoracic aorta: visualization of 3-directional
CHAPTER 6  Blood Flow Velocity Assessment 74.e3

blood flow patterns in healthy volunteers and patients. J Comput Assist 108. Bogren HG, Buonocore MH, Valente RJ. Four-dimensional magnetic
Tomogr. 2004;28:459–468. resonance velocity mapping of blood flow patterns in the aorta in
89. Bogren HG, Mohiaddin RH, Kilner PJ, Jimenez-Borreguero LJ, Yang GZ, patients with atherosclerotic coronary artery disease compared to
Firmin DN. Blood flow patterns in the thoracic aorta studied with age-matched normal subjects. J Magn Reson Imaging. 2004;19:417–427.
three-directional MR velocity mapping: the effects of age and coronary 109. Kvitting JP, Ebbers T, Wigstrom L, Engvall J, Olin CL, Bolger AF. Flow
artery disease. J Magn Reson Imaging. 1997;7:784–793. patterns in the aortic root and the aorta studied with time-resolved,
90. Markl M, Kilner PJ, Ebbers T. Comprehensive 4D velocity mapping of 3-dimensional, phase-contrast magnetic resonance imaging:
the heart and great vessels by cardiovascular magnetic resonance. J implications for aortic valve-sparing surgery. J Thorac Cardiovasc Surg.
Cardiovasc Magn Reson. 2011;13:7. 2004;127:1602–1607.
91. Wigstrom L, Ebbers T, Fyrenius A, et al. Particle trace visualization of 110. Markl M, Draney MT, Miller DC, et al. Time resolved three-dimensional
intracardiac flow using time-resolved 3D phase contrast MRI. Magn magnetic resonance velocity mapping of aortic flow in healthy
Reson Med. 1999;41:793–799. volunteers and patients after valve-sparing aortic root replacement. J
92. Kilner PJ, Yang GZ, Wilkes AJ, Mohiaddin RH, Firmin DN, Yacoub MH. Thorac Cardiovasc Surg. 2005;130:456–463.
Asymmetric redirection of flow through the heart. Nature. 111. Hope MD, Hope TA, Meadows AK, et al. Bicuspid aortic valve:
2000;404:759–761. four-dimensional MR evaluation of ascending aortic systolic flow
93. Bolger AF, Heiberg E, Karlsson M, et al. Transit of blood flow through patterns. Radiology. 2010;255:53–61.
the human left ventricle mapped by cardiovascular magnetic resonance. 112. Frydrychowicz A, Markl M, Hirtler D, et al. Aortic hemodynamics in
J Cardiovasc Magn Reson. 2007;9:741–747. patients with and without repair of aortic coarctation: in vivo analysis
94. Eriksson J, Carlhall CJ, Dyverfeldt P, Engvall J, Bolger AF, Ebbers T. by 4D flow-sensitive magnetic resonance imaging. Invest Radiol.
Semiautomatic quantification of 4D left ventricular blood flow. J 2011;46:317–325.
Cardiovasc Magn Reson. 2010;12:9. 113. Burk J, Blanke P, Stankovic Z, et al. Evaluation of 3D blood flow
95. Toger J, Carlsson M, Soderlind G, Arheden H, Heiberg E. Volume patterns and wall shear stress in the normal and dilated thoracic aorta
Tracking: A new method for quantitative assessment and visualization of using flow-sensitive 4D CMR. J Cardiovasc Magn Reson. 2012;14:84.
intracardiac blood flow from three-dimensional, time-resolved, 114. Geiger J, Arnold R, Herzer L, et al. Aortic wall shear stress in Marfan
three-component magnetic resonance velocity mapping. BMC Med syndrome. Magn Reson Med. 2012;70:1137–1144.
Imaging. 2011;11:10. 115. von Knobelsdorff-Brenkenhoff F, Trauzeddel RF, Barker AJ, Gruettner H,
96. Eriksson J, Bolger AF, Ebbers T, Carlhäll C-J. Four-dimensional blood Markl M, Schulz-Menger J. Blood flow characteristics in the ascending
flow specific markers of LV dysfunction in dilated cardiomyopathy. Eur aorta after aortic valve replacement-a pilot study using 4D-flow MRI.
Heart J Cardiovasc Imaging. 2013;14:417–424. Int J Cardiol. 2014;170:426–433.
97. Zajac J, Eriksson J, Dyverfeldt P, Bolger AF, Ebbers T, Carlhäll CJ. 116. Dyverfeldt P, Hope MD, Tseng EE, Saloner D. Magnetic resonance
Turbulent kinetic energy in normal and myopathic left ventricles. J measurement of turbulent kinetic energy for the estimation of
Magn Reson Imaging. 2014;41:1021–1029. irreversible pressure loss in aortic stenosis. JACC Cardiovasc Imaging.
98. Calkoen EE, Roest AA, Kroft LJ, et al. Characterization and improved 2013;6:64–71.
quantification of left ventricular inflow using streamline visualization 117. Frydrychowicz A, Berger A, Munoz Del Rio A, et al. Interdependencies
with 4DFlow MRI in healthy controls and patients after atrioventricular of aortic arch secondary flow patterns, geometry, and age analysed by
septal defect correction. J Magn Reson Imaging. 2014;41:1512–1520. 4-dimensional phase contrast magnetic resonance imaging at 3 Tesla.
99. Elbaz MS, Calkoen EE, Westenberg JJ, Lelieveldt BP, Roest AA, van der Eur Radiol. 2012;22:1122–1130.
Geest RJ. Vortex flow during early and late left ventricular filling in 118. Geiger J, Markl M, Herzer L, et al. Aortic flow patterns in patients with
normal subjects: quantitative characterization using retrospectively- Marfan syndrome assessed by flow-sensitive four-dimensional MRI. J
gated 4D flow cardiovascular magnetic resonance and three-dimensional Magn Reson Imaging. 2012;35:594–600.
vortex core analysis. J Cardiovasc Magn Reson. 2014;16:78. 119. Barker AJ, Markl M, Bürk J, et al. Bicuspid aortic valve is associated with
100. Eriksson J, Bolger AF, Carlhäll CJ, Ebbers T. Spatial heterogeneity of altered wall shear stress in the ascending aorta: clinical perspective. Circ
four-dimensional relative pressure fields in the human left ventricle. Cardiovasc Imaging. 2012;5:457–466.
Magn Reson Med. 2015;74:1716–1725. 120. Reiter G, Reiter U, Kovacs G, et al. Magnetic resonance-derived
101. Fyrenius A, Wigstrom L, Ebbers T, Karlsson M, Engvall J, Bolger AF. Three 3-dimensional blood flow patterns in the main pulmonary artery as
dimensional flow in the human left atrium. Heart. 2001;86:448–455. a marker of pulmonary hypertension and a measure of elevated
102. Fluckiger JU, Goldberger JJ, Lee DC, et al. Left atrial flow velocity mean pulmonary arterial pressure. Circ Cardiovasc Imaging. 2008;1:
distribution and flow coherence using four-dimensional FLOW MRI: a 23–30.
pilot study investigating the impact of age and pre- and postintervention 121. Geiger J, Markl M, Jung B, et al. 4D-MR flow analysis in patients after
atrial fibrillation on atrial hemodynamics. J Magn Reson Imaging. repair for tetralogy of Fallot. Eur Radiol. 2011;21:1651–1657.
2013;38:580–587. 122. Francois CJ, Srinivasan S, Schiebler ML, et al. 4D cardiovascular
103. Arvidsson PM, Töger J, Heiberg E, Carlsson M, Arheden H. magnetic resonance velocity mapping of alterations of right heart flow
Quantification of left and right atrial kinetic energy using four- patterns and main pulmonary artery hemodynamics in tetralogy of
dimensional intracardiac magnetic resonance imaging flow Fallot. J Cardiovasc Magn Reson. 2012;14:16.
measurements. J Appl Physiol. 2013;114:1472–1481. 123. Bachler P, Pinochet N, Sotelo J, et al. Assessment of normal flow
104. Dyverfeldt P, Kvitting JPE, Carlhäll CJ, et al. Hemodynamic aspects of patterns in the pulmonary circulation by using 4D magnetic resonance
mitral regurgitation assessed by generalized phase-contrast MRI. J Magn velocity mapping. Magn Reson Imaging. 2013;31:178–188.
Reson Imaging. 2011;33:582–588. 124. Canstein C, Cachot P, Faust A, et al. 3D MR flow analysis in realistic
105. Kvitting JP, Dyverfeldt P, Sigfridsson A, et al. In vitro assessment of flow rapid-prototyping model systems of the thoracic aorta: comparison with
patterns and turbulence intensity in prosthetic heart valves using in vivo data and computational fluid dynamics in identical vessel
generalized phase-contrast MRI. J Magn Reson Imaging. geometries. Magn Reson Med. 2008;59:535–546.
2010;31:1075–1080. 125. Harloff A, Albrecht F, Spreer J, et al. 3D blood flow characteristics in the
106. Hope MD, Hope TA, Crook SE, et al. 4D flow CMR in assessment of carotid artery bifurcation assessed by flow sensitive 4D MRI at 3T. Magn
valve-related ascending aortic disease. JACC Cardiovasc Imaging. Reson Med. 2009;61:65–74.
2011;4:781–787. 126. Markl M, Wegent F, Zech T, et al. In vivo wall shear stress distribution in
107. Bogren HG, Buonocore MH. 4D magnetic resonance velocity mapping the carotid artery: effect of bifurcation geometry, internal carotid artery
of blood flow patterns in the aorta in young vs. elderly normal subjects. stenosis, and recanalization therapy. Circ Cardiovasc Imaging.
J Magn Reson Imaging. 1999;10:861–869. 2010;3:647–655.
74.e4 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

127. Meckel S, Leitner L, Bonati LH, et al. Intracranial artery velocity 136. Wåhlin A, Ambarki K, Birgander R, et al. Measuring pulsatile flow in
measurement using 4D PC MRI at 3 T: comparison with transcranial cerebral arteries using 4D phase-contrast MR imaging. AJNR Am J
ultrasound techniques and 2D PC MRI. Neuroradiology. 2013;55: Neuroradiol. 2013;34:1740–1745.
389–398. 137. Frydrychowicz A, Landgraf BR, Niespodzany E, et al. Four-dimensional
128. Tao Y, Rilling G, Davies M, Marshall I. Carotid blood flow measurement velocity mapping of the hepatic and splanchnic vasculature with radial
accelerated by compressed sensing: validation in healthy volunteers. sampling at 3 tesla: a feasibility study in portal hypertension. J Magn
Magn Reson Imaging. 2013;31:1485–1491. Reson Imaging. 2011;34:577–584.
129. Bammer R, Hope TA, Aksoy M, Alley MT. Time-resolved 3D 138. Stankovic Z, Csatari Z, Deibert P, et al. Normal and altered three-
quantitative flow MRI of the major intracranial vessels: initial dimensional portal venous hemodynamics in patients with liver
experience and comparative evaluation at 1.5T and 3.0T in combination cirrhosis. Radiology. 2012;262:862–873.
with parallel imaging. Magn Reson Med. 2007;57:127–140. 139. Stankovic Z, Csatari Z, Deibert P, et al. A feasibility study to evaluate
130. Wetzel S, Meckel S, Frydrychowicz A, et al. In vivo assessment and splanchnic arterial and venous hemodynamics by flow-sensitive 4D MRI
visualization of intracranial arterial hemodynamics with flow-sensitized compared with Doppler ultrasound in patients with cirrhosis and
4D MR imaging at 3T. AJNR Am J Neuroradiol. 2007;28:433–438. controls. Eur J Gastroenterol Hepatol. 2013;25:669–675.
131. Boussel L, Rayz V, Martin A, et al. Phase-contrast magnetic resonance 140. Lum DP, Johnson KM, Paul RK, et al. Transstenotic pressure gradients:
imaging measurements in intracranial aneurysms in vivo of flow measurement in swine–retrospectively ECG-gated 3D phase-contrast
patterns, velocity fields, and wall shear stress: comparison with MR angiography versus endovascular pressure-sensing guidewires.
computational fluid dynamics. Magn Reson Med. 2009;61:409–417. Radiology. 2007;245:751–760.
132. Isoda H, Ohkura Y, Kosugi T, et al. In vivo hemodynamic analysis of 141. Bley TA, Johnson KM, Francois CJ, et al. Noninvasive assessment of
intracranial aneurysms obtained by magnetic resonance fluid dynamics transstenotic pressure gradients in porcine renal artery stenoses by using
(MRFD) based on time-resolved three-dimensional phase-contrast MRI. vastly undersampled phase-contrast MR angiography. Radiology.
Neuroradiology. 2010;52:921–928. 2011;261:266–273.
133. van Ooij P, Guedon A, Poelma C, et al. Complex flow patterns in a 142. Frydrychowicz A, Winterer JT, Zaitsev M, et al. Visualization of iliac
real-size intracranial aneurysm phantom: phase contrast MRI compared and proximal femoral artery hemodynamics using time-resolved
with particle image velocimetry and computational fluid dynamics. 3D phase contrast MRI at 3T. J Magn Reson Imaging. 2007;25:1085–
NMR Biomed. 2011;25:14–26. 1092.
134. Schnell S, Ansari SA, Vakil P, et al. Three dimensional hemodynamics in 143. Uribe S, Bachler P, Valverde I, et al. Hemodynamic assessment in
intracranial aneurysms: influence of size and morphology. J Magn Reson patients with one-and-a-half ventricle repair revealed by four-
Imaging. 2014;39:120–131. dimensional flow magnetic resonance imaging. Pediatr Cardiol.
135. Schrauben E, Johnson K, Huston J, et al. Reproducibility of cerebrospinal 2013;34:447–451.
venous blood flow and vessel anatomy with the use of phase contrast– 144. Geiger J, Hirtler D, Burk J, et al. Postoperative pulmonary and aortic 3D
vastly undersampled isotropic projection reconstruction and contrast- haemodynamics in patients after repair of transposition of the great
enhanced MRA. AJNR Am J Neuroradiol. 2014;35:999–1006. arteries. Eur Radiol. 2014;24:200–208.
7 
Use of Navigator Echoes in Cardiovascular
Magnetic Resonance and Factors Affecting
Their Implementation
David Firmin and Jennifer Keegan

Respiration has been shown to be an important factor influencing the


USE OF NAVIGATOR INFORMATION
quality of cardiovascular magnetic resonance (CMR) images. In addi-
tion to cardiac motion, which can be addressed reasonably well by There are two distinct ways of using navigator echoes to reduce the
electrocardiographic (ECG) triggering, respiratory motion moves the problems of respiratory motion in CMR, which are multiple breath-
position and distorts the shape of the heart by several millimeters between holding with feedback and free-breathing methods. The first of these
inspiration and expiration. In 1991, Atkinson and Edelman1 showed uses the navigator information to provide visual feedback on the dia-
the detrimental effects of breathing on the quality of cardiac studies phragm position to subjects to allow them to hold their breath at the
by showing improved detail (fast low angle shot) in breath-hold seg- same point repeatedly.5 The second uses the navigator echo measurement
mented fast gradient echo images compared with conventional non– as an input to some form of respiratory gating algorithm while the
breath-hold images. Although breath-holding produces images that are patient breathes normally. Fig. 7.1 shows actual respiratory trace data
free of respiratory motion artifact, it is not without problems. The in a subject when performing multiple breath-holds and when free
breath-hold position may vary from one breath-hold scan to the next, breathing. In both cases, a navigator acceptance window, typically 5-mm
giving rise to misregistration effects, and it may also vary during the wide, is defined, and all data acquired when the navigator is outside of
breath-hold period itself,2 resulting in image blurring and artifacts. In this window are ignored. The resulting image therefore consists of data
addition, the scan parameters are limited by the need to perform imaging acquired over a narrow range of respiratory positions. The respiratory
within the duration of a comfortable breath-hold period, and for a or scan efficiency is defined as the percentage of ECG triggers that fall
number of patients, this period may be very short. within the navigator acceptance window and is a measure of the data
An alternative to breath-holding is to monitor respiratory motion rejection rate, which in turn determines the overall scan duration. As
throughout the data acquisition period and to correct the data for the navigator acceptance window is reduced, the rejection rate increases
that motion, either in real time or through postprocessing, with the and the scan efficiency decreases. Fig. 7.2 shows the residual diaphragm
efficacy of both techniques being strongly dependent on the accuracy displacements that occurred during data acquisitions performed during
of the method of motion assessment. During normal tidal respiration, conventional breath-holding, breath-holding with navigator feedback,
the superior-inferior (SI) motion of the diaphragm is approximately and navigator free-breathing in normal subjects.6 Both navigator tech-
four to five times the anterior-posterior motion of the chest wall,3 niques result in images acquired over a reduced range of diaphragm
and consequently, diaphragm motion is the most sensitive measure displacement compared with those acquired using repeated conventional
of respiratory motion. In 1989, Ehman and Felmlee4 were the first breath-holding. In addition, they allow a longer overall scan time. This
to introduce navigator echoes for measuring the displacement of a allows for averaging of data to improve the signal-to-noise ratio, increas-
moving structure and to demonstrate their use in determining dia- ing the k-space coverage for improved spatial resolution, and increasing
phragm motion during respiration. The navigator echo is the signal the temporal resolution by reducing the number of individual image
from a column of material oriented perpendicular to the direction of views acquired per cardiac cycle.
the motion to be monitored. On Fourier transformation, this signal
results in a well-defined edge of the moving structure. The navigator Multiple Breath-Hold Methods
echoes may be interleaved with the imaging sequence and, consequently, Wang and colleagues7 were the first to show the use of a respiratory
enable the motion to be determined throughout the data acquisition feedback monitor to reduce misregistration artifacts in consecutive
period. breath-hold segmented fast gradient echo coronary artery images and
In CMR, there have been a number of developments in the use of to show improved image quality from averaging scans acquired over
navigator measurement to reduce the problems of respiratory motion. multiple breath-holds. When used in informed healthy volunteers, this
This chapter discusses these developments, considers the various choices technique has been shown to produce good results with reasonable
that have been studied in the implementation of navigators, and dis- scan efficiency.8 However, a period of training is required, and the process
cusses their importance. There are many variables in the application can be problematic, particularly with patients who have difficulty holding
and use of navigator echoes, and although there have been some their breath because of a combination of illness and anxiety.6 Although
attempts to study these, it is unlikely that we are close to optimizing it might be expected that breath-holding with respiratory feedback
their application. would enable the completion of a cardiac study much more quickly

75
76 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

5 mm NE acceptance window Standard deviation


±2.4 mm ±0.7 mm ±0.7 mm
0 30
Diaphragm displacement (mm)

−20
20

−40

Diaphragm displacement (mm)


10

−60

0
A 0 Time (s) 60
Diaphragm displacement (mm)

−10
0
BH

LED
−20 −20
+ FR

−40
−30
FIG. 7.2  Mean diaphragm displacement in 17 normal subjects with
B conventional breath-holding (BH; open circles), BH with navigator feed-
FIG. 7.1  Navigator echo (NE) respiratory trace data during multiple back (closed circles), and free respiration (FR; plus marks). The navigator-
breath-holding with navigator feedback (A) and free breathing (B). In controlled studies used a 5-mm navigator acceptance window. (From
each case, the shaded region shows the position of a 5-mm navigator Taylor AM, Keegan J, Jhooti P, Gatehouse PD, Firmin DN, Pennell DJ.
acceptance window outside of which data is rejected. (From Taylor AM, Differences between normal subjects and patients with coronary artery
Keegan J, Jhooti P, Gatehouse PD, Firmin DN, Pennell DJ. Differences disease for three different MR coronary angiography respiratory sup-
between normal subjects and patients with coronary artery disease for pression techniques. J Magn Reson Imaging. 1999; 9:786–793.)
three different MR coronary angiography respiratory suppression tech-
niques. J Magn Reson Imaging. 1999;9:786–793.)

subject’s breathing pattern during the long acquisition period and was
often still unacceptably high.
than when using the free-breathing methods described later, because After the introduction of prospective control techniques, navigators
of the time required for training and the required rest periods between have most commonly been used with a simple accept-reject algorithm
breath-holds, the overall examination times are longer than anticipated. where data are acquired or not, depending on whether the navigator
In fact, in a group of patients with coronary artery disease, there was measurement is within a predefined acceptance window. Oshinski and
no significant difference between the overall examination times with coworkers12 were the first investigators to show high-quality coronary
the two techniques,6 although the same study showed that, in a group artery images with such an approach. The problem with this method,
of normal healthy subjects, multiple breath-holding resulted in a time however, is that for reasonably high scan quality, a narrow acceptance
reduction of 20%. window of 5 mm or less is required, and this generally results in relatively
poor scan efficiency. In addition, as noted earlier, many subjects and
Free-Breathing Methods patients undergo a drift in diaphragm position over time,9 such that
Free-breathing methods require very little cooperation from the patient. the predefined acceptance window becomes less and less suitable as the
The main disadvantage is the potential for respiratory drift, which can study progresses. The diminishing variance algorithm overcomes this
cause considerably reduced scan efficiency.9 Recently, therefore, most problem because it does not use a predefined acceptance window.13 With
effort has gone toward improving scan efficiency with this approach. this method, one complete scan is acquired and the navigator positions
Much of the early work used retrospective respiratory gating.10 With are saved for each line of data. At the end of the initial scan, the most
this method, data acquisition was oversampled, typically by a factor of frequent diaphragm position during that scan is determined, and a
five, and then sorted retrospectively so that the final image was con- process of reacquiring lines of data that were acquired with diaphragm
structed from data acquired over the narrowest possible range of respi- positions furthest offset from this position begins. As time progresses,
ratory positions. In 1995, Hofman and associates11 showed that, using the range of diaphragm positions for the data making up the final set
this approach, the image quality of three-dimensional (3D) coronary is considerably reduced. In addition to the lack of requirement of an
acquisitions was improved over those acquired with multiple averages. acceptance window, this method has the advantage that an image can
However, scan efficiency was poor (20% for an oversampling ratio of be reconstructed at any time after the initial dataset is complete.
five), and although the final image was constructed from the narrowest Another alternative to the simple accept-reject algorithm that can
respiratory window possible, the range was highly dependent on the improve both image quality and scan efficiency is to use a k-space
CHAPTER 7  Use of Navigator Echoes in Cardiovascular Magnetic Resonance 77

ordering that depends on diaphragm position. Two similar approaches general respiratory motion16 and more recently applied to imaging of the
have been suggested, based on the finding that the center of k-space coronary arteries, used a tailored acceptance window through k-space
appears to be more sensitive to motion than the edges.14 Jhooti and as opposed to phase-encode ordering to obtain a very similar result.17
colleagues developed a phase-encode ordered method that used a dual Both of these phase-ordering or windowing techniques use a predefined
acceptance window of 5 mm for the center of k-space and 10 mm for the navigator acceptance window, and scan efficiency is reduced when the
outer regions.15 This approach allowed much greater scan efficiency than respiratory pattern changes during study acquisition. This has been more
other methods, while retaining scan quality (Fig. 7.3 and Table 7.1). An recently addressed by Jhooti and colleagues, who developed a technique
alternative method, initially developed by Sinkus and Bornert to address combining the benefits of phase ordering with an automatic window

A C
Phase reordered (scan eff. = 73%) DVA (scan eff. = 73%)

B D
ARA (scan eff. = 49%) RRG (scan eff. = 20%)
FIG. 7.3  A single slice from a three-dimensional dataset showing a long section of the right coronary artery.
The phase-reordered images (A) are of comparable quality to those acquired with the accept-reject algorithm
(ARA) (B) and better than those acquired with both the diminishing variance algorithm (DVA), (C) and retro-
spective respiratory gating (RRG) (D). Scan efficiency is also significantly higher for phase ordering than for
both the accept-reject algorithm and retrospective respiratory gating techniques. (From Jhooti P, Keegan J,
Gatehouse PD, et al. 3D coronary artery imaging with phase reordering for improved scan efficiency. Magn
Reson Med. 1999;41:555–562.)

TABLE 7.1  Image Quality Scores and Scan Efficienciesa for Three-Dimensional Magnetic
Resonance Angiographyb
Phase Ordered ARA DVAa RRG
Image quality mean score 4.4 4.7 6.6 6.8
Scan efficiency 72 (±11.6) 48 (±11.5) 72 (±11.6) 20
a
Scan time for the DVA technique is set to that of the phase-ordered technique.
Mean image quality scores (1 = excellent, 10 = very poor) and scan efficiencies (± standard deviation) for data acquired using phase ordering, an
b

ARA, the DVA, and RRG in 15 subjects.


ARA, Accept-reject algorithm; DVA, diminishing variance algorithm; RRG, retrospective respiratory gating.
Modified from Jhooti P, Keegan J, Gatehouse PD, et al. 3D coronary artery imaging with phase reordering for improved scan efficiency. Magn
Reson Med. 1999;41:555.
78 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

selection that enables the highly efficient acquisition of high-quality 0


coronary artery images without the need for a predefined acceptance
window.18 3D motion-adapted gating19 is a similar technique that yields

Downward coronary displacement (mm)


y = –0.08 – (0.45 x)
images comparable with standard prospective navigator gating, with
r = 0.99
significantly improved scan efficiency.20 A further development of these 4
ideas, termed continuously adaptive windowing strategy (CLAWS), was
described by Jhooti et al. in 2010.21 This novel and dynamic acquisition
strategy ensured all potential navigator acceptance windows are possible
and acquires an image with the highest possible efficiency regardless 8
of variations in the respiratory pattern. Unnecessary prolongation of
scan durations due to respiratory drift or navigator acceptance window
adjustments are avoided. Because CLAWS requires no setting of the
acceptance window, nor monitoring of the navigator traces during the 12
scan, operator dependence is minimized and ease of use improved. This
method has been applied by Keegan et al.22 to improve the respiratory
efficiency of 3D late gadolinium enhancement imaging and by Jhooti
et al.,23 who combined it with biofeedback to increase the navigator
16
efficiency for imaging the thoracic aorta.
0 10 20 30
Downward diaphragm displacement (mm)
NAVIGATOR ECHO IMPLEMENTATION
FIG. 7.4  Plot of superior-inferior right coronary artery displacement
Method of Column Selection against superior-inferior diaphragm displacement for a single subject.
Two methods have been used for the generation of a navigator echo. The gradient of the linear regression line is the subject-specific correc-
With the spin echo technique, a spin echo signal is generated from tion factor. (From Taylor AM, Keegan J, Jhooti P, Firmin DN, Pennell
DJ. Calculation of a subject-specific adaptive motion-correction factor
the column of material formed by the intersection of two planes, one
for improved real-time navigator echo-gated magnetic resonance coronary
excited by a 90-degree radiofrequency (RF) pulse and the other by a
angiography. J Cardiovasc Magn Reson. 1999;1:131–138.)
180-degree RF pulse. The column cross section may be either rect-
angular or rhombic, depending on the orientation of the two planes.
This approach is very robust and produces an extremely well-defined
column. However, it cannot be repeated rapidly, and care must be taken
to ensure that the column selection planes do not impinge on the variation in the degree of cardiac motion with respiration. This was
region of interest. also observed by Danias and coworkers, who used real-time 2D echo
The alternative approach is to use a selective two-dimensional (2D) planar imaging to study the SI motion of the heart as a function of
RF pulse to excite a column of approximately circular cross section.24 navigator position.27 The accuracy of slice-following techniques will
Although this technique is much more sensitive to factors such as shim- obviously depend on the accuracy of the CF implemented. In 1997,
ming errors, which can potentially cause blurring and distortion of the Taylor and colleagues28 showed how a subject-specific factor could be
column, with a reduced flip angle, it can be repeated more rapidly and measured rapidly with end-inspiratory and end-expiratory breath-
the navigator artifact is less extensive. hold scans before the coronary imaging protocol. Fig. 7.4 shows the
Both methods are used routinely for research studies on coronary relationship between the motion of the right hemidiaphragm and the
imaging, without any reported problems. coronary ostia measured in one subject, with the slope of the graph
giving the CF. Fig. 7.5 shows two examples of subjects with very dif-
Correction Factors ferent CFs, showing how a wider acceptance window can be used,
In CMR, navigator echoes are most frequently used to measure the thus improving scan efficiency. The need for a subject-specific CF has
position of the diaphragm. However, the motion of the heart is not further been confirmed in 3D coronary angiography, where its use was
straightforward, and only the inferior border that sits on the diaphragm found to yield optimal image quality.29 In 2002, Keegan and associ-
will move to the same extent, whereas superiorly, the relative motion will ates further developed this area of work by studying the variability
be reduced. This was first studied by Wang and associates,3 who measured of CFs in the SI, anterior-posterior, and right-left directions for both
the displacement of the right coronary artery root, the origin of the breath-holding and free-breathing.30 The study concluded that subject
left anterior descending artery, and the superior and inferior margins variability in CFs, together with within-subject differences between
of the heart relative to the diaphragm in 10 healthy subjects. For the breath-holding and free-breathing, is such that slice following should be
right coronary artery origin, the mean (± standard deviation) relative performed with subject-specific factors determined from free-breathing
displacement (or correction factor [CF]) was 0.57 (± 0.26). McConnell acquisitions.
and coworkers25 first used this CF to track the position of the imaging An additional or alternative approach to the real-time slice following
slice during breath-holding and showed improved image registration described earlier is to use a postprocessing adaptive motion correction
relative to untracked acquisitions. In free-breathing studies, the CF was technique4 to correct an image retrospectively for movement occurring
first applied by Danias and colleagues,26 who showed that tracked image during data acquisition. This technique, which can be used to correct
quality was maintained as the navigator acceptance window increased a 2D acquisition for in-plane displacement or a 3D acquisition for in-
from 3 mm to 7 mm, whereas in untracked images, it decreased sig- plane and through-plane displacement, may not appear to be an attrac-
nificantly. This technique, called real-time prospective slice following, or tive option initially, but it has the advantages of allowing the CF to be
slice tracking, is now used routinely for both 2D and 3D methods of optimized for each individual patient and provides an alternative
acquisition. Of note, however, is the relatively high standard deviation approach to those centers with scanners that do not have a real-time
of the CF discussed earlier, which reflects considerable intersubject decision-making capability. This approach has been implemented with
CHAPTER 7  Use of Navigator Echoes in Cardiovascular Magnetic Resonance 79

6 mm
16 mm

A
CF 0.70 0.00 1.00
6 mm
16 mm

B
CF 0.25 0.00 1.00
FIG. 7.5  Right coronary artery origin images acquired with navigator acceptance windows of 6 mm and
16 mm in subjects with subject-specific correction factors (CF) of 0.7 (A) and 0.25 (B). For both subjects,
images were also acquired with CFs of 0 and 1. In the absence of slice following (CF = 0), image quality is
reduced as the navigator acceptance window increases from 6 mm to 16 mm. When slice following with
a subject-specific CF is used, however, image quality is maintained. (From Taylor AM, Keegan J, Jhooti P,
Firmin DN, Pennell DJ. Calculation of a subject-specific adaptive motion-correction factor for improved real-time
navigator echo-gated magnetic resonance coronary angiography. J Cardiovasc Magn Reson. 1999;1:131–138.)
80 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

both segmented gradient echo31 and interleaved spiral32 coronary artery anterior left ventricular wall navigator scans to be longer in duration,
acquisitions, with promising results. but the difference did not reach statistical significance. One of the prob-
lems with monitoring the heart itself is the complex anatomy, making
Column Positioning it more difficult to find a suitable position for the navigator column.
The degree of diaphragm motion detected by the navigator echo is More sophisticated methods of positioning the column may further
dependent on the positioning of the navigator column. The dome of improve this method of monitoring cardiac motion.
the right hemidiaphragm is higher than that of the left, and the two
move coherently with respiration, but to differing degrees.33 Motion of
the diaphragm is also greater posteriorly than anteriorly (anterior and
MULTIPLE COLUMN ORIENTATIONS
dome excursions are 56% and 79%, respectively, of posterior excur- There is a linear relationship between the SI and anterior-posterior
sions), and at the level of the dome, it is greater laterally than medially.34 motions of the heart, with the SI motion being approximately five times
The CF implemented in real-time slice following or postprocessing that of the anterior-posterior motion.3 For this reason, the real-time
adaptive motion correction, as described earlier, is strongly dependent slice-following methods first used by McConnell and colleagues25 and
on the positioning of the navigator column and further supports the by Danias and associates26 included a correction for anterior-posterior
use of a subject-specific factor, as described in the previous section. motion of the heart, assuming it to be equal to 20% of the SI motion.
McConnell and colleagues35 studied the effects of varying the navi- Unfortunately, there is not always such a strong relationship between
gator location on the image quality of coronary artery studies. Navigators the directions of motion of the heart with respiration. Sachs and col-
were positioned through the dome of the right hemidiaphragm, through leagues showed this by using three navigators to measure the SI, anterior-
the posterior portion of the left hemidiaphragm, through the anterior posterior, and right-left motions of the heart.36 Fig. 7.7 shows an example
and posterior left ventricular walls, and through the anterior left ven- from this study illustrating the scatter of SI, right-left, and anterior-
tricular wall, as shown in Fig. 7.6. The advantage of the latter navigator posterior measurements, made over a period of approximately 10 minutes.
position is that it would eliminate the need for a CF, as described in The group went on to compare the use of one, two, and three navigators
the previous section, relating the navigator-echo-measured displacement for imaging the right coronary artery and showed an improvement
to the coronary artery motion. The results are summarized in Table when multiple directions of motion were considered. This improvement
7.2 and show no significant differences in the image quality scores in image quality, however, must be offset against the main disadvantage,
obtained with varying navigator location. There was a tendency for the which is that scan efficiency is reduced, potentially introducing more

A B C D
FIG. 7.6  Navigator column locations positioned on transverse, coronal, and sagittal pilot images: (A) through
the dome of the right hemidiaphragm, (B) through the posterior left hemidiaphragm, (C) through both anterior
and posterior left ventricular walls, and (D) through the anterior left ventricular wall.

TABLE 7.2  Image Quality Scores, Registration Errors, and Total Scan Timesa

Right Diaphragm Left Diaphragm Anterior LV Wall


Parameter Navigator Navigator LV Navigator Navigator
Image quality score (0–4) 2.3 ± 0.1 2.3 ± 0.1 2.4 ± 0.1 2.2 ± 0.2
Registration error (mm):
 Craniocaudal 0.5 ± 0.1 0.4 ± 0.1 0.6 ± 0.1 0.4 ± 0.1
 Anteroposterior 0.3 ± 0.1 0.3 ± 0.1 0.3 ± 0.1 0.4 ± 0.1
Total scan timeb (s) 294 ± 28 314 ± 30 342 ± 62 427 ± 111
a
Image quality scores (0 = very poor, 4 = excellent), registration errors, and total scan times for different navigator column positions during
free-breathing magnetic resonance coronary angiography. There were no significant differences between the navigator column locations. Data
are presented as mean ± standard error of the mean.
b
Total scan time is the time from start to finish for six scans.
LV, Left ventricle.
Modified from McConnell MV, Khasgiwala VC, Savord BJ, et al. Comparison of respiratory suppression methods and navigator locations for MR
coronary angiography. Am J Roentgenol. 1997;168:1369.
CHAPTER 7  Use of Navigator Echoes in Cardiovascular Magnetic Resonance 81

80
60

R/L AP

40

A 0 SI 150 B 0 SI 150

FIG. 7.7  Anteroposterior (AP) (A) and right-left (R/L) (B) navigator echo measurements as a function of
superior-inferior (SI) navigator echo measurements in a healthy subject. (Data from Todd Sachs, Stanford
University.)

short gating delay or cine scans, post-only navigators can be used as


an alternative. This approach was recently implemented in left ventricular
function studies, where it was found that image quality in a group of
patients with heart failure was significantly improved over conventional
breath-hold scans.38
D. acq. Repeated navigators allow for improved cine or multislice imaging
and also provide some potential for estimating internavigator respira-
Pre- tory motion. The potential problem with this is that the navigator
Pre- and post- signal-to-noise ratio could be reduced and this may affect the accuracy
D. acq. D. acq. D. acq. D. acq. of navigator edge detection. In addition, as the time for navigator output
increases, the time for imaging decreases and the number of phases or
Rep. slices that can be acquired is reduced.
FIG. 7.8  Timing of the navigators for pre-, pre- and post-, and repeated Precision of Navigator Measurement
(Rep.) navigator echo-controlled data acquisition (D. acq.).
Commonly, a spatial resolution of 1 mm is used along the navigator
echo column, for example, having a field of view of 512 mm and sam-
pling 512 points on the navigator readout. However, the precision of
problems associated with long-term drift in the breathing pattern. A the measurement is dependent to a large extent on the signal-to-noise
more recent study by Jahnke and coworkers used a new cross-correlation- ratio of the navigator measurement. The most important factor affect-
based approach and showed the potential advantage of combining three ing the signal-to-noise ratio is the coil arrangement. If, for example, a
orthogonal navigators.37 single coil is used for imaging and navigator detection, it must be large
enough to cover both the imaging area of interest and the region of
Navigator Timing navigator detection. On the other hand, if phased array coils are used,
Navigator timing is one of the more important parameters; however, it is possible to position one coil specifically for navigator detection,
flexibility to alter this is often limited by the computing architecture possibly over the region of the right diaphragm. Another important
of the scanner being used (discussed later). Fig. 7.8 shows the three factor in the precision of the measurement is the quality of the edge
main alternatives: (1) prenavigators, (2) prenavigators and postnaviga- on the navigator trace. To obtain a well-defined edge of the diaphragm,
tors, and (3) navigators repeated regularly throughout the cardiac cycle. for example, have a reasonably small column cross section and position
A simple prenavigator provides the highest scan efficiency when a navi- it through the dome of the diaphragm, so that the column is perpen-
gator acceptance window is used, but may not be reliable if there is a dicular to the diaphragm edge, rather than more posteriorly, where
sudden change in breathing between the navigator measurement and motion is greatest. Finally, the diaphragm edge may be detected by edge
image data acquisition. Prenavigators and postnavigators overcome this detection, correlation, or least-squares fit algorithms. For rapid tracking
problem, but of course, they also reduce scan efficiency. In our experi- (repetition time <100 ms) or narrower columns, the signal-to-noise
ence, the use of prenavigators only produces acceptable results for free- ratio in the diaphragm trace could be too poor for simple edge detec-
breathing studies, whereas multiple breath-hold acquisitions certainly tion methods to succeed. Of the remaining two techniques, the least-
require both prenavigators and postnavigators. An important factor squares fit method has been shown to be more resistant to the effects
that depends on the computer hardware and architecture is the time of noise and to the diaphragm profile deformation that occurs during
required after the navigator acquisition before the start of the imaging respiration than the correlation method and therefore would be the
sequence. Particularly, if prenavigators only are being used, the longer technique of choice.39 However, most navigator techniques acquire only
this interval, the greater the potential for errors caused by respiratory one or two navigators per cardiac cycle, and in such cases, the signal-
motion. Also, for ECG R-wave-triggered scans, this may also have impli- to-noise ratios are usually relatively high and edge detection algorithms
cations for the minimum gating delay that can be obtained and for are generally adequate.
82 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

MORE RECENT APPROACHES are incorporated into a multislice, multiaverage dataset. This technique
has been shown in a group of six healthy volunteers where increased
Other Forms of Navigators signal-to-noise ratio has been achieved with minimal motion blurring.
As mentioned, there are problems with the conventional navigators An alternate approach to respiratory motion correction that has
described previously because they generally do not give a direct measure been applied is to acquire a rapid low-resolution image to measure the
of the respiratory-related motion of the heart, and they cannot be position of the heart either immediately before or immediately after
implemented simply and efficiently to give a measure of this motion the acquisition of the data contributing to the final high-resolution
in 3D.40 A number of ingenious alternative approaches have been image. In the described approaches the rapid low-resolution images
described. In 2003, Nguyen and colleagues41 described a method that have been single 2D,50,51 orthogonal 2D,52 and 3D.53 Another advantage
selectively excited the epicardial fat, followed by a rapid readout scheme of these image-based navigators is that more degrees of freedom can
that instantaneously gave three one-dimensional (1D) images of its be used for the motion correction, which may not be simply translational
position in the x, y, and z directions. Tested on six subjects, the method but may involve rotational and nonrigid distortions. A potential problem
showed a slight improvement over conventional navigators; however, with these approaches is that the navigator acquisition can result in
the authors noted a number of problems that would need to be resolved signal saturation that can have an impact on the final high-resolution
before it could be in routine use. In the same year, another method of acquisition. In the approach of Keegan et al.53 and Scott et al.,54,55 the
rapidly localizing heart signals for measurement of its position was navigator image was a fat-only image acquired using a spectrally selec-
suggested by Pai and Wen, who used a phase contrast angiographic tive excitation pulse. Clearly this assumes the presence of fat around
approach to selectively image the flowing blood in the heart chambers.42 the moving heart which is generally present. Another problem with
These blood signals were used to define the heart position in the SI image-based navigators, particularly if prospective navigation is required,
and anterior-posterior directions. Despite the potential advantages of is that the time to reconstruct and extract a measure of motion can be
truly tracking the heart position, this method does not appear to have relatively long. Although 2D image-based navigators have been used
been developed further or used. prospectively,50,51 for multi 2D or 3D it is likely that faster processing
will be required.
Projection-Based Self-Navigators The above image-based navigators involve the acquisition of a navi-
In 2004, Larson et al.43 were first to describe the “self-gating” concept gator image with data that is separate and not used in any way for the
in their cine imaging where motion synchronization information for high-resolution motion corrected image. One method that combines
timing the cine data acquisition was taken from a repeated 1D projec- the data and can therefore be termed an image-based self-navigator
tion reconstruction from their radial acquisition. Stehning and associates was described by Henningsson et al.56 In their approach the startup
also used radial imaging for “self-navigation.” They developed an inter- echoes of the balanced steady-state free precession acquisition are phase
leaved 3D radial acquisition modified in such a way that the first readout encoded and used to form a low-resolution navigator image. This has
was always in the SI direction.44 This readout could then be reconstructed the advantage of ensuring the navigator is temporally very close to the
every cardiac cycle to give an SI projection for motion extraction. In motion corrected data. Moghari et al.57 extended this method from 2D
this work, the authors showed improved definition compared with to 3D and showed no difference to the image quality but substantially
conventional navigators when imaging a moving phantom and similar reduced scan times.
image quality on initial in vivo coronary scans. For these self-navigator Another image-based self-navigator method using 3D radial sampling
approaches either the central line of k-space or a particular radial spoke involved acquiring several undersampled images over a number of
has to be repeated rapidly enough to be used to correct for either cardiac cycles and dividing these images into different respiratory states
respiratory or cardiac motion, or both. Generally, as in the work of or bins.58,59 The motion between bins was estimated by image registra-
Stehning et al., for respiratory motion the 1D projection was aligned tion with a 3D affine transform and the undersampled data was motion
in the head-foot direction; however, 3D self-navigation has also been corrected and recombined into a fully sampled dataset for reconstruc-
described.45 The problem with projection self-navigators of static tissue, tion. Again the results showed significant reductions in scan time, with
such as the chest wall, potentially reducing the performance of the little or no impact on image quality. Similar binning approaches had
motion following has been addressed by attempts to suppress the signal in fact been described for Cartesian sampling60,61; however, the image-
from static tissue.45,46 The clinical performance of these methods has based navigators had to be acquired separately in these methods.
been assessed by Piccini et al.47 when they used the sequence to study
the coronary arteries of patients. They concluded that the approach Motion Models
showed promise; however, technical improvements are needed to improve Because of the complexity of cardiac motion and the difficulty in extract-
image quality, especially in the more distal coronary segments. ing measures to correct for it, there has been an interest in developing
modeling methods to assist with this process. Manke and coworkers
Image-Based Navigators compared 1D translation (SI direction), 3D translation, and 3D affine
An alternative method that reduces the problem of overlying signals and transformation motion models in a group of healthy subjects.62 By
allows for multiple dimensions is the use of image-based navigators. using an elastic image registration algorithm on 3D coronary images
Hardy and colleagues48 used cross-correlation of low-resolution real-time acquired at different breath-hold positions, the superiority of the 3D
2D spiral coronary artery images to accept or reject images for averag- translational model over the 1D translational model was shown. The
ing. This adaptive averaging technique was extended to high-resolution authors also used a fast model-based image registration to extract motion
segmented acquisitions by cross-correlating subimages reconstructed information from a time series of low-resolution 3D images. This was
from individual data segments. Breathing autocorrection with spiral used in conjunction with conventional navigators to calibrate a respira-
interleaves (BACSPIN) is a similar technique49 that involves the acqui- tory motion model that allowed the prediction of affine transformation
sition of a multislice spiral dataset during breath-holding, followed by parameters, including 3D translation, rotation, scale, and shear motion
repeated acquisition of the same slices during free breathing. Each heavily from the navigator measurements, and was shown with coronary artery
undersampled free-breathing spiral interleaf is compared with the same imaging.63 McLeish and colleagues64 acquired images at a number of
interleaf acquired during breath-holding, and those that match closely breath-hold positions and studied the accuracy of both rigid and
CHAPTER 7  Use of Navigator Echoes in Cardiovascular Magnetic Resonance 83

nonrigid registration methods in registering the other breath-hold images This arrangement inevitably adds a variable and unknown delay that
with those acquired at end-expiration. They used principal component is dependent on other tasks being performed by the host operating
analysis to produce patient-specific statistical motion models and suggest system. To overcome this, either a direct and rapid link is required,
how this could be used to assist motion correction in CMR. In 2005, allowing transfer of data from the reconstruction computer to the scan
Nehrke and Bornert described their study in which they used a patient- computer, or the scan computer itself must be capable of acquiring and
specific model to control the acquisition.65 Initially, multiple low- reconstructing the navigator data, so that no data transfer is required.
resolution 3D images were preacquired during free breathing. An affine Newer scanners are generally being designed with rapid acquisition,
model of the respiratory-related cardiac motion was then extracted processing, and control in mind.
from these images, and this was steered by real-time navigators to control
the high-resolution acquisition. The method was demonstrated in both
phantoms and volunteers when a 20-mm navigator acceptance window
CONCLUSION
was used. The results in the volunteers showed slightly inferior image Navigator echo has been shown to be an important method for moni-
quality to images acquired simply with a 5-mm navigator acceptance toring respiration used for defining the position of the heart, enabling
window. However, scan efficiency was considerably higher and image improved coronary and other cardiac imaging. The limited number of
quality was also considerably better than for those acquired simply with studies and the many parameters and variables involved in their use
a 20-mm acceptance window. A more recent study showed that the suggest that an optimal method of application may not yet have been
residual coronary artery motion observed using affine navigators with developed. With future system development, there will be minimal cost,
a 10-mm acceptance window is similar to that observed with conven- in imaging time or other factors, involved in obtaining this positional
tional navigator gating with a 5-mm window and that observed using information. Therefore, it would seem worthwhile to collect and use it
a single breath-hold.66 where appropriate. The methods are not robust, probably because of
their relative lack of sophistication.
Computer Architecture One of the major advantages of this technique is that navigators
The computer architecture of modern CMR scanners can be very allow images to be acquired during free respiration, eliminating the
complex, generally incorporating three main computers. The host need for patient cooperation. They also allow longer acquisition times,
computer runs the user interface and allows connection to the image enabling higher spatial and temporal resolution and increasing the
database, the reconstruction computer is a dedicated rapid processor for potential for more sophisticated techniques, such as detailed flow
reconstruction of the CMR image data, and the scan computer allows imaging.67,68 A balance must be maintained, however, and imaging time
control and adjustment of parameters associated with the scanning should not be increased so much that increased respiratory drift cancels
sequence. The architecture of these computers can significantly affect any potential benefit.
the potential and usefulness of navigator echoes. On many systems,
for example, the navigator signal must be reconstructed and processed
on the reconstruction computer, but the measurement made must be
REFERENCES
passed through the host to control the parameters on the scan computer. A full reference list is available online at ExpertConsult.com
CHAPTER 7  Use of Navigator Echoes in Cardiovascular Magnetic Resonance 83.e1

REFERENCES acquisitions: continuously adaptive windowing strategy. Magn Reson Med.


2010;64:1015–1026.
1. Atkinson DJ, Edelman RR. Cineangiography of the heart in a single 22. Keegan J, Jhooti P, Babu-Narayan SV, Drivas P, Ernst S, Firmin DN.
breath hold with a segmented turboFLASH sequence. Radiology. Improved respiratory efficiency of 3D late gadolinium enhancement
1991;178:357–360. imaging using the continuously adaptive windowing strategy (CLAWS).
2. Holland AE, Goldfarb JW, Edelman RR. Diaphragmatic and cardiac Magn Reson Med. 2014;71:1064–1074.
motion during suspended breathing: preliminary experience and 23. Jhooti P, Haas T, Kawel N, Bremerich J, Keegan J, Scheffler K. Use of
implications for breath-holding. Radiology. 1998;209:483–489. respiratory biofeedback and CLAWS for increased navigator efficiency for
3. Wang Y, Riederer SJ, Ehman RL. Respiratory motion of the heart: imaging the thoracic aorta. Magn Reson Med. 2011;66:1666–1673.
kinematics and the implications for the spatial resolution in coronary 24. Pauly J, Nishimura D, Macovski A. A k-space analysis of small tipangle
imaging. Magn Reson Med. 1995;33:713–719. excitation. J Magn Reson. 1989;81:43–56.
4. Ehman RL, Felmlee JP. Adaptive technique for high-definition MR 25. McConnell MV, Khasigawala VC, Savord BJ, et al. Prospective adaptive
imaging of moving structures. Radiology. 1989;173:255–263. navigator correction for breath-hold MR coronary angiography. Magn
5. Liu YL, Riederer SJ, Rossman PJ, Grimm RC, Debbins JP, Ehman RL. A Reson Med. 1997;37:148–152.
monitoring, feedback, and triggering system for reproducible breath-hold 26. Danias PG, McConnell MV, Khasigawal VC, Chuang ML, Edelman RR,
MR imaging. Magn Reson Med. 1993;30:507–511. Manning WJ. Prospective navigator correction of image position for
6. Taylor AM, Keegan J, Jhooti P, Gatehouse PD, Firmin DN, Pennell DJ. coronary MR angiography. Radiology. 1997;203:733–736.
Differences between normal subjects and patients with coronary artery 27. Danias PG, Stuber M, Botnar RM, Kissinger KV, Edelman RR, Manning
disease for three different MR coronary angiography respiratory WJ. Relationship between motion of coronary arteries and diaphragm
suppression techniques. J Magn Reson Imaging. 1999;9:786–793. during free breathing: lessons from real-time MR imaging. Am J
7. Wang Y, Grimm RC, Rossman PJ, et al. Angiography in multiple Roentgenol. 1999;172:1061–1065.
breath-holds using a respiratory feedback monitor. Magn Reson Med. 28. Taylor AM, Keegan J, Jhooti P, Firmin DN, Pennell DJ. Calculation of a
1995;34:11–16. subject-specific adaptive motion-correction factor for improved real-time
8. Keegan J, Gatehouse PD, Taylor AM, Yang GZ, Jhooti P, Firmin DN. navigator echo-gated magnetic resonance coronary angiography. J
Coronary artery imaging on a mobile 0.5Tesla scanner: implementation Cardiovasc Magn Reson. 1999;1:131–138.
of real-time navigator-echo controlled segmented k-space FLASH and 29. Nagel E, Bornstedt A, Schnackenburg B, Hug J, Oswald H, Fleck E.
interleaved spiral sequences. Magn Reson Med. 1999;41:392–399. Optimisation of real-time adaptive navigator correction for 3D
9. Taylor AM, Jhooti P, Wiesmann FW, Keegan J, Firmin DN, Pennell DJ. magnetic resonance coronary angiography. Magn Reson Med. 1999;42:
MR navigator-echo monitoring of temporal changes in diaphragm 408–411.
position: implications for MR coronary angiography. J Magn Reson 30. Keegan J, Gatehouse P, Yang GZ, Firmin D. Coronary artery motion with
Imaging. 1997;7:629–636. the respiratory cycle during breath-holding and free-breathing:
10. Lenz GW, Haacke EM, White RD. Retrospective cardiac gating: a review implications for slice-followed coronary artery imaging. Magn Reson
of technical aspects and future directions. Magn Reson Imaging. Med. 2002;47:476–481.
1989;7:445–455. 31. Wang Y, Ehman RL. Retrospective adaptive motion correction for
11. Hofman MB, Paschal CB, Li D, Haacke EM, van Rossum AC, Sprenger M. navigator-gated 3D coronary MR angiography. J Magn Reson Imaging.
MRI of coronary arteries: 2D breath-hold vs. 3D respiratory-gated 2000;11:208–214.
acquisition. J Comput Assist Tomogr. 1995;19:56–62. 32. Keegan J, Gatehouse PD, Yang GZ, Firmin DN. Adaptive motion
12. Oshinski JN, Hofland L, Mukundan S, Dixon WT, Parks WJ, Pettigrew RI. correction of interleaved spiral images and velocity maps: implications
Two-dimensional coronary MR angiography without breath-holding. for coronary imaging. Proceedings of the 15th Annual Meeting of the
Radiology. 1996;201:737–743. ESMRMB. MAGMA. 1998;6(suppl 1):67.
13. Sachs TS, Meyer CH, Irarrazabal P, Hu BS, Nishimura DG, Macovski A. 33. Korin HW, Ehman RL, Riederer SJ, Felmlee JP, Grimm RC. Respiratory
The diminishing variance algorithm for real-time reduction of motion kinematics of the upper abdominal organs: a quantitative study. Magn
artifacts in MRI. Magn Reson Med. 1995;34:412–422. Reson Med. 1992;23:172–178.
14. Maki JH, Prince MR, Londy FJ, Chenevert TL. The effects of time varying 34. Gierada DS, Curtin JJ, Erickson SJ, Prost RW, Strandt JA, Goodman LR.
intravascular signal intensity and k-space acquisition order on three- Diaphragmatic motion: fast gradient recalled echo MR imaging in
dimensional MR angiography image quality. J Magn Reson Imaging. healthy subjects. Radiology. 1995;194:879–884.
1996;6:642–651. 35. McConnell MV, Khasgiwala VC, Savord BJ, et al. Comparison of
15. Jhooti P, Keegan J, Gatehouse PD, et al. 3D coronary artery imaging with respiratory suppression methods and navigator locations for MR
phase reordering for improved scan efficiency. Magn Reson Med. coronary angiography. AJR Am J Roentgenol. 1997;168:1369–1375.
1999;41:555–562. 36. Sachs TS, Meyer CH, Pauly JM, Hu BS, Nishimura DG, Macovski A. The
16. Sinkus R, Bornert P. Motion pattern adapted real-time respiratory gating. real-time interactive 3D-DVA for robust coronary MRA. IEEE Trans Med
Magn Reson Med. 1999;41:148–155. Imag. 2000;19:73–79.
17. Sinkus R, Börnert P. Extension of real-time MR gating to cope with 37. Jahnke C, Nehrke K, Paetsch I, et al. Improved bulk myocardial motion
changes in motion pattern: making MR gating autarkic. In: Proceedings of suppression for navigator-gated coronary magnetic resonance imaging. J
the Sixth Scientific Meeting of ISMRM. Sydney: 1998:2127. Magn Reson Imaging. 2007;26:780–786.
18. Jhooti P, Gatehouse PD, Keegan J, Bunce HH, Taylor AM, Firmin DN. 38. Bellenger NG, Gatehouse PD, Rajappan K, Keegan J, Firmin DN, Pennell
Phase ordering with automatic window selection (PAWS): a novel DJ. Left ventricular quantification in heart failure by cardiovascular MR
motion-resistant technique for 3D coronary imaging. Magn Reson Med. using prospective respiratory navigator gating: comparison with
2000;43:470–480. breath-hold acquisition. J Magn Reson Imaging. 2000;11:411–417.
19. Hackenbroch M, Nehrke K, Gieseke J, et al. 3D motion adapted gating 39. Wang Y, Grimm RC, Felmlee JP, Riederer SJ, Ehman RL. Algorithms for
(3D MAG): a new navigator gated 3D coronary MR-angiography. Eur extracting motion information from navigator echoes. Magn Reson Med.
Radiol. 2005;1598–1606. 1996;36:117–123.
20. Langreck H, Schnackenburg B, Nehrke K, et al. MR coronary artery 40. Nehrke K, Bornert P, Manke D, Bock JC. Free-breathing cardiac MR
imaging with 3D motion adapted gating (MAG) in comparison to a imaging: study of implications of respiratory motion: initial results.
standard prospective navigator technique. J Cardiovasc Magn Reson. Radiology. 2001;220:810–815.
2005;7:793–797. 41. Nguyen TD, Nuval A, Mulukutla S, Wang Y. Direct monitoring of
21. Jhooti P, Keegan J, Firmin DN. A fully automatic and highly efficient coronary artery motion with cardiac fat navigator echoes. Magn Reson
navigator gating technique for high-resolution free-breathing Med. 2003;50:235–241.
83.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

42. Pai VM, Wen H. Rapid-motion-perception based cardiac navigators: 56. Henningsson M, Koken P, Stehning C, Razavi R, Prieto C, Botnar RM.
using the high flow blood volume as a marker for the position of the Whole-heart coronary MR angiography with 2D self-navigated image
heart. J Cardiovasc Magn Reson. 2003;5:531–543. reconstruction. Magn Reson Med. 2012;67:437–445.
43. Larson AC, White RD, Laub G, McVeigh ER, Li D, Simonetti OP. 57. Moghari MH, Roujol S, Henningsson M, et al. Three-dimensional heart
Self-gated cardiac cine MRI. Magn Reson Med. 2004;51:93–102. locator for whole-heart coronary magnetic resonance angiography. Magn
44. Stehning C, Bornert P, Nehrke K, Eggers H, Stuber M. Free-breathing Reson Med. 2014;71:2118–2126.
whole-heart coronary MRA with 3D radial SSFP and self-navigated 58. Bhat H, Ge L, Nielles-Vallespin S, Zuehlsdorff S, Li D. 3D radial sampling
image reconstruction. Magn Reson Med. 2005;54:476–480. and 3D affine transform-based respiratory motion correction technique
45. Lai P, Bi X, Jerecic R, Li D. A respiratory self-gating technique with for free-breathing whole-heart coronary MRA with 100% imaging
3D-translation compensation for free-breathing whole-heart coronary efficiency. Magn Reson Med. 2011;65:1269–1277.
MRA. Magn Reson Med. 2009;62:731–738. 59. Pang J, Bhat H, Sharif B, et al. Whole-heart coronary MRA with 100%
46. Piccini D, Littmann A, Nielles-Vallespin S, Zenge MO. Respiratory respiratory gating efficiency: self-navigated three-dimensional
self-navigation for whole-heart bright-blood coronary MRI: methods for retrospective image-based motion correction (TRIM). Magn Reson Med.
robust isolation and automatic segmentation of the blood pool. Magn 2014;71:67–74.
Reson Med. 2012;68:571–579. 60. Schmidt JF, Buehrer M, Boesiger P, Kozerke S. Nonrigid retrospective
47. Piccini D, Monney P, Sierro C, et al. Respiratory self-navigated respiratory motion correction in whole-heart coronary MRA. Magn
postcontrast whole-heart coronary MR angiography: initial experience in Reson Med. 2011;66:1541–1549.
patients. Radiology. 2014;270:378–386. 61. Henningsson M, Prieto C, Chiribiri A, Vaillant G, Razavi R, Botnar RM.
48. Hardy CJ, Saranathan M, Zhu Y, Darrow RD. Coronary angiography by Whole-heart coronary MRA with 3D affine motion correction using 3D
real-time MRI with adaptive averaging. Magn Reson Med. image-based navigation. Magn Reson Med. 2014;71:173–181.
2000;44:940–946. 62. Manke D, Rosch P, Nehrke K, Bornert P, Dossel O. Model evaluation and
49. Hardy CJ, Zhao L, Zong X, Saranathan M, Yucel EK. Coronary MR calibration for prospective respiratory motion correction in coronary MR
angiography: respiratory motion correction with BACSPIN. J Magn Reson angiography based on 3D image registration. IEEE Trans Med Imaging.
Imaging. 2003;17:170–176. 2002;21:1132–1141.
50. Henningsson M, Smink J, Razavi R, Botnar RM. Prospective respiratory 63. Manke D, Nehrke K, Bornert P. Novel prospective respiratory motion
motion correction for coronary MR angiography using a 2D image approach for free breathing coronary angiography using patient-adapted
navigator. Magn Reson Med. 2013;69:486–694. affine motion model. Magn Reson Med. 2003;50:122–131.
51. Kawaji K, Spincemaille P, Nguyen TD, et al. Direct coronary motion 64. McLeish K, Hill DL, Atkinson D, Blackall JM, Razavi R. A study of the
extraction from a 2D fat image navigator for prospectively gated motion and deformation of the heart due to respiration. IEEE Trans Med
coronary MR angiography. Magn Reson Med. 2014;71:599–607. Imaging. 2002;21:1142–1150.
52. Wu HH, Gurney PT, Hu BS, Nishimura DG, McConnell MV. Free- 65. Nehrke K, Bornert P. Prospective correction of affine motion for arbitrary
breathing multiphase whole-heart coronary MR angiography using MR sequences on a clinical scanner. Magn Reson Med. 2005;54:1130–1138.
image-based navigators and three-dimensional cones imaging. Magn 66. Fischer RW, Botnar RM, Nehrke K, Boesiger P, Manning WJ, Peters DC.
Reson Med. 2013;69:1083–1093. Analysis of residual coronary artery motion for breath hold and
53. Keegan J, Gatehouse PD, Yang GZ, Firmin DN. Non-model-based navigator approaches using real-time coronary MRI. Magn Reson Med.
correction of respiratory motion using beat-to-beat 3D spiral fat-selective 2006;55:612–618.
imaging. J Magn Reson Imaging. 2007;26:624–629. 67. Nagel E, Bornstedt A, Hug J, Schnackenburg B, Wellnhofer E, Fleck E.
54. Scott AD, Keegan J, Firmin DN. High-resolution 3D coronary vessel wall Noninvasive determination of coronary blood flow velocity with
imaging with near 100% respiratory efficiency using epicardial fat magnetic resonance imaging: comparison of breath-hold and navigator
tracking: reproducibility and comparison with standard methods. J Magn techniques with intravascular ultrasound. Magn Reson Med.
Reson Imaging. 2011;33:77–86. 1999;41:544–549.
55. Scott AD, Keegan J, Firmin DN. Beat-to-beat respiratory motion 68. Keegan J, Gatehouse P, Mohiaddin RH, Yang GZ, Firmin D. Comparison
correction with near 100% efficiency: a quantitative assessment using of spiral and FLASH phase velocity mapping, with and without
high-resolution coronary artery imaging. Magn Reson Imaging. breath-holding, for the assessment of right and left coronary artery blood
2011;29:568–578. flow velocity. J Magn Reson Imaging. 2004;20:953–960.
8 
Cardiovascular Magnetic Resonance
Assessment of Myocardial Oxygenation
Rohan Dharmakumar, Sotirios A. Tsaftaris, Hsin-Jung Yang, and Debiao Li

MYOCARDIAL OXYGENATION: SUPPLY for the diagnosis of ischemic heart disease and the evaluation of thera-
pies for improving the balance between MBF and oxygen demand.
VERSUS DEMAND
If we assume that the blood oxygen saturation of hemoglobin in
Under physiologic conditions, myocardial blood flow (MBF), myocardial the arterial blood is 100% and is Y in the venous pool, then the MVO2
oxygen consumption (MVO2), and myocardial mechanics are intimately can be estimated to first order by Fick’s law as
related. Therefore it is not surprising that the key disease processes
MVO2 = F × Hct × (1− Y )
involving the heart manifest from imbalances between myocardial
oxygen supply and demand. Consequently, the noninvasive assessment where F (in mL/g/min) is the blood flow to the myocardium and Hct
of imbalances in myocardial oxygen supply and demand, particularly on (in percent) is the hematocrit of the blood.31 Hence if F, Y, and Hct are
a regional basis, is of critical importance in both clinical cardiology and known, it is possible to estimate MVO2.
for cardiovascular research. The noninvasive quantification of MVO2 Furthermore knowledge of myocardial venous blood oxygenation
was not possible until it was shown that positron emission tomography can permit noninvasive evaluation of myocardial perfusion reserve,
(PET), using 11C-acetate, permits accurate quantification of MVO2.1–4 defined as the ratio between the peak myocardial perfusion rate at
Using this approach, numerous studies have demonstrated the salutary maximum vasodilation and rest.32 Under pharmacologic vasodilation,
effects of restoring nutritive perfusion on MVO2 and cardiac function such as that in response to dipyridamole or adenosine, normal coronary
and the importance of preserving MVO2 as a descriptor and probable blood flow increases several fold, whereas the oxygen consumption
determinant of myocardial viability in both acute and chronic ischemic remains relatively unaltered, leading to an increase in myocardial venous
disease.5–8 However, PET studies are limited by relatively poor spatial blood oxygen saturation. However, with progressive coronary lumen
resolution, limited availability mainly due to short-life radiotracers, narrowing or with microvascular dysfunction, the maximal increase
and potentially harmful ionizing radiation, especially when repeated in MBF under vasodilatory stress is blunted proportionately relative
examinations are needed. to the healthy myocardium. This is illustrated in the case of CAD in
Magnetic resonance imaging (MRI) has become an important clini- Fig. 8.1. Such perfusion abnormalities lead to regional differences in
cal imaging modality because it is noninvasive, does not require iodin- myocardial venous blood oxygenation, allowing for the assessment of
ated contrast media or ionizing radiation, and is widely available. MRI the functional significance of CAD. Extensive clinical support now exists
can also provide structural and functional information as well as report for imaging myocardial perfusion research with CMR using Gd-based
on several important biomarkers in the same setting. To date, multiple contrast agents (GBCAs).33–36 However, it is contraindicated in severe
cardiovascular magnetic resonance (CMR) applications have been chronic kidney disease (CKD, stage 4 or 5). According to the National
developed, including anatomic imaging of the heart and great vessels, Institutes of Health (NIH) and the United States Renal Data System
coronary artery imaging, methods to characterize myocardial infarction (USRDS), the prevalence and the cost of treating new cases of CKD has
with or without gadolinium (Gd) contrast media, methods for char- more than doubled in the past 10 years and this trend is predicted to
acterizing extracellular space, methods for evaluating myocardial wall grow given the increasing prevalence of diabetes and its contribution to
motion, and first-pass perfusion (FPP) for the identification of perfusion the development of CKD.37 Moreover, recent imaging/autopsy studies
defects in the myocardium. Over the past two decades, efforts have also from Italy,38 the United States,39 and Japan40 have shown that even in
been made to use CMR to determine regional myocardial blood oxy- patients with normal kidney function, there are retained Gd deposits
genation levels.9–30 The blood oxygenation state of the myocardium in the brain—a finding that is contrary to the widely held belief that
reflects the combined effects of MBF and oxygen extraction (which Gd is fully cleared from the body within hours of infusion.41 These
together reflect MVO2). Thus a change in myocardial blood oxygenation observations are central to the growing interest and emphasis on non–
secondary to imbalances in oxygen supply and demand would be useful Gd-based approaches for studying myocardial perfusion, particularly
in evaluating disease processes such as coronary artery disease (CAD) based on changes in blood oxygenation, in all subjects suspected of
or microvascular disease (MVD), which can lead to impaired myocardial having ischemic heart disease.
perfusion reserve. Noninvasive assessment of myocardial venous blood In this chapter, we provide an overview of the basic biophysical
oxygenation may permit the measurement of oxygen extraction. When concepts that allow for the assessment of myocardial changes in blood
coupled with flow, these data would allow for measurement of MVO2. oxygenation. We then summarize the preclinical and clinical literature
Anatomic, functional, and metabolic information can then be obtained to date in the assessment of myocardial oxygenation. This is followed by
in a single CMR study, thereby providing a comprehensive examination growing literature on image-processing methods that have the capacity

84
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 85

blood.44,45 This observation has allowed for the acquisition of oxygen-


sensitive images permitting the discrimination between arteries and
5.0
Hypertrophy veins.46 Its utility for detecting chronic mesenteric ischemia47 and the
↑ Heart rate identification and quantification of cardiac shunts associated with
↑ Preload congenital abnormalities have also been demonstrated.48
Etc.
50% An extension of this phenomenon into the microcirculation opened
the door for assessing myocardial oxygenation changes. In the myocar-
Maximum dium, nearly 90% of the blood volume is within the capillaries49; accord-
vasodilation ingly, our discussions will be limited to capillary beds. A change in
blood oxygenation in the capillary bed leads to changes in magnetic
70%
field variations between the red blood cells and plasma and between
Coronary blood flow

3.0
the intravascular and extravascular spaces. Following the excitation
of the magnetization onto the transverse plane, these field variations
cause the spins to lose coherence, leading to a decay of the magnetic
80% ↑ Flow resonance (MR) signal.50–55 In particular, the severity of the field varia-
prior to tion due to changes in blood oxygen saturation directly determines the
vasodilation
rate of loss of the spin coherence (MR signal). This phenomenon, referred
to as the BOLD effect, implies that when the capillaries contain deoxy-
genated blood, with all else remaining the same, the MR signal associated
85% with the deoxygenated state (rest) will be lower than that of the hyper-
emic state (vasodilation) when the capillary oxygenation is substantially
80% 50%
1.0 higher (~30% [resting] vs. ~80% [hyperemic]). This allows for detecting
85% 70% regional myocardial oxygen differences as regions of signal loss with
90%
imaging sequences sensitive to local field inhomogeneities.9 In addition,
the sensitivity of the MR signal to the BOLD effect is dependent on
the blood volume, hematocrit, and choice of pulse sequences used.50–55
Both gradient and spin echo sequences as well as balanced steady-state
Coronary pressure free precession (bSSFP) methods have been used to probe these effects.50,55
FIG. 8.1  A schematic showing the relationship between the coronary BOLD effect was first demonstrated in the brain,56–59 before being
flow and coronary arterial pressure. The solid curve represents the normal adopted for cardiac applications.9–15 However, there are important bio-
relationship. At a constant level of myocardial metabolic demand, coro- physical differences between BOLD CMR of the heart and brain. Spe-
nary artery flow is maintained constant over a wide range of coronary cifically, the heart has a larger blood volume fraction than the brain
artery pressures, between the bounds of maximum coronary vasodilation (~10% vs. ~4%) and the venous blood oxygen saturation in the heart
and constriction (dashed curves). The solid circle represents the normal
is approximately 30%, compared with approximately 60% in the brain.9
operating point under basal conditions; the solid triangle is the flow
This allows for a wider range of signal change with vasodilator-induced
observed at the same pressure during maximum vasodilation. Myocardial
flow reserve is the ratio of flow during vasodilation to that measured flow in the myocardium compared with the brain, which provides greater
before vasodilation. Note that the flow reserve decreases in a nonlinear BOLD sensitivity in the heart. However, in contrast to brain imaging,
manner with reduction of coronary pressure (or coronary artery stenosis). the challenge for myocardial BOLD CMR has been imaging artifacts
Also note that hypertrophy, increased heart rate, and increased preload from motion (cardiac, respiratory, and pulsatile blood flow), as well as
all decrease the coronary flow reserve. (Modified from Klocke FJ. Mea- bulk susceptibility shifts between heart and lung interface.
surements of coronary flow reserve: defining pathophysiology versus
making decisions about patient care. Circulation. 1987;76:1183–1189.) VASODILATORS IN THE ASSESSMENT OF
MYOCARDIAL OXYGENATION
to enable accurate visualization and quantification of blood-oxygen- Common Pharmacologic Vasodilators
level-dependent (BOLD) signal changes in the myocardium. Finally, we Presently, vasodilators appear to be essential for assessing myocardial
review the emerging methods that show promising evidence into how blood oxygenation, especially in the context of ischemic heart disease.
BOLD CMR can become a reliable tool for examining ischemic heart Their importance was first demonstrated in human subjects with mul-
disease in the clinical arena and conclude with a brief outlook on the tigradient echo methods using two different pharmacologic stress agents:
future of myocardial BOLD CMR. dipyridamole and dobutamine.14,16 Both agents induce hyperemia, but
with different effects on myocardial venous blood oxygenation. Dipyri-
damole is a potent coronary vasodilator that typically induces a 3- to
BIOPHYSICS OF MYOCARDIAL BOLD CONTRAST 4-fold increase in MBF with minimal effect on MVO2.60 Consequently,
Blood is a magnetically inhomogeneous medium in which the magnetic myocardial venous blood oxygen saturation increases as oxygen supply
susceptibility of red blood cells is strongly dependent on the blood (blood flow) exceeds demand (oxygen consumption). In contrast, dobu-
oxygen saturation (%O2), defined as the percentage of hemoglobin that tamine is a potent beta-agonist with a primary pharmacologic effect
is oxygenated.42,43 Because the susceptibility of blood plasma is generally to increase cardiac work.60 This results in a concomitant increase in
invariant, the cooperative binding of oxygen to the heme molecules MVO2, which triggers an increase in MBF. Thus oxygen supply and
within the red blood cells results in a detectable susceptibility difference demand remain largely balanced, and there is little to no change in
between plasma and the red blood cells. This susceptibility variation myocardial venous blood oxygen saturation.61
gives rise to local magnetic field inhomogeneities, resulting in local Vasodilators have also facilitated direct demonstration of the in
frequency variations that lead changes in T2* and apparent T2 of whole vivo correlation between BOLD CMR response (via changes in R2*
86 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

[or 1/T2*]) and venous blood oxygen saturation. In a well-controlled decrease in R2*. However, the blood volume fraction in the myocardium
canine model, a wide range of global myocardial venous blood oxygen also increases, which increases the hematocrit content of a voxel, which
saturation levels were created. Hyperemic conditions were induced by tends to increase myocardial R2*; this is the opposite of the effect of
the intravenous administration of dipyridamole and dobutamine. To increased oxygen saturation. Because a decrease in myocardial R2* was
induce hypoxemia, the oxygen content of the inspired gas was reduced observed in these studies, increased oxygen saturation clearly has the
by ventilating dogs with a mixture of 10% oxygen and 90% nitrogen, dominant effect over increased blood volume, but the apparent R2*
which reduced the oxygen saturation in both arteries and veins. To change as a function of %O2 is reduced because of the accompanied
correlate myocardial R2* with global venous blood oxygenation, venous blood volume effect. In contrast, during hypoxia, both %O2 in coronary
blood oxygen saturation levels were measured directly by coronary sinus sinus and the blood volume fraction increase, and their effects enhance
sampling. MBF was quantified invasively by the administration of radio- each other. As a result, the apparent change in R2* as a function of
labeled microspheres. Measurements of myocardial R2* were obtained %O2 is greater than that if blood volume fraction remained the same.
at baseline, during and after infusion of dipyridamole and dobutamine, These studies showed that by measuring MBV fraction changes using
and when the dogs were ventilated with hypoxic air. Paired arterial technetium-99m-labeled (99mTc-labeled) red blood cells at each of the
and coronary sinus blood samples were withdrawn at the six different interventions and correcting their effects on myocardial R2*, a more
stages of the study. Blood oxygen saturation levels were measured by linear relationship was found between R2* and the blood oxygen satu-
using a blood gas analyzer interfaced with an oximeter. Coronary sinus ration. Thus an accurate assessment of myocardial oxygen saturation
blood oxygen saturation levels ranged from 9% to 80% with experi- using CMR will probably require a correction for blood volume.
mental interventions with dipyridamole, dobutamine, or hypoxic air.
Administration of dipyridamole and dobutamine and ventilation of Hypercapnia as a New Potent Coronary Vasodilator
hypoxic air all increased MBF significantly, but significant decrease in It has long been known that carbon dioxide (CO2) may act as a vaso-
myocardial R2* was observed only with dipyridamole infusion, which dilator in the heart, but its sensitivity to impart sufficient coronary
indicates that myocardial R2* is a reflection of MBF only when myo- vasodilation to assess regional changes in MBF has not been clear.
cardial oxygenation demand is not significantly altered between rest Recent studies, enabled by prospective gas control allowing for rapid
and hyperemia. The relationship between the changes of myocardial and independent control of arterial CO2 and O2, have made it possible
R2* from baseline and the %O2 in the coronary sinus showed a linear to directly relate the influence of arterial CO2 on coronary vasodilation.
regression (r = 0.84), indicating a strong correlation between myocardial The use of targeted increases in arterial CO2 to vasodilate the coronary
R2* and %O2 in the coronary sinus. arteries and invoke changes in myocardial oxygenation and its relation
These studies also showed that both dipyridamole and hypoxic air to adenosine in the preclinical models and healthy human subjects has
increased myocardial blood volume (MBV) fraction in excess of 50%. been demonstrated (Fig. 8.2).62 However, its utility to assess changes in
However, their effects on R2* are manifested differently. With administra- myocardial oxygenation in patients with ischemic/nonischemic heart
tion of dipyridamole, %O2 in coronary sinus increases, which leads to a disease has not been demonstrated.

Baseline Hypercapnia
*
% hy peremic BOLD response

150
*
Raw images

140
130
120
110
Color overlays

100
90
80
70
Rest Plus 5 Plus 10 Adenosine
A B
FIG. 8.2  Effect of changing arterial carbon dioxide (CO2) on blood-oxygen-level-dependent (BOLD) cardio-
vascular magnetic resonance signal intensities in healthy humans. (A) Representative short-axis BOLD car-
diovascular magnetic resonance images collected from a healthy human at baseline (end-tidal CO2 [PETCO2]
= 37 mm Hg) and hypercapnia (PETCO2 = 47 mm Hg). BOLD signal intensity increased during hypercapnia.
For ease of visualization, color overlays of the left ventricle (with color bar showing BOLD signal intensity in
arbitrary units [a.u.]) corresponding to the gray-scale images are shown directly below. (B) Box plot showing
the dependence of % hyperemic BOLD response on PETCO2 and standard dose of adenosine: % hyperemic
response increased at higher PETCO2 values; response at Plus 10 did not differ from that because of standard
adenosine infusion. % hyperemic BOLD response for adenosine is from reported values in the literature.80
Asterisk denotes statistically significant difference relative to rest (P < .05). Top and bottom of boxes indicate
upper limit +1 standard deviation (SD) and lower limit −1 SD, respectively; error bars (whiskers) are maximum
and minimum of data. The mean and median are represented as a point and a band within the box, respec-
tively. For adenosine, the whiskers represent the boundaries of 1st and 99th percentile of the data. (Modified
from Yang HJ, Yumul R, Tang R, et al. Assessment of myocardial reactivity to controlled hypercapnia with
free-breathing T2-prepared cardiac blood-oxygen-level-dependent MR. Radiology. 2014;272:397–406.)
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 87

An alternate approach for modulating PaCO2 is through breathing flow to the affected region decreases the oxygen saturation of hemo-
maneuvers, where a subject is asked to hyperventilate, typically for 30 globin and aids in delineating regional perfusion differences. Over the
seconds, and then suspend breathing during imaging to invoke a hyper- past two decades, both clinical and preclinical animal studies have
emic response. The combined hyperventilation/suspension of breathing demonstrated BOLD CMR may be an alternative to FPP methods.
is expected to provide a significant change in PaCO2 to invoke a hyper- Some of the earliest studies of the utility of BOLD CMR to understand
emic blood flow response in the heart. To date, the value of this approach the perfusion deficits associated with coronary occlusion were performed
in modulating the BOLD CMR response has been demonstrated in with gradient echo sequences. These studies showed that the occlusion
healthy human subjects.63 Comparative studies between this approach of the left anterior descending artery (LAD) resulted in signal reduction
and prospective gas control methods to probe myocardial oxygenation in corresponding regions of the myocardium perfused by the LAD.11,12
changes in response to hypercapnia are yet to be conducted. These initial studies were followed by T2* mapping of the myocardium,
which demonstrated that the myocardial regions with perfusion deficits
MYOCARDIAL BOLD CMR: PRECLINICAL STUDIES also show preferential reduction in T2* under vasodilation. T2-prepared
gradient echo methods in canine models demonstrated that it may be
T2*- and T2-Prepared Methods possible to extend the myocardial BOLD technique to three-dimensional
To date, a number of isolated heart and animal studies have demon- (3D) imaging and acquire multiple slices with BOLD contrast.21 However,
strated that vasodilator interventions that alter the blood oxygenation poor image quality and signal-to-noise ratio (SNR), long scan times,
levels result in BOLD signal changes.9–13,21,22,24–30 One of the first studies and inadequate sensitivity are notable limitations of the gradient echo
to demonstrate the feasibility of assessing myocardial oxygenation on technique at 1.5 T.
the basis of the BOLD effect was performed in a rat model. This study T2-prepared methods have been proposed as an alternative to T2*-
showed significant signal loss in both the left ventricular chamber and based methods. Whereas T2-prepared methods employing spiral readouts
the myocardium during apnea.9 A subsequent study in an isolated rabbit showed the capability to capture the BOLD contrast, off-resonance-based
heart model confirmed a substantial correlation between the gradient blurring was a key limitation.22,23 A subsequent T2-preparation method
echo image intensity of the myocardium and deoxyhemoglobin con- with a bSSFP readout showed significant improvements in SNR, image
centration levels.13 In large animal models, gradient echo myocardial quality, and scan times over the gradient echo techniques29,30 and good
signal enhanced significantly after the infusion of dipyridamole,11 pre- sensitivity for evaluating myocardial BOLD signal changes. This tech-
sumably because of an increase in MBF in the absence of a corresponding nique has been employed in canine models with varying degrees of
increase in oxygen demand, resulting in a decrease in myocardial venous stenosis of the left circumflex coronary arteries (LCXs) in the presence
blood oxygen saturation. Several studies have now independently of systemic adenosine infusion to demonstrate the effect of perfusion
validated these studies in large animal models with T2-prepared abnormalities with BOLD effect.30 Results showed a visually discernible
methods.22,27,28 Canine studies also suggest that it is possible to derive BOLD signal drop in the inferior and inferolateral walls supplied by
myocardial oxygen extraction fraction and quantitative perfusion reserve the LCXs as well as a close correspondence between flow reductions in
values with BOLD CMR.24–26 the same region as identified by the FPP images and microsphere flow
Perhaps the greatest expectation of BOLD CMR imaging is in detect- maps (Fig. 8.4).
ing regional differences in myocardial oxygenation caused by focal CAD. Prospective, targeted control of arterial partial pressure of CO2
Fig. 8.318 shows a schematic of the coronary vessels that supply the (PaCO2), as discussed earlier, has been shown to be valuable in modu-
different myocardial territories of the mid left ventricle in humans.18 lating coronary blood flow. Recent studies using T2-prepared bSSFP-
Luminal narrowing of the coronary vessels can lead to regional perfu- based BOLD CMR demonstrated the capability of physiologically
sion deficits in the associated myocardial territories during pharmacologic tolerable hypercapnia to induce myocardial hyperemia, and compared
stress. Whereas vasodilators are effective at increasing blood flow to the this response with intravenous adenosine. These studies showed that
myocardium perfused by healthy coronary vessels, MBF to territories repeat targeting of a desirable hypercapnic stimulus punctuated by
distal to the coronary artery stenosis is limited. The reduction in blood return of arterial CO2 levels to baseline leads to a reproducible BOLD
response. These findings provide early evidence into novel means of
examining myocardial oxygenation through a benign/repeatable coronary
vasodilator.
Anterior
Most studies to date have employed 2D methods, with the majority
of them using T2*- or T2-based (weighted or mapping) methods. These
Antero- LAD
septal approaches often require suspension of breathing and cannot provide
Lateral RCA full ventricular coverage over the time period over which adenosine is
RCA, typically delivered. To overcome these limitations, a fast, free-breathing
LCX when
3D T2-mapping technique that uses near-perfect imaging efficiency,
left dominant
Infero- with insensitivity to heart-rate changes which take place in the presence
LCX,
septal RCA, when of vasodilator stress, allowing for full coverage of the whole left ventricle
right dominant within 5 minutes, has been developed. This approach demonstrated in
Inferolateral
preclinical studies promises to offer a more refined BOLD CMR method
with the 3D free-breathing T2-mapping technique and has the capability
Inferior
to map the hyperemic BOLD response throughout the left ventricle
FIG. 8.3  A schematic representation of the myocardial segments of
during adenosine administration.
the mid ventricular section with the assigned coronary territories. LAD,
Left anterior descending coronary artery; LCX, left circumflex coronary
artery; RCA, right coronary artery. (Modified from Friedrich MG, Niendorf
Steady-State BOLD CMR: Cardiac Phase–Resolved
T, Schulz-Menger J, Gross CM, Dietz R. Blood oxygen level-dependent Imaging of Myocardial Oxygenation
magnetic resonance imaging in patients with stress-induced angina. To date, systematic studies have demonstrated that bSSFP imaging can be
Circulation. 2003;108:2219–2213.) used in the assessment of oxygen-sensitive imaging. Controlled in vitro
88 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

separated, based on %O2 differences.65 The advantages of the bSSFP


method over other BOLD CMR methods is increased in SNR. In the
heart, however, there are other advantages, including cardiac phase–
resolved (CP) imaging allowing one to capture potential BOLD effects
as well as function in one acquisition. This was first demonstrated in
canines with and without coronary stenosis. Two-dimensional (2D)
cardiac phase–resolved BOLD (CP-BOLD) bSSFP with a relatively long
TR (6.3 ms) was used to demonstrate that occlusion of the LCX leads
to regional myocardial oxygen deficit in the coronary territory sup-
plied by LCX. The results were compared with the FPP technique and
A B validated with microsphere-based perfusion analysis. 2D CP-BOLD
CMR accurately predicted the regional MBF deficit region identified
by the first-pass technique employing an exogenous contrast media.
The benefits of CP-BOLD CMR with this approach permits increased
confidence for evaluating myocardial BOLD signal changes in the pres-
ence of a coronary artery stenosis. Canine studies by Vöhringer et al.66
have demonstrated that CP-BOLD CMR can identify changes in myo-
cardial oxygenation and flow in the presence of intracoronary infusion
of acetylcholine, an endothelium-dependent vasodilator, and adenosine,
an endothelium-independent vasodilator. These studies showed that
CP-BOLD CMR has the capability to report on oxygenation changes
C D in the setting of CAD or MVD.
Several noncardiac studies have shown that BOLD contrast is directly
tied to field strength, and this was demonstrated in the heart as well as
through theoretical simulations, and animal studies have also established
this in the heart.67 These studies showed that at 3 T, a 3-fold increase
in oxygen sensitivity could be expected in comparison to 1.5 T in theory.
Experimental canine studies showed that a 2.5-fold ± 0.2-fold increase
in BOLD sensitivity is possible at 3 T relative to 1.5 T (Fig. 8.5).67 On
the basis of the relationship between BOLD signal changes and micro-
sphere perfusion, it was found that the minimum regional perfusion
differences that can be detected with CP-BOLD CMR at 1.5 T and 3.0
T were 2.9 and 1.6, respectively. These findings suggest that CP-BOLD
E F
CMR at 3 T may have the necessary sensitivity to detect the clinically
FIG. 8.4  Preclinical studies for detecting perfusion defects using T2-pre- required minimum flow difference of 2.0 that is achievable with FPP
pared balanced steady-state free precession cardiovascular magnetic methods.
resonance. Comparison of blood-oxygen-level-dependent (BOLD) magnetic Vasodilatory stress is the standard paradigm for probing myocardial
resonance images acquired with T2-prepared steady-state free preces- oxygenation (O2) changes due to coronary artery stenosis on the basis
sion (TR/TE = 3.0/1.3 ms, 40-ms T2 preparation) at rest (A and C) and of BOLD MRI. However, because vasodilation is typically achieved
during stress stenosis (B and D), with corresponding first-pass image
with provocative stress, approaches that can identify the presence of
(E) (saturation recovery steady-state free precession, TR/TE = 3.0/1.5 ms)
and microsphere flow image (F). Arrows indicate the region of reduced
stenosis on the basis of microvascular alterations at rest are highly
perfusion due to stenosis in the BOLD stress stenosis and first-pass desirable. It is known that MBV varies throughout the cardiac cycle;
images. Note the clear delineation of the myocardial region fed by the MBV increases during diastole and decreases during systole.68,69 It has
stenosed artery in the BOLD images, as well as the excellent match- also been shown that changes in MBV lead to increased O2 extraction
up of the stress stenosis BOLD image with the first-pass image and by cardiomyocytes.70 Thus MBV and O2 are expected to vary at different
microsphere flow map. (The microsphere flow map was generated by parts of the cardiac cycle. In particular, in diastole, it is expected that
calculating flow ratios in each segment in relation to one reference MBV and O2 extraction are maximal, whereas in systole, MBV and O2
segment. The scale indicates black for 0 flow ratio and white for a extraction are minimal. In addition, as MBV increases, even at a stable
5-fold flow ratio.) (Modified from Shea SM, Fieno DS, Schirf BE, et al. level of O2, the number of deoxygenated hemoglobin molecules within a
T2-prepared steady-state free precession blood oxygen level-dependent
voxel increases, causing a proportionate elevation in the local magnetic
MR imaging of myocardial perfusion in a dog stenosis model. Radiology.
2005;236:503–509.)
field inhomogeneities.71 Moreover, with increasing grade of stenosis, the
MBV in the myocardial territory supplied by a stenotic artery increases
in systole.19,72–74 Thus the relative MBV and O2 changes between systole
and diastole are expected to be different between myocardial territo-
studies with blood samples oxygenated to various levels revealed that ries supplied by healthy and stenotic coronary arteries. Moreover, it is
when the off-resonance effects are minimized through the appropriate also known that T1 of myocardium is dependent on MBV and that
choice of flip angle and phase-cycling scheme, oxygen-sensitive contrast the apparent T2 is dependent on blood O2. Because bSSFP signals are
can be realized in whole blood in a repetition time (TR)-dependent approximately T2/T1 weighted, it was expected that CP-BOLD signal
manner.64 The results showed that relatively long TRs (compared with intensities at systole and diastole may reflect changes in MBV and blood
those that are used in conventional bSSFP imaging) are necessary to O2. In addition, because stenosis leads to an increase in systolic MBV
establish oxygen-sensitive contrast in bSSFP images in blood. This and is accompanied by a reduction in blood O2, it was hypothesized
was subsequently used to demonstrate that arteries and veins can be that systolic and diastolic CP-BOLD signal intensities may be used to
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 89

0%

A B C D

100%
A′ B′ C′
FIG. 8.5  Demonstration of increased myocardial blood-oxygen-level-dependent sensitivity at 3 T. Short-axis
two-dimensional balanced steady-state free precession (bSSFP) cardiovascular magnetic resonance images
obtained at 1.5 T (top row, A–C) and at 3 T (bottom row, A′–C′) in canines. Images A and A′ are bSSFP
images obtained without stenosis, images B and B′ are bSSFP images at systole under severe stenosis of
the left circumflex coronary artery (LCX), and images C and C′ are the corresponding first-pass perfusion
(FPP) images acquired under stenosis of similar extent as in B and B′. Images D and D′ represent the spatial
map (scale provided by the gray-scale bar) of percent difference in microsphere-based regional flow between
prestenosis and severe stenosis in the presence of adenosine infusion at 1.5 T and 3 T, respectively. The
arrows subtend the suspected regions (LCX territory), where the perfusion deficits are expected to develop
as a result of LCX stenosis in dogs. Note the discriminating signal loss in these regions in images B and B′
and the close correspondence between the FPP (C and C′) and microsphere-based flow difference maps
(D and D′). (Modified from Dharmakumar R, Mangalathu Arumana J, Tang R, et al. Assessment of regional
myocardial oxygenation changes in the presence of coronary artery stenosis with balanced SSFP imaging at
3.0 T: theory and experimental evaluation in canines. J Magn Reson Imaging. 2008;27:1037–1045.)

detect the ischemic territories at resting states. These hypotheses were tomography (SPECT) with the aim of visualizing regional oxygen deficits
shown to be accurate with simulations and canine experiments with under pharmacologic stress. This study was performed in 25 patients
maximal coronary stenosis using a 2D CP-BOLD sequence (Fig. 8.6) (23 patients with >50% stenosis, 10 of whom had severe stenosis, i.e.,
designed to mitigate against flow-mediated phase changes, which oth- >75%). Results demonstrated that significant changes in suspected
erwise would appear as “ghost” artifacts.75 Although these results are regions of the myocardium were apparent in patients with severe ste-
encouraging, further validation of the proposed method is required. nosis. Receiver operating characteristic (ROC) analysis of both BOLD
Moreover, technical advances in sequence development for 3D CP-BOLD CMR and thallium SPECT methods showed that the area under the
(for minimizing potential signal variations due to through-plane motion), ROC curve is 0.66 and 0.73, respectively. These results indicated that
methods to overcome image artifacts in bSSFP images at 3 T, improve- both methods have limitations in identifying regional perfusion deficits
ments in image processing, along with additional preclinical studies and that the limitations are attributable to suboptimal resolution of
at more modest coronary stenosis may be necessary before clinical SPECT images and the well-known image artifacts associated with
translation. gradient-recalled T2*-weighted methods in the heart, as discussed earlier.
In addition to demonstrating the feasibility of the BOLD method for
studying CAD in patients (Fig. 8.7),18 these studies demonstrated that
CLINICAL EXPERIENCE WITH BOLD CMR improved CMR methods are needed.
Successful demonstrations of the BOLD effect in animal models have Another clinical study by Wacker and colleagues19 examined the
provided the impetus for myocardial BOLD CMR in patients with possibility of evaluating oxygen-sensitive changes in patients with
CAD. In the past decade, there has been an extensive number of studies CAD without pharmacologic stress. Their study included a control
in ischemic and nonischemic patients. group of 16 healthy subjects (age 31 ± 10 years old) with no history of
cardiovascular disease and 16 patients (63 ± 9 years old) with single-
T2*-Based BOLD CMR in Patients vessel CAD (degree of stenosis >70%) identified by x-ray coronary
In one of the first and pioneering studies, T2*-weighted BOLD CMR angiogram. Within the control group, T2* mapping of the mid left
method was compared against single-photon emission computed ventricular myocardium was homogeneous (35 ± 3 ms) and increased
90 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

LCX [Baseline] LCX [Baseline]


1.2 LAD [Baseline] 1.2 LAD [Affected]
Line of identity End-systole End-systole
End-diastole End-diastole

Normalized regional
Normalized regional

1.1 1.1

intensity
intensity

1.0 1.0

0.9 0.9

Line of identity
0.8 0.8
0 25 50 75 100 0 25 50 75 100
A Nominal cardiac phase B Nominal cardiac phase

1.4 Theor y Ex per iment s

1.3
Systolic to diastolic ratio

LCX [ Remote ] LAD [ Affected ]

1.2
Line of identity
(S/D)

1.1

1.0

0.9

0.8
T1 T2 SSFP Baseline Stenosis
FIG. 8.6  Detecting ischemic territories with cardiac phase–resolved blood-oxygen-level-dependent at rest.
Top left (A and B), Regional mean intensities from left anterior descending (LAD) and left circumflex coronary
artery (LCX) regions under baseline (A) and 90% (or more) LAD stenosis (B) normalized by the regional mean
intensities at end diastole (ED) (shown with red circles) as a function of nominal percentage of cardiac cycle
are shown. Green circles indicate end systole (ES). A line of identity (between ES and ED, i.e., segmental
biomarker [S/D] = 1) is also shown for reference. During baseline conditions, both regions have S/Ds >1 (1.1
and 1.07, for LCX and LAD, respectively). Under stenosis, however, only the LCX region (remote territory)
maintained an S/D >1 (at 1.17). Conversely, the S/D for the LAD region decreased significantly below 1 (at
0.93), in line with the hypothesis.100 Top right (bull’s eye plots), S/D under baseline (left) and stenosis (right)
conditions. Under baseline, all S/D ratios are greater than unity (1.11 ± 0.25). Under stenosis, only the regions
corresponding to the LAD territory have a markedly reduced S/D values (1.01 ± 0.01). (Modified from Tsaftaris
SA, Zhou X, Tang R, Li D, Dharmakumar R. Detecting myocardial ischemia at rest with cardiac phase-resolved
CMR. Circ Cardiovasc Imaging. 2013;6:311–319.)

uniformly under the influence of dipyridamole (40 ± 4 ms). However, the ischemic segments (26.7 ± 11.6 ms) compared with normal (31.9
in patients with CAD, the suspected regions of myocardium were at ± 11.9 ms; P < .0001) and nonischemic segments (31.2 ± 12.2 ms; P
a lower T2* than the healthy regions (31% ± 9%). Furthermore, this = .0003). Under adenosine stress T2* values increased significantly in
noted deviation increased under the influence of dipyridamole (43% ± normal segments only (37.2 ± 14.7 ms; P < .0001).
21%). This study also followed patients who underwent coronary artery Myocardial perfusion reserve changes in hypertensive hypertrophic
bypass grafting or stenting of the diseased vessel to reestablish flow to patients have also been studied with BOLD imaging using the T2*
the affected regions of the myocardium. Their findings showed that maps.20 This study included 10 patients (43 ± 4 years old) with hyper-
following the intervention, the T2* homogeneity of the myocardium tension (blood pressure >140/90 mm Hg), hypertrophic hearts (assessed
was improved, probably owing to the improved perfusion facilitated by 2D echocardiography), and no coronary artery stenosis, and 9 healthy
by the more patent coronary vessels. This observation has been linked subjects with no heart disease. The results showed that the change in
to capillary recruitment in the distal beds of stenotic coronary vessels, T2* between basal and dipyridamole-infused states (ΔT2*) was approxi-
which results in an increase in blood volume and thus an elevation mately 3-fold lower in hypertrophic patients than in healthy controls.
of deoxyhemoglobin concentration per unit of imaging voxel in the This observation is consistent with previous reports32 that myocardial
affected territories.19,58 In a subsequent clinical study, T2* BOLD CMR perfusion reserves are reduced in people with hypertrophic hearts (see
contrast was extended to 3 T, where T2* maps were acquired under rest Fig. 8.1). From the perspective of myocardial oxygenation, in hyper-
and vasodilator stress in subjects with and without suspected CAD.76 trophic patients, vasodilatory mechanisms are compromised, depending
The results were compared against invasive angiography. Myocardial on the severity of the disease, and are not fully effective at altering the
segments were separated into nonischemic (territories supplied by coro- oxygenation of the myocardium as in a healthy heart. This inability to
naries with nonsignificant stenosis), ischemic (supplying artery with alter blood oxygenation between the basal and dipyridamole-infused
stenosis ≥50%) and normal (reference segments of subjects without any states in hypertrophic patients is probably the reason for the substantially
CAD). Results demonstrated significant T2* reduction during rest in different T2* between healthy subjects and hypertrophic patients.
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 91

Pat. #1 Pat. #2 Pat. #3


Rest
Stress No significant stenosis Stenosis LAD, R. diag., and R. marg. Critical stenosis RCA

BOLD-CMR SPECT BOLD-CMR SPECT BOLD-CMR SPECT


FIG. 8.7  Gradient echo planar blood-oxygen-level-dependent cardiovascular magnetic resonance (BOLD-CMR)
images obtained before (top) and during (bottom) adenosine infusion and corresponding thallium single-photon
emission computed tomography (SPECT) images from three representative patients. Left, Patient (Pat. #1)
without a 0% to 25% stenosis. The mean signal intensity (SI) change of the six segments during adenosine
was 2.85% ± 0.33%. Middle, Patient (Pat. #2) with a 50% stenosis of the left anterior descending coronary
artery (LAD), a 90% stenosis in a large diagonal branch, and a 99% stenosis in the second marginal branch.
Although there is no obvious visible change of SI, the quantitative evaluation showed an SI decrease of 0.2%
to 7.6% in the segments related to the stenotic arteries. Right, Patient (Pat. #3) with a critical stenosis of
the right coronary artery (RCA). The SI decrease in the inferior segment during adenosine was 7.2%. In this
case, a signal drop can be visually evident (arrow). (Modified from Friedrich MG, Niendorf T, Schulz-Menger
J, Gross CM, Dietz R. Blood oxygen level-dependent magnetic resonance imaging in patients with stress-
induced angina. Circulation. 2003;108:2219–2223.)

T2-Prepared BOLD CMR in Patients evaluation of BOLD imaging on detecting perfusion defect in 105 sub-
Following the demonstration of marked improvement in image quality jects (25 patients, 20 healthy, and 60 suspected patients) at 3 T with
with T2-prepared bSSFP-based BOLD CMR, there have been several 2D T2-prepared bSSFP BOLD MR.80 This study first determined a
clinical studies that have aimed to validate this approach in patients.77–81 threshold for BOLD response using the derivation arm (25 patients
As in preclinical studies, this method was studied in patients at 1.5 T and 20 healthy) and subsequently used it to study 60 suspected patients
using FPP as the ground truth under adenosine stress in 41 patients with and compared the diagnostic performance against angiography. Results
suspected CAD.77 The study successfully demonstrated that T2-weighted showed the prospective evaluation of BOLD imaging yielded an accuracy
signal changes corresponding to pharmacologic stress are significantly of 84%, a sensitivity of 92%, and a specificity of 72% for detecting
different between segments with normal perfusion, nontransmural, and myocardial ischemia compared with QCA. Although the study showed
transmural perfusion deficits. ROC analysis showed an area under the BOLD CMR could achieve favorable accuracy for identifying the ana-
curve of 0.83 for the T2-prepared SSFP BOLD approach with regard tomic and functional significance of CAD in a clinical setting, a com-
to detection of inducible perfusion deficit. Another study from the parison between BOLD response and FPP-based MPRI revealed evidence
same group evaluated the BOLD response in relation to fractional of dissociation between oxygenation and perfusion (r = −0.26), which
flow reserve (FFR)78 cutoff of 0.8 to identify ischemic segments in 42 suggested that myocardial hypoperfusion does not correlate with deoxy-
patients with suspected CAD. Results showed significantly lower BOLD genation. This and other studies from the same group of investigators
response in the ischemic segments (FFR ≤ 0.8: 1.1 ± 0.2% vs. FFR demonstrating a dissociation between BOLD and perfusion are notable
> 0.8: 1.5 ± 0.2; P < .0001) with sensitivity and specificity of 88.2% observations but are discordant with preclinical studies validated at 1.5
and 89.5%, respectively. Although this study was promising, notably T using microspheres. Whether this discrepancy is related to imaging
the small BOLD response observed in human subjects, especially artifacts with bSSFP readouts at 3 T or in FPP approaches used in these
without disease, could present a limitation for T2-prepared BOLD studies, or associated with confounding effects of heart rates from insuf-
SSFP CMR at 1.5 T. ficient signal correction between rest and stress, or indeed the physiologic
With the preclinical studies showing marked improvement in BOLD truth, remains to be confirmed. Recently, T2-prepared BOLD CMR at
sensitivity at 3 T compared with 1.5 T, a significant number of clinical 3 T have also been successfully explored in patients with nonobstructive
BOLD CMR studies have been performed in the past 5 years and have coronary disease (syndrome X), hypertrophic and dilated cardiomy-
all been exclusively at 3 T. Jahnke et al. studied 50 patients with suspected opathies, patients with type 2 diabetes, aortic stenosis, and in those
CAD at 3 T using a free-breathing, T2-prepared gradient echo 3D CMR with CKD.82–87
with quantitative coronary angiography (QCA) serving as the ground
truth. The BOLD response is significantly correlated with the degree CP-BOLD CMR in Patients
of coronary stenosis (r = −0.65, P < .001). Additionally, BOLD response Two-dimensional CP-BOLD CMR has also been used in a feasibility
and myocardial perfusion reserve index (MPRI) from FPP were in study in nine patients suspected of CAD (Fig. 8.9).88 On the basis of
agreement (κ = 0.66) (Fig. 8.8).79 Another study carried out prospective thallium SPECT, segments were classified as healthy, mildly affected,
92 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A B C

D E F

FIG. 8.8  Representative cardiovascular magnetic resonance examples of blood-oxygen-level-dependent


(BOLD) and first-pass perfusion (FPP). Upper row, Subtracted BOLD image (A), stress and rest FPP (B), and
corresponding invasive angiogram of the left and right coronary artery (RCA) (C) in a patient with known
coronary artery disease and prior stenting of the RCA. Subtracted BOLD and FPP imaging show a stress-
induced deficit in the anterior/anterolateral myocardial segments (yellow arrows). Invasive coronary angiography
confirmed severe stenosis of the left anterior descending (LAD) coronary artery (blue arrow) (quantitative
coronary angiography 63%). Bottom row, Subtracted BOLD image (D), stress and rest FPP (E), and corre-
sponding invasive angiogram of the left and right coronary artery (F) in a patient with suspected coronary
artery disease. Subtracted BOLD and FPP imaging detected a stress-induced deficit in the inferoseptal/inferior
wall (yellow arrows). Invasive coronary angiography revealed a complete occlusion of the RCA (blue arrow)
(100%) with retrograde filling of the distal RCA via extensive collaterals. (Modified from Jahnke C, Gebker
R, Manka R, et al. Navigator-gated 3D blood oxygen level-dependent CMR at 3.0-T for detection of stress-
induced myocardial ischemic reactions. JACC Cardiovasc Imaging. 2010;3:375–384.)

or severely affected. Segmental bSSFP signal intensities at rest and segment are equally affected, which need not be the case because the
stress were measured, and the signal intensity ratio (stress/rest) was epicardial coronary artery supplying a given segment is not well defined
calculated. Statistical results showed that there are significant differ- and can vary among individuals and within a segment.18 Because it
ences in stress/rest values computed from healthy, mildly affected, overemphasizes the magnitude of intensity changes over the regional
and severely affected segments (P < .05). More recently this approach extent of the BOLD effect at rest and vasodilation, it may contrib-
has been studied in 37 CAD patients with FFR measurements as the ute to the reduced sensitivity and specificity reported by a number of
ground truth, which showed that in subjects with FFR <0.8, the sensi- studies.18,22,30,67 To date, several attempts have been made to identify new
tivity and specificity for detecting the presence of diminished vasodi- ways to visualize and evaluate the BOLD effect, including the simultane-
lator response in affected segments was 86% and 92%, respectively.89 ous acquisition of BOLD and cine wall motion assessment.90a
However, because of poor image quality, a large minority (~40%)
of segments had to be excluded. Improved CP-BOLD CMR strate- Detecting and Quantifying Pixel-Level BOLD Changes
gies such as those that can overcome flow/motion artifacts may help Under Vasodilator Stress With ARREAS
to overcome this limitation but remain to be studied in the clinical One of the early efforts to improve the visualization and quantification
setting.90 of myocardial BOLD, area-based biomarker for characterizing coro-
nary stenosis (ARREAS) aimed to provide pixel-level assessment of
VISUALIZATION AND QUANTIFICATION OF BOLD ischemia on the basis of a statistical model of myocardial intensity dis-
tribution.91 ARREAS relies on the combination of a statistically derived
EFFECTS VIA IMAGE PROCESSING threshold and connectivity analysis to identify the territories affected
Maximal BOLD CMR response from healthy myocardial segments is by the stenosis. It assumes that the myocardial intensity distribution
typically on the order of 15% to 20% and segments with disease are under rest conditions can be represented as a unimodal scaled distribu-
significantly lower. Without proper windowing, visualizing these BOLD tion (e.g., Student’s t). On the other hand, under stress conditions, the
signal changes is difficult. Manual windowing requires significant time distribution is the same but because of the increase in BOLD contrast
(several minutes), is subjective, and can introduce large intraobserver (attributed to vasodilation), a different mean is used. In the case of
and interobserver variability. Until recently, the evaluation of myocardial disease, the distribution becomes bimodal (one representing healthy
BOLD CMR relied on measuring the relative mean signal intensity myocardium, and the other, affected myocardium) but because the con-
changes in segments of interest under rest and pharmacologically induced trast change is within the deviation, these underlying distributions are
vasodilation. This paradigm assumes that all pixels within the entire not separable. Thus the ARREAS approach, with the aid of a Student’s
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 93

bSSFP BOLD FPP Thallium


1.20

1.15
Rest

Stress/rest
1.10

1.05

1.00
Stress

0.95
Severely affected Mildly affected Healthy
A B Myocardial segments
FIG. 8.9  Patient studies with two-dimensional balanced steady-state free precession (bSSFP) blood-oxygen-
level-dependent (BOLD) cardiovascular magnetic resonance at 1.5 T. Midventricular short-axis steady-state
free precession BOLD, first-pass perfusion (FPP), and thallium single-photon emission computed tomography
(SPECT) images obtained from a patient with 70% stenosis of the left anterior descending (LAD) coronary
artery at rest and stress (A). The windowing and leveling of images obtained at rest and stress are the same.
Myocardial signal in the rest BOLD, FPP, and SPECT images are relatively homogeneous. However, under
stress, the territory supplied by the LAD (larger arc subtended by arrows) does not increase in the BOLD
images as expected. This pattern of regional signal differences is also evident in the FPP and SPECT images.
Statistical results from the myocardial BOLD signal analysis showed that significant differences in stress/
rest values exist between healthy and affected regions (B). In comparison to healthy segments, the stress/
rest values of affected regions are lower, consistent with previous findings in animals that bSSFP signals
obtained under pharmacologic stress are significantly reduced in myocardial territories supplied by stenotic
arteries. (Modified from Dharmakumar R, Green JD, Flewitt J, et al. Blood oxygen-sensitive SSFP imaging
for probing the myocardial perfusion reserves of patients with coronary artery disease: a feasibility study.
J Cardiovasc Magn Reson. 2008;10[suppl 1]:A101.)

ARREAS MICROSPHERE FPP LGE

3.5
3
2.5
2
1.5
1
0.5
sMFR
FIG. 8.10  Relation between myocardial blood-oxygen-level-dependent (BOLD) image processed using area-
based biomarker for characterizing coronary stenosis (ARREAS), segmental myocardial flow ratio (sMFR)
bull’s eye plot, first-pass perfusion (FPP), and late gadolinium enhancement (LGE) images. FPP image obtained
under adenosine stress with critical (70%) left circumflex coronary artery stenosis and the corresponding
BOLD image (processed with the ARREAS) are shown for comparison. LGE image confirms the absence of
any infarction. (Modified from Tsaftaris SA, Tang R, Zhou X, et al. Detecting clinically significant coronary
stenosis by inducing changes in microcirculatory oxygenation: an experimental study in canines using oxygen-
sensitive magnetic resonance imaging. J Magn Reson Imaging. 2012;35:1338–1348.)

t distribution, fitted on myocardial intensities at rest, establishes an This was demonstrated in canines using microsphere as gold
intensity threshold. This threshold is used to find, in rest and stress standard—an example of visualization with ARREAS and compari-
images, pixels that have intensity higher than the threshold (termed, son to microsphere analysis and FPP is shown in Fig. 8.10. This study
for simplicity, “affected” pixels). The ratio of affected pixels over the showed that ischemic extent is exponentially related to microsphere
area of the myocardium is termed as affected fraction, the ratio of which flow and increases with the severity of surgically implemented coronary
defines a biomarker called ischemic extent that aims to characterize stenosis. This indicated that the extent of epicardial narrowing likely
how many more pixels can be identified as hypoperfused following affects the supplied territories in a graded fashion; that is, with a greater
vasodilatory stress. narrowing of a given epicardial coronary artery, a greater fraction of
94 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

the corresponding supply territory experiences an alteration in oxygen- specifically one class learning, and does not rely on any user-defined
ation. It was shown that ARREAS could significantly increase sensitivity parameters (e.g., a threshold, as is necessary with the S/D biomarker).75
and specificity for detecting BOLD signal changes compared with the The algorithm also provides a probabilistic interpretation as to the con-
conventional (segmental) average intensity approach. fidence of the label assignments. The algorithm was evaluated extensively
Because CP-BOLD provides several images throughout the cardiac using simulated and real data from canine experiments. In canines, they
cycle for analysis, ARREAS despite being a single image approach, was showed that ischemic extent (measured as the fraction of segments
evaluated both on end-systolic and end-diastolic images. There were that are anomalous) was very well correlated (0.84, P = .004) with
mild differences between the findings when systolic or diastolic images area at risk.
were used. These could have been from differences in the myocardial
surface available for processing between end-diastolic and end-systolic Need for Better Myocardial Registration and Segmentation
images or from potential differences in underlying physiology between Currently, the majority of BOLD CMR analysis assumes the precise
systole and diastole. delineation of the myocardium (myocardial segmentation), which is
Although ARREAS processes images within the cycle individually, largely performed in a manual fashion; this is time consuming and can
ischemic extent measurements from multiple cardiac phases could have introduce operator bias. Hence there is a need to develop automated
been fused (combined) post-hoc to obtain even higher specificity and approaches that can segment and register the myocardium in the pres-
sensitivity. This idea has been exploited in a more formal framework ence of BOLD contrast.
in the context of rest CP-BOLD CMR and is discussed below. Recent studies showed that the presence of BOLD contrast compli-
cates the process of developing such automated algorithms.95,96 Because
Automating the Detection of Area-at-Risk Under Rest of BOLD effects, temporal (for CP-BOLD) and spatial variations (from
With CP-BOLD CMR disease) in signal intensity collectively violate the principle of appear-
As discussed earlier, ischemic territories of the myocardium can be ance invariance that is commonly assumed. One study considered that
identified on the basis of signal differences in CP-BOLD images. In the one of the principal limitations is the lack of appropriate features that
preclinical validation of this concept, a segmental biomarker (S/D) was can be used to describe the myocardium within a classical treatment
derived on the basis of end-systolic and diastolic images to examine of myocardial segmentation as a supervised learning classification
ratios of average signal intensity within a segment at systole and diastole. problem.95 Using discriminatory dictionaries, the study showed that
It was found that S/D can be used to identify ischemic territories in algorithms with close to 70% myocardial segmentation accuracy could
early ischemia animal model and this S/D biomarker can be visualized be achieved,95 with comparable performance to state-of-the-art algo-
in typical bull’s eye plots showing color coded the values of S/D. The rithms for standard cine MRI myocardial segmentation.97
authors also noted that even at rest, and before any coronary occlusion, Supervised machine-learning algorithms require training sets with
S/D between territories in the heart vary, a fact that they originally annotation (in this case myocardial delineations), which are often hard
attributed to potential phasic variations in BOLD signal intensity. Sub- to obtain in large numbers. In a follow-up work, an unsupervised myo-
sequent work92 later on confirmed these findings. Using a signal processing cardial segmentation algorithm for cardiac BOLD was developed.98 It
technique referred to as shift-invariant dictionary learning they were relies on the premise that the myocardium appears and moves differently
able to show that time series of myocardial BOLD signal intensity (where with respect to surrounding organs (background). Thus the algorithm
time is cardiac phase), derived from segmental averages in the data, proceeds by first obtaining a crude segmentation of the myocardium, by
can be uniquely characterized by projecting to an optimal template. measuring the amount of motion, and then learns a dictionary-driven
They called this effect the characteristic CP-BOLD curve that forms a representation for the other areas using both intensity and motion
phenotype of how BOLD signal intensity varies with cardiac phase. information. A statistical model is built on these representations, and
They also found statistically that phasic variation among territories inference algorithms are used to find parts in the images that do not
exist in canines. fit this model. The final outcome is a segmentation of the myocardium
Building upon these observations, Rusu and Tsaftaris93 devised that parallels the performance of supervised algorithms when evalu-
an algorithm that can provide a probabilistic assessment to ischemia ated in a dataset of 40 acquisitions with and without BOLD contrast
likelihood for CP-BOLD at rest. Their algorithm takes data from a and disease.
single individual and, without any prior knowledge, outputs a map of Because the heart is moving across the cardiac cycle, finding cor-
ischemia likelihood. Bevilacqua et al.94 extended this work to extract respondences between different phases of a CP-BOLD acquisition to
both signal intensity time series and time series that relate to myocar- establish intensity time series necessitates accurate nonlinear myocardial
dial function (e.g., wall thickness) from very fine radial segments of registration. This process is again challenged by the presence of BOLD
the myocardium. These time series are given to an iterative algorithm contrast and the violation of the appearance invariance assumption—
aiming to identify those time series originating from ischemic territories common in many intensity-driven registration algorithms. Another
by considering that they do not fit a normal model. The normal model study devised a new registration algorithm that builds on the previous
is built upon the observations that BOLD time series should follow the work to derive new features and a new metric to compare those features
BOLD characteristic curve and that myocardial function time series do for the purpose of matching correspondences within a nonlinear reg-
have distinct characteristics in systole and diastole. The normal model istration framework.96 This study, also evaluated in preclinical data,
is algorithmically defined as coupled shift invariant dictionaries, and showed that the algorithm outperformed all other intensity-driven
the algorithm proceeds by repeating the following steps: fit the model; algorithms.
project the time series on the model; find the time series that do not
fit the model (and hence are anomalous); isolate the time series that Evolving Importance and Need for Pixel-Level
fit the model and repeat the process. The algorithm converges after a Detection of BOLD Contrast
few iterations to obtain a label for each time series whether it is normal Although ARREAS was developed and tested on the basis of bSSFP
(originates from territories remote to ischemia) or anomalous (origi- BOLD images, extensions of the approach to T2*- or T2-weighted BOLD
nates from an ischemic territory). To find the time series that do not CMR could be straightforward. In fact, ARREAS established the potential
fit the model, the algorithm uses concepts from machine learning and of pixel-level analysis of the BOLD effect in the heart. Given recent
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 95

advances in new sequences, which have superior SNR and resolution, higher gradient-strength clinical scanners may permit shorter TRs that
it is natural to consider analysis techniques that shy away from segmental can facilitate improvements in image quality to allow for more reli-
notions of analysis. With the advent of better registration and segmen- able acquisition of BOLD CMR with bSSFP readouts well within the
tation algorithms and as our understanding of myocardial oxygenation next decade.
improves, pixel-level assessment of territories of myocardium at risk Although field-dependent (B0 and B1) inhomogeneities may be
would be possible in a completely automated fashion. This would enable overcome, the image artifacts from unpredictable cardiac motion during
precise determination of “area at risk” and transmural distribution of stress, leading to significant deterioration of BOLD image quality, is a
perfusion defects without any contrast. difficult problem. Yang et al.99 have shown that this difficulty may be
Furthermore, the approach of Bevilacqua et al.,94 presented previ- overcome by exploiting the long-lasting effects of regadenoson. Using
13
ously, has distinct benefits toward the quest to the pixel level. Regression- N-ammonia PET as the validation standard, they demonstrated in a
based approaches, which adopt ideas from the general linear models hybrid PET/CMR system that stress BOLD CMR performed at ~10
(GLMs) common in functional MRI (fMRI) to analyze, for example, minutes can markedly improve the reliability for detecting myocardial
BOLD data from breathing exercise protocols, require predefined regres- hyperemia in canines; and that myocardial perfusion reserve (MPR)
sors. Instead, this approach has the benefit in that it can identify auto- remains significantly greater than 2-fold (a meaningful hyperemic state
matically all possible regressors (similar to how an independent for ischemic testing) following regadenoson injection. Their studies
component analysis decomposition would). Thus it is able to character- showed that myocardial BOLD images acquired at 10 minutes following
ize both effects and confounders in a principled fashion, paving the regadenoson were free of image artifacts typically observed in images
way to completely automated assessment of area at risk. In addition, acquired at 2 minutes following regadenoson. MPR values at 10 minutes
within the same framework, spatial constraints can be considered without following regadenoson were significantly higher than 2-fold and were
the need of smoothing that is typical of GLM approaches. Furthermore strongly correlated with the myocardial BOLD effect (Fig. 8.11). These
it can consider time series as we saw from different origins, for example, data suggest that delayed BOLD acquisition following regadenoson
BOLD contrast and myocardial function. All these factors should facili- administration can be a practical strategy for increasing the reliability
tate pixel-level analysis and increase robustness to noise (at the pixel of cardiac BOLD CMR for cardiac stress testing. More studies are needed
level the benefits of noise reduction because of segmental averaging to confirm these findings.
are lost). This algorithm can be readily applied to cases where stress is An alternate strategy to increase the reliability of BOLD CMR is
present either via pharmacologic agents or even within a repeated design to rely on multiple measurements, which in a similar fashion to brain
(e.g., with hypercapnia), which is expected to provide greater accuracy activation may provide increased confidence with the detection of
and sensitivity for BOLD CMR. underlying pathology. However, until recently, repeat stimulation of
the coronary vasculature did not seem possible, especially with phar-
TOWARD A MORE RELIABLE STRESS BOLD macologic vasodilators such as adenosine. However, the capability to
repeatedly stimulate the coronaries with a benign stimulus such as CO2,
CMR EXAMINATION as indicated earlier, opens the possibility for repeatedly assessing the
Most clinical studies using BOLD CMR for detection of ischemic and relationship between a known stimulus and a system response through
nonischemic heart disease at the present time have converged on the statistical means. Although many studies remain to be performed to
use of 3 T CMR systems to ensure greater BOLD contrast is available investigate whether or not this strategy will be beneficial, the capabil-
compared with at 1.5 T. However, BOLD CMR at 3 T still requires ity to repeatedly vasodilate the coronaries and study their response
further technical advances to overcome several confounders, both tech- provides an exciting new opportunity to increase the reliability of
nical and physiologic, which can otherwise lead one to make erroneous BOLD CMR.
conclusions. This is naturally evidenced by the marked variability in
BOLD data observed in recently published clinical data. Currently, the
key confounders for BOLD CMR at 3 T are (1) B0 field inhomogeneities,
FUTURE DIRECTIONS OF MYOCARDIAL BOLD CMR
particularly from the heart–lung interface, which lead to inadvertent Although recent advances in BOLD CMR technology are promising,
signal loss within the myocardium; (2) B1 field inhomogeneities, which the newer CMR techniques (such as fast, free-breathing 3D methods
can lead to signal variations in the myocardium that have little or no that are insensitive to bulk B0 inhomogeneities and B1 variations at 3
contributions from the BOLD effect; and (3) complex motion (heart- T), novel vasodilation methods (late effects of regadenoson, targeted
rate variations, changes in cardiac contractility, and variations in blood increases in PaCO2, and breathing maneuvers to alter arterial CO2),
flow) specifically under evolving state of pharmacologic stress that make and rest imaging to identify area at risk using CP-BOLD need to be
it difficult to limit acquisitions to the typical quiescent phase of mid validated and extended through prospective clinical studies. Furthermore
diastole. Much of the ongoing technical development in data acquisition with improvements in imaging methods, image-processing approaches
is centered on overcoming these key obstacles. having the capability to dramatically improve the visualization and
3 T BOLD CMR examinations at the present time primarily use a automatic quantification to estimate the myocardial territories at risk
standard T2-prepration scheme followed by bSSFP readout. However, of ischemia accurately via pixel-level detection also needs to be extended
despite the high SNR efficiency afforded by bSSFP readouts at 3 T, to the clinical realm. Moreover, there is a growing appreciation that
both the preparation and readout scheme are highly sensitive to B0 ischemic heart disease and CAD are not equivalent, because microvas-
and B1 inhomogeneities. Recent studies have shown that this obsta- cular dysfunction in the absence of obstructive coronary disease can
cle can be overcome through the use of a T2 preparation employing be a key source of ischemia. To date, CMR methods have not fully
adiabatic radiofrequency (RF) pulses combined with spoiled gradient examined microvascular dysfunction and this gap needs to be bridged
echo (fast low angle shot [FLASH]) readouts, albeit at the cost of a through animal studies (for validation) and prospective clinical studies
modestly reduced SNR and loss of capability to perform CP-BOLD with advanced image acquisition and image-processing strategies. Finally,
CMR. Further improvements in shimming technology are expected BOLD CMR methods, even if they are quantitative (e.g., via mapping
to be critically important before the BOLD CMR can be performed methods), have yet to be inspired toward yielding quantifiable MBF or
robustly using bSSFP imaging. However, the increasing availability of oxygenation. A BOLD CMR method that enables quantification of MBF
96 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

1.6 (0.6,3.8)
4
M yocar dial T2 (ms)

20 80

variability at stress
Myocardial BOLD
1.0 0.9 (0.6,1.2)

0
A Re s t 2 min 10 min

Rest Stress 0 Rest Stress 20

(mL/min/g)

T2 (ms)
MBF
4 80
B D
Myocardial
4 45
MBF (mL/g/min)

PET 4.5 R = 0.7, P < .05


BOLD
3 42 4.0

PET MPR
T2 (ms)

3.5
2 39
3.0
1 36 2.5
0 33 2.0
Rest 10 min Rest 10 min 1.02 1.08 1.13 1.18
C E F Myocardial BOLD response
FIG. 8.11  Myocardial blood-oxygen-level-dependent (BOLD) variability at stress relative to rest. Box plot of
myocardial BOLD variability and representative images at rest and from 2 and 10 minutes post regadenoson
are shown. Large myocardial BOLD variability is observed at 2 minutes post regadenoson compared with
rest and is markedly reduced at 10 minutes post regadenoson administration. 13N-ammonia positron emission
tomography (PET) myocardial blood flow (MBF) and BOLD. Both PET and BOLD images showed significant
increase in MBF and BOLD response, at 10 minutes postregadenoson administration compared with rest
(A–E). Results from regression analysis showed good correlation between PET myocardial perfusion reserve
(MPR) (MBF[stress)/MBF[rest]) and myocardial BOLD response (R = 0.7, P < .05; panel [F]). MPR, Myocardial
perfusion reserve. (Modified from Yang HJ, Dey D, Sykes J, et al. Towards reliable myocardial blood-oxygen-
level-dependent (BOLD) CMR using late effects of regadenoson with simultaneous 13N-ammonia PET valida-
tion in a whole-body hybrid PET/MR system. J Cardiovasc Magn Reson. 2016;18[suppl 1]:O19.)

or oxygenation is expected to be of paramount importance, because it


ACKNOWLEDGMENT
can enable the detection of myocardial oxygenation changes in patients
with all forms of CAD, especially those with balanced ischemia second- This work was supported in part by National Institutes of Health R01
ary to multivessel CAD. Successful adoption of recent advances through HL91989.
clinical studies and additional technical improvements can propel myo-
cardial BOLD CMR toward becoming a powerful noninvasive diagnostic
method in the early detection and posttreatment monitoring of ischemic
REFERENCES
heart disease. A full reference list is available online at ExpertConsult.com
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 96.e1

REFERENCES 20. Beache GM, Herzka DA, Boxerman JL, et al. Attenuated myocardial
vasodilator response in patients with hypertensive hypertrophy revealed
1. Brown M, Marshall DR, Sobel BE, Bergmann SR. Delineation of by oxygenation-dependent magnetic resonance imaging. Circulation.
myocardial oxygen utilization with carbon-11-labeled acetate. 2001;104:1214–1217.
Circulation. 1987;76:687–696. 21. Wright KB, Klocke FJ, Deshpande VS, et al. Assessment of regional
2. Brown MA, Myears DW, Bergmann SR. Noninvasive assessment of differences in myocardial blood flow using t2-weighted 3D BOLD
canine myocardial oxidative metabolism with carbon-11 acetate and imaging. Magn Reson Med. 2001;46:573–578.
positron emission tomography. J Am Coll Cardiol. 1988;12:1054–1063. 22. Foltz WD, Huang H, Fort S, Wright GA. Vasodilator response assessment
3. Brown MA, Myears DW, Bergmann SR. Validity of estimates of in porcine myocardium with magnetic resonance relaxometry.
myocardial oxidative metabolism with carbon-11 acetate and positron Circulation. 2002;106:2714–2719.
emission tomography despite altered patterns of substrate utilization. J 23. Foltz WD, Al-Kwifi O, Sussman MS, Stainsby JA, Wright GA. Optimized
Nucl Med. 1989;30:187–193. spiral imaging for measurement of myocardial T2 relaxation. Magn
4. Buxton DB, Nienaber CA, Luxen A, et al. Noninvasive quantitation of Reson Med. 2003;49:1089–1097.
regional myocardial oxygen consumption in vivo with [1-11c]acetate 24. Zheng J, Wang J, Rowold FE, Gropler RJ, Woodard PK. Relationship of
and dynamic positron emission tomography. Circulation. 1989;79: apparent myocardial T2 and oxygenation: towards quantification of
134–142. myocardial oxygen extraction fraction. J Magn Reson Imaging. 2004;20:
5. Henes CG, Bergmann SR, Walsh MN, Sobel BE, Geltman EM. 233–241.
Assessment of myocardial oxidative metabolic reserve with positron 25. Zheng J, Wang J, Nolte M, Li D, Gropler RJ, Woodard PK. Dynamic
emission tomography and carbon-11 acetate. J Nucl Med. estimation of the myocardial oxygen extraction ratio during
1989;30:1489–1499. dipyridamole stress by MRI: a preliminary study in canines. Magn Reson
6. Gropler RJ, Siegel BA, Sampathkumaran K, et al. Dependence of Med. 2004;51:718–726.
recovery of contractile function on maintenance of oxidative 26. Zhang H, Gropler RJ, Li D, Zheng J. Assessment of myocardial oxygen
metabolism after myocardial infarction. J Am Coll Cardiol. 1992;19: extraction fraction and perfusion reserve with BOLD imaging in a
989–997. canine model with coronary artery stenosis. J Magn Reson Imaging.
7. Gropler RJ, Geltman EM, Sampathkumaran K, et al. Functional recovery 2007;26:72–79.
after coronary revascularization for chronic coronary artery disease is 27. McCommis KS, Goldstein TA, Zhang H, Misselwitz B, Gropler RJ,
dependent on maintenance of oxidative metabolism. J Am Coll Cardiol. Zheng J. Quantification of myocardial blood volume during
1992;20:569–577. dipyridamole and doubtamine stress: a perfusion CMR study. J
8. Gropler RJ, Geltman EM, Sampathkumaran K, et al. Comparison of Cardiovasc Magn Reson. 2007;9:785–792.
carbon-11-acetate with fluorine-18-fluorodeoxyglucose for delineating 28. McCommis KS, Zhang H, Herrero P, Gropler RJ, Zheng J. Feasibility
viable myocardium by positron emission tomography. J Am Coll Cardiol. study of myocardial perfusion and oxygenation by noncontrast MRI:
1993;22:1587–1597. comparison with PET study in a canine model. Magn Reson Imaging.
9. Wendland MF, Saeed M, Lauerma K, de Crespigny A, Moseley ME, 2008;26:11–19.
Higgins CB. Endogenous susceptibility contrast in myocardium during 29. Fieno DS, Shea SM, Li Y, Harris KR, Finn JP, Li D. Myocardial perfusion
apnea measured using gradient recalled echo planar imaging. Magn imaging based on the blood oxygen level-dependent effect using
Reson Med. 1993;29:273–276. T2-prepared steady-state free-precession magnetic resonance imaging.
10. Atalay MK, Forder JR, Chacko VP, Kawamoto S, Zerhouni EA. Circulation. 2004;110:1284–1290.
Oxygenation in the rabbit myocardium: assessment with susceptibility- 30. Shea SM, Fieno DS, Schirf BE, et al. T2-prepared steady-state free
dependent MR imaging. Radiology. 1993;189:759–764. precession blood oxygen level-dependent MR imaging of myocardial
11. Balaban RS, Taylor JF, Turner R. Effect of cardiac flow on gradient perfusion in a dog stenosis model. Radiology. 2005;236:503–509.
recalled echo images of the canine heart. NMR Biomed. 1994;7:89–95. 31. Wilke N, Simm C, Zhang J, et al. Contrast-enhanced first pass
12. Stillman AE, Wilke N, Jerosch-Herold M. BOLD contrast of the heart myocardial perfusion imaging: correlation between myocardial blood
during occlusion and reperfusion. SMR 1st Meeting; 1994. flow in dogs at rest and during hyperemia. Magn Reson Med. 1993;29:
13. Atalay MK, Reeder SB, Zerhouni EA, Forder JR. Blood oxygenation 485–497.
dependence of T1 and T2 in the isolated, perfused rabbit heart at 4.7T. 32. Klocke FJ. Measurements of coronary flow reserve: defining
Magn Reson Med. 1995;34:623–627. pathophysiology versus making decisions about patient care. Circulation.
14. Li D, Dhawale P, Rubin PJ, Haacke EM, Gropler RJ. Myocardial signal 1987;76:1183–1189.
response to dipyridamole and dobutamine: demonstration of the BOLD 33. Foltz WD, Merchant N, Downar E, Stainsby JA, Wright GA. Coronary
effect using a double-echo gradient-echo sequence. Magn Reson Med. venous oximetry using MRI. Magn Reson Med. 1999;42:837–848.
1996;36:16–20. 34. Wendland MF, Saeed M, Masui T, Derugin N, Higgins CB. First pass of
15. Niemi P, Poncelet BP, Kwong KK, et al. Myocardial intensity changes an MR susceptibility contrast agent through normal and ischemic heart:
associated with flow stimulation in blood oxygenation sensitive gradient-recalled echo-planar imaging. J Magn Reson Imaging. 1993;3:
magnetic resonance imaging. Magn Reson Med. 1996;36:78–82. 755–760.
16. Li D, Oellerich WF, Beck G, Gropler RJ. Assessment of myocardial 35. Edelman RR, Li W. Contrast-enhanced echo-planar MR imaging of
response to pharmacologic interventions using an improved MR myocardial perfusion: preliminary study in humans. Radiology. 1994;
imaging technique to estimate T2 values. AJR Am J Roentgenol. 190:771–777.
1999;172:141–145. 36. Wilke N, Jerosch-Herold M, Wang Y, et al. Myocardial perfusion reserve:
17. Wacker CM, Bock M, Hartlep AW, et al. Changes in myocardial assessment with multisection, quantitative, first-pass MR imaging.
oxygenation and perfusion under pharmacological stress with Radiology. 1997;204:373–384.
dipyridamole: Assessment using T*2 and T1 measurements. Magn Reson 37. United States Renal Data System (USRDS). Annual Data Report:
Med. 1999;41:686–695. Epidemiology of Kidney Disease in the United States. Ann Arbor, MI:
18. Friedrich MG, Niendorf T, Schulz-Menger J, Gross CM, Dietz R. Blood USRDS; 2014.
oxygen level-dependent magnetic resonance imaging in patients with 38. Errante Y, Cirimele V, Mallio CA, Di Lazzaro V, Zobel BB, Quattrocchi
stress-induced angina. Circulation. 2003;108:2219–2223. CC. Progressive increase of T1 signal intensity of the dentate nucleus on
19. Wacker CM, Hartlep AW, Pfleger S, Schad LR, Ertl G, Bauer WR. unenhanced magnetic resonance images is associated with cumulative
Susceptibility-sensitive magnetic resonance imaging detects human doses of intravenously administered gadodiamide in patients with
myocardium supplied by a stenotic coronary artery without a contrast normal renal function, suggesting dechelation. Invest Radiol. 2014;49:
agent. J Am Coll Cardiol. 2003;41:834–840. 685–690.
96.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

39. McDonald RJ, McDonald JS, Kallmes DF, et al. Intracranial gadolinium 61. Massie BM, Schwartz GG, Garcia J, Wisneski JA, Weiner MW, Owens T.
deposition after contrast-enhanced MR imaging. Radiology. Myocardial metabolism during increased work states in the porcine left
2015;275:772–782. ventricle in vivo. Circ Res. 1994;74:64–73.
40. Kanda T, Fukusato T, Matsuda M, et al. Gadolinium-based contrast 62. Yang HJ, Yumul R, Tang R, et al. Assessment of myocardial reactivity to
agent accumulates in the brain even in subjects without severe renal controlled hypercapnia with free-breathing T2-prepared cardiac blood
dysfunction: evaluation of autopsy brain specimens with inductively oxygen level-dependent MR imaging. Radiology. 2014;272:397–406.
coupled plasma mass spectroscopy. Radiology. 2015;276:228–232. 63. Guensch DP, Fischer K, Flewitt JA, et al. Breathing manoeuvre-
41. Messroghli DR, Walters K, Plein S, et al. Myocardial T1 mapping: dependent changes in myocardial oxygenation in healthy humans. Eur
application to patients with acute and chronic myocardial infarction. Heart J Cardiovasc Imaging. 2014;15:409–414.
Magn Reson Med. 2007;58:34–40. 64. Dharmakumar R, Hong J, Brittain JH, Plewes DB, Wright GA.
42. Pauling L, Coryell CD. The magnetic properties and structure of Oxygen-sensitive contrast in blood for steady-state free precession
hemoglobin, oxyhemoglobin and carbonmonoxyhemoglobin. Proc Natl imaging. Magn Reson Med. 2005;53:574–583.
Acad Sci USA. 1936;22:210–216. 65. Brittain JH, Reeder SB, Shimakawa A. Non-contrast-enhanced
43. Thulborn KR, Waterton JC, Matthews PM, Radda GK. Oxygenation angiography at 3T using SSFP and “Dixon” fat-water separation. In:
dependence of the transverse relaxation time of water protons in whole 2004 International Society for Magnetic Resonance in Medicine Conference
blood at high field. Biochim Biophys Acta. 1982;714:265–270. Proceedings; 2004; Kyoto, Japan.
44. Li D, Wang Y, Waight DJ. Blood oxygen saturation assessment in vivo 66. Vöhringer M, Flewitt JA, Green JD, et al. Oxygenation-sensitive CMR
using T2* estimation. Magn Reson Med. 1998;39:685–690. for assessing vasodilator-induced changes of myocardial oxygenation. J
45. Wright GA, Hu BS, Macovski A. 1991 i.I. Rabi award. Estimating oxygen Cardiovasc Magn Reson. 2010;12:20.
saturation of blood in vivo with MR imaging at 1.5T. J Magn Reson 67. Dharmakumar R, Arumana JM, Tang R, Harris K, Zhang Z, Li D.
Imaging. 1991;1:275–283. Assessment of regional myocardial oxygenation changes in the presence
46. Brittain JH, Olcott EW, Szuba A, et al. Three-dimensional flow- of coronary artery stenosis with balanced SSFP imaging at 3.0T: theory
independent peripheral angiography. Magn Reson Med. 1997;38: and experimental evaluation in canines. J Magn Reson Imaging.
343–354. 2008;27:1037–1045.
47. Li KC, Dalman RL, Ch’en IY, et al. Chronic mesenteric ischemia: use of 68. Wu EX, Tang H, Wong KK, Wang J. Mapping cyclic change of regional
in vivo MR imaging measurements of blood oxygen saturation in the myocardial blood volume using steady-state susceptibility effect of iron
superior mesenteric vein for diagnosis. Radiology. 1997;204:71–77. oxide nanoparticles. J Magn Reson Imaging. 2004;19:50–58.
48. Nield LE, Qi XL, Valsangiacomo ER, et al. In vivo MRI measurement of 69. Wansapura J, Gottliebson W, Crotty E, Fleck R. Cyclic variation of T1 in
blood oxygen saturation in children with congenital heart disease. the myocardium at 3T. Magn Reson Imaging. 2006;24:889–893.
Pediatr Radiol. 2005;35:179–185. 70. Bai XJ, Iwamoto T, Williams AG Jr, Fan WL, Downey HF. Coronary
49. Kaul S, Jayaweera AR. Coronary and myocardial blood volumes: pressure-flow autoregulation protects myocardium from pressure-
noninvasive tools to assess the coronary microcirculation? Circulation. induced changes in oxygen consumption. Am J Physiol. 1994;266:
1997;96:719–724. H2359–H2368.
50. Bauer WR, Nadler W, Bock M, et al. Theory of the BOLD effect in the 71. Klocke FJ, Li D. Testing coronary flow reserve without a provocative
capillary region: an analytical approach for the determination of T2 in stress. A “BOLD” approach. J Am Coll Cardiol. 2003;41:841–842.
the capillary network of myocardium. Magn Reson Med. 1999;41:51–62. 72. Wei K, Tong KL, Belcik T, et al. Detection of coronary stenoses at rest
51. Chien D, Levin DL, Anderson CM. Mr gradient echo imaging of with myocardial contrast echocardiography. Circulation. 2005;112:
intravascular blood oxygenation: T2* determination in the presence of 1154–1160.
flow. Magn Reson Med. 1994;32:540–545. 73. Wei K, Le E, Jayaweera AR, Bin JP, Goodman NC, Kaul S. Detection of
52. Kim SG, Ugurbil K. Comparison of blood oxygenation and cerebral noncritical coronary stenosis at rest without recourse to exercise or
blood flow effects in fMRI: estimation of relative oxygen consumption pharmacological stress. Circulation. 2002;105:218–223.
change. Magn Reson Med. 1997;38:59–65. 74. Lindner JR, Skyba DM, Goodman NC, Jayaweera AR, Kaul S. Changes
53. Kennan RP, Scanley BE, Gore JC. Physiologic basis for BOLD MR signal in myocardial blood volume with graded coronary stenosis. Am J
changes because of hypoxia/hyperoxia: separation of blood volume and Physiol. 1997;272:H567–H575.
magnetic susceptibility effects. Magn Reson Med. 1997;37:953–956. 75. Tsaftaris SA, Zhou X, Tang R, Li D, Dharmakumar R. Detecting
54. Boxerman JL, Hamberg LM, Rosen BR, Weisskoff RM. MR contrast due myocardial ischemia at rest with cardiac phase-resolved blood oxygen
to intravascular magnetic susceptibility perturbations. Magn Reson Med. level-dependent cardiovascular magnetic resonance. Circ Cardiovasc
1995;34:555–566. Imaging. 2013;6:311–319.
55. Bauer WR, Nadler W, Bock M, et al. The relationship between the 76. Manka R, Paetsch I, Schnackenburg B, Gebker R, Fleck E, Jahnke C.
BOLD-induced T(2) and T(2)(*): a theoretical approach for the Bold cardiovascular magnetic resonance at 3.0 Tesla in myocardial
vasculature of myocardium. Magn Reson Med. 1999;42:1004–1010. ischemia. J Cardiovasc Magn Reson. 2010;12:54.
56. Belliveau JW, Kennedy DN Jr, McKinstry RC, et al. Functional mapping 77. Bernhardt P, Manzke R, Bornstedt A, et al. Blood oxygen level-
of the human visual cortex by magnetic resonance imaging. Science. dependent magnetic resonance imaging using T2-prepared steady-state
1991;254:716–719. free-precession imaging in comparison to contrast-enhanced myocardial
57. Kwong KK, Belliveau JW, Chesler DA, et al. Dynamic magnetic perfusion imaging. Int J Cardiol. 2011;147:416–419.
resonance imaging of human brain activity during primary sensory 78. Walcher T, Manzke R, Hombach V, Rottbauer W, Wohrle J, Bernhardt P.
stimulation. Proc Natl Acad Sci USA. 1992;89:5675–5679. Myocardial perfusion reserve assessed by T2-prepared steady-state free
58. Ogawa S, Menon RS, Tank DW, et al. Functional brain mapping by precession blood oxygen level-dependent magnetic resonance imaging
blood oxygenation level-dependent contrast magnetic resonance in comparison to fractional flow reserve. Circ Cardiovasc Imaging.
imaging. A comparison of signal characteristics with a biophysical 2012;5:580–586.
model. Biophys J. 1993;64:803–812. 79. Jahnke C, Gebker R, Manka R, Schnackenburg B, Fleck E, Paetsch I.
59. Lai S, Hopkins AL, Haacke EM, et al. Identification of vascular Navigator-gated 3D blood oxygen level-dependent CMR at 3.0-T for
structures as a major source of signal contrast in high resolution 2D and detection of stress-induced myocardial ischemic reactions. JACC
3D functional activation imaging of the motor cortex at 1.5T: Cardiovasc Imaging. 2010;3:375–384.
preliminary results. Magn Reson Med. 1993;30:387–392. 80. Arnold JR, Karamitsos TD, Bhamra-Ariza P, et al. Myocardial
60. McGuinness ME, Talbert RL. Pharmacologic stress testing: experience oxygenation in coronary artery disease: insights from blood oxygen
with dipyridamole, adenosine, and dobutamine. Am J Hosp Pharm. level-dependent magnetic resonance imaging at 3 Tesla. J Am Coll
1994;51:328–346, quiz 404–405. Cardiol. 2012;59:1954–1964.
CHAPTER 8  Cardiovascular Magnetic Resonance Assessment of Myocardial Oxygenation 96.e3

81. Karamitsos TD, Leccisotti L, Arnold JR, et al. Relationship between 91. Tsaftaris SA, Tang R, Zhou X, Li D, Dharmakumar R. Ischemic extent as
regional myocardial oxygenation and perfusion in patients with a biomarker for characterizing severity of coronary artery stenosis
coronary artery disease: insights from cardiovascular magnetic with blood oxygen-sensitive MRI. J Magn Reson Imaging. 2012;35:
resonance and positron emission tomography. Circ Cardiovasc Imaging. 1338–1348.
2010;3:32–40. 92. Rusu C, Morisi R, Boschetto D, Dharmakumar R, Tsaftaris SA. Synthetic
82. Karamitsos TD, Arnold JR, Pegg TJ, et al. Patients with syndrome x have generation of myocardial blood-oxygen-level-dependent MRI time series
normal transmural myocardial perfusion and oxygenation: a 3T via structural sparse decomposition modeling. IEEE Trans Med Imaging.
cardiovascular magnetic resonance imaging study. Circ Cardiovasc 2014;33:1422–1433.
Imaging. 2012;5:194–200. 93. Rusu C, Tsaftaris SA. Structured dictionaries for ischemia estimation in
83. Karamitsos TD, Dass S, Suttie J, et al. Blunted myocardial oxygenation cardiac BOLD MRI at rest. Med Image Comput Comput Assist Interv.
response during vasodilator stress in patients with hypertrophic 2014;17:562–569.
cardiomyopathy. J Am Coll Cardiol. 2013;61:1169–1176. 94. Bevilacqua M, Dharmakumar R, Tsaftaris SA. Dictionary-driven
84. Levelt E, Rodgers CT, Clarke WT, et al. Cardiac energetics, oxygenation, ischemia detection from cardiac phase-resolved myocardial BOLD MRI
and perfusion during increased workload in patients with type 2 at rest. IEEE Trans Med Imaging. 2016;35:282–293.
diabetes mellitus. Eur Heart J. 2015. 95. Mukhopadhyay A, Oksuz I, Bevilacqua M, Dharmakumar R, Tsaftaris
85. Mahmod M, Francis JM, Pal N, et al. Myocardial perfusion and SA. Data-driven feature learning for myocardial segmentation of
oxygenation are impaired during stress in severe aortic stenosis and CP-BOLD-MRI. In: van Assen H, Bovendeerd P, Delhaas T, eds.
correlate with impaired energetics and subclinical left ventricular Functional Imaging and Modeling of the Heart: 8th International
dysfunction. J Cardiovasc Magn Reson. 2014;16:29. Conference, FIMH 2015, Maastricht, The Netherlands, June 25–27, 2015.
86. Dass S, Cochlin LE, Suttie JJ, et al. Exacerbation of cardiac energetic Proceedings. Cham: Springer International Publishing; 2015:189–197.
impairment during exercise in hypertrophic cardiomyopathy: a potential 96. Oksuz I, Bevilacqua M, Mukhopadhyay A, Dharmakumar R, Tsaftaris S.
mechanism for diastolic dysfunction. Eur Heart J. 2015;36:1547–1554. BOLD contrast: a challenge for cardiac image analysis. J Cardiovasc
87. Parnham S, Gleadle JM, Bangalore S, et al. Impaired myocardial Magn Reson. 2016;18:1–2.
oxygenation response to stress in patients with chronic kidney disease. J 97. Bai W, Shi W, Ledig C, Rueckert D. Multi-atlas segmentation with
Am Heart Assoc. 2015;4:e002249. augmented features for cardiac MR images. Med Image Anal.
88. Dharmakumar R, Green J, Flewitt J, et al. Imaging for Probing the 2015;19:98–109.
Myocardial Perfusion Reserves of Patients With Coronary Artery Disease: A 98. Mukhopadhyay A, Oksuz I, Bevilacqua M, Dharmakumar R, Tsaftaris
Feasibility Study. Los Angeles: Society for Cardiovascular Magnetic SA. Unsupervised myocardial segmentation for cardiac MRI. In: Navab
Resonance; 2008. N, Hornegger J, Wells MW, Frangi FA, eds. Medical Image Computing
89. Luu JM, Friedrich MG, Harker J, et al. Relationship of vasodilator- and Computer-Assisted Intervention—MICCAI 2015: 18th International
induced changes in myocardial oxygenation with the severity of Conference, Munich, Germany, October 5-9, 2015, Proceedings, Part III.
coronary artery stenosis: a study using oxygenation-sensitive Cham: Springer International Publishing; 2015:12–20.
cardiovascular magnetic resonance. Eur Heart J Cardiovasc Imaging. 99. Yang H, Dey D, Sykes J, et al. Towards reliable myocardial blood-oxygen-
2014;15:1358–1367. level-dependent (BOLD) CMR using late effects of regadenoson with
90. Zhou X, Tsaftaris SA, Liu Y, et al. Artifact-reduced two-dimensional cine simultaneous 13N-ammonia PET validation in a whole-body hybrid
steady state free precession for myocardial blood-oxygen-level- PET/MR system. J Cardiovasc Magn Reson. 2016;18(suppl 1):O19.
dependent imaging. J Magn Reson Imaging. 2010;31:863–871. 100. Tsaftaris SA, Rundell V, Zhou X, et al. Detecting myocardial ischemia at
90a.  Oksuz I, Mukhopadhyay A, Dharmakumar R. Unsupervised myocardial rest with cardiac phase-resolved BOLD MRI: early findings. In: The 19th
segmentation for cardiac BOLD. IEEE Trans Med Imaging. 2017;36: Meeting of the International Society for Magnetic Resonance Medicine
2228–2238. (ISMRM). Montreal, Canada: ISMRM; 2011.
9 
Cardiac Magnetic Resonance Spectroscopy
Stefan Neubauer and Christopher T. Rodgers

between ~5 cm and ~1 m. The nucleus-specific probe head with the
INTRODUCTION
radiofrequency (RF) coils, which are used for MR excitation and signal
Cardiovascular magnetic resonance (CMR) imaging uses the 1H nucleus reception, is seated within the magnet bore. The magnet is interfaced
in water (H2O) and fat (CH2 and CH3 groups) molecules as its only with a computer, a pulsed magnetic field gradient system, an RF trans-
signal source, and therefore offers little insight into the biochemical mitter, and RF receivers. MRS requires a much higher magnetic field
state of cardiac tissue. In contrast, MR spectroscopy (MRS) of the heart homogeneity than CMR; therefore, before any MRS experiment, the
allows the study of many other nuclei. It is the only available method for magnetic field must first be homogenized with shim gradients. Spin
the noninvasive assessment of cardiac metabolism without needing the excitation is achieved by transmitting a radiofrequency pulse through
application of external radioactive tracers. Information on the major the RF coils into the subject. The resulting MR signal, known as the free
nuclei of interest for the metabolic study of cardiac tissue by MRS is induction decay (FID), is then received from the subject by RF coils and
given in Table 9.1, including 1H, 13C, 23Na, and 31P. Although, theoretically, recorded by the scanner. The signal (or FID) oscillates with time but
many clinical questions can be answered with cardiac MRS, the main within an overall exponentially decaying “envelope.” It typically reaches
reason why MRS has not yet fulfilled its potential in clinical cardiol- negligible levels after tens of milliseconds. An MR spectrum is then
ogy is related to the fundamental physical limitations of the method. computed from the FID (by applying the discrete Fourier transform
MRS is often used to study nuclei other than 1H, which have a much formula10). The MR spectrum describes the signal intensity as a func-
lower intrinsic MR sensitivity than 1H. Furthermore, metabolite con- tion of resonance frequency. Because of the low sensitivity of MRS, in
centrations in vivo are typically in the mM (millimolar) range, which is practice, many FIDs are averaged to obtain MR spectra with an adequate
several orders of magnitude lower than for water (approximately 80 M) “signal-to-noise ratio” (SNR; i.e., the height of a peak in the spectrum
or fat (often >1 M). Therefore the temporal and spatial resolution of after “matched filtering” divided by the standard deviation of baseline
MRS has so far remained far behind that of CMR imaging. Successful noise11). The required number of averages depends on the concentra-
applications of MRS are those in which the unique metabolic insights tion of the metabolite under investigation, its MR relaxation times (T1,
of MRS are more important than obtaining high spatial or temporal T2, T2*), the “filling factor” (the mass of the heart relative to the coil
resolution. size), the natural abundance of the nuclear isotope studied, its relative
Although this chapter focuses on clinical cardiac MRS, some expla- MR sensitivity (see Table 9.1), the pulse angle, and the pulse repetition
nation of the experimental principles of MRS is important even for time (TR). For a perfused rat heart experiment at ≥7 T, 100 to 200 FIDs
the clinical reader: MRS has been a widespread method in experimental are typically acquired. To quantify metabolite concentrations from MR
cardiology, ever since the first 31P-MR spectrum from an isolated heart spectra, it is vital to correct for partial saturation, which depends on the
was obtained by Radda’s group in the 1970s,1 and since then experi- selected pulse angles and TR. This is because the maximum MR signal is
mental MRS studies have offered numerous fundamental insights into only obtained when nuclei are excited from a fully relaxed spin state, that
cardiac metabolism. Furthermore, only with an understanding of the is, when a time of at least 5 × T1 has passed since a previous excitation
major implications of experimental MRS studies are we able to fully (e.g., T1 of phosphocreatine [PCr] at 7 T is ~3 s requiring TR of ≥15 s,
appreciate the potential of the method and extrapolate to clinical cardiac at 1.5 T the PCr T1 ~4.4 s requiring TR of ≥22 s); acquisition of fully
MRS applications that should become feasible in the future, once the relaxed MR spectra therefore requires long TRs, leading to prohibitively
technical challenges presently limiting the clinical utility of MRS have long acquisition times. In practice, we take advantage of the fact that
been overcome. For those interested in greater detail on experimental the initial part of the FID contains most of the signal. Use of shorter
MRS and methodological background, comprehensive reviews of the TRs therefore yields spectra with higher SNR for a given acquisition
subject are available elsewhere.2–4 Complementary clinical reviews are time, but some of the signal is lost because of saturation. Such spectra
also available.5,6 are termed “partially saturated.” The extent of saturation also varies
for different 31P-resonances, because the T1s of 31P-metabolites such
as PCr and adenosine triphosphate (ATP) are significantly different
PHYSICAL PRINCIPLES (T1 of PCr is 2–3 × longer than T1 of ATP). Therefore for metabolite
The basic principles of MRS (see the reference list for textbooks on the quantification from partially saturated spectra, the spectral peak areas
general physical principles of CMR and MRS7–9) are best explained from are determined (e.g., fitted using the Advanced Method for Accurate,
the most extensively studied nucleus, 31P, and from the most widely Robust, and Efficient Spectral Fitting [AMARES]12 or LCModel13), and
used animal model, the isolated buffer-perfused rodent heart. These then correction factors are applied to obtain the metabolite concentra-
principles apply to MRS of all nuclei. An MR spectrometer consists of tion. By comparing fully relaxed and saturated spectra, these factors
a high-field superconducting magnet (currently up to 23.5 T, where can be determined for each metabolite. In practice, TRs and pulse flip
tesla [T] is the unit of magnetic field strength) with a bore size ranging angles for MRS are chosen to yield acceptable measurement times at

97
98 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

at distinct frequencies, allowing their discrimination from each other.


TABLE 9.1  Elements (Nuclei) Most
Chemical shift is given in units of parts per million (ppm) relative to
Frequently Studied by Magnetic Resonance the resonance frequency of a reference compound (e.g., phosphoric acid
Spectroscopy H3PO4). 31P-metabolite resonances extend over a range of ~30 ppm.9 The
Natural Relative MR Myocardial Tissue physical basis of chemical shift is that nuclei at different positions in the
Nucleus Abundance Sensitivity Concentrations molecule have different spatial distributions of electrons around them,
1
H 99.98% 100% H2O 110 M; up to ~90 mM which interact differently with the scanner’s magnetic field.14 Fitting of
(CH3-1H of creatine) spectral peak areas (e.g., with AMARES) and saturation correction gives
13
C 1.1% 1.6% Labeled compounds, metabolite concentrations in arbitrary units. Often metabolite ratios,
several mM such as the PCr/ATP ratio, are then calculated—implicitly assuming that
23
Na 100% 9.3% 10 mM (intracellular); the concentration of the metabolite in the denominator (ATP here) is
140 mM (extracellular) well regulated enough to act as an endogenous reference. Alternatively,
31
P 100% 6.6% Up to ~18 mM (PCr) using an exogenous reference phantom, absolute metabolite concentra-
tions can be computed.15 Frequently, phenylphosphonic acid (PPA) is
MR, Magnetic resonance; PCr, phosphocreatine. used for this purpose because this generates an additional peak in the
spectrum, which does not overlap with the cardiac 31P resonances.16,17
The most significant advantage of MRS over destructive methods such
PCr
as traditional biochemical assays, where the tissue has to be frozen and
extracted, is that the MRS measurement is noninvasive. Therefore, spectra
can be acquired sequentially, and the response of energy metabolites
to ischemia, hypoxia, or inotropic stimulation can be followed longi-
tudinally in the same tissue. With this approach, each heart can serve
ATP as its own control, yielding a more powerful experimental design and
substantially reducing the number of required experiments.

γ– α– β– EXPERIMENTAL FOUNDATIONS
31
P-MRS
With 31P-MRS, cardiac high-energy phosphate metabolism, that is, the
Pi energetic state of the heart, can be monitored noninvasively. ATP is the
PME direct energy source for all energy-consuming reactions in the heart.
PCr acts as a temporal energy buffer and as an energy transport mol-
ecule in the “creatine kinase energy shuttle”18 (Fig. 9.2). For this, the
high-energy phosphate group is transferred from ATP to creatine in
10 0 –10 –20 the mitochondria (where ATP is produced by ATP synthase), yielding
PCr and adenosine diphosphate (ADP) in a reaction catalyzed by the
Chemical shift (ppm)
mitochondrial creatine kinase isoenzyme (CKmito). PCr diffuses through
FIG. 9.1  31P-magnetic resonance spectrum of an isolated, buffer-perfused
the cytoplasm to the site of ATP utilization, the myofibrils, where the
rat heart obtained in 5 minutes at 7 T. ATP, Adenosine triphosphate; PCr,
phosphocreatine; Pi, inorganic phosphate; PME, phosphomonoesters.
back reaction occurs (catalyzed by the myofibrillar-bound MM-creatine
kinase isoenzymes; CKMM), ATP is reformed and is used for contraction.
Free creatine then diffuses back to the mitochondria. The reaction rates
an ~20% to 50% degree of saturation. Larger degrees of saturation, at the creatine kinase (CK) isoenzymes complement each other to
resulting from the use of extremely short TRs, make quantification of maintain this steady state.19 The second important function of PCr and
spectra unreliable because of uncertainties in the metabolite T1 values creatine kinase is to control the thermodynamic state of the cell, that
and the dynamic chemistry of these metabolites. is, to maintain free cytosolic ADP at low concentration on exertion,
A 31P-MR spectrum from an isolated, beating rat heart, obtained instead of raising ADP and consuming Cr in the mitochondria, thereby
in 5 minutes at 7 T with a TR of 1.93 seconds and a pulse angle of 45 up-regulating oxidative phosphorylation.20 This is a requirement for
degrees, is shown in Fig. 9.1. A typical cardiac 31P-spectrum shows six normal cardiac function, because ADP determines the free energy change
resonances, corresponding to the three (γ, α, β) 31P atoms of ATP (the of ATP hydrolysis (ΔG; kJ/mol), a measure of the maximum amount
resonance at the right shoulder of the α-ATP peak represents the 31P of energy released from ATP hydrolysis (see Neubauer21 for details on
atom in nicotinamide adenine dinucleotide [NAD+]), PCr, inorganic the calculation of free ADP and ΔG from creatine kinase shuttle metabo-
phosphate (Pi), and phosphomonoesters (PME), mostly representing lites). In the normal heart, ΔG is ~−58 kJ/mol. Many intracellular enzymes
adenosine monophosphate (AMP) and glycolytic intermediates.9 The such as SR-Ca2+-ATPase and others will not function properly above a
reason why only a small number of 31P resonances is detectable, in spite threshold value for ΔG of about −52 kJ/mol. CK has also been implicated
of many more 31P-containing metabolites being present in the heart, is in minimizing cellular ADP loss, maintaining cellular pH, and activating
that, for 31P nuclei to be visible, they must be present above a certain con- glycogenolysis and glycolysis.20
centration threshold of ~600 µM and free to tumble in solution. Largely Pi is formed when ATP is hydrolyzed: ATP ⇌ ADP + Pi. Pi increases
immobilized metabolites such as plasma membrane phospholipids do when ATP utilization exceeds ATP production, for example, during
not yield a quantifiable MR signal because of their very short T2 values; ischemia. Intracellular pH (pHi) can also be measured with 31P-MRS,
instead, these metabolites contribute to the broad “baseline hump” of from the chemical shift difference between PCr and Pi, which is pH-
31
P spectra, which is particularly pronounced in the brain. Because of sensitive because of shifts in the equilibrium H2PO4− ⇌ HPO42− + H+
the phenomenon termed “chemical shift,” different metabolites resonate near pH 7.
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 99

Mitochondria Cytoplasm Myofibrils

ADP Phosphocreatine ADP

Oxidative
ATP-transfer ATP utilization
phosphorylation

ATP Creatine ATP


CKmito CKMM
FIG. 9.2  Creatine kinase energy shuttle. For details, see text. ADP, Adenosine diphosphate; ATP, adenosine
triphosphate; CKmito, mitochondrial creatine kinase isoenzyme; CKMM, MM-creatine kinase isoenzyme.

Control 15 min ischemia End of reperfusion

Untreated

PCr
ATP
Pi γ− α− β−
ET-1

10 5 0 –5 –10 –15 –20 10 5 0 –5 –10 –15 –20 10 5 0 –5 –10 –15 –20


FIG. 9.3  31P-magnetic resonance spectra from an untreated perfused rat heart and a heart treated with
endothelin-1 (ET-1) during control, at the end of 15 minutes of ischemia and at the end of reperfusion. For
details, see text. ATP, Adenosine triphosphate; PCr, phosphocreatine; Pi, inorganic phosphate. (From de
Groot MC, Illing B, Horn M, et al. Endothelin-1 increases susceptibility of isolated rat hearts to ischemia/
reperfusion injury by reducing coronary flow. J Mol Cell Cardiol. 1998;30:2657–2668.)

In principle, the intracellular magnesium ion concentration can also imaging technology seems likely soon to offer similar insight earlier
be measured by 31P-MRS from the chemical shift difference between in the energetic chain.32,33
PCr and β-ATP.22,23 With the magnetization (saturation) transfer method,34–36 the forward
Cardiac energy metabolism has been investigated by 31P-MRS rate and velocity of the creatine kinase reaction, a measure of ATP
under various experimental conditions. For example, the effect of transfer from mitochondria to myofibrils, can be measured in vivo.
changes in cardiac workload on energetics has been examined. PCr Creatine kinase reaction velocity correlates with cardiac workload37 and
levels do not change with moderate changes in workload,24 but decline with recovery of mechanical function after ischemia.38 In animal models
with substantial increases in cardiac work.25 ATP content remains or skeletal muscle, magnetization transfer also allows measurement of
almost constant with varying workload and during the cardiac cycle, the forward rate and velocity of the ATP hydrolysis reaction in the
because the creatine kinase equilibrium favors ATP synthesis over PCr myofibrils as a measure of ATP consumption.39,40
synthesis by a factor of ~100.26–28 Thus for any situation of myocar- Experimental 31P-MRS studies have substantially contributed to our
dial stress, including ischemia, ATP only decreases when PCr levels understanding of the role of energetics in heart failure.34,41–44 Independent
are substantially depleted.29 This is the fundamental reason why the of the etiology of heart failure, the failing myocardium shows reduced
PCr/ATP ratio is a highly sensitive indicator of the energetic state PCr, unchanged or moderately (by <30%) reduced ATP, unchanged or
of the heart. Changes of myocardial energy metabolism in experi- increased Pi, and substantially reduced creatine kinase reaction veloc-
mental models of ischemia and reperfusion highlight the potential ity and flux. These changes are likely to contribute to the impairment
of 31P-MRS for the detection of ischemia in the human heart. Fig. of contractile reserve in failing myocardium, because of the failure to
9.3 shows an example of 31P-MR spectra during control, ischemia, maintain appropriate ΔG values during inotropic stimulation.45
and reperfusion. After 15 minutes of total, global ischemia, ATP and
1
PCr resonances have disappeared, and almost all the 31P in heart is H-MRS
present as Pi and PME. During reperfusion, PCr and Pi show full, Other than 31P, the nuclei with the greatest potential for clinical cardiac
and ATP partial recovery. We showed that, when hearts are pretreated MRS are 1H and hyperpolarized 13C (of which, more below). Protons
with endothelin-1, a hormone that increases susceptibility to ische- have the highest MR sensitivity of all MR-detectable nuclei as well as high
mia, recovery of high-energy phosphate metabolism is impaired.30 By natural abundance (see Table 9.1). 1H is contained in a large number of
summing up data from several experiments, Clarke et al.31 demon- metabolites, for example, creatine, lactate, carnitine, taurine, and −CH3
strated that the decrease of PCr and the increase of Pi were amongst and −CH2 resonances of lipids.46,47 Measurement of total creatine by
the very earliest metabolic responses in myocardial ischemia, with sig- single voxel spectroscopy48–50 or navigator-gated echo planar spectroscopic
nificant changes occurring within seconds. Thus if we were successful imaging (EPSI)51 or potentially by creatine chemical exchange saturation
in measuring energetics in human myocardium with high temporal transfer (CrCEST) imaging52 should, in conjunction with 31P-MRS, allow
and spatial resolution, we could directly image parameters that detect the noninvasive determination of free ADP and ΔG of ATP hydrolysis,
myocardial ischemia within seconds after its onset. No other diag- as demonstrated by Bottomley and Weiss in dogs48 and humans.53,54 By
nostic approach currently achieves this, although hyperpolarized 13C means of the oxymyoglobin and deoxymyoglobin resonances, tissue
100 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

deoxygenation can be measured.55 Technical challenges for 1H-MRS and the formation of both intracellular and extracellular edema (see
include the need for suppression of the strong 1H signal from water Kim et al.66 and Horn et al.67). Furthermore, 23Na remains significantly
and the complexity of 1H spectra with overlapping resonances, many elevated during chronic scar formation post coronary ligation67 because
of which remain to be characterized, and which means that excellent of the expansion of the extracellular space in scar, and the area of
shimming and motion compensation are needed.56,57 elevated 23Na signal correlates closely with histologically determined
infarct size. Importantly, 23Na content is not elevated in stunned or
13
C-MRS hibernating myocardium.67 Thus 23Na CMR may allow detection of
The 13C nucleus has a low natural abundance (~1%), and for a classical myocardial viability without the use of external contrast agents. Sig-
13
C-MRS experiment, the heart has to be supplied with 13C-labeled com- nificant improvements in image quality were made at 3 T,68–70 and,
pounds such as, for example, 1-13C-glucose. Cardiac substrate utilization,58 recently, high-quality human 23Na images and retrogated cine movies
citric acid cycle flux, pyruvate dehydrogenase flux or beta-oxidation have been acquired at 7 T in the human heart.71,72 An example of the
of fatty acids can then be quantified.59,60 Clinical cardiac studies have high spatial and temporal resolution from 7 T is shown in Fig. 9.4.
yet to be reported because of the low sensitivity of 13C-MRS and the Furthermore, Umathum et al. have recently presented the first results
requirement for high concentrations of exogenous 13C-labeled precursors. from a whole-body 23Na birdcage coil at 7 T, which can image from
However, the technique of hyperpolarization can increase the SNR the pelvis to the heart in one scan.73 The outlook for cardiac sodium
of 13C experiments by a factor of up to 10,000× for a few minutes, until CMR has recently been reviewed by Bottomley.74
the magnetization decays by T1 relaxation back to thermal equilib-
rium.32,33 This is a very active area of research as new pulse sequences, Other Nuclei
analysis methods, and injectable hyperpolarized agents are developed The availability of ultra-high field (7 T) CMR scanners is making it
to capitalize on this extraordinary but brief burst of enhanced signal- possible to perform human in vivo spectroscopy and imaging also of
to-noise. Pyruvate61,62 is a particularly attractive molecule to hyperpolarize fluorine (19F), which is not present naturally in the body in mobile form
because it plays a pivotal role in substrate uptake before oxidative phos- and therefore makes an attractive CMR tracer to study, for example,
phorylation, enabling fluxes into acetyl acetate, lactate, and bicarbonate in drug uptake. Imaging of chlorine (35Cl) and potassium (39K) has
to be quantified. These enable experimental assessment of the balance also been demonstrated at 7 T,75–78 which makes it possible to map cell
between fats, carbohydrates, and ketone bodies being metabolized by membrane potential in vivo.78
the heart, a balance which may well be disturbed in disease. Hyperpo-
larization studies in animal models are revisiting many of the systems CLINICAL MAGNETIC RESONANCE
previously characterized by 31P-MRS to obtain further insight, for SPECTROSCOPIC STUDIES
example, ischemia-reperfusion models.63
Methodologic Considerations
23
Na-CMR Historically, almost all human cardiac MR spectroscopy studies have
23
Na-CMR can evaluate changes in total and intracellular and extra- interrogated the 31P nucleus. The main reason for the slow progress
cellular Na+ during cardiac injury.64 A cardiac 23Na spectrum shows a with clinical cardiac MRS is that the method poses major technical
single peak representing the total Na+ signal. To split the intracellular challenges. In practice, total examination time should not be more than
and extracellular Na+ pools into two resonances, paramagnetic shift 60 minutes, and time for signal acquisition is therefore limited. The
reagents, such as [TmDOTP]5−, are added to the perfusate. This method heart is a rapidly moving organ, currently requiring gating to the elec-
has been used experimentally to examine the mechanisms of intracel- trocardiogram (ECG), and, when resolution is further improved, ulti-
lular Na+ accumulation in ischemia-reperfusion injury,65 but 23Na-MR mately to respiration as well.79 Most clinical MRS studies were performed
shift reagents are not yet available for clinical use. Experimental CMR at 1.5 T until the early 2000s, and since then mostly at 3 T; the first 7 T
of total 23Na shows that in acute ischemia, the total myocardial 23Na studies are now being published.80,81 Although the field strength of
CMR signal increases because of the breakdown of ion homeostasis animal scanners continues to increase, the latest 7 T human systems

A 0.2 s 0.4 s 0.6 s 0.8 s 1.0 s

B 0.2 s 0.4 s 0.6 s 0.8 s 1.0 s


FIG. 9.4  Na-cardiovascular magnetic resonance of the human heart. (A) A four-chamber view with field of
23

view (FOV) zoomed to (210 × 273) mm2 and (B) a short-axis view (FOV = [180 × 150] mm2). The ventricles
and the septum are clearly visible in A and B. The left and right atriums are delineated from the ventricles
in the four-chamber views. The rib cartilage shows the highest signal intensity because of the high sodium
concentration. The temporal resolution is 200 ms, nominal spatial resolution (6 mm)3. The images were
acquired with the ST acquisition scheme (10,000 projections) and reconstructed with a Hanning filter. (From
Resetar A, Hoffmann SH, Graessl A, et al. Retrospectively-gated CINE (23)Na imaging of the heart at 7.0
tesla using density-adapted 3D projection reconstruction. Magn Reson Imaging. 2015;33:1091–1097.)
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 101

have comparable field strength with that used historically for many α
important animal studies. The cardiac muscle lies behind the chest wall RF t
skeletal muscle, which creates a strong 31P signal that must be suppressed Acquire
by spectral localization techniques, outer volume suppression,82 or surface Gy
spoiling gradient coils.83 Such localization methods include depth-resolved A
surface coil spectroscopy (DRESS), rotating frame one-dimensional
chemical shift imaging (1D-CSI), image-selected in vivo spectroscopy nθx α+y
(ISIS), and three-dimensional (3D)-CSI (Fig. 9.5). These methods are t
reviewed in detail elsewhere.84 Bottomley et al.85–87 have pioneered most RF Acquire
of the early methodological development of human cardiac MRS. Most
MRS studies have been performed in prone position rather than supine B RF gradient Uniform RF
because this reduces motion artifacts and may slightly reduce the dis-
tance of the heart from the surface coil, thereby improving sensitivity. α
Most spectroscopic techniques require a stack of 1H scout images to t
RF
be obtained first, which are used to select the spectroscopic volume(s). Acquire
The low sensitivity of 31P-MRS requires relatively large voxel sizes, typi-
cally about 20 to 70 mL (and often rather larger if one considers the Gy
“true” integrated point-spread-function volume).80 A set of 31P-MR C
spectra acquired from a healthy volunteer with 3D-CSI at 1.5 T, 3 T,
and 7 T is shown in Fig. 9.6. Compared with the rat heart spectrum in π π π α
Fig. 9.1, the signal-to-noise is lower at 1.5 and 3 T, and two additional t
resonances are detected: 2,3-diphosphoglycerate (2,3-DPG), because Acquire
of the presence of erythrocytes in the interrogated voxel, and phospho- RF
diesters (PDE), a signal caused by membrane as well as serum phos- Gx
pholipids. The 2,3-DPG peaks overlap with the Pi resonance, which is Gy
D Gz
therefore challenging to discriminate in blood-contaminated human
31
P-MR spectra. Thus intracellular pH is hard to determine; early results87a
suggest that Pi and pH become detectable in human myocardium when α
RF t
spatial resolution and signal-to-noise are increased by scanning at 7 T.
Acquire
By calculating the PCr/ATP and PDE/ATP peak area ratios, relative
quantification of human 31P spectra is simple. PCr/ATP is a powerful Gz
index of the energetic state of the heart (see Experimental Foundations
section), whereas the meaning of the PDE/ATP ratio is poorly under- Gy
stood, and this ratio probably does not change with cardiac disease. 31P
resonances must be corrected for partial saturation, as described for Gx
E
experimental MRS. Appropriate T1 values have been summarized.2,80
Intrinsic 31P-T1 values are believed to remain constant in the presence FIG. 9.5  Basic pulse sequences for localized cardiac spectroscopy with
surface coils. (A) “Depth-resolved surface coil spectroscopy.” A single
of cardiac disease,88 although observed T1 values can change because
section parallel to the plane of the surface coil is selected by applying
of the effects of chemical exchange in the CK system.89 Furthermore,
31
a magnetic resonance (MR) imaging gradient G in the presence of a
P spectra require correction for blood contamination: blood contributes modulated radiofrequency (RF) excitation pulse of flip angle α. (B) The
signal to the ATP, 2,3-DPG and PDE resonances. Because human blood “rotating frame” MR method uses the gradient inherent in a surface coil
spectra have an ATP/2,3-DPG area ratio of ~0.11 and a PDE/2,3-DPG to simultaneously spatially encode spectra from multiple sections parallel
area ratio of ~0.19, for blood correction, the ATP resonance area of to the surface coil by means of application of a θ flip angle pulse, which
cardiac spectra is reduced by 11% of the 2,3-DPG resonance area, and is stepped in subsequent applications of the sequence. (C) The “one-
the PDE resonance area is reduced by 19% of the 2,3-DPG resonance dimensional chemical shift imaging” method similarly encodes multiple
area.90 31P-metabolite ratios in blood also change in the presence of sections but uses an MR imaging gradient whose amplitude is stepped.
disease,91 which should be taken into account. (D) The “image-selected in vivo spectroscopy” method localizes to a
single volume with selective inversion pulses applied with Gx, Gy and Gz
Absolute quantification of PCr and ATP is technically challenging,
MR imaging gradients. All eight combinations of the three pulses must
but is desirable because the PCr/ATP ratio does not detect simultaneous
be applied and the resultant signals added and subtracted. (E) A section-
decreases of both PCr and ATP, which occur in the failing heart92 or in selective “three-dimensional chemical shift imaging” sequence employs
infarcted nonviable myocardium.93 Absolute 31P-metabolite levels can be MR imaging section selection in one dimension and phase encoding in
obtained by acquiring signal from a 31P standard as well as estimates of two dimensions. (From Bottomley P. MR spectroscopy of the human
myocardial mass from CMR.86,94 A different approach calibrates the 31P heart: the status and the challenges. Radiology. 1994;191;593–612.)
signal to the tissue water proton content, measured by 1H-MRS.87 Signals
can be calibrated to an electronically synthesized reference signal using
the “electronic reference to access in vivo concentrations” (ERETIC) time most effectively to find concentrations in a predetermined com-
method.95 Modern “non-Fourier” methods solve for the metabolite con- partment, for example, myocardium segmented from a 1H image. A
centrations using the powerful “inverse problem” mathematical formu- variety of non-Cartesian acquisition methods, such as compressed sensing
lation. For example, spectral localization with optimum pointspread (CS) spectroscopy99 or spiral 31P-MRS,100 also enable a smooth tradeoff
(SLOOP)96 postprocessing can deliver concentrations in noncuboidal between acquisition time, spatial resolution, and spectral resolution.
compartments (e.g., myocardium). Spectroscopy with linear algebraic A long-term goal for human cardiac MRS is to combine 31P- and
modeling (SLAM)97,98 accelerates the acquisition steps to focus the scan 1
H-spectroscopy to measure free ADP and ΔG of ATP hydrolysis, as
102 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

10
1.5 T
3T
8 7T
PCr

Signal/normalized 6

γ-ATP α-ATP
4
β-ATP

2 DPG

0
10 5 0 -5 -10 -15 -20
Chemical shift/ppm
FIG. 9.6  Left, 31
P-magnetic resonance spectra from the interventricular septum of the same volunteer
recorded at 1.5 T, 3 T, and 7 T using a three-dimensional chemical shift imaging (3D-CSI) sequence. Field-
of-view 240 × 240 × 200 mm3 with a 12 × 8 × 8 matrix with 32 averages in 22 minutes at 1.5 T; a 16 × 16
× 8 matrix with 10 averages in 28 minutes at 3 T and 7 T. DPG, 2,3-Diphosphoglycerate (from blood); PCr,
phosphocreatine; γ-, α-, β-ATP, the three phosphorus atoms of adenosine triphosphate. To facilitate com-
parison the spectra are normalized to the baseline noise level, apodized, and the 1.5 T spectrum is scaled
to account for its larger voxel size and shorter acquisition. Right, Corresponding short-axis and four-chamber
localizer images recorded by 1H-cardiovascular magnetic resonance at 3 T. The 3D-CSI voxel grid is overlaid.
The voxel corresponding to the 31P spectra is highlighted.

described in canine myocardium by Bottomley et al.48 Another highly Athlete’s Heart and Hypertension
relevant energetic parameter is the rate and extent of ATP transfer (“CK In patients with hypertension, Lamb et al.110 demonstrated reduced
rate kf ” and “CK-flux”; see Experimental Foundations section). Bot- PCr/ATP ratios both at rest and during dobutamine stress, and PCr/
tomley et al.101 developed the four-angle saturation transfer (FAST) ATP ratios also correlated inversely with indices of diastolic function
method, which allows measurement of creatine kinase rates in ~30 (E deceleration peak). In contrast, two other studies showed no signifi-
minutes at 1.5 T. More recently, Bottomley et al. have introduced the cant changes of cardiac energetics in patients with hypertension.92,104
triple repetition time saturation transfer (TRiST)35 and two repetition Differences in patient characteristics—for example, severity and duration
time saturation transfer (TwiST)88 methods to measure CK flux at 3 T of hypertension—may be responsible for this discrepancy. Experimental
based on dual-TR T1 measurements102 during saturation (illustrated data clearly suggest that cardiac energetics are impaired in long-standing
in Fig. 9.7), and we have introduced Bloch-Siegert four angle saturation hypertension.111 In contrast, physiologic hypertrophy in the athlete’s
transfer (BOAST) to measure 3D-resolved CK rates at 7 T.36 heart does not lead to a decrease of the myocardial PCr/ATP ratio,
The methods of 31P-MRS are still being improved significantly. Nev- either at rest or during stress112; experimental studies in exercise-trained
ertheless, a major effort to standardize protocols and to make routinely rats had predicted this finding.113 In the future, it will be important to
available advanced RF coils and pulse sequences at ultra-high field unravel the molecular mechanisms responsible for these differences in
strengths will be required to bring cardiac MRS into clinical practice. energy metabolism between physiologic and pathologic hypertrophy.

Healthy Volunteers Diabetes and Obesity


Because of differences in the MRS methods used, the range of “normal” Other than via the well-recognized secondary mechanisms (e.g., acceler-
human heart PCr/ATP ratios reported in the literature is considerable, ated coronary disease), diabetes has numerous deleterious effects on
from about 1.1 to 2.4, with an overall average of about 1.8.2 This attests cardiac metabolism that may lead to cardiomyopathy. For example, the
to the need for development of methodologic standards for MRS. SLOOP diabetic heart is insulin resistant and glucose utilization is impaired.
values for absolute PCr levels in normal human myocardium are 9.0 Plasma free fatty acid levels are elevated, leading to increased expression
± 1.2 mmol/kg wet weight, and ATP levels are 5.3 ± 1.2 mmol/kg wet of mitochondrial uncoupling proteins and reduced expression of glucose
weight.103 There is an accumulation of evidence that high-energy phos- transporters (GLUT4). Several studies have examined cardiac energy
phate levels decrease slightly with advanced age,104–107 perhaps because metabolism in patients with maintained left ventricular (LV) ejection
of increasing mitochondrial dysfunction.27,108 During stress, PCr/ATP fraction and type 2114–116 or type 1117,118 diabetes mellitus and have uni-
ratios stay normal for all but extreme levels of inotropic stimulation, formly shown reduced PCr/ATP ratios. Van der Meer et al.119 observed
when there is a small decrease.109 Using FAST (see earlier), high-energy no change in PCr/ATP in 78 type 2 diabetes mellitus men given either
phosphate turnover rates (CK reaction rates) were found to be 0.29 ± pioglitazone or metformin for 24 weeks, despite improvements in LV
0.06 s-1 in healthy volunteers at rest and did not change during doubling function. PCr/ATP ratios were found to correlate inversely with plasma
of the cardiac rate–pressure product.53 free fatty acid levels114 and with indices of diastolic function.115,117 Shivu
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 103

No saturation, TR = 16.24 s Control saturation, TR = 16.23 s g-ATP saturation, TR = 10.23 s g-ATP saturation, TR = 1.94 s
Signal (a.u.)

Signal (a.u.)

Signal (a.u.)

Signal (a.u.)
Frequency (ppm) Frequency (ppm) Frequency (ppm) Frequency (ppm)
A B C D
FIG. 9.7  Illustration of raw data used to measure the creatine kinase rate constant kf at 3 T with the two
repetition time saturation transfer (TwiST) protocol. (A) A 9-minute acquisition without any saturation used
to determine M0 from the peak height of the phosphocreatine (PCr) peak. This spectrum is also used to
determine the saturation frequency of the γ-adenosine triphosphate (ATP) peak in the heart. (B) A 9-minute
acquisition with control saturation at the frequency indicated by the arrow used to determine M0control. (C) A
22-minute acquisition with γ-ATP saturation (arrow) and long TR used to determine M0′ in the TwiST experi-
ment, and M′(TRlong) in the triple repetition time saturation transfer (TRiST) experiment. (D) This 9-minute
acquisition is only used in TRiST but not in TwiST. It is acquired with γ-ATP saturation (arrow) and short
repetition time and used to determine M′(TRshort). TwiST uses acquisitions shown in B and C; TRiST uses
acquisitions shown in B, C, and D; Q-corrected TwiST uses acquisitions shown in A, B, and C; and Q-corrected
TRiST uses acquisitions shown in A, B, C, and D. (From Schar M, Gabr RE, El-Sharkawy AM, Steinberg A,
Bottomley PA, Weiss RG. Two repetition time saturation transfer (TwiST) with spill-over correction to measure
creatine kinase reaction rates in human hearts. J Cardiov Magn Reson. 2015;17:70.)

et al.118 showed that in 25 asymptomatic type 1 diabetic patients, PCr/ independent predictor of LV concentric remodeling and cardiac systolic
ATP was reduced (1.6 ± 0.2 vs. 2.1 ± 0.5 in controls) within 5 years of strain in a study of 46 patients with type 2 diabetes.
diagnosis and that this changed little more than 10 years after diagnosis
(to 1.5 ± 0.4), whereas microvascular dysfunction worsened in diabetics Heart Failure
over time (myocardial perfusion reserve index 1.7 ± 0.6 vs. 2.3 ± 0.4, Experimental studies have firmly established altered energy metabolism
P = .005). We recently showed in a study of 31 type 2 diabetic patients as a hallmark of the chronically failing myocardium (see above). Initial
that exercise further reduced the PCr/ATP ratio (resting PCr/ATP was clinical 31P-MRS studies, however, which included mild stages of heart
1.74 ± 0.26 in patients vs. 2.07 ± 0.35 in controls, and dropped to 1.54 failure, did not find significant reductions of PCr/ATP ratios.132–134 Hardy
± 0.26; P = .005), and that this correlated with myocardial perfusion et al.135 first reported that the myocardial PCr/ATP ratio is significantly
reserve index from adenosine stress CMR imaging.116 reduced (from 1.80 ± 0.06 to 1.46 ± 0.07) in patients with symptomatic
Initial evidence also suggests reduced PCr/ATP ratios in patients heart failure of ischemic or nonischemic etiology. Subsequently, we
with uncomplicated obesity and elevated free fatty acid levels.120 For found that the decrease of PCr/ATP ratios in dilated cardiomyopathy
example, Rider et al.121 demonstrated cardiac energetic differences (DCM) correlated with the New York Heart Association (NYHA) class90
between obese and normal-weight subjects by 31P-MRS at rest: the obese and with LV ejection fraction.136 Thus PCr/ATP ratios decrease in
group had a 15% lower PCr/ATP ratio (1.73 ± 0.40 vs. 2.03 ± 0.28; advanced stages of heart failure (Fig. 9.8) but initially remain normal.
P = .048). During dobutamine stress, a further reduction in PCr/ATP This pattern was observed also by Leme et al. in a study of 39 patients
occurred in the obese group (from 1.73 ± 0.40 to 1.53 ± 0.50; P = .03) with Chagas disease, whose PCr/ATP ratios at rest correlated with NYHA
but not in normal-weight subjects (from 1.98 ± 0.24 to 2.04 ± 0.34; class137 and where PCr/ATP dropped further during isometric handgrip
P = .50). Together, these results suggest that early cardiac metabolic exercise, consistent with the involvement of ischemia in Chagas disease.138
derangement as observed in diabetes and obesity may contribute to PCr/ATP ratios also hold prognostic information on survival of patients
the later development of heart failure, suggesting a possible explanation with heart failure. We showed that in DCM, the myocardial PCr/ATP
for the increased incidence of heart failure in diabetes and obesity. ratio was a better predictor of long-term survival than LV ejection
However, intriguingly, it is also observed that obese patients with heart fraction or NYHA class139 (Fig. 9.9). It is known from experimental
failure survive longer than normal-weight patients with heart failure, work that in heart failure, both PCr and ATP levels decrease in paral-
a phenomenon termed the “obesity paradox.”122,123 lel,140 and this cannot be detected by measurement of PCr/ATP ratios.
Proton spectroscopy (1H-MRS) is an established method to monitor Accordingly, using SLOOP (see above) in patients with heart failure
triglyceride (TG) concentration (“fat fraction”) in the liver124–127 and because of DCM (ejection fraction 18%), Beer et al.141 reported that
heart.128 Van der Meer et al.129 investigated the effect of drinking 800 mL absolute PCr levels were reduced by 51%, ATP levels by 35%, whereas
cream for 3 days on healthy young men, observing a more than doubling the PCr/ATP ratio decreased by 25% only. Furthermore, significant
of hepatic TG concentration, but no significant effects on cardiac TG, correlations between LV volumes/ejection fraction and energetics were
function, or energetics. Holloway et al.130 compared the effect of a 5-day found, with the strongest correlations observed for PCr and the weakest
high-fat or normal-fat diet in healthy males, observing a 9% (P < .01) for the PCr/ATP ratio. Thus ATP levels are reduced in human heart
reduction on cardiac PCr/ATP in the high-fat group. Recently, Levelt failure, and the true extent of changes in energy metabolism in heart
et al.131 demonstrated that myocardial triglyceride content is an failure is underestimated when PCr/ATP ratios rather than absolute
104 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

50
DCM, patient died 7 days
after MR examination
40

Total mortality (%)


30 P = .036

DCM, PCr/ATP <1.6 PCr/ATP <1.60


20 PCr/ATP >1.60

10

DCM, PCr/ATP >1.6


0
0 500 1000 1500
Days of follow-up
2,3-DPG FIG. 9.9  Kaplan-Meier life table analysis for total mortality of dilated
cardiomyopathy patients. Patients were divided into two groups, split
by the myocardial phosphocreatine/adenosine triphosphate (PCr/ATP)
ratio (<1.60 vs. >1.60). Patients with an initially low PCr/ATP ratio showed
PCr increased mortality over the study period of, on average, 2.5 years.
(From Neubauer S,Horn M, Cramer M, et al. Myocardial phosphocreatine-
Volunteer
to-ATP ratio is a predictor of mortality in patients with dilated cardiomy-
γ- α- β-ATP opathy. Circulation. 1997;96:2190–2196.)

PDE

years, and showing that CK flux was a significant predictor of heart


failure outcomes even after correction for NYHA class, LV ejection
fraction, and race. For each increase in CK flux of 1 µmol/g/s, risk of
heart failure–related composite outcomes decreased by 32% to 39%.44
5 0 –10 –20
Overall, therefore, 31P-MRS may be valuable for prognosis evaluation in
Frequency (ppm)
heart failure.
FIG. 9.8  Cardiac 31P-magnetic resonance (MR) spectra. From bottom Heart failure trials from the past two decades show that energy-
to top: volunteer; dilated cardiomyopathy with normal phosphocreatine/
costly treatment, such as beta-receptor mimetics or phosphodiesterase
adenosine triphosphate (ATP) ratio; dilated cardiomyopathy with reduced
phosphocreatine/ATP ratio; dilated cardiomyopathy with severely reduced
inhibitors, increases mortality, whereas energy-sparing treatment, such
phosphocreatine/ATP ratio; this patient died 7 days after the MR exami- as beta-blockers, angiotensin-converting enzyme (ACE) inhibitors, or
nation. 2,3-DPG, 2,3-Diphosphoglycerate; PDE, phosphodiesters; PCr, angiotensin II receptor blockers, improves survival.123 Thus one of the
phosphocreatine; γ-, α-, and β- are P atoms of adenosine triphosphate most promising future applications of clinical 31P-MRS is monitoring
(ATP). (From Neubauer S, Horn M, Cramer M, et al. Myocardial phos- of the cardiac energetic response to new forms of heart failure treat-
phocreatine-to-ATP ratio is a predictor of mortality in patients with dilated ment, and it is likely that the PCr/ATP ratio or absolute concentrations
cardiomyopathy. Circulation. 1997;96:2190–2196.) of PCr and ATP are powerful surrogate parameters for mortality in
this situation. For example, in 6 patients with DCM treated with ACE
inhibitors, digitalis, diuretics and beta-blockers for 3 months, PCr/ATP
concentrations are measured. In a later study, Beer et al.142 applied ratios improved significantly during clinical recompensation, from 1.51
SLOOP 31P-MRS before and after exercise training in a study involving ± 0.32 to 2.15 ± 0.27.90 However, no systematic controlled study has
24 DCM patients. They demonstrated that exercise training improves so far used 31P-MRS to monitor cardiac energetics during heart failure
LV function without causing deterioration in cardiac energetics. Similar treatment. A treatment trial using 31P-MRS to monitor cardiac energet-
results were obtained by Holloway et al.143 in a study of 15 DCM patients ics has been reported for Friedreich ataxia. This primarily neurological
using 3 T 31P-MRS at rest and during exercise, where LV function disease is often associated with cardiomyopathy, as lack of the mito-
improved but no significant changes in PCr/ATP were observed. Nakae chondrial protein frataxin leads to deficient mitochondrial respiration
et al.144 used 1H-MRS to demonstrate significant reductions of total and increased free radical damage. In patients with Friedreich ataxia
creatine levels and a correlation of creatine and LV ejection fraction in treated with antioxidants (coenzyme Q and vitamin E) for 6 months,
patients with DCM. Lodi et al.146 reported that the myocardial PCr/ATP ratio increased from
Weiss et al.53 showed a 50% reduction of CK reaction rates in 1.34 ± 0.59 to 2.02 ± 0.43, demonstrating that cardiac energy metabo-
mild-to-moderate heart failure, indicating that energy turnover rates lism was markedly improved by antioxidative treatment. Hirsch et al.
are depressed to a greater extent than steady-state levels of PCr and demonstrated that 600 mg allopurinol (a xanthine oxidase inhibitor)
ATP. Weiss et al.145 subsequently measured the CK flux (i.e., ATP syn- improved cardiac energetics in patients with heart failure (i.e., PCr/ATP
thesis rate) in failing hypertrophied myocardium, suggesting that the increased by 11%, P < .02), and CK flux increased by 39% (from 2.07
kinetics of ATP turnover through CK distinguish failing from non- ± 1.27 µmol/g/s to 2.87 ± 1.82 µmol/g/s, P < .007).54 However, the 253
failing hypertrophic hearts more clearly than the relative or absolute patient multicenter randomized controlled Xanthine Oxidase Inhibition
CK metabolite pool sizes. Weiss et al. subsequently performed a pro- for Hyperuricemic Heart Failure Patients (EXACT-HF) trial failed to
spective study following 58 heart failure patients for a median of 4.7 observe any improvement in outcomes after 24-week administration
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 105

of allopurinol,147 suggesting that effective treatment requires modi-


fication of more than this one aspect of the cardiac nitroso-redox Valvular Heart Disease
balance.148,149 Experimental studies have shown impaired cardiac energy metabolism
Frenneaux et al.123 investigated the application of the antianginal in advanced left ventricular hypertrophy.169 Similarly, in patients with
drug perhexiline to treat heart failure in a series of studies. Perhexiline left ventricular hypertrophy resulting from aortic stenosis or incom-
is believed to inhibit the metabolism of free fatty acids. In heart failure petence, Conway et al.170 detected reduced PCr/ATP ratios (1.1 ± 0.32
patients, perhexiline increased the rate of PCr recovery in skeletal muscle vs. 1.5 ± 0.2 in volunteers; mean ± standard deviation) when patients
after exercise, indicating improved mitochondrial capacity.150 Recently, were in clinical heart failure, but PCr/ATP ratios were normal (1.56 ±
Frenneaux et al.151 published the results of a clinical trial in 50 patients 0.15) for clinically asymptomatic stages. Likewise, in patients with aortic
with nonischemic heart failure, observing that treatment with perhexiline valve disease, we showed reduced PCr/ATP ratios only for NYHA classes
200 mg once daily for 1 month was associated with a 30% increase in III and IV but not for classes I and II.171 When matched for the degree
PCr/ATP (from 1.16 ± 0.39 to 1.51 ± 0.51; P < .001).152 of heart failure, energy metabolism was more affected in aortic stenosis
(pressure overload) than in aortic incompetence. We also showed that
Specific Gene Defects With Cardiac Pathology in aortic stenosis, altered energetics correlated with left ventricular end-
Clinical cardiac 31P-MRS has major potential for the noninvasive phe- diastolic pressures and with end-diastolic wall stress.171 We reported
notyping of cardiomyopathies because of specific gene defects, which unchanged absolute ATP concentrations and a 28% decrease of PCr
may eventually be identifiable by a specific metabolic profile. Most concentrations in aortic stenosis using SLOOP.141 The time course of
of the work in this area has been on hypertrophic cardiomyopathy recovery of cardiac energetics during regression of hypertrophy after
(HCM), which is, in most cases, because of specific gene mutations surgical valve replacement can also be monitored by 31P-MRS.172 When
associated with structural disarray of myofibrils and often with sub- patients with aortic valve stenosis were studied before and 40 weeks
stantial increases of left ventricular wall thickness. Energetic derange- after surgery, the PCr/ATP ratio increased from 1.28 ± 0.17 to 1.47 ±
ment has been suggested as the common pathophysiologic mechanism 0.14 (control subjects 1.43 ± 0.14); that is, energetic impairment was
underlying all forms of HCM,153 and experimental studies of transgenic completely reversed 9 months after valve replacement. Energetics in
models of HCM confirm this.154 Human studies in HCM134,155–157 have aortic stenosis patients (1.45 ± 0.21 vs. 2.00 ± 0.25 in controls, P < .001)
uniformly shown reduced PCr/ATP ratios. For example, Jung et al.158 have been shown to correlate with perfusion, oxygenation, and LV
demonstrated that in young, asymptomatic patients with HCM, PCr/ functional defects, and to improve markedly 8 months after aortic valve
ATP ratios were reduced, indicating that energetic imbalance occurs replacement energetics (PCr/ATP 1.86 ± 0.48).173 A recent multicenter
early in the disease process. They also reported that HCM patients with a randomized control trial of perhexiline to augment myocardial protec-
familial history of the disease showed a more pronounced derangement tion in patients with left ventricular hypertrophy undergoing cardiac
of energetics than those patients without a family history.159 Abraham surgery was halted early when it became clear that perhexiline did not
et al.160 observed a 24% reduction in myocardial PCr, 26% reduction provide an additional benefit in hemodynamic performance or attenuate
in CK kf , and 44% reduction in forward CK flux in patients with HCM myocardial injury in the hypertrophied heart secondary to aortic stenosis
caused by a point mutation of Arg403Gln compared with volunteers. (AS).174 Nevertheless, long-term prospective clinical studies of the role
They argue that the energetic deficit, even in patients without changes of energetics in valvular disease remain the best strategy to determine
in cardiac function, means that the energetic deficit results from the whether 31P-MRS provides clinical information on the optimum timing
mutation and is not simply a consequence of mechanical dysfunction.160 of valve replacement.
Larger studies at 3 T have confirmed the derangement of energetics in
HCM.161,162 Dass et al.163 demonstrated that in 35 patients with HCM, Ischemic Heart Disease
leg exercise further reduced PCr/ATP (1.56 ± 0.29 vs. 1.71 ± 0.35 at rest; Magnetic Resonance Spectroscopy Stress Testing to
P = .02), whereas there was no change on exercise in normal (2.16 ± 0.26 Detect Ischemia
vs. 2.14 ± 0.35 at rest; P = .98). Frenneaux et al.164 showed that treat- Within seconds after reduction of oxygen supply, PCr levels decrease
ment of HCM patients with perhexiline for 4.6 ± 1.8 months improved and inorganic phosphate increases; that is, these changes are extremely
PCr/ATP (from 1.27 ± 0.02 to 1.73 ± 0.02, P = .003). Using 1H-MRS, rapid indicators of myocardial ischemia (see above). If it were feasible
Nakae et al.144 reported reduced total creatine content in patients with to detect these metabolites in human myocardium with high temporal
HCM. In the future, large patient groups with HCM and known spe- and spatial resolution, a 31P-MRS-based “biochemical stress test” would
cific gene defects will have to be studied to establish whether meta- be a powerful diagnostic tool for detecting exercise-induced regional
bolic phenotyping by 31P- and 1H-MRS can identify the underlying ischemia, only requiring low levels of stress and without the need for
genetic mutation. intravenous agents or radiation. In selected patients with large anterior
Becker muscular dystrophy, an X-chromosome linked disease associ- wall territories, which become ischemic on exercise, the feasibility of
ated with the absence or altered expression of dystrophin in cardiac and this principle has been demonstrated: Weiss et al. (Fig. 9.10) showed
skeletal muscle, may lead to the development of cardiomyopathy and that in patients with high-grade stenosis of the left anterior descending
heart failure. Clarke et al.165 showed that both patients (PCr/ATP ratio or left main coronary arteries, PCr/ATP ratios were normal at rest,
1.55 ± 0.33) and female gene carriers (1.37 ± 0.25) had significantly decreased during handgrip exercise (leading to a 30%–35% increase of
lower PCr/ATP ratios than control subjects (2.44 ± 0.33), although all of cardiac work) from 1.5 ± 0.3 to 0.9 ± 0.2, and returned toward normal
the carriers and most of the patients showed preserved left ventricular during recovery.175 After revascularization, PCr/ATP ratios remained
function. Thus energetic imbalance occurs early in the disease process constant during exercise. These findings were reproduced by Yabe et al.,176
and may contribute to the development of contractile dysfunction in who also demonstrated that a decrease of PCr/ATP ratios was only
Becker disease. Altered cardiac energetics have also been demonstrated detected in patients with reversible defects on thallium scintigraphy
in hereditary hemochromatosis,166 in familial hypercholesterolemia,167 (viable myocardium) but not in those with fixed thallium defects (scar),
where PCr/ATP ratios returned to normal after treatment with statins, where PCr/ATP was already reduced at rest.93 Najjar et al.177 applied
and in Fabry disease where PCr/ATP in 23 patients was 1.68 ± 0.43 31
P-MRS stress testing to investigate potential cardioprotective effects
compared with 1.92 ± 0.50 in volunteers.168 of RSR13 (an allosteric modifier of hemoglobin’s affinity for oxygen)
106 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

2.5 Myocardial Viability Assessment


Controls (N = 11)
Akinetic myocardium, supplied by a stenotic coronary artery, may be
nonviable (scarred), or it may be viable, that is, stunned or hibernating.
2.0 By readjusting the balance between oxygen supply and demand, hiber-
nating myocardium has downregulated its contractility to reduce energy
needs. Stunned myocardium shows transient contractile dysfunction
PCr/ATP

following reperfusion after reversible ischemia. 31P-MRS studies of animal


1.5 ischemia-reperfusion models182–186 have shown that in stunned and
hibernating187 myocardium, ATP levels remain close to normal, whereas
myocardial scar tissue contains negligible amounts of ATP (<1% of
1.0 normal levels). Thus a noninvasive diagnostic test that allows measure-
ment of myocardial ATP levels with high spatial (1–5 mL) and acceptable
temporal resolution (<30 minutes) should be attractive for viability
0.5 assessment. For example, Kalil-Filho et al.188 studied 29 patients with
anterior myocardial infarction (MI) 4 days and 39 days after MI. All
Rest Exercise Recovery
patients showed akinetic anterior myocardium, which had recovered
function at the time of the second examination. PCr/ATP ratios were
2.5 normal in stunned myocardium (1.51 ± 0.17 vs. 1.61 ± 0.18 in volun-
Patients (N = 16) teers) and did not change during the recovery of contractile function
(1.51 ± 0.17 vs. 1.53 ± 0.17). These observations were confirmed by
Beer et al.,189 who also showed that in patients with nonviable infarcts,
2.0
failing to recover regional contractile function after 6 months, 31P-spectra
showed complete absence of PCr. The same group later reported the
detection of inferior infarcts by “acquisition-weighted” 31P-MRS.190
PCr/ATP

1.5 However, loss of myocardial tissue because of necrosis and scar forma-
tion primarily leads to a reduction of both PCr and ATP, and the extent
of viable tissue loss can therefore not be detected using the PCr/ATP
1.0 ratio. Instead, measurement of absolute concentrations of high-energy
phosphates is necessary. Yabe et al.93 showed that absolute myocardial
ATP content was significantly reduced in patients with fixed thallium
defects (nonviable), but was unchanged in patients with reversible defects
0.5
(viable myocardium). Bottomley et al.191 also reported that the CK flux
Rest Exercise Recovery drops in infarcted tissue. Although these initial results are promising,
FIG. 9.10  Phosphocreatine/adenosine triphosphate (PCr/ATP) ratios. At substantial improvement in spatial resolution is necessary if 31P-MRS
rest, during handgrip exercise, and during recovery in controls and in evaluation of viability is to become competitive with current imaging
patients with stenosis of the left anterior descending coronary artery. methods of assessment.192
PCr/ATP decreased in patients but not in healthy volunteers. ATP,
Viability detection may also be feasible by measuring total creatine
Adenosine triphosphate; PCr, phosphocreatine. (From Weiss RG, Bot-
tomley PA, Hardy CJ, Gerstenblith G. Regional myocardial metabolism
content by localized 1H-MRS48–51 or potentially by CrCEST imaging,52
of high-energy phosphates during isometric-exercise in patients with because creatine concentrations in scar tissue are negligible. Due to the
coronary-artery disease. N Engl J Med. 1990;323:1593–1600.) higher MR sensitivity of 1H and because the concentration of CH3-
creatine protons is ~10-fold higher than 31P-concentrations of ATP, the
resolution of 1H-MRS is superior to 31P-MRS, currently at ~1 mL vs.
in seven subjects with coronary artery disease but did not observe sig- ~70 mL. However, several methodologic hurdles (see earlier) remain
nificant changes in energetics,177 in contrast to successful results in to be overcome before clinical 1H-MRS can be more widely applied.
canine models.178 Exercise testing has also been performed at 3 T.116,179 Because the myocardial 23Na signal is elevated in both acute necrosis
With a 31P-MRS stress test, we would also be able to test the efficacy of and in chronic scar (see above), 23Na-CMR may allow detection of
revascularization procedures or of established or new antianginal medi- myocardial viability without the use of external contrast agents. Due
cation. It is conceivable that a “PCr threshold” may become a clinically to 100% natural abundance, relatively high MR sensitivity (9.3% of
1
relevant parameter as the level of exercise achievable without a decrease H) and tissue concentration (~140 mM extracellular, ~10 mM intra-
of myocardial PCr concentrations. This may in the future allow objec- cellular) of 23Na, and due to its short T1 (~30 ms at 1.5 T), allowing
tive fine-tuning of antianginal therapy. for short TR, 23Na yields the second-highest CMR resolution of all
The pathophysiologic mechanisms of exercise-induced chest pain in MR-detectable nuclei. 23Na-CMR has been demonstrated with a reso-
women with a normal coronary angiogram remain unclear, but micro- lution of up to 216 µL with 100 ms cine temporal resolution during a
vascular dysfunction and subsequent tissue ischemia in the absence 19-minute scan in volunteers at 7 T,72 which is a dramatic improvement
of epicardial coronary stenoses has been postulated. Pohost’s group180 compared with 392 µL resolution (in patients193) using an ECG-triggered
showed that in 7 out of 35 women with chest pain and normal coronary 3D spoiled gradient echo sequence, in ~1 hour, at 1.5 T. In patients
arteries, the PCr/ATP ratio decreased by 29 ± 5% during handgrip exer- studied days and again more than 6 months after acute MI, Sandstede
cise, providing direct evidence of exercise-induced myocardial ischemia. et al.194 obtained the first in vivo 23Na-MR images of infarcted human
In a subsequent study, they showed that, at 3-year follow-up, an initially myocardium. All patients after subacute infarction and 12 out of 15
abnormal 31P-MRS stress test was a strong predictor of future cardio- patients with chronic infarction showed an area of elevated 23Na signal
vascular events.181 31P-MRS stress testing may facilitate the development intensity that significantly correlated with wall motion abnormalities. In
and monitoring of treatment for this ubiquitous condition. a follow-up study, we demonstrated that the total Na+ signal remained
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 107

significantly elevated in scar over a time period of at least 1 year.195 studies should be able to probe some of the earlier substrate utilization
Bottomley recently summarized the status of 23Na-CMR as a potential steps. Technologies for genetic manipulation of animal models, for
clinical imaging modality.74 He identified the primary challenge for optical and electron microscopy, and computer modeling of biologic
23
Na-CMR as being a need to demonstrate unique insight beyond that systems have all advanced considerably since Bessman and Geiger pro-
available, for example, by 1H-CMR extracellular volume mapping.196 posed the compartmentalized creatine kinase shuttle18 (reviewed in
Thus 31P-MRS, 1H-MRS, and 23Na-CMR/MRS provide an array of references208–211).
methods that might in the future become the preferred approach for Several recent experiments are challenging our understanding
viability assessment, if substantially higher spatial resolution can be of the role of the CK shuttle in the heart. For example, genetically
achieved. Unlike currently used techniques, such as dobutamine stress manipulated mouse models with selective knockout212–215 or overex-
echocardiography, nuclear imaging, or delayed enhancement CMR, the pression216 of the various components of energy metabolism have
MRS approach provides intrinsic contrast for distinction between viable demonstrated the crucial role of cardiac energetics in normal and
and nonviable myocardium, is radiation-free, does not require intra- failing heart.
venous agents, and can avoid stress testing. Bottomley et al.217 performed diffusion-weighted 31P spectroscopy
confirming a b-value for PCr consistent with the CK shuttle hypothesis
Hypoxia/Altitude in skeletal muscle.
Volunteers exposed to normobaric hypoxia (80% O2 saturation) for 20 However, studies using a combination of high-resolution 3D electron
hours showed PCr/ATP of 1.7 versus 2.0 before exposure (a 15% reduc- microscopy, optical fluorescence microscopy, and focused ultraviolet
tion).197 A group of 14 healthy volunteers scanned 4 days after a 17-day UV laser destruction of mitochondria are lending fresh impetus to
trek to Mount Everest Base Camp (5300 m) showed an 18% (P < .01) the notion of a mitochondrial reticulum218–221 capable of delivering
reduction in cardiac PCr/ATP, which suggests that reduced cardiac PCr/ energy to myofibrils in the form of a mitochondrial electrochemical
ATP ratios may be a general response to hypoxia, whether that be because gradient ΔΨ.
of the environment or disease.198 However, a similar study in skeletal It will be exciting in the years ahead to see how the results from
muscle observed no significant change in exercising energetics in subjects these many methods can be synthesized into an improved understand-
who had climbed Mount Everest.199 ing of cardiac energetics, and particularly the roles of creatine kinase,20,222
as a temporal and spatial energy buffer.26–28,223–225
MAGNETIC RESONANCE SPECTROSCOPY
AT 3 T AND 7 T PERSPECTIVE AND GENERAL CONCLUSIONS
The most promising route for the improvement of spatial and temporal MR spectroscopy allows for the noninvasive assessment of many aspects
resolution of MRS is the use of higher magnetic field strengths, because of cardiac metabolism in the normal and diseased heart, providing a
MRS methods are fundamentally still signal-to-noise limited. wealth of information that should, in theory, be clinically relevant for
Early attempts to increase the field strength200,201 were limited by the patient management. The main obstacle for the widespread implementa-
technical prototype nature of the magnets available at that time. From tion of cardiac MRS is its technical complexity and limited resolution,
2009, significant (~2×) gains were demonstrated for 31P-MRS at 3 T and a major technological effort is required to substantially improve
compared with 1.5 T in volunteers82,202,203 and in HCM patients.161 Proton spatial and temporal resolution. Also, to reduce measurement variability
1
H-MRS also improves significantly at 3 T.128,204 In the last decade, to less than 5%, high signal/noise spectra need to be acquired. Finally,
methods for absolute quantitation102 for measuring the creatine kinase standardized acquisition and quantification protocols will have to be
forward rate constant kf and ATP flux35,88 have also been developed. 3 T developed and agreed upon to provide clinicians with reliable mea-
31
P-MRS has been used in tens of patient studies in Oxford and else- surements, which are also comparable among different centers. These
where to date. goals should be achievable in the coming years by a major dedicated
Since 2011, we have pioneered the use of ultra-high field 7 T scan- technical development effort on coil design, sequence design, and in
ners for cardiac 31P-MRS. We have observed further 2.8× gains in SNR particular on high-field magnets and hyperpolarization methods. High-
(compared with 3 T) in volunteers,80 and a similar 2.6× gain in DCM resolution metabolic imaging would then finally become a reality in
patients.81 We have already demonstrated adiabatic excitation205 and patients with ischemic heart disease, heart failure, valve disease, and
made the first 3D-resolved cardiac creatine kinase kf measurements36 genetic cardiomyopathy.
using the 7 T system. Realizing the potential of 7 T scanners will require
significant continuing methodologic development83,205a to use the
increased SNR, while avoiding excessive RF heating, line broadening,
ACKNOWLEDGMENTS
or spatial inhomogeneity. Nevertheless, non-1H MR spectroscopy and S.N. acknowledges support from the Oxford NIHR Biomedical Research
imaging is expected to be one of the principal beneficiaries of ultra-high Centre and from the Oxford British Heart Foundation Centre of Research
field MR systems, as exemplified in Figs. 9.4 and 9.6.206,207 Excellence. C.T.R. is funded by a Sir Henry Dale Fellowship from the
Wellcome Trust and the Royal Society (098436/Z/12/Z).
MODELING STUDIES
REFERENCES
The changes seen in 31P spectra reflect the latter part of the cardiac energy
supply through the creatine kinase shuttle. Human hyperpolarized 13C A full reference list is available online at ExpertConsult.com
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 107.e1

REFERENCES 26. Wu F, Beard DA. Roles of the creatine kinase system and myoglobin in
maintaining energetic state in the working heart. BMC Syst Biol.
1. Garlick PB, Radda GK, Seeley PJ. Phosphorus NMR studies on perfused 2009;3:22.
heart. Biochem Biophys Res Commun. 1977;74:1256–1262. 27. Saks V, Schlattner U, Tokarska-Schlattner M, et al. Systems level
2. Bottomley PA. NMR spectroscopy of the human heart. In: Harris RK, regulation of cardiac energy fluxes via metabolic cycles: role of creatine,
Wasylishen RE, eds. Encyclopedia of Magnetic Resonance. Chichester: phosphotransfer pathways, and AMPK signaling. Springer Ser Biophys.
John Wiley; 2009. 2014;16:261–320.
3. Bizino MB, Hammer S, Lamb HJ. Metabolic imaging of the human 28. Weiss K, Bottomley PA, Weiss RG. On the theoretical limits of detecting
heart: clinical application of magnetic resonance spectroscopy. Heart. cyclic changes in cardiac high-energy phosphates and creatine kinase
2014;100:881–890. reaction kinetics using in vivo P-31 MRS. NMR Biomed.
4. van Ewijk PA, Schrauwen-Hinderling VB, Bekkers SC, Glatz JF, 2015;28:694–705.
Wildberger JE, Kooi ME. MRS: a noninvasive window into cardiac 29. Clarke K, O’Connor AJ, Willis RJ. Temporal relation between energy-
metabolism. NMR Biomed. 2015;28:747–766. metabolism and myocardial-function during ischemia and reperfusion.
5. Holloway CJ, Suttie J, Dass S, Neubauer S. Clinical cardiac magnetic Am J Physiol. 1987;253:H412–H421.
resonance spectroscopy. Prog Cardiovasc Dis. 2011;54:320–327. 30. de Groot MC, Illing B, Horn M, et al. Endothelin-1 increases
6. Hudsmith LE, Neubauer S. Magnetic resonance spectroscopy in susceptibility of isolated rat hearts to ischemia/reperfusion injury by
myocardial disease. JACC Cardiovasc Imaging. 2009;2:87–96. reducing coronary flow. J Mol Cell Cardiol. 1998;30:2657–2668.
7. Westbrook C. MRI at a Glance. Oxford: Blackwell Science; 2002. 31. Clarke K, O’Connor AJ, Willis RJ. Temporal relation between energy
8. Gadian DG. NMR and Its Applications to Living Systems. 2nd ed. New metabolism and myocardial function during ischemia and reperfusion.
York: Oxford University Press; 1995. Am J Physiol. 1987;253:H412–H421.
9. De Graaf RA. Vivo NMR Spectroscopy: Principles and Techniques. 2nd ed. 32. Schroeder MA, Clarke K, Neubauer S, Tyler DJ. Hyperpolarized
Chichester: John Wiley & Sons; 2007. magnetic resonance a novel technique for the in vivo assessment of
10. Bracewell RN. The Fourier Transform and Its Applications. 3rd ed. cardiovascular disease. Circulation. 2011;124:1580–1594.
Boston: WCB/McGraw-Hill; 2000. 33. Rider OJ, Tyler DJ. Clinical implications of cardiac hyperpolarized
11. Ernst RR, Bodenhausen G, Wokaun A. Principles of Nuclear Magnetic magnetic resonance imaging. J Cardiovasc Magn Reson. 2013;15.
Resonance in One and Two Dimensions. Oxford: Clarendon Press; 1987. 34. Neubauer S, Horn M, Naumann A, et al. Impairment of energy
12. Vanhamme L, van den Boogaart A, Van Huffel S. Improved method for metabolism in intact residual myocardium of rat hearts with chronic
accurate and efficient quantification of MRS data with use of prior myocardial infarction. J Clin Invest. 1995;95:1092–1100.
knowledge. J Magn Reson. 1997;129:35–43. 35. Schar M, El-Sharkawy AMM, Weiss RG, Bottomley PA. Triple repetition
13. Provencher SW. Estimation of metabolite concentrations from localized time saturation transfer (TRiST) P-31 spectroscopy for measuring human
in-vivo proton NMR-spectra. Magn Reson Med. 1993;30:672–679. creatine kinase reaction kinetics. Magn Reson Med. 2010;63:1493–1501.
14. Levitt MH. Spin Dynamics: Basics of Nuclear Magnetic Resonance. 2nd 36. Clarke WT, Robson MD, Rodgers CT. 3D resolved human cardiac
ed. Chichester, England; Hoboken, NJ: John Wiley & Sons; 2008. creatine kinase rate from 31P-MRS at 7T. Paper presented at: ISMRM
15. El-Sharkawy A-MM, Gabr RE, Schär M, Weiss RG, Bottomley PA. 24th Annual Meeting. Singapore: International Society for Magnetic
Quantification of human high-energy phosphate metabolite Resonance in Medicine; 2016.
concentrations at 3 T with partial volume and sensitivity corrections. 37. Bittl JA, Ingwall JS. Reaction rates of creatine kinase and ATP synthesis
NMR Biomed. 2013;26:1363–1371. in the isolated rat heart. A 31P NMR magnetization transfer study. J Biol
16. Ingwall JS. Phosphorus nuclear magnetic resonance spectroscopy of Chem. 1985;260:3512–3517.
cardiac and skeletal muscles. Am J Physiol. 1982;242:H729–H744. 38. Neubauer S, Hamman BL, Perry SB, Bittl JA, Ingwall JS. Velocity of the
17. Clarke K, Stewart LC, Neubauer S, et al. Extracellular volume and creatine kinase reaction decreases in postischemic myocardium: a
transsarcolemmal proton movement during ischemia and reperfusion: a 31P-NMR magnetization transfer study of the isolated ferret heart. Circ
31P NMR spectroscopic study of the isovolumic rat heart. NMR Biomed. Res. 1988;63:1–15.
1993;6:278–286. 39. Kemp GJ, Brindle KM. What do magnetic resonance-based
18. Bessman SP, Geiger PJ. Transport of energy in muscle: the measurements of Pi–>ATP flux tell us about skeletal muscle
phosphorylcreatine shuttle. Science. 1981;211:448–452. metabolism? Diabetes. 2012;61:1927–1934.
19. van Dorsten FA, Reese T, Gellerich JF, et al. Fluxes through cytosolic and 40. Laurent D. The interpretation of abnormal 31P magnetic resonance
mitochondrial creatine kinase, measured by P-31 NMR. Mol Cell saturation transfer measurements of Pi/ATP exchange in insulin-
Biochem. 1997;174:33–42. resistant skeletal muscle. Am J Physiol Endocrinol Metab. 2008;294:E640–
20. Greenhaff PL. The creatine-phosphocreatine system: there’s more than E642; author reply E643–E644.
one song in its repertoire. J Physiol (London). 2001;537:657. 41. Nascimben L, Friedrich J, Liao R, Pauletto P, Pessina AC, Ingwall JS.
21. Neubauer S. Cardiac magnetic resonance spectroscopy. In: Lardo A, Enalapril treatment increases cardiac performance and energy reserve
Fayad Z, Chronos N, Fuster V, eds. Cardiovascular Magnetic Resonance via the creatine kinase reaction in myocardium of Syrian myopathic
Established and Emerging Applications. London: Martin Dunitz; hamsters with advanced heart failure. Circulation. 1995;91:1824–1833.
2003:39–60. 42. Liao R, Nascimben L, Friedrich J, Gwathmey JK, Ingwall JS. Decreased
22. Iotti S, Frassineti C, Alderighi L, Sabatini A, Vacca A, Barbiroli B. In vivo energy reserve in an animal model of dilated cardiomyopathy.
assessment of free magnesium concentration in human brain by P-31 Relationship to contractile performance. Circ Res. 1996;78:893–902.
MRS. A new calibration curve based on a mathematical algorithm. NMR 43. Zhang J, Wilke N, Wang Y, et al. Functional and bioenergetic
Biomed. 1996;9:24–32. consequences of postinfarction left ventricular remodeling in a new
23. Iotti S, Frassineti C, Alderighi L, Sabatini A, Vacca A, Barbiroli B. In vivo porcine model. MRI and 31 P-MRS study. Circulation. 1996;94:
P-31-MRS assessment of cytosolic [Mg2+] in the human skeletal muscle 1089–1100.
in different metabolic conditions. Magn Reson Imaging. 2000;18: 44. Bottomley PA, Panjrath GS, Lai S, et al. Metabolic rates of ATP transfer
607–614. through creatine kinase (CK Flux) predict clinical heart failure events
24. Balaban RS, Kantor HL, Katz LA, Briggs RW. Relation between work and and death. Sci Transl Med. 2013;5:215re3.
phosphate metabolite in the in vivo paced mammalian heart. Science. 45. Ingwall JS. Is cardiac failure a consequence of decreased energy reserve?
1986;232:1121–1123. Circulation. 1993;87(suppl VII):58–62.
25. Zhang J, Duncker DJ, Xu Y, et al. Transmural bioenergetic responses of 46. Ugurbil K, Petein M, Madian R, Michurski S, Cohn JN, From AH. High
normal myocardium to high workstates. Am J Physiol. 1995;268: resolution proton NMR studies of perfused rat hearts. FEBS Lett.
H1891–H1905. 1984;167:73–78.
107.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

47. Balschi JA, Hai JO, Wolkowicz PE, et al. 1H NMR measurement of 70. Ouwerkerk R, Bottomley PA, Solaiyappan M, et al. Tissue sodium
triacylglycerol accumulation in the post-ischemic canine heart after concentration in myocardial infarction in humans: a quantitative Na-23
transient increase of plasma lipids. J Mol Cell Cardiol. 1997;29:471–480. MR imaging study. Radiology. 2008;248:88–96.
48. Bottomley PA, Weiss RG. Noninvasive localized MR quantification of 71. Graessl A, Ruehle A, Waiczies H, et al. Sodium MRI of the human heart
creatine kinase metabolites in normal and infarcted canine myocardium. at 7.0T: preliminary results. NMR Biomed. 2015;28:967–975.
Radiology. 2001;219:411–418. 72. Resetar A, Hoffmann SH, Graessl A, et al. Retrospectively-gated CINE
49. Schneider J, Fekete E, Weisser A, Neubauer S, von Kienlin M. Reduced Na-23 imaging of the heart at 7.0 tesla using density-adapted 3D
(1)H-NMR visibility of creatine in isolated rat hearts. Magn Reson Med. projection reconstruction. Magn Reson Imaging. 2015;33:1091–1097.
2000;43:497–502. 73. Platt T, Fiedler TM, Nagel AM, et al. Development and application of a
50. Bottomley PA, Weiss RG. Non-invasive magnetic-resonance detection of 23Na elliptical body coil for 7 tesla. Paper presented at: ISMRM 24th
creatine depletion in non-viable infarcted myocardium. Lancet. Annual Meeting. Singapore: International Society for Magnetic
1998;351:714–718. Resonance in Medicine; 2016.
51. Weiss K, Martini N, Boesiger P, Kozerke S. Metabolic MR imaging of 74. Bottomley PA. Sodium MRI in human heart: a review. NMR Biomed.
regional triglyceride and creatine content in the human heart. Magn 2016;29:187–196.
Reson Med. 2012;68:1696–1704. 75. Umathum R, Rosler MB, Nagel AM. In vivo 39K MR imaging of human
52. Haris M, Singh A, Cai KJ, et al. A technique for in vivo mapping of muscle and brain. Radiology. 2013;269:569–576.
myocardial creatine kinase metabolism. Nat Med. 2014;20:209–214. 76. Rosler MB, Nagel AM, Umathum R, Bachert P, Benkhedah N. In vivo
53. Weiss RG, Gerstenblith G, Bottomley PA. ATP flux through creatine observation of quadrupolar splitting in K magnetic resonance
kinase in the normal, stressed, and failing human heart. Proc Natl Acad spectroscopy of human muscle tissue. NMR Biomed. 2016.
Sci USA. 2005;102:808–813. 77. Freitag MT, Breithaupt M, Berger M, et al. In vivo visualization of
54. Hirsch GA, Bottomley PA, Gerstenblith G, Weiss RG. Allopurinol acutely mesoscopic anatomy of healthy and pathological lymph nodes using 7T
increases adenosine triphospate energy delivery in failing human hearts. MRI: a feasibility study. J Magn Reson Imaging. 2015;41:1405–1412.
J Am Coll Cardiol. 2012;59:802–808. 78. Nagel AM, Lehmann-Horn F, Weber MA, et al. In vivo 35Cl MR
55. Kreutzer U, Mekhamer Y, Chung Y, Jue T. Oxygen supply and oxidative imaging in humans: a feasibility study. Radiology. 2014;271:585–595.
phosphorylation limitation in rat myocardium in situ. Am J Physiol 79. Schar M, Kozerke S, Boesiger P. Navigator gating and volume tracking
Heart Circ Physiol. 2001;280:H2030–H2037. for double-triggered cardiac proton spectroscopy at 3 tesla. Magn Reson
56. de Heer P, Bizino MB, Lamb HJ, Webb AG. Parameter optimization for Med. 2004;51:1091–1095.
reproducible cardiac 1H-MR spectroscopy at 3 tesla. J Magn Reson 80. Rodgers CT, Clarke WT, Snyder C, Vaughan JT, Neubauer S, Robson
Imaging. 2016;44:1151–1158. MD. Human cardiac 31P magnetic resonance spectroscopy at 7 tesla.
57. Fuetterer M, Stoeck CT, Kozerke S. Second-order motion compensated Magn Reson Med. 2014;72:304–315.
PRESS for cardiac spectroscopy. Magn Reson Med. 2017;77:57–64. 81. Stoll VM, Clarke WT, Levelt E, et al. Dilated cardiomyopathy:
58. Malloy CR, Jones JG, Jeffrey FM, Jessen ME, Sherry AD. Contribution of phosphorus 31 MR spectroscopy at 7 T. Radiology. 2016;281:409–417.
various substrates to total citric acid cycle flux and anaplerosis as 82. Tyler DJ, Emmanuel Y, Cochlin LE, et al. Reproducibility of 31P cardiac
determined by 13C isotopomer analysis and O2 consumption in the magnetic resonance spectroscopy at 3T. NMR Biomed. 2009;22:405–413.
heart. MAGMA. 1996;4:35–46. 83. Schaller B, Clarke WT, Neubauer S, Robson MD, Rodgers CT.
59. Lewandowski ED. Cardiac carbon 13 magnetic resonance spectroscopy: Suppression of skeletal muscle signal using a crusher coil: a human
on the horizon or over the rainbow? J Nucl Cardiol. 2002;9:419–428. cardiac 31p-MR spectroscopy study at 7 tesla. Magn Reson Med.
60. Weiss RG. 13C-NMR for the study of intermediary metabolism. 2016;75:962–972.
MAGMA. 1998;6:132. 84. Bottomley PA. MR spectroscopy of the human heart: the status and the
61. Atherton H, Schroeder M, Dodd M, et al. Real-time assessment of krebs challenges. Radiology. 1994;191:593–612.
cycle metabolism with hyperpolarised [2-13c]pyruvate. Heart. 2010;96. 85. Bottomley PA. Noninvasive study of high-energy phosphate metabolism
62. Dodd M, Atherton H, Schroeder M, et al. Investigating metabolic flux in in human heart by depth-resolved 31P NMR spectroscopy. Science.
the hyperthyroid heart using hyperpolarised magnetic resonance. Heart. 1985;229:769–772.
2010;96. 86. Bottomley PA, Hardy CJ, Roemer PB. Phosphate metabolite imaging and
63. Yoshihara HA, Bastiaansen JA, Berthonneche C, Comment A, Schwitter concentration measurements in human heart by nuclear magnetic
J. An intact small animal model of myocardial ischemia-reperfusion: resonance. Magn Reson Med. 1990;14:425–434.
characterization of metabolic changes by hyperpolarized 13C MR 87. Bottomley PA, Atalar E, Weiss RG. Human cardiac high-energy
spectroscopy. Am J Physiol Heart Circ Physiol. 2015;309:H2058–H2066. phosphate metabolite concentrations by 1D-resolved NMR spectroscopy.
64. Kohler SJ, Perry SB, Stewart LC, Atkinson DE, Clarke K, Ingwall JS. Magn Reson Med. 1996;35:664–670.
Analysis of 23Na NMR spectra from isolated perfused hearts. Magn 87a. Valkovič L, Clarke WT, Purvis LAB, Robson MD, Neubauer S, Rodgers
Reson Med. 1991;18:15–27. CT. Measurement of human cardiac intracellular pH in vivo using long
65. Van Emous JG, Vleggeert-Lankamp CL, Nederhoff MG, Ruigrok TJ, Van TR 31P-MRS with adiabatic excitation at 7T. In: International Society for
Echteld CJ. Postischemic Na(+)-K(+)-ATPase reactivation is delayed in Magnetic Resonance in Medicine 25th Annual Meeting; 2017; Abstract #2933.
the absence of glycolytic ATP in isolated rat hearts. Am J Physiol Heart 88. Schar M, Gabr RE, El-Sharkawy AM, Steinberg A, Bottomley PA, Weiss
Circ Physiol. 2001;280:H2189–H2195. RG. Two repetition time saturation transfer (TwiST) with spill-over
66. Kim RJ, Lima JAC, Chen EL, et al. Fast 23Na magnetic resonance correction to measure creatine kinase reaction rates in human hearts. J
imaging of acute reperfused myocardial infarction. Potential to assess Cardiovasc Magn Reson. 2015;17.
myocardial viability. Circulation. 1997;95:1877–1885. 89. Ouwerkerk R, Bottomley PA. On neglecting chemical exchange effects
67. Horn M, Weidensteiner C, Scheffer H, et al. Detection of myocardial when correcting in vivo P-31 MRS data for partial saturation. J Magn
viability based on measurement of sodium content: a (23)Na-NMR Reson. 2001;148:425–435.
study. Magn Reson Med. 2001;45:756–764. 90. Neubauer S, Krahe T, Schindler R, et al. 31P magnetic resonance
68. Pabst T, Sandstede J, Beer M, Kenn W, Neubauer S, Hahn D. Sodium spectroscopy in dilated cardiomyopathy and coronary artery disease.
T-2* relaxation times in human heart muscle. J Magn Reson Imaging. Altered cardiac high-energy phosphate metabolism in heart failure.
2002;15:215–218. Circulation. 1992;86:1810–1818.
69. Ouwerkerk R, Weiss RG, Bottomley PA. Measuring human cardiac tissue 91. Horn M, Neubauer S, Bomhard M, Kadgien M, Schnackerz K, Ertl G.
sodium concentrations using surface coils, adiabatic excitation, and 31P-NMR spectroscopy of human blood and serum: results from
twisted projection imaging with minimal T-2 losses. J Magn Reson volunteers and patients with congestive heart failure, diabetes mellitus
Imaging. 2005;21:546–555. and hyperlipidemia. MAGMA. 1993;1:55–60.
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 107.e3

92. Beer M, Sandstede J, Landschütz W, et al. Absolute concentrations of of spontaneously hypertensive rats. Heart Vessels. 2000;15:
myocardial high-energy phosphate metabolites in normal, hypertrophied 197–202.
and failing human myocardium, measured non-invasively with 112. Pluim BM, Lamb HJ, Kayser HW, et al. Functional and metabolic
31P-SLOOP-magnetic resonance spectroscopy. J Am Coll Cardiol. evaluation of the athlete’s heart by magnetic resonance imaging and
2002;40:1267–1274. dobutamine stress magnetic resonance spectroscopy. Circulation.
93. Yabe T, Mitsunami K, Inubushi T, Kinoshita M. Quantitative 1998;97:666–672.
measurements of cardiac phosphorus metabolites in coronary artery 113. Spencer RG, Buttrick PM, Ingwall JS. Function and bioenergetics in
disease by 31P magnetic resonance spectroscopy [see comments]. isolated perfused trained rat hearts. Am J Physiol. 1997;272:H409–H417.
Circulation. 1995;92:15–23. 114. Scheuermann-Freestone M, Madsen PL, Manners D, et al. Abnormal
94. Bottomley PA, Hardy CJ. Rapid, Reliable in vivo assays of human cardiac and skeletal muscle energy metabolism in patients with type 2
phosphate metabolites by nuclear magnetic resonance. Clin Chem. diabetes. Circulation. 2003;107:3040–3046.
1989;35(3). 115. Diamant M, Lamb HJ, Groenevelt Y, et al. Diastolic dysfunction is
95. Akoka S, Barantin L, Trierweiler M. Concentration measurement by associated with altered myocardial metabolism in asymptomatic
proton NMR using the ERETIC method. Anal Chem. 1999;71:2554–2557. normotensive patients with well-controlled type II diabetes mellitus. J
96. von Kienlin M, Beer M, Greiser A, et al. Advances in human cardiac Am Coll Cardiol. 2003;42:328–335.
P-31-MR spectroscopy: SLOOP and clinical applications. J Magn Reson 116. Levelt E, Rodgers CT, Clarke WT, et al. Cardiac energetics, oxygenation,
Imaging. 2001;13:521–527. and perfusion during increased workload in patients with type 2
97. Zhang Y, Gabr RE, Schar M, Weiss RG, Bottomley PA. Magnetic diabetes mellitus. Eur Heart J. 2016;37:3461–3469.
resonance spectroscopy with linear algebraic modeling (SLAM) for 117. Metzler B, Schocke MF, Steinboeck P, et al. Decreased high-energy
higher speed and sensitivity. J Magn Reson. 2012;218:66–76. phosphate ratios in the myocardium of men with diabetes mellitus type
98. Zhang Y, Gabr RE, Zhou J, Weiss RG, Bottomley PA. Highly-accelerated I. J Cardiovasc Magn Reson. 2002;4:493–502.
quantitative 2D and 3D localized spectroscopy with linear algebraic 118. Shivu GN, Phan TT, Abozguia K, et al. Relationship between coronary
modeling (SLAM) and sensitivity encoding. J Magn Reson. microvascular dysfunction and cardiac energetics impairment in type 1
2013;237:125–138. diabetes mellitus. Circulation. 2010;121:1209–1215.
99. Maguire ML, Geethanath S, Lygate CA, Kodibagkar VD, Schneider JE. 119. van der Meer RW, Rijzewijk LJ, de Jong HW, et al. Pioglitazone improves
Compressed sensing to accelerate magnetic resonance spectroscopic cardiac function and alters myocardial substrate metabolism without
imaging: evaluation and application to Na-23-imaging of mouse hearts. affecting cardiac triglyceride accumulation and high-energy phosphate
J Cardiovasc Magn Reson. 2015;17. metabolism in patients with well-controlled type 2 diabetes mellitus.
100. Valkovic L, Chmelík M, Meyerspeer M, et al. Dynamic 31P MRSI with Circulation. 2009;119:2069–2077.
spiral readout for quantification of mitochondrial capacity in muscles of 120. Rider OJ, Francis JM, Tyler D, Byrne J, Clarke K, Neubauer S. Effects of
the calf during plantar flexion exercise at 7T. Paper presented at: ISMRM weight loss on myocardial energetics and diastolic function in obesity.
24th Annual Meeting. Singapore: International Society for Magnetic Int J Cardiovasc Imaging. 2013;29:1043–1050.
Resonance in Medicine; 2016. 121. Rider OJ, Francis JM, Ali MK, et al. Effects of catecholamine stress on
101. Bottomley PA, Ouwerkerk R, Lee RF, Weiss RG. Four-angle saturation diastolic function and myocardial energetics in obesity. Circulation.
transfer (FAST) method for measuring creatine kinase reaction rates in 2012;125:1511–1519.
vivo. Magn Reson Med. 2002;47:850–863. 122. Rayner JJ, Neubauer S, Rider OJ. The paradox of obesity
102. El-Sharkawy AM, Schar M, Ouwerkerk R, Weiss RG, Bottomley PA. cardiomyopathy and the potential for weight loss as a therapy. Obes Rev.
Quantitative cardiac P-31 spectroscopy at 3 tesla using adiabatic pulses. 2015;16:679–690.
Magn Reson Med. 2009;61:785–795. 123. Ormerod JOM, Ashrafian H, Frenneaux MP. Impaired energetics in
103. Meininger M, Landschutz W, Beer M, et al. Concentrations of human heart failure—a new therapeutic target. Pharmacol Ther. 2008;119:
cardiac phosphorus metabolites determined by SLOOP 31P NMR 264–274.
spectroscopy. Magn Reson Med. 1999;41:657–663. 124. Hamilton G, Middleton MS, Bydder M, et al. Effect of PRESS and
104. Okada M, Mitsunami K, Inubushi T, Kinoshita M. Influence of aging or STEAM sequences on magnetic resonance spectroscopic liver fat
left ventricular hypertrophy on the human heart: contents of quantification. J Magn Reson Imaging. 2009;30:145–152.
phosphorus metabolites measured by 31P MRS. Magn Reson Med. 125. Hamilton G, Yokoo T, Bydder M, et al. In vivo characterization of the
1998;39:772–782. liver fat (1)H MR spectrum. NMR Biomed. 2011;24:784–790.
105. Schocke MF, Metzler B, Wolf C, et al. Impact of aging on cardiac 126. Reeder SB, Cruite I, Hamilton G, Sirlin CB. Quantitative assessment of
high-energy phosphate metabolism determined by phosphorus-31 liver fat with magnetic resonance imaging and spectroscopy. J Magn
2-dimensional chemical shift imaging (31P 2D CSI). Magn Reson Reson Imaging. 2011;34:729–749.
Imaging. 2003;21:553–559. 127. Hamilton G, Middleton MS, Hooker JC, et al. In vivo breath-hold (1) H
106. Cardellach F, Galofre J, Cusso R, Urbano-Marquez A. Decline in skeletal MRS simultaneous estimation of liver proton density fat fraction, and
muscle mitochondrial respiration chain function with ageing. Lancet. T1 and T2 of water and fat, with a multi-TR, multi-TE sequence. J
1989;2:44–45. Magn Reson Imaging. 2015;42:1538–1543.
107. Esterhammer R, Klug G, Wolf C, et al. Cardiac high-energy phosphate 128. Rial B, Robson MD, Neubauer S, Schneider JE. Rapid quantification of
metabolism alters with age as studied in 196 healthy males with the help myocardial lipid content in humans using single breath-hold H-1 MRS
of 31-phosphorus 2-dimensional chemical shift imaging. PLoS One. at 3 tesla. Magn Reson Med. 2011;66:619–624.
2014;9:e97368. 129. van der Meer RW, Hammer S, Lamb HJ, et al. Effects of short-term
108. Wallace DC. Mitochondrial diseases in man and mouse. Science. high-fat, high-energy diet on hepatic and myocardial triglyceride
1999;283:1482–1488. content in healthy men. J Clin Endocrinol Metab. 2008;93:2702–2708.
109. Pluim BM, Lamb HJ, Kayser HW, et al. Functional and metabolic 130. Holloway CJ, Cochlin LE, Emmanuel Y, et al. A high-fat diet impairs
evaluation of the athlete’s heart by magnetic resonance imaging and cardiac high-energy phosphate metabolism and cognitive function in
dobutamine stress magnetic resonance spectroscopy. Circulation. healthy human subjects. Am J Clin Nutr. 2011;93:748–755.
1998;97:666–672. 131. Levelt E, Mahmod M, Piechnik SK, et al. Relationship between left
110. Lamb HJ, Beyerbacht HP, van der Laarse A, et al. Diastolic dysfunction ventricular structural and metabolic remodeling in type 2 diabetes.
in hypertensive heart disease is associated with altered myocardial Diabetes. 2016;65:44–52.
metabolism. Circulation. 1999;99:2261–2267. 132. Schaefer S, Gober JR, Schwartz GG, Twieg DB, Weiner MW, Massie B. In
111. Perings SM, Schulze K, Decking U, Kelm M, Strauer BE. Age-related vivo phosphorus-31 spectroscopic imaging in patients with global
decline of PCr/ATP-ratio in progressively hypertrophied hearts myocardial disease. Am J Cardiol. 1990;65:1154–1161.
107.e4 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

133. Auffermann W, Chew WM, Wolfe CL, et al. Normal and diffusely 153. Ashrafian H, Redwood C, Blair E, Watkins H. Hypertrophic
abnormal myocardium in humans: functional and metabolic cardiomyopathy: a paradigm for myocardial energy depletion. Trends
characterization with P-31 MR spectroscopy and cine MR imaging. Genet. 2003;19:263–268.
Radiology. 1991;179:253–259. 154. Spindler M, Saupe KW, Christe ME, Seidman CE, Seidman JG, Ingwall
134. de Roos A, Doornbos J, Luyten PR, Oosterwaal LJ, van der Wall EE, den JS. A murine model of familial hypertrophic cardiomyopathy shows a
Hollander JA. Cardiac metabolism in patients with dilated and markedly impaired response to inotropic stimulation (Abstract).
hypertrophic cardiomyopathy: assessment with proton-decoupled P-31 Circulation. 1996;94(8 suppl I):I-433.
MR spectroscopy. J Magn Reson Imaging. 1992;2:711–719. 155. Sakuma H, Takeda K, Tagami T, et al. 31P MR spectroscopy in
135. Hardy CJ, Weiss RG, Bottomley PA, Gerstenblith G. Altered myocardial hypertrophic cardiomyopathy: comparison with Tl-201 myocardial
high-energy phosphate metabolites in patients with dilated perfusion imaging. Am Heart J. 1993;125:1323–1328.
cardiomyopathy. Am Heart J. 1991;122:795–801. 156. Sieverding L, Jung WI, Breuer J, et al. Proton-decoupled myocardial 31P
136. Neubauer S, Horn M, Pabst T, et al. Contributions of 31P-magnetic NMR spectroscopy reveals decreased PCr/Pi in patients with severe
resonance spectroscopy to the understanding of dilated heart muscle hypertrophic cardiomyopathy. Am J Cardiol. 1997;80(3A):34A–40A.
disease. Eur Heart J. 1995;16(suppl O):115–118. 157. Crilley JG, Boehm EA, Blair E, et al. Hypertrophic cardiomyopathy due
137. Leme AM, Salemi VM, Parga JR, et al. Evaluation of the metabolism of to sarcomeric gene mutations is characterized by impaired energy
high energy phosphates in patients with Chagas’ disease. Arq Bras metabolism irrespective of the degree of hypertrophy. J Am Coll Cardiol.
Cardiol. 2010;95:264–270. 2003;41:1776–1782.
138. Betim Paes Leme AM, Salemi VM, Weiss RG, et al. Exercise-induced 158. Jung WI, Sieverding L, Breuer J, et al. 31P NMR spectroscopy detects
decrease in myocardial high-energy phosphate metabolites in patients metabolic abnormalities in asymptomatic patients with hypertrophic
with Chagas heart disease. J Card Fail. 2013;19:454–460. cardiomyopathy. Circulation. 1998;97:2536–2542.
139. Neubauer S, Horn M, Cramer M, et al. Myocardial phosphocreatine-to- 159. Jung WI, Hoess T, Bunse M, et al. Differences in cardiac energetics
ATP ratio is a predictor of mortality in patients with dilated between patients with familial and nonfamilial hypertrophic
cardiomyopathy. Circulation. 1997;96:2190–2196. cardiomyopathy. Circulation. 2000;101:E121.
140. Shen W, Asai K, Uechi M, et al. Progressive loss of myocardial ATP due 160. Abraham MR, Bottomley PA, Dimaano VL, et al. Creatine kinase
to a loss of total purines during the development of heart failure in adenosine triphosphate and phosphocreatine energy supply in a single
dogs: a compensatory role for the parallel loss of creatine. Circulation. kindred of patients with hypertrophic cardiomyopathy. Am J Cardiol.
1999;100:2113–2118. 2013;112:861–866.
141. Beer M, Seyfarth T, Sandstede J, et al. Absolute concentrations of 161. Shivu GN, Abozguia K, Phan TT, Ahmed I, Henning A, Frenneaux M.
high-energy phosphate metabolites in normal, hypertrophied, and P-31 magnetic resonance spectroscopy to measure in vivo cardiac
failing human myocardium measured noninvasively with (31)P-SLOOP energetics in normal myocardium and hypertrophic cardiomyopathy:
magnetic resonance spectroscopy. J Am Coll Cardiol. 2002;40:1267–1274. experiences at 3T. Eur J Radiol. 2010;73:255–259.
142. Beer M, Wagner D, Myers J, et al. Effects of exercise training on 162. Crilley JG, Boehm EA, Blair E, et al. Hypertrophic cardiomyopathy due
myocardial energy metabolism and ventricular function assessed by to sarcomeric gene mutations is characterized by impaired energy
quantitative phosphorus-31 magnetic resonance spectroscopy and metabolism irrespective of the degree of hypertrophy. J Am Coll Cardiol.
magnetic resonance imaging in dilated cardiomyopathy. J Am Coll 2003;41:1776–1782.
Cardiol. 2008;51:1883–1891. 163. Dass S, Cochlin LE, Suttie JJ, et al. Exacerbation of cardiac energetic
143. Holloway CJ, Dass S, Suttie JJ, et al. Exercise training in dilated impairment during exercise in hypertrophic cardiomyopathy: a potential
cardiomyopathy improves rest and stress cardiac function without mechanism for diastolic dysfunction. Eur Heart J. 2015;36:1547–1554.
changes in cardiac high energy phosphate metabolism. Heart. 164. Abozguia K, Elliott P, McKenna W, et al. Metabolic modulator perhexiline
2012;98:1083–1090. corrects energy deficiency and improves exercise capacity in symptomatic
144. Nakae I, Mitsunami K, Omura T, et al. Proton magnetic resonance hypertrophic cardiomyopathy. Circulation. 2010;122:1562–1569.
spectroscopy can detect creatine depletion associated with the progression 165. Crilley JG, Boehm EA, Rajagopalan B, et al. Magnetic resonance
of heart failure in cardiomyopathy. J Am Coll Cardiol. 2003;42:1587–1593. spectroscopy evidence of abnormal cardiac energetics in Xp21 muscular
145. Smith CS, Bottomley PA, Schulman SP, Gerstenblith G, Weiss RG. dystrophy. J Am Coll Cardiol. 2000;36(6):1953–1958.
Altered creatine kinase adenosine triphosphate kinetics in failing 166. Schocke MF, Zoller H, Vogel W, et al. Cardiac phosphorus-31 two-
hypertrophied human myocardium. Circulation. 2006;114:1151–1158. dimensional chemical shift imaging in patients with hereditary
146. Lodi R, Hart PE, Rajagopalan B, et al. Antioxidant treatment improves hemochromatosis. Magn Reson Imaging. 2004;22:515–521.
in vivo cardiac and skeletal muscle bioenergetics in patients with 167. Schocke MF, Martinek M, Kremser C, et al. 3-Hydroxy-3-methylglutaryl
Friedreich’s ataxia. Ann Neurol. 2001;49(5):590–596. coenzyme A reductase inhibitors improve myocardial high-energy
147. Givertz MM, Anstrom KJ, Redfield MM, et al. Effects of xanthine phosphate metabolism in men. J Cardiovasc Magn Reson.
oxidase inhibition in hyperuricemic heart failure patients: the Xanthine 2003;5:595–602.
Oxidase Inhibition for Hyperuricemic Heart Failure Patients 168. Machann W, Breunig F, Weidemann F, et al. Cardiac energy metabolism
(EXACT-HF) study. Circulation. 2015;131:1763–1771. is disturbed in Fabry disease and improves with enzyme replacement
148. Munzel T, Gori T, Keaney JF Jr, Maack C, Daiber A. Pathophysiological therapy using recombinant human galactosidase A. Eur J Heart Fail.
role of oxidative stress in systolic and diastolic heart failure and its 2011;13:278–283.
therapeutic implications. Eur Heart J. 2015;36:2555–2564. 169. Zhang J, Merkle H, Hendrich K, et al. Bioenergetic abnormalities
149. Tamariz L, Hare JM. Xanthine oxidase inhibitors in heart failure: where associated with severe left ventricular hypertrophy. J Clin Invest.
do we go from here? Circulation. 2015;131:1741–1744. 1993;92:993–1003.
150. Lee L, Campbell R, Scheuermann-Freestone M, et al. Metabolic 170. Conway MA, Allis J, Ouwerkerk R, Niioka T, Rajagopalan B, Radda GK.
modulation with perhexiline in chronic heart failure—a randomized, Detection of low phosphocreatine to ATP ratio in failing hypertrophied
controlled trial of short-term use of a novel treatment. Circulation. human myocardium by 31P magnetic resonance spectroscopy. Lancet.
2005;112:3280–3288. 1991;338:973–976.
151. Beadle RM, Williams LK, Abozguia K, et al. Metabolic manipulation in 171. Neubauer S, Horn M, Pabst T, et al. Cardiac high-energy phosphate
chronic heart failure: study protocol for a randomised controlled trial. metabolism in patients with aortic valve disease assessed by
Trials. 2011;12. 31P-magnetic resonance spectroscopy. J Investig Med. 1997;45:453–462.
152. Beadle RM, Williams LK, Kuehl M, et al. Improvement in cardiac 172. Beyerbacht HP, Lamb HJ, van Der Laarse A, et al. Aortic valve
energetics by perhexiline in heart failure due to dilated cardiomyopathy. replacement in patients with aortic valve stenosis improves myocardial
Jacc Heart Fail. 2015;3:202–211. metabolism and diastolic function. Radiology. 2001;219:637–643.
CHAPTER 9  Cardiac Magnetic Resonance Spectroscopy 107.e5

173. Mahmod M, Francis JM, Pal N, et al. Myocardial perfusion and 192. Sandstede JJW. Assessment of myocardial viability by MR imaging. Eur
oxygenation are impaired during stress in severe aortic stenosis and Radiol. 2003;13:52–61.
correlate with impaired energetics and subclinical left ventricular 193. Pabst T, Sandstede J, Beer M, et al. Optimization of ECG-triggered
dysfunction. J Cardiovasc Magn Reson. 2014;16. 3D (23)Na MRI of the human heart. Magn Reson Med. 2001;45:
174. Senanayake EL, Howell NJ, Ranasinghe AM, et al. Multicentre 164–166.
double-blind randomized controlled trial of perhexiline as a metabolic 194. Sandstede JJ, Pabst T, Beer M, et al. Assessment of myocardial infarction
modulator to augment myocardial protection in patients with left in humans with (23)Na MR imaging: comparison with cine MR
ventricular hypertrophy undergoing cardiac surgery. Eur J Cardiothorac imaging and delayed contrast enhancement. Radiology.
Surg. 2015;48:354–362. 2001;221:222–228.
175. Weiss RG, Bottomley PA, Hardy CJ, Gerstenblith G. Regional myocardial 195. Sandstede JJ, Hillenbrand H, Beer M, et al. Time course of 23Na signal
metabolism of high-energy phosphates during isometric-exercise in intensity after myocardial infarction in humans. Magn Reson Med.
patients with coronary-artery disease. N Engl J Med. 2004;52:545–551.
1990;323:1593–1600. 196. Ugander M, Oki AJ, Hsu LY, et al. Myocardial extracellular volume
176. Yabe T, Mitsunami K, Okada M, Morikawa S, Inubushi T, Kinoshita M. imaging by MRI quantitatively characterizes myocardial infarction and
Detection of myocardial ischemia by 31P magnetic resonance subclinical myocardial fibrosis. Circulation. 2010;122.
spectroscopy during handgrip exercise. Circulation. 1994;89:1709–1716. 197. Holloway C, Cochlin L, Codreanu I, et al. Normobaric hypoxia impairs
177. Najjar SS, Bottomley PA, Schulman SP, et al. Effects of a human cardiac energetics. FASEB J. 2011;25:3130–3135.
pharmacologically-induced shift of hemoglobin-oxygen dissociation on 198. Holloway CJ, Montgomery HE, Murray AJ, et al. Cardiac response to
myocardial energetics during ischemia in patients with coronary artery hypobaric hypoxia: persistent changes in cardiac mass, function, and
disease. J Cardiovasc Magn Reson. 2005;7:657–666. energy metabolism after a trek to Mt. Everest Base Camp. FASEB J.
178. Weiss RG, Mejia MA, Kass DA, et al. Preservation of canine myocardial 2011;25:792–796.
high-energy phosphates during low-flow ischemia with modification of 199. Edwards LM, Murray AJ, Tyler DJ, et al. The effect of high-altitude on
hemoglobin-oxygen affinity. J Clin Invest. 1999;103:739–746. human skeletal muscle energetics: P-MRS results from the Caudwell
179. Dass S, Cochlin LE, Suttie JJ, et al. Exacerbation of cardiac energetic Xtreme Everest expedition. PLoS One. 2010;5:e10681.
impairment during exercise in hypertrophic cardiomyopathy: a potential 200. Menon RS, Hendrich K. 31P NMR spectroscopy of human heart at 4T:
mechanism for diastolic dysfunction. Eur Heart J. 2015;36:1547–1554. detection of substantially uncontaminated cardiac spectra and
180. Buchthal SD, den Hollander JA, Merz CN, et al. Abnormal myocardial differentiation of subepicardium and subendocardium. Magn Reson
phosphorus-31 nuclear magnetic resonance spectroscopy in women Med. 1992;26:368–376.
with chest pain but normal coronary angiograms. N Engl J Med. 201. Hetherington HP, Luney DJ, Vaughan JT, et al. 3D 31P spectroscopic
2000;342:829–835. imaging of the human heart at 4.1 T. Magn Reson Med. 1995;33:
181. Johnson BD, Shaw LJ, Buchthal SD, et al. Prognosis in women with 427–431.
myocardial ischemia in the absence of obstructive coronary disease. 202. Tyler DJ, Hudsmith LE, Clarke K, Neubauer S, Robson MD. A
Circulation. 2004;109:2993–2999. comparison of cardiac 31P MRS at 1.5 and 3 T. NMR Biomed.
182. Thompson K, Thompson RT, Sykes J, Wisenberg G. Long-term magnetic 2008;21:793–798.
resonance imaging/spectroscopy study of cariporide in a canine cardiac 203. Rodgers CT, Cochlin LE, Tyler DJ, Neubauer S, Robson MD.
ischemia/reperfusion model. J Cardiovasc Pharmacol. 2003;41:536–543. Performance of a phased array for 31P cardiac MR spectroscopy. Paper
183. Takaoka A, Nakae I, Mitsunami K, et al. Renal ischemia/reperfusion presented at: ISMRM 24th Annual Meeting. Stockholm, Sweden:
remotely improves myocardial energy metabolism during myocardial International Society for Magnetic Resonance in Medicine; 2010.
ischemia via adenosine receptors in rabbits: effects of “remote 204. Schar M, Kozerke S, Boesiger P. Navigator gating and volume tracking
preconditioning”. J Am Coll Cardiol. 1999;33:556–564. for double-triggered cardiac proton spectroscopy at 3 tesla. Magn Reson
184. Farrall AJ, Thompson RT, Wisenberg G, Campbell CM, Drost DJ. Med. 2004;51:1091–1095.
Myocardial infarction in a canine model monitored by two-dimensional 205. Valkovič L, Clarke WT, Schaller B, et al. Adiabatic excitation for 31P
31P chemical shift spectroscopic imaging. Magn Reson Med. 1997;38: spectroscopy in the human heart at 7T. Paper presented at: ISMRM 24th
577–584. Annual Meeting. Singapore: International Society for Magnetic
185. von Kienlin M, Rosch C, Le Fur Y, et al. Three-dimensional 31P Resonance in Medicine; 2016.
magnetic resonance spectroscopic imaging of regional high-energy 205a. Valkovič L, Dragonu J, Almujayyaz S, et al. Using a whole-body 31P
phosphate metabolism in injured rat heart. Magn Reson Med. birdcage transmit coil and 16-element receive array for human cardiac
1998;39:731. metabolic imaging at 7T. PLoS One. 2017;12:e0187153.
186. Ekelof S, Rosenberg J, Jensen JS, Gogenur I. Pharmacological attenuation 206. Niendorf T, Paul K, Oezerdem C, et al. W(h)ither human cardiac and
of myocardial reperfusion injury in a closed-chest porcine model: a body magnetic resonance at ultrahigh fields? Technical advances,
systematic review. J Cardiovasc Transl Res. 2014;7:570–580. practical considerations, applications, and clinical opportunities. NMR
187. Flameng W, Vanhaecke J, Van Belle H, Borgers M, De Beer L, Minten J. Biomed. 2016;29:1173–1197.
Relation between coronary artery stenosis and myocardial purine 207. Trattnig S, Bogner W, Gruber S, et al. Clinical applications at ultrahigh
metabolism, histology and regional function in humans. J Am Coll field (7 T). Where does it make the difference? NMR Biomed.
Cardiol. 1987;9:1235–1242. 2016;29:1316–1334.
188. Kalil-Filho R, de Albuquerque CP, Weiss RG, et al. Normal high-energy 208. Wallimann T, Wyss M, Brdiczka D, Nicolay K, Eppenberger HM.
phosphate ratios in stunned human myocardium. J Am Coll Cardiol. Intracellular compartmentation, structure and function of creatine-
1997;30:1228–1232. kinase isoenzymes in tissues with high and fluctuating energy
189. Beer M, Sandstede J, Landschutz W, et al. Altered energy metabolism demands—the phosphocreatine circuit for cellular-energy homeostasis.
after myocardial infarction assessed by 31P- MR-spectroscopy in Biochem J. 1992;281:21–40.
humans. Eur Radiol. 2000;10:1323–1328. 209. Schlattner U, Tokarska-Schlattner M, Wallimann T. Mitochondrial
190. Beer M, Spindler M, Sandstede JJ, et al. Detection of myocardial creatine kinase in human health and disease. Biochim Biophys Acta.
infarctions by acquisition-weighted 31P-MR spectroscopy in humans. J 2006;1762:164–180.
Magn Reson Imaging. 2004;20:798–802. 210. Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabolism.
191. Bottomley PA, Wu KC, Gerstenblith G, Schulman SP, Steinberg A, Weiss Physiol Rev. 2000;80:1107–1213.
RG. Reduced myocardial creatine kinase flux in human myocardial 211. McClellan G, Weisberg A, Winegrad S. Energy transport from
infarction an in vivo phosphorus magnetic resonance spectroscopy mitochondria to myofibril by a creatine phosphate shuttle in cardiac
study. Circulation. 2009;119:1918–1924. cells. Am J Physiol. 1983;245:C423–C427.
107.e6 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

212. ten Hove M, Lygate CA, Fischer A, et al. Reduced inotropic reserve and 218. Glancy B, Hartnell LM, Malide D, et al. Mitochondrial reticulum for
increased susceptibility to cardiac ischemia/reperfusion injury in cellular energy distribution in muscle. Nature. 2015;523:617–620.
phosphocreatine-deficient guanidinoacetate-N-methyltransferase- 219. Amchenkova AA, Bakeeva LE, Chentsov YS, Skulachev VP, Zorov DB.
knockout mice. Circulation. 2005;111:2477–2485. Coupling membranes as energy-transmitting cables I. Filamentous
213. Saupe KW, Spindler M, Tian R, Ingwall JS. Impaired cardiac energetics mitochondria in fibroblasts and mitochondrial clusters in
in mice lacking muscle-specific isoenzymes of creatine kinase. Circ Res. cardiomyocytes. J Cell Biol. 1988;107:481–495.
1998;82:898–907. 220. Hood DA, Irrcher I, Ljubicic V, Joseph AM. Coordination of metabolic
214. Saupe KW, Spindler M, Hopkins JC, Shen W, Ingwall JS. Kinetic, plasticity in skeletal muscle. J Exp Biol. 2006;209:2265–2275.
thermodynamic, and developmental consequences of deleting creatine 221. Birkedal R, Laasmaa M, Vendelin M. The location of energetic
kinase isoenzymes from the heart. Reaction kinetics of the creatine compartments affects energetic communication in cardiomyocytes.
kinase isoenzymes in the intact heart. J Biol Chem. 2000;275:19742– Front Physiol. 2014;5:376.
19746. 222. Karo J, Peterson P, Vendelin M. Molecular dynamics simulations of
215. van Deursen J, Ruitenbeek W, Heerschap A, Jap P, ter Laak H, Wieringa creatine kinase and adenine nucleotide translocase in mitochondrial
B. Creatine kinase (CK) in skeletal muscle energy metabolism: a study membrane patch. J Biol Chem. 2012;287:7467–7476.
of mouse mutants with graded reduction in muscle CK expression. Proc 223. Meyer RA, Sweeney HL, Kushmerick MJ. A simple analysis of the
Natl Acad Sci USA. 1994;91:9091–9095. phosphocreatine shuttle. Am J Physiol. 1984;246:C365–C377.
216. Wallis J, Lygate CA, Fischer A, et al. Supranormal myocardial creatine 224. van Beek JH, Supandi F, Gavai AK, de Graaf AA, Binsl TW, Hettling H.
and phosphocreatine concentrations lead to cardiac hypertrophy and Simulating the physiology of athletes during endurance sports events:
heart failure: insights from creatine transporter-overexpressing modelling human energy conversion and metabolism. Philos Trans A
transgenic mice. Circulation. 2005;112:3131–3139. Math Phys Eng Sci. 2011;369:4295–4315.
217. Gabr RE, El-Sharkawy A-MM, Schär M, Weiss RG, Bottomley PA. 225. Hettling H, van Beek JH. Analyzing the functional properties of the
High-energy phosphate transfer in human muscle: diffusion of creatine kinase system with multiscale ‘sloppy’ modeling. PLoS Comput
phosphocreatine. Am J Physiol Cell Physiol. 2011;301:C234–C241. Biol. 2011;7:e1002130.
10 
Special Considerations for Cardiovascular
Magnetic Resonance: Safety,
Electrocardiographic Setup, Monitoring,
and Contraindications
Pieter van der Bijl, Victoria Delgado, and Jeroen J. Bax

During the last three decades, cardiovascular magnetic resonance 8. Possible effects of the intravenous use of magnetic resonance (MR)
(CMR) has developed into an important diagnostic clinical tool in contrast agents
cardiology. Not only the anatomy of the heart but also its function, 9. Patient safety during stress conditions
metabolism, and perfusion, as well as the coronary arteries, can be The safety concerns inherent to these issues are discussed.
evaluated with CMR. There are advantages of CMR over other diag-
nostic imaging methods: first, CMR does not use ionizing radiation; Biologic Effects
second, the radiofrequency (RF) radiation penetrates bony and air- Concerning the biologic effects of a static magnetic field, many struc-
filled structures without attenuation; third, CMR gives additional tures in animals and humans are affected by magnetic fields. Many
diagnostic information about tissue characteristics; and, finally, CMR potential biologic effects and different magnitudes of magnetic fields
provides three-dimensional (3D) images or images of arbitrarily have been examined, including the effect of the field on cardiac con-
oriented slices. tractility and function. Gulch and colleagues concluded that static
However, when performing CMR, particular precautions must be magnetic fields used in CMR do not constitute any hazard in terms
taken. Because CMR operates with high static and gradient magnetic of cardiac contractility.3 These magnetic fields do not increase ven-
fields, special safety regulations must be taken into account and certain tricular vulnerability, as assessed by the repetitive response threshold
contraindications must be considered. This chapter reviews the safety, and the ventricular fibrillation threshold.4 In one of the investigations,
electrocardiographic (ECG) setup, patient monitoring, and contrain- however, cardiac cycle length was shown to be altered.5 Two studies
dications to CMR; in particular, the issue of devices (pacemakers, reported that lymphocytes from blood samples obtained in humans
implantable cardioverter defibrillators [ICDs]) as well as coronary stents exposed to a 1.5 tesla (T) field demonstrated DNA double-strand
and prosthetic heart valves, is addressed. breaks and changes consistent with mild inflammation.6,7 However,
these studies have significant limitations and four recent reports
SAFETY OF CARDIOVASCULAR have found no such DNA damage.8–11 Another area of concern is
MAGNETIC RESONANCE CMR during pregnancy, for example, teratogenicity (cellular effects,
for example on differentiation, have been demonstrated in animals),
General Issues damage to the fetal inner ear due to noise exposure and tissue heating
CMR generally takes longer than other diagnostic modalities (although effects, although none have been documented to cause harm.12 A recent
the time is significantly shortened with real-time imaging1), and the study of CMR in pregnancy (including first trimester) found CMR
confined space in which the patient is placed is rather narrow, which to be safe.13 Furthermore, 7 T scanners are becoming more common
some patients find uncomfortable. During CMR, communication with (although currently very rarely used for CMR), and have been associ-
the patient may be difficult because of interfering noise from the gradi- ated with transient dizziness upon movement of the subject into the
ent coils. On the other hand, CMR is entirely noninvasive. Overall, the scanner (postulated to be due to induced currents affecting vestibular
safety issues during CMR that may pose potential safety concerns can hair cells in the inner ear), as well as a metallic taste in the mouth in up
be summarized as follows2: to 11% of patients.14 Numerous biologic effects on other systems have
1. Biologic effects of the static magnetic field been investigated extensively, and it may be concluded that no delete-
2. Ferromagnetic attractive effects of the static magnetic field on certain rious biologic effects from static magnetic fields used in CMR have yet
devices been established.
3. Potential effects on the relatively slow time-varying magnetic field
gradients Ferromagnetism
4. Effects of the rapidly varying RF magnetic fields, including RF power The physical effect of the static magnetic field consists of a potential
deposition concerns health hazard from the attractive effect on ferromagnetic objects. Fer-
5. Auditory considerations of noise from the gradients romagnetic objects can be defined as those in which a strong intrinsic
6. Safety considerations concerning superconducting magnet systems magnetic field can be induced when they are exposed to an external
7. Psychological effects magnetic field. The existence of different kinds of scanners with different

108
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 109

A B E

C D F
FIG. 10.1  Bileaflet, tilting disc, mechanical aortic valve prosthesis (arrow) during diastole on steady-state
free precession (SSFP) imaging (A). Short-axis view of the same prosthesis (arrow) as in A during diastole,
clearly demonstrating an artifact (B). Systolic image of the prosthesis (arrow) in A (C). Systolic, short-axis
view of the same prosthesis (arrow) as in B, with both leaflets visible in the open position (D). Bileaflet,
tilting disc, mechanical mitral valve prosthesis during systole on an SSFP image, demonstrating a regurgitant
jet (arrow) (E). Short-axis view of the same prosthesis (arrow) as in E (F). (From Von Knobelsdorff-Brenkenhoff
F, Trauzeddel RF, Schulz-Menger J. Cardiovascular magnetic resonance in adults with previous cardiovascular
surgery. Eur Heart J Cardiovasc Imaging. 2014;15(3):235–248.)

shielding makes the discussion about this topic even more crucial. When occurs around metallic prosthetic valves (Fig. 10.1) and sternal wires
dealing with a static magnetic field, two types of physical concerns exist. (Fig. 10.2) after bypass surgery, but this does not make the imaging
First, there are concerns about forces exerted on ferromagnetic hazardous.20 Nonstainless steel, which may be ferromagnetic, is not
objects within, on, or distant from the patient. These forces result in used for human implants, but is commonly used for oxygen cylinders,
rotational (torque) or translational (attractive) motion of the object. for example. Finally, batteries are typically attracted to the magnet, and
Within the human body, a ferromagnetic metallic structure may be this is one of the problems of imaging pacemakers.
sufficiently attracted, or have a sufficient amount of torque exerted, to The second type of physical concern deals with magnetically sensi-
create a hazardous situation. These factors should be carefully considered tive equipment, the functioning of which may be adversely affected by
before subjecting a patient with a ferromagnetic implant or material the magnetic field. The most common of these is the cardiac pacemaker.
to CMR, particularly if the device is located in a potentially dangerous Most pacemakers include a reed relay switch whereby the sensing mecha-
area of the body, where movement or dislodgment of the device could nism can be bypassed and pacing in the asynchronous mode can occur.
injure the patient. Another potentially injurious effect is known as the This switch is activated when a magnet of sufficient strength is held
projectile, or missile, effect. This refers to the fact that ferromagnetic over the pacemaker.21 In addition, the function of cardiac pacemakers
objects have the potential to gain sufficient speed during attraction may be influenced by field strengths as low as 17 gauss.21 In practice,
to the magnet that the accumulated kinetic energy could be injurious reed switch closure can be expected in all pacemakers placed in the
or even lethal if the object were to strike a patient. Numerous studies bore of the scanner. Pacemaker and ICD function is considered again
have been performed to assess the ferromagnetic qualities of various later in this chapter.
metallic implants and materials.15–19 The results indicate that patients
with certain metallic implants or prostheses that are nonferromagnetic Effect of Rapidly Switched Magnetic Fields
or are minimally deflected by static magnetic fields can safely undergo CMR exposes the patient to rapid variations of magnetic fields by the
CMR. The literature on this topic has been extensively reviewed and transient application of magnetic gradients during imaging. The effect
compiled.17 However, there are common misconceptions about what of rapidly switched magnetic fields may be the induction of currents
types of objects are ferromagnetic. The most important misconcep- within the body or any other electrical conductor, according to Faraday’s
tion is that stainless steel is ferromagnetic, when it is not. Patients law. The current is dependent on the time rate of change of the magnetic
with stainless steel implants can therefore be imaged safely, except for field (dB/dt), the cross-sectional area of the conducting tissue loop, and
a small number of well-described exceptions, as discussed later. The the conductivity of the tissue. Biologic effects of induced currents can
implant will interfere locally with the images; for example, signal loss be caused either by power deposition by the induced currents (thermal
110 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A B C
FIG. 10.2  Cardiovascular magnetic resonance image of a patient with sternal wires after bypass grafting.
The artifact is clearly seen (arrows) on the horizontal long-axis (A) and short-axis (B) gradient echo cine images,
and to a much lesser extent, on the transaxial spin echo image (C). (Courtesy Dr. Dudley Pennell, Royal
Brompton Hospital, London.)

effects) or by direct effects of the current (nonthermal effects). Thermal magnetostimulation, which has the hyperbolic form of a strength-duration
effects as a result of switching gradients are not believed to be clinically curve and allows an estimation of the lowest possible value of the
significant.22–24 Possible nonthermal effects include stimulation of nerve magnetic flux density capable of stimulating nerves and muscles. Cal-
or muscle cells. The threshold currents for nerve stimulation and ven- culations have shown that the threshold for heart excitation is more
tricular fibrillation are known to be much higher than the estimated than 200 times higher than for nerve and muscle stimulation, depending
current densities induced under clinical CMR conditions. The echo on pulse duration.29 However, in clinical practice, some precautions are
planar imaging method, however, involves more rapidly changing mag- necessary. First and most importantly, the specific absorption rate of
netic field gradients, and peripheral muscle stimulation in humans has the imaging sequence being operated is monitored by the scanner soft-
been reported.25 Such considerations have become more important as ware and must be kept below limits set by such bodies as the US Food
new technology has allowed the introduction of commercially available and Drug Administration (FDA). Second, circumstances that could
ultrafast gradient switching systems, and guidelines for maximum mag- enhance the possibility of heating injury should be avoided. This includes
netic field variation are under development. ensuring the prevention of loops that could act as aerials within the
scanner and enhance the heating effect locally. Therefore, patients should
Radiofrequency Time-Varying Field not be allowed to cross their legs (loop via the pelvis) or clasp their
The transmitted RF time-varying field induces electrical currents within hands (loop via the shoulder and upper chest). The simple use of pillows
the tissue of the patient. The majority of this power is transformed into prevents such problems. Other possible loops include the ECG leads,
heat within the patient’s tissue as a result of ohmic heating. The time- which should always be run out of the scanner parallel to the main
varying magnetic gradients have the potential to cause either thermal field, and not looped across the chest. Finally, pacemaker and ICD leads
or nonthermal biologic effects. The distinction between these two is make excellent aerials. The pacemaker lead can heat significantly during
a matter of frequency, waveform shape, and magnitude. The discus- CMR and become a potential hazard (discussed later). Another con-
sion about nonthermal effects from RF magnetic fields is controversial sideration in patients after cardiac surgery is the effect of retained epi-
because of questions about the relationship between chronic exposure cardial pacemaker leads. These leads can be left in place after surgery,
to electromagnetic fields over many years and the causation of cancer and they may therefore act as an antenna during CMR. Studies have
or developmental abnormalities. The most recent evidence suggests that suggested that such short retained epicardial wires do not pose a sig-
proximity to power lines is not injurious.26 Clearly, acute exposure of nificant problem.30,31
a patient to short-term RF fields for a diagnostic CMR examination is Finally, the use of ECG electrodes, which are essential for cardiac
different from chronic exposure. The induced currents from RF mag- gating, must be considered. Metallic ECG electrodes may cause burns
netic fields are unable to cause nerve excitations. One of the difficulties during CMR, but this risk can be reduced with the use of carbon fiber
faced by medicine is proving that a procedure is not injurious because electrodes, and these have now become standard.32,33
of anecdotal case reports of adverse events and publication bias toward
nonneutral reports.27 This issue is also faced by such a well-established Auditory Considerations
technology as ultrasound, where safety concerns have been raised over During CMR, the gradient coils and adjacent conductors produce a
acoustic exposure.28 repetitive sound because they act essentially as loudspeakers, with current
In contrast to the insignificant thermal effects caused by switched being driven through them while they are in a magnetic field. Auditory
gradients, however, thermal effects as a result of RF pulses are of sig- considerations should therefore be taken into account when imaging
nificant concern. The main biologic effects induced by RF fields are a patient. The amplitude of this noise depends on factors such as the
therefore related to the thermogenic qualities of the RF field. A general physical configuration of the magnet, the pulse sequence type, timing
point of discussion is the appropriate safety regulations for levels of specifications of the pulse sequence, and the amount of current passing
magnetic field strength in CMR imaging. Application of the fundamental through these coils.34 In general, the amplitude of the generated noise
law of electrostimulation is well established, both on theoretical and from the clinical CMR scanners remains between 65 and 95 dB. However,
experimental grounds. Application of this law, in combination with there have been reported instances of temporary hearing impairment
Maxwell’s law, yields an equation called the fundamental law of as a result of CMR. Magnet-safe headphones or wax earplugs are readily
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 111

available and have been shown to prevent hearing loss, and these are administration of gadolinium contrast agents are headache, nausea,
in common use.35 Systems combining sound attenuation with the facility vomiting, local burning or cool sensation, and hives. There have been
to play music of the patient’s choice are also available. Research on the reported incidents of anaphylactoid reactions associated with IV injec-
reduction of noise in MR scanners is ongoing, and the use of antinoise tion, although the frequency of this appears to be approximately 1 per
is one area of interest.36 100,000 doses.45 The safety margins with these agents appear to be
considerably better than with x-ray contrast agents, although issues of
Superconducting System Issues nephrogenic systemic fibrosis are a concern in patients with severely
Most superconducting CMR scanner systems use liquid helium. The impaired renal function, and there are differences in safety between
helium maintains the magnet coils in their superconducting state. Helium the commercially available gadolinium contrast agents favoring the
achieves the gaseous state at approximately −269°C (4 K). If for any tighter-bound chelates (see Chapter 3). Evidence of cerebral deposi-
reason the temperature within the cryostat rises, or in a system quench, tion (especially globus pallidus and dentate nucleus) of gadolinium-
the helium will enter the gaseous state. This means a marked increase based contrast agents has emerged from animal and human studies,
in volume and thereby pressure within the cryostat. A pressure-sensitive although the mechanism(s) and clinical implications are currently
valve is designed to give way to the gaseous helium, which is always unknown.46,47 The risk of cerebral deposition appears related to the
vented outside the CMR scanner room. However, it is possible that number of administrations of gadolinium-based contrast.48 The use
some helium gas is released into the imaging room should the system of gadolinium in pregnancy depends on the risk-benefit ratio, as in
not work perfectly. Asphyxia and frostbite are potential hazards if a general contrast agents are best avoided. Gadolinium can be used and
patient is exposed to helium vapor for a prolonged time, although there safety appears good, but with a possible small increase in adverse out-
are no reports of such an occurrence in the medical community. For comes, the interpretation of which is limited by small numbers.13 For
older scanners that still use a buffer of liquid nitrogen within the system CMR, these agents are used to increase contrast between blood and
(boils at 77 K), an oxygen monitor is recommended in the scanner soft tissue for cine imaging for functional studies or angiography, to
room. Cryogenic dewars should be stored away from the scanner and enhance cardiac tumors and cysts, to assess myocardial perfusion, and
in well-ventilated areas. to examine for myocardial infiltration. In summary, FDA-approved
gadolinium complexes can be safely used in patients with cardiac
Psychological Effects disorders.
Claustrophobia or other psychological problems may be encountered Multiple new CMR contrast agents are being developed and inves-
in up to 10% of patients undergoing CMR, although on average, the tigated. These are mainly gadolinium complexes, sometimes with novel
incidence is closer to 2% to 4%, and this can be reduced further to a binding molecules for special actions, but in addition, iron-based com-
small number of intractably anxious patients by the use of explanation, pounds are being developed. Some of these agents (e.g., gadofosveset)
reassurance, and where necessary, light sedation with, for example, 2 bind to albumin and are retained in the vascular system and do not leak
to 5 mg intravenous (IV) diazepam.37,38 The development of shorter into the extravascular space. The adverse-effect profile of gadofosveset
magnets and open designs are promising, although 39% of patients appears similar to other gadolinium-based agents, although a prolon-
experienced some level of claustrophobia in a short-bore scanner and gation of the corrected QT interval (without any dysrhythmias) has
26% in an open-bore scanner in a direct comparison.39 been reported. These agents may have clinical utility for angiography,
Such problems are related to a variety of factors, including the restric- possibly in the coronaries, and for functional imaging. Hyperpolarized
tive dimensions of the scanner, the duration of the examination, the CMR—executed with tracers such as 1-13C pyruvate—has only recently
noise, and the ambient conditions within the magnet bore.40 Fortunately, been introduced to humans and safety data are limited.49 Molecular
adverse psychological effects with CMR are usually transient. In a study imaging employs “probes,” which bind to molecular targets either
reported by Weinreb and colleagues, based on the experience of 450 actively or passively by accumulation, and trace various anatomical
patients undergoing CMR and computed tomography (CT) examina- and functional processes, such as thrombus formation, angiogenesis,
tions, it was clearly shown that patients often prefer the CMR study, atherosclerotic plaque, and fibrosis development.50,51 Most of these tech-
although CMR took longer.41 Furthermore, the patient is placed into niques have only been tested in animals, and safety data in humans
a confined space and there are difficulties in communicating with the are scarce.
patient during CMR scanning because of the noise from the gradient
coils and the necessity of eliminating all extraneous RF sources from Patient Safety During Stress Conditions
the examination room. To a certain extent, this can be avoided when A concern with stress CMR scans has been the ability to handle emer-
the patient assumes a prone position in the scanner, facilitating com- gency situations. Patient monitoring during stress conditions is a critical
munication with the outside surroundings.42 Simple maneuvers, such issue because myocardial ischemia can be provoked in patients with
as using mirrors, also help in allowing the patient a clear view of the coronary artery disease. Commercial equipment is available for nonin-
scanning room. Allowing the anxious patient to visit the scanner before vasive monitoring of blood pressure, heart rate, oxygen saturation, and
the appointment gives the patient an opportunity to become familiar other vital parameters in CMR scanners. The most crucial difference
with the facility and staff. compared with conventional exercise testing outside a magnetic field
is the lack of a diagnostic ECG, in particular, at high levels of stress,
Safety Considerations Associated With CMR precluding the proper assessment of stress-induced ST-segment changes.
Contrast Agents This holds for both conventional exercise using a specially adapted
The safety profile of the contrast agents containing gadolinium cur- bicycle ergometer and pharmacologically induced stress. Under these
rently on the market is extremely good. Their safety profile is well doc- circumstances, only heart rate can be monitored reliably. When per-
umented.43,44 The median lethal dose of gadopentetate dimeglumine forming pharmacologic stress CMR (e.g., with dipyridamole, adenos-
(Gd-DTPA) is roughly 10 mmol/kg, which is 100 times the diagnostic ine, regadenoson, or dobutamine), an experienced physician should
dose and exemplifies the wide safety margin that these contrast agents be present during the examination, and appropriate treatments for
enjoy. Patient tolerance is also high, and the prevalence of adverse complications should be in direct proximity. Dipyridamole (half-life 30
reactions is approximately 2%. Among the reactions related to the IV minutes), adenosine (half-life 10 seconds) and regadenoson (half-life 3
112 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

minutes) are vasodilators. These agents have similar side effects, such the CMR scanner by lengthening the rubber tubing connected to a
as hypotension, arrhythmias, and bradycardia. During adenosine infu- blood pressure cuff.57 The newest monitoring equipment eliminates
sion, atrioventricular (AV) heart block may develop in a small percent- the need for wires and tubes to leave the scanning room by using a
age of patients (0.7%–2.8%), although this is usually asymptomatic microwave transmitter communicating with a slave display unit in the
and self-limited. When patients are symptomatic, the short physical operating room.
half-life of adenosine means that heart rhythm and symptoms can be The CMR procedure depends on a high-quality ECG signal for
restored very quickly by halting the infusion. As a suitable antagonist routine imaging, and each manufacturer has developed its own solution
to dipyridamole, adenosine, and regadenoson, aminophylline may be to the problems posed. Fiberoptic transmission of ECG signals for gating
given slowly at an initial dose of 50 mg IV up to a maximum dose of is now common, and this significantly reduces RF pulse artifacts in the
250 mg if necessary. In the case of persisting advanced heart block, ECG. Felblinger and coworkers showed that this type of system could
0.5 mg atropine IV should be administered up to a total dose of 3 mg. yield signals almost free from interference, during both conventional
Dipyridamole and adenosine should not be administered to patients with and high gradient activity sequences, such as during echo planar
asthma, whereas regadenoson is safe in patients with asthma and chronic imaging.58–60 From this signal, the authors also developed a method for
obstructive pulmonary disease, because of its more selective action on respiration monitoring during MR sequences. Third-party ECG solu-
A2A receptors. Adenosine, and dipyridamole indirectly (by inhibiting tions are also now being incorporated into the latest generation of
the degradation of adenosine), acts on A1, A2A, A2B, and A3 receptors. scanners, and these come with specific ECG recommendations for lead
A2A receptors mediate coronary and peripheral vasodilation, whereas A1, placement. Carbon fiber electrodes are required to eliminate the risk
A2A, A2B, and A3 receptors cause bronchoconstriction and AV block.52 A of burning that has been reported with standard metallic ECG elec-
further advantage of regadenoson is the fact that it can be administered trodes.32 Typical lead placement is the result of compromise. A better
as a bolus, therefore not requiring a CMR-compatible infusion pump.52 signal results from widely spaced electrodes, but this results in more
Dobutamine (half-life 2 minutes) is a beta-agonist leading to an increase artifact from the gradients. In general, therefore, the leads are kept
in cardiac inotropy (contractility) and chronotropy (heart rate). Common relatively close together, and on the left side, which reduces the mag-
side effects are palpitations, and less commonly, arrhythmias, such as netohydrodynamic effect (the effect of systolic aortic flow causing surface
supraventricular tachycardia and (nonsustained) ventricular tachycar- potentials on the ECG that distort the ST segment; Fig. 10.3). A typical
dia. Dobutamine can be safely administered to patients with asthma.53 lead placement that is commonly adopted is shown in Fig. 10.4. Some
The actions of dobutamine can be counteracted by IV administration centers have found ECG gating using electrodes on the back to be suc-
of a short-acting beta-blocking agent, such as esmolol. In the case of cessful, but this is not widely used. The ECG leads should not be allowed
cardiac arrest or ventricular fibrillation, the recommendations should to form loops, which could present a burning hazard, and they should
be followed according to published guidelines, such as those proposed be braided together and brought out of the magnet while aligned parallel
by the European Resuscitation Council.54 In every CMR facility, an to the bore to reduce electrical interference. Keeping the electrical cables
alarm system and a written flow chart should be visually available, short is helpful, and fiberoptic conversion modules are therefore often
with the necessary instructions in case of emergency. It is necessary very close to the patient’s chest. Switching between the ECG traces
to be able to remove the patient from the examination room quickly sometimes allows flexibility to reduce gating errors from tall T-waves
(preferably within 20 seconds) to an area where emergency treatment or electrical interference. One thing is certain, however, and that is that
can be performed safely, away from the hazards of the magnetic field. time spent ensuring that the ECG is stable and working correctly at
A nonferromagnetic stretcher stored in the scanner room or a detach- the start of the scan is time very well spent.
able scanner table is ideal. A cardiac arrest trolley must be maintained An alternative technique to routine surface ECG recording has been
in close proximity to the scanner room, and all staff should undergo described by Fischer and colleagues using vectorcardiography, and has
regular training in cardiopulmonary resuscitation techniques. Regular been widely implemented.61 The system examines the 3D orientation
checks should be made of both the resuscitation equipment and the of the ECG signal and uses the calculated vector of the QRS complex
alarm system. as a filter mechanism to ignore electrical signals, which are of similar

PATIENT MONITORING AND


ELECTROCARDIOGRAPHIC SETUP
Patient monitoring during CMR poses problems that will not be familiar
Outside
to users of other technologies, such as echocardiography. Ferrous metal, magnet
which is present in most monitoring equipment, can distort the mag- fast
netic field, and such an item has the potential to become a projectile. atrial
fibrillation
In addition, monitoring wires that are attached to the patient, leaving
the scanner, and passing to another room may act as an antenna for
stray RF signals. Electrical equipment in the scanner room also can act
as a source of RF noise. All of these disturbances may result in image
degradation. Therefore, specific solutions to these problems have been Inside
designed. magnet
Commercially available CMR-compatible monitoring equip-
ment, including that used to measure ECG, blood pressure, and chest
wall movements, as well as for general anesthesia, has been tested FIG. 10.3  The magnetohydrodynamic effect. The top trace was recorded
in several studies.55,56 Satisfactory monitoring can be obtained and in a patient with atrial fibrillation outside of the magnet, and the bottom
images obtained during its use can be evaluated adequately.57 For trace, inside. Note the distortion of the ST segment (arrows) caused by
some monitoring, simple solutions work, such as that reported by added potentials arising from systolic flow in the aorta. (Courtesy Dr.
Roth and associates, who measured arterial blood pressure outside Dudley Pennell, Royal Brompton Hospital, London.)
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 113

V2 V3
V4 General Contraindications to Cardiovascular
V1 Magnetic Resonance
There are a number of circumstances in which CMR is better avoided.
V5 This is because of reports of death or harm that have occurred. Promi-
V3R nent among these are patients with cerebral aneurysm clips, which have
become dislodged by MR, causing fatal cerebral hemorrhage. Modern
clips are not ferromagnetic and are safe, but the problem is establishing
the type of clip with certainty before performing CMR. In general,
therefore, CMR should be avoided in these patients unless written
V4R
information is available and appropriate advice from a neurosurgical
V6
center is obtained. There have been cases of bleeding in the eye in
patients with previous injury with metallic shards (usually metal workers),
and again a history of this should be sought. A skull x-ray can be helpful
in cases of doubt. Electronic implants are the other major problem.
These may dysfunction or may be damaged by CMR, which is therefore
FIG. 10.4  Typical electrode placement for cardiovascular magnetic best avoided. This applies to cochlear implants, neurostimulation systems,
resonance. The conventional chest leads (black circles) are shown for non-MR conditional pacemakers/implantable cardioverter-defibrillators
comparison, and the positions of the four carbon electrocardiographic (ICDs), and a number of other modern implants. In general, the main
electrodes over the left chest are indicated (white circles). (Courtesy rule to observe is to determine the risk-benefit ratio for the proposed
Dr. Dudley Pennell, Royal Brompton Hospital, London.) procedure. If the information can only be obtained by CMR, and if it
is very important, then the risk-benefit ratio may be positive for some
patients (as has been shown in some patients with pacemakers). In
timing and similar magnitude in the cardiac cycle, but a different vector. many clinical circumstances, however, the information required can be
The system as reported identified the QRS complex correctly in 100% obtained by other means. Reference texts on the safety of specific devices
of cases, with 0.2% false-positive findings. In a subsequent study in are available (e.g., http://www.mrisafety.com/).
normal individuals and patients with supraventricular extrasystoles, For CMR, a number of other specific device issues require mention.
the same authors showed that vector cardiography-based triggering Swan-Ganz catheters and temporary pacing wires, in general, preclude
provided nearly 100% triggering performance during CMR examina- CMR, but sternal wires and vascular clips on grafts do not present
tions.62 This system represents a significant improvement for CMR in safety problems, although localized artifacts occur on the images. Three
stabilizing this important gating signal. specific areas are discussed in more detail: namely, coronary stents,
Should interpretation prove impossible for technical reasons, a stan- valvular prostheses and structural heart disease intervention devices,
dard vascular Doppler can be used to monitor heart rate during CMR. and cardiac implantable electronic devices. Many of them are not con-
The Doppler and telemetric ECG do not contain enough ferromagnetic sidered absolute contraindications for CMR. However, timing and scan-
material to cause visible image degradation. Jorgensen and associates ning protocol should be adjusted in each clinical scenario.
evaluated whether patients could be monitored during CMR with 1.5 T
machines in a manner that complies with monitoring standards.63 Coronary Stents
The high magnetic field can interfere with normal functioning of The first intracoronary stents were implanted in human coronary arter-
equipment, not only monitoring equipment but also smaller items such ies in 1986 by Sigwart and colleagues.67 The reduced restenosis rate
as infusion pumps used for stress testing. In general, the influence of compared with conventional balloon angioplasty has resulted in routine
the CMR scanner on nearby equipment depends on several factors, use of stent placement for treatment of coronary stenoses.68–71
such as the strength of the CMR magnetic field, the proximity of the Most coronary stents are metallic structures that remain in situ for
equipment to the scanner, the amount of ferromagnetic material in the life. As a result, there have been concerns about stent dislodgment during
equipment, and the design of the electrical circuitry. CMR as well as concern about possible heating effects. Several factors
Finally, simple devices, such as closed-circuit television and a two-way determine the risk of placing these materials in a magnetic field. These
intercommunication system, also aid in monitoring by allowing a con- factors include the ferromagnetism of the material, the strength of the
stant view of the patient and easy communication if the patient is in static and gradient magnetic fields, the metal mass, and the geometry
discomfort. However, the latter may be impaired during imaging as a of the material.19,72 It is generally recommended that 6 to 8 weeks elapse
result of the noise of the gradients. Because sequences are now com- before CMR imaging to allow for tissue ingrowth, although this is not
monly being reduced in duration to a breath-hold, however, this limita- based on robust data.73 The most commonly used stents are made of
tion is becoming much less important. stainless steel or nitinol, which is not ferromagnetic.74 Metallic stents
have been exposed to field strengths up to 7 T in vitro, without signifi-
CONTRAINDICATIONS TO CARDIOVASCULAR cant heating.75 Large studies have subsequently confirmed the safety of
stent imaging with CMR. Gerber and coworkers evaluated 111 patients
MAGNETIC RESONANCE with CMR within 8 weeks of stent implantation (median 8 days; range
In general, there are potential hazards with artifacts of ferromagnetic 0 to 54 days); during follow-up, 4 noncardiac deaths occurred as well
and nonferromagnetic materials in CMR, for example, neurosurgical as 3 revascularization procedures.76 These events were not related to
clips and ocular implants.64–66 The (relative) contraindications of these the CMR procedure. This finding agrees with widespread clinical experi-
materials for CMR are dependent on factors such as the degree of fer- ence in which stent imaging has been performed on the day after
romagnetism, the geometry of the material, the gradient (for force), implantation, or soon afterward.77,78 Moreover, assessing changes in
and the field strength (for torque) of the imaging magnet and many coronary flow reserve after percutaneous intervention with stenting
other factors. was shown to be feasible.79
114 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Another issue regarding CMR procedures in patients with coronary for patients with atrial fibrillation and contraindications or intolerance
stents is the image distortion caused by the ferromagnetic properties to oral anticoagulants. Left atrial occluder devices contain metal, such
of the stent. Generally, the greater the ferromagnetism of a metal- as nitinol mesh, but have been safely scanned at 1.5 T and 3 T.73,96,97
lic implant, the greater its magnetic susceptibility artifact. However, Artifacts are minimal, even allowing qualitative and quantitative evalu-
studies have shown that CMR angiography could reliably be used for ation of residual leaks (see Fig. 10.5).98
noninvasive imaging and evaluation of blood flow after stent place- Severe mitral regurgitation can be successfully treated with percu-
ment.80,81 Bioabsorbable vascular scaffolds (BVS) are constructed from taneous, mitral valve repair in high-risk patients, and CMR has been
polymers, which are hydrolyzed and metabolized to CO2 and H2O.82 safely performed at 1.5 T and 3 T.99,100 The MitraClip device con-
They have a number of potential advantages compared with metallic tains cobalt and chromium, which creates a significant artifact in the
stents, such as more physiologic vasomotion postplacement, facilita- mitral valve area, disallowing accurate evaluation of the mitral valve
tion of percutaneous or surgical reintervention, and little or no artifact leaflet morphology and residual mitral regurgitation postimplant
with CMR, allowing imaging of the stent lumen.82,83 BVS do some- (see Fig. 10.5).101
times contain metallic, radiopaque markers, for example, platinum, Use of septal occlusion devices has become widespread, and such
with the same potential effects as metallic stents when exposed to a devices containing nitinol and cobalt have been tested in vitro at 3 T,
strong magnetic field (i.e., heating and physical motion of the stent).82 showing no torque, maximum deflection of 4 degrees, and a temperature
An in vitro study conducted at 3 T led to no heating (1.6°C, equivalent increase of 0.4°C.100 These devices may therefore be considered safe
to the maximum background change) and no torque or deflection for CMR, but do cause large artifacts in the region of the interatrial
(0 degrees).82 septum.100

Valvular Prostheses and Structural Heart Disease Cardiac Implantable Electronic Devices
Intervention Devices The issues surrounding CMR of pacemakers are complex. In general,
Heart valve prostheses are all safe for CMR.84 These are the most recent three hazardous MR interactions with pacemakers and ICDs should
recommendations, and they supersede those that suggested that pre- be considered. First, the static magnetic fields exert mechanical forces
model 6000 series Starr-Edwards valves might cause problems during on the ferromagnetic components of the devices, including the pace-
MR. This conclusion is backed up by numerous data. Studies up to 3 maker and shock leads; the static magnetic fields can also induce asyn-
T static magnetic fields have shown that for a number of prosthetic chronous pacing. Second, a pulsed RF field may result in oversensing
valves there is no hazardous deflection during exposure of the magnetic or may induce currents in the leads, resulting in thermal damage at the
field.85–87 In general, a waiting period of 6 weeks is advocated for valvular tissue-electrode interface. Third, the gradient magnetic fields may induce
prostheses and structural heart disease devices (allowing tissue to heal) voltages on leads, resulting in over- and undersensing. Combined fields
before CMR evaluation.73 Heating of small metallic implants was tested may also result in device damage and failure.
in a study reported by Davis and associates.64 The authors found no Finally, pacemaker leads may serve as an antenna, which could result
significant increase in temperature in steel and copper clips that were in pacing the heart during scanning at the frequency of the applied
exposed to changing magnetic fields 6.4 times as strong as those expected imaging pulses. This could potentially lead to hypotension and dys-
to be used in the CMR scanner. As a result, there is no contraindication rhythmias. This effect was shown in experiments and in several patients
to CMR in patients with the currently used prosthetic valves.88–90 However, while positioned in a CMR system, but this effect must be separated
prosthetic material may lead to artifacts on CMR images. To evaluate from excitation caused by the pulse generator.102,103 Battery depletion
the influence of prosthetic valves on the interpretation of CMR images is another concern—primarily occasioned by activation of telemetry
and the capability of functional valve analysis, in a group of 89 patients circuitry.104,105
and 100 heart valve prostheses, Bachmann and colleagues showed con- It is estimated that 50% to 70% of patients with a cardiac implant-
vincingly that all patients could be imaged with CMR without any risk able electronic device (CIED) will require MR imaging during the lifetime
and that prosthesis-induced artifacts did not interfere with image inter- of the device, making a rational and safe approach to this issue impera-
pretation.89 In particular, physiologic valvular regurgitation was easy tive.104,106 Asynchronous mode is recommended in pacemaker-dependent
to differentiate from pathologic or transvalvular regurgitation. Di Cesare patients, because temporally varying gradient fields may mimic cardiac
and coworkers studied 14 patients who were surgically treated with electricity and lead to inhibition of pacing output.107 This is not without
nine biologic and seven mechanical aortic and mitral valves.90 Three risk, because competitive intrinsic and pacing rhythms may be dys-
classes of artifacts were distinguished and graded as minimal, moderate, rhythmogenic.105 Sense-only mode is preferred in patients with high
or significant. The biologic valves produced minimal artifacts and the intrinsic heart rates, precisely to avoid competitive pacing if pacemaker
mechanical valves showed only moderate artifacts. In all 16 prosthetic tracking of electromagnetic interference occurs.105,106 Studies of CIEDs
valves, CMR allowed adequate semiquantitative analysis of flow over and MR imaging at 1.5 T (only studies with >100 patients included)
the valve. are summarized in Table 10.1. A limitation shared by many studies is
Transcatheter aortic valve replacement (TAVR) is frequently per- that few CMR scans were performed (in contrast to MR scans of extra-
formed for patients with calcific, aortic stenosis who are considered cardiac anatomy).
high risk for conventional, surgical valve replacement. An in vitro study In the largest study of CIEDs in MR, 438 scans were performed in
has been conducted on a TAVR device (containing nitinol) at 3 T, which 555 patients (54% pacemakers and 46% ICDs) who were subjected to
demonstrated no torque, minimal (3 degrees) deflection and a low- MR scans at 1.5 T.107 In this study, asynchronous mode was selected in
grade heating effect (temperature rise of 2.5°C), leading the authors to pacemaker-dependent patients, and careful monitoring of vital signs
conclude that scanning the particular device in question at a high field during scanning was performed. Two power-on resets (reversion to a
strength is likely to be safe in vivo.91 A number of studies have evaluated default programming mode—usually an inhibited pacing mode) were
paravalvular leaks post-TAVR with CMR, demonstrating safe scanning documented in pacemakers, and one in an ICD.107 Two studies of CIEDs
at a field strength of 1.5 T in humans.92–94 Even though an artifact is at 3 T have been reported (primarily brain imaging), with no serious
created, the lumen of the TAVR device can be visualized (Fig. 10.5).95 adverse effects on pacemaker function noted in either (total of 58 patients
Percutaneous closure of the left atrial appendage has become an option in both studies).105,108 A “burning” sensation was noted by one patient
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 115

A B

AV

C D
FIG. 10.5  Images of valvular prostheses and structural heart disease intervention devices. An aortic bio-
prosthesis is demonstrated in (A), with a transcatheter aortic valve replacement (TAVR) device in (B). A left
atrial occlusion device (Watchman) is shown in situ in the left atrial appendage (arrow) (C). The arrow in (D)
identifies a MitraClip device, causing an artifact in the mitral valve area on a steady-state free precession
image in a four-chamber view. AV, Atrioventricular. (A and B, From Crouch G, Tully PJ, Bennetts J, et al.
Quantitative assessment of paravalvular regurgitation following transcatheter aortic valve replacement. J
Cardiovasc Magn Reson. 2015;17:32. C, From Hong SN, Rahimi A, Kissinger KV, et al. Cardiac magnetic
resonance imaging and the WATCHMAN device. J Am Coll Cardiol. 2010;55(24):2785. D, From Lurz P,
Serpytis R, Blazek S, et al. Assessment of acute changes in ventricular volumes, function, and strain after
interventional edge-to-edge repair of mitral regurgitation using cardiac magnetic resonance imaging. Eur Heart
J Cardiovasc Imaging. 2015;16(12):1399–1404.)

in the above-mentioned study, but no evidence for pacemaker dysfunc- 150 MR conditional ICDs were evaluated.110 A fast gradient echo sequence
tion could be uncovered in the subject in question.108 Sixteen percent delivered moderate-to-good images of the left ventricle in 74% of cases,
of patients experienced a power-on reset in a 3 T study by Naehle et al., and moderate-to-good images of the right ventricle in 84% of cases,
but no emergencies arose as a consequence.105 which can be explained by the fact that the generator is generally placed
Even though concern has been raised about potential coronary sinus closer to the left ventricle, and causes more significant artifacts than
(CS) lead dislodgment in cardiac resynchronization therapy (CRT) the right ventricular lead.111
devices, because of the fact that they do not possess active fixation Loop recorders may be safely scanned, although substantial artifacts
mechanisms, no such dislodgment was noted in a study of 40 patients are generated (more pronounced with gradient echo than spin echo
with nonconditional CS leads, who were scanned at 1.5 T.109 In addition, sequences).112 Rhythm artifacts may be produced, and the theoretical
no evidence was found for lead dysfunction or dysrhythmogenesis.109 risk of electromagnetic fields impacting stored data requires all data to
Inclusion of patients with CRT devices in other trials also did not be downloaded from the loop recorder before MR imaging.113,114
signal alerts.104,107 MR conditional pacemakers have been developed, with adaptations
The largest trial of CIEDs in MR to date included 201 ICD devices to hardware (minimization of ferromagnetic components and changes
at 1.5 T.107 One power-on reset occurred in an ICD (with no attempt in lead design) and software (substitution of a reed switch with Hall
by the ICD to deliver antitachycardia therapy, which is a potential con- sensor, which will not close or open unexpectedly when exposed to a
sequence).107 It has been suggested that formal defibrillation testing be magnetic field, thereby switching to asynchronous pacing or inhibition).
performed after CMR imaging of an ICD device.108 In the Evera trial, Promising safety profiles of such devices have been demonstrated in
116 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TABLE 10.1  Summary of Studies of Cardiac Implantable Electronic Devices and Magnetic
Resonance Imaging at 1.5 T (Only Studies With >100 Patients Are Included)
Year No. of No. of Patients Type of Device(s) and
Trial Published Scans Scanned % of Patients or Scans CMR % Adverse Effects
117
Higgins et al. 2015 256 198 PPM (DC) 87.9% of scans Unknown 9 power-on resets
ICD 2.3% of scans
Bailey et al.118 2015 229 229 PPM (SC) 10.5% of patients 0% No significant
PPM (DC) 89.5% of patients
Higgins et al.117 2015 512 398 Unknown Unknown 1 significant lead threshold change
Gold et al.110 2015 156 156 ICD (SC) 52.6% of patients Unknown No significant
ICD (DC) 47.4% of patients
Gimbel et al.116 2013 150 150 PPM (DC) 100% of patients Unknown No significant
Cohen et al.104 2012 125 109 PPM (SC) 4% of scans 4% Battery voltage decrease in 4% of
PPM (DC) 63% of scans scans, lead threshold change in 3% of
CRT-P 2% of scans scans, and lead impedance change in
ICD (SC) 3% of scans 6% of scans
ICD (DC) 16% of scans
CRT-D 13% of scans
Nazarian et al.107 2011 555 438 PPM 54% of patients 16% 3 power-on resets
ICD 46% of patients
CRT-D 12% of patients
Mollerus et al.119 2010 127 103 Unknown Unknown 1 device reset requiring reprogramming

CMR %, Percentage of cardiac magnetic resonance studies; CRT-D, cardiac resynchronization therapy with defibrillator function; CRT-P, cardiac
resynchronization therapy with pacemaker function; DC, dual chamber; ICD, implantable cardioverter defibrillator; PPM, permanent pacemaker;
SC, single chamber.

RV

LV

RV

A B
FIG. 10.6  A significant imaging artifact created by a cardiac implantable electronic device in a four-chamber
view (A) and a short-axis view (B). LV, Left ventricle; RV, right ventricle. (From Nordbeck P, Ertl G, Ritter O.
Magnetic resonance imaging safety in pacemaker and implantable cardioverter defibrillator patients: how far
have we come? Eur Heart J. 2015;36(24):1505–1511.)

trials.110,115,116 With appropriate selection, programming, monitoring cause significant artifacts, which may hamper image interpretation
and follow-up, CIEDs may therefore be subjected to CMR with an (Fig. 10.6).115
acceptable risk, with the possible exclusion of pacemaker-dependent
patients.109 Pacing dependency may be temporally variable, and it is
therefore challenging to define.108 The European Society of Cardiol-
CONCLUSION
ogy guideline on cardiac pacing and cardiac resynchronization therapy In general, CMR is safe and no long-term ill effects have been reported.
provides a class IIb recommendation for nonconditional CIED systems, Very rapidly changing gradients may induce nerve excitations that
and a class IIa recommendation for conditional devices.106 CIEDs can may result in muscle twitching. However, clinical scanners operate
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 117

below the threshold for such effects. The threshold of excitation of materials used are nonferromagnetic. MR-conditional pacemakers
the myocardium is approximately 200 times higher than that for other may be safely scanned, whereas patients with other CIEDs may be
muscles, and so the heart will not be stimulated by the rapidly chang- considered for CMR if the risk-benefit ratio is in favor of imaging.
ing gradients. Pharmacologic CMR stress testing can be performed
safely with appropriate monitoring. Most metallic implants, such as
intracoronary stents, prosthetic valves, structural heart disease inter-
REFERENCES
vention devices and sternal sutures, present no hazard because most A full reference list is available online at ExpertConsult.com
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 117.e1

REFERENCES 23. Schaefer DJ, Wehrli FW, Shaw D, Kneeland BJ, eds. Safety aspects of
magnetic resonance imaging. In: Biomedical Magnetic Resonance
1. Yang PC, Kerr AB, Liu AC, et al. New real-time interactive cardiac Imaging: Principles, Methodology and Applications. New York: VCH;
magnetic resonance imaging system complements echocardiography. J 1988:553.
Am Coll Cardiol. 1998;32(7):2049–2056. 24. Persson BRR, Stahlberg F, eds. Extremely low frequency (ELF) magnetic
2. Kanal E, Shellock FG, Talagala L. Safety considerations in MR imaging. fields. In: Health and Safety of Clinical NMR Examinations. Boca Raton,
Radiology. 1990;176(3):593–606. FL: CRC Press; 1989:49.
3. Gulch RW, Lutz O. Influence of strong static magnetic fields on heart 25. Cohen MS, Weisskoff RM, Rzedzian RR, Kantor HL. Sensory
muscle contraction. Phys Med Biol. 1986;31(7):763–769. stimulation by time-varying magnetic fields. Magn Reson Med.
4. Doherty JU, Whitman GJ, Robinson MD, et al. Changes in cardiac 1990;14(2):409–414.
excitability and vulnerability in NMR fields. Invest Radiol. 1985; 26. Exposure to power-frequency magnetic fields and the risk of childhood
20(2):129–135. cancer. UK Childhood Cancer Study Investigators. Lancet.
5. Jehenson P, Duboc D, Lavergne T, et al. Change in human cardiac 1999;354(9194):1925–1931.
rhythm induced by a 2-T static magnetic field. Radiology. 1988;166(1 Pt 27. Easterbrook PJ, Berlin JA, Gopalan R, Matthews DR. Publication bias in
1):227–230. clinical research. Lancet. 1991;337(8746):867–872.
6. Lancellotti P, Nchimi A, Delierneux C, et al. Biological effects of cardiac 28. Newnham JP, Evans SF, Michael CA, Stanley FJ, Landau LI. Effects of
magnetic resonance on human blood cells. Circ Cardiovasc Imaging. frequent ultrasound during pregnancy: a randomised controlled trial.
2015;8(9):e003697. Lancet. 1993;342(8876):887–891.
7. Fiechter M, Stehli J, Fuchs TA, Dougoud S, Gaemperli O, Kaufmann PA. 29. Irnich W, Schmitt F. Magnetostimulation in MRI. Magn Reson Med.
Impact of cardiac magnetic resonance imaging on human lymphocyte 1995;33(5):619–623.
DNA integrity. Eur Heart J. 2013;34(30):2340–2345. 30. Hartnell GG, Spence L, Hughes LA, Cohen MC, Saouaf R, Buff B. Safety
8. Brand M, Ellmann S, Sommer M, et al. Influence of cardiac MR imaging of MR imaging in patients who have retained metallic materials after
on DNA double-strand breaks in human blood lymphocytes. Radiology. cardiac surgery. AJR Am J Roentgenol. 1997;168(5):1157–1159.
2015;277(2):406–412. 31. Murphy KJ, Cohan RH, Ellis JH. MR imaging in patients with epicardial
9. Reddig A, Fatahi M, Friebe B, et al. Analysis of dna double-strand breaks pacemaker wires. AJR Am J Roentgenol. 1999;172(3):727–728.
and cytotoxicity after 7 tesla magnetic resonance imaging of isolated 32. Boutin RD, Briggs JE, Williamson MR. Injuries associated with MR
human lymphocytes. PLoS One. 2015;10(7):e0132702. imaging: survey of safety records and methods used to screen patients
10. Fatahi M, Reddig A, Vijayalaxmi, et al. DNA double-strand breaks and for metallic foreign bodies before imaging. AJR Am J Roentgenol.
micronuclei in human blood lymphocytes after repeated whole body 1994;162(1):189–194.
exposures to 7T magnetic resonance imaging. Neuroimage. 2016;133: 33. Jones S, Jaffe W, Alvi R. Burns associated with electrocardiographic
288–293. monitoring during magnetic resonance imaging. Burns. 1996;22(5):
11. Reddig A, Fatahi M, Roggenbuck D, et al. Impact of in vivo high-field- 420–421.
strength and ultra-high-field-strength MR imaging on DNA double- 34. Hurwitz R, Lane SR, Bell RA, Brant-Zawadzki MN. Acoustic analysis of
strand-break formation in human lymphocytes. Radiology. gradient-coil noise in MR imaging. Radiology. 1989;173(2):545–548.
2017;282(3):782–789. 35. Brummett RE, Talbot JM, Charuhas P. Potential hearing loss resulting
12. Ciet P, Litmanovich DE. MR safety issues particular to women. Magn from MR imaging. Radiology. 1988;169(2):539–540.
Reson Imaging Clin N Am. 2015;23(1):59–67. 36. McJury M, Stewart RW, Crawford D, Toma E. The use of active noise
13. Ray JG, Vermeulen MJ, Bharatha A, Montanera WJ, Park AL. Association control (ANC) to reduce acoustic noise generated during MRI scanning:
between MRI exposure during pregnancy and fetal and childhood some initial results. Magn Reson Imaging. 1997;15(3):319–322.
outcomes. JAMA. 2016;316(9):952–961. 37. Flaherty JA, Hoskinson K. Emotional distress during magnetic
14. Versluis MJ, Teeuwisse WM, Kan HE, van Buchem MA, Webb AG, van resonance imaging. N Engl J Med. 1989;320(7):467–468.
Osch MJ. Subject tolerance of 7 T MRI examinations. J Magn Reson 38. Francis JM, Pennell DJ. Treatment of claustrophobia for cardiovascular
Imaging. 2013;38(3):722–725. magnetic resonance: use and effectiveness of mild sedation. J Cardiovasc
15. New PF, Rosen BR, Brady TJ, et al. Potential hazards and artifacts of Magn Reson. 2000;2(2):139–141.
ferromagnetic and nonferromagnetic surgical and dental materials and 39. Enders J, Zimmermann E, Rief M, et al. Reduction of claustrophobia
devices in nuclear magnetic resonance imaging. Radiology. 1983;147(1): during magnetic resonance imaging: methods and design of the
139–148. “CLAUSTRO” randomized controlled trial. BMC Med Imaging.
16. Shellock FG, Crues JV. High-field-strength MR imaging and metallic 2011;11(1):1–15.
biomedical implants: an ex vivo evaluation of deflection forces. AJR Am 40. Quirk ME, Letendre AJ, Ciottone RA, Lingley JF. Anxiety in patients
J Roentgenol. 1988;151(2):389–392. undergoing MR imaging. Radiology. 1989;170(2):463–466.
17. Shellock FG. MR imaging of metallic implants and materials: a 41. Weinreb JC, Maravilla KR, Peshock R, Payne J. Magnetic resonance
compilation of the literature. AJR Am J Roentgenol. 1988;151(4): imaging: improving patient tolerance and safety. AJR Am J Roentgenol.
811–814. 1984;143(6):1285–1287.
18. Randall PA, Kohman LJ, Scalzetti EM, Szeverenyi NM, Panicek DM. 42. Hricak H, Amparo EG. Body MRI: alleviation of claustrophobia by
Magnetic resonance imaging of prosthetic cardiac valves in vitro and in prone positioning. Radiology. 1984;152(3):819.
vivo. Am J Cardiol. 1988;62(13):973–976. 43. Goldstein HA, Kashanian FK, Blumetti RF, Holyoak WL, Hugo FP,
19. Teitelbaum GP, Bradley WG Jr, Klein BD. MR imaging artifacts, Blumenfield DM. Safety assessment of gadopentetate dimeglumine in
ferromagnetism, and magnetic torque of intravascular filters, stents, and U.S. clinical trials. Radiology. 1990;174(1):17–23.
coils. Radiology. 1988;166(3):657–664. 44. Sullivan ME, Goldstein HA, Sansone KJ, Stoner SA, Holyoak WL,
20. von Knobelsdorff-Brenkenhoff F, Trauzeddel RF, Schulz-Menger J. Wiggins JR. Hemodynamic effects of Gd-DTPA administered via rapid
Cardiovascular magnetic resonance in adults with previous bolus or slow infusion: a study in dogs. AJNR Am J Neuroradiol.
cardiovascular surgery. Eur Heart J Cardiovasc Imaging. 2014;15(3): 1990;11(3):537–540.
235–248. 45. Weiss KL. Severe anaphylactoid reaction after i.v. Gd-DTPA. Magn Reson
21. Pavlicek W, Geisinger M, Castle L, et al. The effects of nuclear magnetic Imaging. 1990;8(6):817–818.
resonance on patients with cardiac pacemakers. Radiology. 1983;147(1): 46. Stojanov D, Aracki-Trenkic A, Benedeto-Stojanov D. Gadolinium
149–153. deposition within the dentate nucleus and globus pallidus after repeated
22. Bottomley PA, Edelstein WA. Power deposition in whole-body NMR administrations of gadolinium-based contrast agents-current status.
imaging. Med Phys. 1981;8(4):510–512. Neuroradiology. 2016;58(5):433–441.
117.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

47. Ramalho J, Castillo M, AlObaidy M, et al. High signal intensity in 69. Serruys PW, de Jaegere P, Kiemeneij F, et al. A comparison of balloon-
globus pallidus and dentate nucleus on unenhanced T1-weighted MR expandable-stent implantation with balloon angioplasty in patients with
images: evaluation of two linear gadolinium-based contrast agents. coronary artery disease. Benestent Study Group. N Engl J Med. 1994;
Radiology. 2015;276(3):836–844. 331(8):489–495.
48. Malayeri AA, Brooks KM, Bryant LH, et al. National Institutes of Health 70. Kimura T, Yokoi H, Nakagawa Y, et al. Three-year follow-up after
perspective on reports of gadolinium deposition in the brain. J Am Coll implantation of metallic coronary-artery stents. N Engl J Med.
Radiol. 2016;13(3):237–241. 1996;334(9):561–566.
49. Rider OJ, Tyler DJ. Clinical implications of cardiac hyperpolarized 71. Carrozza JP Jr, Kuntz RE, Levine MJ, et al. Angiographic and clinical
magnetic resonance imaging. J Cardiovasc Magn Reson. 2013;15:93. outcome of intracoronary stenting: immediate and long-term results
50. Botnar RM, Ebersberger H, Noerenberg D, et al. Molecular imaging in from a large single-center experience. J Am Coll Cardiol. 1992;20(2):
cardiovascular diseases. Rofo. 2015;36(2):92–101. 328–337.
51. Spuentrup E, Buecker A, Katoh M, et al. Molecular magnetic resonance 72. Shellock FG, Morisoli S, Kanal E. MR procedures and biomedical
imaging of coronary thrombosis and pulmonary emboli with a novel implants, materials, and devices: 1993 update. Radiology. 1993;
fibrin-targeted contrast agent. Circulation. 2005;111(11):1377–1382. 189(2):587–599.
52. Nguyen KL, Bandettini WP, Shanbhag S, Leung SW, Wilson JR, Arai AE. 73. Levine GN, Gomes AS, Arai AE, et al. Safety of magnetic resonance
Safety and tolerability of regadenoson CMR. Eur Heart J Cardiovasc imaging in patients with cardiovascular devices: an American Heart
Imaging. 2014;15(7):753–760. Association scientific statement from the Committee on Diagnostic and
53. Pennell DJ, Underwood SR, Ell PJ. Safety of dobutamine stress for Interventional Cardiac Catheterization, Council on Clinical Cardiology,
thallium-201 myocardial perfusion tomography in patients with asthma. and the Council on Cardiovascular Radiology and Intervention: endorsed
Am J Cardiol. 1993;71(15):1346–1350. by the American College of Cardiology Foundation, the North American
54. Monsieurs KG, Nolan JP, Bossaert LL, et al. European Resuscitation Society for Cardiac Imaging, and the Society for Cardiovascular Magnetic
Council Guidelines for Resuscitation 2015: Section 1. Executive Resonance. Circulation. 2007;116(24):2878–2891.
Summary. Resuscitation. 2015;95:1–80. 74. Matsumoto AH, Teitelbaum GP, Barth KH, Carvlin MJ, Savin MA,
55. Sellden H, de Chateau P, Ekman G, Linder B, Saaf J, Wahlund LO. Strecker EP. Tantalum vascular stents: in vivo evaluation with MR
Circulatory monitoring of children during anaesthesia in low-field imaging. Radiology. 1989;170(3 Pt 1):753–755.
magnetic resonance imaging. Acta Anaesthesiol Scand. 1990;34(1): 75. Santoro D, Winter L, Muller A, et al. Detailing radio frequency heating
41–43. induced by coronary stents: a 7.0 tesla magnetic resonance study. PLoS
56. Lindberg LG, Ugnell H, Oberg PA. Monitoring of respiratory and heart One. 2012;7(11):e49963.
rates using a fibre-optic sensor. Med Biol Eng Comput. 1992;30(5): 76. Gerber TC, Fasseas P, Lennon RJ, et al. Clinical safety of magnetic
533–537. resonance imaging early after coronary artery stent placement. J Am Coll
57. Roth JL, Nugent M, Gray JE, et al. Patient monitoring during magnetic Cardiol. 2003;42(7):1295–1298.
resonance imaging. Anesthesiology. 1985;62(1):80–83. 77. Nagel E, Hug J, Bunger S, et al. Coronary flow measurements for
58. Felblinger J, Boesch C. Amplitude demodulation of the evaluation of patients after stent implantation. MAGMA. 1998;6(2–3):
electrocardiogram signal (ECG) for respiration monitoring and 184–185.
compensation during MR examinations. Magn Reson Med. 78. Kramer CM, Rogers WJ, Reichek N. Magnetic resonance contrast
1997;38(1):129–136. enhancement versus dobutamine tagging response for assessment
59. Felblinger J, Lehmann C, Boesch C. Electrocardiogram acquisition of myocardial viability after infarction. J Am Coll Cardiol. 1999;33
during MR examinations for patient monitoring and sequence (suppl a):485A.
triggering. Magn Reson Med. 1994;32:523–529. 79. Al-Saadi N, Nagel E, Gross M, et al. Improvement of myocardial
60. Felblinger J, Debatin JF, Boesch C, Gruetter R, McKinnon GC. perfusion reserve early after coronary intervention: assessment with
Synchronization device for electrocardiography-gated echo-planar cardiac magnetic resonance imaging. J Am Coll Cardiol. 2000;36(5):
imaging. Radiology. 1995;197(1):311–313. 1557–1564.
61. Fischer SE, Wickline SA, Lorenz CH. Novel real-time R-wave detection 80. Duerinckx AJ, Atkinson D, Hurwitz R, Mintorovitch J, Whitney W.
algorithm based on the vectorcardiogram for accurate gated magnetic Coronary MR angiography after coronary stent placement. AJR Am J
resonance acquisitions. Magn Reson Med. 1999;42(2):361–370. Roentgenol. 1995;165(3):662–664.
62. Chia JM, Fischer SE, Wickline SA, Lorenz CH. Performance of QRS 81. Kotsakis A, Tan KH, Jackson G. Is MRI a safe procedure in patients with
detection for cardiac magnetic resonance imaging with a novel coronary stents in situ? Int J Clin Pract. 1997;51(6):349.
vectorcardiographic triggering method. J Magn Reson Imaging. 82. Shellock FG, Giangarra CJ. In vitro assessment of 3-T MRI issues for a
2000;12(5):678–688. bioabsorbable, coronary artery scaffold with metallic markers. Magn
63. Jorgensen NH, Messick JM Jr, Gray J, Nugent M, Berquist TH. ASA Reson Imaging. 2014;32(2):163–167.
monitoring standards and magnetic resonance imaging. Anesth Analg. 83. Wiebe J, Nef HM, Hamm CW. Current status of bioresorbable scaffolds
1994;79(6):1141–1147. in the treatment of coronary artery disease. J Am Coll Cardiol. 2014;
64. Davis PL, Crooks L, Arakawa M, McRee R, Kaufman L, Margulis AR. 64(23):2541–2551.
Potential hazards in NMR imaging: heating effects of changing magnetic 84. Shellock F. Pocket Guide to MR Procedures and Metallic Objects: Update
fields and RF fields on small metallic implants. AJR Am J Roentgenol. 1998. Philadelphia: Lippincott-Raven; 1998.
1981;137(4):857–860. 85. Shellock FG. Biomedical implants and devices: assessment of magnetic
65. Laakman RW, Kaufman B, Han JS, et al. MR imaging in patients with field interactions with a 3.0-tesla MR system. J Magn Reson Imaging.
metallic implants. Radiology. 1985;157(3):711–714. 2002;16(6):721–732.
66. Mechlin M, Thickman D, Kressel HY, Gefter W, Joseph P. Magnetic 86. Hassler M, Le Bas JF, Wolf JE, Contamin C, Waksmann B, Coulomb M.
resonance imaging of postoperative patients with metallic implants. AJR Effects of the magnetic field in magnetic resonance imaging on 15 tested
Am J Roentgenol. 1984;143(6):1281–1284. cardiac valve prostheses. J Radiol. 1986;67(10):661–666.
67. Sigwart U, Puel J, Mirkovitch V, Joffre F, Kappenberger L. Intravascular 87. Soulen RL, Budinger TF, Higgins CB. Magnetic resonance imaging of
stents to prevent occlusion and restenosis after transluminal angioplasty. prosthetic heart valves. Radiology. 1985;154(3):705–707.
N Engl J Med. 1987;316(12):701–706. 88. Globits S, Higgins CB. Assessment of valvular heart disease by magnetic
68. Fischman DL, Leon MB, Baim DS, et al. A randomized comparison of resonance imaging. Am Heart J. 1995;129(2):369–381.
coronary-stent placement and balloon angioplasty in the treatment of 89. Bachmann R, Deutsch HJ, Jungehulsing M, Sechtem U, Hilger HH,
coronary artery disease. Stent Restenosis Study Investigators. N Engl J Schicha H. Magnetic resonance tomography in patients with a heart
Med. 1994;331(8):496–501. valve prosthesis. Rofo. 1991;155(6):499–505.
CHAPTER 10  Special Considerations for Cardiovascular Magnetic Resonance 117.e3

90. Di Cesare E, Enrici RM, Paparoni S, et al. Low-field magnetic resonance 105. Naehle CP, Meyer C, Thomas D, et al. Safety of brain 3-T MR imaging
imaging in the evaluation of mechanical and biological heart valve with transmit-receive head coil in patients with cardiac pacemakers:
function. Eur J Radiol. 1995;20(3):224–228. pilot prospective study with 51 examinations. Radiology. 2008;249(3):
91. Saeedi M, Thomas A, Shellock FG. Evaluation of MRI issues at 3-tesla 991–1001.
for a transcatheter aortic valve replacement (TAVR) bioprosthesis. Magn 106. Brignole M, Auricchio A, Baron-Esquivias G, et al. 2013 ESC guidelines
Reson Imaging. 2015;33(4):497–501. on cardiac pacing and cardiac resynchronization therapy: the task force
92. Abdel-Wahab M, Mehilli J, Frerker C, et al. Comparison of balloon- on cardiac pacing and resynchronization therapy of the European
expandable vs self-expandable valves in patients undergoing Society of Cardiology (ESC). Developed in collaboration with the
transcatheter aortic valve replacement: the CHOICE randomized clinical European Heart Rhythm Association (EHRA). Eur Heart J. 2013;
trial. JAMA. 2014;311(15):1503–1514. 34(29):2281–2329.
93. Merten C, Beurich HW, Zachow D, et al. Aortic regurgitation and left 107. Nazarian S, Hansford R, Roguin A, et al. A prospective evaluation of a
ventricular remodeling after transcatheter aortic valve implantation: a protocol for magnetic resonance imaging of patients with implanted
serial cardiac magnetic resonance imaging study. Circ Cardiovasc Interv. cardiac devices. Ann Intern Med. 2011;155(7):415–424.
2013;6(4):476–483. 108. Gimbel JR. Magnetic resonance imaging of implantable cardiac rhythm
94. Hartlage GR, Babaliaros VC, Thourani VH, et al. The role of devices at 3.0 tesla. Pacing Clin Electrophysiol. 2008;31(7):795–801.
cardiovascular magnetic resonance in stratifying paravalvular leak 109. Sheldon SH, Bunch TJ, Cogert GA, et al. Multicenter study of the safety
severity after transcatheter aortic valve replacement: an observational and effects of magnetic resonance imaging in patients with coronary
outcome study. J Cardiovasc Magn Reson. 2014;16:93. sinus left ventricular pacing leads. Heart Rhythm. 2015;12(2):345–349.
95. Crouch G, Tully PJ, Bennetts J, et al. Quantitative assessment of 110. Gold MR, Sommer T, Schwitter J, et al. Full-body MRI in patients with
paravalvular regurgitation following transcatheter aortic valve an implantable cardioverter-defibrillator: primary results of a
replacement. J Cardiovasc Magn Reson. 2015;17:32. randomized study. J Am Coll Cardiol. 2015;65(24):2581–2588.
96. Mohrs OK, Wunderlich N, Petersen SE, Pottmeyer A, Kauczor HU. 111. Schwitter J, Gold MR, Al Fagih A, et al. Image quality of cardiac
Contrast-enhanced CMR in patients after percutaneous closure of the magnetic resonance imaging in patients with an implantable
left atrial appendage: a pilot study. J Cardiovasc Magn Reson. 2011; cardioverter defibrillator system designed for the magnetic resonance
13:33. imaging environment. Circ Cardiovasc Imaging. 2016;9(5).
97. Romero J, Perez IE, Krumerman A, Garcia MJ, Lucariello RJ. Left atrial 112. Blaschke F, Lacour P, Walter T, et al. Cardiovascular magnetic resonance
appendage closure devices. Clin Med Insights Cardiol. 2014;8:45–52. imaging in patients with an implantable loop recorder. Ann Noninvasive
98. Hong SN, Rahimi A, Kissinger KV, Pedrosa I, Manning WJ, O’Halloran Electrocardiol. 2016;21(3):319–324.
TD. Cardiac magnetic resonance imaging and the WATCHMAN device. 113. Gimbel JR, Zarghami J, Machado C, Wilkoff BL. Safe scanning, but
J Am Coll Cardiol. 2010;55(24):2785. frequent artifacts mimicking bradycardia and tachycardia during
99. Krumm P, Zuern CS, Wurster TH, et al. Cardiac magnetic resonance magnetic resonance imaging (MRI) in patients with an implantable loop
imaging in patients undergoing percutaneous mitral valve repair with recorder (ILR). Ann Noninvasive Electrocardiol. 2005;10(4):404–408.
the MitraClip system. Clin Res Cardiol. 2014;103(5):397–404. 114. Wong JA, Yee R, Gula LJ, et al. Feasibility of magnetic resonance imaging
100. Radunski UK, Franzen O, Barmeyer A, et al. Cardiac remodeling in patients with an implantable loop recorder. Pacing Clin Electrophysiol.
following percutaneous mitral valve repair—initial results assessed by 2008;31(3):333–337.
cardiovascular magnetic resonance imaging. Rofo. 2014;186(10): 115. Nordbeck P, Ertl G, Ritter O. Magnetic resonance imaging safety in
951–958. pacemaker and implantable cardioverter defibrillator patients: how far
101. Lurz P, Serpytis R, Blazek S, et al. Assessment of acute changes in have we come? Eur Heart J. 2015;36(24):1505–1511.
ventricular volumes, function, and strain after interventional edge-to- 116. Gimbel JR, Bello D, Schmitt M, et al. Randomized trial of pacemaker
edge repair of mitral regurgitation using cardiac magnetic resonance and lead system for safe scanning at 1.5 tesla. Heart Rhythm.
imaging. Eur Heart J Cardiovasc Imaging. 2015;16(12):1399–1404. 2013;10(5):685–691.
102. Holmes DR Jr, Hayes DL, Gray JE, Merideth J. The effects of magnetic 117. Higgins JV, Sheldon SH, Watson RE Jr, et al. “Power-on resets” in cardiac
resonance imaging on implantable pulse generators. Pacing Clin implantable electronic devices during magnetic resonance imaging.
Electrophysiol. 1986;9(3):360–370. Heart Rhythm. 2015;12(3):540–544.
103. Hayes DL, Holmes DR Jr, Gray JE. Effect of 1.5 tesla nuclear magnetic 118. Bailey WM, Rosenthal L, Fananapazir L, et al. Clinical safety of the
resonance imaging scanner on implanted permanent pacemakers. J Am ProMRI pacemaker system in patients subjected to head and lower
Coll Cardiol. 1987;10(4):782–786. lumbar 1.5-T magnetic resonance imaging scanning conditions. Heart
104. Cohen JD, Costa HS, Russo RJ. Determining the risks of magnetic Rhythm. 2015;12(6):1183–1191.
resonance imaging at 1.5 tesla for patients with pacemakers and 119. Mollerus M, Albin G, Lipinski M, Lucca J. Magnetic resonance imaging
implantable cardioverter defibrillators. Am J Cardiol. 2012;110(11): of pacemakers and implantable cardioverter-defibrillators without
1631–1636. specific absorption rate restrictions. Europace. 2010;12(7):947–951.
11 
Pacemaker and Implantable Cardioverter-
Defibrillator Safety and Safe Scanning
Esra Gucuk Ipek and Saman Nazarian

Magnetic resonance imaging (MRI) has gained popularity over the last upon the intended design and prior experience with the CIED in an
decade because of its excellent soft tissue imaging capability and superior MRI environment (Table 11.1). Potential interactions between CIED
spatial resolution as well as lack of ionizing radiation. These properties systems and the MRI environment are summarized in Table 11.2. The
have led to expanded indications for MRI. At the same time, with the static magnetic field, in addition to radiofrequency (RF) energy gen-
aging of the US population, the need for cardiac implantable electronic erated during MRI, may interact with the CIED and lead to adverse
devices (CIEDs) is increasing in parallel to that for MRI indication. effects. The magnetic field strength, RF power, patient position/
Despite its advantages, MRI is relatively contraindicated in individu- orientation within the scanner, CIED type, and patient character-
als with CIEDs because of the possibility for heating, induction, and istics are all important variables that help quantify the magnitude
electromagnetic interference. This is a public health issue because it of interaction.8
limits diagnostic access to MRI for millions of CIED recipients and
ever-increasing trends for device implantation.1,2 According to a recent CLINICAL STUDIES WITH NONMAGNETIC
61-country survey, there was a significant rise in the use of implantable RESONANCE IMAGING CONDITIONAL DEVICES
cardioverter-defibrillators (ICDs) by the year 2009 compared with 2005,
with most implants occurring in the United States (434 new implants Up to 1.5 T
per million population).3 Moreover, the high prevalence of comorbidities Clinical studies of MRI in the setting of nonconditional devices are
in the same population indicates potential benefits from MRI. Within summarized in Table 11.3. The first study was performed in 1984 by
1-year post implant, 17% of the CIED recipients have an MRI indi- Fetter et al.,9 who examined the in vivo effects of MRI on CIED func-
cation, and estimates indicate a lifetime need for MRI in up to 75% tion.9 Although device parameters did not change, an asynchronous
of recipients.4–6 pacing mode was activated during MRI scanning in one patient. After
Owing to an increased awareness of the public health importance this initial experience, in 1996, Gimbel et al.10 examined five patients
of this issue, device manufacturers have focused on the development with pacemakers undergoing 0.5 T MRI scan and observed a 2-second
of MRI conditional CIEDs, device systems that demonstrate no known pause in a pacemaker-dependent patient. In 2000, Sommer et al.11 evalu-
hazards in a specified MRI environment with specified conditions of ated the effects of 0.5 T MRI on pacemaker generators and leads in
use. Most important, to be considered MRI conditional, all implanted vitro, then examined 44 non–pacemaker-dependent patients in vivo,
CIED components must be tested together for electromagnetic compat- and reported no adverse events. The following year, the same group
ibility and approved for such use by the US Food and Drug Adminis- reported a prospective study that investigated safety at 0.5 T in 32 pace-
tration (FDA). Other CIED systems are considered to be “legacy” systems maker patients undergoing MRI.12 Lead impedance, sensing thresholds
or “nonconditional” for MRI. did not change after MRI. They showed overall safety; however, battery
A decade ago, the presence of a CIED was considered an absolute voltage decreased immediately after MRI, and returned to baseline at
contraindication for MRI. However, this approach was challenged with the 3-month follow-up.
numerous case series demonstrating no significant safety issues when In the last decade, several studies have examined the interaction of
MRI was performed with specific protocols to minimize electromagnetic nonconditional CIED systems with MRI. Roguin et al.13 studied CIED
interference.7 Today, we know that MRI examination is feasible and behavior and changes in system parameters in vitro and in vivo in
can be performed with minimal risk as long as a well-defined and tested animal models. Older (pre-2000) ICD systems were damaged following
safety protocol is followed. According to current guidelines, MRI exami- MRI, but no significant changes were observed in newer (2000 and
nation should be avoided in CIED recipients, but, if necessary, it can later) systems. Pathologic examination showed limited necrosis or fibrosis
be done in experienced centers under specific protocols. adjacent to the lead tip that was not different than that observed in
In this chapter, we review the current data on nonconditional and animals that were not exposed to MRI. Later, Martin et al. performed
conditional CIEDs and experiences with MRI examination, summarize 62 MRI examinations in 54 pacemaker patients. There were no adverse
the Johns Hopkins protocol for MRI safety in the setting of noncon- events; but changes in pacing threshold were observed in 10 leads, 2 of
ditional CIED systems, and review associated MRI-related artifacts. which required a change in programmed output.7
Because of significant adverse events reported with older-generation
CARDIAC IMPLANTED ELECTRONIC DEVICES AND ICDs, it was first thought that MRI was an absolute contraindication
in the setting of ICD systems.14 Junttila et al.15 performed three serial
MAGNETIC RESONANCE IMAGING INTERACTIONS cardiac MRI scans in 10 ICD recipients. In that series, patients reported
Owing to potential interactions with the electromagnetic environ- no symptoms during the MRI examination, and none of the examina-
ment, CIEDs are classified as MRI unsafe or MRI conditional based tions required early termination. No adverse event or change in device/

118
CHAPTER 11  Pacemaker and Implantable Cardioverter-Defibrillator Safety and Safe Scanning 119

troponin levels increased, which was associated with the increased pacing
TABLE 11.1  American Society for Testing
threshold in one case. Decreased battery voltage was again noted imme-
and Materials Categorization of Medical diately after the MRI; but this drop was transient in most cases and
Devices in a Magnetic Resonance Imaging returned to baseline at follow-up. Naehle et al.19 evaluated the effects
Environment of serial MRI in 47 CIED recipients. There was a statistically significant
Category Description decrease in battery voltage and pacing capture threshold after two or
more MRI examinations, but no clinically relevant or cumulative changes
MR safe No known hazards in an MR environment
that would affect device longevity were noted. In 2011, the same group
MR conditional No known hazards in a specified MR environment
reported another series pacemaker and ICD patients that underwent
with specified conditions
safe cardiovascular magnetic resonance (CMR) imaging without any
MR unsafe Known to pose hazards in all MR environments
change in pacing capture threshold, lead impedance, or troponin levels.20
MR, Magnetic resonance. Cardiac resynchronization therapy and coronary sinus leads also
appear to be safe.21 Little is known about the safety of epicardial leads.
In a study by Pulver et al.,22 11 patients with pediatric and adult congenital
TABLE 11.2  Potential Interactions heart disease underwent 1.5 T MRI, including 6 patients with epicardial
Between a Cardiac Implantable Electronic leads. There were no adverse events such as symptoms that could be
Device and Electromagnetic Interference related to lead heating. Of note, three patients with epicardial leads
Force and torque • Magnetic fields can attract ferromagnetic underwent CMR without any safety issue.
materials within the CIEDs that may lead to Cohen et al.23 performed 125 clinically indicated MRI examinations
generator dislodgment in 109 patients with CIED, including ICD systems, and compared the
• The torque is directly related to ferromagnetic outcomes with 50 CIED recipients who did not have an MRI. The
content, distance between ferromagnetic authors did not report MRI-related adverse events; however, they did
material and magnetic source, and field observe small nonsignificant changes in device parameters after MRI
strength of the magnetic field that were similar to their control group. Such insignificant changes may
• In vitro studies showed that maximum torque be unrelated to MRI itself and reflect random physiologic variations
is 0.98 Newton with a 1.5 T scanner13 in the interaction CIED tissue interface.
Induced electrical • Electrical currents induced by MRI-related Muehling et al.24 also examined 356 CIED recipients who underwent
current electromagnetic components may theoretically 1.5 T cranial MRI. There was no significant change in device parameters,
initiate ventricular or atrial arrhythmias61 and troponin levels were stable immediately after MRI. Moreover, patients
Heating and tissue • The pacing leads can act as an antenna under were followed for 1 year and device interrogations were performed at
damage an electromagnetic field, resulting in heating 2 weeks, 2 months, 6 months, and 12 months after the MRI. No significant
and tissue damage around the tip of the lead. change was observed in device-related parameters during follow-up.
The eventual tissue damage may change Finally, the MagnaSafe Registry is a multicenter, prospective cohort
pacing, sensing, or capture thresholds29 study designed to evaluate the safety of non-MRI conditional CIED
• Retained/abandoned or epicardial leads may systems following nonthoracic 1.5 T MRI.25 The MagnaSafe Registry
carry a higher risk of local heating included 1500 cases in which patients had a nonthoracic MRI with a
Reed switch activity • Reed switch activity of CIED may lead to pacemaker (n = 1000) or ICD (n = 500) implanted after 2001. Patients
asynchronous pacing62 with abandoned or inactive lead were excluded. There were no deaths,
Inappropriate function • Radiofrequency energy pulses may lead to lead failures, losses of capture, or ventricular arrhythmias during the
inappropriate inhibition of demand pacing, index MRI. Prespecified changes in lead impedance, pacing threshold,
tracking, or programming changes battery voltage, and P-wave and R-wave amplitude were exceeded in a
• Sensing of electromagnetic interference may small number of cases. Repeat MRI was not associated with an increase
be interpreted as an arrhythmia and may lead in adverse events.25
to attempts by the device to deliver In addition, another large study from our Johns Hopkins group
antitachycardia therapies reported on MRI safety at 1.5 T in 1509 patients who had a pacemaker
(58%) or ICD (42%) and underwent 2103 thoracic and nonthoracic
CIED, Cardiac implantable electronic device; MRI, magnetic MRI. The pacing mode was changed to asynchronous mode for pacing-
resonance imaging. dependent patients and to demand mode for others. No long-term
clinically significant adverse events were reported. In 9 (0.4%) MRI
lead parameters was observed during short-term and long-term follow- examinations, the device reset to a backup mode, which was transient
up. Similarly, in another small cohort of 10 ICD recipients, there were in all but one device. An acute decrease in P wave amplitude occurred
no adverse events after 1.5 T MRI.16 Naehle et al.17 performed 1.5 T in 1% of studies that increased to 4% of patients on long-term follow-
MRI in 18 ICD recipients. Although no significant pacing threshold up. The observed changes in lead parameters were not clinically sig-
and lead impedance changes were observed, battery voltage significantly nificant and did not require device revision or reprogramming.25a
decreased. In a subset of patients, the voltage recovered completely; There are limited data on high-risk groups such as pacemaker-
however, in the remaining patients, decreased battery voltage persisted. dependent patients. Gimbel et al.26 examined 10 pacemaker-dependent
Moreover, RF noise was oversensed as ventricular fibrillation (VF), but patients who underwent 1.5 T MRI. There were no clinical adverse
no therapies were initiated. There was no evidence of overheating or events; however, minor capture threshold changes were observed in 7
tissue damage as assessed by troponin level. patients. Higgins et al.27 reviewed 198 CIED recipients who underwent
Sommer et al.18 performed 115 nonthoracic 1.5 T MRI examinations 1.5 T MRI. Power-on reset events occurred in 8 patients, all patients
in 85 patients with CIED. There were no arrhythmias, but pacing capture having older-generation devices. Importantly, devices reset to an inhibited
threshold increased significantly. After four MRI examinations, serum pacing mode. Finally, 137 (9%) of the 1509 patients in the previously
120 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TABLE 11.3  Clinical Studies With Nonmagnetic Resonance Imaging Conditional Devices
Study Group Year Patient No. Device MRI Scanner Findings
10
Gimbel et al. 1996 5 Pacemaker 0.5 T Pause detected in a pacemaker-dependent patient with unipolar lead
Sommer et al.11 2000 44 Pacemaker 0.5 T No adverse events; no change in device and lead parameters
Vahlhaus et al.12 2001 32 Pacemaker 0.5 T Battery voltage decreased after MRI that turned back to baseline in
3 months; reed switch deactivation was observed in 37.5% of the
patients
Martin et al.7 2004 54 Pacemaker 1.5 T Pacing threshold was changed in 9.4% of the leads
Del Ojo et al.33 2005 13 Pacemaker 2.0 T No adverse events; no change in device and lead parameters
Gimbel et al.63 2005 7 ICD 1.5 T Power-on reset occurred in 1 patient
Gimbel et al.26 2005 10 Pacemaker 1.5 T Minor changes in capture thresholds
Nazarian et al.40 2006 55 Pacemaker and ICD 1.5 T No adverse events; no change in device and lead parameters
Sommer et al.18 2006 82 Pacemaker 1.5 T Capture threshold was increased; increased troponin levels in 4
cases. One of the capture threshold increases was associated with
elevated troponin levels
Gimbel et al.35 2008 14 Pacemaker, ICD and loop 3.0 T No change in device and lead parameters. One patient reported
recorder chest pain
Naehle et al.34 2008 44 Pacemaker 3.0 T No adverse events; no change in device and lead parameters
Mollerus et al.64 2008 37 Pacemaker and ICD 1.5 T No adverse events; no change in device and lead parameters
Naehle et al.17 2009 18 ICD 1.5 T A decrease was observed in battery voltage; electromagnetic
interference was interpreted as VF in 2 cases
Naehle et al.19 2009 47 Pacemaker 1.5 T Pacing threshold and battery decreased after serial MRI
Pulver et al.22 2009 11 Pacemaker 1.5 T Minimal change in device voltage; lead threshold; lead impedance in
6 patients
Mollerus et al.65 2009 52 Pacemaker and ICD 1.5 T No adverse events; no change in device and lead parameters
Halshtok et al.66 2010 18 Pacemaker and ICD 1.5 T Power-on reset occurred in 2 patients
Strach et al.32 2010 114 Pacemaker 0.2 T No adverse events; no change in device and lead parameters
Burke et al.16 2010 38 Pacemaker and ICD 1.5 T No adverse events; no change in device and lead parameters
Mollerus et al.67 2010 103 Pacemaker and ICD 1.5 T Sensing amplitudes and pacing impedances decreased significantly
Nazarian et al.6 2011 438 Pacemaker and ICD 1.5 T Power-on reset in 1.5% of the patients
Juntilla et al.15 2011 10 ICD 1.5 T No adverse events; no change in device parameters
Naehle et al.20 2011 32 Pacemaker and ICD 1.5 T No adverse events; no change in device and lead parameters
Cohen et al.23 2012 109 Pacemaker and ICD 1.5 T Battery voltage decreased in 4%; pacing threshold increased in 3%;
pacing lead impedance changed in 6%
Muehling et al.24 2014 356 Pacemaker 1.5 T No adverse events; no change in device and lead parameters
Higgins et al.27 2015 198 Pacemaker and ICD 1.5 T Power-on reset in 8 patients
Sheldon et al.21 2015 42 CRT-D, CRT-P, pacemaker 1.5 T No adverse events; no change in device and lead parameters
Russo et al.25 2017 1500 Pacemaker and ICD 1.5 T No deaths or lead failures; one ICD generator was replaced because
it could not be interrogated after MRI
Nazarian et al.25a 2017 1509 Pacemaker and ICD 1.5 T No deaths or lead failures; one pacemaker needed replacement
because it could not be reprogrammed; no clinically significant
change in lead parameters

CRT-D, Cardiac resynchronization therapy defibrillator; CRT-P, cardiac resynchronization therapy pacemaker; ICD, implantable cardioverter
defibrillator; MRI, magnetic resonance imaging.

mentioned study of Nazarian et al.25a were pacer-dependent (includ- same study group evaluated troponin levels in 348 patients, including
ing 22 of whom had an ICD with asynchronous programming mode 22 patients with abandoned leads. There was no change in troponin
capability). There was similar safety as in the overall study. Thus MRI levels in these patients, which may serve as a surrogate for myocardial
examination should be carefully considered and closely monitored in thermal damage. Finally, low-field (0.2 T) MRI was performed in 114
pacemaker-dependent patients with older (pre-2000) devices. CIED patients who required MRI examination for urgent indications,
including pacemaker-dependent patients and patients with abandoned
Retained/Orphaned Leads leads.32 No adverse events or changes in device parameters were reported.
Patients with retained/orphaned leads constitute another high-risk
group, because of the possibility of increased heating. An in vitro study High-Field Magnetic Resonance
compared retained/orphaned leads with pacemaker-attached leads, and There are limited studies with high-field MRI. Del Ojo et al.33 performed
showed that heating of the retained lead was significantly higher, and 2 T MRI in 13 patients with pacemakers. This is one of the initial studies
the risk was significantly correlated with lead length.28,29 In another that showed MRI can be safely performed in CIED recipients, even
study by Higgins et al.,30,31 the safety of retained pacemaker lead was though the magnetic field is relatively high. 3 T MRIs are most com-
evaluated in 19 patients who underwent nonthoracic 1.5 T MRI. There monly used in neurologic imaging. Because of the increased magnetic
were no adverse events and in a subset of patients who had their devices field strength, some risks are theoretically higher. However, neurologic
reconnected to the retained leads, lead parameters were unchanged. The imaging with transmit-receive head coils decreases power deposition
CHAPTER 11  Pacemaker and Implantable Cardioverter-Defibrillator Safety and Safe Scanning 121

upon the CIED and mitigates the risk of heating and current generation.
TABLE 11.4  Magnetic Resonance Imaging
In contrast, the CIED will be exposed to more force and torque. Naehle
et al.34 evaluated brain 3 T MRI in CIED recipients. No safety issues,
Conditional Cardiac Implantable Electronic
changes in device parameters, or changes in troponin levels occurred in Devices and Leads
the 44 patients who underwent 55 brain examinations at 3 T. Similarly, Company Device Lead
Gimbel et al.35 safely performed 3 T MRI in 14 patients without adverse Medtronic Revo MRI SureScan CapSureFix/CapsureSense
effects or changes in device parameters. However, in the following year, Advisa MRI Medtronic Sprint Quattro
Gimbel et al.36,37 reported two cases of pacing inhibition with 3 T MRI. Evera MRI SureScan Secure MRI
St. Jude Medical Accent MRI Tendril MRI
Recommendations
Biotronik ProMRI Safio S53/Safio S60
According to current guidelines, patients with MRI nonconditional Lumax 740 Setrox
devices may undergo 1.5 T MRI if the benefit of the MRI overweighs ProMRI ICD Linoxsmart
the risk of the procedure, provided that the CIED is a new-generation Boston Scientific Ingenio Ingevity
device and appropriate supervision is provided.8,38,39 The following are Advantio
recommended by the guidelines.38 Sorin Kora 100 Beflex
1. MRI can be performed if there is no alternative imaging modality,
the CIED is a new-generation system that has been implanted for ICD, Implantable cardioverter defibrillator; MRI, magnetic resonance
at least 6 weeks, and there are no abandoned or epicardial leads. imaging.
2. The CIED should be interrogated before examination, and system
parameters should be adjusted according to patient and device
characteristics. currently available MRI conditional systems are summarized in Table
3. During the MRI examination, experienced personnel should perform 11.4. Those systems pose no known hazards when MRI is performed
continuous monitoring. under specific monitoring conditions. The use of ferromagnetic materi-
4. Device interrogation should be performed after the scan. als is limited when possible; and lead construction materials and internal
circuits are modified. Instead of a reed switch, a Hall sensor is used.
Inappropriate sensing is prevented by filters. There is also a specific
THE JOHNS HOPKINS PROTOCOL device module for MRI mode that enables limited interaction with
Our institutional safety protocol is based upon our experience with over electromagnetic field. With these systems, 1.5 T MRI can safely be per-
2000 MRI examinations since 2003. The protocol has been published formed by following manufacturer specific instructions (class IIa, level
previously and is summarized in Fig. 11.1.40 First, we acquire all the of evidence B).38
CIED-related information, such as the implantation date, type of the The cumulating experience with conditional MRI devices is sum-
device, number of leads, and presence of abandoned or epicardial leads, marized in Table 11.5. In the randomized study performed by Wilkoff
and assess pacemaker dependence. We exclude patients with epicardial et al.41 the safety and efficacy of the EnRhythm MRI SureScan and
and/or abandoned leads. In all examinations, an electrophysiologist or CapSureFix leads was tested under 1.5 T MRI. A total of 464 patients
an experienced registered nurse prepared to respond to device mal- were randomized to either MRI (n = 258) versus control (n = 206)
function needs to be present. The pacemakers are programmed to an groups. There were no MRI-related complications, and electrical param-
inhibited mode (DDI/VVI) or, if the patient is pacemaker dependent, to eters before and after MRI were similar between the two groups. No
asynchronous mode (DOO/VOO). Other pacemaker functions such as MRI-induced power-on reset was observed. Of note, the EnRhythm
rate response or tachyarrhythmia therapies are deactivated. The electro- MRI SureScan and CapSureFix leads were designed for 1.5 T MRI with
cardiogram and pulse oximetry signals are monitored carefully during a maximum specific absorption rate (SAR) value of 2 W/kg. Accord-
MRI, and pacemaker interrogation is performed after the examination. ingly, the FDA approved the EnRhythm MRI SureScan Pacing System
Using this protocol, we initially reported on 55 patients with CIED, (Medtronic, Minneapolis, MN) in 2011. The SureScan and CapSureFix
all of whom underwent MRI without safety issues.40 Later, we reported leads are not approved at higher field strengths.
on 555 1.5 T MRI examinations (16% cardiac) in 438 patients (12% Forleo et al.42 evaluated the safety and feasibility of the EnRhythm
pacemaker dependent) with CIED (54% pacemakers, 46% defibrillators) MRI SureScan and CapSureFix leads in 50 patients and compared the
systems. The major adverse event was power-on reset, which occurred results with a conventional CIED group. The implantation success rates
in 0.7% of the patients.6 Minor changes were observed in lead sensing and complication rates were similar in both groups, and during follow-
and capture thresholds, but none of these required system revision or up there were no significant changes in pacing thresholds. In the MRI
reprogramming. Our most recent report included MRI safety at 1.5 T conditional group, cephalic veins were less likely to be accessed because
in 1509 patients who had a pacemaker (58%) or ICD (42%) and under- of the thickness of conditional leads. Later, the same study group reported
went 2103 thoracic and nonthoracic MRI. No long-term clinically their experience with MRI-conditional CIED systems with a larger
significant adverse events were reported. In 9 (0.4%) MRI examinations, cohort of patients (n = 250). In this study, MRI conditional CIED
the device reset to a backup mode, which was transient in all but one systems from three different manufacturers were implanted in 250
device. An acute decrease in P wave amplitude occurred in 1% of studies patients without a significant difference in complication rates compared
that increased to 4% of patients on long-term follow-up. The observed with conventional devices.43
changes in lead parameters were not clinically significant and did not Rickard et al.44 compared the procedural and long-term performance
require device revision or reprogramming.25a of the 5086 MRI conditional leads (n = 466) with conventional leads
(n = 316). Acute lead dislodgment was higher with MRI conditional
MAGNETIC RESONANCE IMAGING leads (2.6% vs. 0.6%). At 12-month follow-up, ventricular sensing was
lower and ventricular capture threshold was higher with MRI conditional
CONDITIONAL DEVICES leads compared with controls. In a smaller study by Elmouchi et al.,45
Given the increased need for MRI examinations in CIED recipients, 65 patients implanted with MRI conditional leads were compared with
device companies have introduced MRI conditional systems. The 92 patients with conventional leads. Complication rates were higher in
122 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Type of Implantable Cardiac


Device

Pacemaker ICD

Proceed to NO ICD generator NO Proceed to


Alternative Test Peacemaker Alternative Test
implanted after
implanted after 1998?
2000?

YES YES

Patient has:
- Recently implanted leads (<6 weeks before MRI)? and/or YES
- Abandoned leads? and/or
- Nontransvenous epicardial leads?

NO

INTERROGATE Device
Record device variables for comparison after MRI
BEFORE MRI PROCEDURE

- Lead impedance and threshold


- P/R wave amplitude
- Battery voltage

Pacemaker ICD

Patient pace- Patient pace- YES


dependent? dependent?

YES NO NO

DEACTIVATE
REPROGRAM
REPROGRAM Tachyarrhythmia
VOO/DOO
VOO/DOO Therapies
(Asynchronous)
(Asynchronous) (ATP/Defibrillation)

REPROGRAM
VVI/DDI
(Inhibited)

DEACTIVATE
- Magnet
- Rate
- PVC
- Noise
- Ventricular sense
- Conducted atrial fibrillation response
PROCEDURE
DURING MRI

MONITOR
- Blood pressure
- ECG
- Oxygen saturation
- Symptoms
PROCEDURE

+ RECHECK device parameters


AFTER MRI

- Lead impedance and threshold


- P/R wave amplitude
- Battery voltage
+ RESTORE original programming
+ FOLLOW-UP interrogation in 3−6 months

FIG. 11.1  The safety protocol of Johns Hopkins Hospital for magnetic resonance imaging (MRI) scanning
of the patients with a cardiac implantable electronic device. ATP, Antitachycardia pacing; DDI, dual-chamber
inhibited pacing without atrial tracking; DOO, dual-chamber asynchronous pacing; ECG, electrocardiogram;
ICD, implantable cardioverter defibrillator; PVC, premature ventricular contraction; VOO, ventricular asynchro-
nous pacing; VVI, ventricular inhibited pacing.
CHAPTER 11  Pacemaker and Implantable Cardioverter-Defibrillator Safety and Safe Scanning 123

TABLE 11.5  Previous Studies With Magnetic Resonance Imaging Conditional Devices
Study Group Year Patient No. Device MRI Scanner Findings
42
Forleo et al. 2010 107 MRI conditional (EnRhythm) vs. None Successfully implanted without significant
conventional dual chamber pacemaker procedural complications
Wilkoff et al.41 2011 464 MRI conditional (EnRhythm) 1.5 T There were no scan-related events or safety issues
Forleo et al.43 2013 250 MRI conditional pacemaker None Successfully implanted without significant
procedural complications
Gimbel et al.47 2013 263 MRI conditional pacemaker (Advisa MRI) 1.5 T There were no scan-related events or safety issues
Rickard et al.44 2014 466 MRI conditional pacemaker (CapSureFix) None MRI conditional leads had lower ventricular
vs. conventional leads sensing, higher ventricular capture threshold, and
higher acute dislodgment rate
Elmouchi et al.45 2014 157 MRI conditional pacemaker (CapSureFix) None Lead-related complications were higher with
vs. conventional leads CapSureFix leads
Wollman et al.50 2014 36 MRI conditional pacemaker (Advisa MRI) 1.5 T There were no scan-related events or safety issues
Acha et al.46 2015 492 MRI conditional pacemaker (CapSureFix) None Lead-related complication were higher with
vs. conventional leads CapSureFix leads
Klein-Wiele et al.51 2015 24 MRI conditional pacemaker 1.5 T There were no scan-related events or safety issues
Savouré et al.68 2015 29 MRI conditional pacemaker (Kora 100) 1.5 T There were no scan-related events or safety issues
Awad et al.54 2015 153 MRI conditional ICD (ProMRI ICD) 1.5 T There were no scan-related events or safety
issues; one patient demonstrated decreased
R-wave amplitude in 1 month follow-up
Shenthar et al.52 2015 266 MRI conditional pacemaker (Advisa MRI) 1.5 T There were no scan-related events or safety issues
Gold et al.53 2015 275 MRI conditional ICD (Evera MRI) 1.5 T One patient experienced atrial tachycardia during
scanning
Bailey et al.48 2015 272 MRI conditional pacemaker (ProMRI) 1.5 T There were no scan-related events or safety issues
Bailey et al.49 2016 245 MRI conditional pacemaker (ProMRI) 1.5 T Pericardial effusion in one patient that required
lead positioning

ICD, Implantable cardioverter defibrillator; MRI, magnetic resonance imaging.

the MRI conditional group, including perforation, pericarditis, and small changes in system parameters after MRI, which were clinically
lead dislodgment. Acha et al.46 observed similar results in another study irrelevant. In a recent study, adenosine stress perfusion CMR was suc-
where higher lead perforations (5.5%) with MRI conditional leads cessfully performed in 24 patients with MRI conditional CIED systems.51
(0.5%) were reported. Interestingly, MRI conditional leads tended to No significant adverse events were observed.
cause delayed perforations, occurring over 3 weeks after the implanta- To expand MRI conditional status to previously implanted non-
tion. It is likely that the stiffer lead design, shorter length of the lead, conditional MRI CapSureFix Novus 5076 leads, a randomized trial was
and increased diameter were responsible for the observed differences. designed.52 In 266 patients, Advisa SureScan MRI devices and CapSureFix
Another MRI conditional system was tested under 1.5 T MRI in a Novus 5076 leads were implanted, or previously implanted CapSureFix
prospective randomized multicenter trial.47 A total of 263 patients were Novus 5076 leads were connected to newly implanted Advisa SureScan
randomized to MRI versus no MRI at 9 to 12 months after implanta- MRI generators. The patients were randomized to MRI versus control
tion. Notably, the MRI sequences included head and thoracic regions. groups. There were no MRI-related complications or arrhythmias, and
There were no adverse events during MRI, and device interrogation the proportions of the patients with changes in device parameters were
after the MRI showed no significant change in system parameters com- similar in the two groups. This study showed that previously implanted
pared with the control group. Following this trial, the FDA approved conventional CapSureFix Novus 5076 pacemaker leads may safely
the Advisa SureScan MRI (Medtronic, Minneapolis, MN) pacemaker undergo MRI if connected to the Advisa SureScan MRI conditional
system in 2013. generator.
The ProMRI system (Biotronik, Berlin, Germany), another MRI Recently, device manufacturers have also introduced MRI conditional
conditional pacemaker, was tested in two sequential studies. In the first ICD systems. The performance of MRI conditional ICD, Evera MRI
study, 272 patients were enrolled, 226 of whom underwent head and SureScan (Medtronic, Minneapolis, MN) was evaluated in a multicenter
lumbar 1.5 T MRI.48 There were no adverse events immediately after randomized trial.53 A total of 263 patients were randomized to full-body
scan, or during 1-month follow-up; and no significant change was MRI versus control groups. In the MRI group, one patient experienced
observed in device parameters. Later, ProMRI systems were tested under atrial tachycardia during the examination. The scan was stopped and
1.5 T thoracic spine and CMR.49 Of 221 patients studied, one patient the rhythm was converted noninvasively through the CIED; then the
presented with pericardial effusion, in whom lead positioning was scan was continued without any further safety issues. There were minor
required. In others, no adverse events were reported. Based on these changes in system parameters in the MRI group, which did not differ
results, ProMRI systems were approved by the FDA in March 2015. from the control group. Accordingly, FDA approved the Evera MRI
Another MRI conditional CIED, the ImageReady MR-Conditional Pacing SureScan system in September 2015.
System (Boston Scientific) has received FDA approval in April 2016 for Lastly, another MRI conditional ICD, ProMRI ICD system (Biotronik,
full-body 1.5 T MRI. Berlin, Germany), was granted FDA approval in December 2015 after
In a small single-center study, Advisa SureScan MRI was tested under safety and efficacy was demonstrated under 1.5 T thoracic and CMR
1.5 T CMR.50 Clinical adverse events were not observed, but there were in a large prospective study.54
124 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A B
FIG. 11.2  Susceptibility artifact due to a left-sided implantable cardio-
verter defibrillator generator in a patient undergoing 1.5 T cardiovascular
magnetic resonance. (A) Short-axis cine image: lead artifact can be seen
in right ventricle. Generator artifact did not affect the overall diagnostic
quality. (B) Short-axis late gadolinium enhancement sequence. Suscep-
tibility artifact has severely affected the interpretability of the image of
the anterior segment of the left ventricle.

FIG. 11.3  Axial thoracic gradient echo image in a patient with a Medtronic
CARDIAC IMPLANTABLE ELECTRONIC Reveal LINQ long-term monitoring device. Note the anterior image artifact
DEVICE–RELATED ARTIFACTS (arrow).

The potential of image artifacts should be considered when deciding


to perform an MRI examination in CIED recipients, and the benefit- noise recordings that may be misinterpreted as an arrhythmia.35,59 In a
risk ratio should be determined based on the need of the MRI, the case report, two patients with implanted loop recorders were safely
location of the suspected disease, as well as the location of the implanted examined with MRI.60 Importantly, several implantable recorders are
CIED generator. Susceptibility artifacts are more common with inver- considered to be MRI conditional by the FDA. Given the small size of
sion recovery and balanced steady-state free precession (bSSFP) sequences. these devices, the local artifact is also smaller (Fig. 11.3).
In our experience, selecting imaging planes that are perpendicular to
the device generator, shortening echo times, or using spin fast echo
sequences may reduce the artifacts. Specialized artifact reduction
FUTURE DIRECTIONS
sequences have also been developed, which can greatly improve the MRI conditional CIED systems have been a welcome addition in recent
image quality in the setting of CIED systems.55 years. However, several questions remain to be answered. Should we
Kaasalainen et al.56 performed 1.5 T CMR in 16 patients. Overall, implant MRI conditional devices in all patients? If not, how should we
CMR was adequate for diagnostic purposes. In all patients with a right- select the suitable candidate? Because it is likely that legacy leads will
sided generator, there was no compromise in any of the sequences; pose minimal hazards, should MRI conditional generators be implanted
however, in left-sided generators, the artifacts were visible in several of when a generator replacement is needed? Long-term follow-up, addi-
the sequences, and those were typically observed in apical, anterior, tional analyses of safety, cost-effectiveness, and accumulating experience
and anteroseptal left ventricular (LV) segments, especially during myo- will answer these questions.
cardial late gadolinium enhancement (LGE) imaging. Device-related image artifacts seem to affect the decision to order
Similarly, we were able to diagnose patients in 100% of the nontho- an MRI in CIED recipients. Based on our current knowledge, type and
racic MRIs, and in 93% of the thoracic MRIs.40 We also showed that the location of the device, as well as MRI sequence characteristics, may
type of CIED (ICDs are more likely to cause artifacts than pacemakers), affect the presence and quantity of CIED-related artifacts. Ultimately,
side of the device generator (left higher than right), indirect param- newer designs addressing the large local artifact produced by the ICD
eters such as body mass index (BMI) and LV end-diastolic volumes transformer are necessary to mitigate artifacts.
that indicate cardiac distance from the device, and the CMR sequence,
all affect the presence and amount of the artifact.57 We observed that
artifacts were mainly present in LGE sequence, and in short-axis view,
CONCLUSION
with the anterior and apical LV regions most commonly affected (Fig. The public health importance of the ability to perform MRI in CIED
11.2). Lead artifacts did not affect the overall interpretability. recipients has led to laudable efforts by device manufacturers to develop
Junttila et al.15 also reported ICD-related artifacts, especially in the MRI conditional systems, and by investigators to develop safety protocols
anterior and septal LV segments, and most commonly in the apical for improved access in patients with legacy systems. Current guidelines
region in the LGE sequences that limited the diagnostic capabilities of suggest that MRI may be performed in legacy CIED recipients using a
the CMR. Naehle et al.20 performed CMR in 32 patients with permanent strict protocol if there is no alternative imaging and the benefits of the
pacemakers and ICDs. With right-sided implantations, the image quality MRI outweigh the risk of the examination.
and diagnostic value were better compared with left-sided CIED (100%
vs. 35%). With left-sided devices, the image quality is impaired, specifi-
cally in anterior and anterolateral LV segments. In the subanalysis of
CONFLICT OF INTEREST
the Advisa MRI pacemaker trial that was performed with 1.5 T CMR, Esra Gucuk Ipek has no conflict of interest. Saman Nazarian is a sci-
both right and LV acquisitions had enough diagnostic quality to facilitate entific advisor to CardioSolv, St. Jude Medical, Spectranetics, and Biosense
quantitative cardiac assessment.58 Webster Inc. and principal investigator for research funding to Johns
Hopkins University from Biosense-Webster Inc.
Implantable Long-Term Loop Recorders
Although limited data are available on implantable loop recorders com-
pared with pacemakers and ICDs, current data suggest that those patients
REFERENCES
can safely undergo MRI, albeit with a potential for electromagnetic A full reference list is available online at ExpertConsult.com
CHAPTER 11  Pacemaker and Implantable Cardioverter-Defibrillator Safety and Safe Scanning 124.e1

REFERENCES 19. Naehle CP, Zeijlemaker V, Thomas D, et al. Evaluation of cumulative


effects of MR imaging on pacemaker systems at 1.5 tesla. Pacing Clin
1. Borne RT, Peterson PN, Greenlee R, et al. Temporal trends in patient Electrophysiol. 2009;32(12):1526–1535.
characteristics and outcomes among Medicare beneficiaries undergoing 20. Naehle CP, Kreuz J, Strach K, et al. Safety, feasibility, and diagnostic value
primary prevention implantable cardioverter-defibrillator placement in of cardiac magnetic resonance imaging in patients with cardiac
the United States, 2006-2010. Results from the National Cardiovascular pacemakers and implantable cardioverters/defibrillators at 1.5 T. Am
Data Registry’s Implantable Cardioverter-Defibrillator Registry. Heart J. 2011;161(6):1096–1105.
Circulation. 2014;130(10):845–853. 21. Sheldon SH, Bunch TJ, Cogert GA, et al. Multicenter study of the safety
2. Greenspon AJ, Patel JD, Lau E, et al. Trends in permanent pacemaker and effects of magnetic resonance imaging in patients with coronary
implantation in the United States from 1993 to 2009: increasing complexity sinus left ventricular pacing leads. Heart Rhythm. 2015;12(2):345–349.
of patients and procedures. J Am Coll Cardiol. 2012;60(16):1540–1545. 22. Pulver AF, Puchalski MD, Bradley DJ, et al. Safety and imaging quality of
3. Mond HG, Proclemer A. The 11th world survey of cardiac pacing and MRI in pediatric and adult congenital heart disease patients with
implantable cardioverter-defibrillators: calendar year 2009—a World pacemakers. Pacing Clin Electrophysiol. 2009;32:450–456.
Society of Arrhythmia’s project. Pacing Clin Electrophysiol. 2011;34(8): 23. Cohen JD, Costa HS, Russo RJ. Determining the risks of magnetic
1013–1027. resonance imaging at 1.5 tesla for patients with pacemakers and
4. Kalin R, Stanton MS. Current clinical issues for MRI scanning of implantable cardioverter defibrillators. Am J Cardiol. 2012;110:1631–1636.
pacemaker and defibrillator patients. Pacing Clin Electrophysiol. 24. Muehling OM, Wakili R, Greif M, et al. Immediate and 12 months follow
2005;28:326–328. up of function and lead integrity after cranial MRI in 356 patients with
5. Taruya A, Tanaka A, Nishiguchi T, et al. Necessity of magnetic resonance conventional cardiac pacemakers. J Cardiovasc Magn Reson. 2014;16:39.
imaging examinations after permanent pacemaker implantation. Int J 25. Russo RJ, Costa HS, Silva PD, et al. Assessing the risks associated with
Cardiol. 2015;184:497–498. MRI in patients with a pacemaker or defibrillator. N Engl J Med.
6. Nazarian S, Hansford R, Roguin A, et al. A prospective evaluation of a 2017;376:755–764.
protocol for magnetic resonance imaging of patients with implanted 25a.  Nazarian S, Hansford R, Rahsepar AA, et al. Safety of magnetic
cardiac devices. Ann Intern Med. 2011;155:415–424. resonance imaging in patients with cardiac devices. N Engl J Med.
7. Martin ET, Coman JA, Shellock FG, Pulling CC, Fair R, Jenkins K. 2017;377:2555–2564.
Magnetic resonance imaging and cardiac pacemaker safety at 1.5-tesla. 26. Gimbel JR, Bailey SM, Tchou PJ, Ruggieri PM, Wilkoff BL. Strategies for
J Am Coll Cardiol. 2004;43:1315–1324. the safe magnetic resonance imaging of pacemaker-dependent patients.
8. Levine GN, Gomes AS, Arai AE, et al. Safety of magnetic resonance Pacing Clin Electrophysiol. 2005;28:1041–1046.
imaging in patients with cardiovascular devices: an American Heart 27. Higgins JV, Sheldon SH, Watson RE Jr, et al. “Power-on resets” in cardiac
Association scientific statement from the Committee on Diagnostic and implantable electronic devices during magnetic resonance imaging. Heart
Interventional Cardiac Catheterization, Council on Clinical Cardiology, Rhythm. 2015;12(3):540–544.
and the Council on Cardiovascular Radiology and Intervention: endorsed 28. Langman DA, Goldberg IB, Finn JP, Ennis DB. Pacemaker lead tip
by the American College of Cardiology Foundation, the North American heating in abandoned and pacemaker-attached leads at 1.5 tesla MRI.
Society for Cardiac Imaging, and the Society for Cardiovascular Magnetic J Magn Reson Imaging. 2011;33:426–431.
Resonance. Circulation. 2007;116:2878–2891. 29. Luechinger R, Zeijlemaker VA, Pedersen EM, et al. In vivo heating of
9. Fetter J, Aram G, Holmes DR Jr, Gray JE, Hayes DL. The effects of pacemaker leads during magnetic resonance imaging. Eur Heart J.
nuclear magnetic resonance imagers on external and implantable pulse 2005;26:376–383.
generators. Pacing Clin Electrophysiol. 1984;7(4):720–727. 30. Higgins JV, Gard JJ, Sheldon SH, et al. Safety and outcomes of magnetic
10. Gimbel JR, Johnson D, Levine PA, Wilkoff BL. Safe performance of resonance imaging in patients with abandoned pacemaker and
magnetic resonance imaging on five patients with permanent cardiac defibrillator leads. Pacing Clin Electrophysiol. 2014;37(10):1284–1290.
pacemakers. Pacing Clin Electrophysiol. 1996;19(6):913–919. 31. Higgins JV, Watson RE Jr, Jaffe AS, et al. Cardiac troponin T in patients
11. Sommer T, Vahlhaus C, Lauck G, et al. MRI and cardiac pacemakers: with cardiac implantable electronic devices undergoing magnetic
in-vitro evaluation and in vivo studies in 51 patients at 0.5 T. Radiology. resonance imaging. J Interv Card Electrophysiol. 2016;45(1):91–97.
2000;215:869–879. 32. Strach K, Naehle CP, Mühlsteffen A, et al. Low-field magnetic resonance
12. Vahlhaus C, Sommer T, Lewalter T, et al. Interference with cardiac imaging: increased safety for pacemaker patients? Europace. 2010;12(7):
pacemakers by magnetic resonance imaging: are there irreversible 952–960.
changes at 0.5 T? Pacing Clin Electrophysiol. 2001;24:489–495. 33. Del Ojo JL, Moya F, Villalba J, et al. Is magnetic resonance imaging safe
13. Roguin A, Zviman MM, Meininger GR, et al. Modern pacemaker and in cardiac pacemaker recipients? Pacing Clin Electrophysiol.
implantable cardioverter/defibrillator systems can be magnetic resonance 2005;28:274–278.
imaging safe: in vitro and in vivo assessment of safety and function at 34. Naehle CP, Meyer C, Thomas D, et al. Safety of brain 3-T MR imaging
1.5 T. Circulation. 2004;110:475–482. with transmit-receive head coil in patients with cardiac pacemakers: pilot
14. Schmitt C, Brachmann J, Waldecker B, et al. Implantable cardioverter prospective study with 51 examinations. Radiology. 2008;249(3):991–1001.
defibrillator: possible hazards of electromagnetic interference. Pacing Clin 35. Gimbel JR. Magnetic resonance imaging of implantable cardiac rhythm
Electrophysiol. 1991;14(6):982–984. devices at 3.0 tesla. Pacing Clin Electrophysiol. 2008;31(7):795–801.
15. Junttila MJ, Fishman JE, Lopera GA, et al. Safety of serial MRI in patients 36. Gimbel JR. Unexpected asystole during 3T magnetic resonance imaging
with implantable cardioverter defibrillators. Heart. 2011;97:1852–1856. of a pacemaker-dependent patient with a ‘modern’ pacemaker. Europace.
16. Burke PT, Ghanbari H, Alexander PB, Shaw MK, Daccarett M, Machado 2009;11:1241–1242.
C. A protocol for patients with cardiovascular implantable devices 37. Gimbel JR. Unexpected pacing inhibition upon exposure to the 3T static
undergoing magnetic resonance imaging (MRI): should defibrillation magnetic field prior to imaging acquisition: what is the mechanism?
threshold testing be performed post-MRI. J Intev Card Electrophysiol. Heart Rhythm. 2011;8:944–945.
2010;28:59–66. 38. Brignole M, Auricchio A, Baron-Esquivias G, et al. 2013 ESC guidelines
17. Naehle CP, Strach K, Thomas D, et al. Magnetic resonance imaging at on cardiac pacing and cardiac resynchronization therapy: the task force
1.5-T in patients with implantable cardioverter-defibrillators. J Am Coll on cardiac pacing and resynchronization therapy of the European Society
Cardiol. 2009;54(6):549–555. of Cardiology (ESC). Developed in collaboration with the European
18. Sommer T, Naehle CP, Yang A, et al. Strategy for safe performance of Heart Rhythm Association (EHRA). Eur Heart J. 2013;34(29):2281–2329.
extrathoracic magnetic resonance imaging at 1.5 tesla in the presence of 39. Sommer T, Luechinger R, Barkhausen J, et al. German Roentgen Society
cardiac pacemakers in non-pacemaker-dependent patients: a prospective Statement on MR Imaging of Patients with Cardiac Pacemakers. Rofo.
study with 115 examinations. Circulation. 2006;114:1285–1292. 2015;187(9):777–787.
124.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

40. Nazarian S, Roguin A, Zviman MM, et al. Clinical utility and safety of a thoracic spine and cardiac 1.5-T magnetic resonance imaging scanning
protocol for noncardiac and cardiac magnetic resonance imaging of conditions. Heart Rhythm. 2015;12(10):2155–2161.
patients with permanent pacemakers and implantable cardioverter 55. Rashid S, Rapacchi S, Vaseghi M, et al. Improved late gadolinium
defibrillators at 1.5 tesla. Circulation. 2006;114:1277–1284. enhancement MR imaging for patients with implanted cardiac devices.
41. Wilkoff BL, Bello D, Taborsky M, et al. EnRhythm MRI SureScan Pacing Radiology. 2014;270(1):269–274.
System Study Investigators. Magnetic resonance imaging in patients with 56. Kaasalainen T, Kivistö S, Holmström M, et al. Cardiac MRI in patients
a pacemaker system designed for the magnetic resonance environment. with cardiac pacemakers: practical methods for reducing susceptibility
Heart Rhythm. 2011;8(1):65–73. artifacts and optimizing image quality. Acta Radiol. 2016;57(2):178–187.
42. Forleo GB, Santini L, Della Rocca DG, et al. Safety and efficacy of a new 57. Sasaki T, Hansford R, Zviman MM, et al. Quantitative assessment of
magnetic resonance imaging-compatible pacing system: early results of a artifacts on cardiac magnetic resonance imaging of patients with
prospective comparison with conventional dual-chamber implant pacemakers and implantable cardioverter-defibrillators. Circ Cardiovasc
outcomes. Heart Rhythm. 2010;7:750–754. Imaging. 2011;4(6):662–670.
43. Forleo GB, Della Rocca DG, Panattoni G, et al. Clinical and electrical 58. Schwitter J, Kanal E, Schmitt M, Advisa MRI System Study Investigators,
performance of currently available MRI-safe pacing systems. Do all et al. Impact of the Advisa MRI pacing system on the diagnostic quality
devices perform in the same way? Int J Cardiol. 2013;167(5):2340–2341. of cardiac MR images and contraction patterns of cardiac muscle during
44. Rickard J, Taborsky M, Bello D, et al. Short- and long-term electrical scans: Advisa MRI randomized clinical multicenter study results. Heart
performance of the 5086MRI pacing lead. Heart Rhythm. 2014;11(2): Rhythm. 2013;10(6):864–872.
222–229. 59. Gimbel JR, Zarghami J, Machado C, Wilkoff BL. Safe scanning, but
45. Elmouchi DA, Rosema S, Vanoosterhout SM, et al. Cardiac perforation frequent artifacts mimicking bradycardia and tachycardia during
and lead dislodgement after implantation of a MR-conditional pacing magnetic resonance imaging (MRI) in patients with an implantable loop
lead: a single-center experience. Pacing Clin Electrophysiol. recorder (ILR). Ann Noninvasive Electrocardiol. 2005;10(4):404–408.
2014;37(1):4–10. 60. Blaschke F, Lacour P, Walter T, et al. Cardiovascular magnetic resonance
46. Acha MR, Keaney JJ, Lubitz SA, et al. Increased perforation risk with an imaging in patients with an implantable loop recorder. Ann Noninvasive
MRI-conditional pacing lead: a single-center study. Pacing Clin Electrocardiol. 2016;21(3):319–324.
Electrophysiol. 2015;38(3):334–342. 61. Tandri H, Zviman MM, Wedan SR, Lloyd T, Berger RD, Halperin H.
47. Gimbel JR, Bello D, Schmitt M, et al. Advisa MRI System Study Determinants of gradient field-induced current in a pacemaker lead
Investigators. Randomized trial of pacemaker and lead system for safe system in a magnetic resonance imaging environment. Heart Rhythm.
scanning at 1.5 tesla. Heart Rhythm. 2013;10:685–691. 2008;5(3):462–468.
48. Bailey WM, Rosenthal L, Fananapazir L, et al. ProMRI/ProMRI AFFIRM 62. Luechinger R, Duru F, Zeijlemaker VA, Scheidegger MB, Boesiger P,
Study Investigators. Clinical safety of the ProMRI pacemaker system in Candinas R. Pacemaker reed switch behavior in 0.5, 1.5, and 3.0 tesla
patients subjected to head and lower lumbar 1.5-T magnetic resonance magnetic resonance imaging units: are reed switches always closed in
imaging scanning conditions. Heart Rhythm. 2015;12(6):1183–1191. strong magnetic fields? Pacing Clin Electrophysiol. 2002;25:1419–1423.
49. Bailey WM, Mazur A, McCotter C, et al; ProMRI Study Investigators. 63. Gimbel JR, Kanal E, Schwartz KM, Wilkoff BL. Outcome of magnetic
Clinical safety of the ProMRI pacemaker system in patients subjected to resonance imaging (MRI) in selected patients with implantable
thoracic spine and cardiac 1.5-T magnetic resonance imaging scanning cardioverter defibrillators (ICDs). Pacing Clin Electrophysiol. 2005;
conditions. Heart Rhythm. 2016;13(2):464–471. 28(4):270–273.
50. Wollmann CG, Thudt K, Kaiser B, Salomonowitz E, Mayr H, Globits S. 64. Mollerus M, Albin G, Lipinski M, Lucca J. Cardiac biomarkers in patients
Safe performance of magnetic resonance of the heart in patients with with permanent pacemakers and implantable cardioverter-defibrillators
magnetic resonance conditional pacemaker systems: the safety issue of undergoing an MRI scan. Pacing Clin Electrophysiol. 2008;31(10):1241–
the ESTIMATE study. J Cardiovasc Magn Reson. 2014;16:30. 1245.
51. Klein-Wiele O, Garmer M, Urbien R, et al. Feasibility and safety of 65. Mollerus M, Albin G, Lipinski M, Lucca J. Ectopy in patients with
adenosine cardiovascular magnetic resonance in patients with MR permanent pacemakers and implantable cardioverter-defibrillators
conditional pacemaker systems at 1.5 tesla. J Cardiovasc Magn Reson. undergoing an MRI scan. Pacing Clin Electrophysiol. 2009;32(6):772–778.
2015;17:112. 66. Halshtok O, Goitein O, Abu Sham’a R, Granit H, Glikson M, Konen E.
52. Shenthar J, Milasinovic G, Al Fagih A, et al. MRI scanning in patients Pacemakers and magnetic resonance imaging: no longer an absolute
with new and existing CapSureFix Novus 5076 pacemaker leads: contraindication when scanned correctly. Isr Med Assoc J. 2010;12(7):
randomized trial results. Heart Rhythm. 2015;12(4):759–765. 391–395.
53. Gold MR, Sommer T, Schwitter J, et al; Evera MRI Study Investigators. 67. Mollerus M, Albin G, Lipinski M, Lucca J. Magnetic resonance imaging
Full-body MRI in patients with an implantable cardioverter-defibrillator: of pacemakers and implantable cardioverter-defibrillators without
primary results of a randomized study. J Am Coll Cardiol. 2015;65(24): specific absorption rate restrictions. Europace. 2010;12(7):947–951.
2581–2588. 68. Savouré A, Mechulan A, Burban M, Olivier A, Lazarus A. The kora
54. Awad K, Griffin J, Crawford TC, et al. Clinical safety of the Iforia pacemaker is safe and effective for magnetic resonance imaging. Clin Med
implantable cardioverter-defibrillator system in patients subjected to Insights Cardiol. 2015;9:85–90.
12 
Special Considerations: Cardiovascular
Magnetic Resonance in Infants and Children
Mark A. Fogel

Clinical cardiovascular magnetic resonance (CMR) had been in use for versa. Because many pulse sequences are made to image adult
nearly 35 years and has become firmly established in the evaluation of patients, the tradeoffs that can be performed in children can
congenital heart disease (CHD) in many ways, including anatomy, physi- only be taken advantage of to a limited degree. In addition, chil-
ology, ventricular function, blood flow, and tissue characterization.1–29 dren under 10 years of age usually need some form of sedation,
In many instances, it is used as an adjunct to other imaging modali- which make sequences designed for breath-holding useless unless
ties such as noninvasive echocardiography and invasive angiography; the child is paralyzed, intubated, and mechanically ventilated
however, in a number of areas such as vascular rings,25–29 blood flow, under anesthesia. Because the breath-holding technique is the
as well as ventricular volumes and mass,30,31 it has become the clini- mainstay of adult CMR, “work-arounds” have been developed to
cal “gold standard.” In addition, CMR offers several advantages over successfully image the pediatric patient. These modifications, or
other imaging modalities, including lack of ionizing radiation, which new approaches to CMR, must be understood for children to be
is extremely important for infants and children who are more suscep- imaged successfully.
tible to DNA breakage leading to neoplastic disease and have more 2. Anatomy of CHD: the anatomy of native CHD is very different
time to manifest cancer symptomatology than adults. Furthermore, from adult cardiology and demands a rigorous and systematic
with capabilities such as exquisite soft tissue contrast, a capacity for approach to make the correct diagnosis. In addition, because
true three-dimensional (3D) imaging in many areas (e.g., anatomy, many CHDs rely on surgical as well as an increasing number of
ventricular function, blood flow), accurate flow quantification, tissue catheter-based therapies, the anatomic and physiologic informa-
characterization such as myocardial viability, T1 mapping for diffuse tion extracted from the CMR examination needs to keep these
fibrosis and perfusion imaging, noninvasive labeling of the myocardium approaches in mind. As a corollary, the postoperative/interventional
or blood (tagging), coronary imaging and freely selectable imaging anatomy and hemodynamics in CHD must also be familiar to
planes without limitations to “windows” or overlapping structures make the physician performing the examination to assess for adequacy
CMR the “one-stop-shop” for noninvasive imaging in children. There of the therapy and follow-up. For example, the connections
are also unique characteristics to CMR, such as building images over between the major cardiac segments (atria, ventricles, and great
many heartbeats so that long-term function and flow are “built into the vessels) along with venous anatomy can and are altered in many
image” (instead of the imager having to do this mentally by visualizing types of the diseases encountered in CHD (e.g., transposition
hundreds of heartbeats), four-dimensional (4D) flow imaging or internal of the great arteries [TGA]). Communications are present where
checks to flow (e.g., the flows in the branch pulmonary arteries must they should not be (e.g., ventricular septal defects [VSD]), blood
sum to the flow measured in the main pulmonary artery) that make vessels from fetal life may still be present (e.g., patent ductus
hemodynamics especially accurate. These advantages, and continued arteriosus), and some cardiac structures might not even be present
advances in CMR hardware, software, and imaging techniques, are bring- or present in a markedly hypoplastic form (e.g., hypoplastic left
ing CMR into even more widespread use in pediatric cardiology. With heart syndrome). In the evaluation of CHD after surgery, conduits
the advent of relatively new techniques—such as “real-time” cine32 and and baffles constructed to separate the circulations have little
flow imaging,33 dark blood 3D sampling perfection with application- parallel in the adult world.8 As such, anatomy and morphology
optimized contrasts using different flip angle evolution (SPACE)34 must be delineated by CMR in ways that are not required in
and fully navigator accepted coronary imaging—CMR is poised to adult heart disease and the physician interpreting the study needs
become a first-line imaging modality in many situations in pediatric to be schooled in the nuances of both the preoperative and post-
cardiology and CHD. operative anatomy of CHD.
Although the application of CMR to CHD uses nearly all of the 3. Physiology: the unique physiology of CHD is most often a result
techniques discussed in the various chapters of this book, it nevertheless of the altered anatomy. Evaluation of shunts (e.g., VSD) plays a
stands on its own as a separate discipline in the world of CMR. The critical role in assessment of any disease entity in pediatrics and
multiple reasons for this can be divided into a few broad categories. yet has a minor role in the adult world. CMR has recently become
1. Technical challenges: imaging infants and children is very demand- the only imaging modality to quantify aortic to pulmonary col-
ing because they require both increased spatial resolution as a lateral flow development in single ventricles, again, a unique
result of their small size as well as increased temporal resolution province of CHD.35–37 Determination of ventricular function in
because of higher heart rates as compared with adults. CMR ventricles with strange and unusual shapes (single ventricles,
in adults makes many tradeoffs to make imaging quicker and L-looped ventricles) is routine in the practice of CMR in children
simpler without sacrificing diagnostic accuracy, such as obtain- and demands unique solutions. Assessment of the postoperative/
ing spatial resolution at the cost of temporal resolution and vice interventional physiology where the surgeon, for example, creates

125
126 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

a systemic to pulmonary artery shunt or creates an anastomosis exist that can be formulated into a generalized protocol of CMR in
between the cava and the pulmonary artery must be dealt with. CHD (Fig. 12.1). The author does not claim this to be the only way
CMR in CHD and in pediatrics must be adapted to fit these to think about the process, but it is, in the author’s opinion, the most
needs and the physician interpreting the study needs to under- efficient and complete. It should be noted that the protocol needs to
stand the complex physiology. be individualized to the patient and the disease process.
Similar to other imaging modalities, CMR has limitations and chal- In this section, each step of the protocol will demonstrate a different
lenges in the CHD population. Sedation, and in some instances, general form of CHD, giving the reader a broad overview of the spectrum of
anesthesia for infants and children to hold still for a 45- to 60-minute lesions encountered.
scan is always a consideration. Even if remaining motionless in the
scanner is not an issue, cooperation by the preteen or teen may be Anatomic Imaging (Noncontrast)
problematic (e.g., breath-holding). In addition, intravascular coils, wires, After localizers, anatomic data are acquired not only to survey the car-
stents, and clips may all cause artifacts if they are near the structure of diovascular anatomy and make the “anatomic diagnosis” but also to
interest and are found in a number of cases of CHD where catheter act as localizers themselves for future physiologic and functional imaging.
and surgical intervention has occurred. The lack of portability of CMR This is important in that physiologic and functional data must be inter-
is disadvantageous, especially for the critically ill infant or child who preted in light of the prevailing anatomy. A full contiguous set of axial
would need to be moved to the CMR suite. And finally, patients with images from the diaphragm to the thoracic inlet is obtained (typically
arrhythmias may not allow proper data acquisition; other patients may 40−50 images, Figs. 12.2 and 12.3); the author prefers “static” balanced
have bizarre T-waves or bundle branch blocks, which may not allow steady-state free precession (bSSFP) images. This must be extended
for proper triggering. This is becoming less of an issue now that “single outside the thorax for special cases such as to the neck if arch anatomy
shot” CMR, “real-time” CMR, and sequences with “arrhythmia rejec- is being evaluated (e.g., innominate artery branches in the neck) or if
tion” are realities. Pacemakers are still a problem and remain a “relative total anomalous pulmonary venous connections are suspected (e.g.,
contraindication”; though safety of MRI in adult patients with non-MRI connection below the diaphragm). Imaging that uses bSSFP depends
conditional/legacy pacemakers and implanted cardioverter-defibrillators on contrast between blood and the surrounding tissue, which is enhanced
has been demonstrated,37a safety data specific to infants and children by using a high flip angle, and therefore the practice is to allow the
are currently lacking. Patients with them usually do not undergo CMR scanner to select the highest flip angle possible. These axial images form
unless it is the only imaging modality that can obtain the data and the basis of examining the anatomy.
change management (see Chapter 11).38 One disadvantage to using “static” bSSFP is that turbulence may
This chapter discusses the present state-of-the-art of CMR in CHD present on the image as an absence of a structure (signal loss) as in
and some of the newer techniques available. For simplicity, the imaging the case of the pulmonary arteries in a single ventricle patient after a
techniques discussed will be generic; however, some of the “lingo” of Blalock-Taussing shunt; typically, these are very difficult to visualize
CMR will be related to the Siemen’s CMR scanners. The reader should by this technique because the turbulence creates a major signal loss.
understand that other manufacturers, for the most part, have similar This drawback can be compensated for by performing half-Fourier
sequences but under a different name. acquisition single-shot turbo spin echo (HASTE) imaging, which yields
a set of dark blood images rapidly (or another type of dark blood
GENERALIZED PROTOCOL OF CMR FOR imaging such as 3D SPACE, although HASTE is the quickest) and is
performed while multiplanar reconstruction (MPR) is being done (see
CONGENITAL HEART DISEASE later). Alternatively, a set of axial cines can be performed to visualize
CMR for CHD is a technique that constantly requires fine tuning during these structures in the hope that a phase of the cardiac cycle will dem-
an examination, in general, because unexpected findings turn up fre- onstrate the “missing” structure.
quently, especially in pediatrics. Nevertheless, certain basic principles MPR, also called multiplanar reformatting, is a technique where a
set of contiguous images (in this case, axial) are stacked atop each other
and any plane can be reconstructed. This can be used after the axial
stack above is obtained. The exact slice orientation and position can
• Static SSFP-axial
therefore be obtained for any future imaging during the study. In addi-
• HASTE tion, the anatomy can be inspected from multiple views from just the
• Cine set of axial images as a first pass. It can be used for any type of imaging.
• Dark blood Finally, MPR can be used to demonstrate the salient point of the anatomy
• Gadolinium in instances where the scan needed to be terminated prematurely because
of patient instability or technical issues after the stack of contiguous
• T1 mapping
axial images had been obtained. “Curved cuts” can be used to delineate
• Velocity mapping
the important parts of the anatomy as well (see Fig. 12.3).
• Viability If needed, dark blood imaging can complement the bright blood
Specialized techniques { •• Coronary
Tagging
imaging imaging as in instances of vascular rings to visualize the trachea, coarc-
tation of the aorta (Fig. 12.4) or a systemic to pulmonary artery shunt.
• Perfusion This type of imaging is used judiciously because it requires relatively
{ Specialized techniques long scanning times. A set of high-resolution double-inversion recovery
• 4D flow
dark blood images can be obtained or, if available, 3D SPACE, which
FIG. 12.1  Generalized protocol for cardiovascular magnetic resonance can obtain a high-resolution slab in 5 minutes or less.
imaging of congenital heart disease. This is a generalized protocol and
should be individualized to the patient and the specific disease. HASTE, Function/Anatomy: Cine CMR (Fig. 12.5)
Half-Fourier acquisition single-shot turbo spin echo; MPR, multiplanar Either bSSFP or spoiled gradient recalled echo (GRE) sequences are
reconstruction; SSFP, steady-state free precession. used and are tailored to the lesion under study. Cine, along with phase
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 127

RV LV Ao

HB

DAo
1 Az 2 3

SVC Ao

Az
RPA

LPA
4 5 6

FIG. 12.2  Stack of contiguous static balanced steady-state free precession images. This is a set of selected
axial images of a patient with heterotaxy syndrome, tricuspid stenosis with pulmonary atresia (S,D,X) with
a hypoplastic right ventricle (RV) and interrupted inferior vena cava with azygous continuation to the right
superior vena cava (SVC) who is imaged after a Kawashima operation (SVC to pulmonary artery connection
in the presence of an interrupted inferior vena cava with azygous continuation) and a hepatic baffle to the
pulmonary arteries. Images progress from inferior to superior as the panels go from left to right and from
top to bottom (numbers 1–6). Ao, Aorta; Az, azygous vein; DAo, descending aorta; HB, hepatic baffle; LPA,
left pulmonary artery; LV, left ventricle; RPA, right pulmonary artery.

1 2 3 4

5 6

SVC

LA

FIG. 12.3  Curved cuts using multiplanar reconstruction. This patient has a partial anomalous right pulmonary
venous connection to the superior vena cava (SVC) but also a persistent levo-atrio-cardinal vein connecting
the left atrium (LA) to the innominate vein. The top and middle 6 panels are static bright blood images fol-
lowing the levo-atrio-cardinal vein (red arrows) and progress from inferior to superior, going from 1 to 6. The
partial anomalous pulmonary venous connection is outlined by the yellow arrows. When the set of static
axial bright blood images are stacked one atop the other, a curved plane can be drawn (lower left panel)
tracing the course of this levo-atrio-cardinal vein in one image (lower middle panel, red arrows). The panel
in the lower right demonstrates a three-dimensional reconstruction from the bright blood images showing
the levo-atrio-cardinal vein (red arrows) and the anomalous pulmonary venous connections (yellow arrows).
128 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

contrast CMR (PCMR), are the major sequences used to delineate Cine imaging is also used to delineate the anatomy as stated in
physiology and function (ventricular and valvar) in CMR of CHD. For the previous section (Fig. 12.6). If coarctation of the aorta is present,
example, to determine left ventricular (LV) volume overload in a patient turbulence should be noted on cine imaging unless there are multiple
with a VSD, a stack of LV short-axis images are acquired to measure collaterals, the coarctation is not severe, or there is poor LV function.
LV volumes, mass, ejection fraction, stroke volume, and cardiac index; Caution must be used, however, when using this signal void to assess
in the absence of valve regurgitation, the difference between LV and the degree of stenosis (or regurgitation if imaging valves) because it
right ventricular (RV) stroke volumes is the added volume overload. must be viewed in light of CMR physics similar to echocardiography
If the LV volume overload was because of aortic regurgitation, a cine (e.g., frame rate, angle of incidence). The size of the signal void is a
of the LV outflow tract in multiple views will be added to the short-axis function of a number of factors such as the echo time (TE, where
stack to visualize the regurgitant jet. longer TEs increase the signal void size and shorter TEs decrease it).
In addition, the size of the signal void is also a function of the direc-
tion of the stenotic jet relative to the orientation of the image voxel.
Finally, bSSFP imaging may underestimate the signal void seen on GRE
sequences.
Cine is also used to visualize the anatomy of valves, typically en face,
and is obtained many times during PCMR (Fig. 12.7; also see Fig. 12.9).
For example, to visualize a bicuspid aortic valve, the most common
CHD, an en face view of the valve is performed by lining up the imaging
plane perpendicular to the LV outflow tract in two orthogonal views
at the level of the aortic valve. This typically results in the equivalent
of a “parasternal short-axis view” by echocardiography. Either bSSFP
or GRE with a high flip angle (e.g., 25 degrees) can be used, although
the author prefers GRE because the high signal of the blood flowing
into the imaging plane outlines the valve leaflets and gives exception-
ally fine detail of the valve (as obtained in the magnitude images of
PCMR). Examples of a trileaflet, bicuspid, and unicuspid aortic valve
are in Figs. 12.7 and 12.8.

Anatomy: Gadolinium-Based CMR (Figs. 12.9–12.11)


The two types of 3D gadolinium techniques for anatomy (both static
FIG. 12.4  Dark blood imaging. Example of a 3-year-old with a mid- [see Figs. 12.10 and 12.11] and time-resolved39 or “dynamic”39 [see
thoracic long segment aortic coarctation of the aorta (arrows) in the Figs. 12.9 and 12.10] versions) are then performed as a complementary
candy cane view (left). On the right is the coarctation in the orthogonal imaging tool because they not only add a special type of 3D dataset,
view to the candy cane view. which can be rotated and manipulated in many different ways, but also

LV

RV RV
LV

FIG. 12.5  Cine cardiovascular magnetic resonance for ventricular function. A 2-year-old child with corrected
transposition of the great arteries (TGA [S,L,L] or L-TGA) demonstrating the left-sided right ventricle (RV) and
the right-sided left ventricle (LV) in the four-chamber view in diastole (upper left) and systole (lower left).
The right panel demonstrates 12 contiguous short-axis images from base to apex in diastole based on the
four-chamber view on the left.
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 129

AAo
MPA
Ao
RPA
RPA

LPA

RPA
RPA

LPA

FIG. 12.6  Cine cardiovascular magnetic resonance for anatomy. A 4-year-old child with transposition of the
great arteries (TGA [S,D,D]) after an arterial switch operation and a Lecompte maneuver where the branch
pulmonary arteries are draped over to the ascending aorta (AAo). The upper left and lower left images dem-
onstrate the bifurcation view of the right (RPA) and left pulmonary arteries (LPA) with steady-state free
precession (SSFP) and gradient echo imaging, respectively. The upper middle and lower right panels dem-
onstrate the RPA in long axis with SSFP and gradient echo imaging, respectively, as well. Upper right shows
the parallel nature of the main pulmonary artery (MPA) and AAo, which is common in this lesion. Ao, Aorta.

Flow m/s

MPA 450
MPA MPA = 6.2 L/min/m2
400
Ao = 4.4 L/min/m2
350
Qp/Qs = 1.4
300

250

200

150

100
Ao
50
Ao
0 50 100 150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 900
0
Trigger time ms

FIG. 12.7  Phase contrast velocity mapping. Data from a 10-year-old child with a conventricular ventricular
septal defect. The upper left panel shows the phase (top) and magnitude images (bottom) of the main pul-
monary artery (MPA) velocity map, while the lower panel shows the phase (top) and magnitude images
(bottom) of the aortic (Ao) velocity map. Note the trileaflet aortic valve. The right panel shows the flow curves
of both MPA and Ao superimposed; Qp/Qs was 1.4.
130 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

FIG. 12.8  Phase contrast magnetic resonance (PCMR) mapping for morphology and velocity. The upper left
panel shows the phase (top) and magnitude images (bottom) of a 3-year-old child with a unicuspid valve
(arrow) whereas the lower left panel shows the phase (top) and magnitude images (bottom) of a 5-year-old
child with a bicuspid valve in transposition of the great arteries (S,D,D) (arrow). The upper right panel is an
in-plane PCMR image of a 6-year-old child with a secundum atrial septal defect with flow shunting left to
right (blue, arrow). The lower middle panel is an in-plane velocity map in color (red is flow cephalad and blue
is flow caudad) of the patient in Fig. 12.3 with a mid-thoracic coarctation whereas the lower right panel is a
four-dimension flow image of the same patient.

RPA Ao
LPA

RV

FIG. 12.9  Time-resolved, contrast-enhanced cardiovascular magnetic resonance angiogram. From left to
right, the panels are maximum intensity projections that demonstrate the pulmonary phase, the systemic
phase, and the recirculation phase of the patient in Fig. 12.6 with transposition of the great arteries (TGA
[S,D,D]) after an arterial switch operation and a Lecompte maneuver. Each maximum intensity projection is
a three-dimensional image in and of itself. Ao, Aorta; LPA, left pulmonary artery; RPA, right pulmonary artery;
RV, right ventricle.

can be used to image smaller vessels much better than other techniques.
This may also be used to create a shaded surface display, a volume- Blood Flow: Phase Contrast CMR (Figs. 12.7, 12.8,
rendered 3D image (see Figs. 12.10 and 12.11) or a standard tessellation and 12.12)
language (STL) file to be used to build a 3D printed model. A recent After gadolinium imaging, blood flow data are obtained and, similar
development has been an inversion recovery gradient echo (IR-GRE) to cine CMR, are tailored to the lesion under study By performing
technique used in conjunction with either gadofosveset trisodium or PCMR after gadolinium injection, increased signal-to-noise ratio (SNR)
other gadolinium agents in a “slow drip” injection, which is both elec- is obtained in the PCMR sequences (as well as not allowing the scanner
trocardiogram (ECG) based and navigator based; this technique allows to remain idle waiting for 10 minutes to pass for viability imaging—see
for submillimeter resolution and has been extremely useful in infants later). “Through plane retrospectively gated” images are acquired so
and children, especially for small vessels such as the coronary arteries that flow can be measured across the entire cardiac cycle whereas in-
(see Coronary subsection). plane velocity mapping can be used for measuring peak velocities and
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 131

Ao
RPA LPA

Cortra ZZ Corecy 29

RPA LPA Cor

Ao

FIG. 12.10  Time-resolved, contrast-enhanced cardiovascular magnetic resonance angiogram and three-
dimensional (3D) volume rendering. The patient is a 7-year-old child with tetralogy of Fallot with pulmonary
atresia and multiple aorto-pulmonary collaterals who is after unifocalization where the aorto-pulmonary col-
laterals were brought together to form “branch pulmonary arteries.” The top three panels, from left to right,
are maximum intensity projections (MIPs), which demonstrate the pulmonary phase, the systemic phase,
and the recirculation phase of a left arm injection. Because each MIP is a 3D dataset, the bottom left, second
from left and second from right panels are volume-rendered images from each of the phases in the top
panels, respectively. Later, a 3D inversion recovery gradient recalled echo sequence was run with 1 mm
isotropic resolution; this is shown from the anteroposterior view in the lower right panel; the right coronary
artery is shown by the arrow. Ao, Aorta; LPA, left pulmonary artery; RPA, right pulmonary artery.

RPA

RPA LPA

MPA

LPA

RV MPA

FIG. 12.11  Static three-dimensional (3D) contrast-enhanced cardiovascular magnetic resonance angiogram.
These are created from a 3D inversion recovery gradient recalled echo sequence using gadofosveset triso-
dium of the patient in Figs. 12.6 and 12.7 with transposition of the great arteries (TGA [S,D,D]) after an
arterial switch operation and a Lecompte maneuver. The upper left panel is viewing the heart anteroposterior,
while the lower panel and the upper right panel is the heart rotated to further define the long axis of the left
(LPA) and right pulmonary arteries (RPA), respectively. Red arrows point to the proximal origins of the reim-
planted coronary arteries. MPA, Main pulmonary artery; RV, right ventricle.
132 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

MPA = 3.5 L/min/m2


Regurgitant fraction = 33%
Flow m/s
200 Forward stroke volume = 43 cc

150

100

50

0 50 100 150 200 250 300 350 400 450 500 550 600
0

-50
Reverse stroke
volume = 14 cc
-100
Trigger time ms

FIG. 12.12  Phase contrast magnetic resonance. Data from an 8-year-old child with tetralogy of Fallot. The
upper left panel shows the phase (top) and magnitude images (bottom) of the main pulmonary artery velocity
map in systole with forward flow (white and red) while the lower panel shows main pulmonary artery (MPA)
velocity map in diastole with reverse flow (black and blue). The right panel shows the flow curves of MPA
with the data calculating the regurgitant fraction.

visualizing flow directionality. PCMR is extremely useful in CHD for Finally, 4D flow imaging is becoming more commonplace in CHD,
understanding hemodynamics and physiology and is used for most and techniques and tools used are continually being developed. A full
cases. For example, in the patient described previously with a VSD, treatment of 4D flow in CHD is beyond the scope of this chapter;
through-plane PCMR is obtained across the aortic and pulmonary however, one image of 4D flow of a coarctation of the aorta is shown
valves to measure the pulmonary to systemic flow ratio (Qp/Qs), which in Fig. 12.8.
has been validated against oximetry.19 As an internal check (one of the
strengths of the accuracy of PCMR), flow in the branch pulmonary Tissue Characterization (Figs. 12.13 and 12.14)
arteries (should equal flow through the main pulmonary artery) and There are a number of different techniques used in CHD that are helpful
in the cavae (should equal flow in the aorta in the absence of aortic to to characterize myocardial tissue.
pulmonary collaterals) are obtained. As another internal check, LV There are special techniques that do not fall into this generalized
output, as calculated using cine CMR (in the absence of mitral regur- protocol and that may be inserted at different points, depending on
gitation), should equal the forward flow as measured across the aorta the lesion:
using through-plane PCMR, and the same is true of RV output.
PCMR, in conjunction with cine CMR, can calculate other important Late Gadolinium Enhancement Imaging
parameters of function such as valve regurgitation (see Fig. 12.12). For Late gadolinium enhancement (LGE) imaging (fibrosis/scar) (see Figs.
example, patients who have undergone complete common atrioven- 12.13 and 12.14), a gadolinium-based technique that was actually first
tricular canal repair can sometimes manifest left atrioventricular valve described in the mid-1980s40 and can achieve signal intensity differences
regurgitation as a sequela. To assess this quantitatively, the difference between normal and infarcted myocardium of up to 500%,41 is not just
in the LV stroke volume and the forward flow through the aorta would the province of adult heart disease.42–44 Children with congenital abnor-
be the left atrioventricular valve regurgitant volume in the absence of a malities of the coronary arteries (e.g., anomalous left coronary artery
VSD. This volume divided by the stroke volume (multiplied by 100) is from the pulmonary artery), those who have had surgical manipulation
the left atrioventricular valve regurgitant fraction. Forward flow across of the coronaries (e.g., TGA after arterial switch procedure or a Ross
the left atrioventricular valve by using PCMR across it should equal LV procedure), those who have had specific or acquired inflammatory
stroke volume (in the absence of any other residual shunts or aortic diseases (e.g., Kawasaki disease), as well as those who have cardiac
insufficiency) and may be used as an internal check; however, it may tumors (see later) can all benefit from LGE assessment (see Figs. 12.13
be difficult at times to line this imaging plane exactly perpendicular and 12.14). In addition and in general, hearts that have undergone
to flow. surgical reconstruction are candidates for LGE imaging to assess for
As mentioned, PCMR can also be used to assess valve anatomy (see myocardial scar tissue in both the substance of the myocardium as well
Figs. 12.7 and 12.8). Through-plane PCMR can outline the leaflets of as the areas that were reconstructed such as the infundibulum and the
valves when imaged en face (both on the phase and the magnitude pulmonary annulus in a patient after transannular patch repair of tetral-
images) and can be successful when routine cine imaging is not. ogy of Fallot. LGE imaging can also be used to delineate anatomy because
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 133

FIG. 12.13  Tissue characterization: late gadolinium enhancement (LGE) and T2-weighted imaging. Four
different cases are shown in this figure. The upper left and middle panels are from a 6-year-old child with a
full thickness basal inferior wall myocardial infarction demonstrating LGE (arrows) from an anomalous left
coronary artery from the pulmonary artery in the short-axis and long-axis views, respectively. The upper right
image is from another 6-year-old child who had a thoracotomy and thrombectomy a few days before this
imaging; T2 edema imaging was performed demonstrating high signal intensity in the chest wall and RV
free wall (arrows). The lower left and second from left are cine and LGE images, respectively, from a 10-year-
old child with hypertrophic cardiomyopathy and myocardial scarring in the mid-anterior and inferoseptal walls
(arrows). The middle and bottom images are from an 11-month-old baby with scimitar syndrome after repair
with severe ventricular dysfunction. There was dextrocardia because of right lung hypoplasia and significant
lacey myocardium. The middle second from right and middle right panels are cines of the ventricular short-
axis and left ventricular long-axis views, respectively; underneath these are the corresponding LGE images
showing nearly transmural and extensive scarring.

FIG. 12.14  Late gadolinium enhancement (LGE) and perfusion. The left panel is an LGE image of a patient
following complete atrioventricular canal repair and demonstrated high signal intensity over the region of the
patch repair (arrow). The middle panel is an LGE image of a 1-year-old child with a very large right ventricular
fibroma depicting how nicely the tumor lights up (arrow). The right panel is a still frame from a perfusion
series of the same patient in the middle image demonstrating a ring of high-intensity signal in the tumor and
much lower signal intensity in the middle of the tumor (arrows).
134 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

it has been demonstrated that surgically placed patches and valves43 earlier, coronary imaging (Figs. 12.15–12.17) is indicated in a variety
can become bright with this technique. of CHD similar to those who obtain LGE imaging (coronary manipula-
tion such as TGA after arterial switch44 or with native or acquired coro-
Perfusion nary disease such as anomalous left coronary artery from the pulmonary
The same patient population who are candidates for LGE imaging are artery or in Kawasaki disease to assess for aneurysms). In children with
also candidates for myocardial perfusion imaging (see Fig. 12.14), which, fast heart rates (RR intervals <500 ms), minimizing coronary artery
similar to LGE, is not only a consideration for the adult with heart motion by acquiring lines of K-space at the quiescent phase of the
disease.42 It is a gadolinium-based procedure and is typically imaged cardiac cycle may require obtaining data at end systole rather than mid
with vasodilator (e.g., adenosine [“stress”] at 140 µg/kg/min for 4–6 to late diastole, as in adults.
minutes) followed 20 minutes or so later without adenosine (“rest”)
and then followed approximately 10 minutes later by LGE imaging Exercise Cardiovascular Magnetic Resonance
similar to adults. This can also be done with dobutamine stress.45 In With the advent of CMR-compatible ergometers, the patient can exercise
CHD, using adenosine stress perfusion is more common. in the CMR environment with subsequent imaging of function and
flow, generally with “real-time” techniques. This is useful in children
Myocardial Iron with chest pain where perfusion at exercise can be assessed, hypertrophic
The T2* technique is used to assess myocardial iron and hematologists cardiomyopathy (HCM) with poor echocardiographic windows to
use this in conjunction with other parameters such as liver and blood visualize the LV outflow tract, and adolescents with poor exercise per-
iron to adjust chelation therapy. This applies to children with thalas- formance to assess ventricular function. For example, it was discovered
semia, sickle cell disease, and other hematologic disorders of red blood that poor exercise performance of single-ventricle patients after Fontan
cell destruction. was related to increased power loss in the systemic venous pathway in
this manner.49
Myocarditis
Although there are a paucity of data in the pediatric age group for the Myocardial and Blood Tagging
CMR assessment of myocarditis, it is not an uncommon diagnosis to In limited use now, myocardial tagging such as spatial modulation of
entertain in an adolescent with chest pain and elevated troponins. Similar magnetization (SPAMM) divides the ventricle noninvasively into “cubes
to the adult assessment, the Lake Louise criteria are used, including T2 of magnetization” and local deformation of the myocardium can be
imaging (edema), T1 imaging before and after gadolinium administra- visualized and strain calculated. In patients where there is a question
tion (hyperemia and capillary leak), and LGE (see Chapter 35). of regional wall motion abnormalities (e.g., cardiomyopathy such as
in Duchenne muscular dystrophy50 or single ventricle11,22,23) or whether
T1 Mapping or not a focal piece of myocardium is even contracting (e.g., tumor or
The ability to quantify the T1 of the myocardium, both before and HCM), myocardial tagging can be used to assess this both qualitatively
after gadolinium contrast administration, has facilitated determination and quantitatively. With blood tagging, if there are questions about the
of diffuse fibrosis within the heart with a disproportionate amount of presence of an ASD or VSD, a saturation band can be laid down on
collagen. This may be quantified by native T1 time, partition coefficient, the blood to “tag” it as dark on a gradient echo image51 and this dark
and extracellular volume fraction. There is emerging evidence to suggest blood can be followed so that shunting can be visualized; this has been
that this may be of value clinically in CHD (e.g., in tetralogy of Fallot, largely supplanted by in-plane PCMR.
the LV and RV T1 mapping values may be different than in normal
individuals).46 Tumor/Mass Characterization52 (see Fig. 12.14)
Although cardiac tumors are relatively rare, they do occur in pediatrics.
OTHER IMPORTANT TECHNIQUES USED IN Many cardiac tumors and masses can be differentiated from each other
CONGENITAL HEART DISEASE not only by where they occur in the heart, what symptoms they cause,
and at what age they occur but also by their tissue characteristics on
Real-Time Cine Imaging and Phase Contrast CMR CMR. For example, a fibroma will accumulate gadolinium and be signal
Real-time cine imaging acquires single-shot images at a temporal reso- intense on LGE imaging and T1-weighted images after gadolinium
lution of ~40 to 50 ms and can be used in exercise CMR (see later), in administration, whereas lipomas will be signal intense on T1-weighted
patients with arrhythmias, or in those who have a problem remaining images and become signal poor after fat saturation. Tumor character-
motionless in the scanner. In addition, real-time cine CMR can be ization procedures by CMR are a protocol in themselves and typically
performed interactively using a “real-time window” and three other include T1- and T2-weighted images, images with fat saturation, GRE
localizer windows controlled with a cursor and sweeping the ventricle to imaging (e.g., for thrombus), perfusion (e.g., for hemangiomas), LGE
assess ventricular function or finding small atrial septal defect (ASD) and imaging, T1-weighted images after gadolinium administration, and
VSD. In addition, these sweeps can be used to survey the cardiovascular SPAMM. If time permits, functional imaging can be used to assess
anatomy in conjunction with the static bSSFP imaging. for effects of the tumor such as obstruction to flow and decreased
Real-time PCMR acquires single-shot flow images and can have a cardiac output.
similar temporal resolution to real-time cine images. This also may be
used for exercise CMR, patients with arrhythmias, or simply to speed Arrhythmogenic Right Ventricular Cardiomyopathy
up the examination. Arrhythmogenic right ventricular cardiomyopathy (ARVC) is the replace-
Deployment of these two technologies is still limited and analysis ment of myocardium by fatty or fibrofatty tissue53 and imaging fulfills
tools for real-time PCMR are still in their infancy. only one of the criteria set forth in the revised 2010 Task Force manu-
script.54 CMR has been successfully used in adults for this disease;
Coronary Artery Imaging44,47,48 however, in pediatrics, there is a question as to its usefulness.55 The
Using T2-prepared bSSFP imaging combined with the navigator tech- findings by CMR in adults with ARVC studies are highly variable56 but,
nique47 or the IR-GRE technique mentioned in the gadolinium section in general, there is: (1) fatty substitution of the myocardium, which is
Ao

V
Ao

Ao

V V
V

FIG. 12.15  Coronary artery imaging in congenital heart disease. A 2-year-old child with a dilated sinus of
Valsalva aneurysm (V) with the left main coronary artery originating from it. Top left panel is a cine image
showing the dilated sinus. Top middle and right panels are three-dimensional (3D) volume renderings of the
right coronary artery origin (arrows). Lower panels are 3D inversion recovery contrast-enhanced gradient
recalled echo images of the dilated sinus and the origin of the left coronary artery with the raw data (lower
left) and 3D volume renderings (lower middle and right). Ao, Aorta.

RCA LCA RCA LCA


PV
Ao
Ao

Ao

Ao

FIG. 12.16  Coronary artery imaging in congenital heart disease. A 4-year-old child with double outlet right
ventricle (S,D,D) with left juxtaposition of the atrial appendages after Fontan. The top two panels demonstrate
the origins of the coronaries from a three-dimensional (3D) inversion recovery contrast-enhanced gradient
echo sequence in the off-axis axial (left) and off-axis coronal views (right). The right coronary artery (RCA)
originates from the right and posterior facing sinus while the left coronary originates from the left posterior
facing sinus. The bottom two panels are 3D volume-rendered images of the coronary origins and courses
from both anteroposterior (left) and off-axis transverse views (right). Arrows in lower panels point to the
coronaries. Ao, Aorta; LCA, left coronary artery; PV, pulmonary valve.
136 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

LV RV
F
F F

SVC TAo

RPA
LPA

FIG. 12.17  Single ventricle. The patient is a 10-year-old child with supero-inferior ventricles with “pseudo”
criss-cross atrioventricular valves, a right ventricular aorta with pulmonary atresia after hemi-Fontan, and
lateral tunnel Fontan procedures. Panels are static three-dimensional (3D) inversion recovery gradient echo
images after contrast formatted in the axial plane progressing from inferior to superior; images progress
from left to right and from top to bottom. The Fontan baffle (F) is visualized as the circular structure on the
right. The criss-cross of the atrioventricular valves are noted in the upper middle and right panels where the
left atrium connects to the inferior left ventricle (LV) and the right atrium connects to the superior right
ventricle (RV) (note how the arrows cross). The right pulmonary artery (RPA) is moderately hypoplastic in
the lower left panel and there is a right aortic arch, as seen by the transverse aortic arch (TAo) crossing to
the right of the trachea (and over the right bronchus). The lower right panel is a 3D volume-rendered image;
there is a single anterior coronary artery, which gives rise to the left, right, and interventricular coronary
arteries (arrows). LPA, Left pulmonary artery; SVC, superior vena cava.

generally difficult to visualize given the thin RV free wall and the epi- 1.5 T to 3 T (which comes with its own separate issues). These modi-
cardial fat being mistaken for fatty infiltration; (2) ectasia of the RV fications come at the cost of imaging time but are worth it to get high-
outflow tract; (3) dyskinetic bulges or dyskinesia of RV wall motion; quality diagnostic images.
(4) a dilated RV; (5) a dilated right atrium; and (6) fixed RV wall thin- At times, for the smallest voxels, even these strategies cannot be
ning with decreased RV wall thickening. The protocol includes employed and affect SNR to the degree needed. In these instances, the
T1-weighted imaging (debatable), cine for ventricular function, one- CMR imager must be satisfied with larger voxels and this is done by
dimensional RV myocardial tagging if needed to assess regional wall decreasing the matrix size—typically, when imaging the adult, a base
motion, and phase-encoded velocity mapping, and LGE has been shown matrix (in frequency encoding direction) of 256 or 192 is used and this
to be helpful.57 can be decreased in varying steps to as low as 128, which can yield
diagnostic images well in the range of field-of-view (FOV) of ~200 mm
or less, yielding pixels of 1.5 to 2 mm or less depending on the FOV.
TECHNICAL CONSIDERATIONS IN PEDIATRIC CMR Slice thicknesses generally do not go below 2.5 to 3 mm because of
Because of the demands of high spatial and temporal resolution in SNR considerations. Note that for gadolinium images, however, because
infants and children along with the inability to breath-hold, alternative of the T1 shortening from the contrast agent, we can achieve submil-
means are needed to allow the CMR sequences designed for adults to limeter isotropic resolution.
work. Strategies have been created to successfully image these youngsters When imaging small children, a small FOV is needed and, at certain
and have been employed to various degrees for many years. settings, sequences are written that do not allow for the FOV to go
down very low (e.g., 150 mm in a 3-kg baby). Similarly, slice thicknesses
Spatial and Temporal Resolution need to be smaller than when imaging the adult and, because of SNR
Small voxels needed to image infants and small children can be prob- considerations, at times the matrix size needs to be smaller and, at
lematic because of the poor SNR. By employing parallel imaging tech- certain settings, sequences are written that do not allow for these manipu-
niques, SNR may be further impaired. Simply imaging without lations. When that occurs, the imager must find combinations by trial
consideration for SNR will result in a poor-quality “grainy” image, and error of changing bandwidths, TR, matrix, segments of k-space
which will be nondiagnostic. To increase the SNR, a number of strate- (“views”), FOV, and slice thickness to allow for optimal imaging for
gies have been employed either in isolation or in combination such as the size of the patient. Each manufacturer has their own idiosyncrasies
using multiple averages (excitations), phase oversampling, decreasing that make it prohibitive to list all the combinations here. By relaxing
the bandwidth, forgoing using parallel imaging, or even moving from some parameters (e.g., longer TR, increasing the segments), other
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 137

parameters may be allowed to be optimized to image the smaller patients 2. Navigator-based techniques use a coil to monitor diaphragmatic
(e.g., smaller FOV). motion and when the diaphragm is at a certain position (typically
Because heart rates can be very high in infants (not uncommonly, expiration), the lines of k-space are accepted for image reconstruc-
140–170 beats per minute), a successful CMR scan in this age group tion; outside this position, those lines of k-space are discarded.
needs to accommodate this. As a rule of thumb, under an RR interval of 3. “Real-time” and other single-shot techniques.
500 ms, 15 to 20 phases of the cardiac cycle should be used; from 500 to In addition to these three strategies, placing a saturation band over
750 ms, 20 to 25 phases should be obtained; and more than 750 ms, 25 the chest wall can also aid in minimizing any respiratory artifact that
to 30 phases should be used. To be able to obtain this level of temporal does occur. These are the most common strategies that allow high-
resolution at fast heart rates, the TR needs to be low. To accomplish this, quality images to be created, albeit at the cost of a little extra time,
in general, the number of segments needs to be as low as possible. If the which is not prohibitive. It is true that breath-holding will yield much
TR and the number of segments are not low enough, in retrospectively crisper images; however, the marginal benefit in many instances is not
gated imaging, the walls of the heart will appear blurry and with a worth the cost because the discussed strategies for breathing freely yield
double or triple shadow whereas in prospectively triggered imaging, the diagnostic information.
heart motion will look stilted and jerky. Adequate temporal resolution
can be obtained at fast heart rates with three segments at a minimum. Contrast-Based Techniques
In retrospective gating sequences, it is important to understand that The information applies to all types of imaging, but the gadolinium-
the CMR scanner is acquiring data continuously and recording the based techniques have additional considerations.
ECG tracing. After all the lines of k-space are acquired, the computer 1. When performing “static” 3D gadolinium imaging using the “tracking”
then “bins” each line of k-space to the closest phase of the cardiac cycle or combined applications to reduce exposure (CARE) bolus technique,
it is calculating, interpolating the data as it is performing the “bin.” it should be noted that children will have a quick circulatory time.
Therefore be aware that the number of calculated phases should not The gadolinium will reach the structure of interest much quicker
exceed twice the measured phases (essentially the RR interval [ms]/TR than in adults and the imager should not be caught off guard when
[ms], where TR is the line TR × lines of k-space obtained) because this occurs, missing the first-pass gadolinium in the process.
there should be two measured points between each interpolated point 2. For viability imaging, as mentioned, two or more RR intervals may
to obtain robust data. The formula used is: be needed to allow the tissue to relax enough for the next excitation.
3. When performing the “slow-drip” 3D inversion recovery (IR) tech-
2 × RR interval (ms) number of calculated phases = TR (ms)
nique (as well as other contrast-based techniques), even at a “double
where TR is defined as the line TR × lines of k-space obtained. This is dose,” the dosage may be small (e.g., 2–3 mL). In this particular
especially important in PCMR for accurate data to be obtained. case, the contrast is diluted (≥1 : 1) for better overall image quality.
At times, it is advantageous to use “single-shot” imaging in children 4. The perfusion technique obtains images at a single slice position at
because of respiratory motion or arrhythmia, where all the lines of different time points in the cardiac cycle after a delay. With heart
k-space are acquired in one heartbeat. A typical single-shot imaging rates in infants and children so high, it may only be possible to fit
sequence is the HASTE sequence mentioned earlier, but other sequences two slices (or, rarely, one slice with really high heart rates) within
such as static bSSFP and cine bSSFP can be performed in single-shot an RR interval. If this occurs, the imager can perform the sequence
mode. If the heart rate is too fast, it may be advantageous to obtain over 2 RR intervals without degradation of diagnostic quality and
the image over two heartbeats (i.e., imaged at the end of two heart- obtain more slices (3–4) during the scan.
beats [2 × RR interval]) instead of one and this should be considered 5. Given data demonstrating very long-term retention of gadolinium
in imaging sequences such as HASTE, double inversion dark blood in multiple tissues,43a until more safety data are known, the routine
imaging, or viability imaging if the heart cannot be successfully imaged use of gadolinium in infants and children should be individually
in one heartbeat. assessed and likely minimized.
When performing tissue characterization with fast heart rates, it is
imperative that the spins in the tissue are allowed to relax before the WORKED EXAMPLES OF CMR FOR CONGENITAL
next excitation. In these instances, the RR interval set in the acquisition HEART DISEASE
window may be purposely doubled or even tripled and the acquisition
of data occurs at the end of the last heartbeat. This technique has saved Transposition of the Great Arteries
many scans in the past from failure, albeit at the cost of doubling or (see Figs. 12.6, 12.9, and 12.11)
tripling the time involved. With the aorta arising from the RV and the pulmonary artery arising
from the LV, the state-of-the-art surgical procedure for simple TGA is
Inability of Pediatric Patients to Breath-Hold the arterial switch procedure, where both great vessels are surgically
To perform CMR in patients who cannot hold their breath, general transposed to arise normally and the coronary arteries are implanted
anesthesia may be used where the anesthesiologist can sustain the above the native pulmonary (neoaortic) valve. The pulmonary arteries
“breath-hold.” That being said, deep sedation has been successfully used are typically “draped” over the ascending aorta in the Lecompte maneuver
for many years without untoward effects and has allowed the patient (see Figs. 12.6, 12.9, and 12.11), which tends at times to result in left
to breathe freely during the CMR examination. The advantage to deep pulmonary artery stenosis.
sedation and breathing freely is of course the less-invasive nature of For a typical assessment, after the set of axial images, cine of the
this approach and imaging in “normal” day-to-day physiology. pulmonary arteries (see Fig. 12.6) are performed in both long axes to
Imaging the patient who is allowed to breathe freely uses the assess for stenosis, which generally can occur at the takeoff of the left
following: pulmonary artery or in the right pulmonary artery in between the
1. Multiple excitations to “average out” the respiratory motion. Typi- superior vena cava and the aorta. Cine of the RV and LV outflow tracts
cally, three excitations are needed to obtain good image quality are used to evaluate for obstruction. In addition, cines of the four-
(which also increases SNR), although at times, four or five excitations chamber view and a set of short-axis contiguous cines of both ventricles
may be needed for the vigorous breather. are used to quantitate ventricular performance and to search for residual
138 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

VSD. PCMR across the neoaortic and neopulmonary valves are used but also with the various surgical reconstructive techniques of these
to quantitate cardiac output; this same technique is used across the very complex patients. A comprehensive treatment of this evaluation
right and left pulmonary arteries to determine flow distribution to both is beyond the scope of this chapter, and the reader is referred to a more
lungs and to assess gradients across stenotic pulmonary arteries.20 If a specialized text or articles.6,8,11,12,18,21,22,24,59,60
VSD or residual ASD is suggested, PCMR across the cavae and the As a patient with a single ventricle progresses through staged recon-
descending aorta at the level of the diaphragm is used as a check on struction, specific targets of examination change, but the overall goal
neoaortic flow. Gadolinium is used for 3D reconstruction of the pul- of assessing anatomy, physiology, hemodynamics, and function remains.
monary arteries and aorta (see Figs. 12.9 and 12.11) and LGE44 is typi- It goes without saying that each individual patient may have different
cally performed 10 minutes later to evaluate for myocardial scarring. needs and reasons for the CMR examination, which takes precedence
In general, coronary artery imaging is performed to image the manipu- over other considerations. At all stages of surgical reconstruction, includ-
lated coronary arteries in the surgical treatment of this disease44 by ing the single ventricle in the native state, the following is the minimum
either 3D T2-prepared bSSFP coronary imaging or by contrast-enhanced assessment:
IR 3D GRE imaging. • Aortic arch imaging, aimed mostly at patients with an aortic to
Discussion of TGA after an atrial switch (Senning or Mustard) or pulmonary anastomosis, to assess for aortic arch obstruction.
corrected TGA (L-TGA) is outside the scope of this chapter; however, • Pulmonary artery imaging,6,18,24 to assess for pulmonary artery ste-
ventricular function in a child with L-TGA is shown in Fig. 12.5. nosis, hypoplasia, and discontinuity (see Figs. 12.2 and 12.17).
• Assessment of the Fontan baffle7,8 (see Figs. 12.2, 12.17, and
Single Ventricles (Figs. 12.2, 12.16, 12.17, and 12.18) 12.18), atrial septal defect (see Fig. 12.18), ventricular outflow tract
When only one usable ventricle is present to effectively pump blood obstruction (e.g., patients with a bulboventricular foramen), aortic-
while the other is hypoplastic or when both ventricles are linked in pulmonary collaterals, veno-veno collaterals or other anomalous
such a way as separation of the circulations into two pumping chambers venous structures, and pulmonary or systemic venous obstruction.
is impossible, the heart is classified as a functional single ventricle. The • Ventricular function11,21,22,23 (see Fig. 12.18), including biventricular
aorta may be hypoplastic and there may be other associated anomalies regional wall motion abnormalities, ejection fraction, end-diastolic
such as anomalous venous connections. Single ventricles undergo staged volume and mass, stroke volume, cardiac index, and atrioventricular
surgical reconstruction (either in two or three stages, depending on the valve regurgitant fraction.
anatomy and physiology), culminating in the Fontan procedure.58 The • PCMR18 to assess for cardiac index, Qp/Qs, flows to both lungs,
CMR imager must be familiar not only with the various forms of single regurgitant fraction of the semilunar (and indirectly) atrioventricu-
ventricles, associated anomalies, and the physiologic/functional sequelae lar valve to assess for ventricular outflow tract obstruction, and

RV
LV

LV

RV
RV

LV

RV

LV

FIG. 12.18  Single ventricle. Cines and a three-dimensional (3D) volume-rendered image from the patient in
Fig. 12.17. The upper left and second from upper left are cine images in orthogonal planes demonstrating
the left atrium connecting to the inferior left ventricle (LV), whereas the middle left and second from middle
left are cine images in orthogonal planes demonstrating the right atrium connecting to the superior right
ventricle (RV). The directions of the flows appear to be criss-crossed; however, twisting the plane, a cine in
the lower left “uncrosses” the atrioventricular relations, and hence the term “pseudo” criss-cross in Fig.
12.16. Lower middle panel shows the cine short axis of the supero-inferior relationships of the ventricle,
while the lower right panel is a cine of the Fontan baffle (arrows). Upper right is the 3D volume-rendered
image showing the Fontan baffle (arrows).
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 139

to quantify aortic to pulmonary collateral flow (measured as the valves will “light up.” Perfusion may also be performed to evaluate for
difference between aortic flow and caval return or the difference defects. Qp/Qs is as in the stage I patients.
between pulmonary venous and pulmonary arterial flow).35–37 There-
fore PCMR is performed across the cavae, the branch pulmonary Coarctation of the Aorta (Figs. 12.4, 12.8, 12.19,
arteries, the semilunar valves, atrioventricular valve inflow, across and 12.20)
the descending aorta at the level of the diaphragm, and across the One of the more common congenital heart lesions and a very common
pulmonary veins. referral to CMR, coarctation is defined as an obstruction in the isthmus
In the single ventricle’s native state, because much less is known or descending aorta. It may have any number of associated abnormali-
about the anatomy than at other stages, it is important to do an even ties from bicuspid aortic (see Fig. 12.8) valve to complex diseases such
greater detailed anatomic assessment. Anomalous venous structures as Shone complex, double orifice mitral valve, or posterior malalignment
such as a decompressing vein from the left atrium (see Fig. 12.3; even ventricular septal defect. In older children, multiple collaterals can be
though that is not a single ventricle, it demonstrates a levoatrialcardinal visualized, which is the result of the body’s attempt to bypass the obstruc-
vein), the presence of a left superior vena cava, delineation of visceral tion. Surgery for the coarctation itself can be as simple as a patch over
situs for heterotaxy, presence of an inferior vena cava, and the hepatic the narrowed portion to a complex ascending-to-descending aortic
venous drainage, etc., are all important details to sort out (a complex conduit.
anatomical case is seen in Figs. 12.17 and 12.18). In addition, because CMR can provide exquisite detail of the anatomy and physiology
some patients may have needed resuscitation, assessment of ventricular needed for diagnosis and treatment. After the stack of bSSFP static
function, myocardial scarring, and valve insufficiency is also extremely axial images, which will demonstrate a decreased aortic diameter at
important to evaluate. the level of the coarctation, the candy cane view using cine imaging
After the stage I procedure for hypoplastic left heart syndrome, for (bSSFP or gradient echo imaging) is employed to demonstrate the
example, aortic arch imaging is important to evaluate the initial repair. coarctation narrowing; turbulence as indicated by a loss of signal distal
Besides assessment of the distal aortic arch for obstruction and the aortic to the narrowing will occur. It is important to obtain images on either
to pulmonary anastomosis, visualization of the aortic to pulmonary shunt side of the “ideal” candy cane view to ensure that the narrowing is not
(typically it is a right Blalock-Taussig shunt) or the RV to pulmonary artifactual and that the aorta doesn’t get bigger out of the plane of the
artery shunt (i.e., Sano procedure) is important. This is generally done “ideal” image. Cine CMR in the orthogonal plane at the level of the
with double inversion dark blood imaging, 3D SPACE, or gadolinium; narrowing will aid in characterizing the narrowing further. Cine is also
occasionally, gradient echo imaging is successful as the turbulence across obtained in the four-chamber and ventricular short-axis views (a stack
this structure by cine generally causes signal loss. As for Qp/Qs, Qp is of short-axis images from base to apex) to obtain LV mass assessing
the sum of the flows in the pulmonary veins whereas Qs is caval return; for hypertrophy. Aortic valve morphology and the LV outflow tract in
aortic valve flow will overestimate and the sum of pulmonary arterial two orthogonal views are essential assessment and may be visualized by
flow will underestimate Qs and Qp, respectively, because of aortic to cine or PCMR (see Fig. 12.8); the two views across the LV outflow tract
pulmonary collateral flow (and systemic to pulmonary artery shunt are important, especially to determine if subaortic stenosis (membrane,
flow in the case of aortic valve flow). PCMR across the systemic to tunnel) is present. In-plane PCMR and cine can be used to determine
pulmonary artery shunt and in each pulmonary artery is performed where the level of obstruction begins; PCMR can be especially important
using high velocity encoding (VENC), (e.g., 400 cm/s) and as short a to determine the peak velocity. Aortic stenosis or regurgitation secondary
TE as possible. The status of the atrial septal defect should be assessed to a bicuspid or unicuspid valve (Fig. 12.18) can be assessed by cine as
and, because this is a volume-loaded stage, ventricular function is also well as PCMR, and it is important to obtain aortic valve measurements
a key imaging goal (see Figs. 12.17 and 12.18). by long-axis views of the LV outflow tract by cine if valvuloplasty is
After the bidirectional Glenn/hemi-Fontan stage, imaging the superior contemplated. Through-plane phase-encoded velocity mapping across
vena cava to pulmonary artery anastomosis is the major difference with the aortic valve is used to obtain the cardiac index and regurgitant frac-
the stage I patients. This can be done with any one of numerous tech- tion and peak velocity is obtained for aortic regurgitation and stenosis,
niques, including cine or gadolinium sequences, because the flows are respectively. In addition, through-plane PCMR just below or above the
generally low velocity. Qp/Qs is as in the stage I patients. Pulmonary coarctation and at the diaphragm can quantify collateral blood flow.62
arterial flow uses flow in the superior vena cava with flow in both In-plane PCMR can be used to obtain peak instantaneous velocity as
branch pulmonary arteries as an internal check along with flow in the well. 3D gadolinium is used to assess for collaterals and to create a 3D
aorta. If a hemi-Fontan was performed, imaging should assess whether image of the aorta and the coarctation, which is especially important
any leak was present from the superior vena cava–pulmonary artery in tortuous vessels, which cannot be imaged in just one plane.
anastomosis into the atrium.
After the Fontan procedure, the most important structure to image
is the entire systemic venous pathway (Fontan baffle) for obstruction
A TASTE OF THE FUTURE
or clot (see Figs. 12.2, 12.17, and 12.18). Visualizing the fenestration With the many advances in CMR, the future holds great promise in
flow (using cine or in-plane PCMR) and assessment for thrombus in CHD, with work in the area of interventional CMR (e.g., balloon val-
the systemic venous pathway are important points to determine. Quan- vuloplasty, stent placement), molecular imaging, tissue and blood char-
tification of the fenestration flow is obtained by the difference in PCMR acterization techniques to evaluate the heart for myocardial iron, diffuse
flows in the Fontan baffle immediately above the hepatic veins and fibrosis, and oxygen content, as well as continued development of 4D
immediately below the branch pulmonary arteries. Because it is known flow imaging and “real-time” flow assessment. These advances are readily
that patients who have undergone the Fontan procedure have poor seen in the work done to develop the field of functional fetal CMR
ventricular function,21 a cine short-axis stack is an essential part of the (Fig. 12.21). Work published using “real-time” imaging demonstrated
examination. In addition, gadolinium-enhanced imaging can help the feasibility of this approach in patients with in utero diagnosis of
determine the presence of collaterals and to assess the aortic arch. LGE hypoplastic left heart syndrome to quantify ventricular volumes and
imaging61 can be used to visualize any myocardial scarring that has cardiac output.63 With other approaches to advance this field, increased
resulted and both the reconstructed aorta as well as the atrioventricular spatial and temporal resolution along with unique ways of “gating”
140 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

RV

LV
LV

FIG. 12.19  Coarctation of the aorta—multiple techniques. This is the 3-year-old child in Fig. 12.4 with a mid-
thoracic long segment aortic coarctation of the aorta. The top left and middle panels are the four-chamber
and short-axis views, respectively, demonstrating left ventricular (LV) hypertrophy. The upper right and lower
left and second from left show the coarctation at mid-thorax in the candy cane view using contrast-enhanced
inversion recovery gradient echo imaging, cine balanced steady-state free precession (bSSFP) and cine gradient
recalled echo, respectively (arrows). Note how on bSSFP imaging, the turbulence causes much dephasing
distally so that the descending aorta loses signal. The lower second from right panel is an in-plane velocity
mapping of the jet at the coarctation site; peak velocity measured 410 cm/s. The lower right panel is a gradi-
ent echo cine image orthogonal to the “candy cane” view through the coarctation site. RV, Right ventricle.

FIG. 12.20  Coarctation of the aorta—three-dimensional volume rendered. This is the 3-year-old child in Figs.
12.4 and 12.8 with a mid-thoracic long segment coarctation of the aorta. The panel on the left shows only
the left-sided structures, whereas the panel on the right has right-sided structures as well. Arrows point to
the coarctation.
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 141

Sagittal

Axial Coronal

FIG. 12.21  Functional fetal cardiovascular magnetic resonance image. The patient is 36 weeks’ gestation
with the diagnosis of tetralogy of Fallot and pulmonary atresia. The upper left panel is a still frame from a
four-chamber cine of the heart (arrow). The mother’s anterior abdomen is at the top of the image. The upper
right image is a still frame of a left ventricular outflow tract cine (arrow) with the mother’s anterior abdomen
to the left of the image. The head is at the bottom. The lower middle panel is a still frame of a cine (arrow)
and demonstrates a short-axis view of the heart with the mother’s anterior abdomen to the left of the image.
The head is at the bottom.

may allow for phase contrast flow assessments and the routine use of reasons. The imager needs to understand the anatomy, physiology, and
functional fetal CMR in the future. Pioneering work on blood flow in function of native CHD as well as the treatments (surgical, catheter
the fetus and fetal cerebral blood flow by CMR is at the cutting edge based) to perform the examination correctly in addition to tackling
of this field.64,65 the myriad of technical issues associated with imaging in pediatrics.
As mentioned earlier, until more data are known regarding pos- With both hardware and software advances in the field, CMR can only
sible toxicity of long-term gadolinium retention, it is likely that non– become more important in CHD in the years to come.
gadolinium based CMR imaging will become more common in CHD
in the coming years.
REFERENCES
CONCLUSION A full reference list is available online at ExpertConsult.com

CMR of CHD in infants and children as well as in adolescents and


adults is a unique field unto itself in the world of CMR for many
CHAPTER 12  Special Considerations: Cardiovascular Magnetic Resonance in Infants and Children 141.e1

REFERENCES 20. Gutberlet M, Boeckel T, Hosten N, et al. Arterial switch procedure for
D-transposition of the great arteries: quantitative midterm evaluation of
1. Didier D, Higgins CB, Fisher M, Osakai L, Silverman NH, Cheitlin MD. hemodynamic changes with cine MR imaging and phase-shift velocity
Congenital heart disease: gated magnetic resonance imaging in 72 mapping—initial experience. Radiology. 2000;214:467–475.
patients. Radiology. 1986;158:227–235. 21. Fogel MA, Weinberg PM, Chin AJ, Fellows KE, Hoffman EA. Late
2. Fletcher BD, Jacobsteink MD, Nelson AD, Riemenschneider TA, Alfidi RJ. ventricular geometry and performance changes of functional single
Gated magnetic resonance imaging of congenital cardiac malformations. ventricle throughout staged Fontan reconstruction assessed by magnetic
Radiology. 1984;150:137–140. resonance imaging. J Am Coll Cardiol. 1996;28(1):212–221.
3. Higgins CB, Byrd BF, Farmer DW, Osakai L, Silverman N, Cheitlin MD. 22. Fogel MA, Weinberg PM, Fellows KE, Hoffman EA. A study in
Magnetic resonance imaging in patients with congenital heart disease. ventricular-ventricular interaction: single right ventricles compared with
Circulation. 1984;70:851–860. systemic right ventricles in a dual chambered circulation. Circulation.
4. Bank ER. Magnetic resonance of congenital cardiovascular disease. An 1995;92(2):219–230.
update. Radiol Clinic North Am. 1993;31:553–572. 23. Fogel MA, Gupta KB, Weinberg PW, Hoffman EA. Regional wall motion
5. Bornemeier RA, Fellows KE, Fogel MA, Weinberg PM. Anatomic and strain analysis across stages of Fontan reconstruction by magnetic
delineation of congenital heart disease with 3D magnetic resonance resonance tagging. Am J Physiol. 1995;269(Heart and Circulatory
imaging. In: Hoffman EA, Acharya RS, eds. Medical Imaging 1994: Physiology 38):H1132–H1152.
Physiology and Function From Multidimensional Images. Vol. 2168. 24. Fogel MA, Weinberg PM, Hoydu A, et al. The nature of flow in the
Bellingham, WA: SPIE; 1994:184–194. systemic venous pathway in Fontan patients utilizing magnetic resonance
6. Fogel MA, Ramaciotti C, Hubbard AM, Weinberg PW. Magnetic blood tagging. J Thorac Cardiovasc Surg. 1997;114:1032–1041.
resonance and echocardiographic imaging of pulmonary artery size 25. Ho V, Prince M. Thoracic MR aortography: imaging techniques and
throughout stages of Fontan reconstruction. Circulation. 1994;90: strategies. Radiographics. 1998;18(2):287–309.
2927–2936. 26. Beekman R, Hazekamp M, Sobotka M, et al. A new diagnostic approach
7. Bornemeier RA, Weinberg PM, Fogel MA. Angiographic, to vascular rings and pulmonary slings: the role of MRI. Magn Reson
echocardiographic and three-dimensional magnetic resonance imaging of Imaging. 1998;16(2):137–145.
extracardiac conduits in congenital heart disease. Am J Cardiol. 27. van Son J, Julsrud P, Hagler D, et al. Imaging strategies for vascular rings.
1996;78(6):713–717. Ann Thorac Surg. 1994;57(3):604–610.
8. Fogel MA, Hubbard A, Weinberg PM. A simplified approach for 28. Didier D, Ratib O, Beghetti M, et al. Morphologic and functional
assessment of intracardiac baffles and extracardiac conduits in congenital evaluation of congenital heart disease by magnetic resonance imaging.
heart surgery with two- and three-dimensional magnetic resonance J Magn Reson Imaging. 1999;10:639–655.
imaging. Am Heart J. 2001;142(6):1028–1036. 29. Fleenor JT, Weinberg PM, Kramer SS, Fogel M. Vascular rings and their
9. Fogel MA, Rychik J, Chin A, Hubbard A, Weinberg PM. Evaluation and effect on tracheal geometry. Pediatr Cardiol. 2003;24:430–435.
follow-up of patients with left ventricular apical to aortic conduits using 30. Mor-Avi V, Sugeng L, Weinert L, et al. Fast measurement of LV mass with
two- and three-dimensional magnetic resonance imaging and doppler real-time 3-dimensional echocardiography: comparison with MRI.
echocardiography: a new look at an old operation. Am Heart J. Circulation. 2004;110:1814–1818.
2001;141:630–636. 31. Bu L, Munns S, Zhang H, et al. Rapid full volume data acquisition by
10. Weinberg PM, Hubbard AM, Fogel MA. Aortic arch and pulmonary real-time 3-dimensional echocardiography for assessment of LV indexes
artery anomalies in children. Semin Roentgenol. 1998;33(3):262–280. in children: a validation study compared with MRI. J Am Soc
11. Fogel MA, Weinberg PM, Gupta KB, et al. Mechanics of the single left Echocardiogr. 2005;18:299–305.
ventricle: a study in ventricular-ventricular interaction II. Circulation. 32. Lee VS, Resnick D, Bundy JM, Simonetti OP, Lee P, Weinreb JC. MR
1998;98:330–338. Evaluation in one breath hold with real-time true fast imaging with
12. Fogel MA, Weinberg PM, Rychik J, et al. Caval contribution to flow in the steady-state precession. Radiology. 2002;222:835–842.
branch pulmonary arteries of fontan patients using a novel application of 33. Arun J, Kowallick JT, Merboldt KD, et al. Real-time flow MRI of the aorta
magnetic resonance presaturation pulse. Circulation. 1999;99:1215–1221. at a resolution of 40 msec. J Magn Resonan Imag. 2014;40:206–213.
13. Donofrio MT, Clark BJ, Ramaciotti C, et al. Regional wall motion and 34. Edelman RR, Chien D, Kim D. Fast selective black blood MR imaging.
strain of transplanted hearts in pediatric patients using magnetic Radiology. 1991;181:655–660.
resonance tagging. Am J Physiol. 1999;277:R1481–R1487. 35. Whitehead KK, Gillespie MJ, Harris MA, Fogel MA, Rome JJ. Noninvasive
14. Fogel MA, Weinberg PM, Hubbard A, Haselgrove J. Diastolic quantification of systemic to pulmonary collateral flow: a major source of
biomechanics in normal infants utilizing MRI tissue tagging. Circulation. inefficiency in patients with superior cavopulmonary connections. Circ
2000;102:218–224. Cardiovasc Imaging. 2009;2:405–411.
15. Eyskens B, Reybrouck T, Bagaert J, et al. Homograft insertion for 36. Glatz AC, Rome JJ, Small AJ, et al. Systemic-to-pulmonary collateral flow,
pulmonary regurgitation after repair of tetralogy of Fallot improves as measured by cardiac magnetic resonance imaging, is associated with
cardiorespiratory exercise performance. Am J Cardiol. 2000;85:221–225. acute post-Fontan clinical outcomes. Circ Cardiovasc Imaging. 2012;5:
16. Niezen RA, Helgbing WA, van der Wall EE, van der Geest RJ, Rebergen 218–225.
SA, de Roos A. Biventricular systolic function and mass studied with MRI 37. Grosse-Wortmann L, Al-Otay A, Yoo SJ. Aortopulmonary collaterals after
imaging in children with pulmonary regurgitation after repair for bidirectional cavopulmonary connection or Fontan completion:
tetralogy of Fallot. Radiology. 1996;201:135–140. quantification with MRI. Circ Cardiovasc Imaging. 2009;2:219–225.
17. Rebergen SA, Chin JGJ, Ottenkamp J, van der Wall EE, de Roos A. 37a.  Russo RJ, Costa HS, Silva PD, et al. Assessing the risks associated with
Pulmonary regurgitaion in the late postoperative follow-up of tetralogy MRI in patients with a pacemaker or defibrillator. N Engl J Med.
of Fallot. Volumetric quantification by MR velocity mapping. Circulation. 2017;376:755–764.
1993;88:2257–2266. 38. Martin ET, Coman JA, Shellock FG, Pulling CC, Fair R, Jenkins K.
18. Rebergen SA, Ottenkamp J, van der Wall EE, Chin JDJ, de Roos A. Magnetic resonance imaging and cardiac pacemaker safety at 1.5-Tesla.
Postoperative pulmonary flow dynamics after Fontan surgery: assessment J Am Coll Cardiol. 2004;43:1315–1324.
with nuclear magnetic resonance velocity mapping. J Am Coll Cardiol. 39. Finn JP, Baskaran V, Carr JC, et al. Thorax: low-dose contrast-enhanced
1993;21:123–131. three-dimensional MR angiography with subsecond temporal
19. Beerbaum P, Korperich H, Barth P, Esdorn H, Gieseke J, Meyer H. resolution—initial results. Radiology. 2002;224:896–904.
Non-invasive quantification of left-to-right shunt in pediatric patients. 40. McNamara MT, Tscholakoff D, Revel D, et al. Differentiation of reversible
Phase-contrast cine magnetic resonance imaging compared with invasive and irreversible myocardial injury by MR imaging with and without
oxymetry. Circulation. 2001;10:2476–2482. gadolinium-DTPA. Radiology. 1986;158:765–769.
141.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

41. Simonetti OP, Kim RJ, Fieno DS, et al. An improved MRI technique for 53. Basso C, Thiene G, Corrado D, Angelini A, Nava A, Valente M.
the visualization of myocardial injury. Radiology. 2001;218:215–223. Arrhythmogenic right ventricular cardiomyopathy: dysplasia, dystrophy
42. Prakash A, Powell AJ, Krishnamurthy R, Geva T. Magnetic resonance or myocarditis. Circulation. 1996;94:983–991.
imaging evaluation of myocardial perfusion and viability in congenital 54. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic
and acquired pediatric heart disease. Am J Cardiol. 2004;93:657–661. right ventricular cardiomyopathy/dysplasia: proposed modification of the
43. Harris MA, Ghods S, Weinberg PM, Fogel MA. Magnetic resonance task force criteria. Circulation. 2010;121(13):1533–1541.
delayed enhancement for detection of fibrous tissue in postoperative 55. Fogel MA, Weinberg PM, Rhodes L. Usefulness of magnetic resonance
pediatric patients with various forms of congenital heart disease. imaging for the diagnosis of right ventricular dysplasia in children. Am J
J Cardiovasc Magn Reson. 2005;7:157. Cardiol. 2006;97(8):1232–1237.
43a.  Ramalho J, Ramalho M, Jay M, et al. Gadolinium toxicity and treatment. 56. Midiri M, Finazzo M, Brancato M, et al. Arrhythmogenic right
Magn Reson Imaging. 2016;34:1394–1398. ventricular dysplasia: MR features. Eur Radiol. 1997;7:307–312.
44. Taylor AM, Dymarkowski S, Hamaekers P, et al. MR coronary 57. Tandri H, Saranathan M, Rodriguez ER, et al. Noninvasive detection of
angiography and late-enhancement myocardial MR in children who myocardial fibrosis in arrhythmogenic right ventricular cardiomyopathy
underwent arterial switch surgery for transposition of great arteries. using delayed-enhancement magnetic resonance imaging. J Am Coll
Radiology. 2005;234:542–547. Cardiol. 2005;45(1):98–103.
45. Paetsch C, Jahnke A, Wahl R, et al. Comparison of dobutamine stress 58. Fontan F, Baudet E. Surgical repair of tricuspid atresia. Thorax.
magnetic resonance, adenosine stress magnetic resonance, and adenosine 1971;26:240–248.
stress magnetic resonance perfusion. Circulation. 2004;110:835–842. 59. Fogel MA, ed. Ventricular Function and Blood Flow in Congenital Heart
46. Kozak MF, Redington A, Yoo SJ, Seed M, Greiser A, Grosse-Wortmann L. Disease. Malden, MA: Blackwell Futura; 2005.
Diffuse myocardial fibrosis following tetralogy of Fallot repair: a T1 60. Fogel MA. Cardiac magnetic resonance of single ventricles. J Cardiovasc
mapping cardiac magnetic resonance study. Pediatr Radiol. 2014;44(4): Magn Reson. 2006;8(4):661–670.
403–409. 61. Harris M, Johnson T, Weinberg P, Fogel M. Delayed enhancement
47. Kim WY, Danias PG, Stuber M, et al. Coronary magnetic resonance cardiovascular magnetic resonance identifies fibrous tissue in children
angiography for the detection of coronary stenoses. NEJM. after congenital heart surgery. J Thorac Cardiovasc Surg.
2001;345:1863–1869. 2007;133:676–681.
48. Tangcharoen T, Hegde S, Bell A, et al. Feasibility of whole-heart, 62. Steffens JC, Bourne MW, Sakuma H, O’Sullivan M, Higgins CB.
steady-state free precession magnetic resonance coronary angiography Quantification of collateral blood flow in coarctation of the aorta by
(MRCA) in infants and children with congenital heart disease. velocity encoded cine magnetic resonance imaging. Circulation.
J Cardiovasc Magn Reson. 2008;10(suppl 1):A80 (abstract 219). 1994;90:937–943.
49. Khiabani RH, Whitehead KK, Han D, et al. Exercise capacity in single 63. Fogel MA, Wilson DR, Flake A, et al. A new method of functional
ventricle patients after Fontan correlates with hemodynamic energy loss assessment of the fetal heart using a novel application of “real time”
in TCPC. Heart. 2015;101(2):139–143. cardiac magnetic resonance imaging. Fetal Diagn Ther. 2005;20:475–480.
50. Ashford MW, Liu W, Lin SJ, et al. Occult cardiac contractile dysfunction 64. Prsa M, Sun L, van Amerom J, et al. Reference ranges of blood flow in the
in dystrophin-deficient children revealed by cardiac magnetic resonance major vessels of the normal human fetal circulation at term by
strain imaging. Circulation. 2005;112:2462–2467. phase-contrast magnetic resonance imaging. Circ Cardiovasc Imaging.
51. Harris MA, Weinberg PM, Fogel MA. Cardiac magnetic resonance atrial 2014;7:663–670.
level shunt detection utilizing presaturation tagging. Presented at the 4th 65. Sun L, Macgowan CK, Sled JG, et al. Reduced fetal cerebral oxygen
World Congress of Pediatric Cardiology and Cardiac Surgery; 2005; consumption is associated with smaller brain size in fetuses with
Buenos Aires, Argentina. congenital heart disease. Circulation. 2015;131:1313–1323.
52. Kiaffas MG, Powell AJ, Geva T. Magnetic resonance imaging evaluation of
cardiac tumor characteristics in infants and children. Am J Cardiol.
2002;89:1229–1233.
13 
Human Cardiac Magnetic Resonance at
Ultrahigh Fields: Technical Innovations, Early
Clinical Applications and Opportunities
for Discoveries
Thoralf Niendorf, Till Huelnhagen, Lukas Winter, and Katharina Paul

The development of ultrahigh field magnetic resonance (UHF-MR, B0 technical innovations, practical considerations, safety topics, frontier
≥ 7 T, f ≥ 298 MHz) is moving forward at an amazing speed that is studies in healthy subjects, early clinical application in small patient
breaking through technical barriers almost as fast as they appear. cohorts, opportunities for discoveries, and future directions of UHF-CMR.
UHF-MR has become an engine for innovation in experimental and At the moment some of these new concepts and applications are merely
clinical research.1–11 With nearly 40,000 magnetic resonance (MR) of proof-of-principle nature, but they are compelling enough to drive
examinations already performed, the reasons for moving UHF-MR into the field forward with the goal to advance the capabilities of cardiac
clinical applications are more compelling than ever. The value of high imaging. As they are developed, the boundaries of MR physics, biomedi-
field MR at 3 T has already proven itself many times over at lower field cal engineering, and biomedical sciences will be pushed in many ways,
strengths; now 7 T has opened a window on tissues, organs, and (patho) with the implications and potential uses feeding into basic cardiology
physiologic processes that have been largely inaccessible in the past. research and clinical sciences.
The lion’s share of UHF-MR examinations today covers neuroscience In the sections that follow, encouraging developments into multiple
applications where enabling technology has revealed new aspects of channel RF concepts are reviewed. Advances in imaging methodology
the anatomy, functions, and physiometabolic characteristics of the brain and progress in RF pulse design are discussed. Early applications for
at an unparalleled quality. imaging of cardiac anatomy, cardiac chamber quantification, myocar-
The growing number of reports referring to cardiovascular applica- dial mapping, angiography of the large vessels, and real-time imaging
tions is an inherent testament to the advancements of UHF-MR and of the heart are surveyed. Clinical opportunities for high spatial reso-
documents the progress in imaging the heart and large vessels at 7 T.1–45 lution MR and parametric tissue characterization (all being facilitated
These technical and early feasibility studies in healthy subjects and by the traits of UHF-MR) are also discussed. Physiometabolic CMR
patients respond to unsolved problems and unmet needs of today’s applications are explored, including sodium MR and phosphorus
clinical cardiovascular magnetic resonance (CMR). These developments MR. Practical and safety considerations for cardiac MR in humans
are fueled by the signal-to-noise ratio (SNR) advantage and the promise at 7 T are outlined. Current trends in CMR are considered, together
of enhanced relative spatial resolution (voxel per anatomy), and are with their clinical implications. A concluding section ventures a
enabled by novel MR technology. Transferring UHF-CMR into the glance beyond the horizon and explores an even further step into
clinic remains a challenge though.8 Arguably, the benefits of UHF-MR the future, with something called extreme field magnetic resonance
are sometimes offset by concomitant physics-related phenomena and (EF-MR).
by practical obstacles. These impediments include magnetic field inho-
mogeneities, off-resonance artifacts, dielectric effects, radiofrequency
(RF) nonuniformities, localized tissue heating, and RF power deposition ENABLING TECHNICAL INNOVATIONS
constraints. It is no surprise that these effects can make it a challenge
to even compete with the capabilities of CMR at clinical field strengths
FOR UHF-CMR
of 1.5 T and 3 T.8 If these impediments can be overcome, the promises At ultrahigh fields the crux of the matter is that the RF wavelength in
of UHF-CMR will open new avenues for MR-based myocardial tissue tissue λ becomes sufficiently short (λmyocardium ≈ 12 cm) versus the size
characterization and imaging of the myocardial macromorphology and of the upper torso. The heart, being a deep-lying organ surrounded
micromorphology. The benefits of UHF-MR will catalyze explorations by the lung within the comparatively large volume of the thorax, is
that go beyond conventional 1H MR of the heart. This includes imaging a target region that is particularly susceptible to nonuniformities in
and spectroscopy of 23Na, 19F, 31P, 13C, and other X nuclei to enable a the RF transmission field (B1+). These detrimental transmission field
better insight into inflammatory, metabolic, and (nano)molecular phenomena can cause shading, massive signal drop-off, or even signal
processes of the heart. void in the images, and hence bear the potential to offset the benefits of
Realizing the progress and challenges of UHF-CMR, this chapter UHF-CMR because of nondiagnostic image quality. These constraints are
provides an overview of the state of the art of cardiac MR at 7 T. It not a surprise as it might appear at the first glance, because transmission
discusses the clinical relevance of what has been already observed and field nonuniformities, although somewhat reduced, remain significant in
what can be clearly foreseen. The chapter is devoted to surveying CMR at 3 T.46 Further to the challenges imposed by B1+ nonuniformities,

142
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 143

magnetohydrodynamic effects severely disturb the electrocardiogram made good use of building blocks that include stripline elements,1,2,48–50
(ECG) commonly used for cardiac triggering at clinical field strengths. electrical dipoles,22,41,50–54 dielectric resonant antennas,29 and loop ele-
This challenge evoked the need for novel ancillary hardware that sup- ments,5,11,13,14,18,28 with up to 32 independent elements.
ports synchronization of MR data acquisition with cardiac activity. Loop element-based CMR optimized 7 T transceiver configurations
To address the practical obstacles of UHF-CMR, technical innova- were reported for a 4-channel TX/RX11 (Fig. 13.1A), an 8-channel TX/
tions in RF antenna design have evolved in recent years. Novel pulse RX18 (Fig. 13.1B), and a two-dimensional (2D) 16-channel TX/RX design
sequences for transmission field mapping and shaping as well as inno- (Fig. 13.1C).13 The 2D approach was extended to a more sophisticated
vative RF pulse designs along with multichannel RF transmission were modular 32-channel TX/RX array shown in Fig. 13.1D.28
reported with the common goal to overcome the detrimental B1+ phe- Various stripline element-based 7 T transceiver configurations were
nomena at 7 T to enable cardiac imaging. Novel triggering techniques proposed for cardiac UHF-MR. In a pioneering 8-element transverse
that are immune to electromagnetic fields have been established as an electromagnetic field (TEM) transceiver array design, each element was
alternative to ECG. In this light, this section surveys enabling technical independently connected to a dedicated RF power amplifier.1 Other
innovations tailored for UHF-CMR. stripline array configurations run flexible designs consisting of a pair
of 4 or 8 stripline elements.2,48 Another practical solution comprising
Enabling Radiofrequency Antenna Technology an 8-channel stripline transceive array that uses sophisticated automated
A plethora of reports eloquently refer to the development of enabling tuning with piezoelectric actuators was accomplished.33
RF technology tailored for CMR at 7 T. Research directions include Electric dipole configurations have been shown to benefit MR of
local transceiver (TX/RX) arrays and multichannel transmission arrays the upper torso and the heart at 7 T.22,41,51,53,54 Electric dipoles run the
in conjunction with multichannel local receiver arrays. trait of a linearly polarized current pattern, where RF energy is directed
Multichannel RF coil designs tailored for UHF-CMR involve perpendicular to the dipole along the Poynting vector to the subject,
rigid, flexible, and modular configurations. The gestation process resulting in a symmetrical, rather uniform excitation field with increased
revealed a trend toward a larger number of transmit and receive ele- depth penetration.51 Early implementations include straight dipole ele-
ments5,11,13,14,18,28,41 to improve anatomic coverage and to advance the ments.51 The need for densely packed multichannel transceiver coil arrays
capabilities for transmission field shaping.47 Pioneering developments tailored for UHF-CMR has inspired explorations into electric dipole

A. Four-channel B. Eight-channel C. Sixteen- D. Modular 32- E. Modular 8- F. Modular 16-


TX/RX loop coil TX/RX loop coil channel TX/RX channel TX/RX channel TX/RX bow- channel TX/RX bow-
array array loop coil array loop coil array tie antenna array tie antenna array

FIG. 13.1  Examples of multichannel transceiver arrays tailored for cardiovascular magnetic resonance at
7 T. Top row, Photographs of cardiac optimized 7 T transceiver coil arrays including (left to right) a 4-channel,
an 8-channel, a 16-channel, and a 32-channel loop array configuration together with an 8-channel and 16-channel
bow-tie antenna array. For all configurations, the coil elements are used for transmission and reception.
Four-chamber (middle row) and short-axis (bottom row) views of the heart derived from two-dimensional
CINE fast low angle shot acquisitions using the radiofrequency coil arrays shown on top and a spatial resolu-
tion of (1.4 × 1.4. × 4) mm3. (From Niendorf T, Paul K, Ozerdem C, et al. W(h)ither human cardiac and body
magnetic resonance at ultrahigh fields? Technical advances, practical considerations, applications, and clinical
opportunities. NMR Biomed. 2016;29(9):1173–1197.)
144 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

configurations that comprise 8 or 16 bow-tie antenna building blocks, a practical solution that can be made readily available for a large number
as shown in Fig. 13.1E and F.41 Each of the building blocks contains a of MR sites. In today’s research implementations, RFPAs with up to
bow-tie–shaped λ/2-dipole antenna immersed in D2O.22 This approach 16 kW peak power are used for the single-feeding RFPA mode. State-
helps to shorten the effective antenna length. of-the-art multifeeding RFPA implementations support 8 to 16 RF
Improvements in the compactness of dipole antenna arrays were amplifiers, each providing 1 to 2 kW adjustable RF output power. A
also accomplished by incorporating lumped element inductors, by using multifeeding RFPA setup provides software-driven control over phase
fractionated dipoles, or by employing meander structures.54–57 The and amplitude by modulating the applied input signal. Moreover, it
requirements of UHF-CMR along with ultimate intrinsic SNR and enables sophisticated parallel transmission of independent RF waveforms
current pattern considerations has inspired alternative dipole antenna to every channel. On the journey to a broader range of UHF-CMR
designs. One important development is bent electric dipoles.58 Other applications, it is beneficial if RF coil configurations provide flexibility
configurations include circular dipoles, which consist of a circular con- which supports single-feeding as well as multiple-feeding RFPA regimes
ductor with a feed point on one side and a gap on the other.59 Recently, without major changes in cabling and other components.28 To this end,
proposed dipole antenna variants include folded dipoles60 or combina- Fig. 13.2 shows an example of a 16-channel loop array tailored for
tions of loop elements and dipole configuration.53 The snake antenna UHF-CMR which is connected to a universal interface that supports
building block presents an alternative for the design of transmit arrays the single-feeding and multifeeding RFPA modes. Transition between
tailored for UHF-CMR.61 both modes can be accomplished in 5 seconds without the need for
Ultimate intrinsic signal-to-noise ratio (UISNR) considerations moving the patient or the patient table.
outlined that loop and dipole current patterns contribute equally to
UISNR at 7 T.62,63 For field strengths of B0 ≥ 9.4 T, current patterns are Progress in Pulse Sequence Development
dominated by linear (dipole-type) current patterns,62,63 which provide Progress in UHF-CMR pulse sequence methodology includes two main
motivation for shifting the weight to dipoles versus loop and stripline categories: (1) novel approaches for transmission field mapping and
elements at 7 T and beyond. (2) new technologies for transmission field shaping.
The knowledge of the spatial B1+ distribution generated by RF coil
Multichannel Radiofrequency Transmission arrays is pivotal for UHF-CMR to manage transmission fields across
To translate the capabilities of multichannel RF coil configurations into the heart, including magnetization preparation, signal excitation, and
practical value, advances in MR systems hardware abounded that help signal refocusing. B1+ mapping approaches commonly used are mainly
to mitigate signal variations across the heart and to address blood/ magnitude-based and are generally confined to the ratios or the fit
myocardium contrast heterogeneity. State-of-the-art UHF-MR systems of signal intensity images.64–71 For this purpose, sets of images are
architecture supports multichannel transmit arrays to be driven in two acquired using either two flip angles,65–67 identical flip angles but dif-
modes, using (1) an array of independent-feeding RF power amplifiers ferent repetition times (TR),69 variable flip angles,68,70 or signals from
(RFPA) with exquisite control of phase, amplitude, or even complete spin echoes and stimulated echoes,64 as well as signals from gradient
RF waveforms for all individual channels or (2) a single-feeding RFPA. echoes and stimulated echoes.72 It should be noted that B1+ mapping
For the single-feeding RFPA regime, the amplifier output is commonly is not a need limited to transceiver arrays tailored for 1H UHF-MR but
split into fixed-intensity signals using RF power splitters. Phase adjust- includes heteronuclear MR applications. For instance, B1+ mapping has
ments for individual coil elements or groups of coil elements are accom- been applied to 23Na MRI to support quantification of sodium tissue
plished with phase-shifting networks or coaxial cables. This setup provides content.73

FIG. 13.2  Example of a 16-channel radiofrequency (RF) coil array tailored for cardiovascular magnetic reso-
nance at 7 T which is connected to a universal and magnetic resonance vendor-independent interface that
supports two RF transmission modes using (left) an array of up to 16 independent-feeding RF power ampli-
fiers (RFPAs) with exquisite control of phase, amplitude, or even complete RF waveforms for all individual
channels or (right) a single-feeding RFPA connected to a network of RF splitters and phase shifters used for
transmission field shaping. For the single-feeding RFPA mode, the amplifier output is split into 16 fixed-
intensity signals using RF power splitters. Phase adjustments for individual coil elements (or groups of coil
elements) are accomplished with phase-shifting networks incorporated in the coil interface. Transition between
both transmission modes can be accomplished in 5 seconds without the need for moving the patient or the
patient table. (Courtesy Dr. Helmar Waiczies, MRI.TOOLS GmbH, Berlin, Germany.)
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 145

Phase-based techniques present an alternative for B1+ mapping, and of preparation modules common in cardiac imaging such as inversion,
were reported to be more accurate versus magnitude-based methods fat suppression or blood suppression, pencil beam excitation across the
exploiting the low flip angle regime.74 Phase-based techniques com- diaphragm for navigator-gated respiratory motion compensation, and
monly take advantage of a composite or off-resonance RF pulse to rapid imaging modules. Frontier CMR implementations of dynamic B1+
encode the spatial B1+ information into the phase of the magnetization shimming at 7 T include 2D CINE imaging,85 coronary MR angiography,86
vector. Most phase-based methods, however, make use of nonselective and four-dimensional (4D) flow MR angiography of the entire aorta,
pulses for volume excitation in conjunction with three-dimensional which demonstrated improved excitation efficiency, increased SNR of
(3D) imaging schemes.75–77 This approach results in relatively long myocardium and blood, and enhanced blood/myocardium contrast or
acquisition times that often exceed clinically acceptable breath-hold helped to balance the B1+ uniformity with B1+ efficiency.87
periods, along with a pronounced susceptibility for respiratory motion Multichannel transmission88–91 using spoke RF pulses presents a
artifacts. Realizing these limitations, the feasibility of cardiac-gated, major breakthrough for pulse sequence developments tailored for
single-breath-hold B1+ mapping has been demonstrated78 using a 2D UHF-CMR.24 A juxtaposition of the two-spoke approach with B1+
phase-based Bloch-Siegert approach.79 shimming revealed an enhanced excitation fidelity or a reduced RF
Notwithstanding the advances in B1+-mapping methodology, trans- pulse energy for the two-spoke technique.24 This benefit holds the
mission field mapping of individual coil elements at 7 T has been pri- promise to be instrumental for a broader range of CMR applications,
marily established for head imaging80–82 but remains a challenge for including uniform excitation for parametric mapping used for tissue
UHF-CMR because of depth penetration constraints and susceptibility characterization. The pulse designs used for two-spoke parallel transmis-
artifacts. To cope with this challenge, B1+ profiles are often derived from sion RF pulses make use of a single set of B1+/B0 maps. This approach
numerical electromagnetic field (EMF) simulations.13,28,83 For this purpose, may not be suitable for subsequent scans acquired at another respira-
the transmit phases for each coil element are adjusted using iterative tory phase because of organ displacement, which might induce severe
algorithms to improve transmission field homogeneity in a region of excitation profile and B0 degradation. To address this issue, a tailored
interest encompassing the heart using a human voxel model.84 To ensure parallel transmission RF pulse design which is immune to respiration-
that the relative B1+ distributions of the individual coil elements derived induced B1+/B0 variations has been recently proposed.34,43 Multiband
from the EMF simulations accord with reality, it is essential that EMF spoke pulses afford simultaneous multislice acquisitions with enhanced
simulations are validated against MR measurements using phantoms flip angle homogeneity but minimal increase in integrated and peak
filled with a dielectric liquid that resembles the dielectric properties of RF power.
the target organ/tissue, as outlined in Fig. 13.3.
Based upon a priori acquired B1+ maps derived for all individual Ancillary Hardware Tailored for Ultrahigh
transmit channels of a coil array, B1+ shimming can be used to reduce Frenquency-Magnetic Resonance
RF nonuniformities across the heart using the degrees of freedom of In current clinical MR practice, cardiac motion is commonly dealt
multichannel transmission to shape the transmission field for UHF-CMR. with using electrocardiographic gating/triggering techniques92–94 to
Dynamic shimming is the forte of the multifeeding RFPA’s mode synchronize data acquisition with the cardiac cycle. If brought into a
in conjunction with sophisticated pulse sequence developments, which magnetic field, ECGs, being an electrical measurement, are corrupted
support cascades of dynamic B1+ shimming stages that balance the needs by magnetohydrodynamic (MHD) effects.10,95,96 The MHD effect is

A B B1+ fields C B1+ mapping D B1+ difference


(EMF simulations) (experiment) EMF simulation − experiment
EMF simulation

1 3 1 3
1 3

1 3
2 4 2 4
2 4 2 4

80 40 0 +100% 0% -100%
µT/ ÷kW
FIG. 13.3  Example for transmission field assessment with electromagnetic field simulations and experimental
B1+ mapping using a building block equipped with four loop radiofrequency (RF) elements. For this purpose
a cylindrical phantom setup was used in the simulations and in the experiments. (A) Virtual model of the
setup used for validation (RF shield and casing are not shown but included in the simulations). The experi-
mental setup was arranged analogously to the virtual setup. (B) Simulated B1+ distribution (absolute values)
of four channels which form one curved building block. For B1+ evaluation, transversal slices through the
cylindrical phantom were positioned in alignment with the center of the loop elements. (C) Absolute trans-
mission fields derived from B1+ mapping of the experimental setup. (D) Difference maps in percentage of
the simulated results. The results demonstrate the qualitative and quantitative agreement between the
numerical simulations and the measurements. (From Niendorf T, Paul K, Oezerdem C, et al. W(h)ither human
cardiac and body magnetic resonance at ultrahigh fields? Technical advances, practical considerations, appli-
cations, and clinical opportunities. NMR Biomed. 2016;29(9):1173–1197.)
146 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

pronounced during cardiac phases of systolic aortic flow, which results 2


σE
in a distortion of the S-T segment of the ECG. The susceptibility to SAR = Eq. 13.1
EMF interference manifests itself in ECG waveform distortions already ρ
apparent in ECG traces acquired in clinical 1.5 T MR scanners.97 At
ultrahigh fields the propensity of ECG recordings to MHD effects is  the tissue conductivity σ, the root mean square of the electric field
with
pronounced.2,3,6 Artifacts in the ECG trace and severe T-wave elevation E, and the tissue density ρ. Excessive SAR levels might lead to elevated
might be misinterpreted as R-waves, resulting in misdetection of cardiac temperature levels, making a careful SAR evaluation mandatory to
activity or erroneous cardiac gating together with motion-corrupted confirm compliance with the RF power deposition limits given by the
image quality. Realizing the constraints of conventional ECG, an MR International Electrotechnical Commission (IEC) standard 60601-2-
stethoscope has been proposed (Fig. 13.4) for the pursuit of robust and 33:2010 Ed.399 for safe operation of transmit RF coils tailored for
safe clinical gated/triggered CMR.3,10,98 In contrast to ECG-triggering, UHF-CMR.99 The IEC 60601-2-33:2010 Ed.3 technical standard defines
the MR stethoscope employs acoustic instead of electrical signals. For limits for whole-body average SAR (normal mode: 2 W/kg, first-level
cardiac gating/triggering, the first heart tone of the phonocardiogram, controlled mode: 4 W/kg), which is used for large-volume body RF
which marks the onset of the acoustic cardiac cycle, is selected. The coils commonly used for transmission at 1.5 T and 3 T. RF power depo-
phonocardiogram was reported to be immune to interferences with sition of local transceiver RF coil arrays used for UHF-CMR is limited
electromagnetic fields and to provide reliable and stable trigger infor- by a more restrictive local SAR of 20 W/kg for the trunk in first-level
mation3,10,98 in UHF-CMR, as illustrated in Fig. 13.4. controlled mode. This restriction implies (1) that the applicable RF
power based on local SAR limits is lower than that based on whole-body
SAFETY OF HUMAN CARDIOVASCULAR MAGNETIC or partial-body SAR100,101 and (2) that more power has to be applied
RESONANCE AT 7 T to reach the same flip angle. In addition, electrical conductivity is fre-
quency dependent and increases for UHF-MR. This leads to a direct
Radiofrequency Power Deposition SAR increase as seen from Eq. 13.1 and an indirect SAR increase because
Radiofrequency pulses used in MR deposit RF power in tissue, which of lower absolute B1+ per input power resulting from higher transmis-
can be described by the specific absorption rate (SAR): sion losses in tissue.

Galvanic
MRI
isolation
ECG Trigger
ECG- box input
electrodes
Fiber
MRT
optics

Battery

MR system Galvanic Galvanic


decoupling decoupling

Signal Converter Trigger


conditioning input
Acoustic
sensor Acoustic
wave guide MRT

FIG. 13.4  Block diagram (left), clinical setup (center), and four-chamber views of the heart obtained for (top)
vector electrocardiogram (ECG) gating and (bottom) acoustic cardiac triggering (ACT) used for synchronization
of retrospectively gated two-dimensional gradient echo imaging with the cardiac cycle at 7 T.10 Interference
with magnetohydrodynamic effects cause severe distortion in the vector ECG waveform, resulting in erro-
neous trigger recognition and cardiac motion-induced artifacts (top right). For comparison, ACT is free of
interferences from magnetohydrodynamic effects, provides a reliable trigger signal for ultrahigh frequency
cardiovascular magnetic resonance, and images free of cardiac activity-induced motion artifacts (bottom
right). MR, Magnetic resonance; MRI, magnetic resonance imaging; MRT, magnetic resonance tomograph.
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 147

At UHF-MR, the wavelength  becomes sufficiently short versus the expected to include a drive toward controlled E-field steering and tar-
upper torso, which affects the E field distribution inside the body and geted SAR reduction technologies in electrically conductive implants,
offsets some of the SAR predictions governed by102: catheters, and guide wires using parallel transmission to create excita-
tions that induce minimal RF current in elongated conductors.108,109
B12B02τ RF An open-minded look reveals that UHF-CMR parallel transmission
SAR ∝ Eq. 13.2
mTRi
technology perhaps forms an enabling platform to advance interven-
tional CMR applications at 7 T by using an MR antenna array to delib-
RF power deposition is substantially affected by the geometry of erately focus RF power and increase the temperature locally in a controlled
the upper torso, by positioning of the RF coil with respect to the torso way, denoted as thermal magnetic resonance.22,110 In this context, potential
and the heart, by the RF coil design, and by the number of RF trans- applications could include (1) targeted RF-guided drug release and
mission channels, as well as by the RF driving conditions. Here, EMF image-guided monitoring,111 (2) localized contrast agent release and
simulations are essential to provide an insight into the local SAR dis- tracking, or (3) stem cell delivery to the myocardium afforded by local
tribution. Recent simulation studies showed that using the merits of RF heating. In this context new developments of MR thermometry
high RF frequencies for multichannel RF antenna array design, the techniques and their validation will be crucial to MR safety and to
relative SAR increase is even well below a SAR ~ B0 dependency up to advance the field of thermal therapies and thermal MR.
UHF-MR frequencies as high as 1 GHz (23.5 T) (Fig. 13.5).103
Although SAR is key for RF safety evaluations, temperature Radiofrequency-Induced Heating of Passive
distribution—which is the cause of tissue damage based on RF heating— Conductive Devices
does not follow SAR in a straightforward manner.100 Thermoregulation En route to broader clinical UHF-CMR studies, it is essential to gain
and heat transfer inside the body (i.e., blood vessels) need to be con- a better insight into the interaction of passive conducting implants
sidered while moving toward a more realistic scenario. Ongoing research with RF fields. Considering an ever-increasing population of patients
focuses already on temperature modeling instead of SAR models, sug- with a history of stent implantation, detailing the interaction of stents
gesting a thermal tissue damage threshold CEM43 known from thermal with RF fields is of profound relevance for the advancement of
therapies.104,105 The thermal dose concept will be included in edition 4 UHF-CMR. This is not an easy task because of the many RF coil and
of the IEC 60601-2-33 safety guidelines, which is expected to be final- stent configurations available, yet becomes even more relevant when
ized by 2018. Here, realistic thermal modeling based on electromagnetic moving to many element transceiver arrays or even to large-volume
field simulations and MR thermometry is essential to assess thermal coils covering major sections of the body.
distributions in vivo.106,107 These explorations will help to advance toward At UHF-MR the RF wavelength (λ) in myocardial tissue and blood
enhanced MR safety assessment for UHF-CMR. This development is is sufficiently short (λ approximately 12 cm) to induce resonance effects
at λ/4 to λ/2112 in coronary stents with a length of up to approximately
4 cm, a dimension common in clinical practice.113 EMF simulations
Relative SAR increase vs. 300 MHz and RF heating experiments showed that temperature increase due to
coronary stents does not exceed baseline RF heating if IEC standards
10
for local and global SAR limits are strictly obeyed.16 Although being
very important, these early studies did not employ a setup that resembles
8
8ch (00) a clinical scenario.16 The need for RF heating assessment at B0 ≥ 7 T
also prompted valuable research into RF-induced heating of metallic
6 8ch (CP)
(a.u.)

arterial stents.114 The conclusions drawn are valuable but constrained


maxCh (00) to the very specific experimental setup used.
4
maxCh (CP) Recognizing the opportunities for broader clinical UHF-CMR studies,
2 RF-induced heating of coronary stents was examined at 7 T.39 For this
SAR ~ B02
purpose, EMF simulations were performed for a broad range of coronary
SAR ~ B0 stent configurations to detail electric fields and local SAR as a function
0
300 400 500 600 700 800 900 1000 of stent diameter, stent length, stent position with respect to the RF
f (MHz)
transmission source, and stent orientation versus the E-field.39 To trans-
FIG. 13.5  Frequency-dependent relative specific absorption rate (SAR) late these results to arbitrary coronary stent geometries, arbitrary stent
increase covering frequencies ranging from 300 MHz (7 T) to 1 GHz
positions, and arbitrary RF coil configurations, a transfer function for
(23.5 T). For the electromagnetic field simulations, multichannel dipole
antenna arrays were positioned around a cylindrical phantom (diameter
the assessment of EM fields induced in coronary stents was proposed.39
= 180 mm). Discrete radiofrequencies (RF) (f = 300 MHz to 1 GHz) have SAR1g induced by a stent (SAR1g stent) can be described by:
been evaluated for two transmit settings: 00 (0 degree phase shift
between transmit channels) and CP (phase shift between transmit chan- max(SAR1g stent ) = f (diameter, length, orientation) ⋅ SAR1g baseline
nels: 360 degrees/number of elements). At higher RF the number of
Eq. 13.3
transmit channels can be increased from an 8-channel (8ch) to a 19-channel
configuration (maxCh). This improvement in element density increases with SAR1g baseline being the local SAR1g at the assumed stent position
transmit efficiency, lowers SAR, and reduces the relative SAR increase without the presence of a stent.39 Fig. 13.6 shows an example of the
vs. 300 MHz. Taking advantage of high-density multichannel RF antenna dependence of induced stent SAR1g on stent length and stent orientation
arrays, the relative SAR increase (1) is well below the SAR ~ B02 rela-
versus the E-field vector.
tionship described by the literature for low magnetic field strengths and
(2) falls even below a SAR ~ B0 dependency up to ultrahigh frequency
The transfer function provides a fast way to estimate induced stent
magnetic resonance frequencies as high as 1 GHz (23.5 T). (From Winter SAR levels. EMF simulations performed with a stent integrated in a
L, Niendorf T. Electrodynamics and radiofrequency antenna concepts human voxel model prove to be very time consuming. This is a particular
for human magnetic resonance at 23.5 T (1 GHz) and beyond. MAGMA. practical obstacle if many-element RF TX/RX arrays and a set of phase
2016;29(3):641–656.) settings mimicking B1+ shimming or other parallel transmit applications
148 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A B
Baseline SAR simulations without implant (SAR1g(w/o)) Transfer function
[W/kg]
10

<1 s
C D
SAR simulations with implant (SAR1g(/w)) SAR validation
25
[W/kg]
10 20

SAR1g [W/kg]
15
5
10

0 5
>2 h
0
0 1 2 3 4 5 6 7 8 9
Stent Phase setting
Baseline SAR SAR estimation
SAR simulation
with stent

FIG. 13.6  Electromagnetic field (EMF) simulations using the human voxel model “Duke” from the virtual
family and an 8-channel transmit/receive bow-tie electric dipole array. (A) Schematic views of the positioning
of the anterior bow-tie radiofrequency antennas on the anterior chest of the human voxel model and coronal
view of specific absorption rate (SAR)1g baseline distribution for a plane through the target position without the
stent equivalent being present. (B) Surface plot of SAR1g stent derived from the transfer function for baseline
SAR1g for an input power of 1 W/kg. A stent length ranging from 10 to 40 mm and a stent rotation versus
the main E-field vector ranging from 0 to 90 degrees was applied. (C) Coronal view of SAR1g stent distribution
for a plane through the target position with the stent equivalent being present. (D) Simulated maximum
baseline SAR1g baseline and SAR1g stent for the stent equivalent using eight randomly generated phase settings
compared with the SAR estimation deduced from the analytical approach (see Eq. 13.3). Although conser-
vatively overestimating SAR, SAR estimation using Eq. 13.3 was able to predict the induced SAR1g levels
without the need to perform extra time-consuming EMF simulations with a stent being present in the simula-
tion model. (From Niendorf T, Paul K, Oezerdem C, et al. W(h)ither human cardiac and body magnetic reso-
nance at ultrahigh fields? Technical advances, practical considerations, applications, and clinical opportunities.
NMR Biomed. 2016;29(9):1173–1197.)

are included. Benchmarking the analytical approach versus EMF simula- can be conveniently incorporated into state-of-the art SAR prediction
tions showed a good conservative estimation of induced SAR1g peak concepts116,117 to provide SAR estimations induced by coronary stents
levels. A practical example is shown in Fig. 13.6 using an 8-channel and other conductive implants for arbitrary RF pulses used for trans-
bow-tie antenna RF coil array optimized for CMR at 7 T (see Fig. mission field shimming24,118 or parallel transmission.89,90 To generalize,
13.1E). EMF simulations including the stent consumed more than 2 this approach provides a novel metric or design criteria for RF coils:
hours central processing unit (CPU) and graphics processing unit (GPU) for example, to design RF coils (1) with low SAR levels in the vicinity
time per phase setting (not including RF shim field combining and of the stent or (2) with SAR levels during the presence of the stent
SAR calculations) while the analytical approach can be calculated in which do not exceed SAR levels without the presence of the stent.39
real time. This example underscores the value of the proposed analytical The transfer function approach should not end at SAR estimation, but
approach based upon a transfer function for obeying regulatory RF should move toward temperature or CEM43 estimations. In particular,
power limits.99 The approach of using a transfer function is conceptu- for short implants like coronary stents, maximum induced SAR levels
ally translatable to other implant configurations, which is in particular in the implant are much more restrictive to the allowed input power
suitable for short implants (≤λtissue/3) where the influence of the phase than maximum temperature or CEM43 values would be. It was shown
distribution of the RF transmit field is negligible.115 Furthermore, it that even though maximum induced local SAR at the stent tip was by
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 149

a factor >2 higher than in surface regions, the eventual temperature


after 6 minutes of RF heating was by >6°C lower at the stent tip region.39 Cardiac Chamber Quantification
This temperature saturation effect at the stent tip originates from a Early studies demonstrated the feasibility and usefulness of 2D CINE
strong temperature gradient toward colder tissue regions at the stent spoiled gradient echo imaging (fast low angle shot [FLASH]) for cardiac
center and is even more pronounced by physiologic parameters such chamber quantification of the left ventricle (LV)6,9,12 and right ventricle
as blood flow in the stented vessel. These findings are heartening; they (RV)20 at 7 T. 2D CINE FLASH provides high-quality images with
will accelerate further research on safety assessment of coronary stents uniform signal intensity distribution and high blood/myocardium con-
and other implants common in clinical practice and will help to relax trast over the entire heart, as demonstrated for a 32-channel loop element
current UHF-CMR limitations. RF coil array and for a 16-channel bow-tie antenna array in Fig. 13.8.
Fig. 13.9 shows long-axis and short-axis views of an exemplary volunteer
and underlines the signal uniformity and superb blood-to-myocardium
EARLY APPLICATIONS AND CLINICAL STUDIES contrast. The latter is competitive with the blood-to-myocardium con-
This section surveys proof-of-principle and feasibility studies of trast obtained for 2D CINE steady-state free precession (SSFP) imaging
UHF-CMR in healthy human subjects and in small patient cohorts. at 1.5 T.6,9,12 Imaging the right ventricle at 7 T using fast gradient echo
Early UHF-CMR applications include anatomic imaging using black- (FGRE) provided RV dimensions and function comparable with SSFP
blood techniques, cardiac chamber quantification employing 2D CINE imaging at 1.5 T (Fig. 13.10).20 Cardiac chamber quantification at 7 T
techniques, first-pass myocardial perfusion imaging, parametric mapping agrees closely with LV and RV parameters derived at 1.5 T.9
of the effective transversal relaxation time T2* and the longitudinal The baseline SNR gain at 7 T can be translated into spatial resolu-
relaxation time T1, fat-water imaging, coronary artery angiography, tion enhancements versus the kindred counterparts at lower field
vascular imaging, and real-time imaging of the heart. strengths. Fig. 13.11 surveys four-chamber and short-axis views of the
heart obtained with a standardized CMR protocol119,121 and with an
Black-Blood Imaging enhanced spatial resolution protocol. This protocol reduced the voxel
Transfer of rapid acquisition with relaxation enhancement imaging size from 19.4 mm3 to 1.6 mm3, equivalent to a 12-fold improvement
(RARE) (also known as fast spin-echo [FSE] imaging)-based black-blood in the spatial resolution versus a standardized clinical CMR protocol.
imaging to 7 T is of high relevance for advancing the capabilities of This fidelity approaches an effective anatomical spatial resolution—voxel
UHF-CMR for explorations into cardiac morphology. The feasibility size per anatomy—which resembles that demonstrated for animal
of cardiac RARE imaging at 7 T using an 8-channel transceiver array models.122,123 The overall image quality and improvement in spatial
of bow-tie antennas is demonstrated in Fig. 13.7. A spatial resolution resolution enabled the visualization of fine subtle anatomic structures,
as good as (0.8 × 0.8 × 5) mm3 was applied, which represents an order including the compact layer of the free RV wall and the remaining
of magnitude improvement versus standardized protocols of today’s trabecular layer. Pericardium, mitral and tricuspid valves—and their
clinical CMR practice.119 The blood suppression inherent to RARE vari- associated papillary muscles and trabeculae—are identifiable.
ants works well in most regions of the heart. Only slow-flowing blood To put the spatial resolution enhancements to clinical use, a
in areas close to the endocardium remains visible. The refinement of UHF-CMR study was performed in a small cohort of patients with
black-blood preparation modules—including double-inversion recov- hypertrophic cardiomyopathy (HCM)40 (Fig. 13.12). Cardiac chamber
ery preparation—tailored for 7 T is anticipated to further improve the quantification of the left ventricle at 7 T was in accordance with the
image quality of RARE at 7 T. Also, RARE imaging at 7 T presents an results derived from the 3 T LV assessment of the same HCM patient
RF power deposition challenge because of the train of high-peak RF group as well as from the age and body mass index (BMI)-matched
power refocusing pulses (α ≤ 180 degrees). Notwithstanding its utility healthy control group. Thanks to the spatial resolution enhancement
for improving B1+ uniformity in UHF-RARE imaging, recent studies on at 7 T, hyperintense regions were detected in 54% of the HCM patients
using adiabatic pulses for UHF-RARE reported long inter-echo times of within late gadolinium enhancement (LGE) positive regions and were
up to 15 ms120; an approach that does not meet the requirements of CMR identified as myocardial crypts (Fig. 13.13). These crypts were not
taking advantage of breath-hold scans to deal with respiratory motion. detectable at 3 T using a clinical 2D CINE SSFP protocol.40

FIG. 13.7  Examples of high spatial resolution black-blood imaging of end-diastolic short-axis views of the
heart at 7 T using a rapid acquisition with relaxation enhancment imaging technique. The images exhibit a
spatial resolution of (left) (1.2 × 1.2 × 5) mm3, (center) (0.9 × 0.9 × 4) mm3, and (right) (0.8 × 0.8 × 5) mm3,
which is by a factor of 2.7 (left) or 6 (center, right) superior versus the clinical protocols used in today’s
cardiovascular magnetic resonance practice.
150 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

32-channel modular TX/RX


loop array
16-channel modular TX/RX
bow-tie antenna array

FIG. 13.8  End-diastolic short-axis views covering the heart from the apex to the base derived from two-
dimensional CINE fast low angle shot imaging (in-plane resolution 1.1 × 1.1 mm2, slice thickness 2.5 mm,
generalized autocalibrating partial parallel acquisitions reduction factor 2), using (top) a 32-channel modular
transceiver (TX/RX) loop array and (bottom) a 16-channel modular TX/RX bow-tie antenna array. Both array
configurations provided rather uniform signal intensity and no major signal voids for slices covering the heart
from the apex to the base. (From Niendorf T, Paul K, Oezerdem C, et al. W(h)ither human cardiac and body
magnetic resonance at ultrahigh fields? Technical advances, practical considerations, applications, and clinical
opportunities. NMR Biomed. 2016;29(9):1173–1197.)

A B C D
FIG. 13.9  Two-dimensional CINE fast low angle shot images acquired with a 16-channel transceiver (TX/
RX) coil array (see Fig. 13.1C) with an in-plane resolution of 1 × 1 mm2 (slice thickness 4 mm) using 2-fold
acceleration showing a four-chamber view (A), a three-chamber view (B), a two-chamber view (C), and a
mid-ventricular short-axis view of the heart (D). (From Thalhammer C, Renz W, Winter L, et al. Two-dimensional
sixteen channel transmit/receive coil array for cardiac magnetic resonance imaging at 7 T: design, evaluation,
and application. J Magn Reson Imaging. 2012;36(4):847–857.)
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 151

FIG. 13.10  Four-chamber view (top), right ventricle long-axis view (middle), and mid-ventricular transverse
view (bottom) obtained (left) by steady-state free precession at 1.5 T with voxel size 1.2 × 1.2 × 6 mm3 (left),
(center) by fast gradient echo (FGRE) at 7 T with voxel size 1.2 × 1.2 × 6 mm3 and (right) by FGRE at 7 T
with voxel size 1.3 × 1.3 × 4 mm3. (From von Knobelsdorff-Brenkenhoff F, Tkachenko V, Winter L, et al.
Assessment of the right ventricle with cardiovascular magnetic resonance at 7 tesla. J Cardiovasc Magn
Reson. 2013;15:23.)

agents. By making use of the blood-oxygen-level-dependent (BOLD)


First-Pass Myocardial Perfusion Imaging effect,125 T2*-sensitized CMR has been proposed to probe changes in
Clinical CMR assessment of ischemic heart disease includes rapid first- tissue oxygenation and perfusion, and has demonstrated the capability
pass contrast agent enhanced myocardial perfusion CMR.119 Numerous to reveal myocardial ischemia and perfusion deficits.126–130 In clinical
saturation recovery-based perfusion techniques have been proposed to routine, T2* mapping has become the gold standard for myocardial
capture first-pass kinetics with one or two heartbeat temporal resolu- iron assessment, an important parameter for diagnosis and therapy
tions. Saturation methods established at lower fields are suboptimal guiding in patients with myocardial iron overload.131
for myocardial perfusion imaging at 7 T32 because of their propensity The linear relationship between magnetic field strength and micro-
for transmission field nonuniformities. To address this issue a novel scopic susceptibility effects renders the move to B0 = 7 T conceptually
saturation pulse train consisting of four hyperbolic-secant (HS8) RF appealing for T2* mapping.25,132–134 The enhanced susceptibility effects
pulses was proposed for effective saturation of myocardium at 7 T.32 at 7 T may be useful to lower the detection level and to extend the
This approach facilitated an average saturation efficiency of 97.8% in dynamic range of the sensitivity for monitoring T2* changes. To avoid
native myocardium and afforded the first series of human first-pass T2* quantification errors because of signal modulations induced by
myocardial perfusion images at 7 T.32 The ability to produce exquisite fat-water phase shift, echo times when fat and water are in phase are
in-plane spatial resolution at 7 T may offer greater diagnostic value for commonly used for T2* mapping.135 Because of the reduction of the
myocardial perfusion assessment and supports an extension of the in-phase inter-echo time from 4.8 ms at 1.5 T to 1.02 ms at 7 T, UHF-MR
perfusion assessment to the right ventricle. With sufficient acceleration enables rapid acquisition of multiple echoes.17 Taking advantage of
and imaging speed, perfusion imaging is on the verge of 3D whole-heart these properties, high spatial resolution and cardiac phase resolved T2*
coverage acquisitions124 that might be furthered by the traits of UHF-MR. mapping has been demonstrated in the in vivo human heart at 7 T.17
In contrast, studying temporal changes in T2* across the cardiac cycle
Myocardial T2* Mapping at 1.5 T and 3 T is elusive because of scan time constraints which are
A growing number of reports refer to mapping the effective transverse prohibitive for CINE T2* mapping covering the cardiac cycle. The
relaxation time T2* in basic CMR research and clinical CMR applica- improved temporal resolution of T2* mapping at 7 T provides means
tions. Myocardial T2* mapping provides means for noninvasive myo- for monitoring externally controlled variations of blood oxygenation,
cardial tissue characterization without the need for exogenous contrast including short periods of hypoxia and hyperoxia test stimuli.136,137
Four-chamber view
Zoomed view
Short-axis view

A B C D
FIG. 13.11  Four-chamber views (first row) and short-axis views (third row) of the heart derived from two-
dimensional CINE fast low angle shot acquisitions, generalized autocalibrating partial parallel acquisition
reduction factor 2 using (A, B) a 32-channel modular transceiver (TX/RX) loop element array (see Fig. 13.1B
and D) and (C, D) a 16-channel modular TX/RX bow-tie antenna array (see Fig. 13.1C and F). Different resolu-
tions were employed: (A, C) standardized clinical protocol: spatial resolution 1.8 × 1.8 × 6 mm3, (B) spatial
resolution 1.1 × 1.1 × 2.5 mm3 and (D) spatial resolution 0.8 × 0.8 × 2.5 mm3. Second row, Magnified views
of the ventricular trabeculae, demonstrating that spatial resolution enhancements by a factor of 6 or even
12 versus standardized protocols used in current clinical practice improve the delineation of subtle anatomical
details of the heart. (From Niendorf T, Paul K, Oezerdem C, et al. W(h)ither human cardiac and body magnetic
resonance at ultrahigh fields? Technical advances, practical considerations, applications, and clinical oppor-
tunities. NMR Biomed. 2016;29(9):1173–1197.)

FIG. 13.12  Feasibility of cardiovascular magnetic resonance (CMR) in hypertrophic cardiomyopathy (HCM)
patients at 7 T. High-resolution mid-ventricular short-axis CINE images obtained for nine HCM patients enrolled
in an ultrahigh field CMR study demonstrating consistency in image quality. All images showed clinical diag-
nostic quality and were evaluable for cardiac chamber quantification. Spatial resolution is 1.4 × 1.4 × 4 mm3.
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 153

FIG. 13.13  Hypertrophic cardiomyopathy patient showing myocardial crypts detected with two-dimensional
CINE fast low angle shot imaging at 7 T using a spatial resolution of 1.4 × 1.4 × 4 mm3. CINE images of a
short-axis view (left) and a long-axis view (right) exhibit hyperintense signal in areas that were identified as
myocardial crypts (arrows). Fibrosis detected via late gadolinium enhancement at 3 T (not shown) and crypts
do overlap.40

25
Anatomical

Mean septal T2* [ms]


images

Mean septal intravoxel gradient [Hz]


20 Mean septal ∆T2* [ms]

400 15

ms / Hz / ms
Off-center

200
frequency

0 Hz 10
-200
-400 5
100
80
Intravoxel

0
gradient

60
Hz
40
20 −5
0 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8
Relative cardiac phase
A B
FIG. 13.14  Assessment of B0 field and intravoxel gradient over the cardiac cycle derived from a short-axis
view of the heart of a healthy subject obtained at 7 T. (A) Top, Anatomic magnitude images. Middle, Low
pass filtered off-center frequency maps. Bottom, Intravoxel field gradient maps. (B) Plot of mean T2* (blue)
and mean intravoxel B0 gradient (black) in the intraventricular septum over the cardiac cycle for a cohort of
healthy volunteers. An increase of T2* in systole can be observed. The estimated T2* shift caused by the
B0 variation assuming a common myocardial T2* value at 7 T of 15 ms is shown in red. Relative cardiac
phase 0 indicates the beginning of systole. Macroscopic myocardial B0 gradient variations over the cardiac
cycle can be considered minor regarding their effects on T2*. Error bars indicate SD. (From Huelnhagen T,
Hezel F, Serradas Duarte T, et al. Myocardial effective transverse relaxation time T2* correlates with left
ventricular wall thickness: a 7 T MRI study. Magn Reson Med. 2017;77(6):2381–2389.)

For T2* mapping, a reasonable B0 uniformity across the heart B0 gradient in the interventricular septum over the cardiac cycle was
is essential such that susceptibility weighting is not dominated by approximately 0.4 ± 0.3 Hz/voxel. Regarding the mean observed septal
macroscopic B0 field inhomogeneities but rather by microscopic B0 T2* and intravoxel B0 gradient over the cardiac cycle, the observed
susceptibility gradients.17 Because of increased susceptibility effects, maximum intravoxel B0 gradient variation translates into a T2*variation
this is of particular concern for UHF-CMR, but can be managed by of approximately 0.7 ± 0.4 ms. With this B0 uniformity across the heart
dedicated B0 shimming. Using local second-order shimming, a mean and across the cardiac cycle, high spatial resolution and cardiac phase-
peak-to-peak B0 difference of approximately 65 Hz was found across resolved myocardial T2* mapping at 7 T is feasible,17,25 as demonstrated
the left ventricle at 7 T,17 which compares well with 3 T138 and 1.5 T in Fig. 13.15.45
studies.139 For myocardial anterior, anterolateral, and inferoseptal seg- For T2* mapping at 7 T, the longest mean T2* values were found for
ments, a mean in-plane B0 gradient of approximately 3 Hz/mm was the anterior (T2* = 17.3 ms), anteroseptal (T2* = 16.8 ms), and infero-
observed. For CINE T2* mapping also, temporal changes of the mac- septal (T2* = 16.3 ms) segments. Septal segments showed the lowest
roscopic B0 field across the cardiac cycle, induced by changes in cardiac spatial variation in T2*, which is similar to that reported for 1.5 T and
macromorphology and the displacement of the diaphragm, need to be 3 T.25 The inferior (T2* = 12 ms) and inferior lateral (T2* = 11.4 ms)
considered. Using cardiac-gated temporally resolved B0 field mapping, wall yielded lowest T2* values with the spatial variation being signifi-
it was demonstrated that such changes can be considered minor in cantly pronounced versus 1.5 T and 3 T.25 The 7 T setup used facilitated
the ventricular septum with respect to their influence on T2* even at a spatial resolution as good as 1.1 × 1.1 × 2.5 mm3, which helped to
7 T (Fig. 13.14).45 The mean absolute variation of the mean intravoxel reduce intravoxel dephasing along the slice direction.17
154 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TE = 2.04 ms TE = 4.08 ms TE = 6.12 ms TE = 8.16 ms TE = 10.2 ms


50

40

30

T2* in ms
20

10
t = 53 ms t = 197 ms t = 345 ms t = 493 ms t = 638 ms
0

TE = 2.04 ms TE = 4.08 ms TE = 6.12 ms TE = 8.16 ms TE = 10.2 ms


50

40

30

T2* in ms
20

10
t = 18 ms t = 165 ms t = 348 ms t = 495 ms t = 640 ms
0
FIG. 13.15  Example of cardiac phase-resolved myocardial T2* mapping at 7 T. First row, T2*-weighted
short-axis views of the heart for a mid-ventricular slice derived from multiecho gradient echo imaging at 7 T.
Images were obtained at the echo times shown. A spatially adaptive nonlocal means denoising filter was
applied.177 Second row, Temporally resolved myocardial T2* maps of a short-axis view of the same volunteer
derived from a series of T2*-sensitized two-dimensional (2D) CINE acquisitions covering the entire cardiac
cycle (five cardiac phases out of a series of 20 cardiac phases are shown) overlaid onto conventional 2D
CINE fast low angle shot images are displayed. Third row, T2*-weighted short-axis views and temporally
resolved myocardial T2* maps of the heart for a mid-ventricular slice of a patient suffering from hypertrophic
cardiomyopathy. An increase in septal T2* compared with healthy volunteers can be appreciated.

Myocardial T2* changes are commonly regarded to provide a sur- T2* across the cardiac cycle using CINE T2* mapping revealed cyclic
rogate for myocardial tissue oxygenation,140 yet the factors influencing changes in septal T2* with a mean end-systolic T2* increase of about
T2* are of multiple nature.141 Further to blood oxygenation, blood 10% compared with end diastole (Fig. 13.16).45 The periodic changes
volume fraction per tissue volume, hematocrit, the oxyhemoglobin dis- in interventricular septal myocardial T2* correlate well with changes
sociation curve, main magnetic field inhomogeneities, tissue susceptibility, in the septal wall thickness and with alterations in the LV radius through-
and tissue microstructure or micromorphology were reported to govern out the cardiac cycle (see Fig. 13.16). The observed cyclic T2* variations
T2*.142–147 Cardiac macromorphology, including ventricular radius and were suggested to be dominated by changes in myocardial blood volume
ventricular wall thickness, constitutes another category of physiologic fraction rather than oxygenation, indicating that temporally resolved
parameters that are substantially altered throughout the cardiac cycle. MR relaxation mapping could be beneficial for understanding cardiac
These alterations might affect blood volume fraction and the amount (patho)physiology in vivo.45
of deoxygenated hemoglobin (deoxyHb) per tissue volume, resulting The recent progress in in vivo histology and in fiber orientation
in dynamic variation in myocardial T2* over the cardiac cycle. Detailing tracking using T2*-weighted MR143,144,148,149 suggests that the linear
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 155

25 1

20 0.8

Normalized count
mm / ms / mm
15 0.6

10 0.4

5 0.2
Mean septal inner radius (mm)
Mean septal T2* (ms) End systole
Mean septal wall thickness (mm) End diastole
0 0
0.2 0.4 0.6 0.8 1 0 10 20 30
A Relative cardiac phase B T2* [ms]

FIG. 13.16  Relationship of septal myocardial wall thickness, T2* and left ventricular (LV) inner radius in
healthy volunteers at 7 T. (A) Time course of mean septal wall thickness, mean LV inner radius, and mean
septal T2* plotted over the cardiac cycle averaged for 10 volunteers. Error bars indicate standard deviation.
(B) Cumulative frequency plot of ventricular septal T2* in end systole and end diastole for all septal voxels
of all volunteers. A clear shift to higher T2* in systole can be recognized. (From Huelnhagen T, Hezel F,
Serradas Duarte T, et al. Myocardial effective transverse relaxation time T2* correlates with left ventricular
wall thickness: a 7 T MRI study. Magn Reson Med. 2017;77(6):2381–2389.)

T2* distribution in the mid-ventricular septum is qualitative and subjective though, which spurs advancements toward
myocardial T1 mapping.153–155 At 7 T increased RF transmit power and
0.2
improved B0 shimming play complementary roles for myocardial T1
mapping which requires careful transmission field shaping and B0 shim-
Normalized count

0.15 Volunteers
ming. This requirement can be managed if quantification of and cor-
Patients
rection for imperfect inversion is applied.23 For this purpose an adiabatic
0.1
inversion pulse tailored for use in the heart in conjunction with a
0.05 shortened modified Look-Locker inversion recovery (ShMOLLI) variant
was employed.23 Subject-averaged inversion efficiencies ranging from
0 −0.79 to −0.83 (perfect inversion would provide −1) were accomplished
0 10 20 30 40 50 across myocardial segments.23 A T1 of normal myocardium has been
T2* in ms
reported to be 1925 ± 48 ms at 7 T23 versus 1166 ± 60 ms at 3 T156 and
721 ± 37 ms at 1.5 T.155 Another method to address the challenge of
FIG. 13.17  Histogram showing the distribution of mid-ventricular septal
T2* in healthy volunteers and patients suffering from hypertrophic car-
imperfect inversion at UHF is to use saturation recovery methods instead
diomyopathy (HCM). Data were derived from myocardial T2* mapping of inversion recovery. Employing a saturation recovery single-shot
at 7 T. A clear shift toward higher T2* can be observed for HCM patients. acquisition (SASHA) technique with a saturation pulse train and satu-
The data were obtained and grouped for all cardiac phases for six HCM ration delays optimized for 7 T, T1 values comparable with an inversion
patients (red line) and 10 age-matched and body-mass-index–matched recovery approach were obtained.157 A T1 of 1939 ± 73 ms was reported
healthy volunteers (black line). in the intraventricular septum, employing a simple three-parameter fit
model without the need for correction of imperfect inversion as required
for ShMOLLI.
relationship between T2* and B0 might provide means to gain a better The prolonged T1 relaxation times at 7 T provide an enhanced blood/
insight into the myocardial microstructure. Because the susceptibil- myocardium contrast superior to 1.5 T which afforded a revival of
ity effects depend on the orientation of blood-filled capillaries and spoiled gradient echo imaging techniques at 7 T. Slice selective variable
microstructural arrangements with respect to the external magnetic flip angle techniques provide an alternative for rapid parametric T1
field,125,139,144,147,150 T2* mapping at 7 T might contribute to explorations mapping.158 This approach obviates the need for inversion recovery
into visualization of myocardial fibers, into the examination of helical preparation while being fast enough to meet the speed requirements
angulation of myocardial fibers, or into the assessment of fibrotic tissue of cardiac MR.
and other kinds of microstructural tissue changes. Results of the first
cardiac patient study at 7 T investigating T2* in patients suffering from Fat-Water Imaging
HCM demonstrated an increase of septal T2* compared with healthy Fat-water separated cardiac imaging provides a sensitive means of detect-
controls152 (Figs. 13.15 and 13.17). Mean septal T2* averaged over all ing intramyocardial fat, characterizing fibro-fatty infiltration, character-
subjects for all cardiac phases was found to be T2*mean = (17. 5 ± 1.4) ms izing fatty tumors, and delineating epicardial and/or pericardial fat.
for HCM patients and T2*mean = (13.7 ± 1.1) ms for BMI-matched and The concept of fatty myocardium has also received attention because
age-matched healthy subjects. These findings provide encouragement of its role in cardiomyopathy where the mismatch between myocardial
that T2* mapping at UHF could provide an imaging-based biomarker fatty acid uptake and utilization leads to the accumulation of cardiotoxic
that could support diagnosis and risk stratification in cardiomyopathies. lipid species.159 Multiecho Dixon approaches using iterative decomposi-
tion have been shown to provide robust fat-water separation even in
Myocardial T1 Mapping the presence of large field inhomogeneities.160 Equipped with this tech-
Myocardial tissue characterization using T1-weighted LGE imaging is nology, the feasibility of fat-water–separated cardiac imaging at 7 T has
of proven clinical value for the assessment of ischemic heart diseases been demonstrated,161 including its use for fat suppression in coronary
and also for nonischemic myocardial diseases.119 T1-weighted imaging artery imaging (Fig. 13.18). Yet, the Dixon approach constitutes a
156 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

FIG. 13.18  Examples of early ultrahigh field cardiovascular magnetic resonance (UHF-CMR) application in
healthy subjects: Left, center, Fat-water–separated (left, water image; center, fat image) coronary artery
imaging showing the left main coronary artery using the Dixon approach at 7 T. Right, High-resolution image
of the right coronary artery acquired with a spatial resolution of 0.45 × 0.45 × 1.2 mm3, which was facilitated
by a navigator-gated free breathing three-dimensional gradient echo acquisition in conjunction with spectrally
selective adiabatic inversion recovery at 7 T. (Courtesy Maurice Bizino and Hildo Lamb, Department of Radiol-
ogy, Leiden University Medical Center, Leiden, The Netherlands.)

challenge for quantification of fat infiltration in myocardial disease. autocalibrating partial parallel acquisition (GRAPPA) acceleration com-
Chemical shift-resolved MR spectroscopy and imaging techniques provide bined with dynamically applied B1+ shimming, toggling a setting tailored
a valuable alternative for fat quantification in the myocardium. for the navigator used for respiratory gating and the imaging module.44
Fig. 13.19 shows the blood velocity by path lines for different time
Coronary Artery Imaging points of the cardiac cycle within the human aorta derived from a 4D
Coronary artery imaging (CAI) remains one of the most technically flow dataset acquired at 7 T with 1.2-mm isotropic spatial and 40.8 ms
challenging applications in cardiovascular MRI because of small vessel temporal resolution.
size, vessel tortuosity, and physiologic motion.162,163 UHF-CAI using Perhaps UHF-MR forms another important enabling factor to
noncontrast gradient echo imaging techniques suggested that image transform the baseline SNR advantage into improved spatiotemporal
quality already approached that achieved for CAI at 3 T.4,7,26 Coronary resolution of contrast-enhanced MRA. To this end, an SNR gain for
vessel sharpness at 7 T was found to be improved versus 3 T.7 The early contrast-enhanced versus noncontrast-enhanced 4D flow MRA was
studies focused primarily on the right coronary. To afford appropriate reported to be approximately 1.4 at 7 T, which was found to be in
coverage of all main coronary arteries, the capabilities of dynamic B1+ accordance with 3 T and 1.5 T observations.35
shimming were exploited.86 The capabilities of UHF-CAI were furthered
by employing high spatial resolution CAI (see Fig. 13.18).26 For this Real-Time Imaging
approach, coronary vessel edge sharpness was found to be superior Breath-held 2D CINE acquisitions segmented over regular 10 to 16
to the border sharpness accomplished in state-of-the-art CAI at 3 T. heartbeats are the clinical standard for LV morphology and function
Visible vessel length and vessel diameter obtained at 7 T were competi- assessment. However, this approach is limited by physiologic (e.g., cardiac
tive with 3 T.26 arrhythmias) constraints and the ability of the patients to hold their
Traditional frequency selective fat saturation techniques used in CAI breath, which might cause inappropriate diagnostic information. Free-
might suffer from large static field variations at 7 T, which may cause breathing real-time CMR achieves diagnostic quality in a single heart-
nonuniform and imperfect fat suppression over the target region, an beat164 and bears the potential to change the landscape of cardiac
effect which bears the risk of obscuring the delineation of coronary diagnostics.165–167 The SNR gain achieved by ultrahigh magnetic fields
arteries. Here fat-water–separated imaging holds the promise of sub- can help to counteract the loss of signal because of the undersampling
stituting conventional preparatory fat saturation techniques (see Fig. needed for real-time acquisitions.
13.18). This approach also promises to offset some of the RF power The accelerated imaging capabilities and the anatomic coverage of
deposition constraints that come with conventional saturation recovery- multichannel bow-tie antenna arrays (see Fig. 13.1E and F) supported
based fat saturation that makes use of large flip angles. free-breathing real-time imaging of the heart and of the aorta at 7 T
(Fig. 13.20).41 The spatial resolution of 1.2 × 1.2 × 6 mm3 and the frame
Vascular Imaging rate of 30 frames per second fully meet if not excel the requirements
MR angiography (MRA) of the large vessels is a common CMR applica- of standardized LV structure and function assessment protocols used
tion. MRA stands to benefit from UHF-MR. The SNR gain can be used in today’s CMR practice.119 The real-time FLASH images of the aorta
to counter the noise amplification caused by acceleration techniques demonstrate the extended anatomic coverage of the 16-channel bow-tie
employed to meet the high spatiotemporal resolution requirements of antenna array along the head-feet direction, including details of the
MRA. Potential applications include large-volume coverage, time-resolved liver and the spine without B1+ signal voids (see Fig. 13.20).41
phase velocity MRA (4D flow), which is an emerging technique for
studying the flow pattern, or wall shear stress in large vessels. The fea-
OPPORTUNITIES FOR DISCOVERIES
sibility of aortic 4D flow at 7 T was recently demonstrated.35 For this
purpose, a prospectively gated 4D flow sequence was applied. Another Exploiting the benefits of ultrahigh magnetic field strengths, X-nuclei
4D flow MRA implementation at 7 T made good use of kt-generalized imaging of the heart at 7 T moved into the spotlight of interest. In
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 157

t = 104 ms 133 ms 163 ms

t = 232 ms 306 ms 405 ms

1.00

Velocity
0.75

(m/s)
0.50
0.25
0.00
FIG. 13.19  Example of velocity data derived from four-dimensional flow measurements at 7 T using 1.2 mm
isotropic spatial and 40.8 ms temporal resolution. Left, Pathline visualization of the blood velocity within the
human aorta for different time points within the cardiac cycle relative to the R-wave of the electrocardiogram
signal. Right, Enlarged view of time point 306 ms. The high spatial resolution allows for depicting the coronary
artery in the magnitude image (arrow). (Courtesy Sebastian Schmitter, Center for Magnetic Resonance
Research, University of Minnesota Medical School, Minneapolis, MN.)

FIG. 13.20  Examples for free-breathing real-time imaging at 7 T. Top, Mid-ventricular short-axis view of the
heart (left, systole; right, diastole) obtained with an 8-channel bow-tie antenna array (see Fig. 13.1E). Bottom,
Coronal view of the aorta (left, systole; right, diastole) acquired with a 16-channel bow-tie antenna array (see
Fig. 13.1F) demonstrating the 35 cm anatomic coverage of the 16-channel bow-tie antenna array along the
head-feet direction, including details of the liver and the spine without transmission field–induced signal voids.
Images were acquired at a rate of 30 frames per second using highly undersampled radial two-dimensional
fast low angle shot with nonlinear inverse reconstruction at a spatial resolution of 1.2 × 1.2 × 6 mm3.
158 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

particular, sodium and phosphorus are two nuclei that promise to con- deviation for the creatine phosphate/adenosine triphosphate (PCr/ATP)
tribute to the understanding of physio-metabolic processes. This section ratio were observed. This gain resulted in an enhanced quantification
describes pioneering work in the fields of 23Na and 31P cardiac imaging accuracy at 7 T. Myocardial 31P T1 relaxation times are shorter at 7 T
and spectroscopy at 7 T. versus 3 T. These improvements could permit scan time reductions
versus 3 T and might eventually allow metabolic probing of dynamic
Sodium MRI processes. For all these reasons it was concluded that 7 T will become
Sodium MRI (23Na MRI) is an emerging approach for gaining better the field strength of choice for cardiac 31P MR spectroscopy tailored
insights into cellular metabolism, with a broad spectrum of biomedical for probing myocardial energetics.27 This manifests itself in a first 7 T
imaging research applications.168 Previous studies eloquently reported 31
P MR spectroscopy study performed in a cohort of 25 patients with
credible data on 23Na MRI of the heart and demonstrated that 23Na dilated cardiomyopathy (DCM),42 where the PCr/ATP ratio was found
MRI is suitable for the detection and assessment of acute and chronic to be significantly lower than that of healthy control subjects (see Fig.
heart disease.169 The rapidly decaying 23Na signal and the low sensitivity 13.22). These results are a precursor to a broader clinical study aiming
of 23Na MRI versus 1H MR constitute a challenge for clinical 23Na CMR. at the assessment of the effects of energy-sparing drugs in patients with
Once clinically feasible, sodium CMR holds the promise to become a DCM.42
valuable diagnostic tool for classifying viable from nonviable tissue in
ischemic infarct patients. Recognizing the sensitivity gain and yet unim-
paired transmission field homogeneity19 because of the comparably low
LOOKING AT THE HORIZON
23
Na resonance frequency, which is close to 1H MRI at 1.5 T, it is con- Where do we stand? Although the lion’s share of UHF-MR examinations
ceptually appealing to pursue 23Na CMR at 7 T at clinically acceptable today cover brain and neuroscience applications, cardiac imaging is
acquisition times. To approach this goal, a local four-element transceiver another field that can benefit from UHF-MR. The eye-opening quality
RF surface coil customized for 23Na CMR at 7 T was employed37 to of recent anatomic, functional, and physiometabolic UHF-CMR insights
derive 23Na images of the heart (Fig. 13.21) with a resolution of 6 × 6 into the heart have served as a driving force for application develop-
× 6 mm3 using a density-adapted 3D radial acquisition technique.170 ments, which culminated in the breadth and detail outlined in this
These efforts also demonstrated that the sensitivity gain at 7 T enables chapter. UHF-CMR helps to eliminate the main barriers standing in
CINE 23Na imaging of the beating heart using a temporal resolution the way at low fields. Groundbreaking examples include the capability
of 100 ms supported by retrospectively gated, density-adapted 3D pro- to detect subtle myocardial crypts which cannot be seen at lower fields,
jection reconstruction.37,38 as well as unprecedented capacity and speed for real-time imaging.
UHF-CMR opens new avenues into myocardial tissue characteriza-
Phosphorus Magnetic Resonance tion and will yield new insights by permitting phosphorus, sodium,
Phosphorus MR affords in vivo insights into the energy metabolism and potassium MRI, which will reveal new dimensions of metabolism,
and is an ideal candidate to benefit from the sensitivity gain and improved bioenergetics, and tissue function of the heart and allow us to build
frequency dispersion at higher fields. To demonstrate the sensitivity many more bridges to molecular discovery. The pace of discovery is
gain, careful juxtaposition between cardiac 31P MRS at 7 T and 3 T was heartening and a powerful motivator to transfer the lessons learned
conducted.27 These pioneering studies revealed marked superiority of from UHF-CMR research into broader clinical studies. These efforts are
cardiac 31P spectra at 7 T relative to 3 T (Fig. 13.22). SNR improvements fueled by the quest for advancing the capabilities of cardiac imaging,
of 2.8 for creatine phosphate (PCr)27 together with a reduced standard with the implications feeding into MR physics, biomedical engineering,

23
Na MR 1
H MR

FIG. 13.21  Example of 23Na magnetic resonance (MR) imaging of the heart at 7 T. Left, Sodium image of
a four-chamber view of the heart acquired at 7 T. The 23Na image of the heart was acquired using a density-
adapted three-dimensional radial technique139 with TE = 0.4 ms; TR = 11 ms; TRO = 7.1 ms; TX amplitude
115 V (~90% specific absorption rate), equivalent to a tip angle of 30 to 40 degrees; number of projections
= 50,000; number of averages = 2; and a voxel size of 6 × 6 × 6 mm3. Center, Corresponding 1H four-chamber
view of the heart derived from two-dimensional CINE gradient echo imaging at 7 T. Right, Overlay of the
sodium image (color scale) to the anatomical image (gray scale). (Courtesy Dr. Helmar Waiczies, MRI.TOOLS
GmbH, Berlin, Germany.)
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 159

PCr
PCr 3 T 40 3T
7T 7T 20

γ-ATP α-ATP 30 15

Signal/baseline SD
γ-ATP
Signal/baseline SD

2,3-DPG α-ATP 10
2,3-DPG 20

5
β-ATP 10 PDE
PDE 0
10 0
5 -5

0 0

10 5 0 -5 -10 -15 -20 10 5 0 -5 -10


Chemical shift δ (ppm) Chemical shift δ (ppm)

FIG. 13.22  Example of phosphorus spectroscopy in healthy subjects and patients with dilated cardiomyopathy
at 7 T. Top, Comparison of single-voxel 31P magnetic resonance (MR) spectra of the human heart obtained
at 3 T (blue) and at 7 T (red) for a healthy volunteer (left) and for a patient with dilated cardiomyopathy (right).
The voxel used for 31P MR spectroscopy was placed in the middle of the interventricular septum, as illustrated
in the short-axis views (bottom) of the heart obtained from a two-dimensional CINE fast low angle shot
localizer scan at 7 T. 31P MR spectroscopy at 7 T provided a signal-to-noise ratio advantage over 31P MR
spectroscopy at 3 T. (Courtesy Christopher Rodgers, Radcliffe Department of Medicine, University of Oxford,
Oxford, UK.)

cardiology, internal medicine, and other related fields of basic research and body coil transmission can be translated into a reduction in the TR.
clinical science. This 1/TR enhancement is conceptually appealing for the reduction
How can clinical CMR at lower fields benefit from the progress in of banding and off-resonance artifacts frequently encountered for 2D
UHF-CMR? It is safe to state that the benefits of 7 T CMR innovations CINE SSFP imaging at 3 T. TR shortening also benefits rapid imaging
will be more seen at 3 T, where the suboptimal copy-and-paste approach that supports real-time assessment of the heart rather than relying
to protocol migration from 1.5 T is being supplanted by the sort of on the interpolation of data acquired during multiple synchronized
application-targeted redesign which is essential for UHF-CMR. For heartbeats commonly used in today’s segmented CINE acquisitions.
example, whereas most of the effort on transmit-receive array structures What will the foreseeable future bring? It is no surprise that the
for CMR is currently occurring at 7 T, recognition of the benefits of these future of UHF-CMR will not end at 7 T and that the field is moving
structures may result in an eventual migration to 3 T ( λmyocardium
3T
≈ 30 cm), apace to even higher fields. Physicists, engineers, and pioneers from
where RF inhomogeneities and SAR limitations, although of less extent, related disciplines have already taken a further step into the future, in
remain significant in clinical CMR.46,171,172 This is a powerful motivator to their minds, with something they are calling extreme field MR (EF-MR).174
elucidate the performance of local multichannel surface RF coils versus This envisions human MR at 20 T103,110,174–176 and it is an important
large-volume body RF coil transmission for CMR at 3 T. A recent study leap of the imagination because it aims to fill a crucial “resolution gap”
demonstrated that CMR and cardiac chamber quantification at 3 T using in our understanding of human biology. Although discoveries are pouring
local surface RF coil transmission is competitive when benchmarked in on the molecular and cellular level every day, it is extremely difficult
against the today’s 3 T CMR practice of large-volume body RF coil to integrate these findings into a coherent picture of the functions of
transmission.173 CMR of patients equipped with passively conducting tissues and pathologic processes at a mesoscopic level above that of the
implants that constitute an SAR governed contraindication for cardiac cell. There is a wide gap between the view of biologists and clinicians
MR in case of body RF coil transmission presents an application where that is begging to be filled. Extreme field MR is probably an ideal tech-
local surface RF coil transmission provides a vital alternative over RF nology that will reach between these levels in vivo by bridging a crucial
body coil excitation. Another example is interventional CMR employing gap in resolution in space and time. Arguably, 1H CMR at 20 T might
imaging-guided navigation of conducting catheters. It was also shown be challenging in the first run, because of electrodynamic constraints.
that the B1+ efficiency benefits of a local transmission coil array versus Yet, the frequencies of X-nuclei at 20 T are below the 1H resonance
160 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

frequency at 7 T, with the exception of 31P and 19F. This makes technolo-
ACKNOWLEDGMENTS
gies established for 1H MR at 7 T ideal candidates to be perfected and
fine-tuned for heteronuclear CMR at 20 T. For example, the sensitivity The authors wish to acknowledge the members of the Berlin Ultrahigh
gain at 20 T is expected to reduce scan times for 31P and 23Na by a factor Field Facility (BUFF), Berlin, Germany; the working group for Cardiac
of 8 versus today’s 7 T capabilities. This promises 23Na MR with a sub- Magnetic Resonance, Charité, Berlin, Germany; Maurice M. Bizino and
millimeter spatial resolution in 5- to 10-minute scan time and offers Hildo Lamb, Department of Radiology, Leiden University Medical Center,
the potential for probing the myocardial energetics with 31P spectroscopy Leiden, the Netherlands; Thea Marie Niendorf, Aachen, Germany and
MRS in clinically acceptable scan times. Anna Tabea Niendorf, Aachen, Germany; Christopher Rodgers, Radcliffe
Do we need extra resources and a willingness to invest? Yes. Explor- Department of Medicine, University of Oxford, Oxford, UK; Sebastian
ing new territory requires extra resources—and the will to go there. Schmitter, University of Minneapolis, Minnesota, USA; and Helmar
Obviously for a while the most high-end instruments will be costly and Waiczies, MRI.TOOLS GmbH, Berlin, Germany who kindly contributed
only accessible to a few; ideally through National Research Infrastructure examples of their pioneering work or other valuable assistance. This
Facilities. That cannot be an argument, however, against making the work was supported (in part, TN) by the DZHK (German Centre for
leap forward. Although the first 20 T class MR instruments will prob- Cardiovascular Research) and by the BMBF (Federal Ministry of Educa-
ably be devoted to discovery and to proof of principle, the findings will tion and Research). This work was funded (in part, TN) by the Helmholtz
be crucial guides to making the best use of lower-resolution imaging Alliance ICEMED—Imaging and Curing Environmental Metabolic
techniques at 3 T and 7 T. The only thing that could keep the dream Diseases, through the Initiative and Network Fund of the Helmholtz
of human MR at 20 T from becoming reality would be a failure to Association (ICEMED-Project 1210251). LW received support from
follow the path and see what develops. Will the clinic eventually be able the BMBF (Federal Ministry of Education and Research, “KMU-
to follow us to even higher fields? It always does, if a whole community innovativ”: Medizintechnik MED-373-046).
of experts devotes their creative efforts to the task. With this in mind
UHF-CMR remains in a state of creative flux and productive engage-
ment in this area continues to drive further developments for the sake
REFERENCES
of cardiac imaging. A full reference list is available online at ExpertConsult.com
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 160.e1

REFERENCES 22. Winter L, Özerdem C, Hoffmann W, et al. Design and evaluation of a


hybrid radiofrequency applicator for magnetic resonance imaging and
1. Vaughan JT, Snyder CJ, DelaBarre LJ, et al. Whole-body imaging at 7T: RF induced hyperthermia: electromagnetic field simulations up to 14.0
preliminary results. Magn Reson Med. 2009;61(1):244–248. tesla and proof-of-concept at 7.0 tesla. PLoS One. 2013;8(4):e61661.
2. Snyder CJ, DelaBarre L, Metzger GJ, et al. Initial results of cardiac 23. Rodgers CT, Piechnik SK, Delabarre LJ, et al. Inversion recovery at 7 T
imaging at 7 tesla. Magn Reson Med. 2009;61(3):517–524. in the human myocardium: measurement of T(1), inversion efficiency
3. Frauenrath T, Hezel F, Heinrichs U, et al. Feasibility of cardiac gating and B(1) (+). Magn Reson Med. 2013;70(4):1038–1046.
free of interference with electro-magnetic fields at 1.5 tesla, 3.0 tesla and 24. Schmitter S, DelaBarre L, Wu X, et al. Cardiac imaging at 7 tesla:
7.0 tesla using an MR-stethoscope. Invest Radiol. 2009;44(9):539–547. single- and two-spoke radiofrequency pulse design with 16-channel
4. van Elderen SG, Versluis MJ, Webb AG, et al. Initial results on in vivo parallel excitation. Magn Reson Med. 2013;70(5):1210–1219.
human coronary MR angiography at 7 T. Magn Reson Med. 2009; 25. Meloni A, Hezel F, Positano V, et al. Detailing magnetic field strength
62(6):1379–1384. dependence and segmental artifact distribution of myocardial effective
5. Versluis MJ, Tsekos N, Smith NB, Webb AG. Simple RF design for transverse relaxation rate at 1.5, 3.0, and 7.0 T. Magn Reson Med.
human functional and morphological cardiac imaging at 7 tesla. J Magn 2014;71(6):2224–2230.
Reson. 2009;200(1):161–166. 26. Bizino MB, Bonetti C, van der Geest RJ, et al. High spatial resolution
6. Brandts A, Westenberg JJ, Versluis MJ, et al. Quantitative assessment of coronary magnetic resonance angiography at 7 T: comparison with low
left ventricular function in humans at 7 T. Magn Reson Med. 2010; spatial resolution bright blood imaging. Invest Radiol. 2014;49(5):
64(5):1471–1477. 326–330.
7. van Elderen SG, Versluis MJ, Westenberg JJ, et al. Right coronary MR 27. Rodgers CT, Clarke WT, Snyder C, et al. Human cardiac 31P magnetic
angiography at 7 T: a direct quantitative and qualitative comparison resonance spectroscopy at 7 tesla. Magn Reson Med. 2014;72(2):304–315.
with 3 T in young healthy volunteers. Radiology. 2010;257(1):254–259. 28. Graessl A, Renz W, Hezel F, et al. Modular 32-channel transceiver coil
8. Niendorf T, Sodickson DK, Krombach GA, Schulz-Menger J. Toward array for cardiac MRI at 7.0T. Magn Reson Med. 2014;72(1):276–290.
cardiovascular MRI at 7 T: clinical needs, technical solutions and 29. Aussenhofer SA, Webb AG. An eight-channel transmit/receive array of
research promises. Eur Radiol. 2010;20(12):2806–2816. TE01 mode high permittivity ceramic resonators for human imaging at
9. von Knobelsdorff-Brenkenhoff F, Frauenrath T, Prothmann M, et al. 7T. J Magn Reson. 2014;243:122–129.
Cardiac chamber quantification using magnetic resonance imaging at 7 30. Schmitter S, Wu X, Auerbach EJ, et al. Seven-tesla time-of-flight
tesla—a pilot study. Eur Radiol. 2010;20(12):2844–2852. angiography using a 16-channel parallel transmit system with
10. Frauenrath T, Hezel F, Renz W, et al. Acoustic cardiac triggering: a power-constrained 3-dimensional spoke radiofrequency pulse design.
practical solution for synchronization and gating of cardiovascular Invest Radiol. 2014;49(5):314–325.
magnetic resonance at 7 tesla. J Cardiovasc Magn Reson. 2010;12(1):67. 31. Schmitter S, Wu X, Adriany G, et al. Cerebral TOF angiography at 7T:
11. Dieringer MA, Renz W, Lindel T, et al. Design and application of a impact of B1+ shimming with a 16-channel transceiver array. Magn
four-channel transmit/receive surface coil for functional cardiac imaging Reson Med. 2014;71(3):966–977.
at 7T. J Magn Reson Imaging. 2011;33(3):736–741. 32. Tao Y, Hess AT, Keith GA, et al. Optimized saturation pulse train for
12. Suttie JJ, Delabarre L, Pitcher A, et al. 7 tesla (T) human cardiovascular human first-pass myocardial perfusion imaging at 7T. Magn Reson Med.
magnetic resonance imaging using FLASH and SSFP to assess cardiac 2015;73(4):1450–1456.
function: validation against 1.5 T and 3 T. NMR Biomed. 2012;25(1): 33. Keith GA, Rodgers CT, Hess AT, et al. Automated tuning of an
27–34. eight-channel cardiac transceive array at 7 tesla using piezoelectric
13. Thalhammer C, Renz W, Winter L, et al. Two-dimensional sixteen actuators. Magn Reson Med. 2015;73(6):2390–2397.
channel transmit/receive coil array for cardiac MRI at 7.0 T: design, 34. Schmitter S, Wu X, Uğurbil K, Van de Moortele P-F. Design of parallel
evaluation, and application. J Magn Reson Imaging. 2012;36(4): transmission radiofrequency pulses robust against respiration in cardiac
847–857. MRI at 7 tesla. Magn Reson Med. 2015;74(5):1291–1305.
14. Winter L, Kellman P, Renz W, et al. Comparison of three multichannel 35. Hess AT, Bissell MM, Ntusi NAB, et al. Aortic 4D flow: quantification of
transmit/receive radiofrequency coil configurations for anatomic and signal-to-noise ratio as a function of field strength and contrast
functional cardiac MRI at 7.0T: implications for clinical imaging. Eur enhancement for 1.5T, 3T, and 7T. Magn Reson Med. 2015;73(5):
Radiol. 2012;22(10):2211–2220. 1864–1871.
15. Frauenrath T, Fuchs K, Dieringer MA, et al. Detailing the use of 36. Klix S, Els A, Paul K, et al. On the subjective acceptance during
magnetohydrodynamic effects for synchronization of MRI with the cardiovascular magnetic resonance imaging at 7.0 tesla. PLoS One.
cardiac cycle: a feasibility study. J Magn Reson Imaging. 2012;36(2): 2015;10(1):e0117095.
364–372. 37. Graessl A, Ruehle A, Waiczies H, et al. Sodium MRI of the human heart
16. Santoro D, Winter L, Muller A, et al. Detailing radio frequency heating at 7.0 T: preliminary results. NMR Biomed. 2015;28(8):967–975.
induced by coronary stents: a 7.0 tesla magnetic resonance study. PLoS 38. Resetar A, Hoffmann SH, Graessl A, et al. Retrospectively-gated CINE
One. 2012;7(11):e49963. (23)Na imaging of the heart at 7.0 tesla using density-adapted 3D
17. Hezel F, Thalhammer C, Waiczies S, et al. High spatial resolution and projection reconstruction. Magn Reson Imaging. 2015;33(9):1091–
temporally resolved T2* mapping of normal human myocardium at 7.0 1097.
tesla: an ultrahigh field magnetic resonance feasibility study. PLoS One. 39. Winter L, Oberacker E, Özerdem C, et al. On the RF heating of coronary
2012;7(12):e52324. stents at 7.0 Tesla MRI. Magn Reson Med. 2015;74(4):999–1010.
18. Graessl A, Winter L, Thalhammer C, et al. Design, evaluation and 40. Prothmann M, von Knobelsdorff-Brenkenhoff F, Topper A, et al. High
application of an eight channel transmit/receive coil array for cardiac spatial resolution cardiovascular magnetic resonance at 7.0 tesla in
MRI at 7.0 T. Eur J Radiol. 2013;82(5):752–759. patients with hypertrophic cardiomyopathy—first experiences: lesson
19. Niendorf T, Graessl A, Thalhammer C, et al. Progress and promises of learned from 7.0 tesla. PLoS One. 2016;11(2):e0148066.
human cardiac magnetic resonance at ultrahigh fields: a physics 41. Oezerdem C, Winter L, Graessl A, et al. 16-channel bow tie antenna
perspective. J Magn Reson. 2013;229:208–222. transceiver array for cardiac MR at 7.0 tesla. Magn Reson Med.
20. von Knobelsdorff-Brenkenhoff F, Tkachenko V, Winter L, et al. 2016;75(6):2553–2565.
Assessment of the right ventricle with cardiovascular magnetic 42. Stoll VM, Clarke WT, Levelt E, et al. Dilated cardiomyopathy:
resonance at 7 tesla. J Cardiovasc Magn Reson. 2013;15:23. phosphorus 31 MR spectroscopy at 7 T. Radiology. 2016;281:409.
21. Niendorf T, Schulz-Menger J. [Cardiovascular ultrahigh field magnetic 43. Schmitter S, Moeller S, Wu X, et al. Simultaneous multislice imaging in
resonance imaging : challenges, technical solutions and opportunities]. dynamic cardiac MRI at 7T using parallel transmission. Magn Reson
Radiologe. 2013;53(5):422–428. Med. 2017;77(3):1010–1020.
160.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

44. Schmitter S, Schnell S, Uğurbil K, et al. Towards high-resolution 4D flow 67. Stollberger R, Wach P. Imaging of the active B1 field in vivo. Magn Reson
MRI in the human aorta using kt-GRAPPA and B1+ shimming at 7T. J Med. 1996;35(2):246–251.
Magn Reson Imaging. 2016;44(2):486–499. 68. Klose U. Mapping of the radio frequency magnetic field with a MR
45. Huelnhagen T, Hezel F, Serradas Duarte T, et al. Myocardial effective snapshot FLASH technique. Med Phys. 1992;19(4):1099–1104.
transverse relaxation time T2* correlates with left ventricular wall 69. Yarnykh VL. Actual flip-angle imaging in the pulsed steady state: a
thickness: a 7.0 T MRI study. Magn Reson Med. 2017;77(6):2381–2389. method for rapid three-dimensional mapping of the transmitted
46. Mueller A, Kouwenhoven M, Naehle CP, et al. Dual-source radiofrequency field. Magn Reson Med. 2007;57(1):192–200.
radiofrequency transmission with patient-adaptive local radiofrequency 70. Dowell NG, Tofts PS. Fast, accurate, and precise mapping of the RF field
shimming for 3.0-T cardiac MR imaging: initial experience. Radiology. in vivo using the 180 degrees signal null. Magn Reson Med.
2012;263(1):77–85. 2007;58(3):622–630.
47. Adriany G, Ritter J, Vaughan JT, et al. Experimental verification of 71. Stanisz GJ, Odrobina EE, Pun J, et al. T1, T2 relaxation and magnetization
enhanced B1 shim performance with a Z-encoding RF coil array at 7 transfer in tissue at 3T. Magn Reson Med. 2005;54(3):507–512.
tesla. Proc Int Soc Magn Reson Med. 2010. Stockholm, Sweden. 3831. 72. Nehrke K, Bornert P. DREAM–a novel approach for robust, ultrafast,
48. Maderwald S, Orzada S, Schaefer LC, et al. 7T human in vivo cardiac multislice B(1) mapping. Magn Reson Med. 2012;68(5):1517–1526.
imaging with an 8-channel transmit/receive array. Proc Int Soc Magn 73. Linz P, Santoro D, Renz W, et al. Skin sodium measured with (23) Na
Reson Med. 2009. Honolulu, Hawaii. 822. MRI at 7.0 T. NMR Biomed. 2015;28(1):54–62.
49. van den Bergen B, Klomp DW, Raaijmakers AJ, et al. Uniform prostate 74. Morrell GR, Schabel MC. An analysis of the accuracy of magnetic
imaging and spectroscopy at 7 T: comparison between a microstrip resonance flip angle measurement methods. Phys Med Biol. 2010;55(20):
array and an endorectal coil. NMR Biomed. 2011;24(4):358–365. 6157–6174.
50. Ipek O, Raaijmakers AJ, Klomp DW, et al. Characterization of transceive 75. Morrell GR. A phase-sensitive method of flip angle mapping. Magn
surface element designs for 7 tesla magnetic resonance imaging of the Reson Med. 2008;60(4):889–894.
prostate: radiative antenna and microstrip. Phys Med Biol. 2012;57(2): 76. Mugler JPI, Miller GW, Altes TA, et al. A low-flip-angle, phase-based
343–355. method for accurately calibrating the transmitter in hyperpolarized-gas
51. Raaijmakers AJ, Ipek O, Klomp DW, et al. Design of a radiative surface MRI. Proc Int Soc Magn Reson Med. 2005;789.
coil array element at 7 T: the single-side adapted dipole antenna. Magn 77. Santoro D, Rivoire J, Meise F, et al. Three-dimensional mapping of the
Reson Med. 2011;66(5):1488–1497. B1 field using an optimized phase-based method: application to
52. Ipek O, Raaijmakers AJ, Lagendijk JJ, et al. Intersubject local SAR hyperpolarized 3He in lungs. Magn Reson Med. 2011;65(4):1166–1172.
variation for 7T prostate MR imaging with an eight-channel single-side 78. Santoro D, von Samson-Himmelstjerna F, Carinci F, et al. Cardiac
adapted dipole antenna array. Magn Reson Med. 2014;71(4):1559–1567. Triggered B1+ mapping using Bloch Siegert in the heart at 3T. Proc Int
53. Erturk MA, Raaijmakers AJ, Adriany G, et al. A 16-channel combined Soc Magn Reson Med. 2012;3840.
loop-dipole transceiver array for 7 tesla body MRI. Magn Reson Med. 79. Sacolick LI, Wiesinger F, Hancu I, Vogel MW. B1 mapping by Bloch-
2017;77(2):884–894. Siegert shift. Magn Reson Med. 2010;63(5):1315–1322.
54. Raaijmakers AJE, Italiaander M, Voogt IJ, et al. The fractionated dipole 80. Curtis AT, Gilbert KM, Klassen LM, et al. Slice-by-slice B1+ shimming at
antenna: a new antenna for body imaging at 7 tesla. Magn Reson Med. 7 T. Magn Reson Med. 2012;68(4):1109–1116.
2016;75(3):1366–1374. 81. Carinci F, Santoro D, von Samson-Himmelstjerna F, et al.
55. Raaijmakers AJE, El Aidi H, Versluis M, et al. Comprehensive coronary Characterization of phase-based methods used for transmission field
artery imaging at 7.0 T: proof of feasibility. Proc Int Soc Magn Reson uniformity mapping: a magnetic resonance study at 3.0 T and 7.0 T.
Med. 2014;4923. PLoS One. 2013;8(3):e57982.
56. Raaijmakers AJE, Voogt I, Klomp DWJ, et al. Dipole antenna without 82. Nehrke K, Versluis MJ, Webb A, Bornert P. Volumetric B1 (+) mapping
ceramic substrate and still low SAR: the fractionated dipole antenna. of the brain at 7T using DREAM. Magn Reson Med. 2014;71(1):246–256.
Proc Int Soc Magn Reson Med. 2013;4382. 83. Bitz A, Orzada S, Kraff O, et al. Comparison of simulation-based and
57. Chen G, Cloos M, Sodickson DK, Wiggins GC. A 7T 8 channel measurement-based RF shimming for whole-body MRI at 7 tesla. Proc
transmit-receive dipole array for head imaging: dipole element and coil Int Soc Magn Reson Med. 2010;4720.
evaluation. Proc Int Soc Magn Reson Med. 2014;621. 84. Christ A, Kainz W, Hahn EG, et al. The virtual family—development of
58. Chen G, Cloos M, Lattanzi R, et al. Bent electric dipoles: a novel coil surface-based anatomical models of two adults and two children for
design inspired by the ideal current pattern for central SNR at 7 tesla. dosimetric simulations. Phys Med Biol. 2010;55:N23.
ISMRM Proc Int Soc Magn Reson Med. 2014;402. 85. Lindel T, Greiser A, Waxmann P, et al. Cardiac CINE MRI at 7 T using a
59. Lakshmanan K, Cloos M, Lattanzi R, et al. The circular dipole. Proc Int transmit array. Proc Int Soc Magn Reson Med. 2012;1227.
Soc Magn Reson Med. 2014;315. 86. Metzger G, Bi X, DelaBarre L, et al. Left coronary artery imaging at 7T:
60. Lee W, Sodickson DK, Wiggins GC. A new UHF transceiver antenna initial results using multiple B1+ shimming algorithms and targets. Proc
design: modified folded dipole. Proc Int Soc Magn Reson Med. 2013; Int Soc Magn Reson Med. 2011;116.
4367. 87. Setsompop K, Wald LL, Alagappan V, et al. Magnitude least squares
61. Steensma B, Obando Andrade A, Klomp D, et al. Body imaging at 7 tesla optimization for parallel radio frequency excitation design demonstrated
with much lower SAR levels: an introduction of the snake antenna array. at 7 tesla with eight channels. Magn Reson Med. 2008;59(4):908–915.
Proc Int Soc Magn Reson Med. 2016;0395. 88. Katscher U, Bornert P. Parallel RF transmission in MRI. NMR Biomed.
62. Lattanzi R, Sodickson DK. Ideal current patterns yielding optimal 2006;19(3):393–400.
signal-to-noise ratio and specific absorption rate in magnetic resonance 89. Katscher U, Bornert P, Leussler C, van den Brink JS. Transmit SENSE.
imaging: computational methods and physical insights. Magn Reson Magn Reson Med. 2003;49(1):144–150.
Med. 2012;68(1):286–304. 90. Zhu Y. Parallel excitation with an array of transmit coils. Magn Reson
63. Wiggins GJ, Zhang B, Lattanzi R, et al. The electric dipole array: an Med. 2004;51(4):775–784.
attempt to match the ideal current pattern for central SNR at 7 tesla. 91. Ullmann P, Junge S, Wick M, et al. Experimental analysis of parallel
Proc Int Soc Magn Reson Med. 2012;541. excitation using dedicated coil setups and simultaneous RF transmission
64. Akoka S, Franconi F, Seguin F, Le Pape A. Radiofrequency map of an on multiple channels. Magn Reson Med. 2005;54(4):994–1001.
NMR coil by imaging. Magn Reson Imaging. 1993;11(3):437–441. 92. Lanzer P, Barta C, Botvinick EH, et al. ECG-synchronized cardiac MR
65. Cunningham CH, Pauly JM, Nayak KS. Saturated double-angle method imaging: method and evaluation. Radiology. 1985;155(3):681–686.
for rapid B1+ mapping. Magn Reson Med. 2006;55(6):1326–1333. 93. Fischer SE, Wickline SA, Lorenz CH. Novel real-time R-wave detection
66. Insko EK, Bolinger L. Mapping of the radiofrequency field. J Magn algorithm based on the vectorcardiogram for accurate gated magnetic
Reson A. 1993;103:83–85. resonance acquisitions. Magn Reson Med. 1999;42(2):361–370.
CHAPTER 13  Human Cardiac Magnetic Resonance at Ultrahigh Fields 160.e3

94. Chia JM, Fischer SE, Wickline SA, Lorenz CH. Performance of QRS 116. Eichfelder G, Gebhardt M. Local specific absorption rate control for
detection for cardiac magnetic resonance imaging with a novel parallel transmission by virtual observation points. Magn Reson Med.
vectorcardiographic triggering method. J Magn Reson Imaging. 2011;66(5):1468–1476.
2000;12(5):678–688. 117. Graesslin I, Homann H, Biederer S, et al. A specific absorption rate
95. Togawa T, Okai O, Oshima M. Observation of blood flow E.M.F. in prediction concept for parallel transmission MR. Magn Reson Med.
externally applied strong magnetic field by surface electrodes. Med Biol 2012;68(5):1664–1674.
Eng. 1967;5(2):169–170. 118. Metzger GJ, Snyder C, Akgun C, et al. Local B1+ shimming for prostate
96. Stuber M, Botnar RM, Fischer SE, et al. Preliminary report on in vivo imaging with transceiver arrays at 7T based on subject-dependent
coronary MRA at 3 tesla in humans. Magn Reson Med. 2002;48(3): transmit phase measurements. Magn Reson Med. 2008;59(2):396–409.
425–429. 119. Kramer CM, Barkhausen J, Flamm SD, et al. Standardized cardiovascular
97. Becker M, Frauenrath T, Hezel F, et al. Comparison of left ventricular magnetic resonance (CMR) protocols 2013 update. J Cardiovasc Magn
function assessment using phonocardiogram- and electrocardiogram- Reson. 2013;15:91.
triggered 2D SSFP CINE MR imaging at 1.5 T and 3.0 T. Eur Radiol. 120. van Kalleveen IM, Koning W, Boer VO, et al. Adiabatic turbo spin echo
2010;20(6):1344–1355. in human applications at 7 T. Magn Reson Med. 2012;68(2):580–587.
98. Frauenrath T, Niendorf T, Malte K. Acoustic method for synchronization 121. Kramer CM, Barkhausen J, Flamm SD, et al. Standardized cardiovascular
of magnetic resonance imaging (MRI). Acta Acust United Acust. magnetic resonance imaging (CMR) protocols, society for
2008;94(1):148–155. cardiovascular magnetic resonance: board of trustees task force on
99. IEC. 60601-2-33 Medical electrical equipment—Part 2-33: Particular standardized protocols. J Cardiovasc Magn Reson. 2008;10:35.
requirements for the basic safety and essential performance of magnetic 122. Wagenhaus B, Pohlmann A, Dieringer MA, et al. Functional and
resonance equipment for medical diagnosis. Edition 3.0. 2010. morphological cardiac magnetic resonance imaging of mice using a
100. Wang Z, Lin JC, Mao W, et al. SAR and temperature: simulations and cryogenic quadrature radiofrequency coil. PLoS One. 2012;7(8):e42383.
comparison to regulatory limits for MRI. J Magn Reson Imaging. 123. Niendorf T, Pohlmann A, Reimann HM, et al. Advancing cardiovascular,
2007;26(2):437–441. neurovascular, and renal magnetic resonance imaging in small rodents
101. Murbach M, Neufeld E, Capstick M, et al. Thermal tissue damage model using cryogenic radiofrequency coil technology. Front Pharmacol.
analyzed for different whole-body SAR and scan durations for standard 2015;6:255.
MR body coils. Magn Reson Med. 2014;71(1):421–431. 124. Vitanis V, Manka R, Giese D, et al. High resolution three-dimensional
102. Hoult DI. Sensitivity and power deposition in a high-field imaging cardiac perfusion imaging using compartment-based k-t principal
experiment. J Magn Reson Imaging. 2000;12(1):46–67. component analysis. Magn Reson Med. 2011;65(2):575–587.
103. Winter L, Niendorf T. Electrodynamics and radiofrequency antenna 125. Ogawa S, Menon RS, Tank DW, et al. Functional brain mapping by
concepts for human magnetic resonance at 23.5 T (1 GHz) and beyond. blood oxygenation level-dependent contrast magnetic resonance
MAGMA. 2016;29(3):641–656. imaging. A comparison of signal characteristics with a biophysical
104. Murbach M, Neufeld E, Pruessmann KP, Kuster N. Safe MR scan times model. Biophys J. 1993;64(3):803–812.
based on CEM43 tissue damage thresholds, using electromagnetic and 126. Friedrich M, Niendorf T, Schulz-Menger J. Blood oxygen level-
thermal simulations with anatomically correct human models and dependent magnetic resonance imaging in patients with stress-induced
considering local thermoregulation. Proc Int Soc Magn Reson Med. angina. ACC Curr J Rev. 2004;13(4):30.
2012;313. 127. Jahnke C, Gebker R, Manka R, et al. Navigator-gated 3D blood oxygen
105. Yarmolenko PS, Moon EJ, Landon C, et al. Thresholds for thermal damage level–dependent CMR at 3.0-T for detection of stress-induced
to normal tissues: an update. Int J Hyperthermia. 2011;27(4):320–343. myocardial ischemic reactions. JACC Cardiovasc Imaging. 2010;3(4):
106. Oh S, Webb AG, Neuberger T, et al. Experimental and numerical 375–384.
assessment of MRI-induced temperature change and SAR distributions 128. Karamitsos TD, Leccisotti L, Arnold JR, et al. Relationship between
in phantoms and in vivo. Magn Reson Med. 2010;63(1):218–223. regional myocardial oxygenation and perfusion in patients with
107. Oh S, Ryu YC, Carluccio G, et al. Measurement of SAR-induced coronary artery disease insights from cardiovascular magnetic resonance
temperature increase in a phantom and in vivo with comparison to and positron emission tomography. Circ Cardiovasc Imaging.
numerical simulation. Magn Reson Med. 2014;71(5):1923–1931. 2010;3(1):32–40.
108. Eryaman Y, Akin B, Atalar E. Reduction of implant RF heating through 129. Tsaftaris SA, Zhou X, Tang R, et al. Detecting myocardial ischemia at
modification of transmit coil electric field. Magn Reson Med. rest with cardiac phase-resolved blood oxygen level-dependent
2011;65(5):1305–1313. cardiovascular magnetic resonance. Circ Cardiovasc Imaging.
109. Etezadi-Amoli M, Stang P, Kerr A, et al. Controlling radiofrequency- 2013;6(2):311–319.
induced currents in guidewires using parallel transmit. Magn Reson 130. Wacker CM, Bock M, Hartlep AW, et al. Changes in myocardial
Med. 2015;74(6):1790–1802. oxygenation and perfusion under pharmacological stress with
110. Winter L, Oezerdem C, Hoffmann W, et al. Thermal magnetic dipyridamole: assessment using T*2 and T1 measurements. Magn Reson
resonance: physics considerations and electromagnetic field simulations Med. 1999;41(4):686–695.
up to 23.5 tesla (1GHz). Radiat Oncol. 2015;10:201. 131. He T. Cardiovascular magnetic resonance T2* for tissue iron assessment
111. Langereis S, Keupp J, van Velthoven JL, et al. A temperature-sensitive in the heart. Quant Imaging Med Surg. 2014;4(5):407–412.
liposomal 1H CEST and 19F contrast agent for MR image-guided drug 132. Turner R, Jezzard P, Wen H, et al. Functional mapping of the human
delivery. J Am Chem Soc. 2009;131(4):1380–1381. visual cortex at 4 and 1.5 tesla using deoxygenation contrast EPI. Magn
112. Virtanen H, Huttunen J, Toropainen A, Lappalainen R. Interaction of Reson Med. 1993;29(2):277–279.
mobile phones with superficial passive metallic implants. Phys Med Biol. 133. van der Zwaag W, Francis S, Head K, et al. fMRI at 1.5, 3 and 7 T:
2005;50(11):2689. characterising BOLD signal changes. Neuroimage. 2009;47(4):1425–1434.
113. Bakhai A, Booth J, Delahunty N, et al. The SV stent study: a prospective, 134. Donahue MJ, Hoogduin H, van Zijl PC, et al. Blood oxygenation
multicentre, angiographic evaluation of the BiodivYsio phosphorylcholine level-dependent (BOLD) total and extravascular signal changes and
coated small vessel stent in small coronary vessels. Int J Cardiol. 2005; DeltaR2* in human visual cortex at 1.5, 3.0 and 7.0 T. NMR Biomed.
102(1):95–102. 2011;24(1):25–34.
114. Ansems J, Kolk A, Kroeze H, et al. MR Imaging of patients with stents is 135. Hernando D, Vigen KK, Shimakawa A, Reeder SB. R(2) mapping in the
safe at 7.0 tesla. Proc Int Soc Magn Reson Med. 2012;2764. presence of macroscopic B(0) field variations. Magn Reson Med.
115. Yeung CJ, Susil RC, Atalar E. RF heating due to conductive wires during 2011;68(3):830–840.
MRI depends on the phase distribution of the transmit field. Magn 136. Pohlmann A, Arakelyan K, Hentschel J, et al. Detailing the relation
Reson Med. 2002;48(6):1096–1098. between renal T2* and renal tissue pO2 using an integrated approach of
160.e4 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

parametric magnetic resonance imaging and invasive physiological 158. Dieringer MA, Deimling M, Santoro D, et al. Rapid parametric mapping
measurements. Invest Radiol. 2014;49(8):547–560. of the longitudinal relaxation time T1 using two-dimensional variable
137. Pohlmann A, Hentschel J, Fechner M, et al. High temporal resolution flip angle magnetic resonance imaging at 1.5 tesla, 3 tesla, and 7 tesla.
parametric MRI monitoring of the initial ischemia/reperfusion phase in PLoS One. 2014;9(3):e91318.
experimental acute kidney injury. PLoS One. 2013;8(2):e57411. 159. Chiu H-C, Kovacs A, Ford DA, et al. A novel mouse model of lipotoxic
138. Schar M, Vonken EJ, Stuber M. Simultaneous B(0)- and B(1)+-map cardiomyopathy. J Clin Invest. 2001;107(7):813–822.
acquisition for fast localized shim, frequency, and RF power 160. Hernando D, Kellman P, Haldar JP, Liang ZP. Robust water/fat
determination in the heart at 3 T. Magn Reson Med. 2010;63(2):419–426. separation in the presence of large field inhomogeneities using a graph
139. Reeder SB, Faranesh AZ, Boxermann JL, McVeigh ER. In vivo cut algorithm. Magn Reson Med. 2010;63(1):79–90.
measurement of T2* and field inhomogeneity maps in the human heart 161. Kellman P, Hezel F, Shah S, et al. Fat-water separated imaging at 7T:
at 1.5 T. Magn Reson Med. 1998;39:988–998. initial results for cardiac applications. Proc Int Soc Magn Reson Med.
140. Friedrich MG, Karamitsos TD. Oxygenation-sensitive cardiovascular 2011;597.
magnetic resonance. J Cardiovasc Magn Reson. 2013;15:43. 162. Niendorf T, Hardy CJ, Giaquinto RO, et al. Toward single breath-hold
141. Niendorf T, Pohlmann A, Arakelyan K, et al. How bold is blood whole-heart coverage coronary MRA using highly accelerated parallel
oxygenation level-dependent (BOLD) magnetic resonance imaging of imaging with a 32-channel MR system. Magn Reson Med. 2006;56(1):
the kidney? Opportunities, challenges and future directions. Acta Physiol 167–176.
(Oxf). 2015;213(1):19–38. 163. Niendorf T, Saranathan M, Lingamneni A, et al. Short breath-hold,
142. Christen T, Zaharchuk G, Pannetier N, et al. Quantitative MR estimates volumetric coronary MR angiography employing steady-state free
of blood oxygenation based on T2*: a numerical study of the impact of precession in conjunction with parallel imaging. Magn Reson Med.
model assumptions. Magn Reson Med. 2012;67(5):1458–1468. 2005;53(4):885–894.
143. Lee J, van Gelderen P, Kuo LW, et al. T2*-based fiber orientation 164. Voit D, Zhang S, Unterberg-Buchwald C, et al. Real-time cardiovascular
mapping. Neuroimage. 2011;57(1):225–234. magnetic resonance at 1.5 T using balanced SSFP and 40 ms resolution.
144. Dibb R, Qi Y, Liu C. Magnetic susceptibility anisotropy of myocardium J Cardiovasc Magn Reson. 2013;15:79.
imaged by cardiovascular magnetic resonance reflects the anisotropy of 165. Kowallick JT, Joseph AA, Unterberg-Buchwald C, et al. Real-time
myocardial filament alpha-helix polypeptide bonds. J Cardiovasc Magn phase-contrast flow MRI of the ascending aorta and superior vena cava
Reson. 2015;17:60. as a function of intrathoracic pressure (Valsalva manoeuvre). Br J
145. Li TQ, van Gelderen P, Merkle H, et al. Extensive heterogeneity in white Radiol. 2014;87(1042):20140401.
matter intensity in high-resolution T2*-weighted MRI of the human 166. Fasshauer M, Joseph AA, Kowallick JT, et al. Real-time phase-contrast
brain at 7.0 T. Neuroimage. 2006;32(3):1032–1040. flow MRI of haemodynamic changes in the ascending aorta and
146. Shmueli K, de Zwart JA, van Gelderen P, et al. Magnetic susceptibility superior vena cava during Mueller manoeuvre. Clin Radiol. 2014;69(10):
mapping of brain tissue in vivo using MRI phase data. Magn Reson Med. 1066–1071.
2009;62(6):1510–1522. 167. Joseph A, Kowallick JT, Merboldt KD, et al. Real-time flow MRI of the
147. Ziener CH, Kampf T, Melkus G, et al. Signal evolution in the local aorta at a resolution of 40 msec. J Magn Reson Imaging.
magnetic field of a capillary—analogy to the damped driven harmonic 2014;40(1):206–213.
oscillator. Magn Reson Imaging. 2012;30(4):540–553. 168. Madelin G, Regatte RR. Biomedical applications of sodium MRI in vivo.
148. Duyn JH, van Gelderen P, Li TQ, et al. High-field MRI of brain cortical J Magn Reson Imaging. 2013;38(3):511–529.
substructure based on signal phase. Proc Natl Acad Sci USA. 2007; 169. Sandstede JJW, Hillenbrand H, Beer M, et al. Time course of 23Na signal
104(28):11796–11801. intensity after myocardial infarction in humans. Magn Reson Med.
149. Deistung A, Schafer A, Schweser F, et al. Toward in vivo histology: a 2004;52(3):545–551.
comparison of quantitative susceptibility mapping (QSM) with 170. Nagel AM, Laun FB, Weber MA, et al. Sodium MRI using a density-
magnitude-, phase-, and R2*-imaging at ultra-high magnetic field adapted 3D radial acquisition technique. Magn Reson Med. 2009;62(6):
strength. Neuroimage. 2013;65:299–314. 1565–1573.
150. Ziener CH, Kampf T, Reents G, et al. Spin dephasing in a magnetic 171. Rajiah P, Bolen MA, Cardiovascular MR. Imaging at 3 T: opportunities,
dipole field. Phys Rev E Stat Nonlin Soft Matter Phys. 2012;85(5 Pt 1): challenges, and solutions. Radiographics. 2014;34(6):1612–1635.
051908. 172. Dietrich O, Reiser MF, Schoenberg SO. Artifacts in 3-T MRI: physical
151. Deleted in review. background and reduction strategies. Eur J Radiol. 2008;65(1):29–35.
152. Huelnhagen T, Hezel F, Pohlmann A, et al. High Spatial Resolution 173. Weinberger O, Winter L, Dieringer M, et al. Local multi-channel RF
Myocardial T2* Mapping at 7.0 T Reveals Differences Between Healthy surface coil versus body RF coil transmission for cardiac magnetic
Volunteers and Patients with Hypertrophic Cardiomyopathy. Proc Int resonance at 3 tesla: which configuration is winning the game? PLoS
Soc Magn Reson Med. 2015. Milan, Italy. 2599. One. 2016;11(9):e0161863.
153. Messroghli DR, Radjenovic A, Kozerke S, et al. Modified Look-Locker 174. Niendorf T, Barth M, Kober F, Trattnig S. From ultrahigh to extreme
inversion recovery (MOLLI) for high-resolution T1 mapping of the field magnetic resonance: where physics, biology and medicine meet.
heart. Magn Reson Med. 2004;52(1):141–146. MAGMA. 2016;29:309–311.
154. von Knobelsdorff-Brenkenhoff F, Prothmann M, Dieringer MA, et al. 175. Budinger TF, Bird MD, Frydman L, et al. Toward 20 T magnetic
Myocardial T1 and T2 mapping at 3 T: reference values, influencing resonance for human brain studies: opportunities for discovery and
factors and implications. J Cardiovasc Magn Reson. 2013;15(1):53. neuroscience rationale. MAGMA. 2016;29(3):617–639.
155. Messroghli DR, Niendorf T, Schulz-Menger J, et al. T1 mapping in 176. Polenova T, Budinger TF. Ultrahigh field NMR and MRI: science at a
patients with acute myocardial infarction. J Cardiovasc Magn Reson. crossroads. Report on a jointly-funded NSF, NIH and DOE workshop,
2003;5(2):353–359. held on November 12-13, 2015 in Bethesda, Maryland. J Magn Reson.
156. Piechnik SK, Ferreira VM, Dall’Armellina E, et al. Shortened modified 2016;266:81–86.
Look-Locker inversion recovery (ShMOLLI) for clinical myocardial 177. Manjón JV, Coupé P, Martí-Bonmatí L, et al. Adaptive non-local means
T1-mapping at 1.5 and 3 T within a 9 heartbeat breathhold. J Cardiovasc denoising of MR images with spatially varying noise levels. J Magn
Magn Reson. 2010;12:69. Reson Imaging. 2010;31(1):192–203.
157. Rodgers CT, Tao Y, Piechnik SK, et al. Saturation recovery single-shot
acquisition (SASHA) for T1 mapping in the human heart at 7T. Proc Int
Soc Magn Reson Med. 2015. Toronto, Canada. 0758.
14 
Clinical Cardiovascular Magnetic Resonance
Imaging Techniques
David C. Wendell, Wolfgang G. Rehwald, John D. Grizzard, Raymond J. Kim,
and Robert M. Judd

The first dedicated cardiovascular magnetic resonance (CMR) clinical Through the years our philosophy about how to perform CMR has
services opened in the United States in mid-1990s. Since that time, evolved into the concept of a CMR “exam menu” which has not only
CMR imaging has become routine at most academic medical centers, allowed us to streamline our clinical service but has also made it con-
with a number of centers now running multiple CMR scanners dedicated siderably easier for us to teach CMR to cardiology fellows and level 2
to cardiovascular imaging studies. Although different centers still differ trainees. The underlying idea of the examination menu (Fig. 14.3) is
somewhat with regard to the exact clinical CMR imaging protocols that most if not all routine clinical CMR can be performed simply by
employed, in recent years cross-institutional CMR protocols have become combining one or more items from a predetermined menu and per-
much more consistent. In this chapter we focus on the clinical CMR forming these scan protocols/pulse sequences in a single consistent
imaging techniques that we routinely use at our institution, and we manner. As can be seen from Fig. 14.3, the CMR Stress Test is simply
believe these are now representative of most high-volume dedicated one combination of items from the examination menu. Virtually every
CMR clinical services. patient we examine simply undergoes one or more of the items shown
The dedicated CMR clinical service at the Duke Cardiovascular in the menu and, perhaps more importantly, only rarely do we find it
Magnetic Resonance Center (DCRMC) first opened in July 2002. necessary to perform a scan not listed there.
Since that time, we have experimented with and evaluated many Accordingly, the remainder of this chapter focuses on providing
approaches to CMR and have directly observed their effects on more detail about each of the items on the examination menu, with
overall clinical volume. As shown in Fig. 14.1, clinical volume at the an emphasis on providing practical information typically not provided
DCRMC has grown steadily since our service first opened and cur- in other CMR textbooks. In the Conclusion section of this chapter we
rently consists of approximately 4000 billed CMR imaging procedures present typical examples of how the items on the examination menu
per year. can be combined to answer common diagnostic questions in cardiology,
From 2002 to 2010 the percentage of each type of CMR test we such as the detection of coronary artery disease (CAD) and the evalu-
performed varied somewhat as the types of patients referred to us ation of aortic disease.
evolved, and as we ourselves modified the tests we offered. From 2010
to 2015 (the latest year data are available), however, the tests we offer
and perform have remained relatively constant.
SCOUTING (“SCAN” = “SCOUT”)
Fig. 14.2 summarizes the indications for CMR scans, as well as the The thoracic scout examination is the simplest and most commonly
corresponding current procedural terminology (CPT) codes used for used CMR procedure performed during the CMR examination. The
billing. These data were pooled across four US hospitals with dedicated goal of the scout examination is to establish the anatomic position of
CMR clinical services and, as such, portray an arguably representative the heart and the short-axis and long-axis views of the heart. Ana-
picture of clinical practice patterns in the United States. The largest tomic variations from patient to patient cause both the short-axis
referral indication for CMR was heart failure, followed by ischemia and long-axis cardiac views to lie at arbitrary angles with respect to
evaluation and vascular disease. The most common CPT codes were scanner coordinates. These views are then obtained using “double
75561 (morphology and function with/without contrast), 75565 (veloc- oblique” planes.
ity flow), and 71555 (magnetic resonance angiography [MRA]). About The first step in determining the proper double-oblique views is to
20% of all CMR scans involved stress testing (CPT code 75563). Unlike acquire images along the axes of the scanner: that is, axial, sagittal, and
the four-hospital data of Fig. 14.2, however, at our institution roughly coronal planes passing through the thoracic cavity. Fig. 14.4 shows
50% of our CMR scans involve stress testing. The DCMRC definition typical examples of scout images. These scout images are used to pre-
of a “CMR Stress Test” can briefly be described as a group of three scribe single-oblique images: that is, perpendicular to the axial image
individual scans: of Fig. 14.4, going through the left ventricle (LV) parallel to the septum
1. Cines (to examine biventricular contractile function). (dashed yellow line). The single-oblique images (pseudo two-chamber
2. Adenosine stress/rest perfusion (to examine regional myocardial and pseudo four-chamber views) are used to determine the true short
perfusion). axis of the heart (doubly oblique). Therefore the goal of the scout is
3. Late gadolinium enhancement (LGE; to examine viability/infarction). to acquire a series of images similar to that of Fig. 14.4 which define
The DCMRC CMR Stress Test is described in more detail in a paper the short and long axis of the heart.
by Klem et al.,1 and has arguably become the bread and butter of our The pulse sequence we use for the scout images is based on balanced
clinical service. steady-state free precession (bSSFP). The underlying concept of bSSFP

161
162 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Duke hospital CMR clinical procedures


4000

3500

Number of CMR procedures


3000

2500

2000

1500

1000

500

0
20

20

20

20

20

20

20

20

20

20

20

20

20

20
02

03

04

05

06

07

08

09

10

11

12

13

14

15
FIG. 14.1  Annual Duke Cardiovascular Magnetic Resonance (CMR) Center clinical volume. A “Procedure”
corresponds to a current procedural terminology code.

Indications for CMR


CPT
CPT DESCRIPTION # Reports %
CODE

MORPHOLOGY & FUNCTION W/


75561 4,723 66%
WO CONTRAST
Other
Congenital 13% CHF/ 75565 VELOCITY FLOW MAPPINGS 3,714 51%
heart disease cardiomyop-
8% athy
31% 71555 CHEST MRA 3,312 46%
Arrhythmia
9%
STRESS MORPHOLOGY &
75563 1,414 19%
FUNCTION W/WO CONTRAST
Valve
assessment Ischemia
10% evaluation MORPHOLOGY & FUNCTION
75557 270 3%
Vascular 15% WITHOUT CONTRAST
disease
14%
74185
OTHER VASCULAR:
72198
Abdomen, pelvis, neck, lower 761 10%
70549
extremity
73625

CMR clinical procedures (8,242 scans across four US hospitals)


FIG. 14.2  Recent (2010–2014) makeup of cardiovascular magnetic resonance (CMR) clinical referral indica-
tions across four US hospitals. CHF, Congestive heart failure; CPT, current procedural terminology; MRA,
magnetic resonance angiography.

was described in the mid-1980s2 but only since the late 1990s has CMR
1 −− Scout scanner hardware been capable of achieving the bSSFP magnetization
2 −− Left ventricular/right ventricular cine state. Fig. 14.5 shows the bSSFP pulse sequence timing diagram char-
3 −− Stress–rest myocardial perfusion acterized by its elegant simplicity, with symmetry around the data
4 −− Late gadolinium enhancement acquisition window on all three gradient axes. The axis labeled “slice”
5 −− Morphology serves to select the slice to be imaged. The waveforms on the “read”
6 −− Angiography axis create the magnetic resonance (MR) signal as an echo (see “signal”
7 −− Flow/velocity axis on bottom). During the positive portion of the “read” waveform
FIG. 14.3  The Duke Cardiovascular Magnetic Resonance Center “exam the MR signal is digitally sampled. The “phase” encoding axis imposes
menu.” See text for details. a different phase on each echo which allows spatial encoding of the
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 163

Coronal
Axial Sagittal

FIG. 14.4  Scout imaging planes are typically obtained in the axial, coronal, and sagittal orientations.

second dimension called y in Fig. 14.6. In practice, the primary advan- example we assume a heart rate of 60 beats per minute (RR duration
tages of bSSFP imaging are: (1) fast imaging (i.e., short repetition time = 1000 ms). The first “event” the scanner hardware must play out is a
[TR]) and (2) very high signal-to-noise ratio (SNR) images. Accordingly, delay time of approximately 500 ms after the QRS (to get to diastole),
we use bSSFP not only for scouting but also for cine imaging (described followed by the 300 ms of image data acquisition. Fig. 14.7 graphically
in the Contractile Function section). depicts cardiac gating for scout imaging.
Acquiring any CMR image in essence involves filling the raw data
space, often referred to as “k-space.” Fig. 14.6 graphically depicts the
process of filling k-space as a series of “lines” starting at the top and
MORPHOLOGY (“SCAN” = “MORPHOLOGY”)
proceeding to the bottom. Each line is actually one echo (see zoomed- CMR scanning for cardiac morphology is used less often than cine
up box) acquired during the read event (“read” signal in Fig. 14.5). The imaging or even viability imaging, but the technical aspects of morphol-
echo runs in the x-direction. The y-dimension is the phase-encoding ogy, particularly as it relates to cardiac gating, are only modestly more
direction. For a bSSFP pulse sequence running on a typical modern CMR complex than for scouting, so we present these next.
scanner the time needed to acquire each k-space line is approximately The “goal” of morphology scanning is essentially to create a series
3 ms. So, for the 100 k-space lines typical for scouting, the total image of parallel slices which “bread loaf ” the thoracic cavity to examine
acquisition time is ~300 ms (100 × 3) or one-third of a second. This is vascular anatomy. Typically, three sets of black-blood and bright-blood
fast enough to effectively freeze heart motion provided that the image images are acquired in the axial, coronal, and sagittal orientations. Once
data are acquired during diastole when the heart is relatively quiescent. acquired, the scanner operator can then step through each series of
Acquiring the image data in diastole requires that the scanner hard- image slices and understand the patient’s vascular anatomy as well as
ware is “gated” to the cardiac cycle. This is typically achieved by having plan additional scan planes.
the scanner detect the R-wave of the electrocardiographic (ECG) signal It is often desirable to do this both with “black-blood” imaging (Fig.
and initiating the scan sequence at that time. In the following scouting 14.8) and with “bright-blood” imaging (Fig. 14.9). The CMR pulse
164 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

sequences used for black-blood and bright-blood morphology imaging both bSSFP and HASTE slices would take 40 + 80 = 120 heartbeats
are half-Fourier acquisition single-shot turbo spin echo (HASTE, Fig. (i.e., about 2 minutes).
14.10A) and bSSFP (Fig. 14.5), respectively. Fig. 14.10A shows the pulse Black-blood imaging is often advantageous for the examination of
sequence timing diagram of the turbo spin echo sequence, and Fig. morphology because it allows clear differentiation of the inner portion
14.10B shows the simplified spin echo sequence. HASTE is a special of the vessel wall from the blood. Black-blood HASTE can be achieved
variant of the turbo spin echo sequence in which only 60% of k-space by carefully combining CMR physics with the physiology of moving
is sampled to reduce acquisition time. blood. The underlying reason that the blood appears black in HASTE
Cardiac ECG gating for bSSFP morphology images is the same as images is caused by preparatory radiofrequency (RF) pulses before the
that used for scouting, namely, by acquiring the entire image during HASTE readout (see Fig. 14.10). The basic concept is graphically depicted
a single diastolic phase (Fig. 14.11). For example, a set of 40 bSSFP in Figs. 14.12 and 14.13. Soon after the R-wave, a brief (1–3 ms) RF
images for morphology would require 40 consecutive heartbeats. ECG pulse is played, which causes all of the magnetization in the body (“non-
gating for HASTE imaging is also performed in diastole; however, selective” or “hard” pulse) to flip upside down (“180 degrees” or “inver-
these images are typically acquired every other heartbeat (e.g., 80 sion” pulse). Immediately after this, a second RF pulse is played, which
consecutive heartbeats). HASTE images require additional heartbeats causes only the magnetization within the prescribed imaging slice to
to satisfy the black-blood imaging condition (described later). Mor- flip back to where it started (“slice-selective” re-inversion pulse). The
phology imaging typically takes 3 to 5 minutes and is performed with net effect of these preparation pulses results in the magnetization outside
quiet respiration. For example, the acquisition of the 40 slices with the slice being inverted, whereas the magnetization within the prescribed

Symmetry

+α –α
TR One k-space line
RF Acquiring each line
takes ~3 ms (bSSFP).
y Thus, 100 lines
requires ~300 ms
Slice

Read

x
Phase

FIG. 14.6  Filling of raw data space. The raw data space for cardiovas-
cular magnetic resonance is often referred to as “k-space” and must
Signal Time
be filled line by line to generate an image. Each line contains one echo.
Each echo is acquired after application of a different phase-encoding
FIG. 14.5  Balanced steady-state free precession (bSSFP) pulse sequence step (direction y) and thus contains different information. The k-space
timing diagram. In essence, bSSFP consists of rapid gradient echoes is filled using, for example, the pulse sequence of Fig. 14.5. Then this
whose magnetization is preserved across multiple k-space lines, result- raw data undergoes a two-dimensional Fourier transform, resulting in
ing in images with a high signal-to-noise ratio and short scan time. RF, an image of for example, Fig. 14.4. bSSFP, Balanced steady-state free
Radiofrequency; TR, repetition time. precession.

R R R

ECG

Delay Delay
time time
All k-space lines All k-space lines
(e.g., 100 lines) (e.g., 100 lines)
Scout 1 Scout 2

Trigger Trigger
FIG. 14.7  Cardiac electrocardiographic (ECG) gating for scout imaging consists of a delay following the
ECG R-wave and then rapid acquisition of the entire image in diastole. Imaging within a single heartbeat is
made possible in part by the intrinsic speed of the balanced steady-state free precession pulse sequence.
ECG, Electrocardiogram.
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 165

Left Head Posterior

Right Feet Anterior

Sagittal Axial Coronal

FIG. 14.8  The “goal” of “black-blood” half-Fourier acquisition single-shot turbo spin echo imaging. Shown
are a series of parallel images across the entire thoracic cavity which can then be inspected to determine
vascular anatomy. Usually, sagittal, axial, and coronal stacks are acquired.

imaging slice is restored. During the interval between the preparatory ventricular remodeling,6–8 and as a reference method for other imaging
pulses and the HASTE readout (see Fig. 14.13) the heart contracts and techniques.9–14
expels blood from the slice (see Fig. 14.12, middle), which is subsequently The “goal” of cine imaging is to capture a movie of the beating heart
replaced by fresh blood from outside the slice (see Fig. 14.12, right), to visualize its contractile function. Fig. 14.14 shows a representative
which was previously inverted. When the HASTE readout begins, the example of a mid-ventricular short-axis slice during different time points
previously inverted blood is recovering with T1, and its curve crosses within the cardiac cycle, from early systole to late diastole. Images are
zero at the start of the HASTE readout. Consequently, the blood pro- acquired with the same bSSFP sequence used in bright-blood and scout
duces no signal and appears black. Thus black-blood HASTE is made imaging (see Fig. 14.5). The advantage of bSSFP for cardiac function
possible by a clever combination of a “memory” effect related to CMR imaging is high SNR, high temporal resolution, and excellent blood–
physics (T1 recovery) as well as to the physiology of moving blood myocardium contrast, which helps identify the endocardial border.
(and therefore may have intermediate signal with stagnant blood flow). Typically, a stack of multiple short axis slices (5–6 mm thick) are acquired
Such images are quite helpful in detecting abnormalities in large-vessel at 1-cm increments to provide full LV and RV coverage. Short-axis
anatomy, especially when combined with bright-blood bSSFP images views can be planned perpendicular to the long-axis scout images
at the same location. described in the Scouting section. In addition, cine images can be obtained
in multiple long-axis views such as the two-chamber, three-chamber,
and four-chamber views.
CONTRACTILE FUNCTION (“SCAN” = “CINE”) A key consideration for cine imaging is the time required to acquire
Contractile function is a fundamental part of the CMR examination. a movie of a single cardiac cycle. Current CMR scanners are unable to
Contractile function imaging is used for global and regional wall acquire high spatial and temporal resolution cine images in real time.
motion assessment and has been demonstrated to be highly accurate For example, for a movie with 25 frames each with 96 lines, a total of
and reproducible for LV and right ventricular (RV) volume, ejection 25 × 96 = 2400 lines need to be acquired. Considering that the acquisi-
fraction, and mass measurements.3–5 In recent years, CMR has become tion time for one line is approximately 3 ms, the acquisition time to
widely accepted as the clinical gold standard for contractile function create a series of cine images would be 2400 × 3 ms = 7200 ms, which
assessment. It has been used as an endpoint for the evaluation of is considerably longer than the human cardiac cycle. Imaging speed
Left Head Posterior

Right Feet Anterior

Sagittal Axial Coronal

FIG. 14.9  The “goal” of “bright-blood” balanced steady-state free precession imaging. The same image
planes used for half-Fourier acquisition single-shot turbo spin echo are acquired such that the two forms of
images can be compared.

90°
180° 180° 180°
RF

Slice
1st 2nd 3rd echo
Read
Phase Fast spin echo
A sequence

90° 180° 90° 180°

RF

Slice

Read

Phase

Signal
TE/2 TE/2
TE TE
TR
B Spin echo sequence
FIG. 14.10  Pulse sequence timing diagram for half-Fourier acquisition single-shot turbo spin echo (HASTE)
(A) and for spin echo (B). In essence, HASTE consists of a series of spin echoes referred to as turbo spin
echoes. Additional postprocessing steps are required to obtain the HASTE image. The spin echo sequence
(B) is the predecessor of the turbo spin echo sequence (A). In HASTE, blood appears black in the resulting
images because of the preparatory pulse described in Figs. 14.12 and 14.13. RF, Radiofrequency; TE, echo
time; TR, repetition time.
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 167

could be reduced by acquiring fewer phase-encoding lines, but at the of multiple segments acquired during different cardiac cycles; the data
cost of spatial resolution (fewer lines per image) or temporal resolution are then reassembled during image reconstruction to form a movie
(fewer movie frames). For cine imaging, image quality can be improved depicting a single cardiac cycle (although compared with echocardiog-
by using a segmented k-space acquisition assuming the beat-to-beat raphy, the data are not “real time”). For example, the scheme in Fig.
variation in heart rate is minimal. 14.15 acquires 20 segments of 16 lines each in every cardiac cycle.
The segmented k-space data acquisition collects only parts of each Assuming that the scanner operator has chosen to obtain 96 k-space
movie frame over 5 to 8 consecutive cardiac cycles and recombines the lines for each movie frame, the scan will acquire data across 96/16 =
data to form a movie loop. Fig. 14.15 shows the scheme used for CMR six heartbeats. In this example, the green segment is always acquired
in which only a fraction of k-space data is acquired for any given movie immediately after the R-wave and is used to reconstruct the first movie
frame during any single heartbeat. This fraction is referred to as a frame, the orange segment is acquired next and becomes the second
“segment” (e.g., 16 k-space lines per segment in Fig. 14.15) and typically movie frame, and so on. In practice, cine imaging is performed during
is adjusted by the scanner operator to produce an adequate number of breath-holding and takes 5 to 8 seconds for each cardiac view (e.g., one
movie frames (generally 16–20) given the patient’s RR interval. Accord- short-axis movie).
ingly, the full k-space data for any one movie frame actually consists
PERFUSION AT STRESS AND REST
(“SCAN” = “PERFUSION”)
k-space k-space
slice 1 slice 2 Encouraged by a number of early clinical studies1,15–19 adenosine stress–
rest perfusion CMR has become more widespread in CMR centers.
Heartbeat 1 Perfusion CMR has been shown to accurately diagnose coronary artery
Heartbeat 2
disease with high sensitivity and specificity. Our center’s approach of
combining LGE CMR and rest–perfusion imaging with stress perfusion
R Slice 1 R Slice 2 R imaging is important for distinguishing true perfusion defects on stress
images from artifacts.1
ECG The “goal” of CMR perfusion imaging is to create a movie of the
first passage of the CMR contrast media (typically gadolinium based)
Delay Image Delay Image
through the myocardium (e.g., first-pass perfusion). Fig. 14.16 gives an
time slice 1 time slice 2 example of an adenosine stress perfusion scan as well as the corresponding
Trigger Trigger
rest perfusion scan in a patient with a stress-induced perfusion defect.
The blue slice is shown at three representative time points: (1) before
FIG. 14.11  Electrocardiogram (ECG) gating in half-Fourier acquisition
single-shot turbo spin echo (HASTE) and balanced steady-state free
contrast arrival; (2) at the time of the contrast arrival in the RV; and (3)
precession (bSSFP) morphology imaging. Each HASTE and bSSFP mor- shortly after the time of the contrast arrival in the LV. During adenosine
phologic image is acquired in a single diastolic image, similar to scout stress, two perfusion defects appear as dark regions in the anterior and
imaging. To skip systole, the wait period is inserted after electrocardio- inferolateral wall (hypoenhancement; see arrows) surrounded by bright
graphic R-wave trigger detection. contrast-enhanced normally perfused myocardium. In the corresponding

R-wave (end-diastole) Systole Late diastole

Nonselective inversion

Slice-selective “re-inversion”

Imaged slice
FIG. 14.12  Physiology of “black-blood” half-Fourier acquisition single-shot turbo spin echo imaging. Imme-
diately after trigger detection a nonselective inversion (green) is played that is instantly followed by a slice-
selective re-inversion (orange). The so-prepared slice (orange) changes shape and position during cardiac
contraction, but returns to its original geometry during diastole, when the blood is black and the heart has
little motion. Nonblack re-inverted blood (orange) is expelled from the slice during systole. During diastole,
an image is obtained from a slice (blue) that is thinner than and lies inside of the prepared slice (orange).
168 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

• HASTE = Half-Fourier Acquisition Single-Shot Turbo Spin Echo


• Typically used with a black-blood pulse.

ECG

All lines acquired for one complete image.

Noninverted myocardium 90180 180 180 180 180 180 180


Nonselective inversion

re-inversion Time needed


Selective to null blood
Time
od
d blo
erte
wing inv
Inflo

FIG. 14.13  Cardiac electrocardiogram (ECG) gating for “black-blood” half-Fourier acquisition single-shot turbo
spin echo (HASTE) imaging; see text for details. Image acquisition starts when the magnetic relaxation curve
of the inverted blood in the cavity is passing through zero/null signal: that is, when the blood is black. Note
that in HASTE, the center lines of k-space are acquired first (“centric reordering”). Because these lines
contain information of image brightness and contrast, they must be acquired while blood is “black”: hence,
at the beginning of the acquisition train.

Early systole

1 2 3 4

Late diastole

5 6 7 8

FIG. 14.14  The “goal” of cine cardiovascular magnetic resonance imaging. A typical example of a mid-
ventricular series of short-axis cine images at the level of the papillary muscles, acquired with balanced
steady-state free precession, is shown. Typically, the cine series contains about 20 image phases at each
level, of which only eight are shown here because of space constraints.

rest perfusion images, conversely, perfusion appears normal in these angle shot [FLASH], Fig. 14.17).20 Perfusion imaging data are acquired
regions, indicating normal resting perfusion with reduced coronary continuously rather than in the segmented manner previously described
flow reserve, presumably because of the presence of hemodynamically for cine imaging. A single image takes between 100 ms and 150 ms
significant coronary artery stenoses. to acquire and, because of time constraints, imaging is typically per-
The sequences we use for perfusion imaging are based on a 90-degree formed throughout the entire cardiac cycle. All k-space lines for each
saturation pulse followed by a gradient recalled echo readout (fast low of the 4 to 5 short-axis slice locations are acquired every cardiac cycle
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 169

Heartbeat
1 2 3 4 5 6
ECG

Trigger
Acquired in
segments: 1 2 30 1 2 30 1 2 30 1 2 30 1 2 30 1 2 30

30 segments
Each segment
acquired each
contains 10 lines
heartbeat

10 lines

Ordered after
cardiac phase:
Cardiac
1 2 30 phase
FIG. 14.15  Cardiac electrocardiogram (ECG) gating for cine imaging. Raw data lines are acquired in segments
over the course of six successive heartbeats. The segments are then sorted in the k-space arrays for 20
movie frames/cardiac phases. Multiple lines form one segment, multiple segments form one k-space, and
Fourier transform of one k-space yields one movie frame. See text for details.

Stress perfusion slice 1

slice 2

slice 3

slice 4

Time

slice 1

slice 2

slice 3

Rest perfusion slice 4

FIG. 14.16  The “goal” of cardiovascular magnetic resonance perfusion imaging. Stress and rest perfusion
images are shown. In this example four slices (basal red, green, blue, apical yellow) are acquired with each
heartbeat. An apical slice (blue) is shown at different time points throughout image acquisition (3 out of 50
time frames shown: before contrast arrival, at the time of the contrast arrival in the right ventricle, and shortly
after the time of the contrast arrival in the left ventricle). Two stress perfusion defects appear as dark regions
(hypoenhancement, arrows). They are surrounded by bright contrast-enhanced normally perfused myocardium.
The defects are reversible as they are not present (no hypoenhancement, arrows) during rest perfusion.
170 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

α Rephaser to keep
spins aligned

RF
Destroy transverse
A A magnetization to
Slice avoid mixing of
A
longitudinal and
Spoiling transverse
magnetization
Phase
Phase
encoder
B B
Read
B

ADC
Dephasing
Signal
Refocusing Dephasing
Time
FIG. 14.17  Pulse sequence timing diagram for fast low angle shot (FLASH) cardiovascular magnetic reso-
nance perfusion. See text for details. ADC, Analog-to-digital converter; RF, radiofrequency.

Acquire multiple entire slices during each heartbeat

ECG

Trigger

1 2 3 4 5

Heartbeat 1 Beat 2 Beat 3 Beat 4 Beat 5

Slice 1

Slice 2

Slice 3

Slice 4

All lines for each image (one slice and one time point) are acquired in a single shot.
FIG. 14.18  Image acquisition in cardiovascular magnetic resonance perfusion scanning. Images are acquired
in a continuous stream (single shot) over 50 to 60 heartbeats. Depending on the duration of the RR interval,
up to five slices can be acquired during each RR interval. ECG, Electrocardiogram.

(Fig. 14.18) to characterize the first pass of the contrast agent through- rate with adenosine stress. Altogether, perfusion imaging is typically
out the LV myocardium. In general, perfusion imaging sacrifices image performed for 40 to 50 seconds.
quality for the faster scan time required to acquire multiple images To enhance the differences in image intensity between poorly and
within a single cardiac cycle. In general, the total number of slices that normally perfused myocardium a saturation pulse (90-degree pulse)
can be acquired within one cardiac cycle is limited by the patient’s is played as a preparation pulse before each image acquisition (Fig.
RR interval. For example, for a heart rate of 80 beats per minute, the 14.19). The 90-degree pulse tips all longitudinal magnetization into
RR interval would be ~750 ms and five slices of 150 ms/slice could the transverse plane. The transverse magnetization then is dephased
theoretically be acquired. For higher heart rates, the number of short- (spoiled) by a spoiler gradient. The first pass of contrast (e.g., gadolinium-
axis slice locations would have to be reduced to four. In addition, the diethylenetriaminepentaacetic acid [Gd-DTPA]) shortens T1 in regions
heart rate often increases with adenosine, which reduces the available with a higher concentration of gadolinium (e.g., normally perfused
imaging window. Thus it is prudent to allow some extra time within myocardium) compared with regions with reduced perfusion, lower
the resting RR interval to account for the expected increase in heart contrast concentration, and a longer T1. Fig. 14.19 presents the relaxation
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 171

Mz
Myocardium with normal perfusion has
shorter T1 (recovers faster, looks brighter)

+M0
c

Myocardium with relatively reduced perfusion


a has longer T1 (recovers slower, looks darker)

Time
90°

RF

Gradient

Readout one entire image


Spoil transverse
(when curves have maximum separation)
magnetization
FIG. 14.19  Magnetization recovery. The recovery of the longitudinal magnetization is faster in myocardium
with normal perfusion due to T1 shortening as a consequence of high gadolinium contrast agent concentra-
tion. Relatively speaking, contrast concentration is lower in myocardium with reduced perfusion resulting in
myocardial T1 values that are more similar to precontrast values. To maximize the difference in signal intensity
between myocardium with normal (higher concentration of gadolinium) and reduced perfusion (lower con-
centration of gadolinium), the optimal time for image acquisition is when the difference between both curves
is largest (b). RF, Radiofrequency.

curves for both regions. The timing between the saturation pulse and of a significant coronary artery stenosis. The resulting disparity in blood
image readout strongly influences the difference in image intensities flow between regions supplied by stenosed coronary arteries (little or
between myocardium with normal and reduced perfusion, and the no flow increase) and territories supplied by normal arteries (multifold
optimal time for image acquisition is shown at location b in Fig. 14.19. increased flow) is observed as a difference in myocardial contrast agent
In practice, however, the time between saturation pulse and beginning concentration and thus image intensity (e.g., “hypoenhancement” pattern
of image acquisition is less than optimal (e.g., location a) to allow rapid seen in Fig. 14.16 [arrows]).
acquisition of multiple slices within one cardiac cycle. Fig. 14.20A displays the timeline for a comprehensive CMR stress
The basic concept of FLASH timing is shown in Fig. 14.17. The test. After scout and cine imaging, adenosine is infused for at least 2
pulse sequence starts with a slice-selective gradient on the slice encode minutes. This minimum infusion duration is chosen based on physi-
axis and an RF pulse. The RF pulse tips the longitudinal magnetization ological studies in humans demonstrating that, on average, maximum
(by the flip angle α) into the transverse plane to be received by the coronary blood flow is reached 1 minute after the start of adenosine
CMR coils. The flip angle is typically set to 20 to 30 degrees, which is infusion, and in virtually every patient after 2 minutes.23 Then a CMR-
optimized (“Ernst angle”) in the setting of the rapid train of RF pulses contrast agent such as Gd-DTPA (0.075 mmol/kg) is administered fol-
used in FLASH imaging. The FLASH pulse sequence creates an echo lowed by a saline flush (≈35 mL) at a rate of 4.5 mL/s. The adenosine
by first dephasing the spins using the negative read gradient lobe fol- infusion is stopped as soon as image acquisition is completed.
lowed by rephrasing using the first half of the positive read gradient Fig. 14.20B shows the specific steps performed during the stress
lobe. This creates an echo at the center of the readout gradient that is perfusion scan in more detail. Referring to Fig. 14.20B: (1) Adenos-
recorded by the CMR receiver coils. Following data collection, spoiler ine infusion is started. (2) Two minutes later the image acquisition
gradients are applied to destroy any remaining transverse magnetization commences. (3) Contrast agent is injected at a rate of 4.5 mL/s while
before the next RF excitation pulse is played. This is important for adenosine infusion continues. The patient still breathes freely. (4) As
avoiding image artifacts that can result from mixing of longitudinal soon as the contrast media reaches the RV, the patient is asked to hold
and transverse magnetization. their breath for the next 15 seconds to characterize the initial wash-in
For stress perfusion imaging, an adenosine dose of 140 µg/kg/min of contrast agent into the LV myocardium. (5) The remaining image
is typically used. The adenosine causes coronary vasodilation mediated acquisition is completed with shallow breathing. (6) Image acquisition
by the adenosine A2A receptor21 and has a short half-life of less than ends about 50 to 60 seconds after it began. This description assumes
10 seconds. Although symptoms such as chest discomfort can occur, that images are displayed on the scanner in the real-time inline display
serious side effects (e.g., bronchospasm or atrioventricular block) are window to observe contrast agent arrival in the RV.
rare. With normal coronary arteries, adenosine can lead to an increase A wait time of about 15 minutes is required before the rest perfusion
in coronary blood flow of 4- to 5-fold compared with rest/baseline scan to allow the contrast agent to sufficiently clear from the blood
flow.22 In the presence of a significant coronary artery stenosis, this pool. During this time, additional cine scans and/or flow imaging for
large increase in blood flow is blunted. This is because the smaller valvular evaluation can be performed. For the rest perfusion scan, an
arterioles located in myocardium downstream of the stenosed vessel additional dose of 0.075 mmol/kg Gd-DTPA is given. The total scan
are already fully dilated at rest as a result of the autoregulation mecha- time for a comprehensive CMR stress test, including cines, stress and
nism that tries to maintain normal downstream flow in the presence rest perfusion, and LGE is ~45 minutes.
172 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Contrast injection 1

Contrast injection 2
2 min 1 min ≈ 15 min 1 min

Wait or do

perfusion

perfusion
Stress
additional cine

Rest
IMAGING

Scout Cine imaging or LGE


imaging imaging valvular evaluation imaging

0 1 8 15 17 25 33 40 45
Time (minutes)
A
Adenosine
Contrast 2 min 1 min
Saline flush
Breath holding
Image acquisition

B 1 2 3 4 5 6
FIG. 14.20  (A) Timeline for a comprehensive cardiovascular magnetic resonance stress test. (B) The specific
steps performed during the stress perfusion scan in more detail. Time points ❶–❻ are referenced in the
text. Note that the time scale is not linear. See text for details. LGE, Late gadolinium enhancement.

VIABILITY AND INFARCTION (“SCAN” = “LATE


GADOLINIUM ENHANCEMENT”)
The clinical implications of viability imaging are steadily growing. Besides
the assessment of acute and chronic myocardial infarction, LGE24 has
been used for the prediction of contractile improvement after revas-
cularization,25 for measuring the response to beta-blocker treatment,26
for the differentiation of ischemic from nonischemic cardiomyopathies,27
and for the diagnosis of various nonischemic cardiomyopathies.28–30
There is a large body of evidence that LGE can differentiate between
nonviable and viable tissue.24,31–34 Regions of irreversible injury exhibit
high signal intensity (hyperenhancement) on T1-weighted images after
administration of extracellular CMR contrast agent, such as Gd-DTPA.
The underlying mechanism of hyperenhancement is that in acutely
infarcted regions the ruptured myocyte membranes allow the extracel-
lular contrast agent to passively diffuse into the intracellular space,
resulting in an increased tissue-level contrast agent concentration, and
therefore hyperenhancement. Chronic infarcts consist of dense scar
with an increased interstitial space between collagen fibers (relative to
normal myocardium) and therefore an increased distribution volume
of the Gd-DTPA and, consequently, hyperenhancement.
The “goal” of LGE viability imaging is to create images with high
contrast between the hyperenhanced nonviable tissue and normal myo-
cardium for a clear delineation of these regions (Fig. 14.21). This is
currently best achieved by using a segmented inversion recovery
FIG. 14.21  The “goal” of late gadolinium enhancement viability imaging.
sequence.34 So far, the best validated sequence for viability imaging is
The short-axis cardiovascular magnetic resonance image provides a
the segmented inversion recovery TurboFLASH sequence.24,31–34 Fig. clear delineation between nonviable (hyperenhanced) and viable (dark)
14.22 displays the gating for a segmented inversion recovery sequence. myocardium.
The acquisition of multiple k-space lines in each cardiac cycle allows
reductions in imaging times to the point where the entire image can
be acquired during a single breath-hold of 6 to 10 seconds. The images
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 173

R R R

Heartbeat 1 Heartbeat 2
ECG

Trigger
TI TI

Segment 1 Segment 2

180° 180°
inversion inversion

k-space
FIG. 14.22  Electrocardiogram (ECG) gating of the segmented inversion recovery sequence. See text for
details.

Mz
Nonviable (infarcted) myocardium has
shorter T1 (recovers faster, looks brighter)
TI (normal)

+M0

TI
(infarct)

Time

Viable (normal) myocardium has longer


T1 (recovers slower, looks darker)
180° –M0
TI = inversion time
RF

Gradient

Spoil transverse
magnetization
Readout one segment
(when normal myocardium is nulled)
FIG. 14.23  Inversion recovery curve for viable and nonviable myocardium. See text for details. RF,
Radiofrequency.

are acquired in mid-diastole by using a trigger delay to minimize the during the rest interval of a traditional IR image) to determine the
cardiac motion. The magnetization of the heart is prepared by a non- polarity of the magnetization following an inversion pulse. Because
selective 180 degrees inversion pulse to create T1 weighting. The inver- normal myocardium has the longest T1 of the tissues normally imaged
sion time (TI) delay between inversion pulse and data collection (more (normal myocardium, enhanced myocardium, and blood), the PSIR
precise, the center of k-space) is chosen such that the magnetization algorithm renders the normal myocardium black, regardless of the
of viable (normal) myocardium is near its zero crossing, meaning that selected inversion time. A diagram of the pulse sequence and resulting
these regions appear dark (Fig. 14.23). Nonviable myocardium (acutely images are shown in Fig. 14.24.
infarcted or scar), however, appears bright because of the shorter T1
(faster signal recovery curve in areas of Gd-DTPA; see Fig. 14.23). It is FLOW/VELOCITY IMAGING
important to manually adjust the TI for each image to null normal
myocardium to account for wash-out kinetics of the contrast agent.35
(“SCAN” = “FLOW/VELOCITY”)
In recent years phase-sensitive inversion recovery (PSIR) sequences At the DCMRC, velocity-encoded (VENC) CMR imaging of blood
have been developed which relax the stringent constraint of selecting flow is usually performed to measure velocities in arteries, veins, valves,
the appropriate TI on an LGE sequence.36,37 The challenge with tradi- and shunts. Typically, the overall goal is to evaluate the severity of an
tional LGE imaging is the selection of the correct null time for normal aortic valve stenosis or an intracardiac shunt (e.g., atria or ventricular
myocardium, which is continually changing as contrast is being cleared septal defect). Furthermore, cardiac stroke volume can be calculated by
from the body. PSIR imaging uses an extra reference scan (acquired summing up the area under the flow/velocity curve of the ascending aorta.
174 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

ECG

IR
Trigger

Reference
Readout readout

Trigger delay Inversion time


Magnitude
PSIR

FIG. 14.24  Pulse sequence diagram for phase-sensitive inversion recovery (PSIR). A nonselective inversion
pulse (IR) is played after a trigger delay followed by a readout acquired after an additional inversion time
delay. A second readout (reference readout) is acquired during the rest heartbeat. The combination of these
two images restores the polarity of the inversion recovery image. This results in a phase-sensitive readout
that is resistant to improper inversion time. Magnitude images (top row) show different tissues being nulled
at different inversion times (left, too short; center, correct; right, too long). Below are the PSIR images
showing dark or “nulled” myocardium regardless of inversion time selected. ECG, Electrocardiogram.

The “goal” of VENC imaging is to obtain a cine series of gray-scale of flow; in this case flow in the opposite direction would appear darker
images that cover the entire cardiac cycle and where the gray level is than the stationary tissue (black to dark gray).
proportional to the velocity in the measured plane. Both in-plane and Analogous to cine imaging, each VENC image corresponds to a
through-plane flow can be assessed. Fig. 14.25 shows such a series in cardiac phase, and cardiac gating is required. The sequence is retrospec-
the upper row referenced to the electrocardiogram (ECG), and the cor- tively gated to cover the entire cardiac cycle. This is important for the
responding anatomical cine frames in the lower row. The VENC image determination of cardiac output, because prospective triggering, which
exhibits bright image intensity where the stenotic aortic valve opens tends to miss the end of diastole, would result in erroneous volumes.
(high velocity) and a gray intensity where no flow is present (stationary The physics of VENC are complex.38,39 Simply put, in the presence
tissue, aortic sinuses). Flow/velocity imaging can also encode direction of a magnetic field gradient the spins in the image plane acquire a phase
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 175

ECG

Velocity-encoded images

Cine images at corresponding times

FIG. 14.25  The “goal” of cardiovascular magnetic resonance flow (velocity-encoded [VENC]) imaging.
A selection of VENC images at the level of the aortic valve exhibiting stenosis is shown in the middle. Images
are registered with the electrocardiogram (ECG) on top, and corresponding cine frames on the bottom are
included as anatomic reference.

that is proportional to the area under the gradient plotted in a pulse VENC imaging. Occasionally, it may be done in conjunction with a
sequence diagram like the one depicted in Fig. 14.26. For nonmoving more detailed cardiovascular examination (e.g., pulmonary vein imaging,
spins, this phase can be rewound (the spins can be rephased) by a aortic root angiography). In this section we discuss some practical issues
gradient of the same magnitude but opposite sign. If, however, the spins in regards to ECG-gated contrast-enhanced MR angiography (MRA).
change position between experiencing the first gradient and the rewind- We do not discuss different angiography techniques, vessel wall or
ing gradient, as is the case for spins in flowing blood, then they experi- coronary imaging, or advanced forms of motion correction (“navigator
ence a gradient during rewinding that is not only opposite in sign but echoes”).
also different in magnitude. This incomplete or excessive rewinding The “goal” of MRA is to create a three-dimensional (3D) dataset
causes a phase difference that is a linear function of velocity. This phase of a vessel or vascular bed of interest. Use of a T1-shortening para-
difference is displayed as a gray-scale image representing the velocity magnetic contrast agent within the blood pool allows the genera-
of flowing blood (Fig. 14.25, top row of images). tion of high signal within the vascular tree relative to surrounding
Because the phase of spins is also dependent on many other factors, tissue, thus creating a “luminogram.” Fig. 14.27 shows two examples
such as the properties of receiver coils, it is necessary to measure a base of contrast-enhanced angiograms. The images were colored during
phase during flow-insensitive gradient waveforms (see Fig. 14.26, left dotted routine postprocessing. Fig. 14.27A depicts the LV, the aortic arch,
ellipse). The flow-sensitized waveform is shown in the right dotted ellipse. By the descending aorta, the truncus brachiocephalicus, and carotid and
subtracting the base phase from the flow phase and displaying the difference subclavian arteries. Fig. 14.27B shows pulmonary arteries and veins.
on a gray-level scale, the images of Fig. 14.25, upper row, are obtained. The The timing was chosen such that the LV and the aorta were not yet
grey level is proportional to velocity: black corresponds to a maximum filled by contrast-rich blood and are thus not visible. This “proce-
backward flow, white to a maximum forward flow, and gray corresponds dure” would commonly be done as part of a complete evaluation
to no flow. The range from maximum backward to maximum forward of a patient with vascular disease, which includes multiple different
flow can be quantified in cm/s and must be adjusted by the scanner opera- scans from the examination menu in addition to the MRA. Fig. 14.28
tor to maximize sensitivity to differing physiologic blood flow velocities. shows the timeline of the procedure. The noncontrast scan is done
The images allow both a coarse overview and a quantitative assess- for later subtraction from the contrast-enhanced scan to eliminate
ment of abnormal flow patterns. background.
Unlike for lower extremity MRA, ECG gating is critical when imaging
vascular structures that experience motion because of cardiac move-
ANGIOGRAPHY (“SCAN” = “ANGIOGRAPHY”) ment or pulsatile blood flow (e.g., aortic root). In these situations it is
Vascular angiography is often performed independently of stress or important to acquire imaging data only during diastole (see Fig. 14.7)
viability tests. However, this scan is quite frequently combined with because motion even during just a portion of the acquisition will adversely
176 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

ADC
Time

RF

Read

Phase

Slice

Flow-compensated block Flow-sensitive block


FIG. 14.26  Pulse sequence timing diagram of the velocity-encoded sequence. Gradients on the read axis
are flow compensated. On the slice axis two gradient waveforms are shown inside the dotted ellipses; one
that is insensitive to flow (flow-compensated block) and one that is sensitive toward flow (flow-sensitized
block). The figure shows the sequence necessary to acquire one line of data. This is an example for assess-
ment of through-plane flow, but in-plane flow can be imaged as well. ADC, Analog-to-digital converter; RF,
radiofrequency.

Ao

LA
RV

A B
FIG. 14.27  Three-dimensional contrast-enhanced magnetic resonance angiography multiplanar reconstruction
of the aortic root and thoracic aorta. This image was electrocardiogram-gated for high resolution of the aortic
root in a young woman with a history of a bicuspid aortic valve and aortic root dilatation. (A) Anterior view
showing the ventricles, aortic arch, and branch vessels. (B) Posterior view showing the pulmonary arteries
and veins and the left atrium (LA). Ao, Aorta; RV, right ventricle.

affect image quality. In the case of highly dynamic structures such as along the slice-encoding direction to encode the additional dimension.
the ascending aorta, we have also found it important to not only account The data required for the 3D acquisition are much larger than the
for the systolic cycle length but also the period of time in early diastole two-dimensional (2D) FLASH sequence and results in longer scan times
when passive relaxation of the aorta occurs as a result of the elastic (~15–20 seconds). Using parallel imaging techniques, more elegant
recoil of the vessel. This short period of “diastolic vessel relaxation” k-space sampling, and faster gradient sets has decreased scan times
leads to appreciable vascular motion that can blur the edges of an significantly. Using state-of-the-art hardware and newer sequences,
angiogram and compromise its quality. These considerations were taken high-resolution ECG-gated contrast-enhanced MRA can be completed
into account for obtaining the images of Fig. 14.27 because they would within a single breath-hold.
otherwise not be as crisp. Contrast-enhanced 3D MRA is the most common type of angiog-
For contrast-enhanced MRA the most commonly used pulse sequence raphy we use on the clinical service. We have found this scan to be
is a fast 3D spoiled gradient echo sequence. This is a variant of the easily incorporated into the clinical CMR examination without signifi-
FLASH technique that was discussed previously (see Fig. 14.17). The cantly changing the duration of the study or compromising the quality
3D data acquisition requires that an additional gradient be applied of the other components.
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 177

CONTRAST

Contrast
injection
≈ 3 min

OPTIONAL
IMAGING
Selected
bright-
blood
imaging

≈ 2 min ≈ 6 min ≈ 5 min ≈ 5 min

Scout HASTE Noncontrast 3D


3D angio
IMAGING

imaging (2–3 planes) CE-MRA


(GRE) (arterial +/–
venous phase)

0 1 6 12 16 23
Time (min)
FIG. 14.28  Timeline of a typical contrast-enhanced magnetic resonance angiography (CE-MRA). Note that
the time scale is not linear. In addition to scouts, half-Fourier acquisition single-shot turbo spin echo (HASTE)
and selected bright-blood balanced steady-state free precession images are obtained for analysis of extra-
vascular structures and vascular structures which may not be in the angiographic field-of-view. A noncontrast
three-dimensional (3D) angiogram is done before the contrast angiogram so that a subtraction image can be
generated. A comprehensive study can be completed in 20 to 25 minutes. GRE, Gradient recalled echo.

Scout LV cine Perfusion LGE Flow/


Scout Morphology Angiography
velocity
FIG. 14.29  Combination of menu items for the evaluation of a patient
with coronary artery disease. This is the Duke cardiovascular magnetic FIG. 14.30  Combination of menu items for evaluation of the aorta.
resonance center (DCRMC) Cardiovascular Magnetic Resonance (CMR) Magnetic resonance angiography is the third-largest cardiovascular mag-
Stress Test, which comprises approximately 50% of our 4000 annual netic resonance procedure at our institution, after stress testing and
procedures. The DCMRC CMR Viability Test (another 25% of our volume) viability.
is identical, except that perfusion imaging is not performed. LGE, Late
gadolinium enhancement; LV, left ventricular.

In summary, although the definition of routine clinical CMR con-


tinues to evolve at our, and other, institutions, we have found it very
CONCLUSION
useful to assemble a short list of predefined individual CMR scan pro-
The overall goal of this chapter was to describe an approach to routine tocols (the “exam menu”; see Fig. 14.3) from which one can create a
clinical CMR that is based on selecting one or more scans from an “test package” specifically tailored to the diagnostic question. We have
examination menu and assembling these into a test. Fig. 14.29 shows found that this approach substantially decreases the time needed to
how one would assemble a test for a patient referred for the evaluation scan because the operator does not have to select amongst the literally
of coronary artery disease. Specifically, one would perform scouting, hundreds of buttons on the scanner console while the patient waits
cine imaging, perfusion imaging at stress and rest, and finally LGE idly inside the magnet (the buttons are predefined for each menu item).
imaging. The protocol of Fig. 14.29 is, in fact, identical to the CMR We have found that this approach significantly improves the throughput
Stress Test1 discussed in the introductory section of this chapter. Impor- of our clinical CMR service. Perhaps more importantly, however, we
tantly, and as previously noted, this test now comprises approximately have simultaneously found that this approach dramatically reduces the
50% of our 4000 annual CMR procedures. Similarly, Fig. 14.30 sum- historically steep learning curve for new physicians interested in learn-
marizes how one would approach CMR scanning of a patient referred ing CMR.
for the evaluation of aortic disease. Additional information regarding
the latest standards in CMR protocols is continually being updated
through the Society for Cardiovascular Magnetic Resonance consensus
REFERENCES
document.40 A full reference list is available online at ExpertConsult.com
CHAPTER 14  Clinical Cardiovascular Magnetic Resonance Imaging Techniques 177.e1

REFERENCES 19. Nandalur KR, Dwamena BA, Choudhri AF, Nandalur MR, Carlos RC.
Diagnostic performance of stress cardiac magnetic resonance imaging in
1. Klem IHJ, Shah DJ, Sketch MH Jr, et al. Improved detection of coronary the detection of coronary artery disease: a meta-analysis. J Am Coll
artery disease by stress perfusion cardiovascular magnetic resonance with Cardiol. 2007;50:1343–1353.
the use of delayed enhancement infarction imaging. J Am Coll Cardiol. 20. Kellman P, Arai AE. Imaging sequences for first pass perfusion—a review.
2006;47(8):1630–1638. J Cardiovasc Magn Reson. 2007;9(3):525–537.
2. Oppelt A, Graumann R, Barfuss H, et al. Eine neue schnelle Pulssequenz 21. Udelson JE, Heller GV, Wackers FJ, et al. Randomized, controlled
fuer die Kernspintomographie. Electromedica. 1986;54:15–18. dose-ranging study of the selective adenosine A2A receptor agonist
3. Bellenger NG, Burgess MI, Ray SG, et al. Comparison of left ventricular binodenoson for pharmacological stress as an adjunct to myocardial
ejection fraction and volumes in heart failure by echocardiography, perfusion imaging. Circulation. 2004;109(4):457–464.
radionuclide ventriculography and cardiovascular magnetic resonance; 22. Wilson RF, Wyche K, Christensen BV, et al. Effects of adenosine on
are they interchangeable? Eur Heart J. 2000;21(16):1387–1396. human coronary arterial circulation. Circulation. 1990;82(5):1595–1606.
4. Grothues F, Moon JC, Bellenger NG, et al. Interstudy reproducibility of 23. Rossen JD, Quillen JE, Lopez AG, et al. Comparison of coronary
right ventricular volumes, function, and mass with cardiovascular vasodilation with intravenous dipyridamole and adenosine. J Am Coll
magnetic resonance. Am Heart J. 2004;147(2):218–223. Cardiol. 1991;18(2):485–491.
5. Grothues F, Smith GC, Moon JC, et al. Comparison of interstudy 24. Wagner A, Mahrholdt H, Holly TA, Elliot M, Kim RJ, Judd RM. MRI
reproducibility of cardiovascular magnetic resonance with two-dimensional detects subendocardial infarcts which are missed by SPECT. Lancet.
echocardiography in normal subjects and in patients with heart failure or 2003;361(9355):374–379.
left ventricular hypertrophy. Am J Cardiol. 2002;90(1):29–34. 25. Kim RJ, Wu E, Rafael A, et al. The use of contrast-enhanced magnetic
6. Bellenger NG, Rajappan K, Rahman SL, et al. Effects of carvedilol on left resonance imaging to identify reversible myocardial dysfunction. N Engl J
ventricular remodelling in chronic stable heart failure: a cardiovascular Med. 2000;343:1445–1453.
magnetic resonance study. Heart. 2004;90(7):760–764. 26. Bello D, Shah DJ, Farah GM, et al. Gadolinium cardiovascular magnetic
7. Friedrich MG, Dahlof B, Sechtem U, et al. Reduction (TELMAR) as resonance predicts reversible myocardial dysfunction and remodeling in
assessed by magnetic resonance imaging in patients with mild-to- patients with heart failure undergoing beta-blocker therapy. Circulation.
moderate hypertension—a prospective, randomised, double-blind 2003;108:1945–1953.
comparison of telmisartan with metoprolol over a period of six months 27. McCrohon JA, Moon JC, Prasad SK, et al. Differentiation of heart failure
rationale and study design. J Renin Angiotensin Aldosterone Syst. related to dilated cardiomyopathy and coronary artery disease using
2003;4(4):234–243. gadolinium-enhanced cardiovascular magnetic resonance. Circulation.
8. Lamb HJ, Beyerbacht HP, de Roos A, et al. Left ventricular remodeling 2003;108:54–59.
early after aortic valve replacement: differential effects on diastolic 28. Mahrholdt H, Wagner A, Judd RM, Sechtem U, Kim RJ. Delayed
function in aortic valve stenosis and aortic regurgitation. J Am Coll enhancement cardiovascular magnetic resonance assessment of
Cardiol. 2002;40(12):2182–2188. non-ischaemic cardiomyopathies. Eur Heart J. 2005;26:1461–1474.
9. Ioannidis JP, Trikalinos TA, Danias PG. Electrocardiogram-gated single- 29. Mahrholdt H, Goedecke C, Wagner A, et al. Cardiovascular magnetic
photon emission computed tomography versus cardiac magnetic resonance resonance assessment of human myocarditis: a comparison to histology
imaging for the assessment of left ventricular volumes and ejection fraction: and molecular pathology. Circulation. 2004;109:1250–1258.
a meta-analysis. J Am Coll Cardiol. 2002;39(12):2059–2068. 30. Choudhury L, Mahrholdt H, Wagner A, et al. Myocardial scarring in
10. Kjaergaard J, Petersen CL, Kjaer A, et al. Evaluation of right ventricular asymptomatic or mildly symptomatic patients with hypertrophic
volume and function by 2D and 3D echocardiography compared to MRI. cardiomyopathy. J Am Coll Cardiol. 2002;40:2156–2164.
Eur J Echocardiogr. 2006;7(6):430–438. 31. Kim RJ, Fieno DS, Parrish TB, et al. Relationhip of MRI delayed contrast
11. Persson E, Carlsson M, Palmer J, et al. Evaluation of left ventricular enhancement to irreversible injury, infarct age, and contractile function.
volumes and ejection fraction by automated gated myocardial SPECT Circulation. 1999;100:1992–2002.
versus cardiovascular magnetic resonance. Clin Physiol Funct Imaging. 32. Wu E, Judd RM, Vargas JD, Klocke FJ, Bonow RO, Kim RJ. Visualisation
2005;25(3):135–141. of presence, location, and transmural extent of healed Q-wave and
12. Schaefer WM, Lipke CS, Standke D, et al. Quantification of left non-Q-wave myocardial infarction. Lancet. 2001;357:21–28.
ventricular volumes and ejection fraction from gated 99mTc-MIBI 33. Fieno DS, Kim RJ, Chen EL, Lomasney JW, Klocke FJ, Judd RM.
SPECT: MRI validation and comparison of the Emory Cardiac Tool Box Contrast-enhanced magnetic resonance imaging of myocardium at risk:
with QGS and 4D-MSPECT. J Nucl Med. 2005;46(8):1256–1263. distinction between reversible and irreversible injury throughout infarct
13. Thorley PJ, Plein S, Bloomer TN, et al. Comparison of 99mTc healing. J Am Coll Cardiol. 2000;36:1985–1991.
tetrofosmin gated SPECT measurements of left ventricular volumes and 34. Simonetti OP, Kim RJ, Fieno DS, et al. An improved MR imaging technique
ejection fraction with MRI over a wide range of values. Nucl Med for the visualization of myocardial infarction. Radiology. 2001;218:215–223.
Commun. 2003;24(7):763–769. 35. Wagner A, Mahrholdt H, Thomson L, et al. Effects of time, dose and
14. Corsi C, Lang RM, Veronesi F, et al. Volumetric quantification of global inversion time for acute myocardial infarct size measurements based on MRI
and regional left ventricular function from real-time three-dimensional delayed contrast enhancement. J Am Coll Cardiol. 2006;47(10):2027–2033.
echocardiographic images. Circulation. 2005;112(8):1161–1170. 36. Kellman P, Arai AE, McVeigh ER, Aletras AH. Phase-sensitive inversion
15. Giang TH, Nanz D, Coulden R, et al. Detection of coronary artery disease recovery for detecting myocardial infarction using gadolinium-delayed
by magnetic resonance myocardial perfusion imaging with various hyperenhancement. Magn Reson Med. 2002;47:372–383.
contrast medium doses: first European multi-centre experience. Eur 37. Kellman P, Dyke CK, Aletras AH, McVeigh ER, Arai AE. Artifact
Heart J. 2004;25(18):1657–1665. suppression in imaging of myocardial infarction using B1-weighted
16. Paetsch I, Jahnke C, Wahl A, et al. Comparison of dobutamine stress phased-array combined phase-sensitive inversion recovery. Magn Reson
magnetic resonance, adenosine stress magnetic resonance, and adenosine Med. 2004;51:408–412.
stress magnetic resonance perfusion. Circulation. 2004;110(7):835–842. 38. Debatin JF, et al. Renal artery blood flow: quantitation with phase-
17. Wolff SD, Schwitter J, Coulden R, et al. Myocardial first-pass perfusion contrast MR imaging with and without breath holding. Radiology.
magnetic resonance imaging: a multicenter dose-ranging study. 1994;190:371–378.
Circulation. 2004;110(6):732–737. 39. Pelc LR, et al. Arterial and venous blood flow: noninvasive quantitation
18. Lipinski MJ, McVey CM, Berger JS, Kramer CM, Salerno M. Prognostic with MR imaging. Radiology. 1992;185:809–812.
value of stress cardiac magnetic resonance imaging in patients with 40. Kramer CM, Barkhausen J, Flamm SD, Kim RJ, Nagel E. Standardized
known or suspected coronary artery disease: a systematic review and cardiovascular magnetic resonance (CMR) protocols 2013 update. J
meta-analysis. J Am Coll Cardiol. 2013;62:826–838. Cardiovasc Magn Reson. 2013;15:91.
15 
Normal Cardiac Anatomy
Michael L. Chuang, Warren J. Manning, and Ronald M. Peshock

Cardiovascular magnetic resonance (CMR) can be used to obtain images be applied to emphasize or de-emphasize the signal contribution of
of the heart in any plane. Thus to define normal anatomy and function, specific tissues. For example, in the evaluation of arrhythmogenic right
it is useful to define standard imaging planes to develop knowledge of ventricular cardiomyopathy (ARVC), it is important to obtain high-
normal anatomy, anatomic variants, and potential artifacts. Standard resolution spin echo images of the anterior right ventricular (RV) free
CMR planes have evolved from other imaging modalities, including wall that are free from respiratory artifact (see Chapter 7). This goal
body computed tomography (CT) imaging, echocardiography, and can be achieved by using a surface coil to improve signal-to-noise
x-ray contrast angiography; consistent nomenclature across imaging ratio (SNR) compared with the standard body coil, and through the
modalities is important1 for accurate and unambiguous communica- use of spatial and/or fat saturation to reduce artifacts from blood
tion. The problem is often one of determining the appropriate plane and chest wall motion. Breath-hold, double inversion recovery spin
as rapidly as possible to make the diagnosis. As with other cardiac echo techniques can also be very effective in removing respiratory
imaging techniques, it is important to know as much as possible artifacts.
regarding the clinical question before determining the protocol. All Imaging planes oriented with respect to the principal axes of the
examinations therefore should be planned to answer a specific clinical body are particularly useful in the evaluation of the aorta, pericar-
question. dium, anterior RV free wall, and paracardiac masses. Coronal images
The basic imaging planes can be grouped into planes oriented with can be useful because they present tomographic information in an
respect to the heart, such as horizontal and vertical long axis and short orientation similar to the chest x-ray, which is familiar to most clini-
axis, and planes oriented with respect to the major axes of the body, cians (see Fig. 15.2A and B). In general, axial planes are also useful
such as the transaxial, sagittal, and coronal. Cardiac-oriented planes because they are familiar orientations from CT (see Fig. 15.2C to H).
are essential for evaluation of cardiac chamber size and function and Specific vascular structures of interest that can be evaluated well with
are familiar from other cardiac imaging techniques. With CMR the axial imaging include the thoracic aorta and its branches, the pulmo-
position of these planes can be prescribed very accurately. As shown nary artery and veins, and the superior vena cava (see Fig. 15.2C and
in Fig. 15.1A, a breath-hold scout series in the coronal, axial, and sagittal D). Axial images through the heart can be particularly useful in the
plane is the usual starting point. An axial scout (Fig. 15.1B) is used to evaluation of the pericardium and RV free wall (see Fig. 15.2E to H).
define the vertical long axis (also known as the two-chamber view, Fig. They are of limited value in the assessment of LV wall thickness and
15.1C). The horizontal long axis (Fig. 15.1D), which depicts both atria chamber size because of the variable orientation of the LV relative to
and both ventricles but is slightly different from the true four-chamber the principal axes of the body. Sagittal images are, in general, the least
view, is then planned, and followed by the short axis (Fig. 15.1E and familiar to clinicians and can be more difficult to interpret (see Fig.
F), which can be used to generate the left ventricular (LV) outflow tract 15.2I and J). However, sagittal images are useful in depicting the RV
view (Fig. 15.1G), which is similar to the parasternal long-axis view of outflow tract and are therefore helpful in the evaluation of patients
transthoracic echocardiography. with congenital heart disease and ARVC. Oblique sagittal planes are
The main structures of normal cardiac anatomy in the coronal, useful in the evaluation of the thoracic aorta, and these planes can
axial, and sagittal planes are shown for spin echo sequences in Fig. be easily defined from the transaxial images, especially if three-point
15.2A to J. There are many CMR atlases of cross-sectional anatomy plane definition is available using the arch and lower ascending and
that can be helpful,2,3 and on-line resources (e.g., atlas.scmr.org) with descending aorta as the reference points (Fig. 15.2K and L). Black-
interactive learning of the cross-sectional anatomy can be very useful blood images (Fig. 15.2M to P) oriented along the functional axes
teaching aids. The reader is recommended to refer to these for further introduced in Fig. 15.1 are useful in the definition and tissue character-
details. From the standpoint of tissue characterization, the spin echo ization of intracardiac and paracardiac masses. In addition, depiction
images typically permit the differentiation of fat (white) from muscle of these planes with double-inversion recovery black-blood imaging
(intermediate gray). Black regions in spin echo CMR studies represent is useful for characterization of valvular disease and the coronary
several tissues or materials, including air, bone, fibrous tissue, metal, artery wall.4–6
or rapidly moving blood. Note that if fluid moves relatively slowly (for Myocardial function is typically assessed using cine balanced steady-
example, in an aneurysm), its signal intensity will increase, which can state free precession (bSSFP) imaging, which has supplanted the older
mimic more solid tissue such as thrombus. segmented gradient recalled echo (GRE) cine methods. The bSSFP
The placement of imaging planes, slice thickness, and in-plane provides improved contrast between blood pool and myocardium,7
resolution are determined by the size of the structure of interest. As particularly in the presence of impaired ventricular function because
has been indicated in previous chapters, presaturation bands can be it is mainly dependent on T1/T2 ratio rather than inflow of unsaturated
added to remove specific artifacts. Other preparatory prepulses can protons. Both bSSFP and GRE cine methods depict blood as bright

178
CHAPTER 15  Normal Cardiac Anatomy 179

A B

FIG. 15.1  (A) Scout image 1, coronal: typical


breath-hold image used to begin study (alter-
natively a sagittal image could be used). The
white line indicates the location of an axial
image used to locate the mitral valve plane
and interventricular septum. (B) Scout image
2, axial: typical breath-hold image obtained to
C D set up a vertical long-axis (VLA) image. The
white line indicates the position of the VLA
imaging plane and is drawn to pass through
the middle of the mitral valve and the ventricular
apex. (C) Vertical long-axis image from breath-
hold balanced steady-state free precession
(bSSFP) cine cardiovascular magnetic resonance
(CMR) oriented as described above. The white
line indicates the position of the horizontal
long-axis (HLA) imaging plane and is selected
to pass through the left ventricular (LV) apex
and between the attachment points of the
mid-mitral annulus. (D) Horizontal long-axis
breath-hold bSSFP cine CMR image oriented
based on the prescription in panel C. The white
lines indicate the positions of a stack of images
E F oriented in the LV short-axis orientation, which
will be obtained next. (E) A representative end-
diastolic breath-hold bSSFP cine mid-ventricular
short-axis image of the stack shown in panel
D. The white line perpendicular to the imaginary
line between the insertion points of the right
ventricular free wall is used to select the
imaging plane to obtain a four-chamber view
of the heart. (F) Basal image in the LV short-
axis orientation showing an oblique view of
the aortic valve. The white line shows the
orientation of the LV outflow tract (LVOT) view.
(G) An LVOT view at end diastole. This imaging
plane is comparable to the parasternal long-axis
view of transthoracic echocardiography. (H)
G Four-chamber breath-hold bSSFP cine CMR
image at end diastole. This view is similar to
H the HLA view, but typically less of the aortic
outflow tract is seen.
180 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Ascending
aorta
Pulmonary
artery

Left
RVOT ventricle
Right
Left
atrium
ventricular
apex

A B

Ascending
aorta

Superior Superior
vena cava vena cava Main
Transverse pulmonary
aortic arch artery

Trachea

Descending
aorta
C D

Pericardium Pericardium
Epicardial
FIG. 15.2  (A) Coronal breath-hold double inversion recov- Right fat
ery, spin echo images. Fat is white, myocardium inter- ventricular
Tricuspid
mediate gray intensity, and blood is dark. The slice is Right atrial outflow RV
valve
positioned anteriorly and cuts through the right ventricle, appendage tract
RA LV
right ventricular outflow tract (RVOT), interventricular
septum, and left ventricular (LV) apex. (B) Coronal image LA
positioned more posteriorly than the image in panel A. Left Mitral
This shows the right atrium, left ventricle, ascending atrium valve
aorta, and pulmonary artery. The aortic valve leaflets are Pulmonary
vein
also seen. (C) Transverse conventional gated spin echo
image at the left of the transverse aortic arch. The trachea
and superior vena cava are also demonstrated. (D) Trans-
verse conventional gated spin echo image at the level E F
of the main pulmonary artery. The views in panels C and
D are useful in the evaluation of possible aortic dissec-
tion. (E) Transverse conventional spin echo image at the
level of the aortic valve. (F) Transverse conventional spin Right
echo image at the level of the interatrial septum. The ventricular
pericardium and epicardial fat are clearly demonstrated. wall
This view can be useful in evaluating atrial masses and
pericardial disease. (LA, Left atrium; LV, left ventricle;
RA, right atrium; RV, right ventricle.) (G) Transverse con-
Right atrium Inferior
ventional spin echo image at the level of the coronary Coronary vena cava Coronary
sinus. The RV wall, epicardial fat, and pericardium are sinus sinus
also demonstrated. This view can be helpful in evaluating
patients for constrictive pericarditis and arrhythmogenic
right ventricular cardiomyopathy. (H) Transverse conven-
tional spin echo image at the level of the entrance of
G H
the inferior vena cava into the right atrium.
CHAPTER 15  Normal Cardiac Anatomy 181

Ascending
aorta Right
pulmonary RVOT
artery Pericardium
Pericardium
Epicardial
Left atrium fat

Right atrium Right


ventricular wall

I J

Transverse
aortic arch

Descending
Ascending aorta
aorta

K L FIG. 15.2, cont’d  (I) Sagittal conventional spin echo


image obtained through the ascending aorta. The peri-
cardium is clearly demonstrated. This view can be helpful
in the evaluation of the ascending aorta and pericardium.
(J) Sagittal conventional spin echo image obtained through
RCA origin
the RVOT. This view can be helpful in evaluating the
pericardium, RVOT, and RV free wall. (K) Transverse
Aortic valve breath-hold double inversion recovery, spin echo images
leaflets
obtained at the level of the transverse portion of the
PDA
aortic arch (left) and the main pulmonary artery (right).
The white line indicates the position of a parasagittal
oblique plane used to obtain a “candy cane” view of
the aorta (next panel). (L) Parasagittal view of the aorta.
The ascending, transverse, and descending aorta are
seen in a single slice. The vessels to the head and neck
are also well seen. This view can be helpful in the evalu-
M N ation of aortic disease. (M) Long-axis view using breath-
hold double inversion recovery technique. This image is
Left comparable to the parasternal long-axis view in trans-
ventricular thoracic echocardiography. Both the right ventricle and
apex left ventricle are well demonstrated. The origin of the
right coronary artery (RCA) is seen in the fat of the anterior
atrioventricular (AV in O) groove. The aortic valve leaflets
are also well seen. This view can be useful in the evalu-
ation of hypertrophic cardiomyopathy with septal asym-
Anterior AV metry. (N) Short-axis view using breath-hold double
groove
Posterior inversion recovery technique. The LV and RV walls are
AV groove well demonstrated. In this image the posterior descend-
ing coronary artery (PDA) is also seen in cross section
in the posterior interventricular groove. (O) Four-chamber
view using breath-hold double inversion recovery tech-
nique. (P) Vertical long-axis or two-chamber view using
O P
breath-hold double inversion recovery technique.
Continued
182 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

Q R

FIG. 15.2, cont’d  (Q) End-diastolic image in the hori-


zontal long-axis orientation from a balanced steady-state
free precession (bSSFP) breath-hold cine cardiovascular
magnetic resonance sequence. The white lines indicate
the locations of LV short-axis imaging planes in subse-
quent panels R–T, all of which were obtained using
breath-hold cine bSSFP imaging. (R) Basal right ventricle
and left ventricle. The left anterior descending coronary
artery is seen in the anterior interventricular groove. (S)
End-diastolic short-axis image obtained at the mid left
ventricle level. (T) End-systolic image at the same imaging
S T
level as in panel S.

(white), whereas muscle is an intermediate gray, and air, bone, fibrous short-axis orientation; the optimal RV “short-axis” orientation may differ
tissue, and metal are dark. The main findings using bright-blood cine slightly, but the same view can be used for accurate and reliable RV
CMR of the heart are shown in Fig. 15.1C to H and Fig. 15.2Q to T. quantitation as well.
These cines are typically used to assess ventricular and valvular func- Coronary CMR requires yet another set of imaging planes to depict
tion. The LV outflow tract view, for example, is used to show the mitral the coronary arteries in tomographic slices when the targeted slab
and aortic valves (see Fig. 15.1G). One advantage of CMR is the ability approach is used. Alternatively, whole-heart methods which encom-
to precisely position the long-axis planes to avoid the foreshortening pass both ventricles and both coronary trees, yielding a volumetric
that commonly occurs in contrast ventriculography and two-dimensional dataset, may be employed. This subject is discussed in more detail in
(2D) echocardiography (see Fig. 15.1G and H). Short-axis views are Chapters 23 and 24.
planned from the long-axis views to span the entire LV. The short-axis
views in Fig. 15.2R to T are useful in the evaluation of biventricular
size and regional function. Using the same orientation to obtain views
ANATOMIC VARIANTS
of the atria can be useful for assessing atrial masses as well as chamber Given the ability to obtain images in many planes, be aware of normal
size and function. structures and anatomic variants that may complicate interpretation
To most accurately assess LV function and size, it is important of studies. Several potentially confusing features are highlighted here:
to obtain correctly oriented images that encompass the entire LV • Prominence of the lateral border of the right atrial (RA) wall (Fig.
throughout the cardiac cycle. To obtain true LV short-axis views, 15.3A). This structure is a prominence of the trabeculae carneae
we recommend the following steps. From an axial scout image, the and crista terminalis and does not represent an atrial mass.8
vertical long-axis or two-chamber view is obtained. The horizontal • Lipomatous hypertrophy of the interatrial septum (Fig. 15.3B). Fat
long-axis (HLA) view is planned from the two-chamber view, ensur- deposition in the interatrial septum is occasionally seen, particularly
ing that the imaging plane passes through the LV apex and through in the elderly. This process spares the region of the fossa ovalis and
the center of the mitral valve annulus. A stack of short-axis images thus leads to the characteristic dumbbell shape.9 This process is, in
is planned from the HLA view (see Fig. 15.1D), with the short-axis general, considered benign, but it is associated with atrial arrhythmias
planes perpendicular to an imaginary line passing through the LV apex in older patients. More severe and extensive lipomatous hypertrophy
distally and midway between the visualized portions of the mitral valve may extend well outside the heart. Imaging with and without fat
annulus basally. It is important to plan the short-axis stack so that it saturation readily characterizes this abnormality.
extends just distal to the apex and slightly above the base of the LV • Superior pericardial recess (Fig. 15.3C). The pericardium normally
to ensure its entire coverage. Failure to do so results in an incomplete extends up the ascending aorta, and this space may contain fluid.
dataset of limited use for accurate quantitative LV measurements. The This recess can be mistaken for aortic dissection or potentially an
stack of images obtained as described above will be oriented in the LV anomalous coronary vessel in coronary imaging.
CHAPTER 15  Normal Cardiac Anatomy 183

A B

C
FIG. 15.3  (A) Transverse gradient echo image at the level of the aortic valve obtained using respirator gating
with a navigator echo. A right atrial ridge is noted in the lateral wall of the right atrium (arrowhead). This
finding is normal and should not be mistaken for a right atrial mass. (B) Single frame from horizontal long-axis
balanced steady-state free precession cine cardiovascular magnetic resonance (CMR). Lipomatous hypertrophy
of the atrial septum is demonstrated (arrowhead). There is fatty infiltration of the septum that does not
involve the region of the fossa ovalis, resulting in the typical “dumbbell” appearance. (C) Oblique double
inversion recovery breath-hold CMR image obtained at the level of the right pulmonary artery. The extension
of the pericardial space both anterior and posterior to the ascending aorta is demonstrated (arrowheads).
The pericardial recess should not be mistaken for evidence of aortic dissection.

are some examples of commonly-encountered artifacts. Finally, many


COMMON VARIANTS
artifacts are amplified at higher field strengths, and protocols optimized
A number of artifacts related to CMR can complicate interpretation for 1.5 T often are not optimal for imaging at 3 T. However, these chal-
of the images. The acquisition time is often relatively long compared lenges can be addressed so as to benefit from the advantages offered
with physiologic processes, which leads to cardiac and respiratory motion by imaging at higher field strengths.10
artifacts. This problem must be recognized if present, and minimized
at the acquisition stage if possible. Also, because the strength of the Cardiac Motion Artifacts (Fig. 15.4A and B)
local magnetic field determines the position of an object in a CMR Except for single-shot echo planar imaging (EPI) or other real-time
image, if the local magnetic field is altered the position of the structure imaging approaches, CMR requires gating to the electrocardiogram
in the image is also altered. Therefore metal on or in the body can alter (ECG) or peripheral pulse. Problems with gating can result in ghosting
the local magnetic field, leading to distortion and local signal loss. and other noise that degrades the quality of the images. In general,
Hydrogen nuclei in fat experience a slightly different magnetic field focused efforts to obtain the best ECG possible before beginning scan-
compared with hydrogen nuclei in water molecules because of the local ning will minimize cardiac motion artifacts and save time. Surprisingly
chemical environment. This chemical shift is used in CMR spectroscopy good-quality images can be obtained in patients with atrial fibrillation,
to differentiate one compound from another. However, in CMR, this which may be related to the relatively consistent length of systole rela-
results in what is known as a chemical shift artifact at the interface of tive to changes in heart rate.11 Ventricular bigeminy often results in
water and fatty tissues. This artifact results from sharing of fat and poor images, in that every other beat is activated differently, resulting
water components within a pixel, leading to signal cancellation. Below in combining data from two different activation patterns. Many CMR
184 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A B

FIG. 15.4  (A) Artifacts due to respiration and


poor gating. In this gated spin echo image there
is mottling of the ventricular wall and loss of
C D
edge sharpness. (B) The same image as in A,
but with the window and level adjusted to
accentuate the artifact. There are ghosts of
the chest wall related to respiratory motion
and additional artifact over the heart as a result
of poor electrocardiographic gating. (C) Metal
artifact. The upper images were obtained with
a safety pin present on the anterior subject’s
gown. The resultant signal void is very evident.
The bottom row shows corresponding images
after removal of the safety pin. Distortion from
metal artifact is markedly more prominent/larger
in the gradient recalled echo images (right
column) than in the spin echo images (left E F
column). (D) Plain-film x-ray showing sternal
wires (dashed arrow) and metallic coronary
artery bypass graft (CABG) markers (solid arrow)
in a patient with prior CABG surgery. (E) Artifact
from sternal wires (dashed arrow) and CABG
markers (solid arrow) on T1-weighted spin echo
cardiovascular magnetic resonance imaging.
(F) Signal voids (arrows) in two views of a bio-
prosthetic aortic valve replacement by breath-
hold cine balanced steady-state free precession
imaging. The artifact results from the nonor-
ganic struts. (G) Metal in bileaflet mitral valve
prosthesis produces signal voids (arrows). (H)
There is minimal artifact from the tricuspid
(dashed arrow) and mitral (solid arrow) annu- G H
loplasty rings.
CHAPTER 15  Normal Cardiac Anatomy 185

J K
FIG. 15.4, cont’d  (I) Metal artifact from a coronary artery stent in the left anterior descending coronary
artery (arrow) seen on a scout image. (J) Chemical shift artifact. The image on the left is done with a relatively
short signal acquisition time (wide bandwidth). The image on the right is done with a longer signal acquisition
time (narrow bandwidth). This display accentuates the effect of the difference in chemical shift of water and
fat, creating the artifactual space between the aortic wall at fat (arrow). (K) Chemical shift artifact in echo
planar imaging (EPI). In EPI, the chemical shift artifact occurs in the frequency-encoding direction (right to
left in these images). The image on the left is obtained using a multishot EPI sequence with a relatively
short EPI acquisition with each shot. The chemical shift artifact is indicated by the white line in the posterior
chest wall. The image on the right is obtained using fewer shots with a longer EPI acquisition. The chemical
shift is larger, as indicated by the longer white line posteriorly. The image is degraded by superimposition
of anterior subcutaneous fat onto the heart. This problem can be addressed by adding fat saturation to the
sequence.

systems provide arrhythmia rejection in an attempt to reduce these 15.4).14,15 These methods accept cardiac cycles only during some portion
effects; however, use of these tools generally results in increased scan of the respiratory cycle. Respiratory gating can substantially improve
time because of rejection of cardiac cycles. Vectorcardiographic tech- image quality but increases total scan time. Real-time self-gating methods
niques, which exploit the difference between the normal vector and the with continuous data acquisition are gaining increasing interest but are
vector of the artifact from the magnetohydrodynamic effect, have greatly not yet in the clinical realm.15a
facilitated reliable ECG gating.12
Metal Artifact (Fig. 15.4C to I)
Respiratory Motion Artifacts (see Fig. 15.4A and B) Apart from safety considerations, pieces of metal outside or inside the
Respiration is associated with significant bulk cardiac motion. Motion body alter the local magnetic field and can result in artifacts. Patients
in the craniocaudal direction is on the order of a centimeter in normal must be screened carefully for the presence of metal, but despite vigi-
individuals.13 This motion can result in significant image degradation lance, objects common in the hospital may still go with the patient into
with ghosting and blurring, particularly in those with inconsistent the scanner. Fig. 15.4C shows an artifact related to a safety pin on the
respiratory patterns. Strategies to reduce respiratory artifact include patient’s gown. Note that signal loss and distortion are present in both
the use of breath-hold imaging, presaturation of the high-intensity the fast spin echo and GRE images. However, the severity of artifact is
signal from fat in the chest wall, and the use of respiratory gating. worse in the GRE images, severely compromising interpretation of the
Respiratory gating may be accomplished using a thoracic bellows or RV and interventricular septum. Fig. 15.4D and E shows the artifacts
by tracking the diaphragm position using a navigator echo (see Fig. related to sternal wires and coronary artery bypass graft markers. Fig.
186 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

15.4F shows the artifact related to a bioprosthetic aortic valve, whereas volumes. These changes can be estimated from unidimensional (linear)
Fig. 15.4G is a mechanical bileaflet mitral valve prosthesis. Fig. 15.4H measurements as in echocardiography, but with CMR, more accurate
shows the minimal artifacts associated with mitral and tricuspid annu- measures of chamber volume can be made using volumetric (three-
loplasty rings, and Fig. 15.4I depicts artifact from a stent in the left dimensional [3D]) methods. The 2D methods (e.g., the area-length
anterior descending (LAD) coronary artery. method or biplane formulas) have minimal advantages over echocar-
diography and fail to exploit the volumetric advantages of CMR.21 More
Chemical Shift Artifact (Fig. 15.4J to K) accurate measures, particularly in deformed ventricles, which do not
This artifact occurs because the hydrogen nuclei in fat experience a fit common geometric formula-based models, can be obtained using
slightly different magnetic field than hydrogen nuclei in water as a the summation of disks method. (In the cardiac imaging literature, this
result of the different local chemical environment.16 This process results method is commonly referred to as the “Simpson’s rule” method. The
in displacement of the fat signal in the frequency-encoding direction mathematical Simpson’s rule is a fourth-order polynomial approxi-
relative to water and is accentuated with narrow bandwidth sequences, mation for numerical integration22; failure to distinguish between the
which can present a diagnostic problem in spin echo imaging of patients mathematical and medical definitions of “Simpson’s rule” often leads
with suspected aortic dissection.17 It can be addressed by using a wider to confusion between clinicians and engineers or medical physicists.)
bandwidth sequence or repeating the sequence with frequency encoding With the summation of disks method, short-axis images are obtained
in the alternate direction, which will result in changing the orientation spanning the entire ventricle; the cross-sectional area in each slice is
of the artifact and thus help exclude the presence of an aortic dissection. measured, multiplied by the slice thickness (and interslice gap if appli-
In EPI, chemical shift effects lead to artifacts displaced in the phase- cable), and summed over the entire ventricle.23 This approach is accurate
encoding direction. As shown in Fig. 15.4K, this effect can be minimized and reproducible and is widely used both clinically and in research.
using multishot EPI techniques. In single-shot EPI with long acquisi- Regional LV systolic function can be assessed both qualitatively (“by
tion times, the chemical shift effects can be quite large. For this reason, eyeball” similar to echocardiography) or quantitatively. As shown in
single-shot EPI often employs fat saturation to suppress this artifact. Fig. 15.5, the standard long-axis, four-chamber, two-chamber, and short-
axis views can be mapped onto the standard 17-segment American
NORMAL CARDIAC SYSTOLIC AND Heart Association1 model for qualitative assessment of wall thickening.
Wall thickening can also be determined quantitatively using centerline
DIASTOLIC FUNCTION or other methods, and commercial software is available for such analyses.
The management of cardiovascular disease is critically dependent on Importantly, myocardial tagging methods24,25 can be used to quantify
the assessment of cardiac function. CMR provides highly accurate and myocardial contractility using strain without the need to identify endo-
reproducible assessments of global and regional cardiac function and cardial or epicardial borders explicitly. However, software for tracking
CMR is now widely considered as the clinical gold standard for the deformation of the tag lines over time nonetheless requires human
noninvasive evaluation of cardiac function, a standard by which other interaction, prolonging analysis times. The harmonic phase (HARP)
noninvasive methods are now validated. technique was proposed to obviate the human postprocessing issue by
An important consideration in determining function is the temporal computing strain based on local spatial frequency of the tag lines.26
resolution or frame rate of the cine CMR sequence. A frame rate of at With appropriate bandpass filtering and subsequent transformation of
least 25 frames/s (a temporal resolution of 40 ms/frame) is required to the k-space signal, strain can be extracted automatically. An alternative
identify end systole accurately. Historically, x-ray contrast left ventricu- method, displacement encoding with stimulated echoes (DENSE),27
lography has been obtained at a rate of 30 frames/s or a temporal reso- combines a stimulated echo with bipolar gradient to encode displace-
lution of 33 ms. The frame rate in echocardiography is dependent on ment, from which strain can be calculated. Strain-encoded (SENC)
speed of ultrasound in the body and the distance of the heart from the CMR measures through-plane strain and has been applied to stress
transducer, but typically is at least 30 frames/s (temporal resolution of CMR28 for assessment of both systolic and diastolic function.29,30 These
33 ms or better). Partially parallel imaging methods (e.g., SENSE, SMASH, methods have been reviewed in a number of articles31,32 and are detailed
GRAPPA) are now routinely used in clinical imaging to decrease acqui- in Chapters 14 and 22.
sition time and increase frame rate.18
Whereas the CMR cine loop appears to display a single cardiac cycle, Right Ventricle
the image data are most commonly acquired over multiple heartbeats. Historically, measurement of RV volumes by echocardiography has been
As a result, image quality can be markedly degraded in the presence of largely qualitative, because of the lack of standard geometric RV models.
arrhythmias or unreliable ECG triggering or gating. However, an abnor- A major advantage of CMR is that the same LV summation of disks
mal but regular ECG, such as bundle-branch block,18a typically does not approach may be readily applied to the right ventricle. Studies in ven-
adversely affect image quality. Cardiac function is typically assessed during tricular casts have shown an excellent correlation between CMR and
breath-holding, to minimize bulk cardiac translation, but if the goal is to displacement measurements.33
assess the effect of respiration on chamber size or function (e.g., tamponade
or constriction), then nonbreath-hold real-time methods19 may be more Stroke Volume
appropriate because such techniques acquire images during a fraction of Stroke volume is the amount of blood ejected from the ventricle with
a single heartbeat, and not as a composite over multiple cardiac cycles each cardiac cycle. It can be readily calculated by subtracting the end-
as with usual segmented k-space sequences. However, real-time CMR systolic volume from the end-diastolic volume. Multiplying the stroke
has lower spatial and temporal resolution, and may be more prone to volume by the heart rate yields the cardiac output, typically reported
artifacts, as compared with “standard” breath-hold approaches. Real-time in liters per minute. Studies comparing cardiac output determined by
imaging may also be particularly suitable for CMR-based exercise testing.20 CMR with invasive thermodilution methods have shown good correla-
tion.34,35 The cardiac output determined in this way from the stroke
Left Ventricle volume is the same as the cardiac output determined in the catheteriza-
Assessment of LV function includes global and regional function. Assess- tion laboratory. In the setting of aortic or mitral regurgitation, however,
ment of global LV function is based on measuring changes in chamber part of this volume does not result in the net delivery of blood to the
CHAPTER 15  Normal Cardiac Anatomy 187

Regional LV function

Mid- Bs-AS
Bs-Ant
Ap-AS AS Bs-AS

Ap Bs- Bs-
Sept Lat
Ap-Inf/ Bs-
Mid-
Lat
Inf/Lat Inf/Lat Bs-Inf Bs-Inf/Lat
Apex

Ap-Sept Mid- Mid-Ant


Ap-Lat AS
Mid-Sept
Mid- Mid- Mid-
Bs-Sept Lat Sept Lat
Bs-Lat
Mid-Inf Inf/Lat
Apex

Ap-Ant Ap-Sept Ap-Ant


Ap-Inf
Mid-
Mid-Inf Ant
Ap-Inf Ap-Lat
Bs-Inf Bs-Ant

A B
FIG. 15.5  (A) Images in each of the standard planes typically used for scoring wall thickening and segmental
function. The segments visualized correspond to those depicted in the standard American Heart Association
(AHA) 17-segment model. (B) AHA 17-segment left ventricular (LV) model. Ant, Anterior; Ap, apical; AS,
anteroseptal; Bs, basal; Inf, inferior; Lat, lateral; Sept, septal.

periphery. In this setting, the cardiac output based on the summation be more closely associated with LV diastolic function and have greater
of disks LV stroke volume is greater than the forward flow, which can prognostic value.43–45 In contrast to the ventricles, presently there is no
be measured using flow techniques (see Chapters 6 and 14), but the “standard method” for CMR assessment of atrial size and function,
regurgitant volume can be determined by subtracting the forward flow particularly for the RA, but results from selected studies are summarized
from the apparent stroke volume.36,37 in a recent review.46

Ventricular Mass Effect of Imaging Sequence and Magnetic Field


In comparison to clinical echocardiography, which typically assesses Strength on Ventricular Volumes and Mass, and
LV hypertrophy by wall thickness methods, total LV mass can be esti- Implications for Reference Standards
mated by CMR by measuring the volume of the myocardium. This Selecting bSSFP imaging is overwhelmingly the method of choice for
volume is then multiplied by the specific gravity of myocardium, typi- CMR assessment of ventricular function because it provides superior
cally taken as 1.05 g/mL. CMR provides highly accurate and reproducible contrast between blood pool and myocardium compared with GRE
estimates of LV mass over a broad range of heart sizes both in animals cine sequences. Perhaps as a result of this difference in delineation of
and in man.38,39 Intraobserver, interobserver, and interstudy variability endocardial contours, ventricular volumes and stroke volume by bSSFP
are excellent. However, it must be noted that LV mass determined using imaging are slightly but systematically greater than corresponding values
linear measurements and cubed-power geometric formulas generally measured using cine GRE methods.47 Conversely, ventricular mass by
overestimates volumetric mass even when the linear measurements are bSSFP is smaller than GRE mass. Thus reference values for “normal-
made from CMR images.40 Thus comparisons between serial studies range” ventricular volumes and mass must be imaging-sequence specific.
must take into account the imaging and analysis method used. The However, despite greater cavity volumes by bSSFP as compared with
volumetric summation of disks approach has been used to determine GRE methods, left ventricular ejection fraction (LVEF) is similar, regard-
RV mass with good results as well.41 less of which of these imaging sequences is used.
Currently, reference values for GRE-derived ventricular volumes
Left and Right Atria and mass are largely of historical interest only,48,49 but may be useful
Increased left atrial (LA) size is a marker of clinical and subclinical for interpretation of historical data. The Framingham Heart Study has
cardiovascular disease, reflecting increased LV filling pressures over published bSSFP-based reference values for the left and right ventricle50,51
time, among others. LA size and function can be determined using in healthy adults. Study participants were stratified by sex because men
a multislice, short-axis summation of disks method as is used for the have greater ventricular volumes and mass than women, before and
LV, but the LA is often assessed solely from biplane long-axis images after indexation to body surface area (BSA), whereas women have higher
in a manner similar to echocardiography. Increased LA volumes and LVEF. Participants were further stratified by age group because volume
decreased LA ejection fraction (EF) are associated with excess burden and mass decrease, whereas LVEF increases, with greater age among
of incident cardiovascular disease events,42–44 with prognostic value both sexes. Table 15.1 shows bSSFP LV reference values for healthy
above that of standard cardiovascular disease risk factors. Although adults (free of any history of hypertension and cardiac disease) from
both maximal and minimal LA volume are important, the latter may the Framingham population. Table 15.2 presents bSSFP RV reference
188 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

TABLE 15.1  LV Reference Values by Breath-Hold Cine bSSFP, by Sex and Age Group Among
Community-Dwelling Adult Subjects Free of Cardiovascular Disease and Any History of
Hypertension, Drawn From the Framingham Heart Study Offspring Cohort
MEN (AGE IN YEARS) WOMEN (AGE IN YEARS)
≤55 56–65 >65 ≤55 56–65 >65
N = 81 N = 157 N = 102 N = 104 N = 257 N = 151
EDV, mL 159 ± 23 143 ± 24 135 ± 26 118 ± 20 109 ± 18 99 ± 16
ESV, mL 56 ± 12 49 ± 12 46 ± 14 40 ± 10 35 ± 9 31 ± 9
SV, mL 102 ± 15 94 ± 16 89 ± 15 78 ± 13 74 ± 12 69 ± 10
LVM, g 128 ± 21 123 ± 21 120 ± 23 87 ± 16 82 ± 15 80 ± 13
EF, % 64.6 ± 4.3 65.9 ± 5.4 66.2 ± 5.7 66.1 ± 4.3 68.0 ± 4.8 69.4 ± 10.5
LVM/EDV, g/mL 0.82 ± 0.13 0.87 ± 0.14 0.91 ± 0.19 0.74 ± 0.10 0.76 ± 0.11 0.81 ± 0.11
EDV/BSA, mL/m2 77 ± 11 70 ± 12 68 ± 12 66 ± 8 62 ± 9 58 ± 8
ESV/BSA, mL/m2 28 ± 6 24 ± 6 23 ± 7 22 ± 4 20 ± 5 18 ± 5
SV/BSA, mL/m2 50 ± 7 46 ± 8 45 ± 7 43 ± 6 42 ± 6 40 ± 5
LVM/BSA, g/m2 62 ± 9 61 ± 9 59 ± 10 48 ± 7 46 ± 7 47 ± 7

BSA, Body surface area; bSSFP, balanced steady-state free precession; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic
volume; LV, left ventricular; LVM, left ventricular mass; SV, stroke volume.
Data are presented as mean ± standard deviation. The ratio LVM/EDV is a measure of concentricity.
Modified from Yeon SB, Salton CJ, Gona P, et al. Impact of age, sex and indexation method on MR in the Framingham Heart Study offspring
cohort. J Magn Reson Imaging. 2015;4:1038–1045.

TABLE 15.2  RV Reference Values by Breath-Hold Cine bSSFP, by Sex and Age Group Among
Community-Dwelling Adult Subjects Free of Cardiovascular and Pulmonary Disease, Drawn
From the Framingham Heart Study Offspring Cohort
MEN (AGE IN YEARS) WOMEN (AGE IN YEARS)
<55 55–64 65–74 ≥75 <55 55–64 65–74 ≥75
(N = 98) (N = 235) (N = 172) (N = 71) (N = 109) (N = 333) (N = 217) (N = 101)
EDV, mL 159 (2.9) 144 (1.9) 143 (2.2) 132 (3.4) 115 (1.9) 107 (1.1) 100 (1.3) 96 (2)
ESV, mL 63 (1.6) 52 (1) 51 (1.2) 47 (1.9) 39 (1) 35 (0.5) 32 (0.7) 29 (1)
SV, mL 96 (1.8) 91 (1.1) 91 (1.4) 85 (2.1) 76 (1.3) 72 (0.7) 68 (0.9) 67 (1.4)
EF, % 61 (0.6) 64 (0.4) 65 (0.5) 65 (0.8) 66 (0.6) 68 (0.3) 69 (0.4) 70 (0.6)
EDV/BSA, mL/m2 77 (1.3) 70 (0.8) 69 (1) 66 (1.5) 64 (0.9) 60 (0.5) 57 (0.7) 56 (1)
ESV/BSA, mL/m2 30 (0.8) 26 (0.5) 25 (0.6) 23 (0.6) 22 (0.5) 20 (0.3) 18 (0.4) 17 (0.5)
SV/BSA, mL/m2 46 (0.8) 44 (0.5) 44 (0.6) 42 (0.9) 42 (0.7) 41 (0.4) 39 (0.5) 39 (0.7)
BSA, Body surface area; bSSFP, balanced steady-state free precession; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic
volume; RV, right ventricular; SV, stroke volume.
Data are presented as least squares mean (standard error).
Modified from Foppa M, Arora G, Gona P, et al. Right ventricular volumes and systolic function by cardiac magnetic resonance and the impact
of sex, age, and obesity in a longitudinally followed cohort free of pulmonary and cardiovascular disease: the Framingham Heart Study. Circ
Cardiovasc Imaging. 2016;9:e003810.

values from the same population. A systematic review of normal LV had NC/C > 2.3 in at least one of eight myocardial segments,54 sug-
and RV volumes, ejection fraction, and mass is also available.46 gesting that the NC/C > 2.3 criterion might be overly broad. A variety
The superior image quality of bSSFP sequences provides more of methods, both quantitative and semiquantitative, for characterizing
detailed depiction of LV trabeculae than GRE methods, leading to trabecular burden by CMR have been proposed,55–57 but the optimal
increased concern over possible pathologic left ventricular noncom- method for CMR identification of LVNC remains to be determined.
paction (LVNC). LVNC has been defined echocardiographically as an Higher field strength CMR systems (i.e., 3 T) offer SNR advantages
excessive ratio of trabecular, noncompacted (NC) to compacted (C) over “conventional” 1.5 T systems, are well established for neurologic
myocardium (NC/C ≥ 2 at systole).52 A pioneering bSSFP-based study imaging, and are increasingly applied to cardiovascular imaging. With
proposed a CMR-specific ratio of NC/C > 2.3 at diastole53 for diagnosis respect to cardiac size and function, the magnetic field strength of the
of LVNC, but this was based on a relatively small group of patients with scanner (i.e., 1.5 T versus 3 T) does not appear to have any significant
echocardiographically defined LVNC. Subsequently, the Multiethnic effect on measured ventricular volumes and mass, but the systematic
Study of Atherosclerosis (MESA) investigators found that among 323 difference between GRE and bSSFP imaging sequences is preserved
healthy adults without history of hypertension or cardiac disease, 43% regardless of field strength.58,59
CHAPTER 15  Normal Cardiac Anatomy 189

reproducible, leading to loss of signal and motion artifacts. Third,


Aortic Flow valve pathology frequently involves fibrosis and calcification, both of
An important feature of CMR is its sensitivity to motion, and this which are characterized by loss of signal on CMR, making it difficult
feature can be harnessed to allow the measurements of velocity and to detect, particularly in spin echo imaging, Last, regions of turbu-
vessel flow without the constraints of Doppler methods. Although there lence are associated with loss of signal on CMR, which may lead to
is potential for artifacts, CMR allows accurate and reproducible mea- overestimation of the extent of abnormality on GRE imaging. In spite
surement of vessel flow in vivo.37,60,61 The approach for the measurement of these concerns, CMR can be used to obtain high-resolution images
of aortic flow is straightforward. The coronal scout image is used, and of valves using both bright-blood bSSFP techniques or black-blood
the imaging plane is placed perpendicular to the direction of flow several breath-hold, double inversion recovery techniques.65 An example of
centimeters above the aortic valve, typically at approximately the level a normal valve image obtained using the latter technique is shown
of the bifurcation of the main pulmonary artery (Fig. 15.6A). The pulse in Fig. 15.7A. The LV outflow tract (LVOT) view in Fig. 15.7B shows
sequence uses bipolar gradients oriented in the expected direction of aortic and mitral valves by bSSFP imaging and Fig. 15.7C is an en face
flow, so that tissues, such as blood, moving through the imaging plane view of a normal trileaflet aortic valve; this frame from the bSSFP
accrue a nonzero net phase, whereas stationary tissues gain and lose cine loop shows the closed valve at end diastole. Fig. 15.7D is another
phase equally for zero net phase change. The phase change corresponds normal aortic valve open during early systole. Fig. 15.7E is an en face
to velocity. The velocity encoding value (VENC) should be chosen to view of a sclerotic, mildly stenosed aortic valve during early systole.
be just above the anticipated maximum velocity (approximately 1.5 m/s Fig. 15.7F shows a dark signal void caused by turbulent flow of aortic
in normal individuals, and higher in patients with aortic valve disease). regurgitation.
Setting the VENC too low can result in aliasing, where phase change However, imaging the valvular abnormality is only one part of the
exceeds +180 degrees, so that there is an abrupt discontinuity in appar- evaluation of the patient with valvular disease. It is essential to quantify
ent velocity, also known colloquially as wrap-around. For example, a the functional severity of the lesion and to determine its impact on
phase change of, for example, +270 degrees will be misinterpreted as a ventricular size and function. Full details of assessing valvular abnor-
phase of −90 degrees, because of wrap-around. malities are detailed in Chapter 30, but some general comments are
The reconstructed images are typically presented as a set of mag- useful. As noted earlier, CMR is highly accurate in determining ventric-
nitude images that are used to determine the cross-sectional area of ular dimensions and volumes. In addition, phase-contrast quantitative
the aorta in each frame (Fig. 15.6B). There is also a set of phase-encoded flow techniques provide effective means for determining velocity and
(velocity map) images in which the gray scale indicates the velocity of blood flow. Thus in addition to demonstrating that valvular disease
motion in each voxel (Fig. 15.6C). Velocity is measured at each voxel is present, CMR can be used to quantify the regurgitant volume and
across the vessel, integrated over the cross-sectional area of the vessel, to determine its effect on ventricular size and function. Valve pres-
and then integrated over the cardiac cycle (Fig. 15.6D). The integrated sure gradients estimated using CMR correlate well with ultrasound
flow across the slice at each point in time can be graphed. Note that measurements.66,67 In addition, measurements of aortic valve area and
some retrograde flow is normal in the ascending aorta during early cardiac output by CMR agree with measurements of valve area at
diastole because of closure of the aortic valve, diastolic ascent of the catheterization. Regurgitant jets are generally well visualized because
base of the heart, and diastolic coronary flow. of turbulence (dephasing), creating dark regions of signal loss in
GRE cines, but it is hazardous to estimate even qualitative severity of
Pulmonary Artery Flow regurgitation from the size of the region of signal loss on cine CMR
Pulmonary artery flow can be measured using techniques similar to because the apparent size of the jet is highly dependent on the details
those for aortic flow. This is particularly valuable in evaluation of patients of the particular imaging sequence used. Specifically, although bSSFP
with left-to-right intracardiac shunting to determine the ratio of pul- imaging generally provides excellent depiction of myocardial and valve
monary flow to systemic flow (Qp/Qs), which has good correlation anatomy, the size of dephasing jets is smaller (see Fig. 15.7F) than with
with invasive techniques.62,63 Depending on the pulmonary artery ori- GRE sequences. Regurgitant jets can be underestimated or overlooked
entation, the location of the flow images can be planned from the axial entirely based on visual assessment alone. Quantitative flow techniques
and/or sagittal scout with a perpendicular plane positioned several are more useful in determining the regurgitant volume, for example,
centimeters distal to the pulmonary valve. The velocity profile is then by measuring retrograde flow in diastole in the aorta.68,69 Interestingly,
integrated over the cross-sectional area of the artery over time to deter- measures of chamber volume and cardiac output by CMR in patients
mine the volume flow per cardiac cycle in a manner similar to that with atrial fibrillation agree well with invasive measures.11 Mitral
used in the ascending aorta. regurgitation has also been studied using quantitative measures.70,71
Interrogation of the aortic and pulmonic valves is relatively straight-
forward using velocity-encoding methods as described previously, but
NORMAL VALVULAR FUNCTION the application of this method to the mitral and tricuspid valves is not
Assessment of valve function involves evaluation of morphology, necessarily straightforward, as a result of the through-plane translation
motion, competence, and effects on ventricular function. Imaging (10–20 mm in a normal heart) of the base of the ventricles and thus
cardiac valve morphology poses significant problems for CMR.64 First, the mitral and tricuspid annuli. Assessment of mitral regurgitation
the normal valve is a thin, fibrous structure often less than 1 mm thick, is more reliably achieved by calculating the difference between LV
leading to potential for partial volume effects. Second, it is constantly stroke volume, by summation of disks method, and net aortic forward
in motion. In most cine CMR, the image is acquired over a number flow, by phase-encoded velocity mapping, so long as there is no
of cardiac cycles, requiring that the valve return to the same posi- intracardiac shunt.
tion with each cycle. With breath-hold imaging, one can obtain very The presence of a prosthetic valve is not a contraindication for
high-resolution images, indicating that normal valve motion appears CMR72 except potentially in the case of valve dehiscence,73 although
to be quite reproducible over a limited number of cycles when the mechanical valves, or the struts of bioprosthetic valves, will result in
effects of respiratory motion are removed. However, vegetations and artifacts (see Fig. 15.4F). Similarly, rings used for valve repair may
other valve pathology are characterized by valve motion, which is less produce local artifacts (see Fig. 15.4H).
190 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

A B

C
Flux results (slice 1) Scan time 00:02:51
PHILIPS MR 1.5 T
ml/s
SESSION INFORMATION:
400 Q-Flow: AorticFlow (not validated).
350 RESULTS SUMMARY:
Heart rate : 60 beats/min
300 RR-interval : 1000 ms (from heart rate)
FIG. 15.6  (A) Coronal scout image for measur- 250 ANALYSIS RESULTS:
ing aortic flow. The white line indicates the
200 slice 1 Vessel
anatomic position of a flow sequence. The 1
plane is positioned at the level of the bifurca- 150
Stroke volume (ml) 73.5
tion of the main pulmonary artery well above 100 Forward flow vol. (ml) 73.8
Backward flow vol. (ml) 0.3
the aortic valve and perpendicular to the walls 50 Regurgitant fract. (%) 0.4
Abs. stroke volume (ml) 74.1
of the aorta. (B) Magnitude reconstruction from 0 Mean flux (ml/s) 73.5
Stroke distance (cm) 9.5
the flow sequence positioned in panel A. The 0 100 200 300 400 500 600 700 800 9001000 Mean velocity (cm/s) 9.5
time (ms)
imaging plane is transverse and positioned at RR-interval: 1000 ms (from heart rate) Vessel 1, slice 1
the level of the bifurcation of the main pulmo-
Q-Flow: AorticFlow (not validated)
nary artery. The ascending aorta is seen ante- Flux: 7.19 ml/s Peak vel: 12.82 cm/s
riorly (arrow). (C) Velocity map reconstruction 300
Mean vel: 0.94 cm/s Max. vel: 12.82 cm/s cm/s
from the same flow sequence as shown in Area: 7.65 cm 2 Min vel: –6.96 cm/s
panel B. The gray scale in this image indicates Pixels: 557 pixels Vel stddev: 3.38 cm/s 200
the velocity of motion toward the head as bright
(solid arrow, ascending aorta) and away as dark 100
(dashed arrow, descending aorta). (D) Typical
dataset for semiautomated analysis. The area 0
of the ascending aorta is determined in each
frame and the velocity over the area is inte- –100
grated to calculate flow volume per frame. The
top left subpanel shows a graph of flow volume –200
over the cardiac cycle in the ascending aorta.
FFE/M PCA/P –300 cm/s
The forward stroke volume, shown in the SI 1 SI 1
results listed in the upper right subpanel, is Ph 1/000 ms Ph 1/000 ms
calculated by integrating the flow over the
D PCv FH 300 cm/s
cardiac cycle.
CHAPTER 15  Normal Cardiac Anatomy 191

A B

C D

E F
FIG. 15.7  (A) Double inversion recovery spin echo long-axis image. The aortic valve leaflets are demonstrated
(arrowhead). (B) A left ventricular outflow tract view acquired using breath-hold balanced steady-state free
precession (bSSFP) imaging. (C) A normal trileaflet aortic valve depicted en face at end diastole (valve closed)
by bSSFP imaging. (D) An open, normal trileaflet aortic valve seen during early systole. (E) An en face view
of a sclerotic, mildly stenosed, trileaflet aortic valve also during early systole. Note the small opening and
deformed leaflets as compared with panel D. (F) Aortic regurgitation is visualized qualitatively in the dephas-
ing jet (arrow) in this bSSFP image.

be a very effective tool in the noninvasive evaluation of nearly all


CONCLUSION
cardiovascular diseases.
CMR can be used to clearly delineate cardiac structure and assess
function. As with any imaging technique, it is important to have
a strategy for imaging and be familiar with the normal anatomy
REFERENCES
and potential artifacts. With this knowledge in hand, CMR can A full reference list is available online at ExpertConsult.com
CHAPTER 15  Normal Cardiac Anatomy 191.e1

REFERENCES 21. Chuang ML, Hibberd MG, Beaudin RA, et al. Importance of imaging
method over imaging modality in noninvasive determination of left
1. Cerqueira MD, Weissman NJ, Dilsizian V, et al. Standardized myocardial ventricular volumes and ejection fraction: assessment by two and three
segmentation and nomenclature for tomographic imaging of the heart: a dimensional echocardiography and magnetic resonance imaging. J Am
statement for healthcare professionals from the Cardiac Imaging Coll Cardiol. 2000;35:477–484.
Committee of the Council on Clinical Cardiology of the American Heart 22. Kaplan W. Advanced Calculus. New York: Addison-Wesley; 2002.
Association. Circulation. 2002;105:539–542. 23. Strohm O, Schulz-Menger J, Pilz B, et al. Measurement of left ventricular
2. Manning WJ, Braunwald E, eds. Atlas of Cardiovascular Magnetic dimensions and function in patients with dilated cardiomyopathy. J Magn
Resonance. New York: Springer; 2008:304. Reson Imaging. 2001;13:367–371.
3. Kramer CM, Hundley WG. Atlas of Cardiovascular Magnetic Resonance 24. Zerhouni EA, Parish DM, Rogers WJ, Yang A, Shapiro EP. Human heart:
Imaging. Philadelphia: Saunders; 2010:376. tagging with MR imaging—a method for noninvasive assessment of
4. Arai AE, Epstein FH, Bove KE, Wolff SD. Visualization of aortic valve myocardial motion. Radiology. 1988;169:59–63.
leaflets using black blood MRI. J Magn Reson Imaging. 1999;10:771–777. 25. Axel L, Dougherty L. MR imaging of motion with spatial modulation of
5. Fayad ZA, Fuster V, Fallon JT, et al. Noninvasive in vivo human coronary magnetization. Radiology. 1989;171:841–845.
artery lumen and wall imaging using black-blood magnetic resonance 26. Osman NF, Kerwin WS, McVeigh ER, Prince JL. Cardiac motion tracking
imaging. Circulation. 2000;102:506–510. using CINE harmonic phase (HARP) magnetic resonance imaging. Magn
6. Botnar RM, Stuber M, Kissinger KV, et al. Non-invasive coronary vessel Reson Med. 1999;42:1048–1060.
wall and plaque imaging with magnetic resonance imaging. Circulation. 27. Aletras AH, Ding S, Balaban RS, Wen H. DENSE: displacement encoding
2000;102:2582–2587. with stimulated echoes in cardiac functional MRI. J Magn Reson.
7. Thiele H, Nagel E, Paetsch I, et al. Functional cardiac MR imaging with 1999;137:247–252.
steady-state free precession (SSFP) significantly improves endocardial 28. Korosoglou G, Lossnitzer D, Schellberg D, et al. Strain-encoded cardiac
border delineation without contrast agents. J Magn Reson Imaging. MRI as an adjunct for dobutamine stress testing: incremental value to
2001;14:362–367. conventional wall motion analysis. Circ Cardiovasc Imaging.
8. Jarvinen VM, Kupari MM, Hekali PF, Poutanen VP. Right atrial MR 2009;2:132–140.
imaging studies of cadaveric atrial casts and comparison with right and 29. Neizel M, Korosoglou G, Lossnitzer D, et al. Impact of systolic and
left atrial volumes and function in healthy subjects. Radiology. diastolic deformation indexes assessed by strain-encoded imaging to
1994;191:137–142. predict persistent severe myocardial dysfunction in patients after acute
9. Mortele KJ, Mergo PJ, Williams WF. Lipomatous hypertrophy of the atrial myocardial infarction at follow-up. J Am Coll Cardiol. 2010;56:1056–1062.
septum: diagnosis with fat suppressed MR imaging. J Magn Reson 30. Oyama-Manabe N, Ishimori N, Sugimori H, et al. Identification and
Imaging. 1998;8:1172–1174. further differentiation of subendocardial and transmural myocardial
10. Rajiah P, Bolen MA, Cardiovascular MR. imaging at 3 T: opportunities, infarction by fast strain-encoded (SENC) magnetic resonance imaging at
challenges, and solutions. Radiographics. 2014;34:1612–1635. 3.0 tesla. Eur Radiol. 2011;21:2362–2368.
11. Hundley WG, Meshack BM, Willett DL, et al. Quantitation of left 31. el-lbrahim SH. Myocardial tagging by cardiovascular magnetic resonance:
ventricular volumes, ejection fraction and cardiac output in patients with evolution of techniques—pulse sequences, analysis algorithms, and
atrial fibrillation by cine magnetic resonance imaging: a comparison with applications. J Cardiovasc Magn Reson. 2011;13:36.
invasive measurements. Am J Cardiol. 1996;78:1119–1123. 32. Simpson RM, Keegan J, Firmin DN. MR assessment of regional
12. Fischer SE, Wickline SA, Lorenz CH. Novel real-time R-wave detection myocardial mechanics. J Magn Reson Imaging. 2013;37:576–599.
algorithm based on the vectorcardiogram for accurate gated magnetic 33. Boxt LM, Katz J, Kolb T, et al. Direct quantitation of right and left
resonance acquisitions. Magn Reson Med. 1999;42:361–370. ventricular volumes with nuclear magnetic resonance imaging in patients
13. Danias PG, Stuber M, Botnar RM, et al. Relationship between motion of with primary pulmonary hypertension. J Am Coll Cardiol.
coronary arteries and diaphragm during free breathing: lessons from 1992;19:1508–1515.
real-time MR imaging. AJR Am J Roentgenol. 1999;172:1061–1065. 34. Utz JA, Herfkens RJ, Heinsimer JA, et al. Cine MR determination of left
14. Taylor AM, Jhooti P, Wiesmann F, et al. Magnetic resonance navigator- ventricular ejection fraction. AJR Am J Roentgenol. 1987;148:839–843.
echo monitoring of temporal changes in diaphragm position: 35. Culham JAG, Vince DJ. Cardiac output by MR imaging: an experimental
implications for magnetic resonance coronary angiography. J Magn Reson study comparing right ventricle and left ventricle with thermodilution.
Imaging. 1997;7:629–636. J Can Assoc Radiol. 1988;39:247–249.
15. Botnar RM, Stuber M, Danias PG, et al. Improved coronary artery 36. Hundley WG, Li HF, Hillis LD, et al. Quantitation of cardiac output with
definition with T2-weighted, free-breathing, three-dimensional coronary velocity-encoded, phase-difference magnetic resonance imaging:
MRA. Circulation. 1999;99:3139–3148. validation with invasive measurements. Am J Cardiol. 1995;75:
15a.  Usman M, Atkinson D, Heathfield E, et al. Whole left ventricular 1250–1255.
functional assessment from two minutes free breathing multi-slice CINE 37. Hundley WG, Li HF, Willard JE, et al. Magnetic resonance imaging
acquisition. Phys Med Biol. 2015;60:N93–N107. assessment of the severity of mitral regurgitation: a comparison with
16. Babcock EE, Brateman L, Weinreb JC, et al. Edge artifacts in MR images: invasive techniques. Circulation. 1995;92:1151–1158.
chemical shift effect. J Comput Assist Tomogr. 1985;9:252–257. 38. François CJ, Fieno DS, Shors SM, Finn JP. Left ventricular mass: manual
17. Lotan CS, Cranney GB, Doyle M, Pohost GM. Fatshift artifact simulating and automatic segmentation of true FISP and FLASH cine MR images in
aortic dissection on MR images. AJR Am J Roentgenol. 1989;152:385–386. dogs and pigs. Radiology. 2004;230:389–395.
18. Deshmane A, Gulani V, Griswold MA, Seiberlich N. Parallel MR imaging. 39. Katz J, Milliken MC, Stray-Gundersen J, et al. Estimation of human
J Magn Reson Imaging. 2012;36:55–72. myocardial mass with MR imaging. Radiology. 1988;169:495–498.
18a.  Akhtari S, Chuang ML, Berg S, et al. Left and right ventricular volumes 40. Bottini PB, Carr AA, Prisant LM, et al. Magnetic resonance imaging
and global systolic function in isolated left bundle branch block: a cardiac compared to echocardiography to assess left ventricular mass in the
magnetic resonance imaging study. J Cardiovasc Magn Reson. 2016; hypertensive patient. Am J Hypertens. 1995;8:221–228.
18(suppl 1):P296. 41. Kawut SM, Barr RG, Lima JA, et al. Right ventricular structure is
19. Voit D, Zhang S, Unterberg-Buchwald C, et al. Real-time cardiovascular associated with the risk of heart failure and cardiovascular death: the
magnetic resonance at 1.5 T using balanced SSFP and 40 ms resolution. Multi-Ethnic Study of Atherosclerosis (MESA)—right ventricle study.
J Cardiovasc Magn Reson. 2013;15:79. Circulation. 2012;126:1681–1688.
20. La Gerche A, Claessen G, Van de Bruaene A, et al. Cardiac MRI: a new 42. Gupta S, Matulevicius SA, Ayers CR, et al. Left atrial structure and
gold standard for ventricular volume quantification during high-intensity function and clinical outcomes in the general population. Eur Heart J.
exercise. Circ Cardiovasc Imaging. 2013;6:329–338. 2013;34:278–285.
191.e2 SECTION I  Basic Principles of Cardiovascular Magnetic Resonance

43. Wu VC, Takeuchi M, Kuwaki H, et al. Prognostic value of LA volumes 58. Hudsmith LE, Petersen SE, Tyler DJ, et al. Determination of cardiac
assessed by transthoracic 3D echocardiography: comparison with 2D volumes and mass with FLASH and SSFP cine sequences at 1.5 vs. 3 tesla:
echocardiography. JACC Cardiovasc Imaging. 2013;6:1025–1035. a validation study. J Magn Reson Imaging. 2006;24:312–318.
44. Habibi M, Chahal H, Opdahl A, et al. Association of CMR-measured LA 59. Grothues F, Boenigk H, Graessner J, Kanowski M, Klein HU. Balanced
function with heart failure development: results from the MESA study. steady-state free precession vs. segmented fast low-angle shot for the
JACC Cardiovasc Imaging. 2014;7:570–579. evaluation of ventricular volumes, mass and function and 3 tesla. J Magn
45. Russo C, Jin Z, Homma S, et al. Left atrial minimum volume and Reson Imaging. 2007;26:392–400.
reservoir function as correlates of left ventricular diastolic function: 60. Tang C, Blatter DD, Parker DL. Accuracy of phase-contrast flow
impact of left ventricular systolic function. Heart. 2012;98:813–820. measurements in the presence of partial volume effects. J Magn Reson
46. Kawel-Boehm N, Maceira A, Valsangiacomo-Buechel ER, et al. Normal Imaging. 1993;3:377385.
values for cardiovascular magnetic resonance in adults and children. J 61. Buonocore MH, Bogren H. Factors influencing the accuracy and
Cardiovasc Magn Reson. 2015;17:29. precision of velocity-encoded phase imaging. Magn Reson Med.
47. Moon JCC, Lorenz CH, Francis JM, Smith GC, Pennell DJ. Breath-hold 1992;26:141–154.
FLASH and FISP cardiovascular MR imaging: left ventricular volume 62. Brenner LD, Caputo GR, Mostbeck G, et al. Quantification of left to right
differences and reproducibility. Radiology. 2002;223:789. atrial shunts with velocity-encoded cine nuclear magnetic resonance
48. Salton CJ, Chuang ML, O’Donnell CJ, et al. Gender differences and imaging. J Am Coll Cardiol. 1992;20:1246–1250.
normal left ventricular anatomy in an adult population free of 63. Hundley WG, Li HE, Lange RA, et al. Assessment of left-to-right
hypertension: a cardiovascular magnetic resonance study of the intracardiac shunting by velocity-encoded, phase-difference magnetic
Framingham Heart Study offspring cohort. J Am Coll Cardiol. resonance imaging: a comparison with oximetric and indicator dilution
2002;39:1055–1060. techniques. Circulation. 1995;91:2955–2960.
49. Tandri H, Daya SK, Nasir K, et al. Normal reference values for the adult 64. Duerinckx AJ, Higgins CB. Valvular heart disease. Radiol Clin North Am.
right ventricle by magnetic resonance imaging. Am J Cardiol. 1994;32:613–630.
2006;98:1660–1664. 65. Arai AE, Epstein FH, Bove KE, Wolff SD. Visualization of aortic valve
50. Yeon SB, Salton CJ, Gona P, et al. Impact of age, sex, and indexation leaflets using black blood MRI. J Magn Reson Imaging. 1999;10:771–777.
method on MR left ventricular reference values in the Framingham Heart 66. Kilner PJ, Firmin DN, Rees RSO, et al. Valve and great vessel stenosis:
Study offspring cohort. J Magn Reson Imaging. 2015;4:1038–1045. assessment with magnetic resonance jet velocity mapping. Radiology.
51. Foppa M, Arora G, Gona P, et al. Right ventricular volumes and systolic 1991;178:229–235.
function by cardiac magnetic resonance and the impact of sex, age, and 67. Eichenberger AC, Jenni R, von Shulthess GK. Aortic valve pressure
obesity in a longitudinally followed cohort free of pulmonary and gradients in patients with aortic valve stenosis: quantification with
cardiovascular disease: the Framingham Heart Study. Circ Cardiovasc velocity-encoded cine MR imaging. AJR Am J Roentgenol.
Imaging. 2016;9:e003810. 1993;160:971–977.
52. Oechslin EN, Attenhofer Jost CH, et al. Long-term follow-up of 34 adults 68. Dulce MC, Mostbeck GH, O’Sullivan M, et al. Severity of aortic
with isolated left ventricular noncompaction: a distinct cardiomyopathy regurgitation: interstudy reproducibility of measurements with
with poor prognosis. J Am Coll Cardiol. 2000;36:493–500. velocity-encoded cine MR imaging. Radiology. 1992;185:235–240.
53. Petersen SE, Selvanayagam JB, Wiesmann F, et al. Left ventricular 69. Walker PG, Oyre S, Pedersen EM, et al. A new control volume
non-compaction: insights from cardiovascular magnetic resonance method for calculating valvular regurgitation. Circulation. 1995;92:
imaging. J Am Coll Cardiol. 2005;46:101–105. 579–586.
54. Kawel N, Nacif M, Arai AE, et al. Trabeculated (noncompacted) and 70. Fujita N, Chazouilleres AE, Hartiala JJ, et al. Quantification of mitral
compact myocardium in adults: the multi-ethnic study of atherosclerosis. regurgitation by velocity-encoded cine nuclear magnetic resonance
Circ Cardiovasc Imaging. 2012;5:357–366. imaging. J Am Coll Cardiol. 1994;23:951–958.
55. Chuang ML, Gona P, Hautvast GL, et al. Correlation of trabeculae and 71. Hundley WG, Li HF, Willard JE, et al. Magnetic resonance imaging
papillary muscles with clinical and cardiac characteristics and impact on assessment of the severity of mitral regurgitation: a comparison with
CMR measures of LV anatomy and function. JACC Cardiovasc Imaging. invasive techniques. Circulation. 1995;92:1151–1158.
2012;5:1115–1123. 72. Deutsch HJ, Bachmann R, Sechtern U, et al. Regurgitant flow in cardiac
56. Choi Y, Kim SM, Lee SC, et al. Quantification of left ventricular valve prostheses: diagnostic value of gradient echo nuclear magnetic
trabeculae using cardiovascular magnetic resonance for the diagnosis of resonance imaging in reference to transesophageal two-dimensional color
left ventricular non-compaction: evaluation of trabecular volume and doppler echocardiography. J Am Coll Cardiol. 1992;19:1500–1507.
refined semi-quantitative criteria. J Cardiovasc Magn Reson. 2016;18:24. 73. Shellock FG, Crues JV, eds. MRI Bioeffects, Safety, and Patient
57. Captur G, Muthurangu V, Cook C, et al. Quantification of left ventricular Management. Los Angeles: Biomedical Research Publishing Group;
trabeculae using fractal analysis. J Cardiovasc Magn Reson. 2013;15:36. 2014:718.
SECTION II  Ischemic Heart Disease

16 
Assessment of Cardiac Function
Alicia M. Maceira, Nicholas G. Bellenger, and Dudley J. Pennell

The accurate and reproducible assessment of cardiac function is a fun- failure aims to decrease the likelihood of disease progression that could
damental aim of noninvasive cardiac imaging. It forms the foundation result in costly hospital admissions and ultimately death. To allow early
upon which much of the assessment and management of myocardial evaluation and alteration of an individual patient’s management, serial
dysfunction, ischemia, viability, remodeling, valvular, and other cardiac studies need to be performed using a technique that not only is accurate
disorders are based. In this chapter, we discuss the importance of the but also has good interstudy reproducibility. This principle also applies
measurement of global cardiac function, compare techniques, show in considering study populations of trial therapies.
differences according to magnetic fields, provide a practical step-by-step
guide to its assessment by cardiovascular magnetic resonance (CMR) TECHNIQUES FOR ASSESSING
and describe new techniques that can be used for the assessment of
cardiac function.
CARDIAC FUNCTION
Bedside clinical assessment of cardiac function is generally poor,7 and
THE POPULATION IMPACT OF electrocardiogram (ECG) findings are nonspecific, although the pres-
ence of an entirely normal ECG has a 95% likelihood of normal systolic
CARDIAC DYSFUNCTION function.8 In the search for a better assessment, it is worth considering
Cardiac dysfunction can result from a broad spectrum of organ-specific the ideal imaging technique. This would provide a noninvasive, accurate,
and multisystem disorders. The defining property that all these disorders and reproducible assessment of cardiac function without exposure to
have in common is impairment of the ventricle’s ability to eject blood, ionizing radiation. It would be widely available and would be time- and
which, in its broadest sense and omitting any discussion of semantics, cost-effective. Although no technique meets these ideals, there are clear
is known as heart failure. Heart failure is common, with approximately differences between modalities that merit discussion.
1.5% to 2% of the population below 65 years old being affected, rising
to 6% to 10% of those older than 65 years.1 It afflicts 4.8 million people Echocardiography
in the United States, with 400,000 to 700,000 new cases developing each Echocardiography is a widely available but less than ideal imaging tech-
year. It is the leading cause of hospital admission in people older than nique for quantifying cardiac function because the image acquisition
65 years of age, and despite the fact that survival has improved over is operator and acoustic window dependent.9 The quantification of
time, the death rate remains high: ≈50% of people diagnosed with heart ventricular function is limited by a priori geometric assumptions that
failure will die within 5 years.2 The number of any-mention deaths may provide a reasonable assessment in the normal ventricle but are
attributable to heart failure was approximately as high in 1995 as it was less reliable in remodeled hearts, owing to complex irregular shape
in 2011, although projections show that the prevalence of heart failure changes.10,11 M-mode echocardiography was developed in the early 1970s
will increase 46% from 2012 to 2030, resulting in over 8 million people and was immediately applied in practice for LV function assessment,
18 years old and older with heart failure in the United States.3 Conse- because of its simple algorithm and noninvasiveness. In this technique,
quently, heart failure is an enormous consumer of health care budgets mid-LV diameters are measured in the minor axis (Fig. 16.1), and volumes
in the Western world, and treatments to prevent occurrence, to slow are obtained by cubing these values (thereby cubing the errors).12–14
its progression, or to prevent repeated hospitalizations can have an Functional estimates, such as fractional shortening and ejection fraction
important economic impact. (EF), are then derived from these diameters and volumes, respectively.
This method assumes a number of facts that are not always true: that
THE IMPORTANCE OF MEASURING the minor-axis view is strictly perpendicular to the LV long axis, that a
single view is representative of all the myocardial segments, and that
CARDIAC FUNCTION contraction is uniform throughout the ventricle. Two-dimensional echo-
A single assessment of cardiac function can provide important diagnostic cardiography (2DE), with the ability of imaging the heart in tomographic
and prognostic information, whether in the setting of postinfarction views, considerably improved the accuracy of LV volume measurement,
recovery,4 left ventricular (LV) hypertrophy,5 or chronic heart failure.6 providing the opportunity to derive the cardiac function from cardiac
As well as improving symptoms and quality of life, treatment of heart volumes by the area-length and modified Simpson’s method of discs

192
CHAPTER 16  Assessment of Cardiac Function 193

acceptance in clinical trials, owing to its moderate reproducibility and


CW accuracy to define LV ejection fraction (LVEF), because of geometric
assumptions, poor acoustic windows, foreshortening of the ventricle,
RV and inadequate discrimination of the endocardial border. Overall, the
practical difficulties of quantifying global function by echo are underlined
by the fact that, in the “real world,” it is often simply estimated by the
IVS
clinician performing the imaging. This is highly subjective but can be
ESD clinically valid with experience.16 Other echocardiographic methods,
LV
EDD derived from Doppler analysis, have been validated for evaluation of
global myocardial performance that do not require geometric assump-
PVW
tions and do not rely on endocardial border delineation. These include
for instance the Tei index,17 which is less load dependent, the dP/dt,
which requires mitral regurgitation to be present,18 and tissue Doppler
FIG. 16.1  M-mode echo recording at the midventricular level in a patient mitral annular systolic velocity. However, these methods require a good
with tricuspid regurgitation showing paradoxical systolic motion of the alignment of the ultrasound beam with flow or myocardial motion and
interventricular septum and right ventricle. CW, Chest wall; EDD, end- have not been really implemented for daily clinical use. Some years
diastolic dimension; ESD, end-systolic dimension; IVS, interventricular ago, two advances in echocardiography helped improve accuracy and
septum; LV, left ventricle; PVW, posterior ventricular wall; RV, right reproducibility of ventricular function assessment. On the one hand,
ventricle.
contrast echocardiography has been shown to allow improved assess-
ment of LV volumes and LVEF,19 with low interobserver variability,20
thanks to better endocardial border delineation, thus improving the
assessment of LV dimensions and wall motion (Fig. 16.3). On the
other hand, three-dimensional (3D) echocardiography has emerged
as a more accurate and reproducible approach to LV quantitation by
removing the need for geometric assumptions21 and eliminating the
errors caused by foreshortened apical views. Volumetric analysis of
real-time 3D echocardiographic data allows fast dynamic measure-
ment of LV volumes.22 3D echocardiography has good reproducibility
and good agreement with CMR measurements of LVEF, whereas the
agreement of volumes is worse.23 Although in the past this technique
needed a stable cardiac rhythm and constant cardiac function during
a moderately long acquisition, recent developments have allowed a full
pyramidal dataset to be acquired in a single heartbeat, during a short
RV breath-hold, thus eliminating motion artifacts, and without the need for
off-line reconstruction. Still, issues such as its low temporal resolution,
lateral attenuation, dependency on acoustic window and on endocardial
border delineation, and practicality in clinical practice still remain to
be fully assessed as more experience is gained. The use of contrast may
A overcome some of these limitations. A recent multicenter study has
shown that contrast administration on 3D echocardiography results
in improved determination of LV volumes and reduced interreader
variability.23 Finally, speckle-tracking imaging is a relatively new tool
RA
to assess LV function through measurement of myocardial strain, with
LA a high temporal and spatial resolution, and a better inter observer and
intraobserver reproducibility compared with Doppler strain. It is angle
independent, not affected by translation cardiac movements, and can
assess simultaneously the entire myocardium along all the 3D geometri-
FIG. 16.2  Echo measurement by Simpson’s method of discs. This cal (longitudinal, circumferential, and radial) axes.24 Again, there are
represents a more accurate measurement than M-mode or area-length pitfalls such as low spatial and temporal resolution of this technique,
assessment but relies on good endocardial border definition. A, Annulus; reproducibility problems across vendors, and lack of wide availability
LA, left atrium; RA, right atrium; RV, right ventricle. of the technique, which currently is used in the clinical setting, mainly
for monitoring cancer therapy–related cardiotoxicity.25
(Fig. 16.2).12 This, however, relies on assuming that the left ventricle
has a uniform shape with no regional wall motion abnormalities and Nuclear Cardiology
on good visualization of the entire endocardial border, which is fre- Nuclear cardiology with radionuclide ventriculography with either Tc-
quently not possible, although several software-based algorithms for 99m-labeled red blood cells or human serum albumin is commonly
automatic endocardial border detection and online calculation of LV used to measure ventricular function by measuring the LVEF,26 with
volume have been developed.15 For example, in a multicenter study that high accuracy and reproducibility, but it has relatively low spatial and
required good-quality echocardiograms as an entry criterion, the echo temporal resolution, and preparation and scanning times are relatively
core laboratory was unable to perform a confident two-dimensional prolonged. In addition, ventricular volumes are difficult to measure
(2D) analysis in 31% of patients.14 Thus being the most frequently and are rarely performed clinically, and ventricular mass cannot be
used modality in clinical practice, echocardiography has gained limited obtained. Furthermore, the accuracy of the LVEF obtained with a
194 SECTION II  Ischemic Heart Disease

A B

C D
FIG. 16.3  Usefulness of contrast echocardiography for left ventricular opacification. Images A and C show
a four-chamber view in end diastole (A) and end systole (C), whereas B and D show the same frames after
intravenous contrast administration. The visualization of the lateral wall is greatly improved after contrast
(arrows).

multigated acquisition (MUGA) scan decreases in patients with irregular are very low. Tc-99m sestamibi ECG-gated SPECT can predict ventricular
heart rhythms. Last but not least, with this scan the patient is exposed remodeling in patients with ischemic cardiomyopathy,32 but 3D echo-
to an amount of radiation of up to 6 mSv, which is roughly twice the cardiography seems to be more accurate than gated SPECT for estima-
normal background radiation a person receives in 1 year. Consequently, tion of LV remodeling.33 With respect to radiation dose, advances in
this technique is very rarely used today for ventricular function analysis, hardware, including ultrahigh-sensitivity detectors, dedicated geometry,
not even for monitoring cardiotoxicity of cancer therapies which used shorter scan times and optimized reconstruction, and software, with
to be its main indication.27 The use of gated perfusion single-photon implementation of improved reconstruction algorithms,34 have obtained
emission computed tomography (gSPECT) has allowed the develop- total radiation doses of 4.2 to 6.8 mSv.35 Recently, ECG-gated F-18
ment of 3D solutions to global and regional ventricular function,28 and fluorodeoxyglucose positron emission tomography (8F-FDG PET) has
this has achieved widespread use, especially in the United States. This been shown to be reasonably accurate for measurement of cardiac func-
technique is most useful when perfusion needs to be assessed, and it tion,36 but this technique is expensive, time consuming, and is usually
adds prognostic value to the stress perfusion assessment29; however, it performed only in viability studies. Typical radiation doses for cardiac
18
is very rarely performed solely to assess ventricular function (Fig. 16.4). F-FDG PET scans are now around 7 mSv.37 Importantly, for all these
This technique has good reproducibility both for ejection fraction30 nuclear cardiology techniques, the need for repeated radionuclide doses
and ventricular volumes,31 but there are a high number of variables in follow-up studies is highly problematic, especially for research, in
that can affect accuracy, and must be carefully taken care of, including which radiation exposure must be justified in a milieu of competing
(1) acquisition variables, such as count density per frame, frame rate, technologies and public pressure in general to limit radiation burdens.
or acquisition protocol; (2) processing variables, including reconstruc-
tion algorithm, filtering, automation, definition of endocardium and Computed Tomography
epicardium, definition of hypoperfused walls and valve plane; and (3) Electron beam tomography has been used in the past to study both
patient-dependent variables like heart rate variability, arrhythmia, patient function and perfusion, but technical questions prevent its clinical role.
motion, extracardiac uptake close to the LV, small and large left ven- Multidetector computed tomography (MDCT) of the heart is a rapidly
tricles, and hypoperfused areas. There is special concern in both small developing technique that is used mainly to evaluate the coronary arter-
and large ventricles because of the limited spatial resolution and the ies.38 It can also be used for evaluating global and regional ventricular
problems of assigning, either manually or automatically, a ventricular function,39 with presumably higher accuracy than SPECT,40 2D/3D
border in areas of transmural infarction and thinning where counts echocardiography and cineventriculography,41 and with good agreement
CHAPTER 16  Assessment of Cardiac Function 195

FIG. 16.4  Rest gated single-photon emission computed tomography for ventricular function analysis. Rest
perfusion is depicted in the upper row in diastole and systole, contraction is shown in the middle row, both
motion (left) and thickening (right). The bottom row shows a three-dimensional reconstruction in diastole
(left) and systole (right).

with CMR.42 The acquisition time is short and postprocessing tools There are two main methods for measuring the end-diastolic and
allow fast and semiautomatic determination of LV function parameters end-systolic volumes. The earliest semiquantitative method was an
from MDCT data with two methods, the ventricular threshold method adaptation of the echo area-length method, in which the volume of
and, in analogy to known CMR evaluation approaches, myocardial the left ventricle is assumed to constitute an ellipsoid. With a single
analysis (Fig. 16.5). It cannot be considered a first-line modality, owing apical four-chamber view, the area (A) of the LV endocardial border
to the exposure to radiation, the use of potentially nephrotoxic contrast, can be traced, as well as the length (L) from the apex to the mitral
and the low temporal resolution (80–200 ms), but it may be used for annulus. The volume (V) for systole and diastole is then easily calculated:
accessory dynamic information in patients undergoing computed tomog- V = 0.85A2/L. A more accurate adaptation uses both long-axis planes
raphy (CT) coronary angiography.43 New ultralow-dose CT scanners (vertical and horizontal long axis [VLA and HLA, respectively]) to
can acquire a study for coronary artery assessment with as low as 1 mSv44, measure two perpendicular areas and lengths: V = (0.85 × area 1 × area
but for ventricular function, retrospective ECG-gated helical acquisition 2)/smaller length (Fig. 16.6). Although this offers a simple and time-
must be used, which increases the radiation dose significantly. Some efficient volume analysis,48 it suffers from the same limitations that
reference ranges have been established for this technique.45,46 MDCT echocardiography has: the need for geometric assumptions and the
reproducibility for volumes and function is lower than that with CMR, inability to take into account regional differences in wall motion. An
with an interobserver variability of 4.2% for end-diastolic volume, 5.4% example of a distorted heart postinfarction is shown in Fig. 16.7.
for end-systolic volume, and 4.1% for LVEF.47 A better method of measuring volumes and thereby function is by
the use of Simpson’s rule. A stack of contiguous tomographic slices are
Cardiovascular Magnetic Resonance acquired that encompass the entire left ventricle. The ventricular volume
CMR has some fundamental advantages over other imaging techniques, is equal to the sum of the endocardial areas multiplied by the distance
which have fueled the growing enthusiasm for its use in clinical practice between the centers of each slice (Fig. 16.8).49 The volumes that are
and research. CMR offers accurate and reproducible tomographic, static, obtained by this method are independent of geometric assumptions
or cine images of high spatial and temporal resolution in any desired and dimensionally accurate.50 At one time, it was common to use a
plane without exposure to contrast agents or ionizing radiation. As stack of transverse images for this measurement, but although this
such, true two-chamber and four-chamber and short-axis views can makes it easy to define the mitral valve plane and thereby the true base
be easily and rapidly acquired to allow a visual, qualitative assessment of the LV,41 it is also subject to considerable partial volume effects,
of function, similar to that of echocardiography. The main advantage especially in the inferior wall. For this reason, more recently, short-axis
of CMR, however, lies in its quantitative accuracy and reproducibility. slices were employed, and nearly all sites specializing in CMR have
196 SECTION II  Ischemic Heart Disease

A B

C D
FIG. 16.5  Example of ventricular function analysis with computed tomography. A and B show, analogous
to cardiovascular magnetic resonance, myocardial analysis for left ventricular (LV) function quantification both
with ejection fraction and with wall thickening. C and D represent the ventricular threshold method for
measurement of both LV (C) and right ventricular (D) volumes.

Area-length method Biplane area-length method

L L1

A1 A2 L2
A

Volume = 0.85 (A)2/L Volume = (0.85 x A1 x A2)/L


FIG. 16.6  Uniplane (left) and biplane (right) area-length method for measurement of left ventricular volumes
and eventually function. Being a fast method, because only two planes are used, significant geometric
assumptions are made that affect accuracy and reproducibility.

adopted this practice (Fig. 16.9). The question has been raised as to long-axis views have also been proposed as an alternative to short-axis-
whether or not the right ventricular (RV) volumes should be measured derived methods because it has been suggested that they would have
in the axial orientation because this appears to have better interobserver less partial-volume effects and would be less time consuming,54 but
and intraobserver reproducibility.51,52 Yet the interstudy reproducibility this method has not gained wide acceptance so far.
of RV measurements in the short-axis orientation is good,53 with dif- In the past, to achieve full 3D coverage of the ventricle using conven-
ferences not clinically significant and in practice, this orientation allows tional free-breathing gradient echo cine sequences, a total scanning time
the LV and RV dimensions to be measured simultaneously. Rotational of 30 minutes or more was required, but on modern scanners with fast
CHAPTER 16  Assessment of Cardiac Function 197

A B

C D
FIG. 16.7  A patient with ischemic heart disease in whom the left ventricle no longer conforms to geometric
assumptions in either diastole (A, C) or systole (B, D) frames. Both the vertical long-axis (A, B) and the hori-
zontal long-axis (C, D) views are illustrated.

imaging, a single cine can be acquired in just one breath-hold of about SSFP sequences are suboptimal, usually in patients with implanted
8 to 10 seconds, with good spatial and temporal resolution, allowing devices that cause significant artifacts. SSFP sequences rephase the
the whole stack of images to be acquired in 5 to 8 minutes. This also transverse magnetization that undergoes dephasing during phase encod-
has the considerable additional advantage of reducing breathing and ing and readout between radiofrequency (RF) pulses; therefore imaging
movement artifacts. Moreover, real-time steady-state free precession occurs when all transverse and longitudinal magnetization components
(SSFP) imaging can acquire all the ventricular slices in just one breath- are at steady state. SSFP cine imaging eliminates blood saturation arti-
hold with acceptable accuracy and image quality. In patients who are facts and makes the cines independent of in-flow enhancement and
unable to hold their breath consistently or who are orthopneic, solutions based on the ratio of T2 to T1. This results, compared with GRE, in
using the same sequence with more signal averages or combined with higher signal-to-noise ratio (SNR) and substantially improved blood-
navigator echo imaging have been shown to be successful, during free myocardium contrast,61 which may allow easier delineation of the
breathing,55 with a slight increase in the acquisition duration. endocardial borders. At the epicardial border, fat-myocardium delinea-
In addition to the left ventricle, it is important to remember the tion is also improved.
right ventricle, as its function is also known to be an important deter- The SSFP sequence runs at its best with ultrafast gradients because
minant of prognosis, both in coronary artery disease56 and in heart a very short repetition time (TR) is required to reduce the sensitivity
failure,57 congenital heart disease,58 and pulmonary disease.59,60 Global RV of the sequence to movement artifact. Because of these characteristics,
function is difficult to assess adequately by echocardiography, whereas LV end-diastolic and end-systolic volumes are larger and LV mass is
radionuclide ventriculography suffers from assumptions concerning smaller with SSFP than with GRE.62 In terms of reproducibility, only
projection of overlapping structures unless research techniques such as EF shows differences favoring SSFP, whereas SSFP and GRE are equal
first-pass techniques with ultra-short half-life isotopes are used. CMR for volumes and mass reproducibility.
does not experience such problems, and RV function and mass are Nowadays, improved performance is no longer gained through
well characterized. The right ventricle is discussed in greater detail in improvements in gradient hardware. The introduction of parallel imaging,
Chapter 39. either image based such as sensitivity encoding (SENSE) or k-space-
based such as generalized autocalibrating partially parallel acquisitions,
From Gradient Echo to Steady-State Free provides alternative means for increasing acquisition speed. By using
Precession Cine Sequences information from multiple receiver coils, images can be reconstructed
Gradient recalled echo (GRE) cine sequences were the first ones to be from a sparsely sampled set of data, allowing for 2- to 3-fold accelera-
implemented, but nowadays their use is limited to situations where tion of the imaging process. However, further increases in acquisition
198 SECTION II  Ischemic Heart Disease

ED

ES

LV End Diastole 219 mL


LV End Systole 107 mL
LV Stroke Volume 112 mL
LV Ejection Fraction 51%
LV Mass 243 g
LV Mass Index 125 g/m2

FIG. 16.8  The end-diastolic (ED) and end-systolic (ES) slices from multiple contiguous short-axis cines that
encompass the left ventricle (LV), from base to apex, in a patient with ventricular dilatation and hypertrophy
from chronic aortic regurgitation. The epicardial and endocardial borders are traced, and the summation values
are shown. Note that there is one more image at end diastole11 than at end systole,10 reflecting the need to
allow for the systolic descent of the atrioventricular ring, as described in the text. Note also the ingress of
the LV outflow tract in the most basal end-diastolic image, where there is no ventricular mass, and the open
ends of the LV myocardial horseshoe are joined together to form the appropriate volume.

Long axis of left ventricle Short axis

A3

A1 A2
1
2
3
4 Diastole Systole
5
6
7
8
9
10
11 12

13
FIG. 16.9  Schematic of the left ventricular short-axis slices that encompass the entire left ventricle. Each slice
is acquired as a cine. A1, End-diastolic endocardial area; A2, end-systolic endocardial area; A3, myocardium.

speed are difficult to achieve for current clinical field strengths and acceleration) or spatial resolution for a given amount of acquisition,
typical fields of view. More recently developed methods are based on and they also enable the acquisition of 3D k-t BLAST SSFP cine sequences
temporal undersampling, k-t broad-use linear acquisition speed-up in one breath-hold that can be used for ventricular function assess-
technique (BLAST) and k-t SENSE, have been proposed that significantly ment.63 Another recently developed method is unaliasing by Fourier-
improve the performance of dynamic imaging, taking into account the encoding the overlaps using the temporal dimension (UNFOLD),64
similarity of image information at different time points during a dynamic which works by forcing aliased signals to behave in specific ways through
series. These methods improve temporal resolution (5- to 8-fold time, so unwanted signals are detected and removed, thus reducing the
CHAPTER 16  Assessment of Cardiac Function 199

FIG. 16.10  The darkening artifact because of off-resonance effect (top row, arrows) and flow artifacts
(bottom row, arrows) that are very frequent with cardiovascular magnetic resonance at 3 T.

amount of acquired data. This method can in theory be used with k-t between blood and myocardium seems to be reduced in the RV at 3.0
SENSE and k-t BLAST to accelerate the acquisition and/or to suppress T relative to LV, which may affect reproducibility of RV functional
artifacts in free breathing.65 All these techniques are very promising but evaluation.
still in development, and no specific data on their use in LV function
assessment are available. ACCURACY AND REPRODUCIBILITY OF
Steady-State Free Precession Cine Sequences at 1.5 T CARDIOVASCULAR MAGNETIC RESONANCE
vs. 3 T Magnetic Fields It is now widely accepted that CMR offers the reference standard for
There is an overall quantitative improvement in SNR for myocardium the noninvasive assessment of cardiac function, being both accurate72,73
and blood at 3 T, as well as variable increase of blood-to-myocardium (Figs. 16.11 to 16.14) and reproducible in normal as well as abnormal
contrast-to-noise ratio (CNR).66 These SNR improvements can be ventricles, both with nonbreath-hold74 and breath-hold techniques.75,76
translated into higher speed because they facilitate the use of parallel This is illustrated in Fig. 16.15, which shows the intraobserver, interob-
imaging, with higher acceleration factor and multichannel RF receiver server, and interstudy variability for volume and functional assessment
coils, with less noise and almost identical parameters for SSFP imaging by breath-hold CMR in dilated and normal ventricles, compared with
at both magnetic fields. However, accelerated acquisition results in the conventional cine CMR.75
loss of SNR and in the introduction of noise nonuniformly, and there CMR has been shown to have higher reproducibility than 2DE for LV
are some challenges that need to be addressed, including B0 and RF end-systolic volume (4.4% to 9.2% vs. 13.7% to 20.3%, P < .001), LVEF
inhomogeneity, limitation of flip angle and minimum achievable TR, (2.4% to 7.3% vs. 8.6% to 19.4%, P < .001), and LV mass measurements
longer T1, shortened T2* and off-resonance effects because of mag- (2.8% to 4.8% vs. 11.6% to 15.7%, P < .001), and this higher reproduc-
netic susceptibility gradients,67 which together combine to offset the ibility allows for sample size reductions of 55% to 93%.77 The excellent
advantages from SSFP at 3 T, when a reproducible image quality for reproducibility of CMR versus 2DE can be illustrated by considering the
cine SSFP is important for the evaluation of ventricular function (Fig. sample size required for a drug trial designed to show a 10 g decrease in
16.10). Comparative analyses of LV function indexes, mass, and volume LV mass with antihypertensive treatment. A direct comparison of CMR
using cine SSFP has shown no significant difference in values obtained with 2DE for reproducibility has shown that for an 80% power and a P
at 3.0 T compared with 1.5 T.68,69 Nonetheless, reference values for value of .05, the sample size required would be 505 patients with 2DE
LV function parameters have been published.70,71 Importantly, CNR but only 14 patients with CMR.78 Equally, we have found that to show
200 SECTION II  Ischemic Heart Disease

200 250

150 200
CMR LV volume (mL)

Cadaver LV mass (g)


100 150

50 100
R = 0.997 R = 0.99

0 50
0 50 100 150 200 50 100 150 200 250
LV cast volume (mL) CMR LV mass (g)
FIG. 16.11  Validation of left ventricular (LV) volume measurements by FIG. 16.13  Validation of left ventricular (LV) mass with comparison of
cardiovascular magnetic resonance (CMR). CMR-derived volume (vertical cardiovascular magnetic resonance (CMR)-derived mass against human
scale) is compared with the displacement volume of the LV casts from cadaver hearts. (From Katz J, Millikem MC, Stray-Gunderson J, et al.
human postmortem hearts. There is excellent accuracy, and the regres- Estimation of human myocardial mass with MR imaging. Radiology.
sion line slope is close to identity. (From Rehr RB, Malloy CR, Filipchuk 1988;169:495–498.)
NG, et al. Left ventricular volumes measured by MR imaging. Radiology.
1985;156:717–719.)
190

120

110
Cadaver RV mass (g)

170
100

90
LVSV (mL)

80
150
70 R = 0.97

60
R = 0.94
50 130
130 150 170 190
40
CMR RV mass (g)
40 50 60 70 80 90 100 110 120
FIG. 16.14  Validation of right ventricular (RV) mass with comparison
RVSV (mL) of cardiovascular magnetic resonance (CMR)-derived mass against post-
FIG. 16.12  Validation of right and left ventricular stroke volumes (RVSV mortem bovine hearts. (From Katz J, Whang J, Boxt LM, Barst RJ.
and LVSV, respectively) in vivo using cardiovascular magnetic resonance Estimation of right ventricular mass in normal subjects and in patients
(CMR) ventricular volume analysis. There is an excellent agreement with primary pulmonary hypertension by nuclear magnetic resonance
between the stroke volumes of both ventricles, which is strong evidence imaging. J Am Coll Cardiol. 1993;21:1475–1481.)
that both measurements are accurate, because they are equivalent in
vivo in the absence of valve regurgitation or shunting. (From Longmore
DB, Klipstein RH, Underwood SR, et al. Dimensional accuracy of mag- measures but only significantly for EF.79 Three-dimensional echocar-
netic resonance in studies of the heart. Lancet. 1985;1:1360–1362.) diography (3DE) and administration of contrast are claimed to improve
accuracy and reproducibility. In a study80 that compared 2DE, 3DE,
and contrast-enhanced 3DE (CE3DE) with CMR in subjects with poor
a 5% difference in EF with a 90% power and a P value of 0.05 would acoustic window, CE3DE showed the best agreement with CMR and
require only 7 normal subjects or 5 patients with dilated ventricles.77 was claimed to be the only acceptable alternative for CMR. In terms
Similarly, CMR measurements of RV function parameters both in healthy of reproducibility, another study81 compared interreader variability for
subjects and in patients show good interstudy reproducibility (4.2% to LV EF measurement with 2DE, 3DE, CE2DE, CE3DE, and CMR and
7.8% for RV end-diastolic volume, 8.1% to 18.1% for RV end-systolic obtained a variability of 14.3% for 2DE and 3DE, 8.0% for CE2DE,
volume, 4.3% to 10.4% for RV ejection fraction and 7.8% to 9.4% 7.4% for CE3DE and 7.9% for CMR, then showing that 3DE requires
for RV mass), which was lower than that for the left ventricle for all contrast application as much as 2DE to reduce interreader variability.
CHAPTER 16  Assessment of Cardiac Function 201

14 A PRACTICAL GUIDE TO FUNCTIONAL


CARDIOVASCULAR MAGNETIC RESONANCE
Intraobserver % variability
12

10 In modern medicine, a balance must be struck between the information


8 that can be gained from an investigation and the resources it demands.
The following protocol is designed to be as efficient as possible in
6 gaining the volumetric data from the ventricles of the heart83 (see Chapter
4 15). Fig. 16.16 illustrates the sequence of pilot images used to achieve
imaging in the long axis of the left ventricle and thereby the true short
2 axis. A coronal pilot is first taken and used to acquire transverse pilots,
0 which show both the mitral valve and the apex of the left ventricle. By
Bellenger Bellenger Bellenger Semelka Bogaert taking a plane through the center of the mitral valve (halfway between
A normal LVH DCM normal normal the back end of the septum and the back end of the lateral wall) and
the tip of the apex, the vertical long axis (VLA) is acquired. This VLA
14 is used to plan the horizontal long axis (HLA), by again using a plane
through the center of the mitral valve (halfway between the back of
12
the anterior and inferior walls) and the tip of the apex. It should be
Interobserver % variability

10 noted that it is common to find centers describing planes that are


parallel to the septum for the VLA and parallel to the inferior wall for
8 the HLA, but these are not correct because they are likely to lead to the
long-axis plane not passing through the center of the basal ring of the
6
LV, and they may lead to an offset from the tip of the apex, which is
4 also undesirable. This can lead to problems planning the short-axis
cuts to adequately cover the full extent of left and right ventricles; in
2 addition, it reduces the reproducibility of the short-axis plane position-
0
ing for repeated studies. Finally, the short-axis slices are placed on the
HLA to encompass the heart. To achieve the most reliable results, which
rm lka

H ka

B
C ka
a r

H er

M r

al t
rm ge

C e

rm er
LV ng

D leng

LV el

D el

are the most reproducible, attention to detail is required. First, if the


no me

no oga
no llen

Se l
a
Be l

m
M
lle

Se

Se
l

B
Be

Be

short-axis cuts are to be acquired by using a breath-hold cine sequence,


then the VLA and HLA must also have been acquired by using a breath-
14 hold and at end expiration. Second, in using breath-hold techniques,
it is more reproducible to ask the patient to hold the breath at end
12
expiration rather than elsewhere in the respiratory cycle; this applies
Interstudy % variability

10 to the pilots as well as to the cines.84 Third, the first short-axis plane
should be placed at the base of the heart, covering the most basal
8 portion of the left and right ventricles just forward of the atrioventricular
6
(AV) ring, and it should be placed on the end-diastolic HLA image.
Finally, further short-axis planes should then be planned to move api-
4 cally from this plane until the apex is encompassed. Although it is
possible to acquire the VLA and HLA as single pilot images instead of
2
cines, there is little practical merit in this because the time for two
0 breath-hold cines is small, and the contraction pattern in these two
Bellenger Bellenger Bogaert Semelka planes is very useful during qualitative assessment of ventricular func-
C normal DCM normal DCM
tion. Nowadays, with the development of 3D postprocessing software
solutions for analyzing the cines, these long-axis cines are mandatory.
EDV EF
Also, full 3D analysis of atria as well as ventricles is simple and practical
ESV Mass
with automated analysis, in which case cines encompassing the entire
FIG. 16.15  The intraobserver (A), interobserver (B), and interstudy heart should be acquired.
(C) percentage variability for end-diastolic volume (EDV), end-systolic Following are some technical tips:
volume (ESV), ejection fraction (EF), and left ventricular mass (Mass). • Modern CMR scanners with faster gradients allow a shorter TR and
Results using breath-hold gradient echo from our center in patients with
echo time (TE), which improve the breath-hold length so this is no
heart failure and dilated ventricles (DCM) and left ventricular hypertrophy
(LVH) are compared with breath-hold gradient echo in normals (Bogaert)
longer generally a problem for patients. If necessary, increasing the
and traditional, slower gradient echo cine imaging (Semelka).65 Overall, phase-encoded grouping (PEG) can reduce the breath-hold, but a
the results are very similar (probably favoring the breath-hold imaging), compromise is reached because fewer phases will be captured with
both between techniques and between different population groups. a higher PEG. The number of phases should be at least 15 to give
adequate information on wall motion and cover end systole. Decreas-
ing the field-of-view will also reduce the breath-hold time but may
With respect to the right ventricle, a recent study has shown that 3DE result in some wrap-around occurring at the edges of the image. If
underestimates RV volumes, with no significant differences in ejection this remains remote from the heart, it may be considered an accept-
fraction.82 Reproducibility has significant implications for research and able compromise. Typically, the best results are seen when the time
in particular for pharmaceutical companies, for which CMR offers a between cine phases is <30 ms, and this yields a cine with approxi-
more cost-effective and time-effective research tool. mately 25 to 40 frames in clinical practice.
202 SECTION II  Ischemic Heart Disease

A B C

E D
FIG. 16.16  Pilot images used to achieve the true short axis of the left ventricle. A coronal pilot (A) is first
acquired and used to pilot the transverse image (B). The vertical long axis (C) is obtained from this and,
subsequently, the horizontal long axis (D) is obtained, upon which a stack of short-axis cines are placed (E).

• The ECG gating can be prospective or retrospective. In prospective end-diastolic, end-expiratory breath-hold HLA image just forward
gating, images are acquired during systole and the beginning of of the AV ring, the first short-axis cine will, by definition, contain
diastole, whereas with retrospective gating, images from the whole end-diastolic volume and mass within both ventricles. However, at
cardiac cycle are obtained. Although the breath-hold with prospec- end systole, the basal slice will include only atrium, owing to descent
tive gating is shorter, it is advisable to use retrospective gating, which of the AV ring, and, mostly, the systolic area in the basal cine is not
allows diastolic function to be assessed. In patients with arrhythmia, included in the analysis of the systolic volume. In general, the next
retrospective gating may sometimes yield better image quality, but slice down contains both end-diastolic and end-systolic volume. An
when ventricular ectopics are present, a good strategy is to use pro- alternative to this rigorous approach is to oversample with short-axis
spective gating with an acquisition window shorter than the coupling slices into the atrium and attempt to retrospectively differentiate
interval, so the acquisition is not disturbed by the ectopics. ventricle from atrium on the basis of the degree of descent of the
• A slice thickness of 8 mm at 1.5 T provides adequate spatial resolu- AV ring in the long-axis images and whether the chamber dilates
tion without overly increasing the number of slices and thereby the or contracts in systole. In general, we prefer not to oversample but
analysis time. It also limits partial volume effects. A 2 mm slice gap to ensure that the first basal slice is acquired correctly because this
is commonly used to allow easy calculation of volumes because the leads to a reproducible approach and is more time efficient for both
center of each slice is then 1 cm apart. There are no formal studies acquisition and analysis and because oversampling relies more heavily
to aid in the choice of slice thickness, but 8 mm is a reasonable on good image quality to differentiate atrium from ventricle. Also,
consensus. Some centers prefer thinner slices, but it is important to although at some point it has been suggested that a binary cutoff
maintain acceptable SNR and image quality. Three-dimensional should be used for analysis of the basal slice, it has been shown85
imaging may eventually allow more partitions with thinner slices. that fully inclusive quantification, rather than binary cutoffs that
• Analysis of the short-axis slices is relatively straightforward, provided omit basal LV myocardium, yields smallest CMR discrepancy with
that the quality of the images is reasonable (mainly depending on echocardiography-measured LV mass and nonsignificant differences
accurate ECG gating and good breath-holding). The main source with necropsy-measured LV weight.
of error is in separating the ventricles from the atria. Identifying • Papillary muscles and endocardial trabeculae should be excluded
this basal slice is made more difficult by the through-plane descent from the LV volume and included in the LV mass. Although there
of the AV ring in systole, which is usually about 1 cm. This makes is no clear consensus at present, LV mass is usually taken from the
the placement of the first, most basal short-axis slice very important. end-diastolic images. LV mass by CMR varies by a small amount
By ensuring that this basal slice is carefully positioned on the from end diastole to end systole, and this may be due to expulsion
CHAPTER 16  Assessment of Cardiac Function 203

of intramyocardial blood into the venous system, or to partial volume VLA and HLA cines to define accurately the mitral valve position at
effects. The reproducibility of LV diastolic volumes is in general each phase in the cardiac cycle. These softwares provide a fast analysis
better than at end systole because the volumes are larger and delin- of ventricular dimensions in most patients, leaving manual analysis
eation of trabeculae is easier, and this is probably as good a reason limited to studies with poor image quality because of arrhythmia
as any for working from the end-diastolic images to determine mass, or difficulty in breath-holding.
but in addition, if the routine above is followed, there may be doubt
as to whether LV mass is present in the most basal LV slice at end OTHER CARDIOVASCULAR MAGNETIC
systole if its quality is less than ideal, but by definition, LV mass is RESONANCE MEASURES OF GLOBAL FUNCTION:
always present at end diastole in the most basal slice. BRIEF SYNOPSIS
• Normal reference ranges have been reported for LV86,87 and RV88
dimensions and systolic function. This is important because most Systolic Function
LV and RV parameters are dependent on gender, age, and body There are other ways to derive important functional information from
surface area. The reference data are shown in Tables 16.1 (LV) and the heart besides volumetry alone. For example, flow in the major vessels
16.2 (RV) and Fig. 16.17 (LV). is easily measured with great accuracy by using velocity mapping.89 If
• For the very best interstudy reproducibility for follow-up studies the aortic flow is measured over a complete cardiac cycle, this represents
and drug trials, it is necessary to go the extra mile in the analysis the LV stroke volume. The RV stroke volume can likewise be found
and always have the first set of cines with the regions of interest from flow in the pulmonary artery. Cardiac output can be easily derived
(ROIs) on screen alongside the follow-up cines during the analysis. by using the following formula: cardiac output = stroke volume × heart
At least a printout on paper of the first study and the ROIs used for rate. The measurement of stroke volume, which is useful in valve
analysis is required for comparison. For this reason, we routinely disease90,91 and for determining the need for surgery in cardiovascular
print out the diastolic and systolic ROIs in a systematic way for shunting, is noninvasive and quantitative. The peak flow velocity and
every patient on whom volumes are analyzed. acceleration, which are derived from the flow curves, have been used
• Currently, a number of analysis softwares for automated postprocess- to determine the burden of ventricular ischemia during dobutamine
ing of the cines with a 3D analytical approach are available that take stress (see Chapter 17). Recently, tissue tracking technologies such as
into account the motion in systole of the AV ring, by incorporating speckle tracking echocardiography and feature tracking CMR (CMR-FT)

TABLE 16.1  Normal SSFP LV Volumes, Systolic Function, and Mass (Absolute and Indexed
to BSA) by Age Decile (Mean, 95% CI) in Males and Females
20–29 Years 30–39 Years 40–49 Years 50–59 Years 60–69 Years 70–79 Years
Males ABSOLUTE VALUES
EDV (mL), SD 21 167 (126–208) 163 (121–204) 159 (117–200) 154 (113–196) 150 (109–191) 146 (105–187)
ESV (mL), SD 11 58 (35–80) 56 (33–78) 54 (31–76) 51 (29–74) 49 (27–72) 47 (25–70)
SV (mL), SD 14 109 (81–137) 107 (79–135) 105 (77–133) 103 (75–131) 101 (73–129) 99 (71–127)
EF (%), SD 4.5 65 (57–74) 66 (57–75) 66 (58–75) 67 (58–76) 67 (58–76) 68 (59–77)
Mass (g), SD 20 148 (109–186) 147 (109–185) 146 (108–185) 146 (107–184) 145 (107–183) 144 (106–183)

INDEXED TO BSA
2
EDV/BSA (mL/m ), SD 9.0 86 (68–103) 83 (66–101) 81 (64–99) 79 (62–97) 77 (60–95) 75 (58–93)
ESV/BSA (mL/m2), SD 5.5 30 (19–41) 29 (18–39) 27 (17–38) 26 (15–37) 25 (14–36) 24 (13–35)
SV/BSA (mL/m2), SD 6.1 56 (44–68) 55 (43–67) 54 (42–66) 53 (41–65) 52 (40–64) 51 (39–63)
EF/BSA (%/m2), SD 3.3 34 (28–40) 34 (28–40) 34 (28–40) 34 (28–41) 34 (28–41) 34 (28–41)
Mass/BSA (g/m2), SD 8.5 76 (59–93) 75 (59–92) 75 (58–91) 74 (57–91) 73 (57–90) 73 (56–89)

Females ABSOLUTE VALUES


EDV (mL), SD 21 139 (99–179) 135 (94–175) 130 (90–171) 126 (86–166) 122 (82–162) 118 (77–158)
ESV (mL), SD 9.5 48 (29–66) 45 (27–64) 43 (25–62) 41 (22–59) 39 (20–57) 36 (18–55)
SV (mL), SD 14 91 (63–119) 89 (61–117) 87 (59–115) 85 (57–113) 83 (56–111) 81 (54–109)
EF (%), SD 4.6 66 (56–75) 66 (57–75) 67 (58–76) 68 (59–77) 69 (60–78) 69 (60–78)
Mass (g), SD 18 105 (69–141) 106 (70–142) 107 (71–143) 108 (72–144) 109 (73–145) 110 (74–146)

INDEXED TO BSA
2
EDV/BSA (mL/m ), SD 8.7 82 (65–99) 79 (62–96) 76 (59–93) 73 (56–90) 70 (53–87) 67 (50–84)
ESV/BSA (mL/m2), SD 4.7 28 (19–37) 27 (17–36) 25 (16–34) 24 (14–33) 22 (13–31) 21 (12–30)
SV/BSA (mL/m2), SD 6.2 54 (42–66) 53 (40–65) 51 (39–63) 50 (37–62) 48 (36–60) 47 (34–59)
EF/BSA (%/m2), SD 4.7 39 (30–48) 39 (30–49) 40 (30–49) 40 (31–49) 40 (31–49) 40 (31–49)
Mass/BSA (g/m2), SD 7.5 62 (47–77) 62 (47–77) 63 (48–77) 63 (48–78) 63 (48–78) 63 (49–78)

BSA, Body surface area; CI, confidence interval; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic volume; LV, left ventricular;
SD, standard deviation; SSFP, steady-state free precession; SV, stroke volume.
204 SECTION II  Ischemic Heart Disease

TABLE 16.2  Normal SSFP RV Volumes, Systolic Function, and Mass (Absolute and Indexed
to BSA) by Age Decile (Mean, 95% CI) in Males and Females
20–29 Years 30–39 Years 40–49 Years 50–59 Years 60–69 Years 70–79 Years
Males ABSOLUTE VALUES
EDV (mL), SD 25.4 177 (127–227) 171 (121–221) 166 (116–216) 160 (111–210) 155 (105–205) 150 (100–200)
ESV (mL), SD 15.2 68 (38–98) 64 (34–94) 59 (29–89) 55 (25–85) 50 (20–80) 46 (16–76)
SV (mL), SD 17.4 108 (74–143) 108 (74–142) 107 (73–141) 106 (72–140) 105 (71–139) 104 (70–138)
EF (%), SD 6.5 61 (48–74) 63 (50–76) 65 (52–77) 66 (53–79) 68 (55–81) 70 (57–83)
Mass (g), SD 14.4 70 (42–99) 69 (40–97) 67 (39–95) 65 (37–94) 63 (35–92) 62 (33–90)

INDEXED TO BSA
2
EDV/BSA (mL/m ), SD 11.7 91 (68–114) 88 (65–111) 85 (62–108) 82 (59–105) 79 (56–101) 75 (52–98)
ESV/BSA (mL/m2), SD 7.4 35 (21–50) 33 (18–47) 30 (16–45) 28 (13–42) 25 (11–40) 23 (8–37)
SV/BSA (mL/m2), SD 8.2 56 (40–72) 55 (39–71) 55 (39–71) 54 (38–70) 53 (37–69) 52 (36–69)
EF/BSA (%/m2), SD 4 32 (24–40) 32 (25–40) 33 (25–41) 34 (26–42) 35 (27–42) 35 (27–43)
Mass/BSA, (g/m2), SD 6.8 36 (23–50) 35 (22–49) 34 (21–48) 33 (20–46) 32 (19–45) 31 (18–44)

Females ABSOLUTE VALUES


EDV (mL), SD 21.6 142 (100–184) 136 (94–178) 130 (87–172) 124 (81–166) 117 (75–160) 111 (69–153)
ESV (mL), SD 13.3 55 (29–82) 51 (25–77) 46 (20–72) 42 (15–68) 37 (11–63) 32 (6–58)
SV (mL), SD 13.1 87 (61–112) 85 (59–111) 84 (58–109) 82 (56–108) 80 (55–106) 79 (53–105)
EF (%), SD 6 61 (49–73) 63 (51–75) 65 (53–77) 67 (55–79) 69 (57–81) 71 (59–83)
Mass (g), SD 10.6 54 (33–74) 51 (31–72) 49 (28–70) 47 (26–68) 45 (24–66) 43 (22–63)

INDEXED TO BSA
2
EDV/BSA (mL/m ), SD 9.4 84 (65–102) 80 (61–98) 76 (57–94) 72 (53–90) 68 (49–86) 64 (45–82)
ESV/BSA (mL/m2), SD 6.6 32 (20–45) 30 (17–43) 27 (14–40) 24 (11–37) 21 (8–34) 19 (6–32)
SV/BSA (mL/m2), SD 6.1 51 (39–63) 50 (38–62) 49 (37–61) 48 (36–60) 46 (34–58) 45 (33–57)
EF/BSA (%/m2), SD 5.2 37 (27–47) 38 (27–48) 38 (28–49) 39 (29–49) 40 (30–50) 41 (31–51)
Mass/BSA (g/m2), SD 5.2 32 (22–42) 30 (20–40) 29 (19–39) 27 (17–37) 26 (16–36) 24 (14–35)

BSA, Body surface area; CI, confidence interval; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic volume; RV, right ventricular;
SD, standard deviation; SSFP, steady-state free precession; SV, stroke volume.

have enhanced the noninvasive assessment of myocardial deformation throughout the cardiac cycle. The same approach could be applied to
in research and clinical practice. CMR-FT has emerged as a useful tool the analysis of the pulmonary venous flow, but the lack of a dedicated
for the quantitative evaluation of global and regional cardiac function, postprocessing tool makes routine use of the PVF difficult. Cine CMR
through quantification of biventricular mechanics from measures of also allows direct visualization of the abnormal diastolic filling in dilated
myocardial deformation. The technology is common to both modalities, ventricles with inflow directed not toward the apex but toward the free
and derived parameters to describe myocardial mechanics are similar wall, giving rise to a well-developed circular flow pattern turning back
but with different accuracies and reproducibilities.92 CMR-FT provides toward the septum and outflow tract, persisting through diastole.99
a fast assessment of longitudinal strain,93 which is an independent pre- With the current analysis softwares it is possible to measure volumes
dictor of survival in several cardiomyopathies,94 as well as circumferential over a complete cardiac cycle, obtaining a volume curve whose first
and radial strain. Also, normal values of references have been published.95 derivative is the flow curve; normal values of LV diastolic function with
CMR-FT has a reasonable agreement with myocardial tagging96 and a this method have been described and are shown in Fig. 16.18.86 With
there is a high correlation with 2DE and 3DE.97 Although CMR-FT is the same approach, RV diastolic function curves have been produced
a promising technique for evaluation of systolic and diastolic, global, and are shown in Chapter 39.88 Left atrial dilatation is also a good
and regional function, at present more work is needed to accelerate marker of diastolic dysfunction; atrial volumes are easy to obtain with
analysis, which is still time consuming, to assess differences in results CMR.100 Studies of myocardial velocity in diastole with tissue phase
obtained with different software packages, and, more importantly, on contrast allow evaluation of myocardial velocities during diastole and
standardization of acquisition and analysis, applicability, and reference systole, in the same way as Doppler tissue imaging does in echocar-
values. This is discussed in more detail in Chapter 22. diography.101 Other techniques such as tagging, spectroscopy, or elas-
tography have been used to assess myocardial relaxation and/or stiffness
Diastolic Function but more experience is needed before these techniques can be promoted.
For diastolic function, phase-contrast CMR can be applied to measure, More details of the assessment of diastolic function can be found in
analogous to 2DE, both mitral inflow at the tip of the mitral valve Chapter 39.
leaflets and tissue velocity at the annulus. This technique is fast and
reproducible, with good agreement with Doppler echocardiography.98 Regional Function
Still, a mitral valve tracking technique should be implemented to over- Cine CMR sequence allows a qualitative assessment of regional cardiac
come the limitation of not measuring velocities at the same level function in the same way as with echocardiography, but with improved
CHAPTER 16  Assessment of Cardiac Function 205

MALES

volume/BSA - males (mL/m2)

volume/BSA - males (mL/m2)


120 50
110 +95% CI +95% CI
40
100

LV end-diastolic

LV end-systolic
Mean Mean
90 30
80 –95% CI
70 –95% CI 20
60
10
50
40 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

90 120
110
fraction - males (%)

80

LV mass/BSA -
+95% CI 100 +95% CI

males (g/m2)
LV ejection

70 90
Mean
80 Mean
60 –95% CI 70
–95% CI
60
50
50
40 40
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)
FEMALES
volume/BSA - females (mL/m2)

volume/BSA - females (mL/m2)

120 50
110
+95% CI 40 +95% CI
LV end-diastolic

100
LV end-systolic

90 Mean 30 Mean
80 –95% CI
70 –95% CI 20
60
10
50
40 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

90 120
fraction - females (%)

110
80 +95% CI
LV mass/BSA -
females (g/m2)

100
LV ejection

70 Mean 90
80 +95% CI
60 –95% CI 70 Mean
60
50 –95% CI
50
40 40
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)
FIG. 16.17  Left ventricular (LV) volumes, mass, and function in systole normalized to age and body surface
area (BSA) for males and females. CI, Confidence interval.

image quality and a lower loss of nonvisualized segments. In addition, Studies using real-time CMR are now being published; these too show
it is possible to image routinely in the true long and short axis of the that CMR is superior to dobutamine stress echocardiography in patients
heart without compromises that result from restricted angulation caused with limited acoustic access.106,107
by awkward acoustic windows. This wall motion analysis can be per- Several methods based on cine CMR have been suggested to provide
formed at rest, with low-dose dobutamine for detection of viable myo- quantitative assessment of wall motion and wall thickening.108 In reality,
cardium102,103 and high-dose dobutamine for detection of ischemia.104,105 however, myocardial dynamics are more complicated than simple
206 SECTION II  Ischemic Heart Disease

MALES

700 400

rate/BSA - males (mL/s/m2)

rate/BSA - males (mL/s/m2)


600

LV active peak filling


+95% CI

LV early peak filling


500 300
+95% CI
Mean
400
200
300 –95% CI Mean
200 100 –95% CI
100
0 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

8
LV early/active peak filling 7 +95% CI
rate ratio - males 6
5
4 Mean
3
2
1
–95% CI
0
20 30 40 50 60 70 80
Age (years)
FEMALES
rate/BSA - females (mL/s/m2)

rate/BSA - females (mL/s/m2)

700 400
600 +95% CI
LV active peak filling
LV early peak filling

500 300
Mean +95% CI
400
200
300 –95% CI Mean
200 100 –95% CI
100
0 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

8 +95% CI
LV early/active peak filling

7
rate ratio - females

6
5 Mean
4
3
2
1 –95% CI
0
20 30 40 50 60 70 80
Age (years)
FIG. 16.18  Left ventricular (LV) function in diastole normalized to age and body surface area (BSA) for males
and females. CI, Confidence interval.

thickening and 2D motion because of a complex interaction of contrac- using the relation between local loading and deformation, a measure
tion, expansion, twisting, and through-plane motion. This is best mea- of local performance is produced. Tagging is discussed in more detail
sured by the process of tagging,109,110 which quantifies local deformation in Chapter 22. Also, as discussed earlier, myocardial phase contrast and
parameters and accurately measures several determinants of local loading CMR-FT may allow for quantification of local myocardial deformation
conditions, such as wall thickness and curvature. These factors, combined in systole and diastole, with potential applicability in studies for detec-
with systolic blood pressure, give a measure of local loading, and by tion of ischaemia and viability (Fig. 16.19).
CHAPTER 16  Assessment of Cardiac Function 207

Longitudinal strain (%) Radial strain (%) Circumferential strain (%)

FIG. 16.19  Example of strain quantification with cardiovascular magnetic resonance feature tracking. Lon-
gitudinal, radial, and circumferential strain can be measured with this technique. Although longitudinal strain
is usually given as a global value for the whole left ventricle, circumferential and, especially, radial strain can
be provided on a segmental basis. The same approach can be used for the right ventricle and the left atrium.
BSA, Body surface area; CI, confidence interval; LV, left ventricular.

THE FUTURE
mechanics either with CMR-FT or other approaches is expected. As
CMR offers a rapid, accurate, and reproducible assessment of cardiac CMR becomes faster, acquisition protocols will become shorter and
function that is free of geometric assumptions and is truly noninvasive. this will make the technique more cost effective for assessment of car-
At present, CMR analysis of cardiac function is reasonably fast. Auto- diovascular disease.
mated analysis softwares are already in use which allow for a fast 3D
approach to ventricular measurement. Strategies to further decrease
breath-hold time and implementation of 3D volume cine acquisitions
REFERENCES
should be achievable. Generalization of quantification of myocardial A full reference list is available online at ExpertConsult.com
CHAPTER 16  Assessment of Cardiac Function 207.e1

REFERENCES 19. Malm S, Frigstad S, Sagberg E, Larsson H, Skjaerpe T. Accurate and


reproducible measurement of left ventricular volume and ejection
1. Schocken DD, Arrieta MI, Leaverton PE, Ross EA. Prevalence and fraction by contrast echocardiography: a comparison with magnetic
mortality rate of congestive heart failure in the United States. J Am Coll resonance imaging. J Am Coll Cardiol. 2004;44:1030–1035.
Cardiol. 1992;20:301–306. 20. Hoffmann R, von Bardeleben S, ten Cate F, et al. Assessment of systolic
2. Peikes D, Chen A, Schore J, Brown R. Effects of care coordination on left ventricular function: a multi-centre comparison of
hospitalization, quality of care, and health care expenditures among cineventriculography, cardiac magnetic resonance imaging, unenhanced
Medicare beneficiaries: 15 randomized trials. JAMA. 2009;301:603–618. and contrast-enhanced echocardiography. Eur Heart J. 2005;26:
3. Mozaffarian D, Benjamin EJ, Go AS, et al; on behalf of the American 607–616.
Heart Association Statistics Committee and Stroke Statistics 21. Thavendiranathan P, Liu S, Verhaert D, et al. Feasibility, accuracy, and
Subcommittee. Heart disease and stroke statistics—2015 update. A reproducibility of real-time full-volume 3D transthoracic
Report From the American Heart Association. Circulation. 2015;131:e535. echocardiography to measure LV volumes and systolic function: a fully
4. El Aidi H, Adams A, Moons KG, et al. Cardiac magnetic resonance automated endocardial contouring algorithm in sinus rhythm and atrial
imaging findings and the risk of cardiovascular events in patients with fibrillation. JACC Cardiovasc Imaging. 2012;5:239–251.
recent myocardial infarction or suspected or known coronary artery 22. Corsi C, Lang RM, Veronesi F, et al. Volumetric quantification of global
disease: a systematic review of prognostic studies. J Am Coll Cardiol. and regional left ventricular function from real-time three-dimensional
2014;63:1031–1045. echocardiographic images. Circulation. 2005;111:1161–1170.
5. Dunn FG, Pringle SD. Sudden cardiac death, ventricular arrhythmias 23. Hoffmann R, Barletta G, von Bardeleben S, et al. Analysis of left
and hypertensive left ventricular hypertrophy. J Hypertens. ventricular volumes and function: a multicenter comparison of cardiac
1993;11:1003–1010. magnetic resonance imaging, cine ventriculography, and unenhanced
6. Rahimi K, Bennett D, Conrad N, et al. Risk prediction in patients with and contrast-enhanced two-dimensional and three-dimensional
heart failure: a systematic review and analysis. JACC Heart Fail. echocardiography. J Am Soc Echocardiogr. 2014;27:292–301.
2014;2:440–446. 24. Voigt JU, Pedrizzetti G, Lysyansky P, et al. Definitions for a common
7. Marantz PR, Tobin JN, Wassertheil-Smoller S, et al. The relationship standard for 2D speckle tracking echocardiography: consensus
between left ventricular systolic function and congestive heart failure document of the EACVI/ASE/Industry Task Force to Standardize
diagnosed by clinical criteria. Circulation. 1988;77:607–618. Deformation Imaging. J Am Soc Echocardiogr. 2015;28:183–193.
8. O’Keefe JH, Zinsmeister AR, Gibbons RJ. Value of normal 25. Thavendiranathan P, Poulin F, Lim KD, Plana JC, Woo A, Marwick TH.
electrocardiographic findings in predicting resting left ventricular Use of myocardial strain imaging by echocardiography for the early
function in patients with chest pain and suspected coronary artery detection of cardiotoxicity in patients during and after cancer
disease. Am J Med. 1989;86:658–662. chemotherapy. J Am Coll Cardiol. 2014;63(25):2751–2768.
9. Allison JD, Flickinger FW, Wright CJ, et al. Measurement of left 26. Hendel RC, Berman DS, Di Carli MF, et al. ACCF/ASNC/ACR/AHA/
ventricular mass in hypertrophic cardiomyopathy using MRI: ASE/SCCT/SCMR/SNM 2009 appropriate use criteria for cardiac
comparison with echo. Magn Reson Imaging. 1993;11:329–334. radionuclide imaging. A report of the American College of Cardiology
10. Kronik G, Slany J, Mosslacher H. Comparative value of eight M-mode Foundation Appropriate Use Criteria Task Force, the American Society
echocardiographic formulas for determining left ventricular stroke of Nuclear Cardiology, the American College of Radiology, the American
volume. Circulation. 1979;60:1308–1316. Heart Association, the American Society of Echocardiography, the
11. Teichholz LE, Kreulen T, Herman MV, Gorlin R. Problems in Society of Cardiovascular Computed Tomography, the Society for
echocardiographic volume determinations: echocardiographic- Cardiovascular Magnetic Resonance, and the Society of Nuclear
angiographic correlations in the presence or absence of asynergy. Am J Medicine endorsed by the American College of Emergency Physicians. J
Cardiol. 1976;37:7–11. Am Coll Cardiol. 2009;53(23):2201–2229.
12. Bellenger NG, Pennell DJ. Comparison of nuclear and non-nuclear 27. Plana JC, Galderisi M, Barac A, et al. Expert consensus for
techniques for the combined assessment of perfusion and function. In: multimodality imaging evaluation of adult patients during and after
Germano G, Berman DS, eds. Clinical Gated Cardiac SPECT. New York: cancer therapy: a report from the American Society of
Futura; 1999:349–377. Echocardiography and the European Association of Cardiovascular
13. Bellenger NG, Marcus N, Davies LC, Yacoub M, Banner N, Pennell DJ. Imaging. Eur Heart J Cardiovasc Imaging. 2014;15:1063–1093.
Left ventricular function and mass after orthotopic heart 28. Lapeyre AC, Klodas E, Rogers PJ, et al. Quantitation of regional ejection
transplantation: a comparison of cardiovascular magnetic resonance fractions using gated tomographic imaging with 99mTc-sestamibi.
with echocardiography. J Heart Lung Transplant. 2000;19:444–452. Chest. 2005;127:778–786.
14. Bellenger NG, Burgess M, Ray SG, et al; on behalf of the CHRISTMAS 29. Dona M, Massi L, Settimo L, et al. Prognostic implications of post-stress
Steering Committee and Investigators. Comparison of left ventricular ejection fraction decrease detected by gated SPECT in the absence of
ejection fraction and volumes in heart failure by two-dimensional stress-induced perfusion abnormalities. Eur J Nucl Med Mol Imaging.
echocardiography, radionuclide ventriculography and cardiovascular 2011;38:485–490.
magnetic resonance: are they interchangeable? Eur Heart J. 2000;21: 30. Johnson LL, Verdesca SA, Aude WY, et al. Postischemic stunning can
1387–1396. affect left ventricular ejection fraction and regional wall motion on
15. Spencer KT, Bednarz J, Mor-Avi V, DeCara J, Lang RM. Automated post-stress gated sestamibi. J Am Coll Cardiol. 1997;30:1641–1648.
endocardial border detection and evaluation of left ventricular function 31. Iskandrian AE, Germano G, van Decker W, et al. Validation of left
from contrast-enhanced images using modified acoustic quantification. J ventricular volume measurements by gated SPECT 99mTc-labeled
Am Soc Echocardiogr. 2002;15:777–781. sestamibi imaging. J Nucl Cardiol. 1998;5:574–578.
16. Amico AF, Lichtenberg GS, Reisner SA, et al. Superiority of visual versus 32. Skala T, Hutyra M, Vaclavik J, et al. Prediction of long-term reverse left
computerized echocardiography estimation of radionuclide left ventricular remodeling after revascularization or medical treatment in
ventricular ejection fraction. Am Heart J. 1989;118:1259–1265. patients with ischemic cardiomyopathy: a comparative study between
17. Carluccio E, Biagioli P, Alunni G, et al. Improvement of myocardial SPECT and MRI. Int J Cardiovasc Imaging. 2011;27:343–353.
performance (Tei) index closely reflects intrinsic improvement of 33. Mannaerts HF, van der Heide JA, Kamp O, Stoel MG, Twisk J, Visser VA.
cardiac function: assessment in revascularized hibernating myocardium. Early identification of left ventricular remodelling after myocardial
Echocardiography. 2012;29:298–306. infarction, assessed by transthoracic 3D echocardiography. Eur Heart J.
18. Hu K, Liu D, Niemann M, et al. Methods for assessment of left 2004;25:680–687.
ventricular systolic function in technically difficult patients with poor 34. Dey D, Slomka PJ, Berman DS. Achieving very-low-dose radiation
imaging quality. J Am Soc Echocardiogr. 2013;26:105–113. exposure in cardiac computed tomography, single-photon emission
207.e2 SECTION II  Ischemic Heart Disease

computed tomography, and positron emission tomography. Circ 51. Clarke CJ, Gurka MJ, Norton PT, Kramer CM, Hoyer AW. Assessment of
Cardiovasc Imaging. 2014;7:723–734. the accuracy and reproducibility of RV volume measurements by CMR
35. Gimelli A, Bottai M, Genovesi D, Giorgetti A, Di Martino F, Marzullo P. in congenital heart disease. JACC Cardiovasc Imaging. 2012;5:28–37.
High diagnostic accuracy of low-dose gated-SPECT with solid-state 52. Alfakih K, Plein S, Bloomer T, Jones T, Ridgway J, Sivananthan M.
ultrafast detectors: preliminary clinical results. Eur J Nucl Med Mol Comparison of right ventricular volume measurements between axial
Imaging. 2012;39:83–90. and short axis orientation using steady-state free precession magnetic
36. Khorsand A, Gyöngyösi M, Sochor H, et al. Assessment of left resonance imaging. J Magn Reson Imaging. 2003;18:25–32.
ventricular volumes, ejection fraction and mass. Comparison of 53. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ.
model-based analysis of ECG-gated (99m)Tc-SPECT and 18F-FDG-PET. Interstudy reproducibility of right ventricular volumes, function and
Nuklearmedizin. 2011;50:9–14. mass with cardiovascular magnetic resonance. Am Heart J. 2004;147:
37. Einstein AJ, Moser KW, Thompson RC, Cerqueira MD, Henzlova MJ. 218–223.
Radiation dose to patients from cardiac diagnostic imaging. Circulation. 54. Childs H, Ma L, Ma M, et al. Comparison of long and short axis
2007;116:1290–1305. quantification of left ventricular volume parameters by cardiovascular
38. ACCF/SCCT/ACR/AHA/ASE/ASNC/NASCI/SCAI/SCMR 2010 magnetic resonance, with ex-vivo validation. J Cardiovasc Magn Reson.
appropriate use criteria for cardiac computed tomography: a report of 2011;13:40.
the American College of Cardiology Foundation Appropriate Use 55. Bellenger NG, Gatehouse PD, Rajappan K, Keegan J, Firmin DN, Pennell
Criteria Task Force, the Society of Cardiovascular Computed DJ. Left ventricular quantification in heart failure by CMR using
Tomography, the American College of Radiology, the American Heart prospective respiratory navigator gating: comparison with breath-hold
Association, the American Society of Echocardiography, the American acquisition. J Magn Reson Imaging. 2000;11:411–417.
Society of Nuclear Cardiology, the North American Society for 56. Di Bella G, Siciliano V, Aquaro GD, et al. Right ventricular dysfunction:
Cardiovascular Imaging, the Society for Cardiovascular Angiography an independent and incremental predictor of cardiac deaths late after
and Interventions, and the Society for Cardiovascular Magnetic acute myocardial infarction. Int J Cardiovasc Imaging. 2015;31:379–387.
Resonance. J Am Coll Cardiol. 2010;56:1864–1894. 57. Gulati A, Ismail TF, Jabbour A, et al. The prevalence and prognostic
39. Rizvi A, Deaño RC, Bachman DP, Xiong G, Min JK, Truong QA. significance of right ventricular systolic dysfunction in nonischemic
Analysis of ventricular function by CT. J Cardiovasc Comput Tomogr. dilated cardiomyopathy. Circulation. 2013;128:1623–1633.
2015;9(1):1–12. 58. Tobler D, Motwani M, Wald RM, et al. Evaluation of a comprehensive
40. Yamamuro M, Tadamuro E, Kubo S, et al. Cardiac functional analysis cardiovascular magnetic resonance protocol in young adults late after
with multidetector row CT and segmental reconstruction algorithm: the arterial switch operation for d-transposition of the great arteries. J
comparison with echocardiography, SPECT and MR imaging. Radiology. Cardiovasc Magn Reson. 2014;16:98.
2005;234:381–390. 59. Kato S, Sekine A, Kusakawa Y, et al. Prognostic value of cardiovascular
41. Greupner J, Zimmermann E, Grohmann A, et al. Head-to-head magnetic resonance derived right ventricular function in patients with
comparison of left ventricular function assessment with 64-row interstitial lung disease. J Cardiovasc Magn Reson. 2015;17:10.
computed tomography, biplane left cineventriculography, and both 60. van de Veerdonk MC, Kind T, Marcus JT, et al. Progressive right
2- and 3-dimensional transthoracic echocardiography: comparison with ventricular dysfunction in patients with pulmonary arterial
magnetic resonance imaging as the reference standard. J Am Coll hypertension responding to therapy. J Am Coll Cardiol. 2011;58:
Cardiol. 2012;59:1897–1907. 2511–2519.
42. Shuijf JD, Bax JJ, Salm LP, et al. Noninvasive coronary imaging and 61. Fang W, Pereles FS, Bundy J, et al. Evaluating left ventricular function
assessment of left ventricular function using 16-slice computed using real-time TrueFISP: a comparison with conventional MR
tomography. Am J Cardiol. 2005;95:571–574. techniques abstract. J Cardiovasc Magn Reson. 1999;1:310.
43. Takx RA, Vliegenthart R, Schoepf UJ, et al. Computed tomography- 62. Moon JCC, Lorenz CH, Francis JM, Smith GC, Pennell DJ. Breathhold
derived parameters of myocardial morphology and function in black and FLASH and FISP cardiovascular MR imaging: left ventricular volume
white patients with acute chest pain. Am J Cardiol. 2016;117:333–339. differences and reproducibility. Radiology. 2002;223:789–797.
44. Schuhbaeck A, Achenbach S, Layritz C, et al. Image quality of ultra-low 63. Jaroni J, Meier R, Beer A, et al. Three-dimensional magnetic resonance
radiation exposure coronary CT angiography with an effective dose imaging using single breath-hold k-t BLAST for assessment of global left
<0.1 mSv using high-pitch spiral acquisition and raw data-based ventricular functional parameters. Acad Radiol. 2013;20:987–994.
iterative reconstruction. Eur Radiol. 2013;23:597–606. 64. Madore B, Hoge WS, Kwong R. Extension of the UNFOLD method to
45. Mao SS, Li D, Rosenthal DG, et al. Dual-standard reference values of left include free breathing. Magn Reson Med. 2006;55:352–362.
ventricular volumetric parameters by multidetector CT angiography. J 65. Kowalik GT, Knight DS, Steeden JA, et al. Assessment of cardiac time
Cardiovasc Comput Tomogr. 2013;7:234–240. intervals using high temporal resolution real-time spiral phase contrast
46. Stojanovska J, Prasitdumrong H, Patel S, et al. Reference absolute and with UNFOLDed-SENSE. Magn Reson Med. 2015;73:749–756.
indexed values for left and right ventricular volume, function and mass 66. Wintersperger BJ, Bauner K, Reeder SB, et al. Cardiac steady-state free
from cardiac computed tomography. J Med Imaging Radiat Oncol. precession CINE magnetic resonance imaging at 3.0 tesla: impact of
2014;58:547–558. parallel imaging acceleration on volumetric accuracy and signal
47. Mahnken AH, Heuzler D, Klotz E, et al. Determination of cardiac parameters. Invest Radiol. 2006;41:141–147.
output with multislice spiral computed tomography: a validation study. 67. Ferreira PF, Gatehouse PD, Mohiaddin RH, Firmin DN. Cardiovascular
Invest Radiol. 2004;39:451–454. magnetic resonance artefacts. J Cardiovasc Magn Reson. 2013;15:41.
48. Sievers B, Brandts B, Franken U, Trappe HJ. Single and biplane 68. Maroules CD, McColl R, Khera A, Peshock RM. Interstudy
True-FISP cardiovascular magnetic resonance for rapid evaluation of left reproducibility of SSFP cine magnetic resonance: impact of magnetic
ventricular volumes and ejection fraction. J Cardiovasc Magn Reson. field strength and parallel imaging. J Magn Reson Imaging.
2004;6:593–600. 2008;27:1139–1145.
49. Schulz-Menger J, Bluemke DA, Bremerich J, et al. Standardized image 69. Maceira AM, Ripoll C, Cosin-Sales J, et al. Long term effects of cocaine
interpretation and post processing in cardiovascular magnetic on the heart assessed by cardiovascular magnetic resonance at 3T. J
resonance: Society for Cardiovascular Magnetic Resonance (SCMR) Cardiovasc Magn Reson. 2014;16:26.
board of trustees task force on standardized post processing. J 70. Le TT, Tan RS, De Deyn M, et al. Cardiovascular magnetic resonance
Cardiovasc Magn Reson. 2013;15:35. reference ranges for theheart and aorta in Chinese at 3T. J Cardiovasc
50. Longmore DB, Klipstein RH, Underwood SR, et al. Dimensional Magn Reson. 2016;18:21.
accuracy of magnetic resonance in studies of the heart. Lancet. 71. Liu CY, Bluemke DA, Gerstenblith G, et al. Reference values of
1985;1:1360–1362. myocardial structure, function, and tissue composition by cardiac
CHAPTER 16  Assessment of Cardiac Function 207.e3

magnetic resonance in healthy African-Americans at 3T and their 88. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular
relations to serologic and cardiovascular risk factors. Am J Cardiol. systolic and diastolic function normalized to age, gender and body
2014;114:789–795. surface area from steady-state free precession cardiovascular magnetic
72. Katz J, Millikem MC, Stray-Gunderson J, et al. Estimation of human resonance. Eur Heart J. 2006;27:2879–2888.
myocardial mass with MR imaging. Radiology. 1988;169:495–498. 89. Mohiaddin RH, Longmore DB. Functional aspects of cardiovascular
73. Swift AJ, Rajaram S, Condliffe R, et al. Diagnostic accuracy of magnetic resonance imaging: techniques and application. Circulation.
cardiovascular magnetic resonance imaging of right ventricular 1993;88:264–281.
morphology and function in the assessment of suspected pulmonary 90. Bennett CJ, Maleszewski JJ, Araoz PA. CT and MR imaging of the aortic
hypertension results from the ASPIRE registry. J Cardiovasc Magn Reson. valve: radiologic-pathologic correlation. Radiographics.
2012;14:40. 2012;32:1399–1420.
74. Lorenz CH, Walker ES, Morgan VL, Graham TP, Klein SS. Normal 91. Cawley PJ, Maki JH, Otto CM. Valvular heart disease: changing concepts
human right and left ventricular mass, systolic function and gender in disease management. cardiovascular magnetic resonance imaging for
differences by cine magnetic resonance imaging. J Cardiovasc Magn valvular heart disease technique and validation. Circulation.
Reson. 1999;1:7–21. 2009;119:468–478.
75. Bellenger NG, Davies LC, Francis JM, Marcus NJ, Pennell DJ. Reduction 92. Claus P, Omar AM, Pedrizzetti G, Sengupta PP, Nagel E. Tissue tracking
in sample size for studies of remodelling in heart failure by the use of technology for assessing cardiac mechanics: principles, normal values,
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. and clinical applications. JACC Cardiovasc Imaging. 2015;8:1444–
2000;2:271–278. 1460.
76. Walsh R, Salem Y, Shah A, Lai WW, Nielsen JC. Repeatability of 93. Riffel JH, Andre F, Maertens M, et al. Fast assessment of long axis strain
cardiac-MRI-measured right ventricular size and function in congenital with standard cardiovascularmagnetic resonance: a validation study of a
heart disease. Pediatr Radiol. 2011;41:1000–1007. novel parameter with reference values. J Cardiovasc Magn Reson.
77. Grothues F, Smith GC, Moon JCC, et al. Comparison of interstudy 2015;17:69.
reproducibility of cardiovascular magnetic resonance with two- 94. Kalam K, Otahal P, Marwick TH. Prognostic implications of global LV
dimensional echocardiography in normal subjects and in patients with dysfunction: a systematic review and meta-analysis of global
heart failure or left ventricular hypertrophy. Am J Cardiol. longitudinal strain and ejection fraction. Heart. 2014;100:1673–1680.
2002;90:29–34. 95. Taylor RJ, Moody WE, Umar F, et al. Myocardial strain measurement
78. Bottini PB, Carr AA, Prisant M, Flickinger FW, Allison JD, Gottdiener with feature-tracking cardiovascular magnetic resonance: normal values.
JS. Magnetic resonance imaging compared to echocardiography to assess Eur Heart J Cardiovasc Imaging. 2015;16:871–881.
left ventricular mass in the hypertensive patient. Am J Hypertens. 96. Schneeweis C, Lapinskas T, Schnackenburg B, et al. Comparison of
1995;8:221–228. myocardial tagging and feature tracking in patients with severe aortic
79. Grothues F, Moon JC, Bellenger MG, Smith GC, Klein HU, Pennell DJ. stenosis. J Heart Valve Dis. 2014;23:432–440.
Interstudy reproducibility of right ventricular volumes, function, and 97. Obokata M, Nagata Y, Wu VC, et al. Direct comparison of cardiac
mass with cardiovascular magnetic resonance. Am Heart J. 2004;147: magnetic resonance feature tracking and 2D/3D echocardiography
218–223. speckle tracking for evaluation of global left ventricular strain. Eur Heart
80. Saloux E, Labombarda F, Pellissier A, et al. Diagnostic value of J Cardiovasc Imaging. 2016;17:525–532.
three-dimensional contrast-enhanced echocardiography for left 98. Bollache E, Redheuil A, Clément-Guinaudeau S, et al. Automated left
ventricular volume and ejection fraction measurement in patients with ventricular diastolic function evaluation from phase-contrast
poor acoustic windows: a comparison of echocardiography and cardiovascular magnetic resonance and comparison with Doppler
magnetic resonance imaging. J Am Soc Echocardiogr. 2014;27:1029– echocardiography. J Cardiovasc Magn Reson. 2010;12:63.
1040. 99. Mohiaddin R, Hasegawa M. Flow pattern in the dilated ischaemic left
81. Hoffmann R, Barletta G, von Bardeleben S, et al. Analysis of left ventricle studied by MRI in healthy volunteers and in patients with
ventricular volumes and function: a multicenter comparison of cardiac myocardial infarction. J Magn Reson Imaging. 1995;5:493–498.
magnetic resonance imaging, cine ventriculography, and unenhanced 100. Maceira AM, Cosín-Sales J, Roughton M, Prasad SK, Pennell DJ.
and contrast-enhanced two-dimensional and three-dimensional Reference left atrial dimensions and volumes by steady state free
echocardiography. J Am Soc Echocardiogr. 2014;27:292–301. precession cardiovascular magnetic resonance. J Cardiovasc Magn Reson.
82. Kim J, Cohen SB, Atalay MK, Maslow AD, Poppas A. Quantitative 2010;12:65.
assessment of right ventricular volumes and ejection fraction in patients 101. Karwatowski SP, Brecker SJ, Yang GZ, Firmin DN, St John Sutton M,
with left ventricular systolic dysfunction by real time three-dimensional Underwood SR. A comparison of left ventricular myocardial velocity
echocardiography versus cardiac magnetic resonance imaging. in diastole measured by magnetic resonance and left ventricular filling
Echocardiography. 2015;32:805–812. measured by Doppler echocardiography. Eur Heart J. 1996;17:
83. Bellenger NG, Francis JM, Davies CL, Coats A, Pennell DJ. 795–802.
Establishment and performance of a magnetic resonance cardiac 102. Baer FM, Voth E, Schneider CA, Theissen P, Schicha H, Sechtem U.
function clinic. J Cardiovasc Magn Reson. 2000;2:15–22. Comparison of low-dose dobutamine-gradient-echo magnetic resonance
84. Taylor AM, Jhooti P, Wiesmann F, Keegan J, Firmin DN, Pennell DJ. MR imaging and positron emission tomography with [18F]
navigator-echo monitoring of temporal changes in diaphragm position: fluorodeoxyglucose in patients with chronic coronary artery disease: a
implications for MR coronary angiography. J Magn Reson Imaging. functional and morphological approach to the detection of residual
1997;7:629–636. myocardial viability. Circulation. 1995;91:1006–1015.
85. Simprini LA, Goyal P, Codella N, et al. Geometry-independent inclusion 103. Dendale PA, Franken PR, Waldman GJ, et al. Low-dosage dobutamine
of basal myocardium yields improved cardiac magnetic resonance magnetic resonance imaging as an alternative to echocardiography in
agreement with echocardiography and necropsy quantified left the detection of viable myocardium after acute infarction. Am Heart J.
ventricular mass. J Hypertens. 2013;31:2069–2076. 1995;130:134–140.
86. Maceira AM, Prasad SK, Khan M, Pennell DP. Normalized left 104. Pennell DJ, Underwood SR, Manzara CC, et al. Magnetic resonance
ventricular systolic and diastolic function by steady state free precession imaging during dobutamine stress in coronary artery disease. Am J
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. Cardiol. 1992;70:34–40.
2006;8:417–426. 105. Nagel E, Lehmkuhl HB, Bocksch W, et al. Noninvasive diagnosis of
87. Kawel-Boehm N, Maceira A, Valsangiacomo-Buechel ER, et al. Normal ischemia induced wall motion abnormalities with the use of high dose
values for cardiovascular magnetic resonance in adults and children. J dobutamine stress MRI: comparison with dobutamine stress
Cardiovasc Magn Reson. 2015;17:29. echocardiography. Circulation. 1999;99:763–770.
207.e4 SECTION II  Ischemic Heart Disease

106. Yang PC, Kerr AB, Liu AC, et al. New real time interactive magnetic 109. Kramer C, Rogers WJ, Theobald TM, Power TP, Petruolo S, Reichek N.
resonance imaging system complements echocardiography. J Am Coll Remote noninfarcted region dysfunction soon after first anterior
Cardiol. 1998;32:2049–2056. myocardial infarction: a magnetic resonance tagging study. Circulation.
107. Hundley WG, Hamilton CA, Thaomas MS, et al. Utility of fast cine 1996;94:660–666.
magnetic resonance imaging and display for the detection of myocardial 110. Potter DD, Araoz PA, McGee KP, Harmsen WS, Mandrekar JN, Sundt
ischemia in patients not well suited for second harmonic stress TM 3rd. Low-dose dobutamine cardiac magnetic resonance imaging
echocardiography. Circulation. 1999;100:1697–1702. with myocardial strain analysis predicts myocardial recoverability after
108. van Rugge FP, van der Wall EE, Spanjersberg SJ, et al. Magnetic coronary artery bypass grafting. J Thorac Cardiovasc Surg.
resonance imaging during dobutamine stress for detection and 2008;135:1342–1347.
localization of coronary artery disease: quantitative wall motion analysis
using a modification of the centerline method. Circulation.
1994;90:127–138.
17 
Stress Cardiovascular Magnetic Resonance:
Wall Motion
R. Brandon Stacey and W. Gregory Hundley

Master and Oppenheimer described the first stress test in 1929.1 second- and third-degree atrioventricular block, sudden death syndrome,
Several methods of assessing myocardial ischemia have subsequently supraventricular tachycardia, and previous episodes of dobutamine
been developed, including those that incorporate electrocardiograms hypersensitivity.
(ECGs), echocardiography, myocardial scintigraphy, and, most recently, Although dobutamine exhibits positive inotropic activity, its chro-
cardiovascular magnetic resonance (CMR). Since 1987,2 stress CMR has notropic response can be suboptimal. Studies using changes in LV wall
been used to identify inducible ischemia,3 measure contractile reserve,4 motion to identify inducible ischemia exhibit heightened sensitivity in
and identify those at risk of future myocardial infarction (MI) and appreciating flow-limiting epicardial luminal narrowing when the heart
cardiac death.5,6 In this chapter, we review the utility of CMR wall rate response during testing exceeds 85% of the age-predicted maximum
motion stress testing, including the pharmacologic agents used during heart rate (APMHR) response (220 − age in years).17 For this reason,
the procedure, the evolution of the procedure from dobutamine stress intravenous atropine may be needed to achieve an adequate peak heart
echocardiography (DSE), the performance and safety of a dobuta- rate response during testing. Atropine is a naturally occurring alkaloid
mine stress CMR (DCMR) examination, and the efficacy of DCMR that is a competitive antagonist of muscarinic cholinergic receptors; it
in identifying inducible ischemia, myocardial viability, and cardiac increases heart rate by inhibiting vagal tone.18 It is particularly useful
prognosis. in study participants taking beta-blockers, rate-limiting calcium antago-
nists, or those possessing a high parasympathetic drive. Atropine may
be administered in increments of 0.1 to 0.3 mg, up to a total of 2 mg,
INTRAVENOUS DOBUTAMINE AND ATROPINE to facilitate the achievement of >85% the APMHR during testing.
Developed in 1975 by Tuttle and Mills,7 dobutamine is a synthetic Common side effects of atropine include dry mouth, tachycardia, and
beta-1-selective catecholamine agonist that binds to myocyte beta- hallucinations.10 Abnormally high heart rates after atropine administra-
1-receptors and increases intracellular calcium concentrations and tion are best treated with intravenous beta-blockers or rate-limiting
myocardial contraction.8 Dobutamine was first employed as a stress- calcium channel antagonists.
testing agent in 1984.9 In addition to enhancing myocardial contractility,
it increases the heart rate by promoting peripheral vasodilation and a SAFETY PROFILE OF DOBUTAMINE AND ATROPINE
reflex tachycardia.8
Because of rapid metabolism by catechol-O-methyltransferase,
STRESS TESTING
dobutamine’s onset of action and half-life are both approximately 2 Safety is a primary concern for physicians and health care providers
minutes; inactive metabolites are readily excreted by the kidneys and who administer dobutamine/atropine stress examinations.16,19,20 The
liver.10 At low doses, <10 µg/kg/min, myocardial contractility is aug- safety profile of dobutamine/atropine stress has been reported in 6
mented and minor peripheral vasodilation occurs.11 As the dose increases large studies, 3 using DSE and 3 using DCMR (Table 17.1).21,22 Garcia
(20–40 µg/kg/min), heart rate and myocardial oxygen demand are et al.,23 Picano et al.,24 and Geleijnse et al.25 reported on the adverse
increased along with myocardial work.12 In study participants with events associated with DSE. These were classified into major and minor
flow-limiting epicardial stenoses, intravenous dobutamine promotes complications. Major complications included death, ventricular fibrilla-
an oxygen supply/demand mismatch that induces left ventricular (LV) tion, sustained ventricular tachycardia, complete atrioventricular block,
wall motion abnormalities.13,14 acute MI, rupture of the LV free wall or ventricular septal defect, tran-
Intravenous dobutamine increases myocardial oxygen demand in a sient ischemic attack, and severe hypotension. Minor complications
fashion similar to exercise, and thus is useful for stress testing in study included nausea, anxiety, and atropine poisoning with hallucinations
participants whose exercise ability is limited as a result of peripheral lasting up to several hours in the absence of either myocardial ischemia or
vascular disease, physical incapacitation, or chronic deconditioning.15 hypotension.
Other benefits of dobutamine include its tolerance for peripheral Across the 3 DSE studies, the rates of major and minor complica-
vein infusion and its effectiveness in both perfusion and wall motion tions ranged from 0.1% to 4%, and 38% to 71%, respectively. Wahl
imaging.16 et al.,26 Kuijpers et al.,27 and Hamilton et al.28 have reported on the
The common side effects associated with dobutamine administration incidence of major and minor side effects associated with DCMR. In
for stress include chest pain, ventricular ectopy, dyspnea, nausea, hyper- Wahl’s study of 1000 participants, 54% received atropine; in Kuijpers’
tension, and arrhythmias. Despite its relative safety, dobutamine is study of 400 participants, none received atropine; and in Hamilton’s
specifically contraindicated in study participants with hypertrophic study of 469 participants, 27% received atropine. There were no episodes
obstructive cardiomyopathy, epicardial luminal narrowings of >50% of ventricular fibrillation, MI, or death during DCMR (see Table 17.1).
in the left main coronary arterial segment, severe aortic stenosis, The number of study participants sustaining other major events was

208
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 209

TABLE 17.1  Safety Profile of Select DSE and DCMR Studies


Study Modality No. of Patients Minor Events Major Events Deaths
24
Picano DSE 2799 78% 5% 0
Geleijnse25 DSE 2246 71% 5% 0
Garcia23 DSE 325 57% 21% 1
Hamilton28 DCMR 469 67% 0% 0
Kuijpers27 DCMR 400 71% 3% 0
Wahl26 DCMR 1000 64% 6% 0

Major complications included death, ventricular fibrillation, supraventricular tachycardia, atrioventricular block, acute myocardial infarction, rupture
of the left ventricular free wall or ventricular septal defects, transient ischemic attacks, or severe symptomatic hypotension. Minor complications
included nausea/vomiting, anxiety, hypotension, and atropine poisoning with hallucinations lasting several hours in the absence of myocardial
ischemia.
DCMR, Dobutamine stress cardiovascular magnetic resonance; DSE, dobutamine stress echocardiography.
From Mandapaka S, Hundley WG. Dobutamine cardiovascular resonance: a review. J Magn Reson Imag. 2006;24(3):499–512.

0.6%, 0.2%, and 0%, respectively. The percentage of participants sus- hypotension, dysrhythmias, unstable angina, or unexpected neurologic
taining minor events in all three studies combined was 54%. In these findings.21
studies, the rates of major and minor events observed during DCMR DSE is useful for assessing risk of future MI, death, and the need
are similar to those reported with DSE and would be expected as the for coronary intervention.39,40 For many years, it was the modality of
dose administration is similar. choice for risk stratification of cardiovascular events for patients being
It is important to recognize that the major events are usually associ- considered for noncardiac surgery.41,42 However, DSE is of minimal
ated with continued administration of pharmacologic stress in the setting value in study participants with poor acoustic windows, often because
of concurrent myocardial ischemia. For this reason, it is important to of a large body habitus, prior cardiothoracic surgery, or pulmonary
identify ischemia promptly, and to discontinue the DCMR protocol disease.17,43,44 In addition, there is a high interobserver variability for
when ischemia is recognized.29 In the studies reported above, many of image interpretation; consequently, the accuracy of the test depends
the study participants (56%) who developed side effects had known heavily on the proficiency of both the sonographer (acquisition) and
coronary atherosclerosis and a diminished resting LV ejection fraction physician (interpretation).29,45
(LVEF).30 For this reason, a high level of scrutiny is used when review-
ing image or ST-segment data for ischemia in the setting of reduced DOBUTAMINE STRESS CARDIOVASCULAR
LVEF. Because of the risk of major adverse events, certain study par-
ticipants should not receive intravenous dobutamine and atropine.
MAGNETIC RESONANCE
Medications used for testing and emergency medications (Table 17.2) CMR is well suited to overcome the acquisition and interpretative limi-
for treatment of potential cardiac complications need to be readily tations encountered during DSE. It has no acoustic window limitations
available to improve the probability of a successful outcome should and thus can obtain comprehensive visualization of the LV myocardium
complications occur during DSE. regardless of body habitus.46–48 In addition, image acquisition can be
standardized and there is less reliance on an individual technologist’s
technique for acquiring high-quality images.
DOBUTAMINE STRESS ECHOCARDIOGRAPHY DCMR is commonly performed on short, 1.5 tesla (1.5 T) closed-bore
Since 1977, transthoracic echocardiography (TTE) has been used to systems, with bore diameters ranging from 55 to 70 cm. At the initia-
acquire images before, during, and after treadmill or bicycle exercise tion of the DCMR examination, a 12-lead ECG is performed outside of
stress, or pharmacologically induced stress.31 During DSE, images are the magnet. After intravenous access is established, study participants
acquired in four standard imaging planes: parasternal short-axis (PSAX), are told to lay flat on the CMR scanning table with a phased-array
parasternal long-axis (PLAX), apical four-chamber (A4C), and apical surface coil, brachial blood pressure cuff, pulse oximetry monitor, and
two-chamber (A2C) views.32,33 The most common dobutamine infusion respiratory gating belt attached (Fig. 17.1).49 A physician and nurse
protocol used involves initiating dobutamine at 5 to 7.5 µg/kg/min for are present throughout testing to continuously monitor the heart rate
a period of 3 minutes, followed by 10 µg/kg/min for 3 minutes and and rhythm, respiratory rate, oxygen saturation, and blood pressure.
subsequent increments of 10 µg/kg/min in 3-minute increments to a The protocol is similar to that described for DSE in that dobutamine
maximal dose of 50 µg/kg/min. If the patient does not achieve 85% of infusions are started at 5 to 7.5 µg/kg/min, followed by 10, 20, 30, 40,
their APMHR, supplemental atropine is administered in 0.1 mg to and 50 µg/kg/min infusions in 3-minute stages. If study participants
0.3 mg increments per minute (up to 2.0 mg total) until the target do not achieve 85% of their APMHR, atropine is added in 0.1 mg to
heart rate is reached.34 0.2 mg/min increments (up to 2 mg total) to augment the heart rate
Throughout the test, the four echocardiographic planes are reviewed response (Fig. 17.2).
to monitor for ischemia. During testing, LV myocardial wall motion Breath-hold cine balanced steady-state free precession (bSSFP) images
is defined as 1 = normal, 2 = hypokinesis, 3 = akinesis, and 4 = dys- are obtained in three apical views (horizontal, vertical, and apical long
kinesis. Myocardial ischemia is defined as an increase in wall motion axis) and in at least three short-axis LV planes (base, middle, and apex)
score during testing, or the persistence of severe hypokinesis during at each level of pharmacologic stress (Fig. 17.3). If Simpson’s rule is
testing.35,36 The scoring system is determined using a 17-segment model used to calculate LV volumes, short slices spanning the entire LV from
defined by the American Heart Association and American College base to apex can be acquired.50 These views are also obtained after 10
of Cardiology.37,38 A DSE is interrupted when there is evidence of minutes of recovery to confirm that LV wall motion has returned to
ischemia in at least two adjacent segments, severe hypertension or baseline. After the patient is removed from the magnet, the 12-lead
210 SECTION II  Ischemic Heart Disease

TABLE 17.2  Common On-Site Medications for Cardiovascular Magnetic Resonance Stress
Tests
Stress Agent
Dobutamine Stimulates beta-1-receptors to increase myocardial contractility and stroke volume; also acts as a peripheral vasodilator
Atropine Anticholinergic that inhibits acetylcholine at the parasympathetic neuroeffector junction, blocking vagal effects on the sinoatrial and
atrioventricular nodes; enhances conduction through the atrioventricular node and increases the heart rate
Adenosine Peripheral and central vasodilator; increases heart rate and blood flow

Treatment for Ventricular Dysrhythmia


Lidocaine Decreases depolarization, automaticity, and excitability in the ventricles; can suppress ventricular dysrhythmia
Procainamide Decreases excitability, conduction velocity, automaticity, and membrane responsiveness with prolonged refractory period

Treatment for Supraventricular Tachycardia


Metoprolol Beta-1-antagonist that decreases myocardial contractility, heart rate, cardiac output, and blood pressure and reduces myocardial
oxygen demand
Diltiazem Calcium channel antagonist; reduces myocardial contractility and sinoatrial node conductor
Verapamil Calcium channel antagonist; reduces myocardial contractility and oxygen demand; decreases atrioventricular nodal conduction rate;
and dilates coronary arteries and arterioles

Treatment for Chest Pain


Nitroglycerin Reduces cardiac oxygen demand by decreasing left ventricular end-diastolic pressure (preload) and systemic vascular resistance
(afterload)
Aspirin Analgesic/antipyretic that impedes clotting by blocking prostaglandin synthesis
Heparin Anticoagulant that accelerates the formation of antithrombin III–thrombin complex and deactivates thrombin, preventing the
conversion of fibrinogen to fibrin
Morphine sulfate Opium derivative with primary effects on the central nervous system and organs containing smooth muscle

Treatment of Allergic Reaction or Anaphylaxis


Diphenhydramine Prevents histamine-mediated responses
Hydrocortisone Corticosteroid that decreases inflammation, mainly by stabilizing leukocyte lysosomal membranes and suppressing the immune
responses

Anxiolytic
Diazepam Benzodiazepine with relatively long half-life that works through the immune system to exert a calming effect
Midazolam Water-soluble benzodiazepine with a short half-life exerts calming effect at low doses and a central nervous system depressant
effect at high doses

Reversal Agents
Flumazenil Benzodiazepine receptor antagonist that can be used to reverse the effects of midazolam or diazepam
Naloxone Narcotic antagonist that can be used to reverse the effects of morphine sulfate

Other
Furosemide Loop diuretic for treatment of pulmonary congestion due to intravascular volume overload
Albuterol (nebulizer) Inhaled beta-agonist that causes bronchodilation; important for emergent treatment of bronchospasm
Aminophylline Relaxes smooth muscle of the bronchial airways and pulmonary blood vessels for treatment of bronchospasms

ECG is repeated. The DCMR test is stopped if there is development of among the first to report the clinical use of DCMR (Fig. 17.6). In this
ischemia, as evidenced by a new wall motion abnormality (Figs. 17.4 study, 25 study participants underwent DCMR and dobutamine
and 17.5): a fall in systolic blood pressure >40 mm Hg, significant thallium-201 single-photon emission computed tomography (SPECT),34
ventricular arrhythmias, or achievement of the target heart rate of 85% and x-ray coronary angiography. Study subjects received dobutamine
APMHR.51 Although there are multiple image strategies available for infusions of up to 20 µg/kg/min and underwent conventional gradient
the acquisition of bright-blood cine images, in general, most favor the recalled echo (GRE) cine CMR. Twenty of the 22 study participants
use of bSSFP at 1.5 T, with multiple slices acquired during each breath- with significant coronary artery disease (CAD) after angiography had
hold using parallel imaging techniques (Table 17.3).46,52,53 wall motion abnormalities consistent with CMR, and 21 had reversible
ischemia as identified by dobutamine thallium tomography. Thus the
UTILITY OF DOBUTAMINE STRESS sensitivity of DCMR for detection of significant CAD was 91%. Between
CARDIOVASCULAR MAGNETIC RESONANCE FOR DCMR and dobutamine thallium tomography there was a 96% agree-
ment at rest, 90% agreement during stress, and 91% agreement for the
IDENTIFYING INDUCIBLE ISCHEMIA assessment of reversible ischemia. This study was the first to demonstrate
Investigators in the early 1990s began using DCMR to detect LV wall that DCMR could be effectively used to assess regional LV systolic func-
motion abnormalities indicative of ischemia.11,54 Pennell et al.55 were tion in participants with CAD.
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 211

Scanner

Preparation and

recovery area

FIG. 17.1  Images highlighting the preparation/recovery area (left) as well as the cardiovascular magnetic
resonance (CMR) scanning facility (right). Note that by using a preparation and recovery area, CMR imaging
center patient throughput is enhanced, as the scanning facility is not used for time-consuming, nonscan-
related tasks such as insertion of intravenous catheters or placement of special monitoring equipment.

• Review H & P
• Baseline ECG Emphasize target HR 85% APMHR for age
• HR, BP, Ox.
Three apical views and three short-axis views

HR HR HR HR
BP BP BP BP Total scan
time ~25 min

Patient prep B 10 20 + At R

Dobutamine (µg/kg/min)

30 min 3 min 3 min 3 min 3 min


FIG. 17.2  Dobutamine infusion protocol for dobutamine stress cardiovascular magnetic resonance testing.
As shown, after review of the history (H), physical examination (P), and electrocardiogram (ECG), three apical
and three short-axis views are acquired at baseline (B) over a 3-minute time period. Also, heart rate (HR) and
blood pressure (BP) are collected. Next, dobutamine is infused at 10 µg/kg/min, and the three apical and
three short-axis views are repeated. At peak stress, a combination of dobutamine and atropine (At) are
infused to achieve 85% of the age-predicted maximal heart rate (APMHR). At this peak stress level, three
apical views and three short-axis views are repeated. Upon completion of the peak stress image acquisitions,
recovery images (R) are obtained before patient discharge. The total scan time for an individual with a nega-
tive stress test averages 25 minutes using this protocol. Ox, Oxygen saturation.

Subsequently, multiple studies have examined the utility of DCMR assessment with DCMR (5, 10, 15, and 20 µg/kg/min at 1.5 T). Seg-
for a variety of clinical situations. Baer et al.56 reported on the utility mental wall motion analysis was performed in basal, mid, and apical
of DCMR for individuals with no previous infarction or wall motion short-axis planes, as well as two transverse planes. Each of 72 segments
abnormalities by left ventriculography, but with high-grade coronary was related to a coronary artery territory and was graded as normal,
artery stenosis (≥70%). Twenty-eight subjects underwent wall motion hypokinetic, or akinetic. Eighty-seven percent of epicardial coronary
212 SECTION II  Ischemic Heart Disease

Coronal locator Long axis Long axis

Anterior
chest
Antero
septum LV Posterior
wall

Ao LA

Four-chamber
Apical short axis
Septum

LV
Middle short axis RV Lateral
wall
RA
LA

LV Basal short axis Two-chamber


RV

60°

Anterior
Inferior LV wall
wall
LA

FIG. 17.3  Imaging acquisition strategy for obtaining three apical and three short-axis views during dobutamine
stress cardiovascular magnetic resonance. As shown at the top left, a long-axis plane (dashed line, top left
panel) is obtained from a coronal locating plane. This long-axis or apical three-chamber view is similar to the
parasternal long-axis plane acquired during the transthoracic echocardiography (top middle panel). Next, three
short-axis views (apical, middle, and basal) are obtained as plotted off the apical long-axis or three-chamber
view. As shown in the bottom panel (labeled “Basal short axis”), 60-degree rotations are used to obtain
image acquisitions in the four-chamber and two-chamber planes of the left ventricle. Ao, Aorta; LA, left
atrium; LV, left ventricle; RA, right atrium; RV, right ventricle.

artery narrowings were correctly identified in the left anterior descend- 80%, and 100% respectively. In the quantitative portion of the study,
ing (LAD), 62% in the left circumflex (LCX), and 78% in the right 39 study participants with previously demonstrated LV wall motion
coronary artery (RCA) territories. Single vessel and multivessel disease abnormalities and 10 normal volunteers were assessed using a short-
was identified with 73% and 100% sensitivity, respectively. This study axis stress cine and analyzed with a modified centerline method. With
illustrated the utility of DCMR in individuals with known CAD, but preselected cuts, stress wall motion was considered abnormal if four
with normal wall motion at rest. or more adjacent cords (the LV was divided into a total of 100 cords)
Comparison of DCMR and SPECT for the detection of CAD was showed systolic wall thickening below two standard deviations of that
examined in another study by Baer et al.57 In 35 consecutive study obtained from the normal volunteers. This method had high sensitivity
participants, sensitivity for detection of CAD was 84% and 87% for in detecting single- (88%), double- (91%), and triple-vessel (100%)
CMR and SPECT, respectively. This comparison between the two tech- disease, and in specifically detecting stenosis or specific abnormalities in
niques indicated that the efficacy of DCMR is maintained when compared the LAD (75%), RCA (87%), and LCX (63%). Unfortunately, there was
with a well-established clinical tool. no direct comparison between the qualitative and quantitative groups
Van Rugge et al. assessed the efficacy of quantitative58 and qualita- to determine if endocardial and epicardial tracing gave a significant
tive59 measures of LV wall motion during DCMR for identification diagnostic improvement. However, these studies demonstrated the utility
and localization of myocardial ischemia in a study of participants with of DCMR using both quantitative and qualitative methods.
CAD. For the qualitative study of 45 study participants, 37 had CAD Each of the early studies was relatively small, performed in a single
and 30 (81%) showed an LV wall motion abnormality during DCMR institution, and enrolled participants in whom the investigators were
with a specificity of 100%; these values were superior to those from aware of the extent of coronary artery luminal narrowings before stress
exercise electrocardiography (70 [63%]) or DSE (51 [63%]). Sensitiv- CMR. LV wall motion was assessed at baseline and peak stress, and not
ity for detection of single-, double-, or triple-vessel disease was 75%, visualized continuously throughout the course of stress testing. Infusions
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 213

Long axis Four-chamber Two-chamber

Ap-AS Ap-Post Ap-Sept Ap-Lat Ap-Inf Ap-Ant

Mid-AS Mid-Post Mid-Sept Mid-Lat Mid-Inf Mid-Ant

Bs-AS Bs-Post Bs-Sept Bs-Lat Bs-Inf Bs-Ant

Basal short axis Middle short axis Apical short axis

Bs-Ant Mid-Ant
Bs-Lat Mid-Lat Ap-Ant Ap-Lat
Bs-AS
Mid-AS Mid-Post Ap-AS
Bs-Post Ap-Post
Bs-Sept
Bs-Inf Mid-Sept Mid-Inf Ap-Sept Ap-Inf

FIG. 17.4  Delineation of orthogonal left ventricular (LV) myocardial segments. Long axis, four-chamber, and
two-chamber views are displayed in the top series of images. Apical, middle, and basal segments are identi-
fied on each of the corresponding LV myocardial walls. The orthogonal display of the same myocardial
segments in short-axis planes is displayed in the bottom row of images. Recognition of ischemia during
dobutamine stress cardiovascular magnetic resonance is achieved when an observed LV wall motion abnor-
mality occurs during dobutamine/atropine infusions in two orthogonal planes. Ant, Anterior; Ap, apical; AS,
anterior septum; Bs, basal; Inf, inferior; Lat, lateral; Post, posterior; Sept, septal.

End End
diastole systole

Middle
short axis

Normal
response

Apical
view

Middle
short axis

Inducible
ischemia

Apical
view

FIG. 17.5  Examples of normal left ventricular (LV) contractility as well as inducible ischemia during dobuta-
mine stress cardiovascular magnetic resonance. In the top series of images, a middle short-axis plane and
an apical four-chamber plane are demonstrated at end diastole in a sequential series of images proceeding
from diastole (far left images) to end systole (far right images). As shown in the normal situation, LV cavity
dimensions diminish symmetrically in all myocardial segments. In the bottom series of images with evidence
of inducible ischemia, one notices that in both the middle short-axis and the apical three-chamber view, the
middle and apical segments of the septum do not thicken in systole. As shown by the white arrowheads,
the failure to thicken in the same segments in two orthogonal planes is a region consistent with inducible
ischemia.
214 SECTION II  Ischemic Heart Disease

TABLE 17.3  Advantages and Disadvantages of Various Cine Bright-Blood Cardiovascular


Magnetic Resonance Techniques
Technique Advantage Disadvantage
GRE (breath-hold) Reliable ↑ heart rate Breath-hold = 12 s
GRE (respiration triggered) No breath-hold 45-s scan
SSFP Apical views Artifact ↑ heart rate
SENSE Rapid acquisition Unknown reliability
Real time Rapid acquisition Noise, low spatial, and temporal resolution

GRE, Gradient recalled echo; SENSE, sensitivity encoding; SSFP, steady-state free precession.

Rest 10 µg 20 µg Max Rest 10 µg 20 µg Max


End diastole
End systole

FIG. 17.6  Positive dobutamine stress cardiovascular magnetic resonance (DCMR). Top, DCMR cine images
(short-axis and two-chamber view) of a patient with chest pain and suspected coronary artery disease. At
rest and under increasing dobutamine, there is normal regional left ventricular function. However, at maximum
stress, an inferior and inferolateral wall motion abnormality is elicited (white arrowheads). Bottom, X-ray
coronary angiography images demonstrating single-vessel left circumflex coronary artery disease (black
arrows). (From Paetsch I, Jahnke C, Fleck E, Nagel E. Current clinical applications of stress wall motion
analysis with cardiac magnetic resonance imaging. Eur J Echocardiogr. 2005;6(5):317–326.)

were terminated prematurely when study participants developed chest study participants could not be examined by DSE because of poor
pain. Nevertheless, this series of small studies effectively demonstrated image quality and 18 could not be examined by DSMR (adipositas 6
the feasibility and diagnostic utility of DCMR. and claustrophobia 11). Only 4 study participants failed to reach their
Over the last 15 years, advancements in computer software and target heart rate. Results indicated that the diagnostic accuracy of DCMR
CMR hardware have made it possible to study subjects with suspected was higher when compared with DSE (P < .05) because the specificity
CAD more easily, without awareness of the extent of CAD before testing. increased from 69.8% to 85.7%, and sensitivity increased from 74.3%
In 1999, Nagel et al.60 published a landmark study using high-dose to 86.2% (Table 17.4). This was primarily related to patients with sub-
dobutamine (up to 40 µg/kg/min) and atropine during DCMR to detect optimal DSE image quality. The authors concluded that wall motion
CAD; the efficacy of the test was compared with DSE. In this study, abnormalities induced during DCMR could be detected with a signifi-
208 participants (61 women, 147 men) underwent DCMR and DSE cantly higher diagnostic accuracy compared with DSE in study partici-
before x-ray coronary angiography. Dobutamine was infused intrave- pants with suspected CAD.
nously at doses of 5, 10, 20, 30, and 40 µg/kg/min during 3-minute Whereas Nagel et al. compared DSE and DCMR directly, Hundley
periods to achieve ≥85% of the APMHR, with atropine added as an et al.17 used DCMR to study individuals who had poor TTE acoustic
adjunct if necessary. Regional LV wall motion was assessed using a windows that prevented the use of second harmonic contrast-enhanced
16-segment model; 50% of diameter or greater vessel stenosis was defined DSE imaging.17 One hundred and sixty-three subjects (74 women and
as CAD, which was detected in 107 of the study participants. Eighteen 89 men) underwent DCMR after DSE was insufficient to view all
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 215

93 91
TABLE 17.4  Comparison of DSE and 89
DCMR With X-Ray Coronary Angiography 80 80 81
84 82
80 79
75
DSE DCMR P 72
68
Sensitivity 74% 86% < .05 62
Specificity 70% 86% < .05
50
Positive predicting value 81% 91% < .05
Negative predicting value 61% 78% < .05
Accuracy 73% 86% < .005

DCMR, Dobutamine stress cardiovascular magnetic resonance; DSE,


dobutamine stress echocardiography.
From Nagel E, Lehmkuhl HB, Bocksch W, et al. Noninvasive
diagnosis of ischemia-induced wall motion abnormalities with the use
of high-dose dobutamine stress MRI. Circulation. 1999;16;730–732.
Sensitivity Specificity Accuracy PPV NPV
FIG. 17.7  Sensitivity, specificity, diagnostic accuracy, and positive and
endocardial segments as defined by the American Society of Echocar- negative predictive values (PPV and NPV, respectively) of high-dose
diography. Ten percent of study participants undergoing DSE had 5 to dobutamine stress cardiovascular magnetic resonance for coronary artery
8 and 90% had ≥8 endocardial segments not visualized with echocar- disease (>50% diameter stenoses). Orange bars, left anterior descending
diography. Study participants were excluded if they had intracranial coronary artery; peach bar, left circumflex coronary artery, blue bars,
right coronary artery. (From Wahl A, Paetsch I, Roethemeyer S, Klein
metal, a pacemaker, claustrophobia, or a known contraindication to
C, Fleck E, Nagel E. High dose dobutamine-atropine stress cardiovascular
receiving dobutamine or atropine. Images were obtained in three apical
MR imaging after coronary revascularization in patients with wall motion
(horizontal, vertical, and apical long-axis) views and three short-axis abnormalities at rest. Radiology. 2004;233:210–216.)
(base, middle portion, and apex) views of the left ventricle. Images
were continuously acquired while 5-minute dobutamine infusions of
5, 10, 20, and 40 µg/kg/min were given to achieve 85% APMHR (with of 89% and 80%, respectively. These bSSFP imaging techniques allow
atropine administered in 0.3 mg/min increments if 85% APMHR was for the acquisition of multiple slice positions during a single breath-
not reached). After 10 minutes of recovery, additional images were hold and therefore reduce scan time by a factor of 3 to 4 (see Fig. 17.6).
obtained to confirm any inducible LV wall motion had returned to As shown in Table 17.5, the sensitivity and specificity of DCMR for
baseline. Inducible ischemia was evident in 36 study participants and identifying >50% coronary arterial luminal narrowings remains similar
absent in 103 study participants; although 29 study participants devel- or higher to that observed with other noninvasive imaging techniques.
oped chest discomfort, only 8% developed a concurrent new LV wall Recently, qualitative assessment has become more accurate with
motion abnormality. In study participants who later underwent x-ray the incorporation of tagging techniques (e.g., spatial modulation of
coronary angiography, the sensitivity and specificity for detecting a magnetization [SPAMM]) or the use of radiofrequency pulses to create
>50% luminal diameter narrowing were both 83%. Based on the results regular dark bands of selective saturation at the onset of image. Kuij­
of this study, the authors concluded that DCMR provides a useful pers et al.63 reported on the qualitative assessment of tagged images
mechanism to diagnose inducible ischemia and assess LV contraction in comparison with nontagged cine CMR. One hundred ninety-four
in study participants not well suited for stress echocardiography. study participants with chest pain at rest and during increasing doses
Stress echocardiography is difficult when subjects have resting LV of dobutamine were examined during breath-hold, with and without
wall motion abnormalities because there are false positives and a ten- myocardial tagging at three short-axis levels. Study participants with new
dency for the observer to overinterpret. In many of the aforementioned wall motion abnormalities were examined with x-ray coronary angiogra-
studies, study participants with a prior MI or resting wall motion phy. Tagged DCMR images detected new LV wall motion abnormalities
abnormalities were excluded. Wahl et al.61 evaluated a group of 160 in 68 study participants, compared with LV wall motion abnormalities
participants to document the utility of DCMR in individuals with pre- identified in only 58 study participants without tagged images; x-ray
viously documented resting LV wall motion abnormalities. The subjects coronary angiography data corresponded well (P = .002). This was the
had prior revascularization with underlying resting LV wall motion first study to suggest that high-dose DCMR with qualitative assessment
abnormalities, and were difficult to assess with DSE because of the of tag deformation may detect inducible LV wall motion abnormali-
variability in interpreting LV wall motion with poor visualization. The ties with greater accuracy than those images acquired without tagging.
study demonstrated that the sensitivity and specificity of DCMR for In a meta-analysis of studies comparing DCMR with x-ray coronary
detecting coronary artery luminal narrowing ≥50% in this patient angiography in 754 study participants,64 stress-induced wall motion
population was 89% and 84%, respectively (Fig. 17.7). The sensitivity abnormalities yielded a sensitivity of 83% and a specificity of 86% for
of detecting luminal narrowing of one, two, or three epicardial arteries identifying >50% coronary arterial luminal narrowings. From these
was 87%, 91%, and 100%, respectively. Throughout this study, there data, it was concluded that DCMR demonstrates a high overall sensitiv-
was only a single major complication which resulted in external defi- ity and specificity for the diagnosis of CAD in study participants with
brillation. This study demonstrates that high-dose DCMR can be useful high disease prevalence.
even in study participants with resting LV wall motion abnormalities
and prior coronary artery revascularization. UTILITY OF DOBUTAMINE STRESS
Many of the previous studies were unable to use single breath-hold CARDIOVASCULAR MAGNETIC RESONANCE FOR
techniques and consequently led to longer examination times and less
accurate images. Paetsch et al.62 was able to use bSSFP sequences com-
IDENTIFYING CONTRACTILE RESERVE
bined with parallel imaging acquisitions to acquire images to identify Several studies have also examined the utility of DCMR for identify-
≥50% coronary artery diameter narrowing with sensitivity and specificity ing LV myocardial segments that, because of myocardial stunning or
216 SECTION II  Ischemic Heart Disease

TABLE 17.5  Sensitivity and Specificity of Dobutamine Stress Cardiovascular Magnetic


Resonance for Detection of ≥50% Coronary Arterial Luminal Narrowings
Author Dose (µg/kg/min) No. of Patients Sensitivity Specificity
55
Pennell et al. 20 25 91 —
Van Rugge et al.59 20 45 81 100
Van Rugge et al.58 20 39 91 80
Baer et al.57 20 35 84 —
Nagel et al.60 40 + atropine 208 86 86
Hundley et al.17 40 + atropine 163 83 83
Wahl et al.61 40 + atropine 160 89 84
Gebker et al.94 40 455 85a 82a
Summary 1130 86b 84b
a
≥70%.
b
Weighted average.

hibernation, display abnormal wall motion at rest but will improve epicardial coronary artery revascularization as compared with results
in contractility after mechanical coronary arterial revascularization. pertaining to myocardial metabolic activity detected with FDG-PET.
Early studies examining contractile reserve compared DCMR with Sayad et al.68 demonstrated that tissue tagging along with DCMR
other modalities, including DSE and nuclear studies. Development imaging predicted viability of hibernating and stunned myocardium
of tagging has better enabled quantitative assessment of myocardial in 10 individuals displaying LV segmental wall motion abnormalities
viability; numerous studies have used tagging to quantitatively assess at rest and referred for coronary artery revascularization. The results
myocardial viability. Recently, there have been several studies looking showed that quantitative DCMR had a sensitivity of 89%, a specificity
at specific characteristics of the myocardium, including movement of of 93%, a negative predictive value of 82%, and a positive predictive
individual layers, circumferential shortening, and wall thickening, all value of 96% for recovery of segmental function after revascularization.
of which represent important determinants of potential recovery after The authors alluded to the excellent correlation between LV end-systolic
revascularization.65 wall thickness at peak dobutamine infusion and after revascularization
In the past, modalities such as DSE and nuclear medicine studies (Fig. 17.8). Using a combination of three apical views, all myocardial
have been used to assess contractile reserve. Dendale et al.4 explored segments were completely assessed; it was found that LV myocardial
whether low-dose DCMR represents a reasonable alternative. The study segments with an end-systolic wall thickness at rest <7 mm were unlikely
indicated that it is possible to perform a low-dose DCMR examination to demonstrate contractile reserve with dobutamine or recovery of
during the second week after MI without risk of serious adverse events. function after revascularization.
When compared with DSE, there was similar efficacy and overall accu- After an MI, the transmural extent of necrosis and decrease in LVEF
racy in prediction of recovery of contractile function (79% with DCMR can vary considerably. To determine contribution of residual subepi-
and 83% with DSE). However, this study was limited by the lack of cardial viable myocardium to global LV, Bogaert et al.69 studied 12
quantitative analysis of the DCMR images. individuals with single-vessel disease after successful reperfusion of a
Quantitative analysis was performed by Saito et al.66 in a study to first transmural anterior MI with DCMR tagging (Fig. 17.9). Measured
compare DSE with tagged DCMR images. The study used an original parameters included regionally quantified fiber strains, wall thickening,
program which traced all intersecting points automatically; the LV wall and LVEF in study participants at 1 week and 3 months after MI. Normal
motion of study participants with ischemic heart disease and normal and shear strains were calculated based on displacement of node points
subjects was analyzed using low-dose DCMR. Viability of the myocar- along the subepicardial and subendocardial contours from end diastole
dium was subsequently determined by a quantitative analysis of the to end systole. Improved subepicardial fiber shortening during DCMR
improvement of the regional wall motion. The sensitivity of DCMR between week 1 and the 3 months after MI was associated with an
with tagging was 75.9% compared with 65.5% for DSE. The accuracy of improved regional LV wall motion and global LVEF. Consequently, the
DCMR and DSE was 78% and 72%, respectively. In this study there were functional recovery of myocardial strain in viable subepicardial regions
two obese participants, one of whom was misdiagnosed with regards was attributed as a mechanism of late improvement in regional and
to viability of the inferior wall only on echocardiography. global LVEF after transmural infarction. This study confirmed the neces-
DCMR has also been compared with metabolic assessments of viability sity of early reperfusion because restoration of flow in the infarct-related
obtained during radionuclide studies. Baer et al.67 reported that imple- vessel appears to preserve fibers in the subepicardial and lateral border
mentation of DCMR provided more significant information regarding zone of a transmural MI (Fig. 17.10).
viability than resting LV end-diastolic thickness. End-diastolic wall Whereas Bogaert et al. studied mathematically quantified fiber strains,
thickness at rest and dobutamine-induced systolic wall thickening assessed Geskin et al.70 examined the percent of intramyocardial circumferential
by DCMR were compared with positron emission tomography (PET)34 segment shortening. This study demonstrated that an increase in cir-
with 18F-fluorodeoxyglucose (FDG).18 The authors concluded that DCMR cumferential shortening in resting dysfunctional segments during DCMR
was a better predictor of residual metabolic activity (sensitivity of 81%, predicted the improvement in intramyocardial circumferential at 8 weeks
specificity of 95%, and positive predictive accuracy of 96%) than PET after MI. Using low-dose dobutamine, they studied a group of 20 par-
(sensitivity of 72%, specificity of 89%, and positive predictive accuracy ticipants with a first reperfused MI. A normal response to dobutamine
of 91%). From these results, it was concluded that contractile reserve administration was defined as a ≥5% increase in percent intramyocardial
as assessed by DCMR in myocardial segments akinetic at rest conditions circumferential segment shortening. The authors noted that a normal
may more accurately predict recovery of contractile function after increase in shortening elicited by dobutamine within dysfunctional
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 217

Contractile reserve in abnormal segment Correlation between end-systolic thickness at


N = 43 peak dobutamine and post-revascularization
20 N = 43
20

End-systolic thickness (mm)

End-systolic thickness post-


16

revascularization (mm)
12 15

8 10 y = 1.08x − 0.31
r = 0.95
4 5

0 0
Rest Peak Post 0 5 10 15 20
A Dobutamine revascularization B End-systolic thickness at peak dobutamine (mm)
FIG. 17.8  (A) Plot of end-systolic wall thickening at rest, at peak dobutamine, and after revascularization in
all 43 abnormal segments. No segment with end-systolic wall thickness less than 7 mm had contractile
reserve or improved after revascularization. (B) Regression line for end-systolic wall thickening at peak
dobutamine and after revascularization. (From Sayad DE, Willett DL, Hundley WG, Grayburn PA, Peshock
RM. Dobutamine magnetic resonance imaging with myocardial tagging quantitatively predicts improvement
in regional function after revascularization. Am J Cardiol. 1998;82(9):1149–1151, A10.)

A B C D
FIG. 17.9  Short-axis basal views at baseline (rest) and peak dobutamine dose (40 µg/kg/min) before and
after cardiovascular magnetic resonance tagging. Diastolic phase of a normal left ventricle at rest without
tagging (A) and with tagging (B). (C) Early systolic phase of the same left ventricle at peak dobutamine dose.
Wall contraction pattern (wall thickening) appears to be normal. (D) Same phase as panel C. Preservation of
tagging lines at the anterior left ventricular wall. There is akinesia of three segments: septal, anterior-septal,
and anterior (arrows), which indicate myocardial ischemia. Coronary angiography showed significant stenosis
in the left anterior descending. (From Kuijpers D, Ho KY, van Dijkman PR, Vliegenthart R, Oudkerk M. Dobu-
tamine cardiovascular magnetic resonance for the detection of myocardial ischemia with the use of myocardial
tagging. Circulation. 2003;107(12):1592–1597.)

midwall and subepicardium predicted greater circumferential recovery may be superior to LGE as a predictor of recovery of function in seg-
at 8 weeks after MI, but the response within the subendocardium was ments with intermediate degrees of LGE.
not predictive. These results confirm that the response of intramyocardial
function to low-dose DCMR after reperfused MI can be quantified
with CMR tagging and used to forecast function recovery after MI.
DETERMINATION OF CARDIAC PROGNOSIS
Although wall thickening as measured by DCMR is one method of Although being able to diagnose myocardial ischemia has important
assessing contractility, scar quantification by late gadolinium enhance- short-term implications, the first report regarding predictive value
ment (LGE)56 imaging represents another predictor of improvement of DCMR was established in 2002.5 A total of 279 study participants
of wall motion after revascularization. Wellnhofer et al.71 examined 29 were followed over an average of 20 months. After DCMR testing, the
individuals with CAD using DCMR and LGE with gadolinium- occurrence of unstable angina, heart failure, coronary arterial revas-
diethylenetriamine pentaacetic acid (Gd-DTPA). In myocardial segments cularization, cardiac death, and death attributable to any cause were
with transmural extent of infarction <50%, DCMR was more likely determined. Reduced LVEF and evidence of inducible myocardial
than LGE to identify improvement in contractility after revasculariza- ischemia during DCMR were associated with future MI and cardiac
tion (Fig. 17.11). For those with no or extensive infarcts, the techniques death (Fig. 17.12). Using a multivariate analysis, a low LVEF, inducible
had similar efficacy. The reason for this finding is unclear, but may ischemia, or contractile reserve was associated with cardiac death and
relate to the fact that DCMR is a functional test, whereas LGE assesses future MI, independent of the presence of risk factors for coronary
underlying cardiac structure. The authors of this study concluded that arteriosclerosis or MI.
LGE and DCMR provide complementary information. LGE is able to Jahnke et al.6 have also reported on the prognostic utility of stress-
localize and quantify scar, but, with nontransmural scars (1%–74%), induced change in LV wall motion during intravenous dobutamine.
it has impaired specificity as a predictor of recovery. Therefore DCMR This study directly compared the prognostic value of adenosine stress
218 SECTION II  Ischemic Heart Disease

Rest 10 µg 20 µg Max

End diastole
End systole

FIG. 17.10  Contractile reserve in left circumflex (LCX) and right coronary artery (RCA) disease. The format
is the same as in Fig. 14.6. At rest, a severely hypokinetic inferolateral segment was found (white arrows).
Under low-dose dobutamine infusion, up to 20 µg/kg/min, a substantial and gradual improvement in the left
ventricular contractile response of the inferolateral segment could be demonstrated, although at maximum
stress, both the inferolateral and inferior segments became akinetic. The “biphasic profile of contraction” of
the inferolateral segment was compatible with viable myocardium. On x-ray coronary angiography, there is
subtotal occlusion (black arrowheads) of the distal portion of a marginal branch and high-grade stenosis of
the distal LCX (black arrows in lower left image) as well as a high-grade stenosis of the distal RCA23 (black
arrows in lower right image). (From Paetsch I, Jahnke C, Fleck E, Nagel E. Current clinical applications of
stress wall motion analysis with cardiac magnetic resonance imaging. Eur J Echocardiogr. 2005;6(5):317–326.)

CMR perfusion and DCMR wall motion imaging in 461 study partici- stress echocardiography, but with clinical predictors suggesting an inter-
pants after 2.3 ± 1 years follow-up. In their study, the prognostic utility mediate risk for the development of a cardiac event during noncardiac
of the presence or absence of dobutamine-induced LV wall motion surgery, the results of DCMR stress testing can be used to identify those
abnormalities were similar to those identified during adenosine CMR at low and high risk for heart failure, MI, and cardiac death in the
contrast-enhanced perfusion defects. The 3-year event-free survival was perioperative and postoperative period.
99.2% for study participants with normal adenosine or DCMR. Initial studies have also suggested that DCMR may be a suitable
DCMR has also been used to determine preoperative cardiovascular alternative to echocardiography before either renal or liver transplant
risk among those undergoing cardiac and noncardiac surgery. Rerkpat- to identify those at increased risk for perioperative cardiovascular com-
tanapipat et al.72 studied a group of 102 patients for preoperative risk plications. In a series of over 60 study participants who underwent
of heart failure, MI, and death during or after noncardiac surgery. Cardiac DCMR, Dundon et al.73 demonstrated that it was not only feasible
events were experienced in 6 study participants (death in 4, nonfatal but also had a negative predictive value of 100%. These findings dem-
MI in 1, and heart failure in 1) during surgery or in their postoperative onstrate the feasibility of DCMR in study participants with end-stage
course. The presence of inducible ischemia was independently associ- renal disease before transplantation and also suggest that DCMR may
ated with an increase in the future occurrence of a perioperative cardiac be able to reduce the need for invasive x-ray coronary angiography in
event. This study determined that in participants not able to undergo this high-risk, asymptomatic group. In a separate series of over 51 study
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 219

100 N = 86 N = 53 N = 51 N = 45 N = 53

80

60
(%)

40

20

0
No <25 25–49 50–74 ≥75
LGE (% transmurality)

Prevalence Sensitivity
Specificity Correct predictions
FIG. 17.11  Transmurality of late gadolinium enhancement (LGE)56: subgroup analysis. Bars refer to the
prevalence of recovery and sensitivity, specificity, and percentage of correct predictions by dobutamine
stress cardiovascular magnetic resonance (DCMR) and are subgrouped with respect to LGE (cutoff 25%). The
specificity of DCMR remains high irrespective of the extent of LGE. The test retains a high sensitivity in 25%
to 49% LGE. (From Wellnhofer E, Olariu A, Nagel E, et al. Magnetic resonance low-dose dobutamine test is
superior to SCAR quantification for the prediction of functional recovery. Circulation. 2004;109(18):2172–2174.)

Cardiac death or MI after dobutamine/atropine CMR similar, parallel fashion to DSE. More long-term studies with clinical
outcomes after transplantation would help to better define the role of
1.0 LVEF ≥40%; prior CMR stress testing in this clinical scenario.
(–) ischemia
Proportion hard event-free

0.9
LVEF ≥40%;
TISSUE TAGGING DURING DOBUTAMINE STRESS
(+) ischemia CARDIOVASCULAR MAGNETIC RESONANCE
0.8
LVEF <40%
As previously mentioned, the application of tissue tagging (SPAMM)
0.7 facilitates assessment of LV wall motion with a high degree of
precision.75–77 With this technique, it is possible to calculate the local
0.6 Lagrangian strain tensor, which describes the motion around a given
point in the tissue as it transverses through space and time. Different
0.5 tagging schemes have been developed, including lines,78 grids,79 and
radial stripes.80 For the quantitative analysis, tag intersection points are
0 6 12 18 24 30 36 used to assess contraction and relaxation. To date, tissue tagging has
Time (months) been the most widely reported form of quantitative analysis. As described
FIG. 17.12  Kaplan-Meier survival curve indicative of cardiovascular earlier, several studies have documented the utility of tissue tagging for
events. In 279 individuals undergoing dobutamine stress cardiovascular identification of abnormalities of LV systolic thickening associated with
magnetic resonance for chest pain, the proportion of individuals free of myocardial ischemia or contractile reserve.
cardiac death or myocardial infarction (MI) (y-axis) and the 3-year follow- A unique application of tissue tagging is the potential assessment
up (x-axis) are shown. As noted, individuals with a left ventricular ejection of LV diastolic function. Paetsch et al.81 evaluated the diastolic param-
fraction (LVEF) of at least 40% and no inducible ischemia exhibited eters from myocardial tagging of 20 study participants with low- and
fewer cardiac events compared with those individuals with inducible
high-dose DCMR to identify study participants with significant CAD.
ischemia or those with an LVEF <40%.
Diastolic (velocity of circumferential lengthening and diastolic rotation
velocity) and systolic (circumferential shortening, systolic rotation, and
participants, Reddy et al.74 presented the feasibility of using DCMR to systolic rotation velocity) parameters during low-dose DCMR changed
evaluate for obstructive CAD before liver transplantation and evaluate significantly in study participants without CAD, but not in those with
liver pathology at the same time. Although there are few long-term CAD (P < .05). As seen in Fig. 17.13, identification of study participants
outcome data on which to base decisions in CMR stress testing before with and without CAD was possible using the diastolic parameter “time
transplantation, the available data suggest that DCMR performs in a to peak untwist.” Thus the results of this study show that myocardial
220 SECTION II  Ischemic Heart Disease

End diastole End systole End systole

C
FIG. 17.13  Representative tagging end-diastolic and end-systolic apical cardiovascular magnetic resonance
images of a patient without significant coronary artery disease. (A) Rest. (B) Low-dose dobutamine stress.
(C) High-dose dobutamine stress. Note the increase in systolic rotation and endocardial motion from rest to
low-dose stress. (From Paetsch I, Foll D, Kaluza A, et al. Magnetic resonance stress tagging in ischemic
heart disease. Am J Physiol Heart Circ Physiol. 2005;288:H2708–H2714.)

tagging helps in a quantitative analysis of systolic and diastolic function up to 20 minutes, but once the filter is set, a full quantitative analysis of
during both low- and high-dose dobutamine stress. the data typically takes less than 3 minutes. This represents a substantial
Another potential development for the use of tissue tagging during time savings over manual analysis, which can take up to 8 hours. Further
DCMR is the use of automated analyses of the images with harmonic research is proceeding in this area to provide users with a mechanism to
phase magnetic resonance (HARP). Image inspection with HARP enables obtain quantitative tag data in near real time. By standardizing acquisi-
substantial reduction of tag analysis times. Developed by Osman et al.,82,83 tions of tagged images, one can preset the filter and provide very efficient
this technique concentrates on the Fourier transformation of tagged myocardial strain mapping. By extracting HARP images directly from the
images, which is directly related to the motion of the tag lines. The raw data using faster microprocessors, the possibility of on-line detailed
HARP technique can be used with any tagging strategy provided the tag quantitative assessment of two-dimensional (2D) myocardial strains for
pattern is planar and tag lines are placed a uniform distance from one use during stress testing may become a reality in the near future.
another. In addition to wall motion and thickening, myocardial strain Pan et al.84 have proposed a potentially fast and semiautomatic
can be analyzed. Selecting tag filter specifications for the images requires method for tracking three-dimensional (3D) cardiac images. Using a
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 221

Quantification of longitudinal strain and strain rate


Cine imaging Strain encoded MR 0
−2 0 200 400 600 800

−4

Longitudinal strain (%)


−6
−8
−10
Baseline strain
A B −12 Intermediate strain
Baseline −14 Peak strain
−16
−18
I −20 Peak systolic strain values
2
Baseline strain rate
Intermediate strain rate

Longitudinal strain rate (1/8)


C D
1 Peak strain rate
Intermediate stress

0
0 200 400 600 800

−1 Peak systolic strain rate values

E F Time in ma
(after R-wave)
Peak dobutamine stress J −2
Quantitative coronary angiography

K L

G H
LAO 20° RAO 30°
10 minutes after dobutamine stress CRA10° CAU10°

FIG. 17.14  Strain-encoded images (SENC) in a patient with coronary artery disease: mid–short-axis SENC
and cine images are shown (A–H). With cine images, inducible ischemia is detected only during peak stress
in the mid-anterior left ventricular wall (E, arrow). Conversely, with SENC, a strain defect is already observed
during intermediate stages (D, arrow). The arrowheads indicate signal voids on SENC images due to low
signal-to-noise ratio (F and H). Peak systolic strain values decrease as stress increases (I), but peak systolic
strain rate remains unchanged (J). These findings were further investigated by coronary angiography, which
confirmed a 68% stenosis of the left anterior descending artery (K and L). MR, Magnetic resonance. (From
Korosoglou G, Lehrke S, Wochele A, et al. Strain-encoded CMR for the detection of inducible ischemia during
intermediate stress. JACC Cardiovasc Imaging. 2010;3(4):361–371.)

material mesh model that is built to represent a collection of material map.85 This can be done for long- or short-axis images (Fig. 17.14). In
points inside the LV wall, the phase time-invariance property of mate- the setting of a DCMR, images are obtained at baseline and at rest. It has
rial points is then used to track mesh points. With a series of 9 timeframe been studied at both 1.5 T and 3 T.86 Initial analyses are promising and
CMR images, researchers were able to initialize settings, build the mesh, have demonstrated that SENC is as robust as other techniques and may
and track the 3D images in approximately 10 minutes. Eventually, more improve the identification of myocardial ischemia and contractile reserve
complex mesh algorithms will be developed that will allow 3D calcula- during both intermediate and full-dose dobutamine stress testing.87,88
tion of regional and global wall motion in real time. Feature-tracking analysis may be used to measure LV thickening
Another technique used to assess myocardial strain is strain-encoded during DCMR. This technique uses standard bSSFP images to calculate
(SENC) imaging. This technique is a modified SPAMM tagging sequence.85 both longitudinal and radial strain.89 Both LV and right ventricular (RV)
In contrast to other tagging methods, the SENC gradient is applied longitudinal strain are calculated from the four-chamber view (Fig.
orthogonal to the imaging plane. The magnitude of SENC images at 17.15), and the LV radial strain is obtained from the short-axis view.
both a high-tuning frequency and a low-tuning frequency are computed Initial studies demonstrated the feasibility of feature-tracking analysis
and placed over the anatomic slice image, which produces the strain and, subsequently, demonstrated a reduced interobserver variability in
222 SECTION II  Ischemic Heart Disease

Short-Axis
LV Long-Axis
RV Long-Axis

FIG. 17.15  Tracking in the short-axis and long-axis orientation. The figure shows a representative example
of the tracking in short-axis and long-axis orientation of the left ventricle (LV) and right ventricle (RV).30 (From
Schuster A, Kutty S, Padiyath A, et al. Cardiovascular magnetic resonance myocardial feature tracking detects
quantitative wall motion during dobutamine stress. J Cardiovasc Magn Reson. 2011;13:58.)

identifying wall motion abnormalities.90 Even at less than peak doses of from LGE because prior infarcts also have associated perfusion defects
dobutamine, feature tracking has been able to identify the development (Fig. 17.16).
of wall motion abnormalities.91
LIMITATIONS TO LEFT VENTRICULAR WALL
ROLE OF DOBUTAMINE PERFUSION IMAGING MOTION ASSESSMENTS DURING
Although much of the focus during DCMR is on the increase in con-
DOBUTAMINE STRESS
tractility and heart rate, dobutamine also indirectly increases coronary A frequent challenge for the interpretation of LV wall motion stress
blood flow. As cardiac metabolism increases, the metabolites induce testing relates to predobutamine and postdobutamine wall motion
coronary arteriolar vasodilation, resulting in increased coronary blood analyses in individuals with resting LV hypokinesis (e.g., those with LV
flow. Initial studies comparing wall motion and perfusion during DCMR wall motion abnormalities because of prior MI).96 In these circumstances,
showed that perfusion defects had the potential to differentiate LV wall it can be difficult to ascertain whether a segment that is persistently
motion abnormalities because of true myocardial ischemia from heart hypokinetic (rest and stress) exhibits myocardial ischemia or whether
rate–related bundle branch blocks.92 Subsequently, myocardial perfusion residual functioning myocytes within and around a previously scarred
during DCMR has been found to enhance the utility of DCMR for infarct cannot mount a contractile response that overcomes the station-
detecting myocardial ischemia. In a series of 42 study participants, ary forces related to the scar.97–99
Falcao et al.93 found that by adding perfusion imaging to standard wall Those patients with resting LV concentric hypertrophy may not
motion analysis in DCMR, the sensitivity to detect obstructive CAD develop an inducible LV wall motion abnormality in the setting of
went from 80% to 92%. In a larger series of 455 study participants, epicardial coronary artery stenoses.100 In a large DSE series, Smart et al.100
Gebker et al.94 found that by adding perfusion imaging during DCMR, showed that there were fewer recognized stress-induced LV wall motion
the sensitivity for detecting obstructive CAD increased from 85% to abnormalities among individuals with CAD and LV concentric remodel-
91%, but they also found that the specificity was reduced because of ing. Interobserver agreement of the LV wall motion assessment was
artifacts and nonspecific perfusion abnormalities. high when LV systolic function was normal or significantly reduced,
Dobutamine-related perfusion assessments may have clinical but agreement was reduced as the LV systolic function became impaired
utility in other disease states. For those participants with concentric or significant hypertrophy was present.97 In study participants with LV
LV hypertrophy, the addition of perfusion imaging improves the sen- concentric remodeling, it should be recognized that it may be difficult
sitivity of DCMR for obstructive CAD. In a series of 187 participants to fully appreciate the functional changes associated with ischemia. By
who underwent DCMR before invasive x-ray coronary angiography, lowering wall stress, a significant determinant of myocardial oxygen
Gebker et al.95 found that the addition of perfusion imaging to wall demand, LV hypertrophy may make it more difficult to reach the ische-
motion analysis improved the accuracy in identifying obstructive CAD mic threshold needed to produce wall motion abnormalities.101,102
from 71% to 82% in those with concentric LV remodeling. When Finally, when interpreting LV wall motion during intravenous dobu-
interpreting dobutamine perfusion imaging from CMR, it is critical tamine, one assumes that cardiac workload will increase to a sufficient
to interpret any perfusion defects identified with findings obtained degree during dobutamine stress such that a wall motion abnormality
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 223

will occur because of reduced oxygen supply delivered through a stenosis The stress discordance may have resulted from an inadequate stress
epicardial coronary artery. Interestingly, in a prospective series of 278 load because older individuals are often more sensitive to the vascular
older adults who underwent DCMR, Vasu et al.101 demonstrated that effects of dobutamine. In the elderly, dobutamine may reduce both LV
nearly 60% of dobutamine-induced perfusion defects were not associated preload (by reducing the end-diastolic volume) and LV afterload mani-
with corresponding LV wall motion abnormalities during dobutamine. fest as a lower blood pressure with dobutamine. Both effects decrease
their LV wall stress during dobutamine. This situation can occur more
frequently when LV hypertrophy is present, causing further reductions
in LV end-diastolic volume because of the relatively small LV cavity
DCMR Stress Test
size. This finding is demonstrated in Fig. 17.17 in which a 64-year-old
Late gadolinium male who underwent DCMR demonstrated a hyperdynamic LV wall
enhancement response to dobutamine, but displayed an associated apical perfusion
(endocardial)
defect at peak stress. X-ray coronary angiography revealed multivessel
disease, including a 90% proximal left main stenosis. If one did not
assess myocardial perfusion during the test and only focused on LV
wall motion, inducible myocardial ischemia may have been missed.
Stress-related wall Prior myocardial
motion abnormality infarction ADENOSINE AND DIPYRIDAMOLE AS WALL
MOTION STRESS AGENTS DURING
CARDIOVASCULAR MAGNETIC RESONANCE
Dipyridamole, adenosine, and regadenoson exert their effect through
coronary vasodilation, producing up to 5-fold increase in coronary
Stress perfusion defect Stress perfusion defect
artery blood flow. The safety profile of dipyridamole is favorable, with
a well-known and low complication rate. Side effects include chest pain,
shortness of breath, flushing, and dizziness, all of which can be reversed
with aminophylline administration.103 Pennell et al.104 were the first to
Inducible Bundle Inducible report the use of vasodilator dipyridamole with CMR in the assessment
No
myocardial branch myocardial of LV wall motion. In 40 study participants, dipyridamole was infused
ischemia
ischemia block ischemia at a dose 0.56 mg/kg, along with a 10-mg bolus after 10 minutes. Com-
FIG. 17.16  Algorithm to interpret left ventricular wall motion abnor- pared with thallium tomography, the sensitivity for detecting new wall
malities provoked during dobutamine stress cardiovascular magnetic motion abnormalities and identifying perfusion defects associated with
resonance (DCMR). ischemia was 67%.105 After review of the data, however, there was an

FIG. 17.17  Function and cine functional cardiovascular magnetic resonance (CMR) images from a 64-year-
old man obtained during a dobutamine CMR. On the left, an apical perfusion defect is demonstrated. On
the right, still short-axis images of end diastole and end systole demonstrate no left ventricular wall motion
abnormality.
224 SECTION II  Ischemic Heart Disease

apparent inability to detect smaller areas of ischemia. Although the capacity, and hemodynamic response. The ability to extract informa-
procedure was generally well tolerated, mild side effects (flushing, chest tion related to cardiac prognosis is often based on exercise duration.
pain, and headache) were common. In the past, exercise stress CMR studies have been limited by motion
Paetsch et al.62 also compared DCMR with adenosine administration artifacts associated with relatively long image acquisitions. Recently,
for identifying LV wall motion abnormalities indicative of ischemia. however, faster imaging sequences have become available that allow
Seventy-nine study participants with suspected or known CAD, who for rapid image acquisitions after exercise stress. Roest et al.107 studied
were scheduled for x-ray coronary angiography, underwent stress CMR 16 healthy subjects using supine bicycle ergometry stress on a CMR-
with adenosine followed by dobutamine. Segmental perfusion was graded compatible bicycle ergometer (Fig. 17.18). Image blurring was mini-
by the transmural extent of inducible perfusion deficits (<25%, 25%–50%, mized by breath-holding at end expiration for all short-axis images.
51%–75%, and >75%). Fifty-three study participants (67%) had coro- LV stroke volume and LVEF increased in both ventricles in response
nary arterial luminal diameter stenoses ≥50%. The sensitivity and to exercise; as would be expected, the end-systolic volume of the two
specificity for detecting these stenoses by dobutamine or adenosine ventricles decreased, whereas the end-diastolic volumes remained the
wall motion analyses, or adenosine-induced perfusion defects were 89% same. A turbo-field echo planar imaging (EPI) sequence was used
and 80%, 40% and 96%, and 91% and 62%, respectively. Adenosine- during breath-holding as part of the exercise protocol and was com-
induced wall motion abnormalities were seen only in segments with pared with a standard EPI sequence with no significant difference.105
>75% transmural perfusion deficit. Based on the above findings, it was This study demonstrates that exercise CMR can be used to simulta-
concluded that DCMR was superior to adenosine stress for appreciating neously assess physiologic changes in both the LV and RV. Although
inducible LV wall motion abnormalities indicative of ischemia. this study did not examine individuals with CAD, it did address some
In a study of 94 subjects by Pingitore et al.,106 a head-to-head com- of the previous limitations with this modality, including movement
parison was performed between perfusion and function during dipyri- artifacts.
damole CMR stress for the detection of CAD. Wall motion and the Whereas the previous study examined LV and RV function during
perfusion reserve index exhibited similar diagnostic accuracy (86% for exercise CMR, Pedersen et al.108 showed the feasibility of using ultrafast
both) for identifying >50% coronary artery diameter stenoses. Wall CMR during a dedicated exercise protocol to assess hemodynamic
motion analyses had a higher specificity (96% vs. 66%, P < .01) and changes in the abdominal aorta in response to physical exercise. Recent
lower sensitivity (82% vs. 93%, P < .05) than the perfusion reserve theories purport that exercise-induced changes in local wall shear stresses
index. Perfusion had the highest accuracy values for coronary stenoses are responsible for the beneficial effect of exercise on atherosclerotic
<75% (cutoff 59%), whereas wall motion abnormalities exhibited the disease. With 9 healthy subjects, and using fast hybrid phase-contrast
highest accuracy for coronary stenoses >75% (cutoff 84%) (P < .001).91 sequence with k-space segmentation and EPI readouts, this study dem-
The authors concluded that during dipyridamole CMR stress testing, onstrated that retrograde flow patterns in the abdominal aorta, which
perfusion and wall motion abnormalities exhibit a similar diagnostic are associated with oscillating wall shear stresses and development of
accuracy, with perfusion showing higher sensitivity, particularly in the atherosclerosis, disappeared with increased levels of exercise. With further
detection of moderate stenoses, and wall motion showing higher speci- study, exercise CMR has the potential to help practitioners identify
ficity. Both studies demonstrated adenosine and dipyridamole promote study participants that are developing arthrosclerosis and aid in lifestyle
stress-induced regional LV dysfunction in participants with more severe and medication modifications.
(>70% diameter) coronary artery stenoses. Although these prior studies have examined supine exercise testing,
Rerkpattanapipat et al.109 demonstrated the feasibility of upright tread-
LEFT VENTRICULAR WALL MOTION DURING mill exercise in 27 individuals with CMR images obtained <1 minute
EXERCISE STRESS CARDIOVASCULAR postexercise. Ischemia was identified in 14 participants with high overall
sensitivity (79%) and specificity (85%) for detection of coronary artery
MAGNETIC RESONANCE stenosis >70%. By placing the treadmill approximately 20 feet away
Potential advantages of exercise as a stress agent include reduction from the CMR table and having study participants exercise immediately
in stress protocol duration, acquisition of data regarding functional before imaging, the authors were able to counteract one of the previous

Exercise bike attached to the scanner Treadmill positioned outside the scan room
FIG. 17.18  Methods of exercise-induced stress in patients undergoing wall motion analyses during cardio-
vascular magnetic resonance (CMR) stress testing. Left, An individual pedals a nonferromagnetic electronically
braked supine bicycle ergometer (Loda, the Netherlands). Right, An individual performs maximal treadmill
exercise stress on a treadmill located immediately outside the scanning room. Immediately after exercise,
the participant is placed on the CMR imaging table, and images of left ventricular wall motion and perfusion
are collected within 60 seconds of exercise cessation.
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 225

limitations of stress exercise CMR in that they were able to acquire


CONCLUSION
images within 60 to 90 seconds after treadmill exercise cessation. However,
there were limitations with this study in that comprehensive LV cover- Inducible LV wall motion abnormalities during stress CMR has been
age was not achieved, and results were acquired on a small number of found to be useful for identifying inducible ischemia and cardiac prog-
subjects. Further studies in this area are needed to determine the clinical nosis. Improvement in LV wall motion in myocardial segments with
utility of exercise-induced wall motion abnormalities for the assessment resting LV wall motion abnormalities during DCMR accurately identifies
of individuals with cardiovascular disease. Importantly, exercise CMR regions of the LV myocardium that will improve contractility after
can identify those at higher risk for adverse cardiovascular outcomes. coronary arterial revascularization procedures. With new CMR tagging
Using upright exercise CMR, Sukpraphrute et al.110 demonstrated the techniques, qualitative and quantitative assessments of regional LV wall
ability to identify patients who developed unstable angina, MI, or cardiac motion and strain are possible, and although not yet perfected for
death during follow-up. In those individuals with LV wall motion routine clinical use, may in the future provide real-time 3D determina-
abnormalities, 47% experienced MI, unstable angina, or cardiac death tions of regional and global LV systolic function during CMR stress
during follow-up compared with only 14% of individuals without a testing. Recognizing that LV wall motion analysis is regarded as a more
wall motion abnormality (P = .002). Development of upright treadmill specific indication of myocardial ischemia, the addition of perfusion
exercise in the CMR room and immediately adjacent to the gantry table imaging to DCMR improves the sensitivity for detecting obstructive
is also being explored. The multicenter EXercise CMR Accuracy for CAD. In selecting patients for DCMR, select those who have the neces-
Cardiovascular stress Testing (EXACT) trial of 94 patients demonstrated sary physiology to provoke myocardial ischemia, if present.
a sensitivity and specificity of 36% and 98% for exercise stress cine
CMR that improved to 79% and 99% with the inclusion of CMR perfu-
sion with a positive and negative predictive values of 92% and 96%,
REFERENCES
respectively.111 A full reference list is available online at ExpertConsult.com
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 225.e1

REFERENCES 23. Rodriguez Garcia MA, Iglesias-Garriz I, Corral Fernandez F, et al.


Evaluation of the safety of stress echocardiography in Spain and
1. Master AM, Oppenheimer ET. A simple exercise tolerance test for Portugal. Rev Esp Cardiol. 2001;54(8):941–948.
circulatory efficiency with standard tables for individuals. Am J Med Sci. 24. Picano E, Mathias W Jr, Pingitore A, Bigi R, Previtali M. Safety and
1929;177:223. tolerability of dobutamine-atropine stress echocardiography: a
2. Sechtem U, Sommerhoff BA, Markiewicz W, White RD, Cheitlin MD, prospective, multicentre study. Echo Dobutamine International
Higgins CB. Regional left ventricular wall thickening by magnetic Cooperative Study Group. Lancet. 1994;344(8931):1190–1192.
resonance imaging: evaluation in normal persons and patients with 25. Geleijnse ML, Fioretti PM, Roelandt JR. Methodology, feasibility, safety
global and regional dysfunction. Am J Cardiol. 1987;59(1):145–151. and diagnostic accuracy of dobutamine stress echocardiography. J Am
3. Pattynama PM, de Roos A. MR evaluation of myocardial ischemia and Coll Cardiol. 1997;30(3):595–606.
infarction. Top Magn Reson Imaging. 1995;7(4):218–231. 26. Wahl A, Paetsch I, Gollesch A, et al. Safety and feasibility of high-dose
4. Dendale PA, Franken PR, Waldman GJ, et al. Low-dosage dobutamine dobutamine-atropine stress cardiovascular magnetic resonance for
magnetic resonance imaging as an alternative to echocardiography in diagnosis of myocardial ischaemia: experience in 1000 consecutive cases.
the detection of viable myocardium after acute infarction. Am Heart J. Eur Heart J. 2004;25(14):1230–1236.
1995;130(1):134–140. 27. Kuijpers D, Janssen CH, van Dijkman PR, Oudkerk M. Dobutamine
5. Hundley WG, Morgan TM, Neagle CM, Hamilton CA, Rerkpattanapipat stress MRI, part I. Safety and feasibility of dobutamine cardiovascular
P, Link KM. Magnetic resonance imaging determination of cardiac magnetic resonance in patients suspected of myocardial ischemia. Eur
prognosis. Circulation. 2002;106(18):2328–2333. Radiol. 2004;14(10):1823–1828.
6. Jahnke C, Nagel E, Gebker R, et al. Prognostic value of cardiac magnetic 28. Hamilton CA, Link KM, Salido TB, Epstein FH, Hundley WG. Is
resonance stress tests: adenosine stress perfusion and dobutamine stress imaging at intermediate doses necessary during dobutamine stress
wall motion imaging. Circulation. 2007;115(13):1769–1776. magnetic resonance imaging? J Cardiovasc Magn Reson.
7. Tuttle RR, Mills J. Dobutamine: development of a new catecholamine 2001;3(4):297–302.
to selectively increase cardiac contractility. Circ Res. 1975;36(1): 29. Tighe JF Jr, Steiman DM, Vernalis MN, Taylor AJ. Observer bias in the
185–196. interpretation of dobutamine stress echocardiography. Clin Cardiol.
8. Baig MW. Pharmacologic perfusion imaging. Who needs it and why? 1997;20(5):449–454.
Postgrad Med. 1992;91(1):185–187, 190–192. 30. Pedone C, Bax JJ, van Domburg RT, et al. Long-term prognostic value of
9. Rabinovitch MA, Kalff V, Chan W, et al. The effect of dobutamine on ejection fraction changes during dobutamine-atropine stress
exercise performance in patients with symptomatic ischemic heart echocardiography. Coron Artery Dis. 2005;16(5):309–313.
disease. Am Heart J. 1984;107(1):81–85. 31. Autenrieth G, Angermann C, Goss F, Bolte HD. Stress echocardiography
10. Bruton LL, Lazo JS, Parker KL. Goodman & Gilman’s The in patients with coronary heart disease. Verh Dtsch Ges Inn Med.
Pharmacological Basis of Therapeutics. 11th ed. New York: McGraw-Hill; 1977;83:231–236.
2006. 32. Limacher MC, Quinones MA, Poliner LR, Nelson JG, Winters WL Jr,
11. Pennell DJ, Underwood SR. The cardiovascular effects of dobutamine Waggoner AD. Detection of coronary artery disease with exercise
assessed by magnetic resonance imaging. Post Grad Med J. 1991;67. two-dimensional echocardiography. Description of a clinically applicable
12. Iskandrian AS, Verani MS, Heo J. Pharmacologic stress testing: method and comparison with radionuclide ventriculography.
mechanism of action, hemodynamic responses, and results in detection Circulation. 1983;67(6):1211–1218.
of coronary artery disease. J Nucl Cardiol. 1994;1(1):94–111. 33. Paul B, Kasliwal RR. Dobutamine stress echocardiography—
13. Kugiyama K, Inobe Y, Ohgushi M, et al. Comparison of coronary methodology, clinical applications and current perspectives. J Assoc
hemodynamics during infusions of dobutamine and adenosine in Physicians India. 2004;52:653–657.
patients with angina pectoris. Jpn Circ J. 1998;62(1):1–6. 34. Pingitore A, Picano E, Colosso MQ, et al. The atropine factor in
14. Warltier DC, Zyvoloski M, Gross GJ, Hardman HF, Brooks HL. pharmacologic stress echocardiography. Echo Persantine (EPIC) and
Redistribution of myocardial blood flow distal to a dynamic coronary Echo Dobutamine International Cooperative (EDIC) Study Groups.
arterial stenosis by sympathomimetic amines: comparison of dopamine, J Am Coll Cardiol. 1996;27(5):1164–1170.
dobutamine and isoproterenol. Am J Cardiol. 1981;48(2):269–279. 35. Kamaran M, Teague SM, Finkelhor RS, Dawson N, Bahler RC.
15. Dagianti A, Penco M, Agati L, Sciomer S, Rosanio S, Fedele F. Stress Prognostic value of dobutamine stress echocardiography in patients
echocardiography: comparison of exercise, dipyridamole and referred because of suspected coronary artery disease. Am J Cardiol.
dobutamine in detecting and predicting the extent of coronary artery 1995;76(12):887–891.
disease. J Am Coll Cardiol. 1995;26(1):18–25. 36. Mathias Junior W, Doya EH, Ribeiro EE, et al. Detection of myocardial
16. Paetsch I, Jahnke C, Fleck E, Nagel E. Current clinical applications of ischemia during dobutamine stress echocardiography: correlation with
stress wall motion analysis with cardiac magnetic resonance imaging. coronary cineangiography. Arq Bras Cardiol. 1993;60(4):229–234.
Eur J Echocardiogr. 2005;6(5):317–326. 37. Armstrong WF, Pellikka PA, Ryan T, Crouse L, Zoghbi WA. Stress
17. Hundley WG, Hamilton CA, Thomas MS, et al. Utility of fast cine echocardiography: recommendations for performance and
magnetic resonance imaging and display for the detection of myocardial interpretation of stress echocardiography. Stress Echocardiography Task
ischemia in patients not well suited for second harmonic stress Force of the Nomenclature and Standards Committee of the American
echocardiography. Circulation. 1999;100(16):1697–1702. Society of Echocardiography. J Am Soc Echocardiogr. 1998;11(1):
18. Pinto Pereiro LM. Atropine. Practical Procedures. 1996;(6):1. 97–104.
19. Fetiveau R, Lanzarini L, Poli A, et al. The side effects and safety of the 38. Cerqueira MD, Weissman NJ, Dilsizian V, et al. Standardized myocardial
echo-dobutamine test. The experience with 373 patients. G Ital Cardiol. segmentation and nomenclature for tomographic imaging of the heart.
1995;25(2):193–201. A statement for healthcare professionals from the Cardiac Imaging
20. Krenning BJ, Geleijnse ML, Poldermans D, Roelandt JR. Methodological Committee of the Council on Clinical Cardiology of the American
analysis of diagnostic dobutamine stress echocardiography studies. Heart Association. Circulation. 2002;105(4):539–542.
Echocardiography. 2004;21(8):725–736. 39. Poldermans D, Arnese M, Fioretti PM, et al. Improved cardiac risk
21. Cortigiani L, Picano E, Coletta C, et al. Safety, feasibility, and prognostic stratification in major vascular surgery with dobutamine-atropine stress
implications of pharmacologic stress echocardiography in 1482 patients echocardiography. J Am Coll Cardiol. 1995;26(3):648–653.
evaluated in an ambulatory setting. Am Heart J. 2001;141(4):621–629. 40. Davila-Roman VG, Waggoner AD, Sicard GA, Geltman EM, Schechtman
22. Mertes H, Sawada SG, Ryan T, et al. Symptoms, adverse effects, and KB, Perez JE. Dobutamine stress echocardiography predicts surgical
complications associated with dobutamine stress echocardiography. outcome in patients with an aortic aneurysm and peripheral vascular
Experience in 1118 patients. Circulation. 1993;88(1):15–19. disease. J Am Coll Cardiol. 1993;21(4):957–963.
225.e2 SECTION II  Ischemic Heart Disease

41. Kertai MD, Boersma E, Bax JJ, et al. A meta-analysis comparing the 61. Wahl A, Paetsch I, Roethemeyer S, Klein C, Fleck E, Nagel E. High-dose
prognostic accuracy of six diagnostic tests for predicting perioperative dobutamine-atropine stress cardiovascular MR imaging after coronary
cardiac risk in patients undergoing major vascular surgery. Heart. revascularization in patients with wall motion abnormalities at rest.
2003;89(11):1327–1334. Radiology. 2004;233(1):210–216.
42. Fiuza M, Pereirinha A, de Padua F, Lopes MG. The classic exercise test 62. Paetsch I, Jahnke C, Wahl A, et al. Comparison of dobutamine stress
or by imaging? New applications of Doppler echocardiography. Rev Port magnetic resonance, adenosine stress magnetic resonance, and
Cardiol. 1993;12(5):483–493, 409. adenosine stress magnetic resonance perfusion. Circulation.
43. Chaudhry FA, Tauke JT, Alessandrini RS, Greenfield SA, Tommaso CL, 2004;110(7):835–842.
Bonow RO. Enhanced detection of ischemic myocardium by 63. Kuijpers D, Ho KY, van Dijkman PR, Vliegenthart R, Oudkerk M.
transesophageal dobutamine stress echocardiography: comparison with Dobutamine cardiovascular magnetic resonance for the detection of
simultaneous transthoracic echocardiography. Echocardiography. myocardial ischemia with the use of myocardial tagging. Circulation.
2000;17(3):241–253. 2003;107(12):1592–1597.
44. Fedele F, Scopinaro F, Montesano T, et al. Characterization of reversible 64. Nandalur KR, Dwamena BA, Choudhri AF, Nandalur MR, Carlos RC.
myocardial dysfunction by magnetic resonance imaging. Herz. 1994; Diagnostic performance of stress cardiac magnetic resonance imaging in
19(4):210–220. the detection of coronary artery disease: a meta-analysis. J Am Coll
45. Vignola PA, Bloch A, Kaplan AD, Walker HJ, Chiotellis PN, Myers GS. Cardiol. 2007;50(14):1343–1353.
Interobserver variability in echocardiography. J Clin Ultrasound. 65. Johnson T, Hahn D, Sandeste J. Quantitative analysis of left ventricular
1977;5(4):238–242. wall motion with MRI tagging. Radiologe. 2004;44:158–163.
46. Hundley WG, Hamilton CA, Rerkpattanapipat P. Magnetic resonance 66. Saito I, Watanabe S, Masuda Y. Detection of viable myocardium by
imaging assessment of cardiac function. Curr Cardiol Rep. dobutamine stress tagging magnetic resonance imaging with three-
2003;5(1):69–74. dimensional analysis by automatic trace method. Jpn Circ J.
47. Kaufman L, Crooks L, Sheldon P, Hricak H, Herfkens R, Bank W. The 2000;64(7):487–494.
potential impact of nuclear magnetic resonance imaging on 67. Baer FM, Voth E, Schneider CA, Theissen P, Schicha H, Sechtem U.
cardiovascular diagnosis. Circulation. 1983;67(2):251–257. Comparison of low-dose dobutamine-gradient-echo magnetic resonance
48. Shan K, Constantine G, Sivananthan M, Flamm SD. Role of cardiac imaging and positron emission tomography with [18F]
magnetic resonance imaging in the assessment of myocardial viability. fluorodeoxyglucose in patients with chronic coronary artery disease. A
Circulation. 2004;109(11):1328–1334. functional and morphological approach to the detection of residual
49. Darty SN, O’Neal J, Wesley-Farrington D, Davis AD, Link KM, Hundley myocardial viability. Circulation. 1995;91(4):1006–1015.
G. Cardiovascular magnetic resonance imaging. Prog Cardiovasc Nurs. 68. Sayad DE, Willett DL, Hundley WG, Grayburn PA, Peshock RM.
2004;19(2):60–67. Dobutamine magnetic resonance imaging with myocardial tagging
50. Vogel M, Stern H, Bauer R, Buhlmeyer K. Comparison of magnetic quantitatively predicts improvement in regional function after
resonance imaging with cross-sectional echocardiography in the revascularization. Am J Cardiol. 1998;82(9):1149–1151, A10.
assessment of left ventricular mass in children without heart disease and 69. Bogaert J, Maes A, Van de Werf F, et al. Functional recovery of
in aortic isthmic coarctation. Am J Cardiol. 1992;69(9):941–944. subepicardial myocardial tissue in transmural myocardial infarction
51. Kuijpers D, van Dijkman PR, Janssen CH, et al. Risk stratification with after successful reperfusion: an important contribution to the
dobutamine cardiovascular magnetic resonance in patients suspected of improvement of regional and global left ventricular function.
myocardial ischemia. Eur Radiol. 2004;14(11):2046–2052. Circulation. 1999;99(1):36–43.
52. Mazur W, Brucks S, Darty SN, Rerkpattanapipat P. Magnetic resonance 70. Geskin G, Kramer CM, Rogers WJ, et al. Quantitative assessment of
imaging in detection and functional assessment of coronary artery myocardial viability after infarction by dobutamine magnetic resonance
disease. Curr Atheroscler Rep. 2002;4(2):128–133. tagging. Circulation. 1998;98(3):217–223.
53. Cigarroa CG, deFilippi CR, Brickner ME, Alvarez LG, Wait MA, 71. Wellnhofer E, Olariu A, Klein C, et al. Magnetic resonance low-dose
Grayburn PA. Dobutamine stress echocardiography identifies hibernating dobutamine test is superior to SCAR quantification for the prediction of
myocardium and predicts recovery of left ventricular function after functional recovery. Circulation. 2004;109(18):2172–2174.
coronary revascularization. Circulation. 1993;88(2):430–436. 72. Rerkpattanapipat P, Morgan TM, Neagle CM, Link KM, Hamilton CA,
54. van Rugge FP, van der Wall EE, Bruschke AV. New developments in Hundley WG. Assessment of preoperative cardiac risk with magnetic
pharmacologic stress imaging. Am Heart J. 1992;124(2):468–485. resonance imaging. Am J Cardiol. 2002;90(4):416–419.
55. Pennell DJ, Underwood SR, Manzara CC, et al. Magnetic resonance 73. Dundon BK, Pisaniello AD, Nelson AJ, et al. Dobutamine stress cardiac
imaging during dobutamine stress in coronary artery disease. Am J MRI for assessment of coronary artery disease prior to kidney
Cardiol. 1992;70(1):34–40. transplantation. Am J Kidney Dis. 2015;65(5):808–809.
56. Baer FM, Theissen P, Smolarz K, et al. Dobutamine versus dipyridamole 74. Reddy ST, Thai NL, Fakhri AA, et al. Exploratory use of cardiovascular
magnetic resonance tomography: safety and sensitivity in the detection magnetic resonance imaging in liver transplantation: a one-stop shop
of coronary stenoses. Z Kardiol. 1993;82(8):494–503. for preoperative cardiohepatic evaluation. Transplantation. 2013;96(9):
57. Baer FM, Voth E, Theissen P, Schneider CA, Schicha H, Sechtem U. 827–833.
Coronary artery disease: findings with GRE MR imaging and Tc-99m- 75. Garot J, Bluemke DA, Osman NF, et al. Fast determination of regional
methoxyisobutyl-isonitrile SPECT during simultaneous dobutamine myocardial strain fields from tagged cardiac images using harmonic
stress. Radiology. 1994;193(1):203–209. phase MRI. Circulation. 2000;101(9):981–988.
58. van Rugge FP, van der Wall EE, Spanjersberg SJ, et al. Magnetic 76. Ryf S, Spiegel MA, Gerber M, Boesiger P. Myocardial tagging with
resonance imaging during dobutamine stress for detection and 3D-CSPAMM. J Magn Reson Imaging. 2002;16(3):320–325.
localization of coronary artery disease. Quantitative wall motion analysis 77. Pan L, Lima JA, Osman NF. Fast tracking of cardiac motion using
using a modification of the centerline method. Circulation. 1994;90(1): 3D-HARP. Inf Process Med Imaging. 2003;18:611–622.
127–138. 78. Mosher TJ, Smith MB. A DANTE tagging sequence for the evaluation
59. van Rugge FP, van der Wall EE, de Roos A, Bruschke AV. Dobutamine of translational sample motion. Magn Reson Med. 1990;15(2):
stress magnetic resonance imaging for detection of coronary artery 334–339.
disease. J Am Coll Cardiol. 1993;22(2):431–439. 79. Axel L, Dougherty L. Heart wall motion: improved method of spatial
60. Nagel E, Lehmkuhl HB, Bocksch W, et al. Noninvasive diagnosis of modulation of magnetization for MR imaging. Radiology.
ischemia-induced wall motion abnormalities with the use of high-dose 1989;172(2):349–350.
dobutamine stress MRI: comparison with dobutamine stress 80. Bolster BD Jr, McVeigh ER, Zerhouni EA. Myocardial tagging in polar
echocardiography. Circulation. 1999;99(6):763–770. coordinates with use of striped tags. Radiology. 1990;177(3):769–772.
CHAPTER 17  Stress Cardiovascular Magnetic Resonance: Wall Motion 225.e3

81. Paetsch I, Foll D, Kaluza A, et al. Magnetic resonance stress tagging in 97. Herzog CA, Marwick TH, Pheley AM, White CW, Rao VK, Dick CD.
ischemic heart disease. Am J Physiol Heart Circ Physiol. 2005;288(6): Dobutamine stress echocardiography for the detection of significant
H2708–H2714. coronary artery disease in renal transplant candidates. Am J Kidney Dis.
82. Osman NF, Kerwin WS, McVeigh ER, Prince JL. Cardiac motion 1999;33(6):1080–1090.
tracking using CINE harmonic phase (HARP) magnetic resonance 98. Macieira-Coelho E, Dionisio I, Garcia-Alves M, et al. Comparison
imaging. Magn Reson Med. 1999;42(6):1048–1060. between dobutamine echocardiography and thallium-201 scintigraphy
83. Osman NF, McVeigh ER, Prince JL. Imaging heart motion using in detecting residual stenosis, ischemia, and necrosis in patients with
harmonic phase MRI. IEEE Trans Med Imaging. 2000;19(3):186–202. prior myocardial infarction. Clin Cardiol. 1997;20(4):351–356.
84. Pan L, Prince JL, Lima JA, Osman NF. Fast tracking of cardiac motion 99. Klein C, Nagel E, Gebker R, et al. Magnetic resonance adenosine
using 3D-HARP. IEEE Trans Biomed Eng. 2005;52(8):1425–1435. perfusion imaging in patients after coronary artery bypass graft surgery.
85. Pan L, Stuber M, Kraitchman DL, Fritzges DL, Gilson WD, Osman NF. JACC Cardiovasc Imaging. 2009;2(4):437–445.
Real-time imaging of regional myocardial function using fast-SENC. 100. Smart SC, Knickelbine T, Malik F, Sagar KB. Dobutamine-atropine stress
Magn Reson Med. 2006;55(2):386–395. echocardiography for the detection of coronary artery disease in patients
86. Korosoglou G, Youssef AA, Bilchick KC, et al. Real-time fast strain- with left ventricular hypertrophy. Importance of chamber size and
encoded magnetic resonance imaging to evaluate regional myocardial systolic wall stress. Circulation. 2000;101(3):258–263.
function at 3.0 tesla: comparison to conventional tagging. J Magn Reson 101. Vasu S, Little WC, Morgan TM, et al. Mechanism of decreased sensitivity
Imaging. 2008;27(5):1012–1018. of dobutamine associated left ventricular wall motion analyses for
87. Korosoglou G, Lehrke S, Wochele A, et al. Strain-encoded CMR for the appreciating inducible ischemia in older adults. J Cardiovasc Magn
detection of inducible ischemia during intermediate stress. JACC Reson. 2015;17:26.
Cardiovasc Imaging. 2010;3(4):361–371. 102. Mirelis JG, Paetsch I, Jahnke C, et al. Impact of diastolic wall stress on
88. Korosoglou G, Futterer S, Humpert PM, et al. Strain-encoded cardiac the diagnostic value of visually interpreted dobutamine stress MR
MR during high-dose dobutamine stress testing: comparison to cine imaging. Int J Cardiol. 2014;172(1):e39–e42.
imaging and to myocardial tagging. J Magn Reson Imaging. 2009;29(5): 103. Lette J, Tatum JL, Fraser S, et al. Safety of dipyridamole testing in 73,806
1053–1061. patients: the Multicenter Dipyridamole Safety Study. J Nucl Cardiol.
89. Schuster A, Kutty S, Padiyath A, et al. Cardiovascular magnetic 1995;2(1):3–17.
resonance myocardial feature tracking detects quantitative wall motion 104. Pennell DJ, Underwood SR, Longmore DB. Detection of coronary artery
during dobutamine stress. J Cardiovasc Magn Reson. 2011;13:58. disease using MR imaging with dipyridamole infusion. J Comput Assist
90. Schuster A, Paul M, Bettencourt N, et al. Myocardial feature tracking Tomogr. 1990;14(2):167–170.
reduces observer-dependence in low-dose dobutamine stress 105. Pennell DJ, Underwood SR, Ell PJ, Swanton RH, Walker JM, Longmore
cardiovascular magnetic resonance. PLoS One. 2015;10(4):e0122858. DB. Dipyridamole magnetic resonance imaging: a comparison with
91. Schneeweis C, Qiu J, Schnackenburg B, et al. Value of additional strain thallium-201 emission tomography. Br Heart J. 1990;64(6):362–369.
analysis with feature tracking in dobutamine stress cardiovascular 106. Pingitore A, Lombardi M, Scattini B, et al. Head to head comparison
magnetic resonance for detecting coronary artery disease. J Cardiovasc between perfusion and function during accelerated high-dose
Magn Reson. 2014;16:72. dipyridamole magnetic resonance stress for the detection of coronary
92. Lubbers DD, Janssen CH, Kuijpers D, et al. The additional value of first artery disease. Am J Cardiol. 2008;101(1):8–14.
pass myocardial perfusion imaging during peak dose of dobutamine 107. Roest AA, Kunz P, Lamb HJ, Helbing WA, van der Wall EE, de Roos A.
stress cardiac MRI for the detection of myocardial ischemia. Int J Biventricular response to supine physical exercise in young adults
Cardiovasc Imaging. 2008;24(1):69–76. assessed with ultrafast magnetic resonance imaging. Am J Cardiol.
93. Falcao SN, Rochitte CE, Junior WM, et al. Incremental value of 2001;87(5):601–605.
perfusion over wall-motion abnormalities with the use of dobutamine- 108. Pedersen EM, Kozerke S, Ringgaard S, Scheidegger MB, Boesiger P.
atropine stress myocardial contrast echocardiography and magnetic Quantitative abdominal aortic flow measurements at controlled levels of
resonance imaging for detecting coronary artery disease. ergometer exercise. Magn Reson Imaging. 1999;17(4):489–494.
Echocardiography. 2013;30(1):45–54. 109. Rerkpattanapipat P, Gandhi SK, Darty SN, et al. Feasibility to detect
94. Gebker R, Jahnke C, Manka R, et al. Additional value of myocardial severe coronary artery stenoses with upright treadmill exercise magnetic
perfusion imaging during dobutamine stress magnetic resonance for the resonance imaging. Am J Cardiol. 2003;92(5):603–606.
assessment of coronary artery disease. Circ Cardiovasc Imaging. 110. Sukpraphrute BDB, Rerkpattanapipat P, Morgan TM, et al. Prognostic
2008;1(2):122–130. utility of cardiovascular magnetic resonance upright maximal treadmill
95. Gebker R, Mirelis JG, Jahnke C, et al. Influence of left ventricular exercise testing. J Cardiovasc Magn Reson. 2015;17:103.
hypertrophy and geometry on diagnostic accuracy of wall motion and 111. Raman SV, Dickerson JA, Mazur W, et al. Diagnostic performance of
perfusion magnetic resonance during dobutamine stress. Circ Cardiovasc treadmill exercise cardiac magnetic resonance: the prospective,
Imaging. 2010;3(5):507–514. multicenter Exercise CMR’s Accuracy for Cardiovascular Stress Testing
96. Hoffmann R, Lethen H, Marwick T, et al. Analysis of interinstitutional (EXACT) trial. J Am Heart Assoc. 2016;116.
observer agreement in interpretation of dobutamine stress
echocardiograms. J Am Coll Cardiol. 1996;27(2):330–336.
18 
Stress Cardiovascular Magnetic Resonance:
Clinical Myocardial Perfusion
Juerg Schwitter

Considerable progress in myocardial perfusion cardiovascular mag- exponential increase in the likelihood of a coronary occlusion associated
netic resonance (CMR) has been achieved recently and its diagnostic with a plaque rupture with an increasing degree of coronary artery
performance has been documented in single-center1–3 and multicenter stenosis. Accordingly, vulnerable plaques were defined by four major
trials.4–8 From single-center trials, and particularly from multicenter criteria34: (1) thin cap with large lipid core, (2) stenosis degree >90%,
trials, the knowledge of how to perform and interpret perfusion (3) endothelial denudation with superficial platelet aggregation, and
CMR studies has improved considerably. Also, convincing evidence (4) fissured plaque.
has been obtained regarding the robustness of the technique when In a large autopsy study evaluating the entire coronary tree of
performed at different sites,9 the importance of the reader experi- sudden cardiac death patients, active (vulnerable) plaques were found
ence for data analysis, and the excellent prognostic power of perfu- in ~60%, and in all patients severe stenoses (>75% diameter reduc-
sion CMR.10–16 Finally, an increasing number of studies are available tion) were observed. In another 20% healed postinfarct scares were
that now also address the economic consequences of introducing found (most likely causing malignant arrhythmias) and in another
this new technique for diagnosing and managing coronary artery 20% severe nonactive stenoses were present.35 Given these events in
disease (CAD).16–18 the development of CAD described earlier, a major goal of noninvasive
The CMR technique offers an almost unbeatable versatility for imaging is to identify vulnerable plaques in the coronary circulation that
exploring any aspect of cardiac disease. Therefore this chapter starts are at risk for occlusion in case of rupture: that is, severely stenosing
by illustrating how perfusion CMR is embedded in the concepts of plaques. Ideally, identification of these plaques would include determi-
atherosclerosis development and the vulnerable plaque theory. It then nation of both plaque mass (to some degree related to stenosis sever-
addresses established and not yet established aspects of the perfusion ity) and plaque composition. Although direct visualization of plaques
CMR protocol, including the different approaches proposed for data in the coronary circulation and assessment of plaque components is
interpretation and analysis. In this chapter, the current performance still in an early clinical phase, assessment of stenosis degree by means
of clinical perfusion CMR is presented to diagnose CAD and predict of perfusion CMR has been evaluated in large clinical single-center
outcome. A perspective for the future of perfusion CMR is provided and even multicenter trials, which convincingly demonstrated its high
at the end of the chapter. prognostic power to predict cardiac death and nonfatal myocardial
infarction.
THE RATIONALE FOR PERFUSION IMAGING THE PERFUSION CARDIOVASCULAR MAGNETIC
Endothelial dysfunction is generally recognized as a crucial initial step RESONANCE PROTOCOL
in the development of atherosclerosis.19–21 Among many alterations in
dysfunctional endothelium, the production of nitric oxide is reduced, Stress-Only Versus Stress-Rest Examination for the
which promotes the recruitment of leukocytes to the vessel intima22,23 Detection of Hemodynamically Significant Lesions
through enhanced expression of endothelial adhesion molecules.22 In In fundamental canine experiments, a reduction of coronary artery cross-
the vessel wall, mononuclear leukocytes become foam cells by lipid sectional area was related to resting and hyperemic flow in the vessel.36
accumulation and form reversible fatty streaks. In addition, recruitment To assess the hemodynamic consequences of a coronary artery stenosis,
of smooth muscle cells and their production of extracellular matrix the maximum hyperemic coronary blood flow during intracoronary
induce the formation of fibrous lesions. During the course of athero- adenosine infusion was measured. To compare maximum hyperemic
genesis, inflammatory triggers and other factors can affect the balance flow in different coronary vessels, hyperemic flow was normalized with
between the production of matrix-degrading enzymes24 and collagen resting flow to correct for differences in mass of the vessel-dependent
production by macrophages,25 which can cause fibrous cap destabiliza- myocardium. Thus the assessment of hemodynamic significance of a
tion. Subsequent repetitive plaque ruptures (and healing), even when coronary artery stenosis was achieved by dividing hyperemic blood flow
clinically silent, may cause a stepwise progression of atherosclerotic by resting blood flow: that is, by calculating the coronary flow reserve
lesions.26–28 A final rupture then of larger plaques can cause the ultimate (CFR). However, the influence of resting hemodynamics on resting
thrombus formation and acute vessel occlusion.29 In line with this flow, and thus on CFR, was recognized. By altering resting heart rate
concept, invasive studies conducted mainly in the preinterventional era and blood pressure, CFR ranged from 2 to 12 in their experiments.36 To
demonstrated a positive correlation between stenosis degree in coronary solve this problem, the relative CFR was proposed as hyperemic blood
angiography and risk for plaque rupture and acute myocardial infarc- flow with stenosis divided by hyperemic blood flow without stenosis.
tion.27,30–33 Fig. 18.1 demonstrates in approximately 17,000 patients an This relative CFR eliminates the influence of resting hemodynamics

226
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 227

30 Physical stress, dobutamine


Pooled data
n = 16,884
25 y = 0.17 e0.55x O2 demand ↑ Vasodilator

20 Vasodilation Vasodilation
Occlusion rate (%)

Stenosis

15 Hyperemia ↓ Hypoperfusion

10 O2 supply ≠ O2 demand
“Ischemia”

5
Dysfunction
FIG. 18.2  The two major options to provoke perfusion deficits in the
0 presence of a stenosis. Ischemia can be detected by wall motion analysis
0 10 20 30 40 50 60 70 80 90 100 as well as by perfusion analysis. A compromised hyperemic response
is detected by perfusion analysis but not by wall motion analysis unless
Diameter reduction (%)
the vasodilator induces ischemia by a steal effect. O2, Oxygen.
FIG. 18.1  The relationship between the degree of diameter stenosis
in invasive coronary angiography and occlusion rate per year in 4554
patients (light15,17 and dark12 brown bars) as well as the relationship
between pathologic/normal single-photon emission computed tomography measurements are affected by resting hemodynamics, two additional
(SPECT) scans (brown bars) and complication rate per year (deaths or problems should be kept in mind: (1) matching myocardial regions,
nonfatal myocardial infarctions) in 12,360 patients.156 For this bar graph,
for example, the subendocardial layer, for both rest and hyperemic
the finding of ischemia in SPECT scans was assigned to the stenosis
class of ≥50% diameter reduction, the normal scans to <50% diameter
conditions may be difficult because geometry of the heart is chang-
reduction. For the coronary angiography studies, occlusions at sites of ing with changing heart rate and loading, and (2) to obtain accurate
stenoses were considered. For the SPECT studies, the occlusions cannot results for the CFR calculation, the technique must guarantee a linear
be related to the location of stenoses; therefore the SPECT data are relationship between CMR-derived perfusion parameters and true flow
indirect support for the idea that low-grade and high-grade stenoses are over a wide range of flow conditions covering resting and hyperemic
associated with low and high risk for plaque rupture, respectively. Overall, flow. A synopsis on advantages and disadvantages on the stress-only
these compiled data from almost 17,000 patients27,31,33,156 demonstrate and stress-rest protocols is shown in Table 18.1.
a considerable (i.e., exponential) increase in risk of complications with
increasing stenosis severity. This graph also illustrates that the risk of Options for Inducing Stress in Cardiac
a plaque rupture is not zero in mild stenoses (in line with smaller ret-
rospective studies). Nevertheless, diagnostics in this low-risk subpopula-
Perfusion Studies
tion would require a very high “number to test” to identify patients with
From the considerations mentioned earlier, it appears mandatory to
significant stenosis: that is, patients at increased risk. apply some kind of stress to the myocardium to assess stenoses that
reduce maximum blood flow, but not resting flow. If oxygen demand
of the myocardium is increased either by positive inotropic agents such
as dobutamine or by direct physical stress, the supply-demand imbal-
on CFR because hyperemic stenosis flow is normalized by maximum ance induces a vasodilation and a hyperemic reaction in the myocardium
adenosine-induced hyperemic flow that is uncoupled from myocardial to meet demand (Fig. 18.2). However, in myocardial regions supplied
oxygen demand. Consequently, this relative CFR remained stable during by a stenosed coronary artery, increased oxygen demand is not met by
varying resting hemodynamics (standard deviation [SD] of 17% for supply, causing ischemia and consequently regional hypokinesia or
relative CFR vs. 45% for the absolute CFR). These findings are in line akinesia. In this latter situation, assessments of both myocardial (con-
with a recent CMR study in humans, demonstrating the important tractile) dysfunction and reduced hyperemic reaction allow detection
effect of resting hemodynamics such as heart rate, contractility, and of a hemodynamically significant stenosis. Alternatively, hyperemia can
loading conditions on resting myocardial blood flow and, hence, on be induced directly by administration of vasodilators such as dipyri-
absolute CFR.37 Further evidence that absolute CFR is affected by resting damole or adenosine. This strategy detects coronary artery stenoses by
hemodynamics is provided by positron emission tomography (PET) assessment of a compromised hyperemic flow, although ischemia (O2
studies on myocardial perfusion performed in patients with CAD. With supply-demand imbalance) is very rarely induced by vasodilators. To
this technique, a correlation of CFR with percent area stenosis was cause a steal effect by vasodilators, the stenosis has to be severe (i.e.,
demonstrated; however, there was a tendency toward even better cor- >90%) as demonstrated in experimental studies.42 Consequently, when
relations for hyperemic myocardial flow alone.38–41 These data suggest contractile function was assessed during vasodilator-induced stress, the
that relating hyperemic flow supplied by stenosed coronary arteries sensitivity for detection of CAD in clinical studies was very low.43 More-
to hyperemic flow supplied by nonstenosed coronary arteries (e.g., over, in comparison with positive inotropic approaches, the vasodilator
represented in a normal database) is advantageous because it elimi- approach as used for perfusion tests appears advantageous by reducing
nates the influence of resting hemodynamics as encountered with the ischemia-associated side effects such as angina pectoris and arrhythmias.
CFR approach. This stress-only approach, which compares hyperemic Adenosine causes hyperemia via A1 receptors, but it also induces nega-
stenosis flow with hyperemic flow of a normal database, has been suc- tive chronotropic, dromotropic, and inotropic effects and, in addition,
cessfully applied in several clinical CMR studies.1,4 Although the CFR it can cause bronchospasm, atrioventricular (AV) block, and mast cell
228 SECTION II  Ischemic Heart Disease

TABLE 18.1  Assessment of Coronary Artery Disease: Characteristics of Stress-Only Protocol


and Stress-Rest Protocol
Stress-Only Perfusion Protocol Stress-Rest Perfusion Protocol
Acquisitions Hyperemic data only Hyperemic and rest data waiting time between acquisitions
Oxygen demand–perfusion relation
• For resting perfusion Not applicable Determined by “confounding factors”
• Heart rate
• Contractility
• Loading condition
• For hyperemic perfusion Oxygen demand–supply uncoupled Oxygen demand–supply uncoupled
Analysis Only hyperemic data Two analyses (stress and rest) matching anatomy for rest/
stress condition “late enhancement effect” for second
acquisition
“Ideal” MR parameter–perfusion relation Linear for low perfusion range (no perfusion Linear for both resting and hyperemic perfusion, because
increase during hyperemia in myocardium CFR is calculated as hyperemic data/rest data
supplied by significantly stenosed vessel)
Normal/abnormal discrimination With normal database for regional hyperemic With normal database for regional CFR
perfusion
Viability assessment With late enhancement acquisition advantage: Low resting perfusion indicates scar disadvantage:
acquisition window: minutes acquisition window: seconds (first pass)
Assessment of perfusion and viability
• CMR: • Perfusion Hyperemic perfusion With resting perfusion yielding CFR
• Viability Late enhancement Resting perfusion
• SPECT: • Perfusion Hyperemic perfusion (tracer injection at stress) —
• Viability Rest-tracer redistribution (tracer injection at rest) —
• PET: • Perfusion Perfusion (flow tracer injection at stress) With resting perfusion yielding CFR
• Viability Rest FDG uptake (metabolic tracer injection at rest) —

“Stress” indicates hyperemic condition induced by adenosine or dipyridamole.


CFR, Coronary flow reserve; CMR, cardiovascular magnetic resonance; FDG, fluorodeoxyglucose; MR, magnetic resonance; PET, positron
emission tomography; SPECT, single-photon emission computed tomography.

degranulation. Despite this profile, it is a very safe stressor as was shown fiber orientations change during contraction and relaxation, making
in the EuroCMR registry with only two severe reactions in 18,840 this approach problematic in the beating heart.
patients.44 In clinical routine, an insufficient hemodynamic response In blood-oxygen-level-dependent (BOLD) imaging, deoxygenated
to adenosine can occur. Karamitsos and colleagues45 proposed a high- and thus paramagnetic hemoglobin shortens T2 relaxation time and
dose regimen for adenosine (by increasing the standard dose of 140 µg/ therefore can be used as an endogenous contrast media (CM) (although
kg/min for 3 minutes to 210 µg/kg/min for 7 minutes), which was safely oxygenated hemoglobin is slightly diamagnetic, causing less T2 shorten-
applied in patients with suspected CAD, increasing the proportion of ing). A T2- or T2*-weighted pulse sequence allows for an estimation
adequate hemodynamic response (heart rate increase >10 beats per of an increased content of oxygenated blood. During pharmacologically
minute or systolic blood pressure decrease >10 mm Hg) from 82% to induced hyperemia, oxygen content will increase in well-perfused myo-
98%. Conversely, dipyridamole induces hyperemia by blocking adenosine cardium but not in myocardium supplied by a stenosed vessel (which
re-uptake. Finally, regadenoson represents a more selective adenosine is associated with a higher O2 extraction). However, signal differences
2A receptor, which is administered as a single intravenous (IV) bolus. in normally perfused regions versus hyperemic regions (with a 4-fold
This compound was shown to induce hyperemia at a similar degree to increase in flow) were reported to be as low as 32% in an experimental
adenosine46 and it was also successfully used in studies on prognosis study.53 These BOLD data correlated closely with microsphere data, but
(see later).47,48 the slope of the correlation was as low as 0.08. This limitation in signal
difference might be problematic, because CM first-pass studies suggested
ENDOGENOUS VERSUS EXOGENOUS several hundred percent of signal change being required for a reliable
CONTRAST MEDIA stenosis detection.54 In another study applying BOLD, ΔR2 measure-
ments yielded an adequate slope of 0.94.55 But again, the sensitivity to
Endogenous Contrast Media absolute flow changes was low, because a 100% increase in flow yielded
Arterial spin labeling exploits the fact that unsaturated protons entering a signal increase of only 5% (i.e., a change in myocardial R2 of 0.94/s)
saturated tissue shorten the tissue T1.49–52 Using appropriate electro- in that study.55 Accordingly, in a recent study in patients, R2 increased
cardiogram (ECG)-triggered pulse sequences, T1 measurements are by only 16% during adenosine stress in nonstenotic areas (although
performed after global and slice-selective spin preparation. Absolute adenosine is typically increasing flow 300%–500%).56 The BOLD
tissue perfusion is then calculated by assuming a two-compartment approach is also sensitive to magnetic field inhomogeneities, predomi-
model. It should be kept in mind that this approach assumes that the nantly occurring in the posterior wall close the coronary sinus draining
direction of flowing blood in intramural vessels is orthogonal to the slice deoxygenated blood. Considering these aspects, the robustness of the
orientation, which is not the case for all myocardial layers. Furthermore, method is not yet fully explored.57,58
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 229

higher CM doses can cause susceptibility artifacts at the subendocar-


Exogenous Contrast Media for Perfusion Cardiovascular dium, where differences in CM concentrations between blood and
Magnetic Resonance myocardium are high during first pass. Strongly T1-weighted sequences
These CM are typically injected into a peripheral vein, and the signal showed an absence of a susceptibility-induced signal drop in the sub-
change in the myocardium occurring during the first-pass of the CM endocardial layer up to doses of 0.15 mmol/kg of an extravascular Gd
is measured by a fast CMR acquisition. All CM first-pass techniques chelate, which resulted in superior diagnostic performance of doses of
either under development or in clinical routine are designed to meet 0.1 and 0.15 mmol/kg versus 0.05 mmol/kg for semiautomatic analysis
the following requirements: (1) provide high spatial resolution to permit of the upslope parameter4 (Fig. 18.3).
detection of small subendocardial perfusion deficits, (2) provide adequate
cardiac coverage to allow for assessment of the extent of perfusion Intravascular Contrast Media
deficits, (3) feature high CM sensitivity to generate optimum contrast For albumin-targeted MS-32565 and polylysine-Gd compounds66,67 dif-
between normally and abnormally perfused myocardium during CM ferences between normally perfused and stenosis-dependent hypoperfused
first pass, and (4) allow acquisition of perfusion data every one to two myocardium were reported in animals. However, these intravascular
heartbeats to yield signal intensity–time curves of adequate temporal Gd-based CM have not advanced into clinics. Ultrasmall superpara-
resolution that allow for extraction of various perfusion parameters magnetic particles of iron oxide (USPIO) nanoparticles were used for
(see later). To reach these goals, high-speed data acquisition and time- perfusion studies in humans (however, this CM is no longer under
efficient magnetization schemes are most important. Because the first investigation because its iron accumulation in the liver).68 For T2*-
pass of CM during hyperemia lasts only 5 to 10 seconds, breathing enhancing CM, not only the concentration of CM in the voxel but also
motion is minimized by a breath-hold maneuver, although cardiac its intravoxel distribution (homogeneous vs. inhomogeneous) determines
motion is eliminated by ECG triggering. This control of cardiac and its T2*-shortening effect, rendering such a T2*-approach susceptible
breathing motion in perfusion CMR preserves its high spatial resolution to vessel architecture (vessel orientation, intervessel distance), which
of data acquisition: on the order of 1 to 2 mm × 1 to 2 mm. This is also may compromise quantitative approaches.
not the case for scintigraphic techniques with acquisition windows of
several minutes, which preclude breath-holding for elimination of Hyperpolarized Contrast Media
respiratory motion, and ECG triggering during single-photon emission Conventional Gd chelates modulate the magnetic resonance (MR) signal
computed tomography (SPECT) studies requires higher tracer amounts by accelerating the relaxation rates of surrounding water protons.
to improve counts statistics. However, at a field strength of 1.5 T, the polarization level of the spin
population of the MR active nuclei at the thermal equilibrium is low
Extravascular Contrast Media (several parts per million), and only the vast abundance of water mol-
For perfusion CMR techniques, the relationship between myocardial ecules in the human body enables generation of a measurable signal.
CM concentration and myocardial signal depends on a variety of factors. Because the polarization level of water protons increases with magnetic
Normal perfusion can cause a signal increase during first pass of a flux density, a higher signal is achieved at higher field strength. Alter-
gadolinium (Gd) chelate when combined with a T1-weighted pulse natively, however, the polarization level of the spin population of specific
sequence, although a T2-weighted sequence with a higher dose of a Gd nuclei (such as liquid 13C in various compounds) could be increased
chelate can even cause a signal drop during first pass.59,60 This is fun- by a factor of up to 100,000 (compared with polarization of water
damentally different from ischemia detection based on the assessment protons at the thermal equilibrium) by dynamic nuclear polarization69
of wall motion, where new onset of dysfunction unambiguously indicates or parahydrogen-induced polarization.70 With these hyperpolarized CM,
the presence of ischemia.61 signal is received only from the 13C-nuclei; therefore no signal from
Today, extravascular Gd chelates are most commonly used for first- background tissue is obtained. This feature appears ideal for absolute
pass perfusion CMR in combination with heavily T1-weighted pulse quantification of perfusion because signal is directly proportional to
sequences. These CM are excluded from the intracellular compartment the amount of 13C molecules similar to radioactive tracers, where radio-
(i.e., from viable cells with intact cell membranes); therefore a perfu- activity is directly proportional to tracer concentration. Similarly to
sion deficit during the first pass reflects either hypoperfused viable radioactive tracers, the signal from hyperpolarized 13C-nuclei also decays
myocardium (which would become ischemic during inotropic stress) (with specific time constants depending on the type of the 13C-compound),
and/or scar tissue (with severe reduction of perfusion even at rest). To because the spin population of the 13C-molecules is far away from
differentiate hypoperfused tissue further, it is recommended to inject thermal equilibrium. In addition, repetitive radiofrequency pulsing
another dose of CM and to wait for the establishment of a dynamic further destroys longitudinal magnetization depending on the pulse
equilibrium of CM concentrations in the various compartments (blood, sequence type and the imaging parameters. Johansson and colleagues
viable myocardium, scar tissue), to obtain conditions where tissue CM showed that depolarization can be approximated by a monoexponential
concentrations are governed by distribution volumes (and no longer function with a time constant TD and successfully applied this concept
by perfusion).62 During this condition, which typically occurs within for cerebral perfusion quantification.71 Recently, sophisticated spiral
10 to 20 minutes after CM injection in humans, late enhancement pulse sequences became available, which allowed for myocardial perfu-
imaging (with the inversion time set to null normal myocardium) is sion and metabolism quantification in small animals72 and for the first
ideal to discriminate hypoperfused but viable myocardium as dark tissue time in humans.73
from scar, which appears bright.63,64 For viability assessment, scinti-
graphic techniques also exploit the equilibrium distribution of tracers, PERFUSION CARDIOVASCULAR MAGNETIC
which is observed after rest injection or rest reinjections. However, the RESONANCE: WHAT IS ESTABLISHED AND
radioactive tracers are not taken up by scar tissue, which consequently WHAT IS NOT
appears as a cold spot, whereas viable tissue appears as a hot spot
(see Table 18.1). Cardiovascular Magnetic Resonance Data Readout
Several multicenter studies were performed for the assessment of Echo planar readout strategies are well suited to speed up data acqui-
the optimal CM dose for perfusion imaging. This is important because sition,74,75 particularly when segmented to reduce the echo time and
230 SECTION II  Ischemic Heart Disease

Subendocardial stress data ROC analysis


400
1

300
0.75
SI change (%)

Sensitivity
200 0.50
<0.002
<0.05

100 D1: 0.56 + 0.12


0.25
D3: 0.86 + 0.08
0.15 mmol/kg
0.10 mmol/kg P < .05 D1 vs. D3
0.05 mmol/kg
0 0
0 10 20 0 0.25 0.5 0.75 1
A Time (image #) B 1-specificity
FIG. 18.3  (A) Increasing doses of the extravascular contrast medium gadolinium diethylenetriamine penta-
acetic acid from 0.05 to 0.10 and 0.15 mmol/kg administered intravenously induce an increasing myocardial
signal in the subendocardium (inner half of myocardial wall) during first pass.4 (B) Even a dose as high as
0.15 mmol/kg did not cause measurable susceptibility-induced signal loss in the subendocardial layer using
a hybrid echo planar pulse sequence (echo time: 1.3–2.2 ms). Accordingly, the highest dose (D3) detected
stenoses with ≥50% diameter reduction significantly better than the 0.05 mmol/kg dose (D1). ROC, Receiver
operator characteristics; SI, signal intensity.

consequently to render the pulse sequence less prone to susceptibility (RF) deposition homogeneity, a dual-source parallel RF transmission
artifacts (hybrid echo planar pulse sequences).1,54,65,76,77 With these accel- approach was applied. To allow for 3D undersampling with acquisi-
erated pulse sequences, several k-lines are acquired following a single tion windows of 191 ms and 211 ms for FGE and SSFP, respectively,
radiofrequency excitation, reducing the repetition time (TR) per k-line k-t (k-space) BLAST (broad linear speed-up technique) and k-t PCA
down to less than 2 ms and consequently reducing the total acquisition (principal component analysis) reconstructions were used. In 15 vol-
window per slice substantially. This enables the acquisition of a stack of unteers, overall image quality was good, but more susceptibility, that
slices every one to two heartbeats, allowing for multislice acquisitions. is, dark-rim artifacts, were observed in the SSFP acquisitions compared
Conventional nonsegmented fast gradient echo (FGE; or Turbo fast low with FGE.89 Currently, SSFP-based techniques are not recommended
angle shot [TurboFLASH]) pulse sequences were also used for perfusion for clinical use because of a relatively high level of artifacts compared
imaging, however, resulting in relatively long readout windows.61,78–84 with (hybrid) FGE readouts.
Alternatively, FGE can be combined with parallel imaging techniques85
or strategies that exploit the spatiotemporal correlations of image data to Magnetization Preparation
considerably speed up the data acquisition.86 Improving cardiac coverage Although 180-degree preparation was used in the past,61,78–84,90 a 90-degree
without the need to reduce spatial and/or temporal resolution would magnetization preparation is now generally accepted as the most efficient
be beneficial because the extent of CAD correlates with outcome. In way to achieve T1 weighting because it shortens the delay time to 100
a recent study in human volunteers, the loss in signal-to-noise ratio to 150 ms1,54 and, additionally, it renders the sequence heart rate inde-
(SNR) given by g * √R (with g being the so-called geometry factor and pendent. Partial preparation flip angles (e.g., of 45–60 degrees) were
R the accelerating factor) was compensated for by a longer TR (due to suggested, which allow for shorter delay times.65,76 However, this limited
the implementation of temporal sensitivity encoding, a modification of the dynamic range of signal response54 and it is no longer recommended
sensitivity encoding [SENSE]85) and increasing the readout flip angle (Fig. 18.4).
from 20 to 30 degrees. This accelerated hybrid echo planar saturation The relationship between CM concentration and MR signal is
recovery technique yielded twice as many slices as that obtained with the not only dependent on the flip angle of the preparation pulse but
nonparallel approach whereas SNR improved by approximately 20%.87 also on the readout flip angle, as shown in Fig. 18.4. To shorten the
Readout strategies during steady-state conditions of magnetiza- delay time, it is also important to place the readout of the data into
tion (steady-state free precession [SSFP] sequences) appear promising phases of minimal cardiac motion, into late systole and mid-diastole,
because they preserve magnetization and, thus a high SNR. In a human which is achieved with a delay time of about 120 ms. Because length
volunteer study, however, banding artifacts (probably because of longer of systole varies relatively little with a changing heart rate (as occurs
readout periods or off-resonance) were problematic and myocardial during pharmacologically induced hyperemia), these delay times are
signal increase was rather low (approximately 40% of baseline signal).88 robust to place the data collection into stable heart phases irrespective
Recently, Jogiya et al.89 compared an FGE with a SSFP readout in a three- of heart rate. To further accelerate data collection, it was proposed
dimensional (3D) acquisition at 3 T. To optimize the radiofrequency to play out one single nonslice-selective 90-degree saturation pulse,
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 231

Effect of imaging parameters: Perfusion data


αprep, delay time, αread-out
16
Visual analysis Quantitative analysis

13
Perfusion-related parameters Absolute perfusion
e.g., upslope, peak SI, etc. unit: mL/min/g
10
SNR

Manual Semi- Automatic Manual Semi- Automatic


automatic automatic
7
FIG. 18.5  A scheme for better definition of possible analysis strategies.
In this scheme, “quantitative” results are given in numbers, which allow
4 for an objective comparison of studies both longitudinally (e.g., moni-
toring disease activity) as well as from patient to patient (e.g., patient
data vs. normal database). Such quantitative results are obtained either
1 manually (and thus are associated with some observer dependence), in
0 0.2 0.4 0.6 0.8 1.0 a semiautomatic fashion—that is, with some observer interaction—or
automatically, thus eliminating any observer interaction with the data and
Gd-DTPA concentration (mmol/L) thereby completely eliminating analysis variability. SI, Signal intensity.

90°/50°/200 ms
90°/50°/150 ms—4 slices be required to demonstrate a potential advantage of perfusion imaging
90°/50°/100 ms at 3 T. In a recent study, a novel spatiotemporal correlation approach
90°/50°/50 ms was implemented (5× kt-SENSE) on a 1.5 T and a 3 T machine.93 In
90°/30°/150 ms patients with suspected CAD, the 1.5 T and 3 T machines yielded spatial
90°/10°/150 ms resolutions of 1.5 × 1.5 mm2 and 1.3 × 1.3 mm2 (with otherwise identi-
90°/10°/10 ms cal temporal resolution and coverage). The areas under the receiver
60°/10°/10 ms—6 slices operator characteristics (ROC) are under the curve (AUC) for CAD
FIG. 18.4  These phantom measurements demonstrate that optimization detection (defined as >50% diameter reduction on coronary angiog-
of a hybrid echo planar pulse sequence76 with respect to the delay time raphy) were not different with 0.80 and 0.89, respectively (P = .21),
between magnetization preparation and readout, readout flip angle, and despite slightly higher SNR on 3 T, indicating that the high diagnostic
preparation flip angle can improve signal response of gadolinium diethy- performance of perfusion CMR at 1.5 T is hard to improve.93 Accord-
lenetriamine penta-acetic acid (Gd-DTPA)-doped phantoms from 80% ingly, in the EuroCMR registry, out of 27,781 examinations, only 5.9%
(of nondoped phantoms) to 250%, which ultimately increased diagnostic were performed on 3 T machines, whereas 29.3% of all examinations
performance in patients.54 SNR, Signal-to-noise ratio. were adenosine stress studies.9

ANALYSIS OF PERFUSION DATA


and to acquire the entire stack of slices thereafter.2 With this scheme,
however, the delay time varies from slice to slice, and consequently, With increasing experience, an observer will be able to visually dif-
CM sensitivity becomes dependent upon slice position and acquisition ferentiate hypoperfused regions from artifacts, both typically causing
order, which is likely to affect data analysis. In a modified version of lower signal during first-pass conditions.5,84,90,94 However, the advantage
a saturation recovery preparation, the entire myocardium is prepared of experience is traded for subjectivity, that is, reduced interreader
by a 90-degree saturation pulse except the slice which is immediately reproducibility. Therefore a computer-assisted or automatic analysis of
imaged after preparation (“notch technique”).91 With this scheme, the perfusion data would be highly desirable and could potentially render
readout time for slicen equals the preparation time for the next slice, CMR perfusion analyses fully reproducible. Because many different
that is, slicen+1, and so on. This approach allows acquisitions of up to 7 analysis procedures are currently in use, some common definitions for
slices every 2 heartbeats (at a heart rate up to 115 beats per minute). analysis characteristics are proposed in Fig. 18.5.
Currently, identical saturation recovery preparation of each acquired
slice is recommended to allow for consistent signal response and easy Visual Assessment
data analysis. Although visual reading can provide accurate diagnoses, adequate repro-
ducibility should not rely on individual experience but on standard-
Field Strength: 1.5 T Versus 3 T ized criteria that in particular would differentiate signal loss due to
Systematic data comparing the diagnostic performance of perfusion hypoperfusion from artifacts. This indicates that reading criteria should
CMR at 1.5 T and 3 T are still sparse. Increasing the field strength from also assess image quality, which in turn depends on the pulse sequence,
1.5 T to 3 T increases the available magnetization, as was demonstrated the CM type and dose, and, importantly, the experience and care in data
in a study in volunteers, in which myocardial signal during the first acquisition. Signal loss in the subendocardium, which is most sensitive
pass (at 0.1 mmol/kg Gd-diethylenetriamine penta-acetic acid [DTPA] for perfusion abnormalities, can be caused by susceptibility artifacts
IV) was 2.6 times higher at 3 T than at 1.5 T (relative to baseline signal).92 occurring during the time window when CM concentration differs
However, this increase was observed in the anterior wall, whereas signal most between the blood pool and the myocardial tissue. Thus long
increased by only 1.7 times in the posterior wall. This inhomogeneous persistence of signal loss during first pass is suggestive of the presence
signal increase might be disadvantageous, and larger clinical trials would of true hypoperfusion in the subendocardium. However, these timing
232 SECTION II  Ischemic Heart Disease

criteria also depend on the dose of CM injected, the injection rate and software upgrade). On the basis of such a normal database, perfusion
site, and the dispersion of the bolus within the pulmonary circulation. studies can be compared between patients, and even more impor-
Currently, CM doses of 0.075 to 0.10 mmol/kg of a Gd chelate (with tantly, the perfusion situation; that is, the activity or progression of
injection rates of 4–8 mL/s intravenously [IV]) are recommended and CAD can be monitored by serial perfusion CMR studies in the same
typically used in clinical routine. With such a setting, a reduced signal patient. To allow for better comparisons between various analysis strat-
increase in the subendocardium lasting for ≥3 frames (i.e., during egies, it appears reasonable to clarify some confusion that may exist
≥3–6 heartbeats) during hyperemic first pass is highly suggestive for with expressions such as semiquantitative and semiautomatic. In Fig.
ischemia as established in trials on diagnostic performance.3,4,6–8 Most 18.5 quantitative relates to the analysis output obtained as numbers
trials on outcome10–15,47,48,95,96,153 use this or similar criteria demonstrat- (which may be related to perfusion or may represent perfusion itself).
ing an excellent prognostic power (for more details, see also Table 18.4 These aspects should be separated from the issue of how the data are
later). A signal reduction consistent with a territory supplied by an extracted from the imaging set, which may occur manually (with con-
individual coronary artery is also supporting the diagnosis of CAD, siderable observer interaction with the data and corresponding time
whereas artifacts predominantly distribute along the phase direction requirements), semiautomatically (with some observer interaction), or
(because spatial resolution is often reduced along phase) or may occur in fully automatically. A fully automatic approach is time saving (no labor
regions with inhomogeneous magnetic field characteristics as observed needed for analysis) and eliminates any observer variability, although
in the inferior wall (where air is extending between diaphragm and it is anticipated that high-quality data would be required for an auto-
inferior wall of the left ventricle).97 Criteria for quality assessment of matic approach. A quantitative approach is also ideal to generate ROC,
perfusion data were derived from multicenter studies4,5 and validated which are the adequate means for assessment of test performance.
for interobserver reproducibility in multicenter registry data,98 as given Once a ROC curve has been determined (for a given acquisition
in Table 18.2. and analysis protocol), the optimum cost-effectiveness of the pro-
tocol can be calculated (optimum point on the ROC), because cost-
Quantitative Approach effectiveness changes with changing portions of false and true negatives
Dedicated algorithms for perfusion data analysis would allow compari- and positives.
son of signal responses in an individual patient with a normal database,
rendering the technique less observer dependent. Because myocar- Quantitative Approach: Perfusion-Related Parameters
dial signal response is strongly dependent on imaging parameters, a First-pass perfusion studies are typically performed during breath-
normal database should be updated in case the pulse sequence and/ holding, and therefore any type of quantitative analysis should start with
or the imaging parameters would be modified (e.g., by a hardware or a registration of the first-pass data over time: that is, motion in the data

TABLE 18.2  Perfusion Cardiovascular Magnetic Resonance: Acquisition—Data


Quality—Reading/Interpretation
Acquisition Quality Reading
• Saturation recovery preparation • Severe artifacts (3 points each) The perfusion deficit is:
• Myocardial SI increase during FP • Abrupt breathing motion during first pass • clearly visible (minimal SI increase during
>250%–300% of precontrast baseline SI • >2 mistriggers and/or ES during FP or AF FP) during ≥3 phases (i.e., ≥3 heartbeats
• CM dose: (0.075)—0.10 mmol/kg IV • Wrap-around artifacts, ghosts, blurring, and and ≥6 heartbeats, respectively, in 1–2 RR
• Acquisition window (readout): ≤150 ms metallic artifacts in ≥3 slices acquisitions)
in mid-late systole and/or mid-diastole • Intermediate artifacts (2 points) • extends to ≥50% of wall thickness
• Spatial resolution: ≤1.5–2 × ≤1.5–2 mm • Wrap-around artifacts, ghosts, blurring, and • encompasses ≥50% of segment
• Slice thickness: ≤10 mm metallic artifacts in 2 slices • matches a coronary artery territory (except
• Acquisition of ≥3 slices every 1–2 RR • Minor artifacts (1 point each) CABG patients)
• No breathing during FP • Respiratory drift (small respiratory excursions) • resides within viable tissue (LGE-negative
• Reliable ECG triggering during FP tissue)
• Adequate vasodilation • 1–2 mistriggers and/or ES during FP
• Wrap-around artifacts, ghosts, blurring, and
metallic artifacts in 1 slice

Comments
The recommended acquisition parameters These quality criteria were derived from multicenter Visual reading criteria are derived from large
were derived from single-center3 and studies4,5 and were validated with multicenter registry single-center1,153 and multicenter trials4,6–8
multicenter trials,4,6–8 which confirmed data.98 Criteria are modified from Klinke et al.98 and most studies on the prognostic power
their high diagnostic performance The maximum score is 9, which would indicate severe of perfusion CMR (as given in Table 18.4)
artifacts in 3 slices or more. A score <4 indicates use these or similar reading criteria.
good quality, scores 4–6 indicate intermediate quality Perfusion deficits are typically assessed in
(and AUC for CAD detection decreases by ~10% vs. the 17-segment AHA model (not including
good quality), scores >6 indicate inadequate quality the LV apex)157

AF, Atrial fibrillation; AHA, American heart Association; AUC, area under the curve; CABG, coronary artery bypass grafting; CAD, coronary artery
disease; CMR, cardiovascular magnetic resonance; ECG, electrocardiogram; ES, extra-systoles; FP, first pass; LGE, late gadolinium
enhancement; LV, left ventricular; SI, signal intensity.
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 233

caused by breathing and/or diaphragmatic drift should be eliminated for differences in arterial input by dividing myocardial upslope data by
either by a manual procedure or by (semi-)automatic algorithms.99,100 the blood-pool signal upslope.1,54,82,83,103,104 This approach is suboptimal
To improve SNR, cardiac perfusion studies are performed with for input correction because the upslope–CM concentration relation-
phased-array coils. Therefore analyses of signal intensity–time curves ship is flat at higher CM concentrations, which occur in the blood pool
have to correct for inhomogeneous coil sensitivities. For this purpose, during first pass and which are even higher during hyperemic condition.
it has been suggested to subtract precontrast signal from the first-pass An experimental study demonstrated linearity between the upslope
signal intensities.2 However, signal reception by a phased array coil does parameter and perfusion measured by microspheres for low perfusion
not cause a constant offset of signal over the field of view; therefore values only (below approximately 1.5 mL/min/g) (Fig. 18.6A).108 A
a division of first-pass signal by precontrast signal for correction is similar limitation for upslope estimates was described for humans using
required.1,4 PET perfusion measurements as the standard of reference (Fig. 18.6B).1
From the resulting signal intensity–time curves calculated for various
transmural or subendocardial segments covering ideally the entire left Quantitative Approach: Absolute Tissue Perfusion
ventricular myocardium, many parameters can be extracted. The appli- For blood-pool CM, which do not mix homogeneously in the tissue
cability was demonstrated for the peak signal intensity,61,65,78,101 signal compartment but are restricted to the intravascular compartment, the
change over time (upslope),1,54,81–83,101–104 arrival time, time to peak signal, so-called bolus tracking approach can be applied, which is based on
mean transit time,101,102,105,106 area under the signal intensity–time curve,107 the central volume principle given by
and others. For the upslope of the signal intensity–time curve, a relatively
close correlation with perfusion data is reported in both animal102,108 VB = F × MTT Eq. 18.1
and human studies,1 at least for the low-flow range. Its robustness could
also be demonstrated in a multicenter trial.4 This upslope parameter where MTT is the mean transit time (expected distribution of transit
is relatively insensitive to CM recirculation because it uses the initial times for the blood through the tissue volume) and VB is blood volume.
portion of the signal intensity–time curve only, which also reduces its Because the conventional extravascular Gd chelates do not cross the
sensitivity for motion (most patients are able hold their breath for this intact blood-brain barrier, this model is generally used for cerebral
short time period). The upslope parameter is also proposed to correct perfusion measurements. Also, hyperpolarized 13C-CM (see later) can

2.5
6 2.25
Slopemeasured/slopethreshold

2
Myocardial/LV upslope ratio

5
1.75
4
(normalized)

1.5
3 1.25
1
2
0.75 y = 1.122 log (x) + 1.124
1 r = 0.69
0.5
P < .0001
0 0.25
0 1 2 3 4 5 6 0 0.5 1 1.5 2 2.5 3 3.5 4 4.5
Microsphere blood flow (mL/min/g) MBFPET (mL/min/g)

A B Stenosis: 50%–69%
Stenosis: 70%–99%
Occlusion
Volunteers (mean + SD)
Patients without CAD
Remote region of CAD patients
(supplied by nonstenosed vessel)
FIG. 18.6  (A) The nonlinear relationship between the myocardial upslope parameter (“corrected” by the
signal upslope in the left ventricular [LV] blood pool) and the microsphere blood-flow measurements in canine
experiments.108 Linearity exists for low-flow situations only (up to approximately 1.5 mL/min/g). (B) Similar
nonlinearity between the myocardial upslope parameter (normalized by the threshold upslope value) and the
myocardial blood flow measured by positron emission tomography (PET) in patients with and without coronary
artery stenoses. Normalization of the myocardial upslope by the threshold upslope value (which discriminates
between stenotic and nonstenotic flow) results in a ratio of ≥1 for flow in nonstenosed vessels.1 CAD, Coro-
nary artery disease; MBF, myocardial blood flow; SD, standard deviation. (Reproduced with permission from
Christian TF, Rettmann DW, Aletras AH, et al. Absolute myocardial perfusion in canines measured by using
dual-bolus first-pass MR imaging. Radiology. 2004;232:677–684; Schwitter J, Nanz D, Kneifel S, et al. Assess-
ment of myocardial perfusion in coronary artery disease by magnetic resonance: a comparison with positron
emission tomography and coronary angiography. Circulation. 2001;103:2230–2235.)
234 SECTION II  Ischemic Heart Disease

act as an intravascular tracer71; hence, this model could be applied for compartments must be taken into account, because conventional Gd
cardiac studies using these CM. chelates exert their effect indirectly through water proton relaxation,
In the myocardial tissue, however, the conventional Gd chelates which modifies the MR signal during first pass.120–123 An extravascular
behave as extravascular CM. Therefore the CM first-pass techniques CM mixed homogeneously in the extravascular space by diffusion would
with residue detection will be discussed next. It is assumed that CM generate a maximum signal during first pass of 20% of fully relaxed
diluted in blood enters the tissue via the artery, transits the capillaries, magnetization (assuming an extracellular compartment of 20% and the
and leaves the tissue through the venous system. Then the Fick principle absence of water exchange between compartments). For an intravascular
relates the CM concentration in the tissue to the arterial input and CM, the maximum achievable signal during first pass would further
venous outflow by a convolution integral. This model takes both CM decline to approximately 10% of fully relaxed magnetization (assuming
inflow and outflow into account and assumes a freely diffusible CM a blood volume in tissue of 10% and no water exchange between com-
that is homogeneously mixed within a single tissue compartment.109 It partments). In rat experiments, however, extravascular and intravascular
is given by CM yielded approximately 70% and 50%, respectively, of fully relaxed
magnetization during first pass, indicating that water exchange across
t
both capillary vessel wall and cell membranes strongly affects signal.120
CT (t ) = F ⋅ ∫ C A ⋅ (τ ) ⋅ e −F λ (t − τ )dτ
= C A (t ) ⊗ F ⋅ e −F λ ⋅t
Eq. 18.2
Water exchange conditions can be categorized into fast, intermediate,
0
and slow. If tissue 1/T1 (= r1) increases linearly with intravascular r1
where CT is the tissue CM concentration at time t, CA is the CM con- and hence myocardial signal response in the presence of a CM would
centration in blood, F is tissue perfusion (in milliliters per minute be unaffected by water exchange, the water exchange regimen is called
per gram), λ is the tissue-blood partition coefficient, and ⊗ denotes fast. Studies by Larsson and colleagues124 and others121–123 demonstrated
convolution. If CT and CA are known, a two-parameter fit yields F and that intravascular–extravascular water exchange is the rate-limiting step
λ (by application of a three-parameter fit, blood volume VB can be for signal behavior in tissue (slow exchange regime). Similar to radioac-
incorporated into the model as well). Several deconvolution procedures tive tracers, E of extravascular CM varied with increasing myocardial
have been used to obtain F.108,110–113 For nonfreely diffusible CM, the perfusion.116,125,126 By assuming rather small changes in E for resting
extraction fraction E must also be taken into account,114–116 where E and hyperemic conditions and therefore using E as a constant, Ki at rest
is related to F by and during hyperemia was calculated, from which a perfusion index
or CFR was derived.127,128 In asymptomatic patients, myocardial per-
K i = E × F = F (1− e −PS F ) Eq. 18.3 fusion was quantitatively assessed by first-pass perfusion CMR and a
relation was found for hyperemic perfusion impairment and reduced
where PS is the permeability-surface-area product (in milliliters per segmental contractile function at rest, demonstrating the feasibility to
gram per second) and Ki is the unidirectional influx constant (in mil- quantify myocardial perfusion by first-pass techniques; however, no
liliters per gram per second). For all these models, knowledge about attempt was undertaken to use these perfusion data to detect CAD in
the arterial input function (i.e., the measurement of the arterial CM this population.129 In a subgroup of the CE-MARC (Clinical Evaluation
concentration over time) is mandatory. Consequently, the signal intensity– of Magnetic Resonance Imaging in Coronary heart disease) popula-
CM concentration relationship over the full range of CM concentrations tion, myocardial first-pass perfusion data were subject to four different
occurring in the blood pool during first-pass conditions must be models to quantitate stress mean blood flow and myocardial perfusion
known.66,111,117,118 Although higher CM doses yield an appropriate CNR reserve and to evaluate the potential to detect CAD.130 In a cohort of 50
level for the signal response in the myocardium, such high doses are patients, CAD was defined as an ischemia-positive SPECT result and
likely to cause a clipping of the signal intensity–time curve in blood a ≥70% diameter stenosis in quantitative coronary angiography, and
from which a conversion of signal intensities into CM concentration absence of CAD was defined as an ischemia-negative SPECT and no
is problematic. To solve this problem, a dual-bolus approach was pre- diameter stenosis >50% in quantitative coronary angiography. Thus
sented,108 in which a small CM bolus is injected first for determination patients with severe ischemia were compared with normals, whereas
of an arterial input function, followed by a larger CM bolus to achieve “intermediate” ischemia patients were excluded from the study. In this
an adequate signal response in the myocardium. In dogs, this approach highly preselected patient population, quantitative stress mean blood flow
yielded absolute values of tissue perfusion in close agreement with yielded high sensitivities and specificities to discriminate the ischemic
microsphere measurements. Alternatively, Gatehouse and colleagues119 from nonischemic patients and the four methods (Fermi model, uptake
proposed a dual T1 sensitivity method. This elegant approach uses a model, one-compartment model, and model-independent deconvolu-
single high-dose CM bolus injection, which provides a high signal tion) yielded no difference in performance with AUCs of 0.86, 0.85,
response in the myocardium although preventing clipping of the blood 0.85, and 0.87, respectively. To our knowledge, no larger clinical studies
pool signal at peak effect of the bolus. To measure blood signal (with are available that tested a quantitative approach in a suspected CAD
low T1 sensitivity for very short T1), a short saturation recovery time population at intermediate risk and, therefore, a quantitative approach
is applied, although it is longer for measurement of myocardial signal cannot (yet) be recommended for clinical application.
(high T1 sensitivity for longer myocardial T1). Furthermore, the low Besides precision, the variability of parameter estimates is another
T1-sensitive blood-pool measurements are performed at a low spatial important aspect of perfusion measurements. On the basis of simula-
resolution to accelerate acquisition. With this dual T1 sensitivity single- tions for the determination of regional blood volume, intravascular
bolus approach, CFR estimates in volunteers closely matched those CM131,132 were less sensitive for noise than were extravascular CM.114
from dual bolus measurements. Although this dual T1 sensitivity single- Despite a large body of computer simulations114,131 and experimental
bolus technique is available as work-in-progress packages for clinical data,66,111 the clinical value of absolute quantitative measures of perfu-
application, to our knowledge, no larger clinical studies evaluated its sion for the detection of CAD in patients remains unproven. A 95%
performance for CAD detection in comparison to nonquantitative confidence interval (CI) of −45% to −82% to +45% to +82% for absolute
“visual” approaches. perfusion quantification66,110 suggests that the sensitivity of these tech-
Once a reliable arterial input function has been obtained, diffu- niques to detect changes in perfusion might be limited. Also, short
sion of water molecules between the intravascular and extravascular periods of ischemia can cause endothelial leakage,133,134 which would
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 235

complicate absolute perfusion assessment by intravascular CM in patients. assessment of MR stress and rest dynamic data of 69 patients yielded a
For intravascular CM based on hyperpolarization (hyperpolarized sensitivity and specificity of 90% and 85%, respectively, for the detec-
13
C-CM), water exchange does not affect the MR signal; consequently, tion of stenoses with 70% or more diameter reduction on quantitative
perfusion quantification models could become easier. However, for these coronary angiography (QCA) at 0.075 mmol/kg of Gd-DTPA (injected
compounds, the time course of depolarization has to enter the formula at 4 mL/s into a peripheral vein). CMR performed significantly better
for perfusion models (see later).71 Similar to the approach used by with areas under the ROC curves (AUC) of 0.89 to 0.91 for two observ-
Klocke et al., that is, to use the area under the signal intensity time ers versus AUC of 0.71 to 0.75 for SPECT.135 In the large single-center
curve during first pass as a measure of perfusion,107 Lau and co-workers CE-MARC study, 647 patients were investigated with first-pass per-
applied this concept successfully to measure myocardial perfusion in fusion CMR, SPECT, and invasive x-ray coronary angiography (see
rats after co-injection of hyperpolarized 13C-pyruvate and 13C-urea to Table 18.3).3 An excellent diagnostic performance to detect CAD
simultaneously quantify myocardial metabolism and perfusion, respec- (defined as ≥50% diameter stenosis) was observed with an AUC of
tively.72 Because the hyperpolarized 13C-technique is applicable in patients, 0.84. Furthermore, this CE-MARC trial also confirmed the superiority
this perfusion approach may be considered as an alternative to con- of perfusion CMR over SPECT to detect CAD as demonstrated in the
ventional Gd-based techniques. However, to our knowledge, this tech- multicenter MR-IMPACT trial (see later and Fig. 18.7B).6 Recently,
nique has not yet been tested in humans. perfusion CMR was also evaluated at 3 T. Plein et al.93 reported an
excellent sensitivity and specificity to detect CAD of 90% and 83%,
CLINICAL PERFORMANCE OF PERFUSION respectively, which was, however, not superior to that obtained at
CARDIOVASCULAR MAGNETIC RESONANCE 1.5 T. This might be explained among other factors by the lower field
homogeneities typically encountered at higher fields strengths that
Single-Center Studies: Visual Interpretation can cause irregular RF deposition and, consequently, segmental signal
Visual discrimination of normal from severely abnormal findings is variabilities during first pass. However, the higher field strength yields
certainly feasible; however, it should be kept in mind that development better SNR, which could be “invested” in 3D techniques. Accordingly,
of CAD is a progressive process that passes from mild to moderate to three studies demonstrated high sensitivities and specificities to detect
severe lesions (most likely by repetitive plaque ruptures). It might be CAD ranging from 88% to 94% and from 74% to 81%, respectively, in
in this intermediate range of lesions where visual assessment becomes comparison to anatomy (see Table 18.3).140–142 Particularly high sensi-
difficult, and relating the patient data to a normal database might tivity and specificity of 3D perfusion CMR was achieved versus frac-
be particularly advantageous for this population advancing from tional flow reserve (FFR).141 One of these 3D perfusion CMR studies
nonobstructive to mildly obstructive CAD. Results of several studies also compared CMR versus SPECT to detect CAD, but no superior-
applying a visual reading are given in Table 18.3.3,90,94,135–142 In a recent ity of CMR versus SPECT was found in that small population of
study, first-pass perfusion CMR was compared with SPECT.135 A visual 33 patients.142

1 1

0.75 0.75
Sensitivity

Sensitivity

0.5 0.5

0.25 Adequate quality 0.25 Single center


Intermediate quality 3 centers—single vendor
All quality 18 centers—multivendor

0 0
0 0.25 0.5 0.75 1 0 0.25 0.5 0.75 1
A 1-Specificity B 1-Specificity
FIG. 18.7  (A) The important influence of data quality on test performance given by the area under the
receiver operating characteristic curve (AUC).4 If all categories of data quality are entered in the analysis,
AUC is rather low. Applying a blinded read to eliminate data with low quality (most of them caused by
breathing motion) resulted in 86% of data (intermediate category), and AUC improves. In this study, a hybrid
echo planar pulse sequence was applied and yielded five to seven slices every two RR intervals (with trig-
gering on every other R-wave only). Thus only some slices are acquired during cardiac phases with minimal
motion (end-systolic and mid-diastolic phase), whereas other slices fall within rapid motion phases. Eliminating
these slices degraded by motion further increased test performance, yielding an AUC of 0.91 (adequate
quality). (B) A similar trend is demonstrated for quality reduction in studies with increasing numbers of par-
ticipating sites,1,3 which is most evident for multivendor data.4,6,7
236 SECTION II  Ischemic Heart Disease

TABLE 18.3  Clinical Performance of Perfusion Cardiovascular Magnetic Resonance: Detection


of Coronary Artery Disease in Patients
N and Prep/ Reference
Authors Centers Vendora Slicesb CM Dose Restc Stress Analysis Standard Sens Spec AUC
Single-Center Studies
Hartnell et al.94 14/1 S– IR-2D/1 Gd-DTPA 0.04 + Dip Visual QCA (≥70%) 83%d 100% —
Eichenberger et al.82 8/1 S– 2D/3 Gd-DOTA 0.05 + Dip Upslope QCA (≥80%) 65%e 76%e —
Walsh et al.90 46/1 S– IR-2D/3 Gadoteridol 0.10 + Dip Visual 201
Th/99m Tc 89% 44% —
scinti
Bertschinger et al.54 24/1 S– SR-2D/4 Gd-DTPA-BMA 0.10 − Dip Upslopetrans QCA (≥50%) 82% 73% 0.76
Al-Saadi et al.83 34/1 S– IR-2D/1 Gd-DTPA 0.025 + Dip Upslope CFR QCA (≥75%)f 90% 83% —
Schwitter et al.1 57/1 S– SR-2D/4 Gd-DTPA-BMA 0.10 − Dip Upslopesubendo QCA (≥50%) 87% 85% 0.91
Schwitter et al.1 43/1 S– SR-2D/4 Gd-DTPA-BMA 0.10 − Dip Upslopesubendo 13
NH3-PET 91% 94% 0.93
(CFR)
Nagel et al.2 84/1 S– SR-2D/5 Gd-DTPA 0.025 + Ado Upslope CFR QCA (≥75%)f 88% 90% 0.93
Ishida et al.135;g 104/1 S– SR-multi Gd-DTPA 0.075 + Ado Visual QCA (≥70%) 90% 85% 0.90h
Doyle et al.104 138/1 S– SR-multi Gd-BOPTA 0.04 + Dip Upslope CFR QCA (≥70%) 57% 85% —
Plein et al.136
  1.5 T 37/1 S– SR-2D/4 Gd-Butrol 0.10 − Ado Visual QCA (≥50%) 90% 83% 0.80
  3 T 37/1 S– SR-2D/4 Gd-Butrol 0.10 − Ado Visual QCA (≥50%) 90% 83% 0.89
Rieber et al.144 43/1 S– SR-2D/3 Gd-DTPA-BMA 0.05 + Ado Upslope CFR FFR (≤0.75) 88% 90% 0.93
Costa et al.158 11/1 S– SR-2D/3 Gd-DTPA 0.10 + Ado Fermi CFR FFR (≤0.75) 86%i 60%i —
Lockie et al.137
  3 T 42 /1 S– SR-2D/3 Gd-DTPA 0.05 + Ado Visual FFR (<0.75) 82% 94% 0.92
  Per segment 38/1 Fermi CFR 80% 89% 0.89
Watkins et al.138 101/1 S– SR-2D/3 Gd-DTPA-BMA 0.10 + Ado Visual QCA (≥70%) 97% 78% —
FFR (<0.75) 91% 94% —
Greenwood et al.3 647/1 S– SR-2D/3 Gd-DTPA 0.05 + Ado Visual QCA (≥50%) — — 0.84
Greenwood et al.139 235♀/1 S– SR-2D/3 Gd-DTPA 0.05 + Ado Visual QCA (≥50%) 82% 89% 0.90
Manka et al.140 3T 146/1 S– SR-3D Gd-DTPA 0.10 +LGE Ado Visual 3D QCA (≥50%) 92% 74% —
Visual 3 slices 88% 74% —
Jogiya et al.141 3T 53/1 S– SR-3D Gd-Butrol 0.075 +LGE Ado Visual 3D FFR (<0.75) 91% 90% 0.89
Visual 3 slices FFR (<0.75) 85% 84% 0.85
Visual 3D QCA (≥50%) 88% 80% —
Jogiya et al.142 3T 33/1 S– SR-3D Gd-Butrol 0.075 +LGE Ado Visual QCA (≥50%) 94% 81% —

Multicenter Studiesj
Wolff et al.5 99/3 S+ SR-2D/7 Gd-DTPA 0.05 + Ado Visual QCA (≥50%) 93% 75% 0.90
Giang et al.4 99/3 S+ SR-2D/7 Gd-DTPA 0.10 − Ado Upslopesubendo QCA (≥50%) 91% 78% 0.91
Schwitter et al.6 241/18 M+ SR-2D/3 Gd-DTPA-BMA 0.10 + Ado Visual QCA (≥50%) 90% 73% 0.87
Schwitter et al.7 425/33 M+ SR-2D/3 Gd-DTPA-BMA 0.075 − Ado Visual QCA (≥50%) — — 0.75
112♀/33 M+ SR-2D/3 Gd-DTPA-BMA 0.075 − Ado Visual QCA (≥50%) — — 0.76
Manka et al.146 120/2 S– SR-3D Gd-DTPA 0.10 +LGE Ado %Volhypo at peak FFR (<0.75) 86% 86% 0.90
Manka et al.147 3T 150/5 S– SR-3D Gd-Butrol 0.075 + Ado Visual FFR (<0.80) 85% 91% 0.91
a
Single vendor (S) vs. multivendor (M): − indicates without and + indicates with external supervision by European Medicines Agency and/or US
Food and Drug Administration.
b
Prep slices: magnetization preparation by saturation recovery (SR) and inversion recovery (IR), respectively. Single and multi, single-slice, and
multislice acquisition, respectively. Multislice acquisitions indicated as 4 or 7 refer to 4 or 7 slices acquired every 2-RR interval.
c
+ indicates rest perfusion performed; − indicates rest perfusion not performed.
d
Comparison on a regional basis (5 segments/heart evaluated).
e
Comparison on a regional basis (48 segments/heart evaluated).
f
Vessel area reduction (in all other studies: vessel diameter reduction).
g
Patients with history of myocardial infarction and/or resting wall motion abnormalities excluded.
h
AUC (mean of two readers) for subset of 69 patients who also had a single-photon emission computed tomography (SPECT) study (AUC for
SPECT: 0.73; P < .001 versus magnetic resonance).
i
Per segment analysis (44 segments in 11 patients)
j
Multicenter studies, diagnostic performance is given for best CM dose (for dose ranges tested, see text).
AUC, Area under the curve; CM, contrast media (all gadolinium chelates); Fermi CFR, calculation of coronary flow reserve by quantitation based
on Fermi-constrained deconvolution using cutoffs between 1.6–2; FFR, fractional flow reserve; LGE, late gadolinium enhancement; QCA,
quantitative coronary angiography (diameter reduction); PET-CFR, positron emission tomography coronary flow reserve; scinti, scintigraphy;
Upslopetrans, upslope data derived from full wall thickness; Upslopesubendo, upslope data derived from subendocardial layer; Upslope CFR, coronary
flow reserve calculated from upslope data (ratio of slope:rest/hyperemia).
All studies performed on 1.5 T, if not mentioned otherwise. Studies with N ≥ 7 included.
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 237

1
Single-Center Studies: Quantitative
Semiautomatic Analysis
The rate of myocardial signal change (upslope) during CM first pass
is most widely used to quantitatively assess perfusion data.1,2,54,80-83,101,143,144 0.75
From a rest-stress protocol, a CFR index (slopehyperemia/sloperest) was
derived and yielded a sensitivity and specificity of 90% and 83%, respec-

Sensitivity
tively, for the detection of stenoses with 75% or more area reduction
0.50
on QCA (at a CFR threshold of 1.5).83 For an arterial input correction,
the upslope of signal in the left ventricular blood pool was used; fur- Single center
thermore, the dose of the extravascular CM Gd-DTPA was kept at Single center “CE-MARC”
0.025 mmol/kg body weight to minimize clipping of the blood-pool Multicenter single vendor, n = 3
0.25 Multicenter multivendor, n = 18
signal intensity–time curve. The same CFR index derived from multislice “MR-IMPACT I”
perfusion data (with peripheral administration of the same CM dose) Multicenter multivendor, n = 33
yielded a similar performance with a sensitivity and specificity of 88% “MR-IMPACT II”

and 90%, respectively, for the detection of stenoses with 75% or more 0
area reduction on QCA,2 and it also correlated well with FFR.144 When 0 0.25 0.5 0.75 1
CFR was determined by perfusion CMR in women, a specificity of 85%
1-Specificity
for the detection of CAD (defined as ≥70% diameter reduction on
QCA) was achieved, although sensitivity was only 57%.104 This test FIG. 18.8  These receiver operating characteristic curves demonstrate
performance at 0.04 mmol/kg of Gd-BOPTA compares well with the superiority of subendocardial first-pass perfusion data over transmural
perfusion data for detection of stenoses with ≥50% diameter reduction
results of a recent dose-finding study for the dose of 0.05 mmol/kg of
on quantitative coronary angiography,1,54 supporting the general knowl-
Gd-DTPA (see Fig. 18.3B).4
edge of the subendocardium being most sensitive for hypoperfusion.
A stress-only protocol provided a comparable performance of the In these two studies, perfusion data of patients were compared with a
hyperemic upslope parameter when thresholds were derived from a normal database. If susceptibility artifacts were to reduce signal response
normal database, resulting in a sensitivity and specificity of 87% and in the subendocardium, this approach would correct, to a certain extent,
85%, respectively, for the detection of stenoses of ≥50% diameter reduc- because the normal database would also contain lower threshold values
tion on QCA.1 This stress-only protocol avoids the need for matching (provided that both patient data and normal data are acquired and ana-
myocardial regions for rest and stress condition and therefore offers lyzed with the identical protocol and software).
easy analysis of the subendocardial layer, where perfusion abnormalities
are most severe (Fig. 18.8).1,143 In a comparison of the subendocardial
CMR upslope data with PET perfusion data, a sensitivity and specificity Because this analysis is technically easy to accomplish, this approach
of 91% and 94%, respectively, were reported.1 warrants further testing in a multivendor setting.
As discussed previously, perfusion CMR has advantages over SPECT
Multicenter Studies with respect to spatial resolution, motion suppression, and lack of
Perfusion CMR appears to represent the most accurate method cur- attenuation artifacts. This suggests superiority of perfusion CMR over
rently available for the noninvasive assessment of myocardial perfusion SPECT.135 In the MR-IMPACT multicenter program (MR-Imaging for
in humans, as evidenced in multicenter trials. Myocardial Perfusion Assessment in Coronary Artery Disease Trial),6
In a multicenter single-vendor trial, a stress-only protocol combined 241 patients underwent both invasive coronary angiography and SPECT
with a semiautomatic data analysis (upslope) performed best with CM within ± 4 weeks of a perfusion CMR study (see Fig. 18.7B).6 In this trial,
doses as high as 0.10 to 0.15 mmol/kg Gd-DTPA, yielding an AUC of monitored by official authorities (US Food and Drug Administration
0.91 ± 0.07 and 0.86 ± 0.08, respectively, with corresponding sensitivity/ [FDA], European Medicines Agency [EMA]), no serious adverse events
specificity of 91%/78% and 94%/71%, respectively.4 Thus this multi- occurred in the 234 patients dosed with Gd-DTPA-BMA. MR-IMPACT
center trial confirmed the high diagnostic performance of the stress-only confirmed the high diagnostic performance of the MR first-pass perfu-
approach proposed in an earlier single-center study.1 However, in this sion approach at a dose of 0.1 mmol/kg Gd-DTPA-BMA with an AUC
multicenter study, the high diagnostic performance was achieved in of 0.86 for the detection of CAD (defined as ≥50% diameter reduction
data with adequate quality only. For this purpose, a blinded quality on QCA, see Fig. 18.7B). Most importantly, it also showed superiority
reading was performed first, which eliminated 14% of all studies (see of perfusion CMR over nongated SPECT imaging for the detection of
Fig. 18.7A). An example from a study at the dose of 0.15 mmol/kg is CAD.6 From this MR-IMPACT trial, detailed reading and quality cri-
shown in Fig. 18.9. With adequate data quality, a κ-value of 0.73 was teria were derived and tested in the MR-IMPACT II trial. To compare
obtained for interobserver agreement,4 which compares favorably with perfusion CMR with gated SPECT the MR-IMPACT II was launched
κ-values of 0.80 for multicenter SPECT data.145 As expected with an in 33 centers in the United States and Europe (see Fig. 18.7B). The
increasing number of participating sites, homogeneity of data quality, MR-IMPACT II also tested the “minimal effective” CM dose of 0.075
and hence test performance, is likely to deteriorate, as demonstrated mml/kg to detect CAD (in comparison to 0.1 mmol/kg in MR-IMPACT
in a comparison between single-center and multicenter perfusion CMR I), and it is also the largest multicenter SPECT trial using 99mTc-tracers
studies (see Fig. 18.7B). available so far.8 This trial (monitored by the regulatory authorities
Recently, the diagnostic performance of a 3D perfusion approach FDA and EMA) analyzed 425 patients all undergoing perfusion CMR,
was evaluated at 1.5 T in two centers146 and at 3 T at five centers147 in SPECT, and invasive x-ray coronary angiography. The MR-IMPACT II
a single-vendor design. Interestingly, in the two-center study at 1.5 T, confirmed the superiority of perfusion CMR versus gated SPECT with
high sensitivity and specificity of 86% each were achieved versus FFR significantly larger AUCs for the detection of CAD.7 Perfusion CMR also
of ≤0.75 by quantifying the ischemic myocardium with a threshold <2 performed better than gated SPECT in the multivessel disease group, in
SD of remote myocardium measured at the peak myocardium signal. men, in women, as well as in the subgroup without infarction.7 Because
238 SECTION II  Ischemic Heart Disease

Anterior
Hypoperfusion

Normal

Lateral
Septal
Low
Normal
Hypo-
perfusion
A B E Inferior
G
Anterior
Scar
Thrombus
Viable
myocardium

Lateral
Septal
Scar

C D F Inferior H
FIG. 18.9  In this example of a 59-year-old patient, a first-pass perfusion deficit is shown in panel A (at the
peak effect of the bolus), encompassing approximately two-thirds of the left ventricular circumference. From
the time series of first-pass data, an upslope map is generated and compared with a normal database, allow-
ing the color coding of pixels with reduced and normal wash-in kinetics in shades of blue and red, respectively
(B). Late enhancement imaging for detection of scar tissue is shown in panel C. Combining perfusion
(A) and viability (C) data into a single map (D) allows discrimination of hypoperfused viable (jeopardized) tissue
(blue area in panel D) from hypoperfused scarred tissue (white area overlaid in panel D). Perfusion and viability
are also demonstrated in polar map format of the subendocardial layer (inner half of the myocardial wall) in
panels E and F, respectively. (G) Coronary angiography reveals an occlusion of the left anterior descending
coronary artery in this patient (arrows), with collateral vessels that protected the anterior and septal wall
from transmural infarction. The large circumflex artery is not stenosed, explaining the normal perfusion pattern
in the lateral wall in the polar map (E), whereas a stenosis at the crux of the right coronary artery (arrow in
H) causes hypoperfusion of the inferior wall. With this stress-only perfusion approach combined with a late
enhancement approach, the various tissue components (normal viable, ischemic, and scar) can be defined
within various myocardial layers with a single magnetic resonance one-stop-shop examination. Owing to the
extensive areas of hypoperfusion in viable myocardium, the patient was revascularized.

these results were obtained from a large network of 33 participating CMR. Interestingly, there is a trend toward higher complication rates
centers, representing data quality currently achievable over a substan- in CMR-positive patients in the earlier studies compared with more
tial spectrum of centers and MR machines, these data together with recent ones (inlet in Fig. 18.10), which might reflect the clinical situa-
MR-IMPACT I data6 and large single-center data3 were important to tion that, nowadays, patients with ischemia proven by CMR have better
integrate the perfusion CMR techniques into international guidelines chances to get revascularized than in the beginnings of perfusion CMR
for the work-up of CAD.148–152 when this method was not yet accepted to guide treatment. This view
is supported by the low event rate observed in the EuroCMR registry
Perfusion Cardiovascular Magnetic Resonance and population for both ischemia-negative and ischemia-positive patients,
Prediction of Outcome clearly indicating that the use of CMR in patients with suspected CAD
To recommend perfusion CMR for the management of patients with can yield a high event-free survival rate.16 In this context, it is also
known or suspected CAD, it is crucial to demonstrate that the technique noteworthy to mention that CMR-guided management of suspected
is able to reliably detect CAD: that is, to detect relevant coronary artery CAD can reduce costs in comparison to invasive strategies, even when
stenoses in patients. This goal was undoubtedly achieved with the many FFR is used to guide interventions by ischemia testing.16
positive studies performed during the last decades, as summarized in
Table 18.3. Nevertheless, a method is of most value if it can discriminate
patients with good prognosis from those with an increased risk for
PERSPECTIVES
cardiovascular complications; that is, if it can identify patients in need A main feature of CAD is its chronic course over several decades. CAD
for a specific treatment as, for example, revascularizations. In all studies may become clinically overt by symptoms in an early stage of disease,
listed in Table 18.4,10–13,15,16,47,48,95,153 the annual event rate for cardiac but it may also remain silent for many years, becoming symptomatic
death and nonfatal myocardial infarction (MI) was below 0.9%, with suddenly by an acute MI. Because symptoms are not always present
the exception of two studies with a percentage of known CAD of at heralding an imminent infarction or symptoms may be atypical and
least 43%14,96 and one study performed early (in 2009).154 Fig. 18.10 therefore may be misinterpreted, up to 60% of cardiac deaths occurred in
shows a very low event rate in ischemia-negative patients in terms of the United States outside the hospital or before the patients reached the
cardiac death and nonfatal MI (bars to the right), although the event catheterization laboratory for rescue intervention in the year 2004.155 It
rate is high in all studies for ischemia-positive patients (bars to the can be concluded that such a reactive strategy (i.e., evaluating a patient
left), providing clear evidence for the prognostic power of perfusion primarily when the patient is exhibiting symptoms) is often inadequate.
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 239

Cardiac Death and Nonfatal Myocardial Infarction


After Pathologic and Normal CMR

Pathologic CMR Normal CMR


21 20 19 18 17 16 15 14 13 12 11 10 9 8 7 6 5 4 3 2 1 0 0 1 2 ( %/y)

Specific Populations
Obesity: BMI: 35 kg/m2, I+ Sha 255, I- and S-
Men + Women: 20% known CAD. I+ 405, I-
Men, I+, 25% known CAD Co 237, I-
Women, I+, 14% known CAD 168, I-
Acute Coronary Syndrome, I+ Ing 135, I-

26% known CAD, I+ and S+


Single Center
St 254, I- and S-
43% known CAD, I+ Be 362, I- and S-
51% known CAD, I+ Ja 461, I-

56% known CAD, I+ Po 793, I-

16% known CAD, I+ Kri 1232, I- and S-

51% known CAD, I+ Bu 1152, I-


(% cardiac death + nonfatal Ml/year)

r = 0.56, P = .09
St I+ Ab 346, I-
Events in Ischemia-pos CMR

20
56% known CAD, I+ Vi 1024, I- and S-
Co Pi 218, I- and S-
15

Be EuroCMR Registry
10 3647
Ja Po Suspected CAD 59 centers
Kri
5 Bu I+ I-
Vi and/or Mo and
Ab
Mo Registry S+ S-
0 and/or and
2006 2008 2010 2012 2014 2016 LV dysfunction normal LV
function
Publication year

FIG. 18.10  Event rates are shown for patients with ischemia-positive cardiovascular magnetic resonance
(CMR; bars to the left) and ischemia-negative CMR (bars to the right). Bar thickness is proportional to the
sample size. Smaller studies (thinner bars) tend to yield larger differences in outcome between ischemia-
positive and ischemia-negative patients than large studies (thicker bars). Irrespective of study size, the
outcome is excellent for ischemia-negative patients. Insert, Studies performed earlier tend to yield a worse
outcome in ischemia-positive patients than studies performed recently. This may be explained by the fact
that, nowadays, treating physicians are aware of the prognostic power of perfusion CMR and therefore they
revascularize CMR-positive patients regularly. In the EuroCMR registry, 20.4% of the patients were ischemia
positive in at least 1 segment and 17.4% of these patients underwent revascularization (typically with 2 or
more ischemic segments). These excellent EuroCMR registry outcome data of 3647 patients demonstrate
the value of CMR to manage patients with suspected coronary artery disease (CAD). In this registry, the
CMR approach also reduced costs versus an invasive strategy.16 Abbreviations between bars and in the inset
refer to the first author’s name (as used in Table 18.4). I+ and S+ indicate patients positive for ischemia and
scar (by late gadolinium enhancement), respectively. Numbers to the right of bars indicate size of study
population. For details on imaging parameters, contrast media dose, stress agent, etc., see Table 18.4. BMI,
Body mass index; LV, left ventricular; MI myocardial infarction.

A more successful strategy would focus on an earlier detection of CAD to monitor CAD over time because it can be repeated when needed,
(i.e., a noninvasive detection of severe and, thus dangerous coronary owing to a lack of radiation. It yields a high sensitivity and specificity
lesions by CMR) and treating these lesions before they rupture, thereby for the detection of obstructing lesions and, as a short examination, it
preventing acute myocardial infarctions. Such an active strategy would is well tolerated by patients. An active strategy could potentially reduce
monitor the disease process over many years and would indicate the need the high number of “prehospital” cardiac deaths significantly, which
for action in case of a significant lesion. Because the factors triggering would represent a major success in cardiac care. Large single-center
the progression of CAD are not yet well known, a repetitive evaluation and registry data indicate that perfusion CMR can reduce costs sub-
of the coronary circulation is required. Perfusion CMR is an ideal tool stantially in comparison with invasive strategies in a population of
240 SECTION II  Ischemic Heart Disease

TABLE 18.4 Prognosis
Follow-Up
(Median Known
Study Year Tesla Sequence Stress CM Dose Analysis Months) CAD N
Steel 2009 1.5 SR-7 (2RR) Ado Gd-DTPA 0.1 Visual S+r+LGE 17 26% 254
(St) Hybrid FGE
Bertaso 2013 1.5 SR-3 Ado Gd-DTPA 0.1 Visual S+r+LGE 22 43% 362
(Be)
Jahnke 2007 1.5 SR-3 Ado Gd-DTPA 0.05 Visual S+r 27 51% 461
(Ja) (FGE/SSFP)
Pontone 2015 1.5 SR-3 Dip Gd-BOPTA 0.1 Visual S 27 56% 793
(Po)
Krittayaphong 2011 1.5 InvR-3 Ado Gd-DTPA 0.05 Visual S+LGE 35 16% 1232
(Kri)
Buckert 2013 1.5 SR-3 Ado Gd-DTPA 0.1 Visual S+r+LGE 50 51% 1229
(Bu)
Abbasi 2014 3 SR-3 Rega Gd-DTPA 0.1 Visual S+LGE 23 — 346
(Ab)
Vincenti 2016 1.5/3 SR-4 (2RR) Ado Gadobutrol 0.1 Visual S+LGE 30 45% 1024
(Vi)
Pilz 2008 1.5 SR-5 (2RR) Ado Gd-DTPA-BMA 0.1 Visual S+LGE 12 — 218
(Pi) hybridFGE

Specific Patient Populations


Shah 2014 1.5 SR-3 Ado Gd-DTPA 0.1 Visual S+LGE 25 BMI 255
(Sha) 3 Rega 35 kg/m2
Coelho- 2011 1.5 SR-5 (2RR) Ado Gd-DTPA 0.075–0.1 Visual S+LGE 30 20% 405
Filho (Co) 3 SR-5 (1RR) Dip Men: 25% 237
Women: 14% 168
Ingkanisorn 2006 1.5 SR-9 (2RR) Ado Gd-DTPA-BMA 0.1 Visual Fn+S+LGE 15 ACS 135
(Ing) Hybrid FGE

Registry
Moschetti 2016 1.5 SR-3–5 Ado Gd chelates — Visual S+LGE 12 Suspected 3647
(Mo) or 3 (1–2 RR) Dip or S+r+LGE CAD

For abbreviations of first authors refer to Fig. 18.3.


2RR, Acquisition on every other heartbeat; ACS, acute coronary syndrome; Ado, adenosine (at standard dose of 140); Analysis, S+r+LGE: all
datasets, that is, stress perfusion, rest perfusion, and LGE (late gadolinium enhancement) are used for analysis; BMI, body mass index; CAD,
coronary artery disease; Dip, dipyridamole (at standard dose of 0.84); Fn, left ventricular systolic function; Gd-DTPA (Magnevist); Gd-BOPTA
(Multihance); Gd-DTPA-BMA (Omniscan); hybridFGE, hybrid fast-gradient echo echo-planar pulse sequence; InvR, inversion recovery; Rega,
regadenoson; SR, saturation recovery; SR-3, the number indicates the number of slices acquired per R-R interval; SSFP, steady-state free
precession.

low16 to intermediate risk18 (i.e., in a suspected CAD population), while of view, (semi-)automatic analyses of perfusion CMR data would be
an excellent outcome is achieved.16 This is of paramount importance desirable to replace visual approaches to increase reproducibility and
because costs for cardiovascular disease management are expected to reduce costs through reduction of reading time.
increase from $315 billion in 2010 (direct and indirect costs in the United
States) to $918 billion in 2030.155 To determine the cost effectiveness
of perfusion CMR in various disease populations, future prospective
REFERENCES
randomized trials on economics are warranted. From a technical point A full reference list is available online at ExpertConsult.com
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 240.e1

REFERENCES angiography combined with fractional flow reserve for the diagnosis of
coronary artery disease. J Cardiovasc Magn Reson. 2014;16.
1. Schwitter J, Nanz D, Kneifel S, et al. Assessment of myocardial perfusion 18. Walker S, Girardin F, McKenna C, et al. Cost-effectiveness of
in coronary artery disease by magnetic resonance: a comparison with cardiovascular magnetic resonance in the diagnosis of coronary heart
positron emission tomography and coronary angiography. Circulation. disease: an economic evaluation using data from the CE-MARC study.
2001;103:2230–2235. Heart. 2013;99(99):873–881.
2. Nagel E, Klein C, Paetsch I, et al. Magnetic resonance perfusion 19. Celermajer DS, Sorensen KE, Gooch VM, et al. Non-invasive detection
measurements for the noninvasive detection of coronary artery disease. of endothelial dysfunction in children and adults at risk of
Circulation. 2003;108:432–437. atherosclerosis. Lancet. 1992;340:1111–1115.
3. Greenwood J, Maredia N, Younger J, et al. Cardiovascular magnetic 20. Suwaidi JA, Hamasaki S, Higano ST, Nishimura RA, Holmes DR Jr,
resonance and single-photon emission computed tomography for Lerman A. Long-term follow-up of patients with mild coronary artery
diagnosis of coronary heart disease (CE-MARC): a prospective trial. disease and endothelial dysfunction. Circulation. 2000;101:948–954.
Lancet. 2012;379:453–460. 21. Clarkson P, Celermajer DS, Donald AE, et al. Impaired vascular
4. Giang T, Nanz D, Coulden R, et al. Detection of coronary artery disease reactivity in insulin-dependent diabetes mellitus is related to disease
by magnetic resonance myocardial perfusion imaging with various duration and low density lipoprotein cholesterol levels. J Am Coll
contrast medium doses: first European multicenter experience. Eur Cardiol. 1996;28:573–579.
Heart J. 2004;25:1657–1665. 22. Lefer AM, Ma XL. Decreased basal nitric oxide release in
5. Wolff S, Schwitter J, Coulden R, et al. Myocardial first-pass perfusion hypercholesterolemia increases neutrophil adherence to rabbit coronary
magnetic resonance imaging: a multicenter dose-ranging study. artery endothelium. Arterioscler Thromb. 1993;13:771–776.
Circulation. 2004;110:732–737. 23. Barker SGE, Beesley JE, Baskerville PA, Martin JF. The influence of the
6. Schwitter J, Wacker CM, van Rossum AC, et al. MR-IMPACT: magnetic adventitia on the presence of smooth muscle cells and macrophages in
resonance imaging for myocardial perfusion assessment in coronary the arterial intima. Eur J Vasc Endovasc Surg. 1995;9:222–227.
artery disease trial: comparison of perfusion CMR with single photon 24. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased expression of
emission computed tomography for the detection of coronary artery matrix metalloproteinases and matrix degrading activity in vulnerable
disease in a multicenter, multivendor, randomized trial. Eur Heart J. regions of human atherosclerotic plaques. J Clin Invest. 1994;94:
2008;29:480–489. 2493–2503.
7. Schwitter J, Wacker CM, Wilke N, et al. Superior diagnostic performance 25. Libby P. Molecular bases of the acute coronary syndromes. Circulation.
of perfusion-CMR versus SPECT to detect coronary artery disease: the 1995;91:2844–2850.
secondary endpoints of the multicenter multivendor MR-IMPACT II. 26. Davies MJ. A macro and micro view of coronary vascular insult in
J Cardiovasc Magn Reson. 2012;14:61–71. ischemic heart disease. Circulation. 1990;82:Ii38–Ii46.
8. Schwitter J, Wacker CM, Wilke N, et al. MR-IMPACT II: magnetic 27. Bruschke AV, Kramer JR Jr, Bal ET, Haque IU, Detrano RC, Goormastic
resonance imaging for myocardial perfusion assessment in coronary M. The dynamics of progression of coronary atherosclerosis studied in
artery disease trial: perfusion-cardiac magnetic resonance vs. single- 168 medically treated patients who underwent coronary arteriography
photon emission computed tomography for the detection of coronary three times. Am Heart J. 1989;117:296–305.
artery disease: a comparative multicentre, multivendor trial. Eur Heart J. 28. Burke AP, Kolodgie FD, Farb A, et al. Healed plaque ruptures and
2012;34:775–781. sudden coronary death: evidence that subclinical rupture has a role in
9. Bruder O, Wagner A, Lombardi M, et al. European Cardiovascular plaque progression. Circulation. 2001;103:934–940.
Magnetic Resonance (EuroCMR) registry—multi national results from 29. Wilcox JN, Smith KM, Schwartz SM, Gordon D. Localization of tissue
57 centers in 15 countries. J Cardiovasc Magn Reson. 2013;15:9. factor in the normal vessel wall and in the atherosclerotic plaque. Proc
10. Ingkanisorn WP, Kwong RY, Bohme NS, et al. Prognosis of negative Natl Acad Sci USA. 1989;86:2839–2843.
adenosine stress magnetic resonance in patients presenting to an 30. Ellis S, Alderman E, Cain K, Fisher L, Sanders W, Bourassa M.
emergency department with chest pain. J Am Coll Cardiol. 2006;47: Prediction of risk of anterior myocardial infarction by lesion severity
1427–1432. and measurement method of stenoses in the left anterior descending
11. Jahnke C, Nagel E, Gebker R, et al. Prognostic value of cardiac magnetic coronary distribution: a CASS Registry Study. J Am Coll Cardiol.
resonance stress tests: adenosine stress perfusion and dobutamine stress 1988;11:908–916.
wall motion imaging. Circulation. 2007;115:1769–1776. 31. Waters D, Lesperance J, Francetich M, et al. A controlled clinical trial to
12. Coelho-Filho OR, Seabra LF, Mongeon F-P, et al. Stress myocardial assess the effect of a calcium channel blocker on the progression of
perfusion imaging by CMR provides strong prognostic value to cardiac coronary atherosclerosis. Circulation. 1990;82:1940–1953.
events regardless of patient’s sex. JACC Cardiovasc Imaging. 32. Ojio S, Takatsu H, Tanaka T, et al. Considerable time from the onset of
2011;4:850–861. plaque rupture and/or thrombi until the onset of acute myocardial
13. Krittayaphong R, Chaithiraphan V, Maneesai A, Udompanturak S. infarction in humans: coronary angiographic findings within 1 week
Prognostic value of combined magnetic resonance myocardial perfusion before the onset of infarction. Circulation. 2000;102:2063–2069.
imaging and late gadolinium enhancement. Int J Cardiovasc Imaging. 33. Kemp H, Kronmal R, Vlietstra R, Frye R. Seven year survival of patients
2011;27:705–714. with normal or near normal coronary arteriograms: a CASS registry
14. Pontone G, Andreini D, Bertella E, et al. Prognostic value of study. J Am Coll Cardiol. 1986;7:479–483.
dipyridamole stress cardiac magnetic resonance in patients with known 34. Naghavi M, Libby P, Falk E, et al. From vulnerable plaque to vulnerable
or suspected coronary artery disease: a mid-term follow-up study. Eur patient: a call for new definitions and risk assessment strategies: part I.
Radiol. 2015;1–11. Circulation. 2003;108:1664–1672.
15. Buckert D, Dewes P, Walcher T, Rottbauer W, Bernhardt P. Intermediate- 35. Farb A, Tang AL, Burke AP, Sessums L, Liang Y, Virmani R. Sudden
term prognostic value of reversible perfusion deficit diagnosed by coronary death: frequency of active coronary lesions, inactive coronary
adenosine CMR: a prospective follow-up study in a consecutive patient lesions, and myocardial infarction. Circulation. 1995;92:1701–1709.
population. JACC Cardiovasc Imaging. 2013;6:56–63. 36. Gould KL, Kirkeeide RL, Buchi M. Coronary flow reserve as a
16. Moschetti K, Petersen SE, Pilz G, et al. Cost-minimization analysis of physiologic measure of stenosis severity. J Am Coll Cardiol. 1990;15:
three decision strategies for cardiac revascularization: results of the 459–474.
“suspected CAD” cohort of the european cardiovascular magnetic 37. Schwitter J, DeMarco T, Kneifel S, et al. Magnetic resonance-based
resonance registry. J Cardiovasc Magn Reson. 2016;18:1–10. assessment of global coronary flow and flow reserve and its relation to
17. Moschetti K, Favre D, Pinget C, et al. Comparative cost-effectiveness left ventricular functional parameters: a comparison with positron
analyses of cardiovascular magnetic resonance and coronary emission tomography. Circulation. 2000;101:2696–2702.
240.e2 SECTION II  Ischemic Heart Disease

38. Uren NG, Melin JA, De Bruyne B, Wijns W, Baudhuin T, Camici PG. 57. Li D, Dhawale P, Rubin PJ, Haacke EM, Gropler RJ. Myocardial signal
Relation between myocardial blood flow and the severity of coronary response to dipyridamole and dobutamine: demonstration of the BOLD
artery stenosis. N Engl J Med. 1994;330:1782–1788. effect using a double-echo gradient-echo sequence. Magn Reson Med.
39. Di Carli M, Czernin J, Hoh CK, et al. Relation among stenosis severity, 1996;36:16–20.
myocardial blood flow, and flow reserve in patients with coronary artery 58. Atalay MK, Reeder SB, Zerhouni EA, Forder JR. Blood oxygenation
disease. Circulation. 1995;91:1944–1951. dependence of T1 and T2 in the isolated, perfused rabbit heart at 4.7T.
40. Sambuceti G, Parodi O, Marcassa C, et al. Alterations in regulation of Magn Reson Med. 1995;34:623–627.
myocardial blood flow in one-vessel coronary artery disease determined 59. Saeed M, Wendland MF, Yu KK, Li HT, Higgins CB. Dual effects of
by positron emission tomography. Am J Cardiol. 1993;72:538–543. gadodiamide injection in depiction of the region of myocardial
41. Picano E, Parodi O, Lattanzi F, et al. Assessment of anatomic and ischemia. J Magn Reson Imaging. 1993;3:21–29.
physiological severity of single-vessel coronary artery lesions by 60. Sakuma H, O’Sullivan M, Lucas J, et al. Effect of magnetic susceptibility
dipyridamole echocardiography. Comparison with positron emission contrast medium on myocardial signal intensity with fast gradient-
tomography and quantitative arteriography. Circulation. recalled echo and spin-echo MR imaging: initial experience in humans.
1994;89:753–761. Radiology. 1994;190:161–166.
42. Fung AY, Gallagher KP, Buda AJ. The physiologic basis of dobutamine as 61. Schwitter J, Saeed M, Wendland MF, et al. Assessment of myocardial
compared with dipyridamole stress interventions in the assessment of function and perfusion in a canine model of non-occlusive coronary
critical coronary stenosis. Circulation. 1987;76:943–951. artery stenosis using fast magnetic resonance imaging. J Magn Reson
43. Marwick T, Willemart B, D’Hondt AM, et al. Selection of the optimal Imaging. 1998;9:101–110.
nonexercise stress for the evaluation of ischemic regional myocardial 62. Schwitter J, Saeed M, Wendland MF, et al. Influence of the severity of
dysfunction and malperfusion. Comparison of dobutamine and myocardial injury on the distribution of macromolecules: extra- versus
adenosine using echocardiography and 99mTc-MIBI single photon intravascular gadolinium-based MR contrast media. J Am Coll Cardiol.
emission computed tomography. Circulation. 1993;87:345–354. 1997;30:1086–1094.
44. Bruder O, Schneider S, Pilz G, et al. 2015 update on acute adverse 63. Kim RJ, Fieno DS, Parrish TB, et al. Relationship of MRI delayed
reactions to gadolinium based contrast agents in cardiovascular MR. contrast enhancement to irreversible injury, infarct age, and contractile
Large multi-national and multi-ethnical population experience with function. Circulation. 1999;100:1992–2002.
37788 patients from the EuroCMR registry. J Cardiovasc Magn Reson. 64. Knuesel PR, Nanz D, Wyss C, et al. Characterization of dysfunctional
2015;17:1–7. myocardium by positron emission tomography and magnetic resonance:
45. Karamitsos TD, Ntusi NA, Francis JM, Holloway CJ, Myerson SG, relation to functional outcome after revascularization. Circulation.
Neubauer S. Feasibility and safety of high-dose adenosine perfusion 2003;108:1095–1100.
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 65. Kraitchman DL, Chin BB, Heldman AW, Solaiyappan M, Bluemke DA.
2010;12:1–8. MRI detection of myocardial perfusion defects due to coronary
46. Vasu S, Bandettini WP, Hsu L-Y, et al. Regadenoson and adenosine are artery stenosis with MS-325. J Magn Reson Imaging. 2002;15:
equivalent vasodilators and are superior than dipyridamole—a study of 149–158.
first pass quantitative perfusion cardiovascular magnetic resonance. 66. Wilke N, Kroll K, Merkle H, et al. Regional myocardial blood volume
J Cardiovasc Magn Reson. 2013;15:1–9. and flow: first-pass MR imaging with polylysine-Gd-DTPA. J Magn
47. Abbasi SA, Heydari B, Shah RV, et al. Risk stratification by regadenoson Reson Imaging. 1995;5:227–237.
stress magnetic resonance imaging in patients with known or suspected 67. Kraitchman DL, Wilke N, Hexeberg E, et al. Myocardial perfusion and
coronary artery disease. Am J Cardiol. 2014;114:1198–1203. function in dogs with moderate coronary stenosis. Magn Reson Med.
48. Shah RV, Heydari B, Coelho-Filho O, et al. Vasodilator stress perfusion 1996;35:771–780.
CMR imaging is feasible and prognostic in obese patients. JACC 68. Bjerner T, Johansson L, Ericsson A, Wikstrom G, Hemmingsson A,
Cardiovasc Imaging. 2014;7:462–472. Ahlstrom H. First-pass myocardial perfusion MR imaging with
49. Reeder SB, Atalay MK, McVeigh ER, Zerhouni EA, Forder JR. outer-volume suppression and the intravascular contrast agent
Quantitative cardiac perfusion: a noninvasive spin-labeling method that NC100150 injection: preliminary results in eight patients. Radiology.
exploits coronary vessel geometry. Radiology. 1996;200:177–184. 2001;221:822–826.
50. Poncelet BP, Koelling TM, Schmidt CJ, et al. Measurement of human 69. Hall DA, Maus DC, Gerfen GJ, et al. Polarization-enhanced NMR
myocardial perfusion by double-gated flow alternating inversion spectroscopy of biomolecules in frozen solution. Science. 1997;276:
recovery EPI. Magn Reson Med. 1999;41:510–519. 930–932.
51. Wendland MF, Saeed M, Lauerma K, de Crespigny A, Moseley ME, 70. Golman K, Axelsson O, Johannesson H, Mansson S, Olofsson C,
Higgins CB. Endogenous susceptibility contrast in myocardium during Petersson JS. Parahydrogen-induced polarization in imaging: subsecond
13
apnea measured using gradient recalled echo planar imaging. Magn C angiography. Magn Reson Med. 2001;46:1–5.
Reson Med. 1993;29:273–276. 71. Johansson E, Mansson S, Wirestam R, et al. Cerebral perfusion
52. Belle V, Kahler E, Waller C, et al. In vivo quantitative mapping of cardiac assessment by bolus tracking using hyperpolarized 13C. Magn Reson
perfusion in rats using a noninvasive MR spin-labeling method. J Magn Med. 2004;51:464–472.
Reson Imaging. 1998;8:1240–1245. 72. Lau AZ, Miller JJ, Robson MD, Tyler DJ. Simultaneous assessment of
53. Fieno DS, Shea SM, Li Y, Harris KR, Finn JP, Li D. Myocardial perfusion cardiac metabolism and perfusion using copolarized [1-13C]pyruvate
imaging based on the blood oxygen level-dependent effect using and 13C-urea. Magn Reson Med. 2016;n/a-n/a.
T2-prepared steady-state free-precession Magnetic Resonance imaging. 73. Cunningham CH, Lau JY, Chen AP, et al. Hyperpolarized 13C metabolic
Circulation. 2004;110:1284–1290. MRI of the human heart: initial experience. Circ Res. 2016;119:
54. Bertschinger KM, Nanz D, Buechi M, et al. Magnetic resonance 1177–1182.
myocardial first-pass perfusion imaging: parameter optimization for 74. Edelman RR, Li W. Contrast-enhanced echo-planar MR imaging of
signal response and cardiac coverage. J Magn Reson Imaging. 2001;14: myocardial perfusion: preliminary study in humans. Radiology.
556–562. 1994;190:771–777.
55. Wright KB, Klocke FJ, Deshpande VS, et al. Assessment of regional 75. Schwitter J, Debatin JF, von Schulthess GK, McKinnon GC. Normal
differences in myocardial blood flow using T2-weighted 3D BOLD myocardial perfusion assessed with multishot echo-planar imaging.
imaging. Magn Reson Med. 2001;46:573–578. Magn Reson Med. 1997;37:140–147.
56. Manka R, Paetsch I, Schnackenburg B, Gebker R, Fleck E, Jahnke C. 76. Ding S, Wolff SD, Epstein FH. Improved coverage in dynamic contrast-
BOLD cardiovascular magnetic resonance at 3.0 tesla in myocardial enhanced cardiac MRI using interleaved gradient-echo EPI. Magn Reson
ischemia. J Cardiovasc Magn Reson. 2010;12:1–9. Med. 1998;39:514–519.
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 240.e3

77. Schwitter J, Sakuma H, Saeed M, Wendland MF, Higgins CB. Very fast 98. Klinke V, Muzzarelli S, Lauriers N, et al. Quality assessment of
cardiac imaging. Magn Reson Imaging Clin N Am. 1996;4:419–432. cardiovascular magnetic resonance in the setting of the European CMR
78. Saeed M, Wendland MF, Sakuma H, et al. Coronary artery stenosis: registry: description and validation of standardized criteria. J Cardiovasc
detection with contrast-enhanced MR imaging in dogs. Radiology. Magn Reson. 2013;15:1–13.
1995;196:79–84. 99. Holland AE, Goldfarb JW, Edelman RR. Diaphragmatic and cardiac
79. Manning WJ, Atkinson DJ, Grossman W, Paulin S, Edelman RR. motion during suspended breathing: preliminary experience and
First-pass nuclear magnetic resonance imaging studies using implications for breath-hold MR imaging. Radiology. 1998;209:
gadolinium-DTPA in patients with coronary artery disease. J Am Coll 483–489.
Cardiol. 1991;18:959–965. 100. Chuang ML, Chen MH, Khasgiwala VC, McConnell MV, Edelman RR,
80. Lauerma K, Virtanen KS, Sipila LM, Hekali P, Aronen HJ. Multislice Manning WJ. Adaptive correction of imaging plane position in
MRI in assessment of myocardial perfusion in patients with single-vessel segmented k-space cine cardiac MRI. J Magn Reson Imaging.
proximal left anterior descending coronary artery disease before and 1997;7:811–814.
after revascularization. Circulation. 1997;96:2859–2867. 101. Keijer JT, van Rossum AC, van Eenige MJ, et al. Magnetic resonance
81. Matheijssen NA, Louwerenburg HW, van Rugge F, et al. Comparison of imaging of regional myocardial perfusion in patients with single-vessel
ultrafast dipyridamole magnetic resonance imaging with dipyridamole coronary artery disease: quantitative comparison with (201)thallium-
SestaMIBI SPECT for detection of perfusion abnormalities in patients SPECT and coronary angiography. J Magn Reson Imaging. 2000;11:
with one-vessel coronary artery disease: assessment by quantitative 607–615.
model fitting. Magn Reson Med. 1996;35:221–228. 102. Wilke N, Simm C, Zhang J, et al. Contrast-enhanced first pass
82. Eichenberger AC, Schuiki E, Kochli VD, Amann FW, McKinnon GC, von myocardial perfusion imaging: correlation between myocardial blood
Schulthess GK. Ischemic heart disease: assessment with gadolinium- flow in dogs at rest and during hyperemia. Magn Reson Med.
enhanced ultrafast MR imaging and dipyridamole stress. J Magn Reson 1993;29:485–497.
Imaging. 1994;4:425–431. 103. Panting JR, Gatehouse PD, Yang GZ, et al. Abnormal subendocardial
83. Al-Saadi N, Nagel E, Gross M, et al. Noninvasive detection of myocardial perfusion in cardiac syndrome X detected by cardiovascular magnetic
ischemia from perfusion reserve based on cardiovascular magnetic resonance imaging. N Engl J Med. 2002;346:1948–1953.
resonance. Circulation. 2000;101:1379–1383. 104. Doyle M, Fuisz A, Kortright E, et al. The impact of myocardial flow
84. Sensky PR, Jivan A, Hudson NM, et al. Coronary artery disease: reserve on the detection of coronary artery disease by perfusion imaging
combined stress MR imaging protocol-one-stop evaluation of methods: an NHLBI WISE study. J Cardiovasc Magn Reson.
myocardial perfusion and function. Radiology. 2000;215:608–614. 2003;5:475–485.
85. Pruessmann KP, Weiger M, Scheidegger MB, Boesiger P. SENSE: 105. Keijer JT, van Rossum A, van Eenige M, et al. Semiquantitation of
sensitivity encoding for fast MRI. Magn Reson Med. 1999;42:952–962. regional myocardial blood flow in normal human subjects by first-pass
86. Tsao J, Kozerke S. MRI temporal acceleration techniques. J Magn Reson magnetic resonance imaging. Am Heart J. 1995;130:893–901.
Imaging. 2012;36. 106. Lombardi M, Jones RA, Westby J, et al. Use of the mean transit time of
87. Kellman P, Derbyshire JA, Agyeman KO, McVeigh ER, Arai AE. Extended an intravascular contrast agent as an exchange-insensitive index of
coverage of first-pass perfusion imaging using slice-interleaved TSENSE. myocardial perfusion. J Magn Reson Imaging. 1999;9:402–408.
Magn Reson Med. 2004;51:200–204. 107. Klocke FJ, Simonetti OP, Judd RM, et al. Limits of detection of regional
88. Schreiber WG, Schmitt M, Kalden P, Mohrs OK, Kreitner KF, Thelen M. differences in vasodilated flow in viable myocardium by first-pass
Dynamic contrast-enhanced myocardial perfusion imaging using magnetic resonance perfusion imaging. Circulation. 2001;104:
saturation-prepared TrueFISP. J Magn Reson Imaging. 2002;16:641–652. 2412–2416.
89. Jogiya R, Schuster A, Zaman A, Motwani M, Kouwenhoven M, Nagel E. 108. Christian TF, Rettmann DW, Aletras AH, et al. Absolute myocardial
Three-dimensional balanced steady state free precession myocardial perfusion in canines measured by using dual-bolus first-pass MR
perfusion cardiovascular magnetic resonance at 3T using dual-source imaging. Radiology. 2004;232:677–684.
parallel RF transmission: initial experience. J Cardiovasc Magn Reson. 109. Kety S. The theory and application of the exchange of inert gas in lung
2014;16. and tissues. Pharmacol Rev. 1951;3:1–41.
90. Walsh EG, Doyle M, Lawson MA, Blackwell GG, Pohost GM. Multislice 110. Jerosch Herold M, Hu X, Murthy NS, Rickers C, Stillman AE. Magnetic
first-pass myocardial perfusion imaging on a conventional clinical resonance imaging of myocardial contrast enhancement with MS-325
scanner. Magn Reson Med. 1995;34:39–47. and its relation to myocardial blood flow and the perfusion reserve. J
91. Slavin GS, Wolff SD, Gupta SN, Foo TK. First-pass myocardial perfusion Magn Reson Imaging. 2003;18:544–554.
MR imaging with interleaved notched saturation: feasibility study. 111. Kroll K, Wilke N, Jerosch Herold M, et al. Modeling regional myocardial
Radiology. 2001;219:258–263. flows from residue functions of an intravascular indicator. Am J Physiol.
92. Araoz P, Glockner J, McGee K, et al. 3 Telsa MR imaging provides 1996;271:H1643–H1655.
improved contrast in first-pass myocardial perfusion imaging over a 112. Jerosch-Herold M, Swingen C, Seethamraju RV. Myocardial blood flow
range of gadolinium doses. J Cardiovasc Magn Reson. 2005;7:559–564. quantification with MRI by model-independent deconvolution. Med
93. Plein S, Schwitter J, Suerder D, Greenwood J, Boesiger P, Kozerke S. k-t Phys. 2002;29:886–897.
SENSE-accelerated myocardial perfusion MR imaging at 3.0 Tesla— 113. Wilke N, Jerosch HM, Wang Y, et al. Myocardial perfusion reserve:
comparison with 1.5 Tesla. Radiology. 2008;249:493–500. assessment with multisection, quantitative, first-pass MR imaging.
94. Hartnell G, Cerel A, Kamalesh M, et al. Detection of myocardial Radiology. 1997;204:373–384.
ischemia: value of combined myocardial perfusion and cineangiographic 114. Larsson HB, Fritz Hansen T, Rostrup E, Sondergaard L, Ring P,
MR imaging. Am J Roentgenol. 1994;163:1061–1067. Henriksen O. Myocardial perfusion modeling using MRI. Magn Reson
95. Pilz G, Jeske A, Klos M, et al. Prognostic value of normal adenosine- Med. 1996;35:716–726.
stress cardiac magnetic resonance imaging. Am J Cardiol. 2008;101: 115. Diesbourg L, Prato F, Wisenberg G, et al. Quantification of myocardial
1408–1412. blood flow and extracellular volumes using a bolus injection of
96. Bertaso AG, Richardson JD, Wong DTL, et al. Prognostic value of Gd-DTPA: kinetic modelling in canine ischemic disease. Mag Reson
adenosine stress perfusion cardiac MRI with late gadolinium Med. 1992;23:239–253.
enhancement in an intermediate cardiovascular risk population. Int J 116. Tong CY, Prato FS, Wisenberg G, et al. Measurement of the extraction
Cardiol. 2013;167:2055–2060. efficiency and distribution volume for Gd-DTPA in normal and diseased
97. Reeder S, Faranesh A, Boxerman J, McVeigh E. In vivo measurement of canine myocardium. Magn Reson Med. 1993;30:337–346.
T2* and field inhomogeneity maps in the human heart at 1.5 T. Magn 117. Fritz-Hansen T, Rostrup E, Larsson HB, Sondergaard L, Ring P,
Reson Med. 1998;39(39):988–998. Henriksen O. Measurement of the arterial concentration of Gd-DTPA
240.e4 SECTION II  Ischemic Heart Disease

using MRI: a step toward quantitative perfusion imaging. Magn Reson 136. Plein S, Kozerke S, Suerder D, et al. High spatial resolution myocardial
Med. 1996;36:225–231. perfusion cardiac magnetic resonance for the detection of coronary
118. Larsson HB, Stubgaard M, Sondergaard L, Henriksen O. In vivo artery disease. Eur Heart J. 2008;29:2148–2155.
quantification of the unidirectional influx constant for Gd-DTPA 137. Lockie T, Ishida M, Perera D, et al. High-resolution magnetic resonance
diffusion across the myocardial capillaries with MR imaging. J Magn myocardial perfusion imaging at 3.0-Tesla to detect hemodynamically
Reson Imaging. 1994;4:433–440. significant coronary stenoses as determined by fractional flow reserve.
119. Gatehouse PD, Elkington AG, Ablitt NA, Yang G-Z, Pennell DJ, Firmin J Am Coll Cardiol. 2011;57:70–75.
DN. Accurate assessment of the arterial input function during high-dose 138. Watkins S, McGeoch R, Lyne J, et al. Validation of magnetic resonance
myocardial perfusion cardiovascular magnetic resonance. J Magn Reson myocardial perfusion imaging with fractional flow reserve for the
Imaging. 2004;20:39–45. detection of significant coronary heart disease. Circulation.
120. Wendland MF, Saeed M, Yu KK, et al. Inversion recovery EPI of bolus 2009;120:2207–2213.
transit in rat myocardium using intravascular and extravascular 139. Greenwood JP, Motwani M, Maredia N, et al. Comparison of
gadolinium-based MR contrast media: dose effects on peak signal cardiovascular magnetic resonance and single-photon emission
enhancement. Magn Reson Med. 1994;32:319–329. computed tomography in women with suspected coronary artery disease
121. Judd RM, Atalay MK, Rottman GA, Zerhouni EA. Effects of myocardial from the Clinical Evaluation of Magnetic Resonance Imaging in
water exchange on T1 enhancement during bolus administration of MR Coronary Heart Disease (CE-MARC) trial. Circulation.
contrast agents. Magn Reson Med. 1995;33:215–223. 2014;129:1129–1138.
122. Judd RM, Reeder SB, May-Newman K. Effects of water exchange on the 140. Manka R, Jahnke C, Kozerke S, et al. Dynamic 3-dimensional stress
measurement of myocardial perfusion using paramagnetic contrast cardiac magnetic resonance perfusion imaging: detection of coronary
agents. Magn Reson Med. 1999;41:334–342. artery disease and volumetry of myocardial hypoenhancement
123. Donahue KM, Burstein D, Manning WJ, Gray ML. Studies of Gd-DTPA before and after coronary stenting. J Am Coll Cardiol. 2011;57:
relaxivity and proton exchange rates in tissue. Magn Reson Med. 437–444.
1994;32:66–76. 141. Jogiya R, Kozerke S, Morton G, et al. Validation of dynamic
124. Larsson HB, Rosenbaum S, Fritz-Hansen T. Quantification of the effect 3-dimensional whole heart magnetic resonance myocardial perfusion
of water exchange in dynamic contrast MRI perfusion measurements in imaging against fractional flow reserve for the detection of significant
the brain and heart. Magn Reson Med. 2001;46:272–281. coronary artery disease. J Am Coll Cardiol. 2012;60:756–765.
125. Ishida M, Ichihara T, Nagata M, et al. Quantification of myocardial 142. Jogiya R, Morton G, De Silva K, et al. Ischemic burden by 3-dimensional
blood flow using model based analysis of first-pass perfusion MRI: myocardial perfusion cardiovascular magnetic resonance: comparison
extraction fraction of Gd-DTPA varies with myocardial blood with myocardial perfusion scintigraphy. Circ Cardiovasc Imaging.
flow in human myocardium. Magn Reson Med. 2011;66:1391– 2014;7:647–654.
1399. 143. Keijer JT, van Rossum AC, Wilke N, et al. Magnetic resonance imaging
126. Broadbent DA, Biglands JD, Larghat A, et al. Myocardial blood flow at of myocardial perfusion in single-vessel coronary artery disease:
rest and stress measured with dynamic contrast-enhanced MRI: implications for transmural assessment of myocardial perfusion.
comparison of a distributed parameter model with a Fermi function J Cardiovasc Magn Reson. 2000;2:189–200.
model. Magn Reson Med. 2013;70:1591–1597. 144. Rieber J, Huber A, Erhard I, et al. Cardiac magnetic resonance perfusion
127. Fritz-Hansen T, Rostrup E, Sondergaard L, Ring PB, Amtorp O, Larsson imaging for the functional assessment of coronary artery disease: a
HB. Capillary transfer constant of Gd-DTPA in the myocardium at rest comparison with coronary angiography and fractional flow reserve. Eur
and during vasodilation assessed by MRI. Magn Reson Med. Heart J. 2006;27:1465–1471.
1998;40:922–929. 145. Wackers FJT, Bodenheimer M, Fleiss JL, Brown M. Multicenter Study on
128. Cullen JH, Horsfield MA, Reek CR, Cherryman GR, Barnett DB, Samani Silent Myocardial Ischemia Investigators. Factors affecting uniformity in
NJ. A myocardial perfusion reserve index in humans using first-pass interpretation of planar thallium-201 imaging in a multicenter trial. J
contrast-enhanced magnetic resonance imaging. J Am Coll Cardiol. Am Coll Cardiol. 1993;21:1064–1074.
1999;33:1386–1394. 146. Manka R, Paetsch I, Kozerke S, et al. Whole-heart dynamic three-
129. Rosen BD, Lima JAC, Nasir K, et al. Lower myocardial perfusion reserve dimensional magnetic resonance perfusion imaging for the detection of
is associated with decreased regional left ventricular function in coronary artery disease defined by fractional flow reserve: determination
asymptomatic participants of the multi-ethnic study of atherosclerosis. of volumetric myocardial ischaemic burden and coronary lesion
Circulation. 2006;114:289–297. location. Eur Heart J. 2012;33:2016–2024.
130. Biglands J, Magee D, Sourbron S, Plein S, Greenwood J, Radjenovic A. 147. Manka R, Wissmann L, Gebker R, et al. Multicenter evaluation of
Comparison of the diagnostic performance of four quantitative dynamic three-dimensional magnetic resonance myocardial perfusion
myocardial perfusion estimation methods used in cardiac MR imaging: imaging for the detection of coronary artery disease defined by
CE-MARC substudy. Radiology. 2015;275:393–402. fractional flow reserve. Circ Cardiovasc Imaging. 2015;8.
131. Neyran B, Janier MF, Casali C, Revel D, Canet Soulas EP. Mapping 148. Fihn SD, Gardin JM, Abrams J, et al. 2012 ACCF/AHA/ACP/AATS/
myocardial perfusion with an intravascular MR contrast agent: PCNA/SCAI/STS guideline for the diagnosis and management of
robustness of deconvolution methods at various blood flows. Magn patients with stable ischemic heart disease. J Am Coll Cardiol.
Reson Med. 2002;48:166–179. 2012;60:e44–e164.
132. Bjornerud A, Bjerner T, Johansson LO, Ahlstrom HK. Assessment of 149. Fihn S, Blankenship J, Alexander K, et al. ACC/AHA/AATS/PCNA/SCAI/
myocardial blood volume and water exchange: theoretical considerations STS focused update of the guideline for the diagnosis and management
and in vivo results. Magn Reson Med. 2003;49:828–837. of patients with stable ischemic heart disease. Circulation. 2014;130:
133. Dauber IM, VanBenthuysen KM, McMurtry IF, et al. Functional 1749–1767.
coronary microvascular injury evident as increased permeability due to 150. Montalescot G, Montalescot G, Sechtem U, et al. ESC guidelines on the
brief ischemia and reperfusion. Circ Res. 1990;66:986–998. management of stable coronary artery disease: the task force on the
134. Svendsen JH, Bjerrum PJ, Haunso S. Myocardial capillary permeability management of stable coronary artery disease of the European Society
after regional ischemia and reperfusion in the in vivo canine heart. of Cardiology. Eur Heart J. 2013;34:2949–3003.
Effect of superoxide dismutase. Circ Res. 1991;68:174–184. 151. McMurray JJV, Adamopoulos S, Anker SD, et al. ESC Guidelines for the
135. Ishida N, Sakuma H, Motoyasu M, et al. Noninfarcted myocardium: diagnosis and treatment of acute and chronic heart failure 2012. Eur
correlation between dynamic first-pass contrast-enhanced myocardial Heart J. 2012;33:1787–1847.
MR imaging and quantitative coronary angiography. Radiology. 152. Windecker S, Kolh P, Alfonso F, et al. 2014 ESC/EACTS Guidelines on
2003;229:209–216. myocardial revascularization. Eur Heart J. 2014;35:2541–2619.
CHAPTER 18  Stress Cardiovascular Magnetic Resonance: Clinical Myocardial Perfusion 240.e5

153. Vincenti G, Masci PG, Monney P, et al. Stress perfusion CMR in patients 157. Cerqueira MD, Weissman NJ, Dilsizian V, Jacobs AK, Kaul S, Laskey WK.
with known and suspected CAD: prognostic value and optimal ischemic Standardized myocardial segmentation and nomenclature for
threshold for revascularization. JACC Cardiovasc Imaging. tomographic imaging of the heart a statement for healthcare
2017;10:526–537. professionals from the Cardiac Imaging Committee of the Council on
154. Steel K, Broderick R, Gandla V, et al. Complementary prognostic values Clinical Cardiology of the American Heart Association. Circulation.
of stress myocardial perfusion and late gadolinium enhancement 2002;105.
imaging by cardiac magnetic resonance in patients with known or 158. Costa MA, Shoemaker S, Futamatsu H, et al. Quantitative magnetic
suspected coronary artery disease. Circulation. 2009;120:1390–1400. resonance perfusion imaging detects anatomic and physiologic coronary
155. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ. artery disease as measured by coronary angiography and fractional flow
Heart disease and stroke statistics—2014 update: a report from the reserve. J Am Coll Cardiol. 2007;50:514–522.
AHA. Circulation. 2014;129.
156. Iskander S, Iskandrian AE. Risk assessment using single-photon
emission computed tomographic technetium-99m sestamibi imaging.
J Am Coll Cardiol. 1998;32:57–62.
19 
Acute Myocardial Infarction: Cardiovascular
Magnetic Resonance Detection
and Characterization
Andrew Arai

Cardiovascular magnetic resonance (CMR) can characterize acute myo- 98% for ischemic heart disease.7 One important reason cine CMR per-
cardial infarction (AMI) in unique ways and with high quality. Cine formed so well may have related to how early patients were imaged
CMR can assess left ventricular (LV) volumes, ejection fraction (LVEF), after presentation to the emergency department. Specifically, 87% of
and regional wall motion in ways comparable to a high-quality trans- subjects were scanned before the 4-hour troponin was available. By
thoracic echocardiogram. Late gadolinium enhancement (LGE) is gen- studying patients within 6 hours of presentation to the emergency
erally considered the reference standard in imaging chronic myocardial department, stunned myocardium offers an additional mechanism that
infarction and the method works well for AMI as well. CMR stress is capable of detecting infarction or unstable angina. When combined
testing is capable of detecting residual myocardial ischemia. Various with LGE, the sensitivity for AMI increased to 100%. An example of a
important complications of myocardial infarction can be imaged, such rest perfusion abnormality in a patient with a normal initial electro-
as LV dysfunction, thrombus, ventricular septal defect, aneurysm, pseu- cardiogram (ECG) and normal initial troponin is shown in Fig. 19.2.
doaneurysm, and valvular complications. Newer CMR methods for However, a single CMR scan of regional wall motion does not dif-
quantifying myocardial T1, T2, T2*, and extracellular volume fraction ferentiate acute from chronic conditions. In patients with no history
(ECV) are providing quantitative insights into the pathophysiology and of myocardial infarction, a definite regional wall-motion abnormality
characterize AMI in ways that are not currently possible with alternative is reasonably specific for coronary artery disease (CAD) even though
modalities. Thus CMR is highly relevant and a powerful diagnostic tool there are other conditions that can cause regional wall-motion abnor-
for complex problems in AMI patients. malities, such as myocarditis. However, using T2-weighted imaging to
characterize the acuity of regional wall-motion abnormalities, Cury
et al.8 replicated the primary findings of Kwong et al.7 but improved
CARDIOVASCULAR MAGNETIC RESONANCE FOR the specificity for diagnosing ACS.
DETECTING ACUTE CORONARY SYNDROME AND Plein et al. formally studied the additive value of each available
CMR method for diagnosing ACS. In that study, an LV wall motion
ACUTE MYOCARDIAL INFARCTION by cine CMR had lower sensitivity (68%), perhaps as a result of the
Balanced steady-state free precession (bSSFP) cine CMR1 has been later timing of the CMR scan during the index hospitalization. In that
accepted as a reference standard for assessing LV mass, volumes, and setting, stress perfusion and CMR coronary angiography were the most
LVEF.2,3 Cine CMR is a powerful method for detecting regional wall sensitive components, and the comprehensive analysis of all methods
motion abnormalities, as is evidenced by the diagnostic accuracy of used had 96% sensitivity.9 Ingkanisorn et al. also found that stress per-
dobutamine stress tests.4–6 This is an important set of validations because fusion CMR had high diagnostic accuracy for detecting patients with
dobutamine stress testing poses some of the most extreme challenges possible ACS presenting to the emergency department.10 An example of
for assessing regional wall motion (see Chapter 17). The diagnostic a patient with a stress-induced perfusion defect that extends beyond the
information must be acquired quickly (usually within about 2 minutes) myocardial infarction is shown in Fig. 19.3. In that study, CMR did not
at a time when the heart rate is 85% of age-predicted maximum and miss any patients with major adverse cardiac events over about 1 year of
patients may be experiencing angina. follow-up.
Cine CMR can assess regional wall motion with high image quality, A series of studies from Wake Forest University examined the diag-
allowing diagnosis of small wall-motion abnormalities with confidence nostic accuracy and cost effectiveness of CMR to detect CAD in patients
that might be missed by other methods. For example, Fig. 19.1 illustrates presenting to the emergency department with chest pain. Miller et al.
a case in which a presumptive diagnosis of clinically unrecognized found that use of adenosine stress CMR to assess patients with acute chest
myocardial infarction could be made based on a focal anteroseptal pain who were triaged to a chest pain observation unit had lower short-
wall-motion abnormality that was missed on a good-quality noncontrast term11 and 1-year costs12 than those managed with standard inpatient
enhanced transthoracic echocardiogram. care. In lower risk patients presenting to the emergency department with
Cine CMR also performed well in detecting acute coronary syndrome chest pain, provider-selected evaluation reduced costs compared with
(ACS) in patients presenting to an emergency department with at least mandated stress CMR.13 That study was interesting because the primary
30 minutes of chest pain. Both qualitatively and quantitatively, regional stress test modality used was transthoracic echocardiography in 62% of
wall-motion abnormalities by cine CMR had an accuracy of 82% for patients. The primary provider used stress CMR in 32%, single proton
ACS, 89% for non–ST elevation myocardial infarction (NSTEMI), and emission computed tomography (SPECT) in only 2%, and computed

241
242 SECTION II  Ischemic Heart Disease

End diastole End systole

FIG. 19.1  A small dyskinetic wall motion abnormality associated with myocardial infarction was not seen
on transthoracic echocardiography. The cine cardiovascular magnetic resonance is shown at end diastole
and end systole. The red arrow points to the wall-motion abnormality.

Cine

Rest perfusion

FIG. 19.2  Perfusion defect (red arrowheads) associated with acute myocardial infarction in a patient with
no prior coronary disease. This patient presented to the emergency department within the 1st hour after
onset of chest pain. Cardiovascular magnetic resonance (CMR) was performed within 1 hour after arrival in
the hospital, and the initial electrocardiogram and troponin-I were normal. The diagnosis of acute myocardial
infarction was subsequently confirmed by troponin-I assays 8 hours after presentation (6 hours after CMR)
and by coronary angiography. Multiple parallel short axis images allow determination of the amount of myo-
cardium at risk.

Cine Late gadolinium enhancement Adenosine perfusion

FIG. 19.3  Adenosine stress perfusion defect (three red arrows) clearly extends beyond the edges of the
very small inferior myocardial infarction on the late gadolinium enhancement image.
CHAPTER 19  AMI: Cardiovascular Magnetic Resonance Detection and Characterization 243

tomography (CT) in 2%. In the setting of intermediate-risk patients Acutely infarcted myocardium enhances more strongly with Gd-
with chest pain in an observation unit with a negative initial ECG and based contrast agents than viable myocardium because damage to cell
negative initial troponin, CMR was again a cost-effective modality.14 membranes allows Gd into what used to be the intracellular space. This
Although CMR is not commonly used to evaluate patients with chest is represented by yellow in both the intracellular and extracellular spaces
pain in the emergency department, these studies demonstrated several in Fig. 19.4. Finally, chronically infarcted myocardium also enhances
important concepts. Cine CMR is a powerful method of assessing global strongly with Gd-based contrast agents because collagen scar has rela-
and regional LV systolic function and is useful in patients with acute tively little cellular volume and thus a large volume of distribution as
presentations of possible CAD. However, the multiple ways that CMR represented by extensive yellow patches in Fig. 19.4.
can characterize the heart, including stress perfusion, LGE of myocardial
infarction, and other methods create an even more powerful diagnostic VALIDATION OF LATE GADOLINIUM
tool. Despite the relatively high costs of CMR, the modality has high ENHANCEMENT IN ACUTE
diagnostic accuracy and can be cost effective in the management of
patients with possible ACS when applied to appropriate risk categories.
MYOCARDIAL INFARCTION
LGE-CMR is currently the highest-resolution method for detecting
LATE GADOLINIUM ENHANCEMENT OF ACUTE AMI and provides an exquisitely sensitive diagnostic test. In animal
studies in which histopathology was used as a reference standard for
MYOCARDIAL INFARCTION determining what is normal or infarcted myocardium, the bright zones
LGE-CMR is accepted as a reference standard method for imaging on LGE CMR correlate closely in size and location to the infarct delin-
myocardial scar. Before discussing applications of LGE, it is important eated by triphenoltetrazolium chloride (TTC) histopathology.16 These
to understand the different mechanisms leading to hyperenhancement experiments examined acutely infarcted canine hearts and some experi-
of AMI versus the fibrotic or collagen scar associated with chronic ments included a transient coronary occlusion in a different coronary
myocardial infarction. As summarized in Fig. 19.4, after intravenous territory to document that stunned myocardium did not enhance whereas
injection, gadolinium (Gd)-based contrast agents rapidly enter the infarcted myocardium did enhance. The bright myocardium on
interstitial space, as represented by the yellow between cardiomyocytes. LGE-CMR corresponded closely to the infarcted myocardium in reper-
Thus contrast-enhanced normal or viable myocardium has a T1 that fused and nonreperfused canine infarction at 4 hours, 1 day, 3 days, 10
is shorter than native T1. The intact cell membranes of viable cells days, 4 weeks, and 8 weeks postinfarction.17 That study also found no
exclude Gd-based contrast agents from the intracellular space; hence difference in signal intensity between viable myocardium in the area
the dark intracellular space in the diagram. Using inversion recovery at risk (AAR) versus remote myocardium.17 In another study using
methods,15 the inversion time is adjusted to null normal myocardium, electron probe x-ray microanalysis, Gd concentration was high in the
making it appear dark on the LGE images. infarcted myocardium but not statistically different between viable

Normal Acute MI Chronic MI

A B C

FIG. 19.4  In the setting of acute myocardial infarction (MI), different types of myocardium have distinctive
appearances on late gadolinium enhancement (LGE) images. (A) Normal: The myocardium outside the ische-
mic area at risk is characterized by healthy cardiomyocytes (striped cylinders) with intact cell membranes.
Extracellular gadolinium contrast agents arrive via the bloodstream and rapidly enter the interstitial space
(yellow regions) but are excluded from the intracellular space (black regions). Thus normal myocardium has
a shorter T1 after contrast administration, but the amount of enhancement is modest, and the inversion
recovery time can be adjusted to make normal myocardium appear uniformly dark (nulled). Viable or salvaged
myocardium within the area at risk has intact cell membranes and therefore excludes gadolinium contrast
agents from the intracellular space. However, some aspect of the ischemic period reversibly damaged this
part of the heart enough to raise the water content and thus lengthen the T2 of the tissue. Therefore the
area at risk is brighter than normal myocardium on T2-weighted images. The salvaged myocardium appears
essentially normal on LGE cardiovascular magnetic resonance (CMR). (B) Acute MI: The myocardium is
characterized by loss of cell membrane integrity. As long as there has been adequate reperfusion, gadolinium
contrast agents rapidly enter not only the interstitial space but also what used to be the intracellular space.
As indicated by the extensive yellow in the diagram, acutely infarcted myocardium enhances significantly
more than normal myocardium and appears bright on LGE CMR. Acutely infarcted myocardium also appears
bright on T2-weighted images, owing to the tissue swelling. (C) Chronic MI: Chronically infarcted myocardium
enhances strongly as a result of the relatively small number of viable cells and extensive interstitial space
within the collagen scar.
244 SECTION II  Ischemic Heart Disease

myocardium in the AAR versus remote myocardium.18 The elevations imaging in patients with chronic CAD undergoing revascularization.
in Gd concentration were 2 to 3 times higher still in the chronically Bullock et al.27 recommend using a higher transmural extent of hyper-
infarcted myocardium than in acutely infarcted myocardium. Failing enhancement in the context of AMI than recommended for chronic
to find an elevation in Gd concentration in the viable portion of the infarction.
AAR is likely related to the detection limit of the assay and underpow- With a slightly different interpretation of the LGE findings, Ichikawa
ered statistics owing to small sample size. This concept becomes important et al.28 found that the amount of viable-appearing myocardium was a
when one considers newer data finding CMR abnormalities in the sal- better predictor of functional recovery postinfarction than the trans-
vaged myocardium19; techniques that detect abnormalities in the viable mural extent of hyperenhanced myocardium. In a study of patients
AAR are described later in this chapter. Beyond this subtlety, there is with AMI, Baks et al.29 found that LGE could predict recovery of func-
general agreement that the hyperenhanced myocardium on LGE images tion distal to a totally occluded coronary artery, but the effect was
corresponds closely to infarcted myocardium by pathological standards modulated by the presence or absence of microvascular obstruction.
in animals. LGE-CMR imaging for AMI appears to be more sensitive than SPECT
Clinical trials helped validate that LGE imaging of AMI is a clini- imaging. This concept was already known for chronic infarction.30 In
cally validated measurement of infarct size (Table 19.1). The infarct the setting of AMI, three studies showed that LGE-CMR was more
size visualized by LGE-CMR correlated with the degree of elevations sensitive than SPECT for detecting the infarct.31–33 These studies showed
of creatine kinase-MB (CKMB) or troponin.20–22 Several studies dem- that CMR was more likely to detect small infarcts, as evidenced by
onstrated that the recovery of function could be predicted by the trans- lower troponin elevations, NSTEMI, and infarcts in the circumflex
mural extent of LGE-CMR in AMI patients.20,21,23–25 The relationship territory.
between transmural extent of infarction and recovery of function was As further evidence of the benefits of high-resolution CMR imaging
used by Kim et al.26 and Selvanayagam et al.22 to validate CMR viability of AMI, Ricciardi et al.34 helped prove the sensitivity of LGE-CMR

TABLE 19.1  Clinical Validations of Late Gadolinium Enhancement Cardiovascular Magnetic


Resonance in Myocardial Infarction
Reference N Acute ± Chronic Major Findings
Ricciardi et al.34 14 Acute Microinfarcts were detected in patients who had PCI-related elevations in CKMB. Two patterns of
6 Chronic MI were observed: small side branch occlusion and distal embolization.
Choi et al.20 24 Acute Within 7 days of AMI, infarct size correlates with peak CKMB. There is an inverse relationship
Chronic between the transmural extent of infarct and the likelihood of regional recovery of function.
Gerber et al.25 20 Acute In patients 4 days after AMI, LGE predicted recovery of regional function better than did perfusion
Chronic images, which significantly underestimated the amount of irreversible injury.
Beek et al.24 30 Acute In patients 1 week after AMI, there was an inverse relationship between the transmural extent of
Chronic infarction and the likelihood of regional recovery of function.
Ingkanisorn et al.21 33 Acute In patients 2 days after acute PCI, MI size correlated with peak troponin. AMI size predicted chronic
20 Chronic LVEF and regional function. Infarct size decreased from 16%–11% of the LV.
Lund et al.33 60 Acute SPECT and CMR infarct size correlated, but SPECT had 80% sensitivity and CMR had 100%
sensitivity. SPECT missed 6 of 30 inferior AMI.
Ibrahim et al.32 33 Acute LGE-CMR was superior to SPECT in detecting myocardial necrosis after reperfused AMI because
CMR detects small infarcts missed by SPECT.
Baks et al.23 22 Acute LGE predicted recovery of function after AMI better than perfusion imaging.
Chronic
Selvanayagam et al.35 50 Acute All patients with a troponin elevation (as low as 3.7 mg/L) associated with PCI exhibited new
24 Chronic abnormal LGE. Troponin level correlated with AMI size.
Ichikawa et al.28 18 Acute Thickness of nonenhanced myocardium had better diagnostic accuracy than percent transmural
Chronic enhancement for predicting improvement in systolic wall thickening.
Baks et al.29 22 Acute Segments without MVO had early increased wall thickness and late partial functional recovery.
Chronic Segments with MVO showed late wall thinning and no functional recovery at five months.
Gerber et al.99 16 Acute Infarct size by CT and CMR correlated well (r = 0.89), although the two standard deviation limits of
21 Chronic agreement were about 35 g. Interobserver agreement and interstudy agreement were good.
Kumar et al.100 37 Acute In patients with inferior AMI, LGE CMR detected right ventricular infarction more frequently than
Chronic echocardiography or electrocardiography.
Ibrahim et al.31 78 Acute AMI was detected more often by CMR than by SPECT, specifically for non–Q-wave AMI and for the
left circumflex territory.
Plein et al.101 25 Acute LGE mass is highest in Q-wave AMI, intermediate in non–Q ST elevation AMI, and lowest in non-ST
elevation AMI.
Kim et al.36 282 Acute Multicenter validation of LGE imaging of both acute and chronic infarction.
284 Chronic
Total 764 Acute

AMI, Acute myocardial infarction; CKMB, creatine kinase-MB; CMR, cardiovascular magnetic resonance; CT, computed tomography; LGE, late
gadolinium enhancement; LV, left ventricle; LVEF, left ventricular ejection fraction; MI, myocardial infarction; MVO, microvascular obstruction;
PCI, percutaneous coronary intervention; SPECT, single photon emission computed tomography.
CHAPTER 19  AMI: Cardiovascular Magnetic Resonance Detection and Characterization 245

imaging by detecting two types of embolic infarction in patients under- may open and allow red blood cells into the extravascular space, a situ-
going percutaneous coronary intervention (PCI) who had mild eleva- ation that will lead to abnormal iron in the interstitial space of the
tions in creatine kinase-MB. The microinfarcts ranged in size from myocardium. Ischemic injury to cardiomyocytes can lead to edema and
0.7 to 12.2 g. Selvanayagam et al.35 also found new abnormalities by increased pressure in the tissue. When tissue pressures exceed capillary
LGE-CMR in patients with post-PCI elevations in troponin and could perfusion pressure, edema can further decrease perfusion through the
detect abnormities in all patients, including troponin levels as low as myocardium. Reperfusion injury is accompanied by white blood cells,
3.7 µg/L. Before those studies, there had been debate over whether the platelets, and accompanying vasoconstrictors such as endothelin and
post-PCI biomarker elevations truly represented infarction because older thromboxane-A2. Reperfusion injury to the myocardium may exacerbate
imaging methods generally could not reliably detect abnormalities in tissue edema and may cause release of oxygen free radicals that cause
<10% of the myocardium. further damage to cardiomyocytes and the vasculature. Distal emboliza-
The culmination of validation studies of LGE as an imaging marker tion of thrombus and cholesterol plaque can compromise microvascular
of infarction was a multicenter clinical trial led by Kim et al.36 As a reperfusion despite effective epicardial PCI. Finally, some patients may
study intended to get US Food and Drug Administration approval for have genetic or other susceptibilities to microvascular injury that modu-
the contrast agent gadoversetamide, participants were randomized to late the likelihood of this complication of AMI.
one of four doses of: 0.05, 0.1, 0.2, or 0.3 mmol/kg. The sensitivity of Judd et al.40 reported that the hypoenhanced regions within an oth-
CMR for detecting infarction increased with rising dose of gadoverse- erwise contrast-enhanced AMI corresponded to regions of no reflow
tamide reaching 99% for AMI and 94% chronic infarction. Likewise, in a 2-day reperfused canine model. That same year, Lima et al.41 pub-
the accuracy of LGE-CMR for identifying infarction location also lished that similar findings were observed in patients after AMI when
increased with rising dose of gadoversetamide and was 99% for AMI imaged by contrast-enhanced CMR. Shortly thereafter, Wu et al.42 pub-
and 91% for chronic infarction. lished one of the first CMR prognosis studies. They found that patients
Overall, the transmural extent of infarction predicts recovery of with AMI characterized by having MVO had higher adverse cardiovascular
function in patients with either acute and chronic infarction although event rates than patients without MVO.
the relationship is tighter for chronic infarction. The relative prognostic significance of different CMR imaging char-
acteristics will be addressed more completely later in this chapter.
MICROVASCULAR OBSTRUCTION AFTER ACUTE
MYOCARDIAL INFARCTION T1, T2, T2*, AND ECV AS QUANTITATIVE CMR
LGE is dependent on enough perfusion to get Gd to the myocardium.
CHARACTERISTICS OF AMI
In a nonreperfused AMI, there will be a severe perfusion defect (see Signal intensity on CMR depends on the T1, T2, T2*, and proton density
Fig. 19.2) unless there is significant collateral circulation. In the setting characteristics of different tissues and fluids in the body. In the last
of successfully opening the epicardial coronary artery, microvascular several years, it has become possible to produce quantitative maps of
obstruction (MVO) and related “no-reflow” phenomena can prevent myocardial T1, T2, and T2* and ECV.43 The application of these methods
reperfusion in the core of the MI37 as shown in Fig. 19.5. CMR is gener- to characterization of AMI has led to interesting and objective observa-
ally accepted as the best noninvasive imaging test for detecting MVO. tions that indicate CMR can image and differentiate infarcted myocar-
There are four interrelated mechanisms that can lead to MVO or dium, salvaged myocardium, and remote myocardium.
no reflow: ischemic injury, reperfusion injury, distal embolization, and Using LGE imaging alone to estimate the AAR in AMI, Hillenbrand
individual susceptibility.38,39 Although vascular tissue has lower energetic et al.44 reported that one can infer the presence of salvaged myocardium
requirements than cardiomyocytes, if the duration and severity of based on a transmural extent of AMI that is less than the full wall
ischemia is severe enough, then severe ischemic capillary damage may thickness. The concept behind this method is based on the wavefront
occur. Endothelial protrusions, swelling of endothelial cells, and endo- theory of AMI45 where the infarction starts at the endocardium and
thelial blebs may block the capillary lumen. Gaps in the endothelial progresses toward the epicardium with longer durations of coronary

Cine Perfusion Delayed enhancement

FIG. 19.5  Appearance of acute myocardial infarction complicated by severe microvascular obstruction. The
signal intensity of myocardium appears relatively uniform on the cine image in this acute anteroseptal myo-
cardial infarction. A first pass perfusion cardiovascular magnetic resonance (CMR) image reveals a dark zone
consistent with low perfusion (red arrow). Severe microvascular obstruction is present, because the perfusion
defect is still present on late gadolinium enhancement CMR 10 minutes after the injection of contrast. As
is often seen, the dark patch of microvascular obstruction is surrounded by a rim of bright infarcted myocar-
dium. One can also see that the resolution of the late gadolinium enhancement image and overall image
quality is better than the perfusion image.
246 SECTION II  Ischemic Heart Disease

occlusion. One should note that the sensitivity and specificity for pre- that the infarcted subset of the AAR is represented by the LGE-CMR
dicting recovery of function by that method was about 70% based on performed at least 10 to 20 minutes after injection of Gd. In general,
the optimal point on the receiver operating characteristic (ROC) curve the AAR appears bright on T2-weighted images (long T2) or dark on
but was substantially better than occlusion time. That study also did T1-weighted images (long T1). On quantitative T1 and T2 maps, the
not make a measurement of the size of the perfusion defect. AAR will be in the bright range of the color scale because T2 and T1
Another method for estimating the AAR in AMI using only LGE are long (Fig. 19.6).53 The mechanisms behind these signal intensity
imaging is based on a measurement of the endocardial surface area of changes include the observations that recent ischemic myocardium is
the hyperenhanced AMI on LGE-CMR. This method is also based on either edematous or has abnormal tissue characteristics associated with
the wavefront theory of how MI develops.45 The endocardial surface postinfarction inflammation.
area method extrapolates transmurally from the endocardial extent of The original noncontrast method for estimating the size of the AAR
AMI to define the AAR. This method worked well in the first clinical used dark-blood, native T2-weighted imaging and was validated in a
study of 83 patients with AMI when compared with angiographic esti- reperfused canine infarct model19 as well as a nonreperfused model.54
mates of the AAR and as a constraint of the AMI size by LGE.46 Using Dark-blood prepared T2-weighted imaging was validated against human
CMR endocardial surface area and the same angiographic indices of SPECT imaging of the AAR by Carlsson et al.55 on day 1 and day 7 after
AAR, Wright et al.47 found weaker correlations between these measures AMI. This is the most widely used CMR method for imaging the AAR
of AAR than with T2-weighted estimates. In a study of 197 patients, and has been used in many clinical trials. An example of a T2-weighted
Fuernau et al.48 found that AAR was reliably assessed by T2-weighted image from a patient with AMI is shown in Fig. 19.7.
CMR. They found that the endocardial surface area underestimated It is important to recognize that there has been controversy in the
the AAR in patients with high myocardial salvage, such as aborted field.56,57 Recent studies have clarified that the T2 abnormalities follow
infarction. Ubachs et al. also found that the endocardial surface area a bimodal time course in a swine model of ischemia and reperfusion
underestimated the AAR, particularly in patients with aborted AMI. that appear to correspond to an initial wave of postischemic edema
These effects are also seen in the careful validation studies of LGE by followed by a second wave of post-AMI inflammation and healing.58,59
Fieno et al. where the endocardial surface area underestimates the cir- The T2 abnormalities fade at about 24 hours post-AMI but rebound
cumferential extent of the fluorescent marker of the AAR in multiple a day or more later and are quite obvious at day 4 and day 7, with the
cases in those canine infarcts. methods used in those papers delineating the bimodal course of edema.
Several alternative CMR tissue characterization methods have T2 abnormalities also persist for several weeks into the healing phase
been proposed and validated to estimate AAR and myocardial salvage after AMI.60,61 Thus T2 abnormalities in the setting of AMI need to be
including T2-weighted imaging,19 T2 mapping,49 T1 mapping, early interpreted carefully.
Gd enhancement,50,51 and contrast enhanced bSSFP cine CMR after Gd Methodologically, the dark-blood T2-weighted images were limited by
administration.52 In general, these methods are based on the concept two types of artifacts: lower than expected signal intensity in portions of

T1 T2 Microspheres TTC
1.75
1.50
1.00
0.50

0.00
mL/min/g
FIG. 19.6  Quantitative parametric maps of T1 and T2 to detect the area at risk were validated against
microspheres and triphenoltetrazolium chloride (TTC) histopathology. (From Ugander M, Bagi PS, Oki AJ,
et al. Myocardial edema as detected by pre-contrast T1 and T2 CMR delineates area at risk associated with
acute myocardial infarction. JACC Cardiovasc Imaging. 2012;5:596–603.)

Cine Late gadolinium enhancement T2-weighted black blood

AAo

LA

LV

FIG. 19.7  Appearance of acute anteroseptal myocardial infarction on cine cardiovascular magnetic resonance
(CMR) (left), late gadolinium enhancement (LGE) (middle), and T2-weighted images (right). Note that the
LGE-CMR shows a thin rim of viable dark myocardium on the right ventricular side of the interventricular
septum (red arrows). The bright zone on the T2-weighted black-blood images is nearly transmural in this
entire distribution. AAo, Aortic arch; LA, left atrium; LV, left ventricle.
CHAPTER 19  AMI: Cardiovascular Magnetic Resonance Detection and Characterization 247

the LV wall that moved too quickly through the cardiac cycle, and bright enhancement in 37 patients, 15 of whom had nontransmural infarc-
blood near the endocardium that could be confused with edematous or tion and 22 with transmural infarction as defined by conventional LGE
infarcted myocardium. Bright-blood T2-weighted methods improved images. In both types of infarction, early Gd enhancement showed
the diagnostic reliability of this type of imaging.62,63 The bright-blood transmural defects whereas the transmural extent of infarction and the
T2-weighted methods have been validated in patients for imaging the later images during the time course converged on the findings shown
AAR64 and can detect intramyocardial hemorrhage.65 Ultimately, the by standard LGE methods.
utility of the T2-weighted imaging methods depends on the expertise Contrast-enhanced cine CMR can also delineate the AAR associated
of the user in adjusting the brightness and contrast on the computer with AMI while also providing measurements of LV size, mass, regional
monitor used to display the images, a factor that is difficult because of wall motion, and LVEF. Contrast-enhanced cine CMR has been validated
the relative subtlety of the findings in the AAR. against SPECT measurements of the AAR in a swine model.77 Contrast-
Imaging T2* becomes important to evaluate intramyocardial hemor- enhanced cine CMR has been validated in patients compared with
rhage because the hemoglobin breakdown products alter the T2* in SPECT measurements of the AAR.78 Contrast-enhanced cine CMR can
detectable ways. Unlike thalassemia where a minimal number of myo- be automatically segmented.78 Contrast-enhanced cine CMR can be
cardial measurements of T2* can be used to manage a patient, intra- performed on scanners from manufacturers.79 Furthermore, the success
myocardial hemorrhage involves a subset of patients with AMI and a rate of imaging the AAR was higher with contrast-enhanced cine CMR
subset of the infarcted myocardium. Furthermore, T2* associated with than dark-blood prepared T2-weighted imaging.79
intramyocardial hemorrhage anywhere other than the interventricular Ultimately, the rationale for imaging the AAR relates to reducing
septum is not always measurable. Research methods for producing maps sample size for early clinical trials exploring new therapies to reduce
of T2* are being explored. Clinically, these issues are not so important the infarct size. This concept has been formally analyzed in real clinical
because clinicians infer the presence of intramyocardial hemorrhage data from published trials. Engblom et al.80 used data from the CHILL-
from some types of T2-weighted images65 based on signal intensity MI and MITOCARE trials to estimate sample size needed to detect a
changes within the core of LGE images and any other available methods. treatment effect equivalent to a 25% decrease in endpoint. In brief,
Quantitative native T1 and T2 maps (see Fig. 19.6) have improved myocardial salvage measured using a combination of CMR infarct size
the objectivity of imaging and analyzing abnormalities in the setting and AAR had a smaller needed sample size (N = 50 per arm) versus
of AMI. T2 maps have been developed by at least two methods.49,66 infarct size alone (N = 100), peak troponin (N = 120), and peak CKMB
Motion-corrected versions have facilitated using these methods in patients (N = 143). Smaller sample size makes phase II clinical trials more feasible
who cannot hold their breath.67 T2 mapping has been validated to than jumping straight to larger clinical trials based on mortality or
measure the AAR in preclinical models against microspheres53 and in major adverse cardiovascular events.
patients against 18F-fluorodeoxyglucose (FDG).68 There is also a need to come to consensus on the methods used to
Similarly, T1 mapping methods69 have been refined over the years70 measure infarct size. Comparisons of measurements of infarct size made
and now include motion-correction methods that can facilitate imaging by three different clinical trial core laboratories revealed sources of
of patients unable to perform breath-holds. T1 mapping methods have variability in methods used for making these critical measurements.81
been validated for imaging the AAR associated with AMI against refer- The concept of imaging AAR, infarct size, and myocardial salvage is a
ence standards of microspheres, fluorescent dyes, and CT perfusion significant step forward in clinical trials focused on improving treat-
images.53,71 There are even suggestions that T1 mapping will be able to ment of AMI, but the field needs to come to consensus on how to use
differentiate reversibly-injured versus irreversibly-injured myocardium these methods.
in AMI.72
Estimates of myocardial ECV can be determined by measuring myo-
cardial and blood T1 before and after Gd injection when calibrated with PROGNOSIS IN PATIENTS WITH ACUTE
a measurement of hematocrit.73 ECV measurements have the conceptual MYOCARDIAL INFARCTION RELATED TO
benefit in that they convert CMR signal intensities and terminology (such CARDIOVASCULAR MAGNETIC RESONANCE
as T1, which is unfamiliar to most practitioners) into a physiological
context related to the pharmacology of where the Gd-based contrast
CHARACTERISTICS
agents distribute in the myocardium (the extracellular volume). Garg As summarized in Table 19.2, there are many studies that have examined
et al.74 found that ECV maps could determine both the AAR and infarct the prognostic significance of various CMR parameters in predicting
size in patients. At an ECV threshold >33%, the extent of abnormal ECV the outcomes of patients after AMI. In this summary, results from 3447
correlated closely with T2-weighted estimates of the AAR. At an ECV patients are available. Most studies made basic measurements of LVEF
threshold >46%, the extent of ECV abnormality agreed well with final and some estimate of the size or an index of the extent and/or trans-
infarct size as determined by LGE repeated at 3 months. Finally, acute LGE murality of AMI.
imaging as defined by the 5-standard deviation threshold method over- If there are some generalizations that can be gleaned from the prog-
estimated final infarct size at 3 months. Thus quantitative ECV mapping nosis studies, infarct size and LVEF are generally predictors of adverse
may bring objectivity to both AAR imaging and acute infarct sizing. prognosis, at least in univariable analysis.42,82–90 Perhaps more intriguing,
Early Gd enhancement imaging correlated with the size of the the presence of microvascular obstruction or the no-reflow phenomenon
T2-weighted abnormality in a study by Matsumoto et al.51 They also often is a stronger predictor of adverse prognosis than AMI size or
showed the Gd-enhanced abnormality was larger on the early images LVEF42,85,86,91 although there are some exceptions to this generaliza-
than the infarct size as expected for a measure of AAR. In a separate tion.82,88 The rationale for why MVO might portend worse prognosis
study, they found that the optimal timing for early Gd enhancement is an enigma because LGE is supposed to represent irreversible injury.
was about 2 minutes after Gd injection when the reference standard MVO also tends to affect a small subset of the myocardium, frequently
was the size of the T2-weighted abnormality.75 Hammer-Hansen et al.50 only 1% to 4% of the left ventricle compared with MI sizes that are 3
provided microsphere and TTC validation of the early Gd and LGE to 15 times larger. MVO is associated with longer durations of ischemia
findings being consistent with AAR and infarct size, respectively, in a that can result in ischemic damage to the blood vessels and allow intra-
canine model. Hammer-Hansen et al.76 did a kinetic study of early Gd myocardial hemorrhage. Disrupted microvasculature may impair healing.
248 SECTION II  Ischemic Heart Disease

TABLE 19.2  Prognosis Related to Cardiovascular Magnetic Resonance Findings After Acute
Myocardial Infarction
Reference N Independent Prognostic Variables and Outcomes
42
Wu et al. 44 MVO predicted MACE even controlling for infarct size.
Infarct size predicted 16-month MACE.
Hombach et al.87 110 LVEF, LVEDV, MVO were independent predictors of MACE.
Infarct size, MVO were predictors of LV remodeling.
Bruder et al.102 67 Only MVO was associated with 1.2-year MACE in models that included age, gender, diabetes, infarct size, and LVEF.
Wu et al.90 122 MI size, LVEDV, LVESV, LVEF were all independent predictors of 2-year MACE.
Infarct size was the only significant predictor of MACE in the multivariable model.
Bodi et al.84 214 Transmural necrosis/segment and WMA/segment were the only predictors of 1.5-year MACE in models that included
multiple clinical and CMR variables.
Cochet et al.85 184 Infarct size >10% of LV, LVEF <40%, MVO were univariable predictors of MACE.
MVO or persistent MVO were the only significant predictors of MACE in multivariable models.
Bodi et al.83 119 An index of the extent of transmural infarct was as good a predictor of 1.7-year MACE and recovery of systolic
function as a low dobutamine CMR.
de Waha et al.86 438 The presence of MVO was the strongest 19-month MACE in multivariable models that included post-PCI TIMI-flow,
ST-resolution, LVEF, and infarct size.
Eitel et al.92 208 Less than median myocardial salvage was the strongest predictor of 6-month MACE. Predictors of myocardial
salvage were ST-segment resolution, time from symptom to PCI, anterior infarct, and TIMI flow ≥2 before PCI.
Jensen et al.94 RV infarct was common in both anterior and inferior STEMI (65% and 47%, respectively). RV infarct modulated risk
of all infarcts, inferior infarcts, and anterior infarcts.
Larose et al.88 103 Infarct size was a better predictor of long-term LV function than transmurality of infarct, MVO, or myocardial salvage.
Infarct size was an independent predictor of 2-year MACE.
Miszalski-Jamka et al.95 99 The extent of RV infarct size and RV dysfunction provided incremental prognostic value for 3-year MACE over LVEF,
LV infarct size, LV transmural extent of infarct.
Amabile et al.103 114 Intramyocardial hemorrhage from T2 images was associated with larger infarct size, MVO extent, and lower LVEF.
Intramyocardial hemorrhage was an independent predictor of 1-year MACE.
de Waha et al.91 438 The ratio of MVO/ infarct size was a better predictor of MACE than MVO or infarct size alone.
de Waha et al.104 322 Transmural infarct versus nontransmural MI did not discriminate risk of MACE.
Grothoff et al.93 421 RV infarction had a higher rate of MACE (31.9%) compared with those patients with RV infarction (7.3%). RV infarct,
TIMI-flow before PCI, and incomplete ST-segment resolution were all predictors of MACE. This study may have
been the first to assess RV area at risk and RV salvage.
Ahn et al.82 135 Extent of transmural infarction was a better predictor of LV remodeling (>20% increase in LVEDV at 6-month
follow-up) than LVEF, MVO, or infarct location.
Lønberg et al.89 309 MI size measured 3-months postinfarct was an independent predictor of 2.2-year MACE in multivariable models that
included age, peak troponin, LVEF, LVEDV, and heart rate.
Total 3447

CMR, Cardiovascular magnetic resonance; LV, left ventricle or left ventricular; EF, left ventricular ejection fraction; EDV, either end diastolic
volume or end diastolic volume index; MACE, a composite end point of major adverse events; MI, myocardial infarction; MVO, microvascular
obstruction or “no reflow”; N, number of subjects in the study; PCI, percutaneous coronary intervention; RV, right ventricle or right ventricular;
STEMI, ST-elevation myocardial infarction; TIMI, the “Thrombolysis In Myocardial Infarction” group.

Iron deposition in the myocardium may have adverse local metabolic images of mechanical complications of AMI and may be more sensitive
consequences. than echocardiography for certain problems.
There are a few studies that have looked at the prognostic significance LV thrombus is important to detect, owing to the risk of systemic
of myocardial salvage; some found this was an important predictor of embolism and the need for more aggressive anticoagulation. LGE-CMR
prognosis92 whereas others found myocardial salvage was not as strong is more sensitive than transthoracic echocardiography or transesophageal
as other markers.86 One study was able to examine the prognosis related echocardiography.96,97 In general, LV thrombus does not enhance early
to myocardial edema and myocardial salvage in the right ventricle.93 or late after administration of Gd-based contrast agents (Fig. 19.8).
Infarction or injury to the right ventricle is an important modulator CMR can also detect mechanical complications after AMI. Ven-
of prognosis after AMI.93–95 tricular aneurysm and pseudoaneurysm can readily be identified and
characterized by CMR (Fig. 19.9). Infarct related ventricular septal
CARDIOVASCULAR MAGNETIC RESONANCE TO defect can be identified and characterized with velocity-encoded phase-
ASSESS COMPLICATIONS OF ACUTE contrast cine CMR (Fig. 19.10). Rupture of papillary muscles can be
detected with a combination of cine CMR, phase-contrast CMR, and
MYOCARDIAL INFARCTION LGE-CMR. Detailed CMR images of these complications may help to
Although transthoracic echocardiography is still the first-line test for guide surgical repair of these uncommon but serious complications
assessing most complications of AMI, CMR can provide high-quality of AMI.
CHAPTER 19  AMI: Cardiovascular Magnetic Resonance Detection and Characterization 249

Cine Late gadolinium enhancement

LV
LV
LA
LA

FIG. 19.8  Left ventricular apical thrombus associated with acute anterior myocardial infarction shown in the
two-chamber view (left, end-systolic frame from cine cardiovascular magnetic resonance; right, late gadolinium
enhancement images). The black arrow points to the apical thrombus. LA, Left atrium; LV, left ventricle.

Cine Late gadolinium enhancement

RV
LV

RA

LA

FIG. 19.9  Pseudoaneurysm of the basal lateral wall after acute myocardial infarction shown in the four-
chamber view. The pseudoaneurysm is partially filled with a thrombus (arrow) on late gadolinium enhance-
ment but not seen on the cine image, owing to a slightly different image plane. LA, Left atrium; LV, left
ventricle; RA, right atrium; RV, right ventricle.

Cine Late gadolinium enhancement Magnitude image (PC) Velocity PC (y)

FIG. 19.10  Ventricular septal defect 1 day after percutaneous intervention for large anterior and septal acute
myocardial infarction. The mid to apical septum already shows an aneurysmal contour with severe thinning
of some portions of the septum on cine cardiovascular magnetic resonance (left-most image). The septum
and apex are transmurally infarcted on late gadolinium enhancement (green arrows) with dark patches con-
sistent with microvascular obstruction (second image). The magnitude images from the velocity-encoded
phase contrast images show a dark jet consistent with turbulent flow through the interventricular septum
(red arrow). On the velocity-encoded images, the red arrow points to a white patch that appears to indicate
that the left-to-right flow angles toward the apex in this image, where white represents high velocities toward
the apex, gray is zero velocity, and black means high velocity toward the base of the heart. The asterisk
indicates systolic blood flow through the left ventricular outflow tract.
250 SECTION II  Ischemic Heart Disease

TABLE 19.3  Indications for Cardiovascular Magnetic Resonance in the Guidelines Published
by the European Society of Cardiology That May Be Applicable to Patients After Myocardial
Infarctiona
Class Level
Recommendations for CMR in Patients With STEMI
If echocardiography is not feasible, CMR may be used as an alternative for assessment of infarct size and resting LV function. IIb C
For patients with multivessel disease, or in whom revascularization of other vessels is considered, stress testing or imaging I A
(e.g., using stress myocardial perfusion scintigraphy, stress echocardiography, positron emission tomography, or CMR) for
ischemia and viability is indicated before or after discharge.

CMR in Patients With Suspected Non–ST-Elevation Acute Coronary Syndromes


In patients with no recurrence of chest pain, normal ECG findings and normal levels of cardiac troponin (preferably high I A
sensitivity), but suspected acute coronary syndrome, a noninvasive stress test (preferably with imaging) for inducible
ischemia is recommended before deciding on an invasive strategy.

CMR in the Context of Myocardial Revascularization


Stress CMR, stress-echo, SPECT or PET are recommended in subjects with intermediate pretest probability for suspected I A
coronary artery disease and stable symptoms.
To achieve a prognostic benefit by revascularization in patients with coronary artery disease, ischemia has to be documented I A–C
by noninvasive imaging.

Recommendations for CMR in Acute and Chronic Heart Failure


CMR imaging is recommended to evaluate cardiac structure and function, to measure LVEF, and to characterize cardiac I C
tissue, especially in subjects with inadequate echocardiographic images or where the echocardiographic findings are
inconclusive or incomplete (but taking account of cautions/contraindications to CMR).
Myocardial perfusion/ischemia imaging (echocardiography, CMR, SPECT, or PET) should be considered in patients thought to IIa C
have coronary artery disease, and who are considered suitable for coronary revascularization, to determine whether there is
reversible myocardial ischemia and viable myocardium.
a
The guidelines were abstracted by von Knobelsdorff-Brenkenhoff and Schulz-Menger.98 Note that there are other potential indications such as
evaluating for intracardiac thrombus that are not currently mentioned in European Society of Cardiology guidelines.
CMR, Cardiovascular magnetic resonance; LV, left ventricle; PET, positron emission tomography scan; SPECT, single proton emission computed
tomography; STEMI, ST-elevation myocardial infarction.

of reasonable clinical indications for CMR after AMI, which are a subset
CONCLUSION
of recently published appropriateness criteria.98 Note that postinfarct
CMR offers a wide range of imaging methods suitable for detecting patients must be imaged efficiently and quickly.
AMI and assessing many clinically important questions. The combina-
tion of cine, perfusion, and LGE-CMR is a powerful and useful test.
Some patients may benefit from more detailed imaging with velocity-
REFERENCES
encoded CMR or T2-weighted images. Table 19.3 provides a summary A full reference list is available online at ExpertConsult.com
CHAPTER 19  AMI: Cardiovascular Magnetic Resonance Detection and Characterization 250.e1

REFERENCES reversible and irreversible ischemic injury. Circulation. 2002;105:


224–229.
1. Barkhausen J, Ruehm SG, Goyen M, Buck T, Laub G, Debatin JF. MR 19. Aletras AH, Tilak GS, Natanzon A, et al. Retrospective determination of
evaluation of ventricular function: true fast imaging with steady-state the area at risk for reperfused acute myocardial infarction with
precession versus fast low-angle shot cine MR imaging: feasibility study. T2-weighted cardiac magnetic resonance imaging: histopathological and
Radiology. 2001;219:264–269. displacement encoding with stimulated echoes (DENSE) functional
2. Bellenger NG, Davies LC, Francis JM, Coats AJ, Pennell DJ. Reduction in validations. Circulation. 2006;113:1865–1870.
sample size for studies of remodeling in heart failure by the use of 20. Choi KM, Kim RJ, Gubernikoff G, Vargas JD, Parker M, Judd RM.
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2000;2: Transmural extent of acute myocardial infarction predicts long-term
271–278. improvement in contractile function. Circulation. 2001;104:1101–1107.
3. Bellenger NG, Grothues F, Smith GC, Pennell DJ. Quantification of right 21. Ingkanisorn WP, Rhoads KL, Aletras AH, Kellman P, Arai AE.
and left ventricular function by cardiovascular magnetic resonance. Gadolinium delayed enhancement cardiovascular magnetic resonance
Herz. 2000;25:392–399. correlates with clinical measures of myocardial infarction. J Am Coll
4. Nagel E, Lehmkuhl HB, Bocksch W, et al. Noninvasive diagnosis of Cardiol. 2004;43:2253–2259.
ischemia-induced wall motion abnormalities with the use of high-dose 22. Selvanayagam JB, Kardos A, Francis JM, et al. Value of delayed-
dobutamine stress MRI: comparison with dobutamine stress enhancement cardiovascular magnetic resonance imaging in predicting
echocardiography. Circulation. 1999;99:763–770. myocardial viability after surgical revascularization. Circulation.
5. Paetsch I, Jahnke C, Ferrari VA, et al. Determination of interobserver 2004;110:1535–1541.
variability for identifying inducible left ventricular wall motion 23. Baks T, van Geuns RJ, Biagini E, et al. Recovery of left ventricular
abnormalities during dobutamine stress magnetic resonance imaging. function after primary angioplasty for acute myocardial infarction. Eur
Eur Heart J. 2006;27:1459–1464. Heart J. 2005;26:1070–1077.
6. Rerkpattanapipat P, Link KM, Hamilton CA, Hundley WG. Detecting 24. Beek AM, Kuhl HP, Bondarenko O, et al. Delayed contrast-enhanced
left ventricular myocardial ischemia during intravenous dobutamine magnetic resonance imaging for the prediction of regional functional
with cardiovascular magnetic resonance imaging (MRI). J Cardiovasc improvement after acute myocardial infarction. J Am Coll Cardiol.
Magn Reson. 2001;3:21–25. 2003;42:895–901.
7. Kwong RY, Schussheim AE, Rekhraj S, et al. Detecting acute coronary 25. Gerber BL, Garot J, Bluemke DA, Wu KC, Lima JA. Accuracy of
syndrome in the emergency department with cardiac magnetic contrast-enhanced magnetic resonance imaging in predicting
resonance imaging. Circulation. 2003;107:531–537. improvement of regional myocardial function in patients after acute
8. Cury RC, Shash K, Nagurney JT, et al. Cardiac magnetic resonance with myocardial infarction. Circulation. 2002;106:1083–1089.
T2-weighted imaging improves detection of patients with acute 26. Kim RJ, Wu E, Rafael A, et al. The use of contrast-enhanced magnetic
coronary syndrome in the emergency department. Circulation. 2008; resonance imaging to identify reversible myocardial dysfunction. N Engl
118:837–844. J Med. 2000;343:1445–1453.
9. Plein S, Greenwood JP, Ridgway JP, Cranny G, Ball SG, Sivananthan MU. 27. Bulluck H, Rosmini S, Abdel-Gadir A, et al. Redefining viability by
Assessment of non-ST-segment elevation acute coronary syndromes cardiovascular magnetic resonance in acute ST-segment elevation
with cardiac magnetic resonance imaging. J Am Coll Cardiol. myocardial infarction. Sci Rep. 2017;7:14676.
2004;44:2173–2181. 28. Ichikawa Y, Sakuma H, Suzawa N, et al. Late gadolinium-enhanced
10. Ingkanisorn WP, Kwong RY, Bohme NS, et al. Prognosis of negative magnetic resonance imaging in acute and chronic myocardial infarction.
adenosine stress magnetic resonance in patients presenting to an Improved prediction of regional myocardial contraction in the chronic
emergency department with chest pain. J Am Coll Cardiol. 2006;47: state by measuring thickness of nonenhanced myocardium. J Am Coll
1427–1432. Cardiol. 2005;45:901–909.
11. Miller CD, Hwang W, Hoekstra JW, et al. Stress cardiac magnetic 29. Baks T, van Geuns RJ, Biagini E, et al. Effects of primary angioplasty for
resonance imaging with observation unit care reduces cost for patients acute myocardial infarction on early and late infarct size and left
with emergent chest pain: a randomized trial. Ann Emerg Med. ventricular wall characteristics. J Am Coll Cardiol. 2006;47:40–44.
2010;56:209–219.e2. 30. Wagner A, Mahrholdt H, Holly TA, et al. Contrast-enhanced MRI and
12. Miller CD, Hwang W, Case D, et al. Stress CMR imaging observation routine single photon emission computed tomography (SPECT)
unit in the emergency department reduces 1-year medical care costs in perfusion imaging for detection of subendocardial myocardial infarcts:
patients with acute chest pain: a randomized study for comparison with an imaging study. Lancet. 2003;361:374–379.
inpatient care. JACC Cardiovasc Imaging. 2011;4:862–870. 31. Ibrahim T, Bulow HP, Hackl T, et al. Diagnostic value of contrast-
13. Miller CD, Hoekstra JW, Lefebvre C, et al. Provider-directed imaging enhanced magnetic resonance imaging and single-photon emission
stress testing reduces health care expenditures in lower-risk chest pain computed tomography for detection of myocardial necrosis early after
patients presenting to the emergency department. Circ Cardiovasc acute myocardial infarction. J Am Coll Cardiol. 2007;49:208–216.
Imaging. 2012;5:111–118. 32. Ibrahim T, Nekolla SG, Hornke M, et al. Quantitative measurement of
14. Miller CD, Case LD, Little WC, et al. Stress CMR reduces infarct size by contrast-enhanced magnetic resonance imaging early after
revascularization, hospital readmission, and recurrent cardiac testing in acute myocardial infarction: comparison with single-photon emission
intermediate-risk patients with acute chest pain. JACC Cardiovasc tomography using Tc99m-sestamibi. J Am Coll Cardiol. 2005;45:
Imaging. 2013;6:785–794. 544–552.
15. Simonetti OP, Kim RJ, Fieno DS, et al. An improved MR imaging 33. Lund GK, Stork A, Saeed M, et al. Acute myocardial infarction:
technique for the visualization of myocardial infarction. Radiology. evaluation with first-pass enhancement and delayed enhancement MR
2001;218:215–223. imaging compared with 201Tl SPECT imaging. Radiology.
16. Kim RJ, Fieno DS, Parrish TB, et al. Relationship of MRI delayed 2004;232:49–57.
contrast enhancement to irreversible injury, infarct age, and contractile 34. Ricciardi MJ, Wu E, Davidson CJ, et al. Visualization of discrete
function. Circulation. 1999;100:1992–2002. microinfarction after percutaneous coronary intervention associated
17. Fieno DS, Kim RJ, Chen EL, Lomasney JW, Klocke FJ, Judd RM. with mild creatine kinase-MB elevation. Circulation. 2001;103:
Contrast-enhanced magnetic resonance imaging of myocardium at risk: 2780–2783.
distinction between reversible and irreversible injury throughout infarct 35. Selvanayagam JB, Porto I, Channon K, et al. Troponin elevation after
healing. J Am Coll Cardiol. 2000;36:1985–1991. percutaneous coronary intervention directly represents the extent of
18. Rehwald WG, Fieno DS, Chen EL, Kim RJ, Judd RM. Myocardial irreversible myocardial injury: insights from cardiovascular magnetic
magnetic resonance imaging contrast agent concentrations after resonance imaging. Circulation. 2005;111:1027–1032.
250.e2 SECTION II  Ischemic Heart Disease

36. Kim RJ, Albert TS, Wible JH, et al; Gadoversetamide Myocardial 55. Carlsson M, Ubachs JF, Hedstrom E, Heiberg E, Jovinge S, Arheden H.
Infarction Imaging Investigators. Performance of delayed-enhancement Myocardium at risk after acute infarction in humans on cardiac
magnetic resonance imaging with gadoversetamide contrast for the magnetic resonance: quantitative assessment during follow-up and
detection and assessment of myocardial infarction: an international, validation with single-photon emission computed tomography. JACC
multicenter, double-blinded, randomized trial. Circulation. Cardiovasc Imaging. 2009;2:569–576.
2008;117:629–637. 56. Croisille P, Kim HW, Kim RJ. Controversies in cardiovascular MR
37. Kloner RA, Ganote CE, Jennings RB. The “no-reflow” phenomenon after imaging: T2-weighted imaging should not be used to delineate the area
temporary coronary occlusion in the dog. J Clin Invest. 1974;54: at risk in ischemic myocardial injury. Radiology. 2012;265:12–22.
1496–1508. 57. Arai AE, Leung S, Kellman P. Controversies in cardiovascular MR
38. Niccoli G, Scalone G, Lerman A, Crea F. Coronary microvascular imaging: reasons why imaging myocardial T2 has clinical and
obstruction in acute myocardial infarction. Eur Heart J. 2016;37: pathophysiologic value in acute myocardial infarction. Radiology.
1024–1033. 2012;265:23–32.
39. Reffelmann T, Kloner RA. The “no-reflow” phenomenon: basic science 58. Fernandez-Jimenez R, Garcia-Prieto J, Sanchez-Gonzalez J, et al.
and clinical correlates. Heart. 2002;87:162–168. Pathophysiology underlying the bimodal edema phenomenon after
40. Judd RM, Lugo-Olivieri CH, Arai M, et al. Physiological basis of myocardial ischemia/reperfusion. J Am Coll Cardiol. 2015;66:816–828.
myocardial contrast enhancement in fast magnetic resonance images of 59. Fernandez-Jimenez R, Sanchez-Gonzalez J, Aguero J, et al. Myocardial
2-day-old reperfused canine infarcts. Circulation. 1995;92:1902–1910. edema after ischemia/reperfusion is not stable and follows a bimodal
41. Lima JA, Judd RM, Bazille A, Schulman SP, Atalar E, Zerhouni EA. pattern: imaging and histological tissue characterization. J Am Coll
Regional heterogeneity of human myocardial infarcts demonstrated by Cardiol. 2015;65:315–323.
contrast-enhanced MRI. Potential mechanisms. Circulation. 1995;92: 60. Smulders MW, Bekkers SC, Kim HW, Van Assche LM, Parker MA, Kim
1117–1125. RJ. Performance of CMR methods for differentiating acute from chronic
42. Wu KC, Zerhouni EA, Judd RM, et al. Prognostic significance of MI. JACC Cardiovasc Imaging. 2015;8:669–679.
microvascular obstruction by magnetic resonance imaging in patients 61. Arai AE. Healing after myocardial infarction: a loosely defined process.
with acute myocardial infarction. Circulation. 1998;97:765–772. JACC Cardiovasc Imaging. 2015;8:680–683.
43. Messroghli DR, Moon JC, Ferreira VM, et al. Clinical recommendations 62. Kellman P, Aletras AH, Mancini C, McVeigh ER, Arai AE. T2-prepared
for cardiovascular magnetic resonance mapping of T1, T2, T2* and SSFP improves diagnostic confidence in edema imaging in acute
extracellular volume: a consensus statement by the Society for myocardial infarction compared to turbo spin echo. Magn Reson Med.
Cardiovascular Magnetic Resonance (SCMR) endorsed by the European 2007;57:891–897.
Association for Cardiovascular Imaging (EACVI). J Cardiovasc Magn 63. Aletras AH, Kellman P, Derbyshire JA, Arai AE. ACUT2E TSE-SSFP: a
Reson. 2017;19:75. hybrid method for T2-weighted imaging of edema in the heart. Magn
44. Hillenbrand HB, Kim RJ, Parker MA, Fieno DS, Judd RM. Early Reson Med. 2008;59:229–235.
assessment of myocardial salvage by contrast-enhanced magnetic 64. Berry C, Kellman P, Mancini C, et al. Magnetic resonance imaging
resonance imaging. Circulation. 2000;102:1678–1683. delineates the ischemic area at risk and myocardial salvage in patients with
45. Reimer KA, Lowe JE, Rasmussen MM, Jennings RB. The wavefront acute myocardial infarction. Circ Cardiovasc Imaging. 2010;3:527–535.
phenomenon of ischemic cell death. 1. Myocardial infarct size vs 65. Payne AR, Berry C, Kellman P, et al. Bright-blood T(2)-weighted MRI
duration of coronary occlusion in dogs. Circulation. 1977;56:786–794. has high diagnostic accuracy for myocardial hemorrhage in myocardial
46. Ortiz-Perez JT, Meyers SN, Lee DC, et al. Angiographic estimates of infarction: a preclinical validation study in swine. Circ Cardiovasc
myocardium at risk during acute myocardial infarction: validation study Imaging. 2011;4:738–745.
using cardiac magnetic resonance imaging. Eur Heart J. 2007;28: 66. Fernandez-Jimenez R, Sanchez-Gonzalez J, Aguero J, et al. Fast T2
1750–1758. gradient-spin-echo (T2-GraSE) mapping for myocardial edema
47. Wright J, Adriaenssens T, Dymarkowski S, Desmet W, Bogaert J. quantification: first in vivo validation in a porcine model of ischemia/
Quantification of myocardial area at risk with T2-weighted CMR: reperfusion. J Cardiovasc Magn Reson. 2015;17:92.
comparison with contrast-enhanced CMR and coronary angiography. 67. Giri S, Shah S, Xue H, et al. Myocardial T(2) mapping with respiratory
JACC Cardiovasc Imaging. 2009;2:825–831. navigator and automatic nonrigid motion correction. Magn Reson Med.
48. Fuernau G, Eitel I, Franke V, et al. Myocardium at risk in ST-segment 2012;68:1570–1578.
elevation myocardial infarction comparison of T2-weighted edema 68. Bulluck H, White SK, Frohlich GM, et al. Quantifying the area at risk in
imaging with the MR-assessed endocardial surface area and validation reperfused ST-segment-elevation myocardial infarction patients using
against angiographic scoring. JACC Cardiovasc Imaging. 2011;4:967–976. hybrid cardiac positron emission tomography-magnetic resonance
49. Giri S, Chung YC, Merchant A, et al. T2 quantification for improved imaging. Circ Cardiovasc Imaging. 2016;9:e003900.
detection of myocardial edema. J Cardiovasc Magn Reson. 2009;11:56. 69. Messroghli DR, Radjenovic A, Kozerke S, Higgins DM, Sivananthan MU,
50. Hammer-Hansen S, Leung SW, Hsu LY, et al. Early gadolinium Ridgway JP. Modified Look-Locker inversion recovery (MOLLI) for
enhancement for determination of area at risk: a preclinical validation high-resolution T1 mapping of the heart. Magn Reson Med. 2004;52:
study. JACC Cardiovasc Imaging. 2017;10:130–139. 141–146.
51. Matsumoto H, Matsuda T, Miyamoto K, Shimada T, Mikuri M, Hiraoka 70. Piechnik SK, Ferreira VM, Dall’Armellina E, et al. Shortened modified
Y. Peri-infarct zone on early contrast-enhanced CMR imaging in Look-Locker inversion recovery (ShMOLLI) for clinical myocardial
patients with acute myocardial infarction. JACC Cardiovasc Imaging. T1-mapping at 1.5 and 3 T within a 9 heartbeat breathhold. J Cardiovasc
2011;4:610–618. Magn Reson. 2010;12:69.
52. Jablonowski R, Engblom H, Kanski M, et al. Contrast-enhanced CMR 71. Dall’Armellina E, Karia N, Lindsay AC, et al. Dynamic changes of edema
overestimates early myocardial infarct size: mechanistic insights using and late gadolinium enhancement after acute myocardial infarction and
ECV measurements on day 1 and day 7. JACC Cardiovasc Imaging. their relationship to functional recovery and salvage index. Circ
2015;8:1379–1389. Cardiovasc Imaging. 2011;4:228–236.
53. Ugander M, Bagi PS, Oki AJ, et al. Myocardial edema as detected by 72. Liu D, Borlotti A, Viliani D, et al. CMR native T1 mapping allows
pre-contrast T1 and T2 CMR delineates area at risk associated with differentiation of reversible versus irreversible myocardial damage in
acute myocardial infarction. JACC Cardiovasc Imaging. 2012;5:596–603. ST-segment-elevation myocardial infarction: an OxAMI study (Oxford
54. Tilak GS, Hsu LY, Hoyt RF Jr, Arai AE, Aletras AH. In vivo T2-weighted Acute Myocardial Infarction). Circ Cardiovasc Imaging. 2017;10.
magnetic resonance imaging can accurately determine the ischemic area 73. Ugander M, Oki AJ, Hsu LY, et al. Extracellular volume imaging by
at risk for 2-day-old nonreperfused myocardial infarction. Invest Radiol. magnetic resonance imaging provides insights into overt and sub-
2008;43:7–15. clinical myocardial pathology. Eur Heart J. 2012;33:1268–1278.
CHAPTER 19  AMI: Cardiovascular Magnetic Resonance Detection and Characterization 250.e3

74. Garg P, Broadbent DA, Swoboda PP, et al. Acute infarct extracellular 90. Wu E, Ortiz JT, Tejedor P, et al. Infarct size by contrast enhanced cardiac
volume mapping to quantify myocardial area at risk and chronic infarct magnetic resonance is a stronger predictor of outcomes than left
size on cardiovascular magnetic resonance imaging. Circ Cardiovasc ventricular ejection fraction or end-systolic volume index: prospective
Imaging. 2017;10. cohort study. Heart. 2008;94:730–736.
75. Matsumoto H, Matsuda T, Miyamoto K, et al. Temporal change of 91. de Waha S, Desch S, Eitel I, et al. Relationship and prognostic value of
enhancement after gadolinium injection on contrast-enhanced CMR in microvascular obstruction and infarct size in ST-elevation myocardial
reperfused acute myocardial infarction. J Cardiol. 2015;65:76–81. infarction as visualized by magnetic resonance imaging. Clin Res Cardiol.
76. Hammer-Hansen S, Bandettini WP, Hsu LY, et al. Mechanisms for 2012;101:487–495.
overestimating acute myocardial infarct size with gadolinium-enhanced 92. Eitel I, Desch S, Fuernau G, et al. Prognostic significance and
cardiovascular magnetic resonance imaging in humans: a quantitative determinants of myocardial salvage assessed by cardiovascular magnetic
and kinetic study. Eur Heart J Cardiovasc Imaging. 2016;17:76–84. resonance in acute reperfused myocardial infarction. J Am Coll Cardiol.
77. Nordlund D, Kanski M, Jablonowski R, et al. Experimental validation of 2010;55:2470–2479.
contrast-enhanced SSFP cine CMR for quantification of myocardium at 93. Grothoff M, Elpert C, Hoffmann J, et al. Right ventricular injury in
risk in acute myocardial infarction. J Cardiovasc Magn Reson. 2017;19:12. ST-elevation myocardial infarction: risk stratification by visualization of
78. Tufvesson J, Carlsson M, Aletras AH, et al. Automatic segmentation of wall motion, edema, and delayed-enhancement cardiac magnetic
myocardium at risk from contrast enhanced SSFP CMR: validation resonance. Circ Cardiovasc Imaging. 2012;5:60–68.
against expert readers and SPECT. BMC Med Imaging. 2016;16:19. 94. Jensen CJ, Jochims M, Hunold P, Sabin GV, Schlosser T, Bruder O. Right
79. Nordlund D, Klug G, Heiberg E, et al. Multi-vendor, multicentre ventricular involvement in acute left ventricular myocardial infarction:
comparison of contrast-enhanced SSFP and T2-STIR CMR for prognostic implications of MRI findings. AJR Am J Roentgenol.
determining myocardium at risk in ST-elevation myocardial infarction. 2010;194:592–598.
Eur Heart J Cardiovasc Imaging. 2016;17:744–753. 95. Miszalski-Jamka T, Klimeczek P, Tomala M, et al. Extent of RV
80. Engblom H, Heiberg E, Erlinge D, et al. Sample size in clinical dysfunction and myocardial infarction assessed by CMR are
cardioprotection trials using myocardial salvage index, infarct size, or independent outcome predictors early after STEMI treated with primary
biochemical markers as endpoint. J Am Heart Assoc. 2016;5:e002708. angioplasty. JACC Cardiovasc Imaging. 2010;3:1237–1246.
81. Klem I, Heiberg E, Van Assche L, et al. Sources of variability in 96. Weinsaft JW, Kim HW, Shah DJ, et al. Detection of left ventricular
quantification of cardiovascular magnetic resonance infarct size— thrombus by delayed-enhancement cardiovascular magnetic resonance
reproducibility among three core laboratories. J Cardiovasc Magn Reson. prevalence and markers in patients with systolic dysfunction. J Am Coll
2017;19:62. Cardiol. 2008;52:148–157.
82. Ahn KT, Song YB, Choe YH, et al. Impact of transmural necrosis on left 97. Srichai MB, Junor C, Rodriguez LL, et al. Clinical, imaging, and
ventricular remodeling and clinical outcomes in patients undergoing pathological characteristics of left ventricular thrombus: a comparison
primary percutaneous coronary intervention for ST-segment elevation of contrast-enhanced magnetic resonance imaging, transthoracic
myocardial infarction. Int J Cardiovasc Imaging. 2013;29:835–842. echocardiography, and transesophageal echocardiography with surgical
83. Bodi V, Husser O, Sanchis J, et al. Contractile reserve and extent of or pathological validation. Am Heart J. 2006;152:75–84.
transmural necrosis in the setting of myocardial stunning: comparison 98. von Knobelsdorff-Brenkenhoff F, Schulz-Menger J. Role of
at cardiac MR imaging. Radiology. 2010;255:755–763. cardiovascular magnetic resonance in the guidelines of the European
84. Bodi V, Sanchis J, Lopez-Lereu MP, et al. Prognostic and therapeutic Society of Cardiology. J Cardiovasc Magn Reson. 2016;18:6.
implications of dipyridamole stress cardiovascular magnetic resonance 99. Gerber BL, Belge B, Legros GJ, et al. Characterization of acute and
on the basis of the ischaemic cascade. Heart. 2009;95:49–55. chronic myocardial infarcts by multidetector computed tomography:
85. Cochet AA, Lorgis L, Lalande A, et al. Major prognostic impact of comparison with contrast-enhanced magnetic resonance. Circulation.
persistent microvascular obstruction as assessed by contrast-enhanced 2006;113:823–833.
cardiac magnetic resonance in reperfused acute myocardial infarction. 100. Kumar A, Abdel-Aty H, Kriedemann I, et al. Contrast-enhanced
Eur Radiol. 2009;19:2117–2126. cardiovascular magnetic resonance imaging of right ventricular
86. de Waha S, Desch S, Eitel I, et al. Impact of early vs. late microvascular infarction. J Am Coll Cardiol. 2006;48:1969–1976.
obstruction assessed by magnetic resonance imaging on long-term 101. Plein S, Younger JF, Sparrow P, Ridgway JP, Ball SG, Greenwood JP.
outcome after ST-elevation myocardial infarction: a comparison with Cardiovascular magnetic resonance of scar and ischemia burden early
traditional prognostic markers. Eur Heart J. 2010;31:2660–2668. after acute ST elevation and non-ST elevation myocardial infarction. J
87. Hombach V, Grebe O, Merkle N, et al. Sequelae of acute myocardial Cardiovasc Magn Reson. 2008;10:47.
infarction regarding cardiac structure and function and their prognostic 102. Bruder O, Breuckmann F, Jensen C, et al. Prognostic impact of
significance as assessed by magnetic resonance imaging. Eur Heart J. contrast-enhanced CMR early after acute ST segment elevation
2005;26:549–557. myocardial infarction (STEMI) in a regional STEMI network: results of
88. Larose E, Rodes-Cabau J, Pibarot P, et al. Predicting late myocardial the “Herzinfarktverbund Essen.” Herz. 2008;33:136–142.
recovery and outcomes in the early hours of ST-segment elevation 103. Amabile N, Jacquier A, Shuhab A, et al. Incidence, predictors, and
myocardial infarction traditional measures compared with prognostic value of intramyocardial hemorrhage lesions in ST
microvascular obstruction, salvaged myocardium, and necrosis elevation myocardial infarction. Catheter Cardiovasc Interv. 2012;79:
characteristics by cardiovascular magnetic resonance. J Am Coll Cardiol. 1101–1108.
2010;55:2459–2469. 104. de Waha S, Eitel I, Desch S, et al. Time-dependency, predictors and
89. Lønborg J, Vejlstrup N, Kelbaek H, et al. Final infarct size measured by clinical impact of infarct transmurality assessed by magnetic resonance
cardiovascular magnetic resonance in patients with ST elevation imaging in patients with ST-elevation myocardial infarction reperfused
myocardial infarction predicts long-term clinical outcome: an by primary coronary percutaneous intervention. Clin Res Cardiol.
observational study. Eur Heart J Cardiovasc Imaging. 2013;14:387–395. 2012;101:191–200.
20 
Acute Myocardial Infarction:
Ventricular Remodeling
David Lopez and Christopher M. Kramer

Approximately 550,000 Americans will suffer an acute myocardial infarc- radionuclide techniques expose patients to potentially harmful ionizing
tion (AMI) each year.1 Despite major advances in AMI therapies, mortality radiation, making their routine repeated use in humans less desirable.
and incident heart failure (HF) remain significant problems. Five-year Echocardiography is limited in a subset of patients by the availability
mortality is estimated at 36% in men and 47% in women.1 The five-year of acoustic windows for proper endocardial definition. Finally, two-
incidence of HF ranges from 16% in men to 22% in women.1 These dimensional (2D) modalities, such as x-ray left ventriculography and
adverse outcomes have been unequivocally linked to the development echocardiography, rely on geometric assumptions to calculate ventricu-
of significant left ventricular (LV) dilation and reduced systolic func- lar volumes that may not apply in hearts regionally deformed by MI.
tion after AMI, otherwise known as post-MI negative LV remodeling. Cardiovascular magnetic resonance (CMR) is a technique that
LV remodeling refers to structural and functional myocardial changes can overcome many of the limitations mentioned above. As a result
that can occur in response to physiologic stress such as exercise or it has emerged as a valuable noninvasive modality for the assessment
because of a pathologic insult such as AMI. Postinfarction LV remodel- of cardiac disease. CMR provides a comprehensive, volumetric, accu-
ing is an intricate cascade of biologic events set forth by the release of rate, and reproducible cardiovascular evaluation beyond ventricular
intracellular chemokines from necrotic myocytes and increased wall volumes. With the use of CMR it is possible to characterize the myocar-
stress due to infarct region contractile and diastolic impairment.2 Remod- dial tissue for the localization and quantification of myocardial edema,
eling is a healing and compensatory process that restricts myocardial microvascular obstruction (MVO), intramyocardial hemorrhage, and
damage while maintaining cardiac output. infarct burden. Hence, CMR is particularly well suited for the evalua-
Multiple studies have demonstrated different patterns of post-MI tion of post-MI remodeling. In this chapter we will review the research
remodeling with varying degrees of LV dilation. LV dilation may be and clinical utility of CMR in the evaluation of post-MI ventricular
transient, limited or progressive.3–5 Differences in the onset of dila- remodeling,
tion have been observed. It can occur acutely (within 10 days) and/
or later during the healing process.4,5 Progressive dilation has been VENTRICULAR VOLUMES, EJECTION
observed even years after MI.6 Controlled ventricular remodeling is
ultimately achieved by the formation of a strong collagen scar and
FRACTION, AND MASS
remote myocardium hypertrophy which counteract intracavitary LV cavity dilation and systolic dysfunction are the hallmarks of postin-
forces, thereby limiting progressive ventricular dilation.2 In this case, farct remodeling and can be accurately quantified with high spatial and
ventricular remodeling is compensatory. However, in many instances, temporal resolution using cine CMR. Early breath-hold cine imaging
particularly with large transmural infarcts, the remodeling process is used fast low-angle shot (FLASH), a spoiled gradient recalled echo
overwhelmed by the increased LV wall stress.7 Ongoing dilation and (GRE) sequence. Currently, balanced steady-state free precession (bSSFP)
remote contractile impairment ensues8,9 (Fig. 20.1) in a process known is the most commonly used pulse sequence because of faster acquisition
as negative postinfarction LV remodeling, which increases risk of HF times, improved temporal resolution, and optimal contrast between
and mortality. the myocardium and the blood pool.15,16 The use of bSSFP facilitates
The prognostic significance of negative post-MI remodeling has qualitative and quantitative assessment of cardiac anatomy and function
been demonstrated with the use of various imaging modalities, includ- mainly by improving endocardial border definition.
ing chest radiographs,3 invasive x-ray left ventriculography,10 radionuclide Unlike echocardiography, acoustic windows do not limit CMR;
ventriculography,9 and echocardiography.6,11–13 In 1973 Kostuk and col- therefore images can be acquired in any desired orientation to accurately
leagues showed various patterns of LV remodeling and the association depict LV morphology and function. A carefully prescribed stack of
between the extent of negative remodeling and clinical outcomes by short-axis slices extending from the cardiac base to the apex provides
measuring serial left heart dimensions using calibrated chest radio- a three-dimensional (3D) dataset that can be used to make accurate
graphs.3 Subsequent investigations established LV ejection fraction (EF) ventricular morphologic measurements at end diastole (ED) and end
as a strong predictor of survival after AMI.11,14 By 1987 White and systole (ES) without the need for geometric assumptions, as is the case
colleagues found that LV end-systolic volume (ESV) measured by x-ray with 2D modalities (Fig. 20.2). Ventricular volumes are calculated using
left ventriculography 1 to 2 months after MI was a more powerful the summation of disks method.
predictor of survival than end-diastolic volume (EDV) and LVEF.10 CMR derived LV volumes and mass have been validated with
Although these modalities have laid the foundation of our understand- phantom, animal and human studies17–19 and currently considered the
ing of post-MI LV remodeling, their utility has been hindered by technical clinical reference standard for other imaging modalities.20 Although
and safety limitations. Chest radiographs are insensitive to RV and LV accuracy is important, reliability may be even more important because
volumes and systolic function. Invasive x-ray left ventriculography and it dictates the validity of differences between serial examinations.

251
252 SECTION II  Ischemic Heart Disease

FIG. 20.1  Representative cine images from a swine model of percutaneous left anterior descending artery
90-minute occlusion followed by reperfusion illustrate negative ventricular remodeling 8 weeks after infarc-
tion. Note the anterior infarct region wall thinning and akinesis (arrows). ED, End diastole; ES, end systole.

Volumetric measurements by CMR provide high-interstudy, interscan, were included in the study. Infarct transmurality and size were defined
and interobserver reproducibility in normal hearts and those deformed by clinical, electrocardiographic, and enzymatic criteria. The extent of
by prior MI.21–23 Interstudy, interobserver, and intraobserver reproduc- negative remodeling was not only dependent on the enzymatic size but
ibility of CMR-derived RV volumes is also excellent in healthy controls, also on the location of the infarct. Small infarcts caused modest but
patients with HF, ventricular hypertrophy, and congenital heart disease.24,25 concordant ED and ES dilation, so that LVEF remained stable. Individu-
Because of improved reproducibility compared with echocardiography, als with large anterior infarcts developed an unbalanced, progressive
CMR facilitates the identification of clinically significant ventricular diastolic and systolic dilation that resulted in a marked reduction in
changes in serial examinations after AMI. The reduced variability of LVEF. Infarct region thinning was almost always observed at 6 months
the measurement significantly reduces the sample size needed to detect except for small posterior infarcts. LV stroke volume index (SVI) was
differences between treatment arms in clinical trials.26 smaller in large anterior MI, but there was no statistically significant
Cine CMR has been successfully used to evaluate post-MI remodel- difference in LV SVI between 1 and 26 weeks post-MI. Despite a sig-
ing in animal models and humans. Konermann and colleagues27 studied nificant LVEF reduction in those with large infarcts, stroke volume was
the reliability of CMR in the evaluation of LV morphology and function maintained at the expense of ventricular dilation. No clinical difference
in post-MI patients. They compared cine CMR LV volumes and LVEF between the groups was observed when analyzing New York Heart
with 2D echocardiography (2DE), radionuclide, and x-ray left ventricu- Association (NYHA) symptom classification. Four patients died before
lography in a group of 65 patients who suffered a transmural MI and the completion of the study and all had large anterior MI.
did not undergo thrombolysis or angioplasty. Good correlation was In a cohort of 26 patients with reperfused anterior AMI, Kramer
found between x-ray and radionuclide ventriculography. However, cor- et al.28 used cine tissue tagging to evaluate morphologic changes, LVEF,
relation with 2D echocardiography was limited because of inconsistent and regional intramyocardial function of myocardium adjacent and
2DE image quality. In this study, the investigators also demonstrated remote to the infarct. All patients received either angiotensin-converting
good correlation between creatine kinase (CK)-derived infarct size and enzyme inhibitor (ACEI) or beta-blocker. Most patients received ACEI
CMR infarct mass determined from cine images. and about half received both agents. Imaging was done on day 5 ± 2
In a follow-up publication, Konermann et al.7 reported the natural and week 8 ± 1 after MI. There was a significant increase in LV end-
history of LV remodeling in the same group of 65 patients at 1, 4, and diastolic volume index (EDVI) with stable end-systolic volume index
26 weeks post-MI. Medical therapy was limited to nitrates, beta-blockers, (ESVI). Therefore there was an improvement in global LVEF 8 weeks
aspirin, and diuretics. Only those patients with first transmural infarcts post-MI. The increased LVEF was mediated by contractile improvement
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 253

A B

C D
FIG. 20.2  A carefully prescribed stack of short-axis slices extending from the cardiac base to the apex
(A) provides a three-dimensional dataset (B and D) that can be used to make accurate left ventricular (LV)
morphologic measurements at end diastole (ED) and end systole (ES) without the need for geometric
assumptions. It is important to account for in-plane motion of the mitral annulus (C) to avoid overestimation
of the end-systolic volume and underestimation of the LV ejection fraction.

and normalization in the adjacent and remote regions, respectively. heart can be determined by measuring strain with the use of myocardial
Enzymatic infarct size correlated with the degree of diastolic dilation. tissue tagging sequences such as spatial modulation of magnetization
(SPAMM)30 and phase-based sequences such as displacement encod-
ing with stimulated echoes (DENSE).31 More recently, feature tracking
REGIONAL CONTRACTILITY analysis has allowed the measurement of circumferential and longitudinal
Although LVEF is the most commonly used global metric of LV systolic strain from bSSFP cine images.32,33
function, it does not provide information regarding regional perfor-
mance. In fact, segmental systolic abnormalities because of AMI may Wall Thickening and Endocardial Displacement: Cine
sometimes be concealed by a normal global LVEF. Accordingly, to under- Cardiovascular Magnetic Resonance
stand the contributing factors to global functional alterations during the In their chronicle of nonreperfused AMI, Konermann et al.7 assessed
remodeling process, it is important to characterize regional contractile regional LV contractility by measuring wall thickening and the endo-
performance in the adjacent and remote noninfarcted myocardium. cardial motion toward the center of the LV cavity using cine CMR.
In clinical practice this is usually done by subjective visual scoring of Progressive changes in endocardial motion were identified in the infarct
segmental endocardial motion and thickening using cine CMR images. and remote regions during the 6-month follow-up period. The extent
However, visual assessment can be insensitive to subtle abnormalities and of regional LV dysfunction correlated with infarct size and location.
is subject to greater interobserver variability. Thus objective methods to The infarct region of large anterior AMI had the greatest reduction in
quantify contractile function have been developed. Wall thickening and endocardial motion, which progressed to dyskinesis (displacement away
endocardial motion can be measured from cine CMR images. A more from the cavity center) at 6 months. Changes in wall thickening were
accurate29 assessment of intramyocardial contractility in the infarcted not observed in the viable myocardium. It is worth noting that the
254 SECTION II  Ischemic Heart Disease

authors averaged the adjacent and remote myocardium wall thickness reperfusion of a first anterior MI compared with 31 age-matched con-
in their analysis. Hence, regional differences could not be identified trols.39 All patients had received optimal medical therapy with beta-
between these segments. blockers and ACEIs. Imaging was performed 1 and 12 weeks postinfarction.
Holman et al.34 investigated the regional contractility of 25 patients The LV myocardium was divided into 32 cuboid segments, defined by
3 weeks after anterior MI. An optimized 3D analysis of the myocardium 4 endocardial and 4 epicardial node points derived from short- and
centerline was implemented to improve accuracy of wall thickness long-axis tagged images. Regional LVEF was calculated using a pie-
measurement. Some 100 equidistant chords were constructed between shaped volume defined by the endocardium and the center of the LV.
the endocardial and epicardial contours of short-axis cine images. The This parameter was viewed as a composite marker of regional deforma-
starting point was defined as the inferior RV insertion site, which was tion. A relative metric of regional load was defined as the product of
labeled for ED and ES, allowing to correct for rotational motion. Wall the systolic blood pressure and the mean radius of curvature in the
thickening was reduced in the left anterior descending (LAD) artery short and long axes divided by the segment wall thickness. In healthy
territory compared with a normal database. The dysfunctional LV myo- controls, an inverse relationship between regional LVEF and load was
cardial mass 3 weeks after MI, when any myocardial stunning should demonstrated. In contrast for the MI patients, the average load values
have resolved, correlated with enzymatic infarct size. significantly increased in a graded fashion from remote to infarct seg-
ments between the acute and chronic time points. At 12 weeks, remote
Strain Imaging regional LVEF decreased with a similar correlation coefficient to changes
Tissue Tagging in load as controls; therefore the change in LVEF was because of an
CMR strain imaging in animal and human studies has contributed increase in loading conditions and a worsening of myocardial function.
greatly to our understanding of regional myocardial function during No change in regional LVEF was observed in the adjacent segments,
the post-MI remodeling process. In an ovine model of surgical LAD indicating some improvement in myocardial function. The infarct seg-
ligation,35 SPAMM myocardial tissue tagging was used to measure cir- ments demonstrated an average increase in regional LVEF even with
cumferential and longitudinal myocardial shortening at baseline, 1 week, significant increases in load representing an even greater true improve-
8 weeks, and 6 months after AMI. Shortening within infarcted regions ment in myocardial function. This study demonstrated that CMR could
was reduced throughout the study period. A persistent difference in be used to assess LV function post-MI and differentiate improvement
intramyocardial shortening was found between noninfarcted regions in intrinsic myocyte function from changes in deformation that occur
adjacent to and remote from the infarct border. Function in adjacent as a response to alterations in Frank-Starling conditions.
noninfarcted regions fell markedly at 1 week after AMI and partially
improved by 8 weeks, but remained depressed relative to baseline and to Phase-Based Strain Imaging
remote regions at 6 months. These findings were reproduced by Moulton In 1999 advanced, phase-based, strain imaging techniques were intro-
et al.36 using a similar ovine model and DANTE (delays alternating with duced to facilitate and optimize strain analysis compared with tissue
nutations for tailored excitation) myocardial tagging. In addition to tagging methods. These included harmonic phase magnetic resonance
systolic strain, Moulton et al. evaluated diastolic and isovolemic strains. (HARP)40 and DENSE.31 These methods were validated with traditional
They found abnormal, positive strain during isovolemic contraction in tagging sequences in animal and human studies.41–45
the border zone, suggesting that isovolemic myocardial fiber stretching Azevedo et al.46 used tissue tagging with HARP analysis in combina-
contributes to LV systolic dysfunction in these segments. tion with contrast-enhanced (CE)-CMR and radioactive microspheres
Epstein et al.37 used myocardial tissue tagging to characterize acute to evaluate the relationship between strain properties and myocardial
regional function in a murine model of LAD occlusion and reperfusion. injury in a canine model of 90-minute LAD or left circumflex (LCX)
The percent circumferential shortening was measured for the infarct, occlusion followed by 24 hours of reperfusion. In this early period the
adjacent and remote regions. A gradient of contractile dysfunction was areas at risk as defined by radioactive microspheres with preserved
found from the infarct to the remote region 1 day post-AMI compared systolic strain and strain rate demonstrated significantly reduced diastolic
with baseline. Thomas et al.37a used tissue tagging to characterize regional strain rate compared with the remote regions. Similar to the findings
contractile changes in a rat model of LAD ligation at 1 to 2, 3 to 4, 6 of Gerber et al.,47 infarct segments with MVO demonstrated even greater
to 8, and 9 to 12 weeks post-AMI. In this study, changes in the maximum reduction in systolic and diastolic strain than those without.
and minimum principal stretches and strains, and the orientation of In a canine model of reperfused LAD, Ashikaga et al.48 used DENSE
the principal stretch angle were measured. At 1 to 2 weeks post-AMI, to generate 3D-displacement maps, CE CMR imaging to generate infarct
significant changes were found in a gradient fashion from the infarct maps, and epicardial electrical recordings to generate electrical activa-
to the remote region. These abnormalities persisted relatively unchanged tion maps. CMR examinations were completed between 3 and 8 weeks
up to 12 weeks post-MI. The principal strain direction became more post-AMI. Electrical activation times were significantly delayed in the
circumferentially oriented during the study period. infarct zone, but preserved in the border and remote areas. Conversely,
Using CMR tissue tagging in a cohort of patients within the first all strain parameters were impaired in the border zone, which was not
week after reperfused LAD AMI without significant disease in other different from the infarct region. Hence, contractile abnormalities in
coronary territories and with an EF of <50%, Kramer et al.38 found a the border segments were not mediated by impaired electrical activa-
significant reduction in remote region intramyocardial shortening com- tion. In the remote segments there was a typical42 transmural strain
pared with controls. Once again, there was a decrease in contractile gradient from subendocardium to subepicardium, which was not
function in all myocardial segments in a gradient fashion similar to the observed in the infarct or border zone regions. Positive circumferential
findings in animal studies. When this patient group was re-imaged at and longitudinal strains were observed in the border zone indicative
8 weeks after AMI, there was improvement in regional function in all of abnormal systolic stretch.
segments, including normalization of contractile function in the remote In a canine model that typically created subendocardial infarcts
myocardium.28 The impact of early reperfusion is evident by the improve- with large areas at risk, recovery of systolic function was evaluated
ment in infarct region strain. with DENSE imaging and correlated to T2-weighted (T2W) imaging
CMR myocardial tagging was used to assess the correlation between representing the area at risk.49 T2W area at risk was validated with
regional function and loading conditions in 16 patients after successful microsphere measurements. At 2 months post-AMI there was resolution
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 255

of the T2W abnormalities representing the area at risk. This correlated anisotropy returned to baseline 3 weeks after the injury; however, fiber
with a significant improvement in radial and circumferential strain architecture remained abnormal, albeit relatively preserved compared
compared with the acute post-MI setting. However, contractile func- with infarcted hearts.
tion in the area at risk remained significantly depressed compared with
remote sectors.
TISSUE CHARACTERIZATION
Feature Tracking To thoroughly understand post-MI remodeling, it is crucial to precisely
More recently anatomical feature tracking of cine bSSFP images or characterize the location, size, transmural extent, and type of ischemic
feature tracking CMR has been introduced as a practical solution for injury. In the event of epicardial vessel occlusion, the myocardium sub-
strain analysis.32 Although the information obtained via this method tended by the vessel segments distal to the occlusion is at risk of infarc-
is mostly limited to the endocardium and epicardium, it still provides tion. This territory is known as the area at risk. A sufficiently prolonged
relevant and comparable information when compared with traditional occlusion leads to irreversible injury, or infarction, of the entire area
tissue tagging.33 Its application to post-MI remodeling has been limit- at risk. If a vessel is opened before the occurrence of permanent injury,
ed33a; however, given its ease of use, it is likely to rapidly gain popularity a percentage of the area at risk is salvaged. The salvaged myocardium
as it has in many other disease states. exhibits reversible contractile dysfunction, or stunning. A more deleteri-
ous form of injury can occur within the infarct core because of persistent
MYOCARDIAL FIBER STRUCTURE: DIFFUSION ischemia or “no-reflow” caused by MVO. In a number of infarcts with
MVO, intramyocardial hemorrhage may also occur. CMR stands out
TENSOR MAGNETIC RESONANCE IMAGING from other imaging modalities because of its superior tissue charac-
An evolving technique to study post-MI remodeling is diffusion- terization capability. With the use of various noncontrast and contrast-
weighted magnetic resonance imaging (MRI). The ability to visualize enhanced CMR sequences, we can identify and quantify the area at
and characterize the myocardial fiber structure is important to better risk, areas of irreversible infarction, salvaged myocardium, MVO, and
understanding cardiomyopathies, including post-MI remodeling. Dif- intramyocardial hemorrhage, which are all important parameters that
fusion tensor imaging (DTI) is a form of diffusion-weighted CMR influence post-MI remodeling.
which has been validated against histology as a rapid and nondestructive
method to depict the myocardial fibers’ architecture.50,51 Early animal Infarct Characterization and Predictors of Left
and human studies have consistently shown that early after MI, the Ventricular Remodeling
infarct region is characterized by increased diffusivity and decreased Infarcted myocardium can be identified by various CMR pulse sequences,
fractional anisotropy.52–54 Investigators have also shown a difference including cine, CE-CMR, and, more recently, T1 mapping. Today,
in the distribution of right- and left-handed helix fibers between the CE-CMR, specifically LGE, is considered the reference standard for
infarct and remote regions. In a study of 37 patients imaged about acute and chronic infarct characterization.
4 weeks after their first MI, a transmural gradient of left- and right-
handed helical fibers was observed between the infarct and remote Late Gadolinium Enhancement
regions.52 In the infarct region the percent of right-handed fibers was LGE images are acquired at least 10 minutes after gadolinium (Gd)
lowest in association with a greater percent of left-handed fibers. Using administration using an inversion-recovery T1-weighted (T1W) pulse
3D DTI tractography, Sosnovik et al.53 performed in vivo imaging of sequence. Gd shortens the T1 relaxation of the surrounding protons;
mouse hearts 24 hours and 3 weeks after mid-LAD infarct or ischemia- therefore areas of increased Gd appear hyperintense on T1W imaging.
reperfusion injury. DTI imaging findings were compared with CMR Although Gd-based contrast media are extracellular, the loss of cell
and histologically determined infarct and area at risk regions. Areas membrane integrity and altered Gd wash-in and wash-out kinetics lead
of increased T2, representing tissue edema, demonstrated increased to increased Gd in acute infarcts.55,56 In chronic MI, Gd is increased
diffusivity and decreased fractional anisotropy as well as loss of fiber because of an increase in extracellular space, with formation of a col-
tracts. The loss of tract coherence was uniform across the area at risk and lagenous scar as well as alterations in contrast kinetics. Consequently,
included infarct and viable fibers when compared with late gadolinium both AMI and chronic MI will appear as bright myocardial regions on
enhancement (LGE) and histopathology. In the infarct group there was LGE imaging (Fig. 20.3).
persistent loss of fiber tracts 3 weeks post-MI. Therefore presence of LGE imaging precisely and reproducibly demonstrates acute and
collagen fibers did not restrict diffusion adequately to resolve tracts chronic infarcts in comparison to histology.55,57 This was best demon-
by DTI. In the ischemia-reperfusion group, diffusivity and fractional strated in an elegant canine study by Kim et al.55 in which acute infarcts

FIG. 20.3  Representative late gadolinium enhancement images from a swine model of left anterior descend-
ing artery occlusion and reperfusion. Note the increased wall thickness and hypoenhanced core 2 days
postinfarction, followed by infarct resorption/wall thinning at 8 weeks.
256 SECTION II  Ischemic Heart Disease

were created by coronary ligation. A second artery was instrumented to with prior MI (at least 3 months), Di Bella et al.68 found that LGE
create reversible ischemic injury. CMR examinations were performed >12.7% of LV mass, EDV >105 mL/m2, and a wall motion score index
at 1 day, 3 days, and 8 weeks postischemic injury. LGE images were >1.7 were independently associated with cardiac death or appropriate
acquired 30 minutes after Gd administration. Compared with histol- intracardiac defibrillator shocks. Patients with all three factors had a
ogy, the extent of LGE by ex vivo imaging was the same as the extent 4-year event rate of 29.6%, whereas those with none of these factors
of myocardial necrosis in the acute phase and the extent of collagenous had an event rate of 3.5%.
scar at 8 weeks after reperfused and nonreperfused ischemic injury.
Therefore areas of LGE specifically represent permanent myocardial No-Reflow Phenomenon: Microvascular Obstruction and
injury and not stunned or salvaged myocardium. It should be noted Intramyocardial Hemorrhage
that the accuracy of in vivo LGE is influenced by multiple factors, It has been shown in animal and clinical studies that opening an occluded
including imaging delay after contrast injection,56,58 spatial resolution,57 epicardial artery does not always result in homogeneous reperfusion
and postprocessing methodology.59 When a standardized approach is of the area at risk.69 This is known as the “no-reflow” phenomenon.
applied, LGE imaging reproducibly quantifies acute and chronic infarcts Areas of no-reflow can be detected in vivo by various modalities, includ-
with excellent interscan, intraobserver and interobserver agreement.60,61 ing angiography, single-photon emission computed tomography (SPECT),
This technique has been useful to define the evolution of infarct positron emission tomography (PET), and contrast echocardiography,
size in vivo. Rochitte et al.62 demonstrated an increase in infarct size but are best evaluated by CE-CMR. In the CMR literature, no-reflow
from 2 to 48 hours after reperfused 90-minute LAD occlusion in a zones have been referred to as areas of MVO. Areas of MVO are detected
canine model. However, the investigators did not report the changes by CE-CMR as hypoenhanced regions within the infarct core on dynamic
in wall thickness or circumferential expansion index to further char- first-pass perfusion imaging during Gd administration or on LGE imaging
acterize the pattern of infarct size growth in this very early phase after after Gd administration (Fig. 20.4). LGE imaging 2 to 5 minutes after
ischemic injury. They also observed up to 3-fold increase in the size contrast injection reveals so-called early MVO and imaging after 10
of MVO in the infarct core, which correlated with thioflavin-S myo- minutes reveals late MVO. It has been demonstrated that areas of early
cardial blood flow assessment. Fieno et al.63 assessed chronic infarct MVO tend to be larger than late MVO, which suggests that early MVO
remodeling in canines after 45-minute, 90-minute, or permanent LAD illustrates a penumbra of slow collateral flow in addition to true no-
occlusion. Imaging was performed on day 3, 4 weeks, and 8 weeks after reflow regions. Therefore early MVO is more sensitive, whereas late
MI. The infarct size at 8 weeks post-AMI decreased on average by 75% MVO is more specific for no-reflow. A recent systematic review shows
of the size on day 3, but as much as 90% in the case of small infarcts that the prevalence of early MVO is greater than that of late MVO (65%
caused by the 45-minute occlusion. True infarct expansion was not vs. 54%, respectively).70
observed. Although the overall myocardial mass decreased, the non- The mechanisms of the no-reflow phenomenon are incompletely
infarcted myocardium mass increased on average by 15%. The infarct understood but may include vasoconstriction mediated by inflammatory
involution findings were consistent with those of Richard et al.,64 who cells, external compression from surrounding edema, and microemboliza-
measured infarct size on pathologic specimens 4 days, 2 weeks, and tion of atherosclerotic cellular debris leading to small vessel plugging.69
6 weeks after 6 hours of reperfused or permanent circumflex occlu- Regardless of the exact mechanism, studies have demonstrated that this
sion in an open chest canine model. In both experiments, reperfusion phenomenon is mediated by reperfusion injury. Rochitte et al.62 and
appeared to accelerate infarct healing, but did not impact the final infarct Wu et al.71 showed that MVO size, as detected by in vivo CE-CMR and
size. Early infarct expansion and cardiac rupture were not observed in validated with thioflavin-S, peaks 48 hours after reperfusion and plateaus
these studies. from 2 to 9 days after reperfusion. Areas of MVO involute thereafter
Infarct involution has been demonstrated in humans as well. Ing- and are rarely seen on follow-up CMR 1 month or later after AMI.
kanisorn et al.65 evaluated infarct size within 5 days of an AMI in 33 The presence of any MVO has been repeatedly associated with
patients, 20 of whom returned for follow-up imaging more than 8 worse myocardial remodeling and clinical outcomes. This was first
weeks after MI. Acute infarct size correlated well with troponin-I. At demonstrated by Wu et al.,72 who found that early MVO predicts
follow-up, infarct size decreased from 16% to 11% of the LV mass. infarct transmurality, LV function, and remodeling after 16 months,
Choi et al.66 also reported a significant decrease in infarct size between and was also a powerful independent prognostic indicator of MACE
week 1 and week 8 post-MI in 25 patients after reperfused AMI. The even after controlling for infarct size. Gerber el al.47 studied the effect
involution of MI size tended to be greater in those with MVO. of MVO on myocardial strain and reported that the extent of MVO
LGE CMR has been recognized as a powerful predictor of LV remod- correlates with significant decreases in infarct region stretch and
eling, regional and global functional recovery, and major adverse cardiac reduced radial strain in the adjacent regions. Nijveldt et al.73 evaluated
events (MACE) following AMI. Choi et al.67 performed LGE on 24 the impact of early, mid, and late MVO in 63 patients who received
patients within 7 days of revascularization post-MI. Scans were repeated percutaneous coronary intervention (PCI) and optimal medical man-
at 8 to 12 weeks to assess functional recovery. There was an inverse agement. They found that MVO was a better predictor of adverse
relationship between transmural extent of LGE and segmental recovery. remodeling at the 4-month follow-up CMR. Cochet et al.74 compared
Interestingly, the extent of dysfunctional myocardium with zero or <25% the prognostic significance of MVO as detected by first-pass perfusion
transmural enhancement was a better predictor of global functional to late MVO in 187 patients and showed that by multivariate analysis
recovery than peak cardiac enzyme level or total infarct size by LGE. both methods are predictors of 1-year MACE. However, late MVO
Ingkanisorn et al.65 found a better correlation between acute LGE infarct had an odds ratio (OR) over 3 times that of first-pass perfusion (8.7
size and follow-up LVEF than between acute LVEF and follow-up LVEF. vs. 2.5). These results are summarized in a recent systematic review
This is probably caused by the presence of stunned, yet viable, myo- by Hamirani et al.,70 showing that both early MVO and late MVO are
cardium in the setting of early reperfusion. Lund et al.67a imaged 55 associated with lower LVEF, increased ventricular volumes and larger
patients within 1 week of reperfused MI and approximately 8 months infarct size, and predict worse adverse remodeling. However, late MVO
later. LGE infarct size of 24% of the LV area predicted LV remodeling, appears to portend greater risk of MACE compared with early MVO
as defined by an increase in EDV index of ≥20%, with a sensitivity and (OR 4.3 vs. 2.6), which may be a function of its improved specificity for
specificity of 92% and 93%, respectively. In a cohort of 231 patients true no-reflow zones. Furthermore, late MVO is more likely to contain
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 257

FIG. 20.4  Hypoenhanced infarct core may represent intramyocardial hemorrhage or microvascular obstruc-
tion (MVO). Intramyocardial hemorrhage (* on Pig A panels) can be seen with T2 mapping, but is best
demonstrated on T2* mapping. Areas of MVO are not seen with T2 or T2* mapping (Pig B). LGE, Late gado-
linium enhancement.

intramyocardial hemorrhage,70 which may represent a more severe hemorrhage on LV remodeling, and the pooled analysis demonstrates
ischemic injury. that intramyocardial hemorrhage is associated with larger ventricular
volumes, infarct size, and lower LVEF. In a recent study of 151 patients,
Intramyocardial Hemorrhage Kandler et al.81 also report that intramyocardial hemorrhage, as detected
Intramyocardial hemorrhage occurs as the result of capillary endothelial by T2*, was independently associated with larger infarct size, larger
necrosis.75 In the early era of reperfusion therapy, intramyocardial hem- LV volumes, smaller salvage index, and lower LVEF. In a study of 346
orrhage was considered a deleterious effect of reperfusion.76,77 This idea patients with reperfused ST elevation MI (STEMI) within 12 hours of
was later challenged by Fishbein et al.,78 who demonstrated that intra- symptom onset, Eitel et al.82 evaluated the prognostic significance of
myocardial hemorrhage only occurs in areas with existing microvascular intramyocardial hemorrhage, as defined by hypointense core on T2W
damage before epicardial reperfusion. Therefore intramyocardial hem- imaging. Intramyocardial hemorrhage was observed in 35% of the
orrhage is probably a manifestation of preexisting capillary injury and patients and it was associated with larger acute infarct size, MVO size,
not caused by reperfusion. It is worth noting that whether intramyocardial less myocardial salvage, and lower LVEF. Using stepwise multiple Cox-
hemorrhage is a consequence of reperfusion or a manifestation of regression analysis, the presence of intramyocardial hemorrhage was
underlying injury, it is always dependent on the duration of ischemia.76–78 an independent predictor of MACE 6 months after AMI (hazard ratio
Intramyocardial hemorrhage is mostly observed in areas of no-reflow 2.04), whereas late MVO alone was not. The addition of intramyocardial
and cannot be differentiated from nonhemorrhagic MVO by CE-CMR. hemorrhage to a predictive model that included clinical history, LV
However, CMR is the only modality that allows in vivo visualization function, and infarct size increased the c-statistic (area under the curve
of intramyocardial hemorrhage with the use of T2-weighted or T2*- [AUC]) from 0.76 to 0.8 (P = .046). The results of this study require
weighted imaging (see Fig. 20.4). T2*W imaging has been validated in confirmation in larger multicenter studies.
animals and is considered the preferred method because of improved Bulluck et al.83 reported a new insight regarding the fate of intra-
sensitivity compared with T2W imaging.79–81 myocardial hemorrhage in a cohort of STEMI patients who had CMR
An increasing body of literature indicates that the presence of intra- within the first week post-MI and a follow-up examination 5 months
myocardial hemorrhage may represent a more severe form of injury in later. Review of T2 and T2* maps at both time points indicated that
the ischemic spectrum. However, its clinical prognostic value is not well about 87% of those with intramyocardial hemorrhage had evidence of
defined. In their systematic review Hamirani et al.70 identified 9 studies residual iron deposits in the infarct zone. The T2 relaxation times were
comprising 1106 patients that evaluated the impact of intramyocardial higher in infarcts with residual iron deposits. Adverse remodeling was
258 SECTION II  Ischemic Heart Disease

equally associated with infarct size, extent of MVO, and intramyocardial at risk, such as the amount of salvaged myocardium, is preferred. Com-
hemorrhage. Hence, myocardial iron deposits could represent an impor- pared with other markers of LV remodeling, controversy remains regard-
tant therapeutic target in those with intramyocardial hemorrhage. ing the optimal CMR method to assess the area at risk. The main
proposed techniques include T2W imaging93–96 and “early” LGE with
T1 Mapping: Infarct Characterization and an inversion recovery sequence97 or cine bSSFP.98
Extracellular Volume Aletras et al.49 evaluated the use of in vivo T2W imaging as a method
Although LGE is an excellent technique for infarct characterization, to retrospectively measure the area at risk after reperfused MI. The area
there is a need for noncontrast methods that facilitate the evaluation of increased T2 signal correlated well with the fluorescent microspheres,
of patients with advanced renal disease who are at potential risk of and both were reported as consistently greater than the LGE and tri-
nephrogenic systemic fibrosis if exposed to Gd-based contrast agents. phenyl tetrazolium chloride (TTC) areas. At 2 months post-MI there
Recently, native T1 mapping has been proposed as a noncontrast method was evidence of functional recovery in the T2 areas, as demonstrated
to characterize acute and chronic MI.84 In the early 1980s a series of by DENSE radial strain maps, suggesting that this region contained
studies indicated that T1 relaxation times are prolonged in acute stunned myocardium.
infarcts.85–88 However, technical difficulties prevented further investiga- With the availability of LGE imaging for infarct quantification and
tions in this area. With the introduction of T1-mapping techniques, it the mounting evidence pointing to T2W imaging as a measure of the
is now possible reproducibly89 to measure myocardial T1 relaxation area at risk, Friedrich et al.96 used dark blood T2W imaging in a cohort
times with a breath-hold acquisition. of 92 patients presenting with their first MI to measure the salvaged
So far, T1-mapping studies after MI have reproduced earlier findings myocardium after reperfusion. Imaging was obtained about 3 days after
showing that native T1 relaxation times are increased in AMI and chronic presentation. They found that the area of T2 was consistently greater
MI compared with remote regions.84,90,91 Furthermore, native T1 values than the area of LGE by about 16%. Although lacking a reference method,
are significantly higher acutely post-MI than in the chronic phase, indi- the investigators concluded that T2W imaging can be used to image
cating histopathologic differences between AMI and chronic MI. Acute the area at risk and calculate the salvaged myocardium in clinical practice
T1 alterations may be most influenced by tissue edema, whereas in the and trials. Eitel et al.82 studied 208 patients with acute STEMI who
chronic phase replacement fibrosis represents the underlying mecha- underwent PCI and used CMR with T2W imaging and LGE to calculate
nism.84 The diagnostic performance of native T1 mapping for the detec- myocardial salvage. They showed that patients with myocardial salvage
tion of STEMI and non-STEMI (NSTEMI) appears to be high (receiver index below the median had a higher MACE rate (22.1%) over 6 months
operating characteristic = 0.91).90 The accuracy of infarct size and follow-up than those with an index greater than or equal to the median
transmurality with T1 mapping has been inferior to LGE and seems (2.9%). In that study, myocardial salvage index was the strongest pre-
dependent on field strength. Kali et al.91 were the first to prospectively dictor of MACE, outperforming MVO and infarct size.
compare infarct size by native T1 mapping to LGE in a canine model Despite a growing number of studies supporting the use of T2W
of reperfused LAD MI. Examinations were performed 7 days and 4 imaging for area at risk and salvaged myocardium, some investigators
months post-MI at 1.5 T and 3 T. The infarct region was defined as have questioned the validity of the technique. The group of Kim et al.99,100
the mean T1 value of the remote myocardium plus 5 standard devia- have listed a number of concerns, including inconsistencies in the
tions (mean ± 5 SD criterion). The LGE infarct was similarly defined pathophysiologic basis for T2W-derived area at risk, technical limita-
by the mean ± 5 SDs of the remote signal intensity. Whereas T1 maps tions, and the incongruent relationship of the T2 area at risk and infarct
at 1.5 T underestimated AMI and chronic MI size, 3 T-derived maps region. Based on these concerns, Kim et al.101 recently conducted a
overestimated acute infarct size, but accurately represented chronic validation study in canines to compare the area of increased T2 signal
infarcts. In a follow-up study of chronic MI in humans at 3 T, the with fluorescence microsphere area at risk, TTC, and LGE infarct ter-
investigators showed high sensitivity and specificity for infarct size using ritory. They imaged 21 canines with variable ischemic injuries and 24
the mean ± 5 SD threshold method.92 However, sensitivity by visual patients about 3 days after ischemia using dark-blood and bright-blood
assessment was significantly compromised (60% for STEMI, 64% for T2W pulse sequences. Their analysis was based on quantitative mea-
NSTEMI). This is because of the significantly lower contrast between surement of the mean transmural extent of the abnormality and picture
the infarcted and normal myocardium native T1 as compared with the matching of contours. They found that the area of increased T2 signal
contrast between LGE regions and remote nulled myocardium. Thus correlated better with LGE and TTC regions than with histopathologic
further efforts are needed to optimize image contrast to improve infarct area at risk. Thus they concluded that T2W imaging does not depict
detection and quantification. Furthermore, studies of intrascan, interob- the area at risk and should not be used to measure myocardial salvage.
server and intraobserver reproducibility of infarct size by T1 mapping
are needed. CARDIOVASCULAR MAGNETIC RESONANCE
IN THE ASSESSMENT OF POST-MI
Area at Risk and Myocardial Salvage
In the study of interventions intended to minimize ischemic injury,
REMODELING THERAPIES
infarct size is not an adequate measure of therapeutic effect. This is The ultimate goal of post-MI therapies is to reduce hard clinical end-
primarily because of wide variability in the area at risk of infarction, points such as cardiac death and HF. However, to identify interventions
which defines the maximum potential infarct size. The main determi- that may potentially impact clinical outcomes, pilot studies with sur-
nants of size of the area at risk include the location of the occlusion rogate endpoints are used. As described in prior sections, CMR offers
along the length of a coronary vessel and the extent of available collateral the ability to characterize post-MI remodeling with a multiparametric
circulation, both of which vary greatly between individuals in human approach that facilitates such investigations. Because of the reproduc-
studies. Furthermore, as mentioned previously, most infarcts will natu- ibility of CMR parameters, the sample size needed to detect the effect
rally undergo significant involution. However, there is great variability of therapeutic interventions can be significantly reduced compared with
in the extent of infarct resorption and it is not possible to reproducibly other modalities.21,26,102 Furthermore, CMR has been proven accurate
predict the final infarct size based on acute measurements.63 Hence, an in a range of species, which facilitates the study of the effect and mecha-
endpoint that evaluates the ischemic injury as a function of the area nisms of therapeutic interventions from small animal investigations to
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 259

large clinical trials. The following sections highlight some of the animal with cardiac overexpression of the AT2R. CMR was performed at 1, 7,
and human studies conducted to date using CMR to evaluate the effects and 28 days post-MI. At baseline, ESVI was lower and LVEF was higher
and mechanisms of post-MI remodeling interventions. in the transgenic mice. Initial infarct size was similar in both groups.
Controlling for baseline differences, the AT2R overexpressing mice had
Animal Studies a significantly lower ESVI and a significantly better LVEF at 1 week and
The use of ACEIs has been intensely studied with CMR techniques. An 4 weeks.107 The prevention of adverse remodeling was found to be, at
early study in a rat model of permanent LAD occlusion evaluated the least in part, because of preservation of contractile function in the
effects of 3 to 4 months of cilazapril therapy initiated 3 days post-MI.103 periinfarct regions.108 Nitric oxide mediates the benefit of AT2R over-
CMR measurements correlated well with histology. Cilazapril preserved expression, as demonstrated in a study of AT2R-overexpressing mice
wall thickness in infarcted myocardial segments and limited the increase treated with a nitric oxide synthase inhibitor in which these mice remod-
in chamber surface area, a surrogate of LV volume. Using the ovine eled similarly to wild-type mice.109 These investigators demonstrated
model previously mentioned, Kramer et al.104 demonstrated that ACEI equivalent remodeling in AT2R-overexpressing mice and wild-type mice
during the first 8 weeks after LAD MI limited the increase in LV EDV treated with losartan, an AT1R antagonist, supporting the hypothesis
compared with untreated controls. The limitation in LV dilatation was that much of the benefit of AT1R antagonism is mediated through the
associated with maintenance of regional LV function in viable myo- AT2R.110 Isbell et al.111 used AT2R-overexpressing mice with and without
cardium adjacent to the infarction, suggesting that dysfunction in this knockout of the bradykinin 2 receptor to ascertain whether bradykinin
region may be an important determinant of the remodeling process. 2 was involved in the attenuation of remodeling ascribed to AT2R after
Using the same model, Kramer et al.105 demonstrated that the addition MI. The lower baseline ESVI and LVEF previously observed in AT2R-
of beta-blockade to ACEI during LV remodeling did not change LV overexpressing mice appeared to be mediated by the bradykinin 2 as
EDV. However, LVEF did improve over the first 8 weeks compared with mice with bradykinin 2 knockout did not have this phenotype. However,
ACEI alone. In the same model, the most powerful pharmacologic the reduction in adverse remodeling attributed to AT2R appeared to
regimen for attenuating LV remodeling and preserving regional and be independent of bradykinin 2.
global function was the combination of ACEI and angiotensin receptor Gilson et al.112 evaluated the role of inducible nitric oxide synthase
antagonism106 (Fig. 20.5). in limiting LV remodeling after MI using a murine model of reper-
The role of the angiotensin 2 receptor (AT2R) in LV remodeling fused LAD MI and CMR. This group found that LV volumes were
has been investigated in a series of studies involving transgenic mice significantly lower and LVEF was significantly higher in inducible

RV

LA LV

A B

C D E
FIG. 20.5  Representative end-diastolic three-chamber view from an ovine model of late gadolinium enhance-
ment infarction that tested the effects of angiotensin-converting enzyme inhibitor (ACEI) and angiotensin
receptor 1 (AT1) antagonism: no therapy (A); AT1 antagonism (B); standard-dose ACEI (C); high-dose ACEI
(D); and combination therapy (E). Left ventricular remodeling was best attenuated with combination therapy.
LA, Left atrium; LV, left ventricle; RV, right ventricle. (From Mankad S, D’Amato TA, Reichek N, et al.
Combined angiotensin II receptor antagonism and angiotensin-converting enzyme inhibition further attenuates
postinfarction left ventricular remodeling. Circulation. 2001;103(23):2845–2850.)
260 SECTION II  Ischemic Heart Disease

nitric oxide synthase knockout mice compared with wild-type mice. noninfarct endocardial segment length. Other parameters measured
Furthermore, the circumferential extent of wall thinning was reduced included infarct segment length and wall thickness. ACEI therapy was
in inducible nitric oxide synthase knockout mice. This effect was associated with a reduced infarct segment length (7.9 ± 1.0 cm vs. 10.6
evident by day 7 post-MI and persisted at the final CMR scan on day ± 0.9 cm in controls) and a lower infarct expansion index (1.1 ± 0.3
28 post-MI. vs. 1.8 ± 0.3 in controls). The greatest impact of ACEI therapy on infarct
Pharmacologic agents that may limit reperfusion injury and MVO expansion was found in the subgroup with anterior infarcts. Johnson
might also be useful in limiting LV remodeling post-MI. Nicorandil is et al.125 used CMR to assess ACEI therapy post-MI in a study of 35
an adenosine triphosphate-sensitive potassium channel agonist which patients who had an LVEF >40% after their first acute Q-wave MI.
has been used as an antianginal agent in Asia and Europe because of Studies were performed at 1 week and 3 months post-MI. Volumetric
its nitrate-like effect.113 Krombach et al.114 demonstrated that nicor- cine CMR was used to quantify LV EDVI, ESVI, and mass. Therapy
andil treatment in rats after coronary artery ligation before reperfu- with the ACEI ramipril contributed to a fall in LV mass index (LVMI;
sion reduced infarct size assessed by CMR LGE. Subsequently, they from 82 ± 18 to 79 ± 23 g/m2), whereas there was no significant change
fed nicorandil to rats before coronary ligation and reperfusion and for in control patients (77 ± 15 to 79 ± 23 g/m2). No significant change in
8 weeks post-MI. At 8 weeks post-MI the nicorandil-treated groups LV EDVI was noted in either the treatment group or controls in these
had improved LVEF, EDV, ESV, and wall thickening in remote infarct patients with mild LV dysfunction at baseline.
regions and periinfarct regions relative to controls that experienced CMR was used to evaluate the effects of beta-blockade adminis-
initial infarcts of the same size.115 tered before revascularization in those presenting with Killip Class II or
Nahrendorf et al.116 studied the role of 3-hydroxy-3-methylglutaryl- less anterior STEMI in the METOCARD-CNIC (Effect of Metoprolol
coenzyme A (HMG-CoA) reductase inhibition in LV remodeling to in Cardioprotection During an Acute Myocardial Infarction) trial.126
determine whether the mechanism of any positive effect on remod- Infarct size was smaller and ejection fraction higher in the treatment
eling was nitric oxide synthase dependent. Adult rats underwent left arm without a difference in 24-hour outcomes. Groenning et al.127
coronary ligation followed by treatment with placebo, cerivastatin, performed CMR three times over 6 months on 41 patients enrolled in
or cerivastatin plus N-methyl-L-arginine methyl ester (L-NAME), the MERIT-HF (Metoprolol CR/XL Randomized Intervention Trial in
a nitric oxide synthase inhibitor. Rats fed cerivastatin had a signifi- Congestive Heart Failure) study. These were patients with chronic stable
cantly lower LV mass at 4 and 12 weeks post-MI. However, LV volume HF who had been randomized to metoprolol succinate or placebo. At
was similar among all three groups. L-NAME abolished the effects of 6 months the metoprolol group experienced a significant decrease in
cerivastatin. The authors concluded that HMG-CoA reductase inhibi- LVEDVI (150 mL/m2 at baseline to 126 mL/m2 at 6 months, P = .007)
tors diminished LV hypertrophy and helped to preserve function but and LVESVI (107 mL/m2 at baseline to 81 mL/m2 at 6 months, P =
not LV dilatation in a mechanism that appears to involve nitric oxide .001), with a concomitant increase in LVEF (29% at baseline to 37% at
synthase. 6 months, P = .005). There was no change among these variables in the
The same group also tested the effect of testosterone on LV remodel- placebo group. Dubach et al.128 performed CMR on 26 patients with a
ing based on data demonstrating a higher 2-year mortality post-MI in mean LVEF of 26 ± 6% who were randomized to bisoprolol fumarate
women than in men. Rats were treated with placebo, testosterone, or or placebo. After 1 year, the treatment group experienced an increase in
orchiectomy, starting 2 weeks before sham procedure or LAD occlu- LVEF (25.0 ±7 vs. 36.2 ± 9%; P < .05) and a trend toward smaller LVEDV
sion.117 CMR was performed 2 weeks and 8 weeks after LAD ligation. and LVESV. The control group experienced no change. More recently,
Hemodynamic measurements including LV end-diastolic pressure Bellenger et al.129 performed CMR on 34 patients with chronic stable
(LVEDP) and mean arterial pressure (MAP) were recorded. At 8 weeks, HF who were participating in the CHRISTMAS (Carvedilol Hibernation
testosterone therapy augmented LV hypertrophy with associated reduc- Reversible Ischaemia Trial, Marker of Success) trial. This is a double-
tion in LVEDP. No difference in LVEF, cardiac output, or MAP was blind study comparing carvedilol and placebo. The patients underwent
observed between the groups. CMR at baseline and at 6 months. The carvedilol group experienced a
CMR has also been used to study the role of mechanical restraint decrease in LVESVI (−9 vs. + 3 mL/m2, P = .0004) and in LVEDVI (−8
devices to limit LV remodeling after acute MI. Blom et al.118 used an vs. 0 mL/m2, P = .05) with a concomitant increase in LVEF (+3 vs. −2%,
ovine model of LAD occlusion to create an anterior MI in 10 animals. P = .003) relative to control. These studies demonstrate that differences
Five sheep received the Acorn device 1 week after acute MI. At 12 weeks in LV structure because of pharmacologic therapy of acute MI can be
after MI animals treated with the Acorn device had a significantly smaller demonstrated with CMR in relatively small-sized patient cohorts. Studies
LV epicardial surface area compared with controls. Furthermore, border using echocardiography post-MI have required a much larger group of
zone systolic regional radial strain was improved at 12 weeks in sheep patients (on the order of several hundreds) to demonstrate quantifiable
that were treated with the Acorn device. differences between treatment and control groups.130
In the past decade multiple investigators have used CMR techniques It is well established that early revascularization minimizes infarct
to evaluate the effects of stem cell transplantation in small- and large- size and reduces MACE. On the other hand, the benefits of late infarct
animal models.119–123 CMR has been useful to assess changes in volumes, artery revascularization in clinical practice is less evident. A CMR pilot
contractile function, infarct size, energetics,122 and cell engraftment with study of 16 patients showed that reperfusion within 2 weeks of MI
the use of iron-oxide labeling.120,123 mitigated ventricular remodeling compared with delayed intervention
3 months post-MI. Subsequently, Silva et al.131 published a study in
Human Studies which 36 patients with an occluded infarct related artery were random-
Numerous studies have used CMR to assess the effects of post-MI ized to PCI or optimal medical management. At 6 months there was
antiremodeling therapies and many more are underway. Schulman no difference in LV volumes or regional contractility. However, global
et al.124 randomized 43 patients with a Q-wave AMI within 24 hours LVEF improved in the PCI group and became worse in the conservative
of symptom onset to intravenous enalaprilat and then oral ACEI therapy management group. Bellenger et al.132 used CMR to assess whether the
or placebo for 1-month post-MI. Twenty-three of the patients underwent presence of infarct zone viability would affect LV remodeling after late
CMR at 1 month post-MI for evaluation of infarct expansion. The recanalization of an infarct related artery. Twenty-six patients were
infarct expansion index was defined as the ratio of the infarct to randomized to PCI (14 patients) or medical management (12 patients)
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 261

after an anterior MI. PCI was performed on an average of 26 days 2 months of exercise in a rehabilitation program or control. Cine CMR
post-MI. The initial CMR study, which included a dobutamine stress was performed and LV volumes, mass, and EF measured. At the end
protocol to assess myocardial viability, occurred at an average of 7.7 of the training period, no differences were noted between groups in
weeks post-MI. The follow-up CMR study occurred at an average of any of the aforementioned parameters. In addition, no differences were
10.8 months post-MI. In the PCI group, there was a significant correla- noted within groups over time. Exercise capacity increased in the treated
tion between the number of viable segments and improvement in LVEF group, but no deleterious effects on global parameters of LV remodeling
and ESV. No significant relationship existed between EDV and viable were found, contrary to previously published data.136
segments in the PCI group. No correlation between viability and any Finally, Heydari and colleagues examined high-dose omega-3 fatty
of the aforementioned metrics of remodeling was noted in the conser- acids in a double blind, placebo-controlled study of 358 patients with
vatively treated group. Despite these findings, the Occluded Artery Trial AMI.137 By intention-to-treat analysis, those randomized to omega-3
(OAT), which randomized 2166 stable patients to PCI versus medical fatty acids had a significant reduction in LV systolic volume index and
therapy 3 to 28 days post-MI, found no difference in MACE between noninfarct myocardial fibrosis.
the groups.133
CMR has become an important tool to assess the effect of stem cell
transplantation. Following a series of animal studies, multiple clini-
CONCLUSION
cal trials have been conducted to assess stem cell therapy after MI. With the use of CMR it is possible to accurately and reproducibly
Unfortunately the results of stem cell therapy, particularly with bone measure post-MI remodeling, including ventricular volumes, mass, EF,
marrow-derived mononuclear cells, have been disappointing. A meta- regional contractile function, and infarct size. CMR has proved instru-
analysis by de Jong et al.134 comprised 22 randomized controlled trials mental in the understanding of the mechanism of remodeling and plays
between 2002 and 2013 and found that stem cell treatment increased an important role in the development of therapeutic interventions
LVEF by 2.1% in the treatment arm as a result of the preservation of intended to minimize remodeling. CMR will undoubtedly be used with
ESV. However, no beneficial effect was observed in trials that used CMR increasing frequency as surrogate endpoints for clinical trials to prevent
to assess ventricular remodeling. Furthermore, no reduction in MACE post-MI remodeling.
was observed after a median follow-up of 6 months.
CMR has also been used in studies assessing nontraditional therapies
REFERENCES
of LV remodeling postinfarction. Dubach et al.135 studied 25 patients
with LV dysfunction after MI (EF 32 ± 6%) and randomized them to A full reference list is available online at ExpertConsult.com
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 261.e1

REFERENCES imaging, and ventriculography. J Magn Reson Imaging. 1992;2(2):


135–142.
1. Mozaffarian D, Benjamin EJ, Go AS, et al. Executive summary: heart 20. Lang RM, Badano LP, Mor-Avi V, et al. Recommendations for cardiac
disease and stroke statistics—2016 update. Circulation. 2016;133(4): chamber quantification by echocardiography in adults: an update from
447–454. the American Society of Echocardiography and the European
2. Sutton MGSJ, Sharpe N. Left ventricular remodeling after myocardial Association of Cardiovascular Imaging. J Am Soc Echocardiogr.
infarction: pathophysiology and therapy. Circulation. 2000;101(25): 2015;28(1):1–39, e14.
2981–2988. 21. Grothues F, Smith GC, Moon JC, et al. Comparison of interstudy
3. Kostuk WJ, Kazamias TM, Gander MP, Simon AL, Ross J Jr, Ross J. Left reproducibility of cardiovascular magnetic resonance with two-
ventricular size after acute myocardial infarction: serial changes and dimensional echocardiography in normal subjects and in patients with
their prognostic significance. Circulation. 1973;47(6):1174–1179. heart failure or left ventricular hypertrophy. Am J Cardiol. 2002;90(1):
4. Warren SE, Royal HD, Markis JE, Grossman W, McKay RG. Time course 29–34.
of left ventricular dilation after myocardial infarction: influence of 22. Matheijssen NA, Baur LH, Reiber JH, et al. Assessment of left ventricular
infarct-related artery and success of coronary thrombolysis. J Am Coll volume and mass by cine magnetic resonance imaging in patients with
Cardiol. 1988;11(1):12–19. anterior myocardial infarction intra-observer and inter-observer
5. Jeremy RW, Allman KC, Bautovitch G, Harris PJ. Patterns of left variability on contour detection. Int J Card Imaging. 1996;12(1):11–19.
ventricular dilation during the six months after myocardial infarction. J 23. Moon JCC, Lorenz CH, Francis JM, Smith GC, Pennell DJ. Breath-hold
Am Coll Cardiol. 1989;13(2):304–310. FLASH and FISP cardiovascular MR imaging: left ventricular volume
6. St John Sutton M, Pfeffer MA, Moye L, et al. Cardiovascular death and differences and reproducibility. Radiology. 2002;223(11):789–797.
left ventricular remodeling two years after myocardial infarction: 24. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ.
baseline predictors and impact of long-term use of captopril: Interstudy reproducibility of right ventricular volumes, function, and
information from the Survival and Ventricular Enlargement (SAVE) mass with cardiovascular magnetic resonance. Am Heart J. 2004;147(2):
trial. Circulation. 1997;96(10):3294–3299. 218–223.
7. Konermann M, Sanner BM, Horstmann E, et al. Changes of the left 25. Clarke CJ, Gurka MJ, Norton PT, Kramer CM, Hoyer AW. Assessment
ventricle after myocardial infarction—estimation with cine magnetic of the accuracy and reproducibility of RV volume measurements by
resonance imaging during the first six months. Clin Cardiol. CMR in congenital heart disease. JACC Cardiovasc Imaging. 2012;5(1):
1997;20(3):201–212. 28–37.
8. Pfeffer MA, Braunwald E. Ventricular remodeling after myocardial 26. Bellenger NG, Davies LC, Francis JM, Coats AJ, Pennell DJ. Reduction in
infarction. Experimental observations and clinical implications. sample size for studies of remodeling in heart failure by the use of
Circulation. 1990;81(4):1161–1172. cardiovascular magnetic resonance. J Cardiovasc Magn Reson.
9. Gaudron P, Eilles C, Kugler I, Ertl G. Progressive left ventricular 2000;2(4):271–278.
dysfunction and remodeling after myocardial infarction. Potential 27. Konermann M, Grotz J, Hotzinger H, et al. Cine-MRT in the functional
mechanisms and early predictors. Circulation. 1993;87(3):755–763. and morphological diagnosis of the heart after a myocardial infarct: a
10. White HD, Norris RM, Brown MA, Brandt PW, Whitlock RM, Wild CJ. comparison with angiocardiography, 2-dimensional echocardiography,
Left ventricular end-systolic volume as the major determinant of intracardiac scintigraphy and enzymatic infarct size estimation. Rofo.
survival after recovery from myocardial infarction. Circulation. 1995;163(1):24–31.
1987;76(1):44–51. 28. Kramer CM, Rogers WJ, Theobald TM, Power TP, Geskin G, Reichek N.
11. Risk stratification and survival after myocardial infarction. N Engl J Dissociation between changes in intramyocardial function and left
Med. 1983;309(6):331–336. ventricular volumes in the eight weeks after first anterior myocardial
12. Nicolosi GL, Latini R, Marino P, et al. The prognostic value of infarction. J Am Coll Cardiol. 1997;30(7):1625–1632.
predischarge quantitative two-dimensional echocardiographic 29. Kuijer JPA, Ms C, Marcus JT, et al. Quantification of regional contractile
measurements and the effects of early lisinopril treatment on left function after infarction: strain analysis superior to wall thickening
ventricular structure and function after acute myocardial infarction in analysis in discriminating infarct from remote myocardium. J Am Coll
the GISSI-3 Trial. Gruppo Italia. Eur Heart J. 1996;17(11):1646–1656. Cardiol. 2001;37(3):808–817.
13. Migrino RQ, Young JB, Ellis SG, et al. End-systolic volume index at 90 30. Axel L, Dougherty L. Heart wall motion: improved method of spatial
to 180 minutes into reperfusion therapy for acute myocardial infarction modulation of magnetization for MR imaging. Radiology. 1989;172(2):
is a strong predictor of early and late mortality. Circulation. 349–350.
1997;96(1):116–121. 31. Aletras AH, Ding S, Balaban RS, Wen H. DENSE: displacement encoding
14. Sanz G, Castaner A, Betriu A, et al. Determinants of prognosis in with stimulated echoes in cardiac functional MRI. J Magn Reson.
survivors of myocardial infarction: a prospective clinical angiographic 1999;137(1):247–252.
study. N Engl J Med. 1982;306(18):1065–1070. 32. Maret E, Todt T, Brudin L, et al. Functional measurements based on
15. Thiele H, Nagel E, Paetsch I, et al. Functional cardiac MR imaging with feature tracking of cine magnetic resonance images identify left
steady-state free precession (SSFP) significantly improves endocardial ventricular segments with myocardial scar. Cardiovasc Ultrasound.
border delineation without contrast agents. J Magn Reson Imaging. 2009;7:53.
2001;14(4):362–367. 33. Hor KN, Gottliebson WM, Carson C, et al. Comparison of magnetic
16. Barkhausen J, Ruehm SG, Goyen M, Buck T, Laub G, Debatin JF. MR resonance feature tracking for strain calculation with harmonic phase
evaluation of ventricular function: true fast imaging with steady-state imaging analysis. JACC Cardiovasc Imaging. 2010;3(2):144–151.
precession versus fast low-angle shot cine MR imaging: feasibility study. 33a.  Khan JN, Nazir SA, Singh A, et al. Relationship of myocardial strain
Radiology. 2001;219(1):264–269. and markers of myocardial injury to predict segmental recovery
17. Shapiro EP, Rogers WJ, Beyar R, et al. Determination of left ventricular following acute ST-segment elevation myocardial infarction. Circ
mass by magnetic resonance imaging in hearts deformed by acute Cardiovasc Imaging. 2016;9:e003457.
infarction. Circulation. 1989;79(3):706–711. 34. Holman ER, Buller VG, de Roos A, et al. Detection and quantification of
18. Bloomgarden DC, Fayad ZA, Ferrari VA, Chin B, Sutton MG, Axel L. dysfunctional myocardium by magnetic resonance imaging. A new
Global cardiac function using fast breath-hold MRI: validation of new three-dimensional method for quantitative wall-thickening analysis.
acquisition and analysis techniques. Magn Reson Med. Circulation. 1997;95(4):924–931.
1997;37(5):683–692. 35. Kramer CM, Lima JA, Reichek N, et al. Regional differences in function
19. Debatin JF, Nadel SN, Paolini JF, et al. Cardiac ejection fraction: within noninfarcted myocardium during left ventricular remodeling.
phantom study comparing cine MR imaging, radionuclide blood pool Circulation. 1993;88(3):1279–1288.
261.e2 SECTION II  Ischemic Heart Disease

36. Moulton MJ, Downing SW, Creswell LL, et al. Mechanical dysfunction 54. Chen J, Song SK, Liu W, et al. Remodeling of cardiac fiber structure
in the border zone of an ovine model of left ventricular aneurysm. Ann after infarction in rats quantified with diffusion tensor MRI. Am J
Thorac Surg. 1995;60(4):986–998. Physiol Heart Circ Physiol. 2003;285(3):H946–H954.
37. Epstein FH, Yang Z, Gilson WD, Berr SS, Kramer CM, French BA. MR 55. Kim RJ, Fieno DS, Parrish TB, et al. Relationship of MRI delayed
tagging early after myocardial infarction in mice demonstrates contrast enhancement to irreversible injury, infarct age, and contractile
contractile dysfunction in adjacent and remote regions. Magn Reson function. Circulation. 1999;100(19):1992–2002.
Med. 2002;48(2):399–403. 56. Kim RJ, Chen EL, Lima JA, Judd RM. Myocardial Gd-DTPA kinetics
37a.  Thomas D, Ferrari VA, Janik M, et al. Quantitative assessment of determine MRI contrast enhancement and reflect the extent and severity
regional myocardial function in a rat model of myocardial infarction of myocardial injury after acute reperfused infarction. Circulation.
using tagged MRI. MAGMA. 2004;17(3–6):179–187. 1996;94(12):3318–3326.
38. Kramer CM, Rogers WJ, Theobald TM, Power TP, Petruolo S, Reichek 57. Fieno DS, Kim RJ, Chen E-L, Lomasney JW, Klocke FJ, Judd RM.
N. Remote noninfarcted region dysfunction soon after first anterior Contrast-enhanced magnetic resonance imaging of myocardium at risk.
myocardial infarction. A magnetic resonance tagging study. Circulation. J Am Coll Cardiol. 2000;36(6):1985–1991.
1996;94(4):660–666. 58. Judd RM, Lugo-Olivieri CH, Arai M, et al. Physiological basis of
39. Rademakers F, Van de Werf F, Mortelmans L, Marchal G, Bogaert J. myocardial contrast enhancement in fast magnetic resonance images of
Evolution of regional performance after an acute anterior myocardial 2-day-old reperfused canine infarcts. Circulation. 1995;92(7):1902–1910.
infarction in humans using magnetic resonance tagging. J Physiol. 59. Hsu LY, Ingkanisorn WP, Kellman P, Aletras AH, Arai AE. Quantitative
2003;546(Pt 3):777–787. myocardial infarction on delayed enhancement MRI. Part II: clinical
40. Osman NF, Kerwin WS, McVeigh ER, Prince JL. Cardiac motion application of an automated feature analysis and combined thresholding
tracking using CINE harmonic phase (HARP) magnetic resonance infarct sizing algorithm. J Magn Reson Imaging. 2006;23(3):309–314.
imaging. Magn Reson Med. 1999;42(6):1048–1060. 60. Thiele H, Kappl MJE, Conradi S, Niebauer J, Hambrecht R, Schuler G.
41. Garot J, Bluemke DA, Osman NF, et al. Fast determination of regional Reproducibility of chronic and acute infarct size measurement by
myocardial strain fields from tagged cardiac images using harmonic delayed enhancement-magnetic resonance imaging. J Am Coll Cardiol.
phase MRI. Circulation. 2000;101(9):981–988. 2006;47(8):1641–1645.
42. Kim D, Gilson WD, Kramer CM, Epstein FH. Myocardial tissue tracking 61. Mahrholdt H, Wagner A, Holly TA, et al. Reproducibility of chronic
with two-dimensional cine displacement-encoded MR imaging: infarct size measurement by contrast-enhanced magnetic resonance
development and initial evaluation. Radiology. 2004;230(3): imaging. Circulation. 2002;106(18):2322–2327.
862–871. 62. Rochitte CE, Lima JA, Bluemke DA, et al. Magnitude and time course of
43. Feng L, Donnino R, Babb J, Axel L, Kim D. Numerical and in vivo microvascular obstruction and tissue injury after acute myocardial
validation of fast cine displacement-encoded with stimulated echoes infarction. Circulation. 1998;98(10):1006–1014.
(DENSE) MRI for quantification of regional cardiac function. Magn 63. Fieno DS, Hillenbrand HB, Rehwald WG, et al. Infarct resorption,
Reson Med. 2009;62(3):682–690. compensatory hypertrophy, and differing patterns of ventricular
44. Neizel M, Lossnitzer D, Korosoglou G, et al. Strain-encoded (SENC) remodeling following myocardial infarctions of varying size. J Am Coll
magnetic resonance imaging to evaluate regional heterogeneity of Cardiol. 2004;43(11):2124–2131.
myocardial strain in healthy volunteers: comparison with conventional 64. Richard V, Murry CE, Reimer KA. Healing of myocardial infarcts in
tagging. J Magn Reson Imaging. 2009;29(1):99–105. dogs. Effects of late reperfusion. Circulation. 1995;92(7):1891–1901.
45. Liu W, Chen J, Ji S, et al. Harmonic phase MR tagging for direct 65. Ingkanisorn WP, Rhoads KL, Aletras AH, Kellman P, Arai AE.
quantification of Lagrangian strain in rat hearts after myocardial Gadolinium delayed enhancement cardiovascular magnetic resonance
infarction. Magn Reson Med. 2004;52(6):1282–1290. correlates with clinical measures of myocardial infarction. J Am Coll
46. Azevedo CF, Amado LC, Kraitchman DL, et al. Persistent diastolic Cardiol. 2004;43(12):2253–2259.
dysfunction despite complete systolic functional recovery after 66. Choi CJ, Haji-Momenian S, Dimaria JM, et al. Infarct involution and
reperfused acute myocardial infarction demonstrated by tagged improved function during healing of acute myocardial infarction: the
magnetic resonance imaging. Eur Heart J. 2004;25(16):1419–1427. role of microvascular obstruction. J Cardiovasc Magn Reson.
47. Gerber BL, Rochitte CE, Melin JA, et al. Microvascular obstruction and 2004;6(4):917–925.
left ventricular remodeling early after acute myocardial infarction. 67. Choi KM, Kim RJ, Gubernikoff G, Vargas JD, Parker M, Judd RM.
Circulation. 2000;101(23):2734–2741. Transmural extent of acute myocardial infarction predicts long-term
48. Ashikaga H, Mickelsen SR, Ennis DB, et al. Electromechanical analysis improvement in contractile function. Circulation. 2001;104(10):
of infact border zone in chronic myocardial infarction. Am J Physiol 1101–1107.
Heart Circ Physiol. 2005;289(3):H1099–H1105. 67a.  Lund GK, Stork A, Muellerleile K, et al. Prediction of left ventricular
49. Aletras AH, Tilak GS, Natanzon A, et al. Retrospective determination of remodeling and analysis of infarct resorption in patients with reperfused
the area at risk for reperfused acute myocardial infarction with myocardial infarcts by using contrast-enhanced MR imaging. Radiology.
T2-weighted cardiac magnetic resonance imaging: histopathological and 2007;245(1):95–102.
displacement encoding with stimulated echoes (DENSE) functional 68. Di Bella G, Siciliano V, Aquaro GD, et al. Scar extent, left ventricular
validations. Circulation. 2006;113(15):1865–1870. end-diastolic volume, and wall motion abnormalities identify high-risk
50. Scollan DF, Holmes A, Winslow R, Forder J. Histological validation of patients with previous myocardial infarction: a multiparametric
myocardial microstructure obtained from diffusion tensor magnetic approach for prognostic stratification. Eur Heart J. 2013;34(2):104–111.
resonance imaging. Am J Physiol. 1998;275(6 Pt 2):H2308–H2318. 69. Reffelmann T, Kloner RA. The no-reflow phenomenon: a basic
51. Holmes AA, Scollan DF, Winslow RL. Direct histological validation of mechanism of myocardial ischemia and reperfusion. Basic Res Cardiol.
diffusion tensor MRI in formaldehyde-fixed myocardium. Magn Reson 2006;101(5):359–372.
Med. 2000;44(1):157–161. 70. Hamirani YS, Wong A, Kramer CM, Salerno M. Effect of microvascular
52. Wu MT, Tseng WYI, Su MYM, et al. Diffusion tensor magnetic obstruction and intramyocardial hemorrhage by CMR on LV
resonance imaging mapping the fiber architecture remodeling in human remodeling and outcomes after myocardial infarction: a systematic
myocardium after infarction: correlation with viability and wall motion. review and meta-analysis. JACC Cardiovasc Imaging. 2014;7(9):
Circulation. 2006;114(10):1036–1045. 940–952.
53. Sosnovik DE, Mekkaoui C, Huang S, et al. Microstructural impact of 71. Wu KC, Kim RJ, Bluemke DA, et al. Quantification and time course of
ischemia and bone marrow-derived cell therapy revealed with diffusion microvascular obstruction by contrast-enhanced echocardiography and
tensor magnetic resonance imaging tractography of the heart in vivo. magnetic resonance imaging following acute myocardial infarction and
Circulation. 2014;129(17):1731–1741. reperfusion. J Am Coll Cardiol. 1998;32(6):1756–1764.
CHAPTER 20  Acute Myocardial Infarction: Ventricular Remodeling 261.e3

72. Wu KC, Zerhouni EA, Judd RM, et al. Prognostic significance of 92. Kali A, Choi EY, Sharif B, et al. Native T1 mapping by 3-T CMR imaging
microvascular obstruction by magnetic resonance imaging in patients for characterization of chronic myocardial infarctions. JACC Cardiovasc
with acute myocardial infarction. Circulation. 1998;97(8):765–772. Imaging. 2015;8(9):1019–1030.
73. Nijveldt R, Hofman MB, Hirsch A, et al. Assessment of microvascular 93. García-Dorado D, Oliveras J, Gili J, et al. Analysis of myocardial oedema
obstruction and prediction of short-term remodeling after acute by magnetic resonance imaging early after coronary artery occlusion
myocardial infarction: cardiac MR imaging study. Radiology. with or without reperfusion. Cardiovasc Res. 1993;27(8):1462–1469.
2009;250(2):363–370. 94. Aletras AH, Tilak GS, Natanzon A, et al. Retrospective determination of
74. Cochet AA, Lorgis L, Lalande A, et al. Major prognostic impact of the area at risk for reperfused acute myocardial infarction with
persistent microvascular obstruction as assessed by contrast-enhanced T2-weighted cardiac magnetic resonance imaging: histopathological and
cardiac magnetic resonance in reperfused acute myocardial infarction. displacement encoding with stimulated echoes (DENSE) functional
Eur Radiol. 2009;19(9):2117–2126. validations. Circulation. 2006;113(15):1865–1870.
75. Kloner RA, Ganote CE, Jennings RB. The “no-reflow” phenomenon after 95. Tilak GS, Hsu L-YY, Hoyt RF Jr, et al. In vivo T2-weighted magnetic
temporary coronary occlusion in the dog. J Clin Invest. 1974;54(6): resonance imaging can accurately determine the ischemic area at risk for
1496–1508. 2-day-old nonreperfused myocardial infarction. Invest Radiol.
76. Bresnahan GF, Roberts R, Shell WE, Ross J, Sobel BE. Deleterious effects 2008;43(1):7–15.
due to hemorrhage after myocardial reperfusion. Am J Cardiol. 96. Friedrich MG, Abdel-Aty H, Taylor A, Schulz-Menger J, Messroghli D,
1974;33(1):82–86. Dietz R. The salvaged area at risk in reperfused acute myocardial
77. Capone RJ, Most AS. Myocardial hemorrhage after coronary reperfusion infarction as visualized by cardiovascular magnetic resonance. J Am Coll
in pigs. Am J Cardiol. 1978;41(2):259–266. Cardiol. 2008;51(16):1581–1587.
78. Fishbein MC, Y-Rit J, Lando U, Kanmatsuse K, Mercier JC, Ganz W. The 97. Matsumoto H, Matsuda T, Miyamoto K, Shimada T, Mikuri M, Hiraoka
relationship of vascular injury and myocardial hemorrhage to necrosis Y. Peri-infarct zone on early contrast-enhanced CMR imaging in
after reperfusion. Circulation. 1980;62(6):1274–1279. patients with acute myocardial infarction. JACC Cardiovasc Imaging.
79. Kumar A, Green JD, Sykes JM, et al. Detection and Quantification of 2011;4(6):610–618.
Myocardial Reperfusion Hemorrhage Using T2*-Weighted CMR. JACC 98. Tufvesson J, Carlsson M, Aletras AH, et al. Automatic segmentation of
Cardiovasc Imaging. 2011;4(12):1274–1283. myocardium at risk from contrast enhanced SSFP CMR: validation
80. Kali A, Tang RLQ, Kumar A, Min JK. Detection of acute reperfusion against expert readers and SPECT. BMC Med Imaging. 2016;16(1):19.
myocardial hemorrhage with cardiac MR imaging: T2 versus T2. 99. Friedrich MG, Kim HW, Kim RJ. T2-weighted imaging to assess
Radiology. 2013;269(2):387–395. post-infarct myocardium at risk. JACC Cardiovasc Imaging. 2011;4(9):
81. Kandler D, Lucke C, Grothoff M, et al. The relation between 1014–1021.
hypointense core, microvascular obstruction and intramyocardial 100. Croisille P, Kim HW, Kim RJ. Controversies in cardiovascular MR
haemorrhage in acute reperfused myocardial infarction assessed by imaging: T2-weighted imaging should not be used to delineate the area
cardiac magnetic resonance imaging. Eur Radiol. 2014;24(12): at risk in ischemic myocardial injury. Radiology. 2012;265(1):12–22.
3277–3288. 101. Kim HW, Van Assche L, Jennings RB, et al. Relationship of T2-weighted
82. Eitel I, Kubusch K, Strohm O, et al. Prognostic value and determinants MRI myocardial hyperintensity and the ischemic area-at-risk. Circ Res.
of a hypointense infarct core in T2-weighted cardiac magnetic resonance 2015;117(3):254–265.
in acute reperfused ST-elevation-myocardial infarction. Circ Cardiovasc 102. Engblom H, Heiberg E, Erlinge D, et al. Sample size in clinical
Imaging. 2011;4(4):354–362. cardioprotection trials using myocardial salvage index, infarct size, or
83. Bulluck H, Rosmini S, Abdel-Gadir A, et al. Residual myocardial iron biochemical markers as endpoint. J Am Heart Assoc. 2016;5(3):e002708.
following intramyocardial hemorrhage during the convalescent phase of 103. Saeed M, Wendland MF, Seelos K, Masui T, Derugin N, Higgins CB.
reperfused ST-segment–elevation myocardial infarction and adverse left Effect of cilazapril on regional left ventricular wall thickness and
ventricular remodeling. Circ Cardiovasc Imaging. 2016;9(10):e004940. chamber dimension following acute myocardial infarction: in vivo
84. Messroghli DR, Walters K, Plein S, et al. Myocardial T1 mapping: assessment using MRI. Am Heart J. 1992;123(6):1472–1480.
application to patients with acute and chronic myocardial infarction. 104. Kramer CM, Ferrari VA, Rogers WJ, et al. Angiotensin-converting
Magn Reson Med. 2007;58(1):34–40. enzyme inhibition limits dysfunction in adjacent noninfarcted regions
85. Williams ES, Kaplan JI, Thatcher F, Zimmerman G, Knoebel SB. during left ventricular remodeling. J Am Coll Cardiol.
Prolongation of proton spin lattice relaxation times in regionally 1996;27(1):211–217.
ischemic tissue from dog hearts. J Nucl Med. 1980;21(5):449–453. 105. Kramer CM, Nicol PD, Rogers WJ, Seibel PS, Park CS, Reichek N.
86. Higgins CB, Herfkens R, Lipton MJ, et al. Nuclear magnetic resonance Beta-blockade improves adjacent regional sympathetic innervation
imaging of acute myocardial infarction in dogs: alterations in magnetic during postinfarction remodeling. Am J Physiol. 1999;277(4 Pt 2):
relaxation times. Am J Cardiol. 1983;52(1):184–188. H1429–H1434.
87. Wesbey GE, Higgins CB, McNamara MT, et al. Effect of gadolinium- 106. Mankad S, D’Amato TA, Reichek N, et al. Combined angiotensin II
DTPA on the magnetic relaxation times of normal and infarcted receptor antagonism and angiotensin-converting enzyme inhibition
myocardium. Radiology. 1984;153(1):165–169. further attenuates postinfarction left ventricular remodeling. Circulation.
88. Johnston DL, Brady TJ, Ratner AV, et al. Assessment of myocardial 2001;103(23):2845–2850.
ischemia with proton magnetic resonance: effects of a three hour 107. Yang Z, Bove CM, French BA, et al. Angiotensin II type 2 receptor
coronary occlusion with and without reperfusion. Circulation. overexpression preserves left ventricular function after myocardial
1985;71(3):595–601. infarction. Circulation. 2002;106(1):106–111.
89. Messroghli DR, Plein S, Higgins DM, et al. Human myocardium: 108. Bove CM, Gilson WD, Scott CD, et al. The angiotensin II type 2 receptor
single-breath-hold MR T1 mapping with high spatial resolution— and improved adjacent region function post-MI. J Cardiovasc Magn
reproducibility study. Radiology. 2006;238(3):1004–1012. Reson. 2005;7(2):459–464.
90. Dall’Armellina E, Piechnik SK, Ferreira VM, et al. Cardiovascular 109. Bove CM, Yang Z, Gilson WD, et al. Nitric oxide mediates benefits of
magnetic resonance by non contrast T1-mapping allows assessment of angiotensin II type 2 receptor overexpression during post-infarct
severity of injury in acute myocardial infarction. J Cardiovasc Magn remodeling. Hypertension. 2004;43(3):680–685.
Reson. 2012;14(Mi):15. 110. Voros S, Yang Z, Bove CM, et al. Interaction between AT1 and AT2
91. Kali A, Cokic I, Tang RLQ, et al. Determination of location, size, and receptors during postinfarction left ventricular remodeling. Am J Physiol
transmurality of chronic myocardial infarction without exogenous Heart Circ Physiol. 2006;290(3):H1004–H1010.
contrast media by using cardiac magnetic resonance imaging at 3 T. Circ 111. Isbell DC, Voros S, Yang Z, et al. Interaction between bradykinin subtype
Cardiovasc Imaging. 2014;7(3):471–481. 2 and angiotensin II type 2 receptors during post-MI left ventricular
261.e4 SECTION II  Ischemic Heart Disease

remodeling. Am J Physiol Heart Circ Physiol. 2007;293(6):H3372– 125. Johnson DB, Foster RE, Barilla F, et al. Angiotensin-converting enzyme
H3378. inhibitor therapy affects left ventricular mass in patients with ejection
112. Gilson WD, Epstein FH, Yang Z, et al. Borderzone contractile fraction >40% after acute myocardial infarction. J Am Coll Cardiol.
dysfunction is transiently attenuated and left ventricular structural 1997;29(1):49–54.
remodeling is markedly reduced following reperfused myocardial 126. Ibanez B, Macaya C, Sánchez-Brunete V, et al. Effect of early metoprolol
infarction in inducible nitric oxide synthase knockout mice. J Am Coll on infarct size in st-segment-elevation myocardial infarction patients
Cardiol. 2007;50(18):1799–1807. undergoing primary percutaneous coronary intervention: the Effect
113. Roland E. Safety profile of an anti-anginal agent with potassium channel of Metoprolol in Cardioprotection During an Acute Myocardial
opening activity: an overview. Eur Heart J. 1993;14(suppl B):48–52. Infarction (METOCARD-CNIC) trial. Circulation. 2013;128(14):
114. Krombach GA, Higgins CB, Chujo M, Saeed M. Blood pool contrast- 1495–1503.
enhanced MRI detects suppression of microvascular permeability in 127. Groenning BA, Nilsson JC, Sondergaard L, Fritz-Hansen T, Larsson HB,
early postinfarction reperfusion after nicorandil therapy. Magn Reson Hildebrandt PR. Antiremodeling effects on the left ventricle during
Med. 2002;47(5):896–902. beta-blockade with metoprolol in the treatment of chronic heart failure.
115. Saeed M, Watzinger N, Krombach GA, et al. Left ventricular remodeling J Am Coll Cardiol. 2000;36(7):2072–2080.
after infarction: sequential MR imaging with oral nicorandil therapy in 128. Dubach P, Myers J, Bonetti P, et al. Effects of bisoprolol fumarate on left
rat model. Radiology. 2002;224(3):830–837. ventricular size, function, and exercise capacity in patients with heart
116. Nahrendorf M, Hu K, Hiller K-H, et al. Impact of failure: analysis with magnetic resonance myocardial tagging. Am Heart
hydroxymethylglutaryl coenzyme a reductase inhibition on left J. 2002;143(4):676–683.
ventricular remodeling after myocardial infarction: an experimental 129. Bellenger NG, Rajappan K, Rahman SL, et al. Effects of carvedilol on left
serial cardiac magnetic resonance imaging study. J Am Coll Cardiol. ventricular remodelling in chronic stable heart failure: a cardiovascular
2002;40(9):1695–1700. magnetic resonance study. Heart. 2004;90(7):760–764.
117. Nahrendorf M, Frantz S, Hu K, et al. Effect of testosterone on 130. St John Sutton M, Pfeffer MA, Plappert T, et al. Quantitative two-
post-myocardial infarction remodeling and function. Cardiovasc Res. dimensional echocardiographic measurements are major predictors of
2003;57(2):370–378. adverse cardiovascular events after acute myocardial infarction. The
118. Blom AS, Pilla JJ, Arkles J, et al. Ventricular restraint prevents infarct protective effects of captopril. Circulation. 1994;89(1):68–75.
expansion and improves borderzone function after myocardial 131. Silva JC, Rochitte CE, Júnior JS, et al. Late coronary artery recanalization
infarction: a study using magnetic resonance imaging, three-dimensional effects on left ventricular remodelling and contractility by magnetic
surface modeling, and myocardial tagging. Ann Thorac Surg. 2007;84(6): resonance imaging. Eur Heart J. 2005;26(1):36–43.
2004–2010. 132. Bellenger NG, Yousef Z, Rajappan K, Marber MS, Pennell DJ. Infarct
119. Limbourg FP, Ringes-Lichtenberg S, Schaefer A, et al. Haematopoietic zone viability influences ventricular remodelling after late recanalisation
stem cells improve cardiac function after infarction without permanent of an occluded infarct related artery. Heart. 2005;91(4):478–483.
cardiac engraftment. Eur J Heart Fail. 2005;7(5):722–729. 133. Hochman JS, Lamas GA, Buller CE, et al. Coronary intervention for
120. Amado LC, Saliaris AP, Schuleri KH, et al. Cardiac repair with persistent occlusion after myocardial infarction. N Engl J Med.
intramyocardial injection of allogeneic mesenchymal stem cells after 2006;355(23):2395–2407.
myocardial infarction. Proc Natl Acad Sci USA. 2005;102(32): 134. de Jong R, Houtgraaf JH, Samiei S, Boersma E, Duckers HJ.
11474–11479. Intracoronary stem cell infusion after acute myocardial infarction: a
121. van den Bos EJ, Thompson RB, Wagner A, et al. Functional assessment meta-analysis and update on clinical trials. Circ Cardiovasc Interv.
of myoblast transplantation for cardiac repair with magnetic resonance 2014;7(2):156–167.
imaging. Eur J Heart Fail. 2005;7(4):435–443. 135. Dubach P, Myers J, Dziekan G, et al. Effect of exercise training on
122. Zeng L, Hu Q, Wang X, et al. Bioenergetic and functional consequences myocardial remodeling in patients with reduced left ventricular function
of bone marrow-derived multipotent progenitor cell transplantation in after myocardial infarction: application of magnetic resonance imaging.
hearts with postinfarction left ventricular remodeling. Circulation. Circulation. 1997;95(8):2060–2067.
2007;115(14):1866–1875. 136. Jugdutt BI, Michorowski BL, Kappagoda CT. Exercise training after
123. Williams AR, Hatzistergos KE, Addicott B, et al. Enhanced effect of anterior Q wave myocardial infarction: importance of regional left
combining human cardiac stem cells and bone marrow mesenchymal ventricular function and topography. J Am Coll Cardiol. 1988;12(2):
stem cells to reduce infarct size and to restore cardiac function after 362–372.
myocardial infarction. Circulation. 2013;127(2):213–223. 137. Heydari B, Abdullah S, Potalla JV, et al. Effect of omega-3 acid ethyl
124. Schulman SP, Weiss JL, Becker LC, et al. Effect of early enalapril therapy esters on left ventricular remodeling after acute myocardial infarction.
on left ventricular function and structure in acute myocardial infarction. The Omega-Remodel randomized clinical trial. Circulation.
Am J Cardiol. 1995;76(11):764–770. 2016;134:378–391.
21 
Myocardial Viability
Udo P. Sechtem and Heiko Mahrholdt

The detection of residual myocardial viability in a patient with regional often surprisingly little angina. This type of dysfunction is often more
or global severe left ventricular (LV) dysfunction is of clinical importance chronic, and previous myocardial infarction may or may not be reported
to plan the therapeutic strategy because revascularization of dysfunc- in the history. Pathology may reveal regions of transmural scar, regions
tional but viable myocardium may improve LV function.1 Several imaging with predominantly subendocardial scar, and regions with mixtures of
techniques have been shown to be successful in detecting myocardial scar and viable myocardium.
viability; these include LV angiography using appropriate interventions,2 Severe wall thinning is the hallmark of transmural chronic myocardial
perfusion scintigraphy,3 positron emission tomography (PET),4 and infarction. However, wall thinning is the end result of infarct healing,
echocardiography.5 More recently, cardiovascular magnetic resonance and it may take up to 4 months before the remodeling process is com-
(CMR) has gained widespread acceptance as a technique to identify pleted. In contrast to the severe thinning of chronic transmural scar,
viable myocardium and distinguish it from myocardial necrosis and the best example for which is the thin-walled anterior LV aneurysm,
scar.6–9 This chapter reviews the current knowledge of how these tech- acute and subacute transmural infarcts may not yet have reached the
niques can be used in humans to guide clinical decision making and stage of thinning because local infarct remodeling is incomplete.12 In
predict recovery of function after revascularization of dysfunctional contrast with transmural myocardial infarction, which may or may not
myocardium. appear thinned, depending on infarct age, healed nontransmural infarcts
All scientific papers on the value of imaging techniques for detecting usually do not develop severe thinning. Some thinning may be observed,
myocardial viability use the well-known statistical terms of sensitivity, however, depending on the degree to which the endocardially located
specificity, and the predictive values. The sensitivity of a test is com- infarct extends throughout the wall. Therefore the finding of preserved
monly defined as the number of true positives divided by the sum of myocardial wall thickness in diastole in a patient with a known chronic
true positives and false negatives. In other words, the sensitivity of a infarct that is more than 4 months old will likely represent nontrans-
test is the number of diseased persons with a positive test divided by mural infarction with a substantial rim of viable myocardium sur-
the total number of diseased persons. Common sense might suggest rounding the endocardial scar. If the infarct is more recent than
that scar tissue and, hence, absence of viability might indicate the pres- approximately 4 months, preserved end-diastolic wall thickness cannot
ence of disease. The total number of diseased people or segments would be used to distinguish between viable and nonviable myocardium.
thus be the number of people with scar or segments without recovery Patients with small subendocardial infarcts may, however, also
of function. The presence of viability would be synonymous with a present with regional wall thinning despite the presence of substantial
healthy state (relatively speaking), and the total number of viable, healthy amounts (>50% of wall thickness) of viable myocardium.13,14 In a case
people or segments would appear as the denominator in the formula described by Kim and Shah,13 diastolic thickness of the anterior wall on
for calculating specificity. In the viability literature, however, sensitivity a long-axis late gadolinium enhancement (LGE) CMR image measured
indicates the ability of a test to identify viable myocardium, and speci- only 5 mm. However, the subendocardial rim of scar was only 1.5 mm
ficity is an indicator of how well the test performs when it comes to of total wall thickness, indicating the presence of substantial amounts
detecting scar. Positive and negative predictive values are used accord- of viable myocardium. Indeed, recovery of the myocardium occurred
ingly. This needs to be borne in mind when it comes to the interpretation following revascularization. Shah et al.14 systematically looked at
of test results reported later in this chapter. more than 1000 consecutive patients who had LGE CMR for viabil-
ity assessment and found that 19% of them had regional wall thin-
FEATURES OF VIABLE MYOCARDIUM DETECTABLE ning. Within these regions, the extent of scarring was 72%. However,
BY CARDIOVASCULAR MAGNETIC RESONANCE 18% of thinned regions had only limited scar burden (≤50% of total
extent). Among patients with thinning undergoing revascularization
Scar Formation and Left Ventricular Wall Thickness and follow-up cine CMR (n = 42), scar extent within the thinned
Myocardium is commonly defined as viable if it shows severe dysfunc- region was inversely related to regional and global contractile improve-
tion at baseline but recovers function with time either spontaneously ment. End-diastolic wall thickness in thinned regions with limited scar
(myocardial stunning) or following revascularization (hibernating burden increased from 4.4 mm to 7.5 mm after revascularization (P <
myocardium).10,11 Clinically, stunned myocardium may be found in .001) with resolution of wall thinning (Fig. 21.1). Thus regional wall
patients with early reperfusion of an infarct-related artery. If there is thinning may be possible in myocardium that has only little suben-
no residual high-grade stenosis, blood flow at rest will be normal and docardial scarring probably because of ventricular remodeling, and
the myocardium will recover spontaneously after a few days. Patients full recovery of function may occur following revascularization. The
with hibernating myocardium often present with severe triple vessel ratio of viable to total myocardium (viable plus nonviable) irrespective
disease, globally depressed LV function, and prominent dyspnea but of wall thickness in the dysfunctional region may therefore be more

262
CHAPTER 21  Myocardial Viability 263

Before revascularization CMR After revascularization


Long-axis view Long-axis view
Short-axis view 4-Chamber 2-Chamber Short-axis view 4-Chamber 2-Chamber

Cine diastole Cine diastole

Cine systole Cine systole

Region of thinning before


revascularization (shown before
and after revascularization)
Delayed
enhancement
FIG. 21.1  A patient with subendocardial scar and regional akinesia and wall thinning. Left, Before revascu-
larization, cine-cardiovascular magnetic resonance (CMR) still frames in systole and diastole demonstrate
akinesis and thinning of the anteroseptal, anterior, and apical walls. Delayed-enhancement images demonstrate
limited scar burden (≤50%) within the thinned region. Right, After revascularization, cine-CMR still frames
demonstrate improvement in myocardial contractility along with reversal of thinning in the previously thinned
region. End-diastolic wall thickness changed from 4.5 to 9.5 mm after revascularization. (Modified from Shah
DJ, Kim HW, James O, et al. Prevalence of regional myocardial thinning and relationship with myocardial
scarring in patients with coronary artery disease. JAMA. 2013;309;909–918.)

accurate than end-diastolic wall thickness in predicting functional and red blood cells becomes evident. Small blood vessels undergo necro-
improvement. sis, and karyolysis of muscle cell nuclei can be observed. Plugging of
capillaries by erythrocytes is most pronounced in the center of the
Contractile Reserve of Viable Myocardium infarct. If reperfusion can be achieved at an early stage, the resulting
A well-known feature of viable myocardium is augmented contractility infarcts contain a mixture of necrosis and hemorrhage within zones of
in response to a suitable stimulus.11 Such stimuli include sympathomi- irreversibly injured myocytes.
metic agents5 or postextrasystolic potentiation.2 In contrast, necrotic Myocardial edema is associated with prolonged magnetic resonance
or scarred tissue will not respond to such stimulation. Today, the most (MR) relaxation times, and this leads to characteristically increased
widely used mode of stimulation is to infuse low doses of dobutamine signal intensity on MR images, which are sensitive to such changes.16
up to 10 µg/kg/min. If a contractile reserve can be elicited, the responsive By using modern T2-weighted pulse sequences, the edema associated
myocardium will usually recover function after appropriate revascu- with acute infarcts can be depicted, and this can be used to differentiate
larization.15 However, it appears that there is also some spontaneous between acute and chronic infarcts.17
improvement in the response to dobutamine over the course of infarct
healing after reperfused myocardial infarction, which may affect the No-Reflow Phenomenon and Early Hypoenhancement
accuracy of this viability marker after myocardial infarction.12 With Gadolinium
A feature of the central necrotic region within a myocardial infarct is
Noninvasive Observation of Tissue Edema intracapillary red blood cell stasis.18 Plugging of the capillaries leads to
Irreversible myocardial damage occurs after approximately 30 to 120 tissue hypoperfusion. This hypoperfusion is primarily related to the
minutes of ischemia. Very early changes can be observed by electron resulting reduced functional capillary density rather than reduced
microscopy; these changes include intracellular edema and swelling of microvascular flow rates.19 This decrease in functional capillary density
the entire cell, including the mitochondria. The sarcolemma ruptures, results in a prolonged washin time constant. This lack of reperfusion
and there is free exchange between the extracellular and intracellular despite restoration of flow in the epicardial vessel is known as the no-
compartments. In some infarcts, light microscopy reveals changes just reflow phenomenon. When the myocardium is imaged by CMR early
a few hours after the onset of ischemia; these changes are most pro- after injection with gadolinium (early gadolinium enhancement), no-
nounced at the periphery of the infarct. After 8 hours, there is edema reflow zones appear dark in comparison with the surrounding rim
of the interstitium, and infiltration of the infarct zone by neutrophils regions of the infarct.20
264 SECTION II  Ischemic Heart Disease

intensity in the myocardium because of respiratory motion or residual


Late Gadolinium Enhancement in Infarcted Tissue cardiac motion. Thus careful optimization of all imaging parameters
Gadolinium chelates are commonly used as CMR contrast agents. These is required for achieving interpretable results during T2-weighted image
metabolically inert molecules are distributed extracellularly, and they acquisition (Fig. 21.2).
shorten both T1 and T2 relaxation times. Rupture of myocyte mem-
branes leads to an increased volume of distribution of CMR contrast Late Enhancement With Gadolinium in Acute Infarction
agents with a corresponding increase in the effective voxel concentra- In the mid and late 1990s, Kim and Judd developed late enhancement
tion of such agents.21,22 Thus a higher concentration of gadolinium imaging.19,23 Within a short time, LGE CMR became widely used because
contrast agents leads to a more pronounced shortening of relaxation of its high image quality, simplicity, and high resolution allowing clear
times. CMR images are usually T1 weighted (because the RR interval demarcation of the transmural extent of necrosis and scar. The
is approximately 800 ms, which corresponds to the T1 value of myo- T1-weighted segmented inversion recovery pulse sequence that is
cardium), and this will result in a higher signal intensity of infarcted employed in most centers acquires images in mid diastole when cardiac
as compared with normal tissue once the contrast material has fully motion is minimal (Fig. 21.3).29 Segmentation of k-space makes it pos-
penetrated the infarct region. The time–concentration curve of MR sible to acquire images during a breath-hold, which reduces motion
contrast agents in infarct tissue does not correspond to that in blood or artifacts as compared with the older T1-weighted techniques. With the
normal tissue kinetics. Thus, while early hypoenhancement of infarcted appropriate choice of the inversion time and imaging 10 to 20 minutes
regions after injection of contrast material is caused by delayed contrast after the intravenous application of contrast material, the signal intensity
penetration,19 late enhancement in infarction is due to both increased of normal myocardium is nulled but the infarcted tissue becomes very
volume of distribution and slow contrast washout. The enhancement bright (Fig. 21.4). Kim and colleagues23 clearly demonstrated that this
pattern that is seen will depend on regional differences in tissue washin/ technique provides precise images of acute and subacute infarcts irre-
washout kinetics, as well as the time after injection of contrast when the spective of transmurality and reperfusion status (Fig. 21.5).
image is acquired. LGE has now been extensively validated in animal The time between contrast injection and starting the imaging process
and human studies, and with improved imaging sequences (notably is important, as contrast concentration and hence enhancement of a
the use of inversion recovery to null signal from normal myocardium), region vary over time. Normal regions with normal contrast washin
the signal-to-noise ratio of enhanced to unenhanced tissue is dramati- and washout reach a constant enhancement within 2 to 3 minutes.19
cally higher than with previous sequences, at approximately 500%. This The border zones of an infarct, however, have longer time constants
has led to greatly improved image quality and a substantial increase for washin and washout as compared with normal, and these time
in use of the technique. In animal experiments, the area of LGE has constants are even longer in the center of the infarct (see later). Con-
been shown to correlate closely with areas of infarction,23 and for the sequently, both core and border zone of the infarct appear dark during
first time in vivo, high-quality imaging of the distribution of scar the early phase of contrast washin (Fig. 21.6). Later, core and normal
is possible. myocardium have similar signal intensities, but the border zone begins
showing some enhancement. When contrast washout begins in normal
High-Energy Phosphates and Viability myocardium at about 10 minutes after injection, core and border zones
The primary energy reserve in living myocardial cells is stored in the of the infarct are enhanced, and this persists in the core of the infarct
form of creatine phosphate and adenosine triphosphate (ATP). Deple- at late stages of contrast washout. At this time, the border zones have
tion of total myocardial creatine, creatine phosphate, and ATP follows already returned to near-normal signal intensities.
severe ischemic injury, as shown in biopsy samples obtained from patients There has been debate about whether LGE occurs exclusively in
during cardiac surgery or necropsy. Using 31P magnetic resonance spec- regions with myocardial necrosis or also in edematous viable border
troscopy (MRS), it is possible to measure the myocardial content of zones around infarcted areas. However, the observation by Saeed and
phosphocreatine and ATP.24 1H-MRS has a higher sensitivity than 31P-MRS colleagues30 that the Gd-DTPA-enhanced region overestimates true
and has the ability to detect the total pool of phosphorylated plus infarct size by approximately 8% was not supported by other animal
unphosphorylated creatine in skeletal and cardiac muscle.25 MRS is studies.31 Fieno and colleagues32 compared ex vivo CMR with triphen­
currently not used clinically and is not discussed further in this chapter. yltetrazolium chloride–stained sections and confirmed that the spatial
extent of enhancement was the same as the spatial extent of infarction
CARDIOVASCULAR MAGNETIC RESONANCE TO at every stage of healing from day 1 to 8 weeks. The ischemic area at
DETECT VIABLE MYOCARDIUM IN ACUTE risk was defined by fluorescent microparticles injected into the left
MYOCARDIAL INFARCTION atrium with the infarct-related artery occluded. Enhanced regions were
smaller than the ischemic area at risk at every stage of healing (Fig.
Signal Intensity Changes on T2-Weighted Images 21.7). Image intensities of viable myocardium within the risk region
Myocardial edema accompanies acute myocardial necrosis. On were the same as those of remote, normal myocardium. A possible
T2-weighted spin echo images, the increased water content leads to an explanation for the discrepant findings of Saeed and colleagues and
increase in signal intensity.26 T2-weighted spin echo images acquired the group of Kim and Judd is that Saeed and colleagues used a different
early after myocardial infarction (within 10 days) demonstrate that the pulse sequence than that found to be highly and reproducibly accurate
infarct site is a region of high signal intensity in comparison with normal for infarct sizing.
myocardium.27 The advent of modern rapid pulse sequences, which It is important to note that accurate infarct imaging requires constant
produce a higher contrast between edematous and normal myocardium, adjustment of the TI if imaging cannot be completed within 5 minutes.
has led to some revival of T2-weighted CMR.28 This is necessary because the null point of normal myocardium depends
However, it is important to keep in mind that there are several on the concentration of the contrast agent at any given point in time.
potential pitfalls to the acquisition and interpretation of T2-weighted As contrast washout from normal myocardium occurs at a comparatively
images, including the necessity to differentiate the signal from slowly rapid pace, TI increases continuously, necessitating corresponding
flowing blood in the ventricle from increased signal intensity from a increases of TI to achieve perfect nulling of normal myocardium. Typical
region of infarction and to recognize artifactual variation of signal TI values range from 310 ms at 10 minutes to 385 ms at 30 minutes
CHAPTER 21  Myocardial Viability 265

Black-Blood T2-Weighted MRI: Procedure for Optimizing Image Timing

Step 1: At the same short-axis location, acquire multiple images in 50 ms increments throughout diastole.
R-wave R-wave
Systole Diastole

ECG

Step 2: Select optimal imaging time in cardiac cycle when motion (signal dropout) is minimized.
Step 3: Repeat Steps 1−2 if signal dropout recurs for different short-axis location.

Example
Diastole
Image readout time 450 ms 500 ms 550 ms 600 ms
after the R-wave

T2-weighted MRI

Infarction

Pathology Delayed-
enhancement MRI

FIG. 21.2  The steps used to optimize image timing for black-blood T2-weighted cardiovascular magnetic
resonance (CMR). This involved choosing a midventricular short-axis slice, obtaining test images throughout
diastole, and inspecting the images to determine the best timing of readout based on the absence of signal
loss (dropout) artifact. Signal loss was determined visually and defined as a myocardial region with obviously
reduced signal on one but no other test images. Example images from one subject demonstrate that small
changes in timing (≈50 ms) can affect the presence and location of dropout artifact (green arrowhead). Note
that when dropout artifact is present, the remaining myocardium appears hyperintense (yellow arc), which
could confound interpretation. This usually leads to overestimation of T2-abnormality size because dropout
typically affects only a fraction (<50%) of the myocardium in the slice. The image with optimal timing (red box)
demonstrates no region with dropout and an obvious area of hyperintensity (red arc) in the region of acute
infarction as demonstrated by pathology and in vivo late enhancement CMR (red arrows). ECG, Electrocar-
diogram; MRI, magnetic resonance imaging. (From Kim HW, Van Assche L, Jennings RB, et al. Relationship
of T2-weighted MRI myocardial hyperintensity and the ischemic area-at-risk. Circ Res. 2015;117:254–265.)

R R R
ECG

Trigger
Nonselective Nonselective
α1 α2 α12 α23
180° inversion 180° inversion
… …
1 2 12 23 …

Trigger TI
delay
FIG. 21.3  Segmented inversion recovery fast gradient echo sequence
with TI set to null normal myocardium after contrast agent administra-
tion. ECG, Electrocardiogram. (From Simonetti OP, Kim RJ, Fieno DS,
et al. An improved MR imaging technique for the visualization of myo-
cardial infarction. Radiology. 2001;218:215–223.)
266 SECTION II  Ischemic Heart Disease

FIG. 21.4  Nontransmural inferior myocardial infarct (left panel). This patient presented with acute onset
of ST-elevation myocardial infarction and was revascularized by catheterization approximately 120 minutes
after first onset of symptoms. Note the preserved wall thickness (arrow) in the region with subendocardial
late gadolinium enhancement. The transmural inferolateral myocardial infarct displayed in the right panel is
several months old. Note the thinning (arrow) of the infarcted area, which is considered to be a hallmark of
transmural infarcts.

FIG. 21.5  Postmortem images of a dog with a 3-day-old myocardial infarct. Left, Triphenyltetrazolium chloride
(TTC)-stained slices of the left ventricle showing the infarct as nonstained white areas, whereas normal
myocardial cells stain red. Slices are arranged from base to apex, starting in the upper left and advancing
from left to right and then from top to bottom. Right, Insert showing a magnification. There is excellent
matching of the enhanced areas in the ex vivo magnetic resonance images with the necrotic zones in the
necropsy slices. (From Kim RJ, Fieno DS, Parrish TB, et al. Relationship of MRI delayed contrast enhance-
ment to irreversible injury, infarct age, and contractile function. Circulation. 1999;100[19]:1992–2002.)
A B C
FIG. 21.6  Temporal changes in signal intensity after 0.2 mmol/kg gadolinium-diethylenetriaminepentaacetic
acid was administered intravenously in a patient after primary percutaneous transluminal coronary angioplasty
plus stenting of the left anterior descending coronary artery for anterior myocardial infarction. Magnified
three-chamber view at 2 minutes (A), 15 minutes (B), and 30 minutes (C). (From Beek AM, Kühl HP, Bonda-
renko O, et al. Delayed contrast-enhanced magnetic resonance imaging for the prediction of regional functional
improvement after acute myocardial infarction. J Am Coll Cardiol. 2003;42[5]:895–901.)

MRI 1

100 µm
TTC 2

UV 3

FIG. 21.7  Dog with a 1-day-old reperfused infarct. Upper left, The ex vivo magnetic resonance enhancement
image. Middle left, The matching triphenyltetrazolium chloride (TTC)-stained myocardial slice. Bottom left,
The myocardium at risk as the blue-appearing region without fluorescent microparticles. Right, Light microscopy
views of region 1 (not at risk, not infarcted), region 2 (at risk but not infarcted), and region 3 (infarcted).
Arrows point to contraction bands. MRI, Magnetic resonance imaging; UV, ultraviolet. (From Fieno DS, Kim
RJ, Chen EL, et al. Contrast-enhanced magnetic resonance imaging of myocardium at risk: distinction between
reversible and irreversible injury throughout infarct healing. J Am Coll Cardiol. 2000;36[6]:1985–1991.)
268 SECTION II  Ischemic Heart Disease

(Fig. 21.8). With appropriate adjustment of TI, there are no differences


in infarct size measurement by contrast-enhanced MR between 10 and No-Reflow by Cardiovascular Magnetic Resonance
30 minutes after contrast injection (Fig. 21.9).33 If TI is not adjusted, CMR of acutely necrotic myocardium sometimes reveals a central core
the spatial extent of enhancement decreases by up to 30% over time of low signal intensity surrounded by the typical bright infarct zone (Fig.
after contrast application.34 21.10). In the center of the infarct region, myocytes and capillaries may
undergo necrosis simultaneously because of profound and sustained
ischemia. In that situation, capillaries become occluded by dying blood
1.0 cells and debris, to the extent that even with restoration of epicardial
Plasma (Gd-DTPA) (mmoI/L)

0.9 blood flow, the infarct core will not promptly reperfuse. This area of
l 420
retica microvascular obstruction is called the no-reflow region.18 Microvascular
Theo

Inversion time (ms)


0.8 385 obstruction following acute infarction correlates with greater myocardial
0.7 370 damage by echocardiography and poorer global LV function in the
early postinfarction phase.35 The central low signal intensity region
0.6
seen on CMR images within the first 2 to 3 minutes after contrast
0.5 316 320 injection corresponds to experimentally produced no-reflow regions.36
CMR enables in vivo monitoring of the time course of microvascular
0.4
obstruction. It could be shown that this zone increases 3-fold during
0.3 270 the 48 hours after reperfusion, indicating progressive microvascular
0 5 10 15 20 25 30 35 40 45 50 55 60 injury well beyond coronary occlusion and reflow. Moreover, the extent
Time after injection of Gd-DTPA (minutes) of microvascular obstruction is the strongest predictor of LV volume
FIG. 21.8  Solid line, Monoexponential fit to serum contrast agent con- at 10 days after reperfusion. Large areas of microvascular obstruction
centration (left-hand y-axis) as function of time after administration cal- within infarcts lead to significant reductions of radial thickening in
culated (0.125 mmol/kg gadolinium diethylenetriaminepentaacetic acid adjacent noninfarcted regions.37 CMR could also prove in a dog model
[Gd-DTPA]). (Based on data of Weinmann HJ, Laniado M, Mützel W. that intraaortic balloon counterpulsation improves myocardial perfu-
Pharmacokinetics of GdDTPA/dimeglumine after intravenous injection sion at the tissue level and reduces the extent of no-reflow caused by
into healthy volunteers. Physiol Chem Phys Med NMR. 1984;16:167–
microvascular obstruction.38
172.) Dashed line, Magnetic resonance inversion time (right-hand y-
axis) calculated based on data of solid line. Solid circles indicate data CMR may also be used to study the effects of the no-reflow phe-
measured in the study of Mahrholdt and colleagues. (From Mahrholdt nomenon in patients with acute infarcts. Wu and co-workers39 studied
H, Wagner A, Holly TA, et al. Reproducibility of chronic infarct size 44 patients by using perfusion CMR. Almost all of these patients had
measurement by contrast-enhanced magnetic resonance imaging. Cir- thrombolysis or direct angioplasty. To study LV remodeling, 17 patients
culation. 2002;106[18]:2322–2327.) underwent repeated CMR 6 months after the initial study. Microvascular

SCAN 1

SCAN 2 - Patient repositioned, later time, different scanner operator

FIG. 21.9  Full set of short-axis views of cardiovascular magnetic resonance (CMR) scan 1 performed
9 minutes after contrast injection and CMR scan 2 acquired after removing and repositioning of the patient
in the scanner with images acquired 32 minutes after contrast injection. Inversion time was increased
continuously to achieve perfect nulling of the myocardium. Presence, location, and size of the enhanced
region (arrows) were similar in both CMR scans. (From Mahrholdt H, Wagner A, Holly TA, et al. Reproduc-
ibility of chronic infarct size measurement by contrast-enhanced magnetic resonance imaging. Circulation.
2002;106[18]:2322–2327.)
CHAPTER 21  Myocardial Viability 269

FIG. 21.10  Zone of no reflow represented by a central core of low signal intensity surrounded by the typical
bright infarct zone a few days after transmural right circumflex infarct (left panel, arrow). The no-reflow
phenomenon is detectable up to 2 to 4 weeks after acute infarction and is associated with poor prognosis
(see text for details). Subsequently, at follow-up 12 weeks after infarction (right panel), the no-reflow zone
has resolved.

100 Late Gadolinium Enhancement and Recovery of Function


The transmural extent of myocardial infarction as defined by LGE CMR
Event-free survival (%)

80 predicts the likelihood of improvement of contractile function. Spon-


P < .01
taneous recovery of function can be expected after ischemic injury in
60 a segment with a limited amount of subendocardial scar. In contrast,
a segment with transmural infarction will usually not recover wall
40 thickening.42 This concept was confirmed in a dog experiment of acute
myocardial infarction. Segments with less than 25% of enhancement
20 on day 3 after induction of the infarct had an 87% probability of improve-
ment by day 28.43 In contrast, no segment with 100% transmurality of
0 enhancement improved.
0 5 10 15 20 25 Choi and colleagues44 confirmed these experimental findings in 24
patients who presented with their first myocardial infarction and were
Months
successfully revascularized. Improvement in segmental contractile func-
FIG. 21.11  Event-free survival (clinical course without cardiovascular
tion at 2 to 3 months was inversely related to the transmural extent of
death, reinfarction, congestive heart failure, or stroke) for patients with
and without cardiovascular magnetic resonance microvascular obstruc- infarction as demonstrated by LGE within 7 days of the acute event.
tion. (From Wu KC, Zerhouni EA, Judd RM, et al. Prognostic significance Improvement in global contractile function, as assessed by ejection
of microvascular obstruction by magnetic resonance imaging in patients fraction and mean wall thickening score, was not predicted by peak
with acute myocardial infarction. Circulation. 1998;97:765.) creatine kinase-MB or by total infarct size, as defined by CMR. Instead,
the best predictor of global improvement was the extent of dysfunctional
myocardium that was not infarcted or had infarction comprising less
obstruction was defined as hypoenhancement seen 1 to 2 minutes than 25% of LV wall thickness.
after contrast injection. Infarct size was assessed as percent LV mass
enhanced 5 to 10 minutes after contrast. Patients with microvascular Combining T2-Weighted Images and Late Enhancement
obstruction (N = 11) had more cardiovascular events than did those Images for Evaluation of Myocardial Salvage After
without (45% vs. 9%, P = .02). The risk of adverse events increased Early Reperfusion
with infarct extent (30%, 43%, and 71% for small [N = 10], mid-sized Friedrich and colleagues studied patients with reperfused acute myo-
[N = 14], and large [N = 14] infarcts, respectively; P < .05). Even after cardial infarction using a T2-weighted breath-hold short TI inversion
infarct size was controlled for, the presence of microvascular obstruc- recovery pulse sequence and late enhancement images.45 In comparison
tion remained a prognostic marker for postinfarction complications with the zone of high signal intensity on late enhancement images cor-
(Fig. 21.11). Rogers and colleagues40 confirmed the adverse outcome responding to the area of irreversible ischemic damage, the high signal
of patients with low signal intensity regions within the infarct zone intensity zone on T2-weighted images was larger, and was speculated
after injection of gadolinium contrast. However, Gerber and associates41 to correspond to the ischemic area of reversible damage (ischemic area
reported that early gadolinium enhancement in the infarct area was at risk). Thus Friedrich et al.,45 as well as other authors,46 concluded
inferior to LGE for predicting functional improvement in dysfunctional that the combination of these two techniques may allow for quantify-
segments in patients studied 4 days after acute myocardial infarction. ing the extent of the salvaged myocardial area after early reperfusion
Receiver operating characteristic analysis showed an accuracy of 74% therapy (Fig. 21.12).
for the absence of late enhancement but only 49% for the absence of However, for this concept to hold true it is necessary that the entire
early hypoenhancement. ischemic area at risk has substantial edema and that the high signal
270 SECTION II  Ischemic Heart Disease

FIG. 21.12  Short-axis slices of a T2-weighted image (upper left panel) demonstrating edema contours (red,
lower left panel), which are speculated to represent the ischemic area risk, and the corresponding late
enhancement image (upper right panel) with endocardial (green), epicardial (yellow), papillary (dark blue), and
infarcted (red) contours (lower right panel) in a patient with anterior myocardial infarction. The suspected
myocardial salvage was calculated from the ischemic area at risk on T2-weighted images minus infarct area
on late enhancement images. (From Eitel I, Desch S, Fuernau G, et al. Prognostic significance and determi-
nants of myocardial salvage assessed by cardiovascular magnetic resonance in acute reperfused myocardial
infarction. J Am Coll Cardiol. 2010;55:2470–2479.)

intensity zone on T2-weighted images is related to the ischemic area Thus based on the pathophysiology and the recent results from
at risk measured by microspheres in an animal model as the gold stan- the Duke group, T2-weighted imaging should not be used to measure
dard. Whalen et al.47 reported that there is an increase in tissue water myocardial salvage, either to inform patient management decisions, or
of about 88% in myocardial regions that are completely infarcted (post- to evaluate novel therapies for acute myocardial infarction. The earlier
mortem data). In contrast, Jennings et al.48 found an increase of tissue reports of other groups45,46 (see also Fig. 21.12), which are not in line
water of only 9% in reversibly injured myocardium which persisted for with the pathophysiology and the recent gold standard animal data
less than 24 hours after reperfusion. from Duke described above, are most likely explained by the frequent
Based on the data from Whalen and Jennings, there is at least a 9-fold pitfalls in the acquisition and interpretation of T2-weighted images
difference in tissue water content between reversibly and irreversibly (see also Fig. 21.2). In the future new T2-weighted mapping techniques
injured myocardium, casting serious doubt that there is any pathophysi- may overcome the limitations of conventional T2-weighted imaging
ologic basis for the quantification of the area at risk, or the salvaged area described earlier.50
using T2-weighted imaging. These doubts are supported by experimental
data from the Duke group,49 who recently studied 21 canines and 24 Wall Thickness and Wall Thickening Measurements
patients to determine whether T2-weighted images delineate the area at After an acute ischemic event, structural changes occur within the
risk. Matching the pathophysiology described by Whalen and Jennings, infarct zone, and infarct healing with scar formation is completed
they found no relationship between the transmural extent of T2-intense by approximately 3 to 4 months.51 Thinning of the infarct region
regions and that of the ischemic area at risk (P = .97) because the tissue may occur early, especially in large anterior myocardial infarcts, but
water content of reversibly injured myocardium is only increased by transient thickening of the infarcted segment because of edema52 has
9% for less than 24 hours after the acute event. Instead, there was a also been observed. The consequence of infarct thinning is an increase
strong correlation with that of infarction (P < .0001) because tissue in the size of the infarcted segment, known as infarct expansion.53
water content in infarcted myocardium is increased by 88%. There was However, even in transmural infarcts, infarct expansion may not occur
a fingerprint match of T2-intense regions with the intricate contour in patients with open infarct-related arteries. Such patients are encoun-
of infarcted regions by late enhancement and pathology (Fig. 21.13). tered more often today with the widespread use of thrombolysis and
CHAPTER 21  Myocardial Viability 271

Pathology: Microspheres
Transmural AAR

Pathology: TTC staining


Subendocardial MI

T2-weighted-MRI
(black blood)
Subendocardial T2-hyperintensity

T2-weighted-MRI
(bright blood)
Subendocardial T2-hyperintensity

Delayed-enhancement-MRI
Subendocardial MI

FIG. 21.13  A comparison between cardiovascular magnetic resonance (CMR) and pathology at multiple
short-axis level in one subject with substantial salvage. The area at risk (AAR) by histopathology (top row,
orange outline) is nearly 100% transmural at every short-axis location, whereas T2 intense (third and fourth
rows) and infarcted regions (second and bottom row) are clearly nontransmural (yellow arrows). The most
basal slice shows a particularly large discrepancy between the full-thickness AAR and the region of infarction,
which is tiny and subendocardial. For all slices, T2 intense regions closely resemble the shape of infarction,
not the AAR. MI, Myocardial infarction; MRI, magnetic resonance imaging; TTC, triphenyltetrazolium chloride.
(From Kim HW et al. Relationship of T2-weighted MRI myocardial hyperintensity and the ischemic area-at-
risk. Circ Res. 2015;117:254–265.)

angioplasty of the infarct artery.54 Therefore transmural necrosis in order to not assume that the entire region perfused by an occluded
and nontransmural necrosis may have the same wall thickness early coronary artery is completely scarred. Frequently, myocardial cells in
after myocardial infarction. Both conditions may also be associated the border zone survive, and ischemia of this border zone alone may
with complete absence of resting function early after the acute event. cause substantial symptoms in a patient. Moreover, wall thinning may
However, it should be remembered that even a small amount of wall also occur in some patients who only have thin subendocardial scar
thickening in a region of interest indicates the presence of residual yet show substantial end-diastolic wall thinning in a region of severely
contracting cells and hence of viable myocardium. reduced wall motion. Therefore in a patient with single-vessel disease,
previous myocardial infarction, and anginal symptoms, restoration of
CARDIOVASCULAR MAGNETIC RESONANCE IN blood flow by re-establishing patency of the occluded artery may be
justified in the presence of ischemia to improve symptoms, despite
CHRONIC MYOCARDIAL INFARCTION evidence of substantial wall thinning of the infarct zone.
As previously discussed, chronic myocardial infarcts are structurally
different from acute myocardial infarcts. The most obvious difference Myocardial Wall Thickness as a Feature of
is that chronic transmural infarcts may be very thin, owing to infarct Viable Myocardium
expansion and remodeling.55 Consequently, this feature can be detected The hypothesis that thinned and akinetic myocardium usually14 rep-
by CMR and can be used to distinguish between chronic transmural resents chronic scar has been tested by comparing CMR findings with
scar and residual viable myocardium in the infarct area. However, caution those obtained by PET and single-photon emission computed tomog-
must be used when observing a small area of pronounced wall thinning raphy (SPECT) in identical myocardial regions.56,57 Comparison of CMR
272 SECTION II  Ischemic Heart Disease

with scintigraphic techniques is easily accomplished because identical


regions can be matched, owing to the three-dimensional nature of both Contractile Reserve During Low-Dose
techniques. To define transmural scar by end-diastolic wall thickness, Dobutamine Infusion
a cutoff value of 5.5 mm was selected. This value corresponded to the Although severely reduced end-diastolic wall thickness is helpful in
mean end-diastolic wall thickness in normal individuals minus 2.5 identifying scarred myocardium, the positive predictive value of a pre-
standard deviations. It also corresponded well to the wall thickness of served end-diastolic wall thickness for predicting recovery of function
less than 6 mm found in a histopathologic study of transmural chronic following revascularization is disappointingly low. However, CMR can
scar.53 Regions with a mean end-diastolic wall thickness of less than measure wall thickening not only at rest but also during low-dose dobu-
5.5 mm had a significantly reduced fluorodeoxyglucose (FDG) uptake tamine infusion. The advent of fast CMR sequences now permits comple-
as compared with regions with an end-diastolic wall thickness of ≥5.5 mm tion of such a protocol within approximately 30 to 45 minutes. The
(Fig. 21.14).57 In 29 of 35 patients (83%) studied, the diagnosis of viability sensitivity of dobutamine CMR for detection of viable myocardium as
based on FDG uptake was identical to the one based on myocardial defined by a normalized FDG uptake on PET images is 81% with a
wall thickness as assessed by CMR. Importantly, relative FDG uptake specificity of 95%.57 When recovery of wall thickening following revas-
did not differ between segments with systolic wall thickening at rest or cularization was considered to be the gold standard, the sensitivity of
akinesia at rest as long as wall thickness was preserved. However, 18% dobutamine CMR in predicting recovery of function after revascular-
of patients in whom viability is studied and who show diastolic wall ization was 89% with a specificity of 94%. The latter analysis was patient
thinning of ≤5.5 mm have nevertheless substantial amounts of viable related, which is clinically more meaningful than a segment-by-segment
myocardium, and improvement of wall thickening will occur following analysis.
revascularization.14 Using a healthier patient group and a different methodology, Trent
These findings were extended in another population of patients who and colleagues59 found less satisfactory values for sensitivity and speci-
underwent revascularization and control CMR at 3 months after revas- ficity of wall motion (50% and 72%, respectively) or wall thickening
cularization.58 Of 125 segments with an end-diastolic wall thickness (50% and 68%, respectively). This was possibly related to the fact that
less than 5.5 mm in 43 patients with chronic infarcts, only 12 segments they used higher dobutamine doses. They also included segments with
recovered (corresponding to a negative predictive accuracy of 90% for worsening wall dynamics, which were considered viable. In contrast,
the finding of end-diastolic wall thinning to predict transmural scar). Baer and colleagues58 looked only at akinetic segments, which by defini-
In contrast, the positive predictive accuracy was only 62% for preserved tion could not become worse.
end-diastolic wall thickness of ≥5.5 mm for predicting the presence of
viable myocardium with the potential for recovery. This is likely explained Late Gadolinium Enhancement in Chronic Infarction
by the fact that simply looking at wall thickness by CMR will not define Similar principles apply to the assessment of myocardial infarction in
the thickness of the remaining viable rim, which is the most important the chronic phase as to assessment shortly after the event. Late enhance-
factor in determining whether a segment will recover function or not.12,14 ment of the infarct zone is also seen in chronic infarcts7,60,61 because of
Definition of this viable rim is possible only by using Gd-enhanced the continuing increase in partition coefficient and delayed contrast
CMR and will be discussed later. agent kinetics. The tissue in chronic scar consists mainly of fibroblasts
and collagen surrounded by a large intercellular space. Because gado-
linium contrast agents will diffuse into this space, enhancement may
P < .0001 simply be related to the larger volume of distribution in comparison
100 to normal myocardium with its tightly packed myocytes, which the
n.s. P < .001
contrast agent cannot enter.
Rel. FDG-uptake (%)

80 LGE CMR is able to identify patients with healed myocardial infarction


with high accuracy.62 Moreover, this technique may permit noninvasive
60 74% differentiation between ischemic and nonischemic cardiomyopathy. In
67%
a study of 71 subjects, 40 patients with healed myocardial infarction
40 were prospectively enrolled after enzymatically proven necrosis and
47% were imaged 3 ± 1 months (N = 32) and/or 14 ± 7 months (N = 19)
20 WTh WTh WTh later.61 They were compared with 20 patients with nonischemic car-
> 1 mm < 1 mm < 1 mm diomyopathy and 11 normal volunteers. A total of 29 of 32 patients
N = 1713 N = 234 N = 248
0 (91%) with 3-month-old infarcts (13 non-Q-wave) and all 19 with
DWT > 5.5 mm DWT > 5.5 mm DWT < 5.5 mm 14-month-old infarcts (8 non-Q-wave) exhibited LGE. In patients in
(viable) (viable) (scar) whom the infarct-related artery was determined at angiography, 24 of 25
FIG. 21.14  Normalized uptake of 18F-fluorodeoxyglucose (FDG) stratified patients with 3-month-old infarcts (96%) and all 14 with 14-month-old
by end-diastolic wall thickness as measured from gradient echo magnetic infarcts had LGE in the correct territory. None of the 20 patients with
resonance images. Regions with preserved wall thickening (WTh) of nonischemic cardiomyopathy or the 11 volunteers had LGE. Regardless
≥1 mm and preserved end-diastolic wall thickness (DWT) of >5.5 mm of the presence or absence of Q-waves, the majority of patients with
had a relative FDG uptake similar to that of regions without wall thicken- LGE had only nontransmural involvement. Normal LV contraction was
ing (WTh <1 mm) but preserved end-diastolic wall thickness. In contrast, visualized in 7 patients with 3-month-old infarcts (22%) and 3 with
akinetic regions (WTh <1 mm) with reduced end-diastolic wall thickness 14-month-old infarcts (16%), but in these cases, LGE was limited to
had significantly reduced FDG uptake (mean 47%), indicating scar for-
the subendocardium. McCrohon and colleagues63 demonstrated in a
mation. (Data from Baer FM, Voth E, Schneider CA, et al. Comparison
of low-dose dobutamine–gradient-echo magnetic resonance imaging
cohort of 90 patients with heart failure that all patients with coronary
and positron emission tomography with [18F] fluorodeoxyglucose in artery disease had LGE primarily with a subendocardial or transmu-
patients with chronic coronary artery disease: a functional and morpho- ral pattern, whereas patients with dilated cardiomyopathy (no stenosis
logical approach to the detection of residual myocardial viability. Circula- by coronary angiography) had either no enhancement (59%), patchy
tion. 1995;91:1006–1015.) or mid wall enhancement not corresponding to a coronary perfusion
CHAPTER 21  Myocardial Viability 273

bed (28%), or enhancement patterns indistinguishable from those of segments (14%) showed evidence of hypoenhancement. At 6 months
patients with ischemic cardiomyopathy (13%). The patchy and mid wall after revascularization, 57% of dysfunctional segments improve con-
enhancement patterns are often found in patients with biopsy-proven traction by at least one grade. There was a strong correlation between
myocarditis.64 This indicates that CMR may give profound new insights the transmural extent of hypoenhancement and the recovery in regional
into the causes of LV failure.65 function at 6 months. Of a total of 96 previously dysfunctional but
nonenhancing or minimally hypoenhancement myocardial segments
Viability by Late Gadolinium Enhancement that did not improve regional function at 6 months, 35 (36%) dem-
Cardiovascular Magnetic Resonance and onstrated new perioperative hypoenhancement in the early postoperative
Recovery Following Revascularization CMR scan, indicating that failure of recovery may be related to peri-
Prediction of the myocardial response to revascularization remains the operative injury of recoverable myocardium. In a secondary analysis
holy grail of viability testing. Late enhancement CMR performs well of these patients, Pegg et al.68 showed that the 21 of 33 patients who
in this discipline. In the first study looking at the ability of contrast improved ejection fraction by more than 3% at 6 months had ≥10/16
CMR to predict improvement in LV function after revascularization, viable and normal segments (based on the segmental transmural team
Kim and colleagues66 showed that the likelihood of functional recovery viability cutoffs of <50%). The presence of this amount of viable and
decreased progressively as the transmural extent of LGE observed before normal segments predicted improvement in left ventricular ejection
revascularization increased (Fig. 21.15). Approximately 80% of segments fraction (LVEF) with a sensitivity of 95% and a specificity of 75%. In
with no LGE improved function after revascularization (Fig. 21.16), contrast, patients with fewer such segments do not usually improve
whereas if more than 75% of the transmural tissue was enhanced, only global LV function postrevascularization.
a small percentage improved with revascularization (Fig. 21.17). The More recently, Gerber and coworkers69 further extended the obser-
second important study looking at the value of LGE CMR in predicting vations by Kim66 and Selvanayagam,67 which were made in patients
myocardial viability after revascularization extended the data reported with moderately reduced ejection fractions (EFs) (43 ± 13% and 62
by Kim by adding a second LGE CMR examination following coronary ± 12%, respectively), by focusing on patients with severely reduced
artery bypass surgery.67 Some 52 patient were studied by preoperative EF (24 ± 7%). It is really the latter group in whom viability testing is
day 6 and 6 months postoperative cine CMR for functional assessment clinically most meaningful because these patients have most to gain
and for assessment of irreversible myocardial injury. Preoperatively, from successful revascularization of viable myocardium and most to
611 segments (21%) had abnormal regional function, whereas 421 lose from perioperative morbidity and mortality if only scarred tissue

Segments with
All dysfunctional severe hypokinesia, Segments with
segments akinesia, or dyskinesia akinesia or dyskinesia
)
12
of
2
(1
8)

100
14
of

)
9)

28
28
32

of
(1
of

3
(2
56

80
(2

)
86
3)
18
Improved contractility (%)

of
of

6
(5
09
(1

60
0)

)
)

20
68
11

of
of
of

(9
9
6

(2
(4

40
)
4)

03
12

20
of
of

5 4)
0
3

(1
(1

of
58

)
)

46
57

(4
of

of
of
(1

(0
(0

0
00

00

00
0

51 0

51 0

51 0
25

25

25
76 5

76 75

76 75
–5

–5

–5
–7
–1

–1

–1
1–

1–

1–


26

26

26

Transmural extent of hyperenhancement (%)


FIG. 21.15  Relationship between the transmural extent of enhancement before revascularization and the
likelihood of increased contractility after revascularization. Data are shown for all 804 dysfunctional segments
and separately for the 462 segments with at least severe hypokinesia and the 160 segments with akinesia
or dyskinesia before revascularization. For all three analyses, there was an inverse relationship between the
transmural extent of enhancement and the likelihood of improvement in contractility. (From Kim RJ, Wu E,
Rafael A, et al. The use of contrast-enhanced magnetic resonance imaging to identify reversible myocardial
dysfunction. N Engl J Med. 2000;343:1445, with permission. Copyright 2000 Massachusetts Medical Society.
All rights reserved.)
274 SECTION II  Ischemic Heart Disease

BEFORE REVASCULARIZATION
End diastole End systole No hyperenhancement

A
AFTER REVASCULARIZATION
End diastole End systole

B
FIG. 21.16  Representative cine images and contrast-enhanced images obtained by magnetic resonance in
a patient with reversible ventricular dysfunction. This patient had severe hypokinesia of the anteroseptal wall
(arrows), and this area was not enhanced before revascularization (A). The contractility of the wall improved
after revascularization (B). (From Kim RJ, Wu E, Rafael A, et al. The use of contrast-enhanced magnetic reso-
nance imaging to identify reversible myocardial dysfunction. N Engl J Med. 2000;343:1445, with permission.
Copyright 2000 Massachusetts Medical Society. All rights reserved.)

is revascularized. This cohort of patients was similar to the cohort of death of 4.56 (95% confidence interval [CI], 1.93 to 10.8). Thus in
studied in the Surgical Treatment for Ischemic Heart Failure (STICH) patients with ischemic heart disease and severely reduced ejection frac-
trial (see later). However, treatment allocation was not randomized tion the presence of dysfunctional viable myocardium as assessed by
and all patients underwent LGE CMR instead of echocardiography/ LGE CMR is an independent predictor of mortality in medically treated
SPECT for assessment of viability. Of the 144 consecutive patients, 86 patients and outcome is improved by complete revascularization. The
patients underwent complete revascularization of dysfunctional myo- findings in the study of Gerber et al.70 were similar to the results of a
cardium, most by coronary artery bypass grafting (CABG), and 58 previous metaanalysis where patients with myocardial viability treated
patients were treated medically. As in most viability studies the type of medically had the worst outcome which could significantly be improved
medical treatment was not specified and it is possible that some of the by revascularization. Patients without viability had intermediate rates
apparent benefits of revascularization in earlier observational studies, of mortality regardless of treatment.
at least in part, contributed to deficits in the quality of medical therapy A recent metaanalysis looking at 24 CMR studies evaluating myo-
compared with current best practice.8 Patients were considered to have cardial viability with 698 patients (11 studies using LGE, 9 studies using
viable myocardium when four or more dysfunctional segments were low-dose dobutamine, and 4 studies using end-diastolic wall thickness)
viable (transmural of LGE ≤50%). Three-year survival was worse in came to the conclusion that LGE CMR provides the high sensitivity as
medically treated patients with dysfunctional viable (N = 26) than with well as the highest negative predictive value (95% and 90%, respectively)
nonviable myocardium (N = 20) (survival 48% vs. 77%, P = .02). In for predicting improved segmental LV contractile function after revas-
contrast, survival in revascularized patients was similar whether myo- cularization.71 CMR looking at end-diastolic wall thickness followed,
cardium was viable or not (88% and 71% survival, respectively, P = ns whereas low-dose dobutamine CMR demonstrated the lower sensitivity/
[nonsignificant]) (Fig. 21.18). Patients with viable myocardium under negative predictive value among the three modalities. On the other
medical treatment versus complete revascularization had a hazard ratio hand, low-dose dobutamine CMR offered the highest specificity and
CHAPTER 21  Myocardial Viability 275

BEFORE REVASCULARIZATION
End diastole End systole No hyperenhancement

A
AFTER REVASCULARIZATION
End diastole End systole

B
FIG. 21.17  Representative cine images and contrast-enhanced images obtained by cardiovascular magnetic
resonance in a patient with irreversible ventricular dysfunction. This patient had akinesia of the anterolateral
wall (arrows), and this area was enhanced before revascularization (A). The contractility of the wall did not
improve after revascularization (B). (From Kim RJ, Wu E, Rafael A, et al. The use of contrast-enhanced
magnetic resonance imaging to identify reversible myocardial dysfunction. N Engl J Med. 2000;343:1445,
with permission. Copyright 2000 Massachusetts Medical Society. All rights reserved.)

positive predictive value (91% and 93%, respectively), followed by LGE months later. Receiver operating characteristic analysis demonstrated
CMR, whereas CMR looking at end-diastolic wall thickness showed that the measurement of thickness of nonenhanced myocardium had a
the lowest of these parameters (Fig. 21.19). slightly better diagnostic accuracy for predicting improved systolic wall
thickening from scan 1 to 2 in dysfunctional segments than percent
Thickness of the Viable Epicardial Rim and transmural enhancement. Convincing evidence that the thickness
Recovery of Function of viable myocardium (viable rim) is more predictive of functional
The clear depiction of subendocardial myocardial scar by gadolinium- recovery than total regional myocardial thickness comes from the
enhanced CMR also permits clear identification of epicardially located work of Shah et al.14 They demonstrated that 18% of myocardial seg-
viable tissue. Knuesel and colleagues72 determined the segmental ments thinned to an end-diastolic wall thickness of ≤5.5 mm had
amount of viable tissue by LGE CMR and compared this to 18F-FDG only limited or no scar tissue and showed excellent recovery following
uptake and resting perfusion using 13N-ammonia. FDG uptake of revascularization.
≥50% corresponded to a viable rim thickness of 4.5 mm by LGE CMR.
Segments that had both a thick viable rim and an FDG uptake of ≥50% Comparison of Late Contrast Enhancement With Other
showed functional recovery in 85% of segments (Fig. 21.20), whereas Imaging Modalities
thin metabolically nonviable segments (FDG uptake <50%) improved The comparison of gadolinium-enhanced CMR with other modalities
function in only 13% of segments following revascularization (P < for the discrimination of viable and nonviable myocardium has been
.0005). Segments with thin viable rims but preserved FDG uptake favorable. Segmental LGE CMR correlates well with diminished
and those with a thick viable rim but reduced FDG uptake improved thallium-201 uptake measured by rest-redistribution thallium-201 SPECT
function only in 36% and 23%, respectively. Similar findings were in patients with globally reduced LV function.74 However, the much
reported by Ichikawa and colleagues,73 who studied 18 patients with better spatial resolution that permits discrimination of subendocardial
a first myocardial infarct within 1 week of the event and more than 5 and transmural infarcts62 and the lack of ionizing radiation favor CMR.
276 SECTION II  Ischemic Heart Disease

100 IR NV

CR V
80
M NV
CR NV

60
Survival (%)
IR V
Incomplete revascularization nonviable (IR NV) MV
40 Complete revascularization viable (CR V)
Medical R/ nonviable (M NV)
Complete revascularization nonviable (CR NV)
20 Incomplete revascularization viable (IR V)
Medical R/ viable (M V)

P (Mantel Cox) = .01


0
0 1 2 3
Time (years)
IR NV 5 5 3 3
CR V 68 61 48 35
M NV 20 19 14 12
CR NV 18 15 14 10
IR V 7 6 3 2
MV 26 18 14 9
FIG. 21.18  Kaplan-Meier survival curves comparing overall 3-year survival in subgroups of patients according
to treatment and presence of myocardial viability in dysfunctional myocardium. Survival was significantly
worse in patients with dysfunctional viable myocardium remaining under medical treatment or undergoing
incomplete revascularization not including dysfunctional myocardium than in other subgroups. CR, Complete
revascularization; IR, incomplete revascularization not including dysfunctional region; M, medical treatment;
NV, nonviable myocardium; R/, remaining under medical treatment; V, viable myocardium. (From Gerber B,
Rousseau MF, Ahn SA, et al. Prognostic value of myocardial viability by delayed-enhanced magnetic resonance
in patients with coronary artery disease and low ejection fraction—impact of revascularization therapy. J Am
Coll Cardiol. 2012;59:825–835, with permission of the publisher.)

Klein and colleagues75 found that the area of LGE measured by CMR 50% or more. Thus in severely impaired ventricles, low-dose dobutamine
correlated closely with myocardial infarcts defined by PET in patients CMR has a suboptimal sensitivity but a preserved high specificity at
with ischemic cardiomyopathy, but CMR showed the extent of infarct around 90%. This has been known from studies using dobutamine
transmurality more clearly (Fig. 21.21). echocardiography in which similar low sensitivity values of 50% or less
were reported. The lower sensitivity of dobutamine echocardiography
Comparison of Different Cardiovascular Magnetic Resonance in detecting viable myocardium in regions with reduced function and
Techniques for the Diagnosis of Viability perfusion may indicate that some regions of hibernating myocardium
Wellnhofer and colleagues76 compared LGE CMR with dobutamine have such delicately balanced reductions in flow and function, with
CMR (5 and 10 µg/kg/min) for assessment of myocardial viability. Both exhausted coronary flow reserve, that any catecholamine stimulation
techniques were performed in 29 patients with chronic CAD and resting to increase oxygen demands will merely result in ischemia and inability
LV dysfunction (mean LVEF, 32% ± 8%). Cine CMR imaging at rest to elicit enhanced contractile function.78 However, it is just the patients
was repeated 3 months after revascularization to determine wall motion with global severe depression of function in whom viability testing is
improvement. The transmurality of LGE was assessed visually, using a clinically most meaningful. In such patients, it has been demonstrated
five-grade scale. Similarly, wall motion was assessed visually. Using a that LGE CMR predicts improvement in severely dysfunctional seg-
cutoff value of 25% transmurality of LGE, the authors found that ments.79 In an editorial accompanying the paper by Wellnhofer and
contrast-enhanced CMR correctly identified 73% of hibernating seg- colleagues, Kim and Manning80 comment that contractile reserve has
ments, but dobutamine CMR was better and identified 85% correctly. a reduced predictive accuracy if more severe dysfunction is present at
The results for sensitivity and specificity of Wellnhofer and colleagues rest. In this most important subgroup of patients, LGE CMR appears
for dobutamine CMR are slightly worse that those reported by Baer to have a higher accuracy than dobutamine CMR. From a practical
and associates,57 whose patient group had a higher EF (42 ± 16%). point of view, LGE CMR has the additional advantage in that it does
However, the sensitivity found by Wellnhofer and colleagues for scar not require pharmacologic stress and thus involves less risk and sub-
transmurality less than 50% is much better than the 50% found by sequently less monitoring of the patient. Furthermore, it is likely easier
Gunning and colleagues,77 who studied a patient group with more and less observer dependent for interpretation.
severely depressed LV function (mean LVEF, 24% ± 8%). In contrast, Dobutamine and LGE CMR can also be used as complementary
the sensitivity of dobutamine CMR in the Wellnhofer study dropped techniques to make the diagnosis of viability. Kaandorp and associates81
sharply to values below 60% in segments with a scar transmurality of studied 48 patients with chronic coronary artery disease and a mean
CHAPTER 21  Myocardial Viability 277

LGE Studies LGE Studies


Becker 2008 Becker 2008
Bordarenko 2007 Bordarenko 2007
Gutberlet 2005 Gutberlet 2005
Kim 2000 Kim 2000
Kuhl 2006 Kuhl 2006
Pegg 2010 Pegg 2010
Sandstede 2000 Sandstede 2000
Schvartzman 2003 Schvartzman 2003
Selvanayagam 2004 Selvanayagam 2004
Wellnhofer 2004 Wellnhofer 2004
Wu 2007 Wu 2007

Summary Summary

0.8 0.85 0.9 0.95 1 0.2 0.4 0.6 0.8 1


A Sensitivity Specificity

LDD Studies LDD Studies


Baer 1998 Baer 1998
Baer 2000 Baer 2000
Gutberlet 2005 Gutberlet 2005
Lauerma 2000 Lauerma 2000
Sandstede 1999 Sandstede 1999
Sayad 1998 Sayad 1998
Schmidt 2004 Schmidt 2004
Van Hoe 2004 Van Hoe 2004
Wellnhofer 2004 Wellnhofer 2004

Summary Summary

0.4 0.5 0.6 0.7 0.8 0.9 1 0.6 0.7 0.8 0.9 1
B Sensitivity Specificity

EDWT Studies EDWT Studies


Baer 1998 Baer 1998
Gutberlet 2005 Gutberlet 2005
Klow 1997 Klow 1997
Schmidt 2004 Schmidt 2004

Summary Summary

0.7 0.75 0.8 0.85 0.9 0.95 1 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8
C Sensitivity Specificity
FIG. 21.19  Forest plots of sensitivity and specificity from 24 prospective cardiovascular magnetic resonance
studies evaluating myocardial viability. Eleven studies used late gadolinium enhancement (LGE) (A), nine
studies used low-dose dobutamine (LDD) (B), and four studies used end-diastolic wall thickness (EDWT) (C)
as viability parameters. The size of the square plotting symbol is proportional to the same size for each study.
Horizontal lines are the 95% confidence intervals, and the summary sensitivity and specificity are calculated
based on the bivariate approach. (From Romero J, Xue X, Gonzalez W, Garcia MJ. CMR imaging assessing
viability in patients with chronic ventricular dysfunction due to coronary artery disease. JACC Cardiovasc
Imaging. 2012;5:494–508, with permission.)
278 SECTION II  Ischemic Heart Disease

Contrast-enhanced MR FDG-Uptake (%) 100 Baseline MR: Diastole Baseline MR: Systole

50

A B E F
0
75
C D G H

50

Thick metabolically viable segments


with functional recovery
Scar (contrast-enhanced MR) Perfusion (K1, mL/min/100 mL) Follow-up MR: Diastole Follow-up MR: Systole
Thrombus 25
FIG. 21.20  A 59-year-old patient 5 months after an anteroseptal myocardial infarction. Contrast-enhanced
magnetic resonance (MR) (A) shows a small subendocardial zone of scar (bright) with adjacent thrombus
(black, segment 8). A thick rim of viable tissue is observed (segments 1 and 6 through 8 in A) that corre-
sponds to segments with preserved 18F-fluorodeoxyglucose (FDG) uptake (B), whereas perfusion in these
segments is reduced (D). A schematic in C illustrates this situation, with segments 1 and 6 through 8 con-
sisting of metabolically viable segments with a thick rim of viable tissue on MR. Contractile function is
severely reduced in these segments before revascularization (E and F), but has normalized 9 months after
coronary artery graft bypass surgery (G and H). (From Knuesel PR, Nanz D, Wyss C, et al. Characterization
of dysfunctional myocardium by positron emission tomography and magnetic resonance: relation to functional
outcome after revascularization. Circulation. 2003;108[9]:1095–1100.)

ejection fraction of 37% ± 10%. Regional dysfunction was present in among patients with viable myocardium was 0.64 (P = .003) (Fig. 21.22).
41% of segments, and 61% of those had contractile reserve. The likeli- However, after adjustment for other baseline variables, this significant
hood of a contractile improvement with dobutamine was highest (75%) association with mortality was no longer significant. Hence there was
in segments with small amounts of subendocardial scar, lowest (17%) no significant interaction between viability status and treatment assign-
in those with transmural scar, but intermediate (42%) in segments with ment with respect to mortality (Fig. 21.23).
intermediate infarct transmurality. The authors suggest an approach The viability substudy of STICH has been criticized for several
where LGE CMR is sufficient at both extreme ends but dobutamine methodologic problems.83 First, there were significant baseline differ-
CMR should be added to optimally predict outcome in segments with ences between the patient groups that did and did not undergo viability
intermediate amounts of scar transmurality. testing. Second, 81% of patients undergoing viability testing showed
viability, which is much higher than in previous studies where only
Viability Testing After Surgical Treatment for Ischemic about 50% of patients demonstrated significant myocardial viability.
Heart Failure Trial Thus it is conceivable that the patients enrolled in the STICH trial
Despite promising data pointing to substantial value of LGE CMR for represented a different population from previous viability studies and
predicting recovery of chronically dysfunctional myocardium, the recent that this fact may have influenced the results, which contradicted the
STICH trial employed only SPECT imaging or dobutamine echocar- results of these earlier studies. Third, unusual viability criteria were
diography for preoperative viability assessment. The STICH study employed in the STICH trial. These criteria were rather conservative,
randomized 1212 patients with an ejection fraction of less than 35% which makes it more surprising that such a high percentage of patients
and coronary artery disease suitable for CABG to receive optimal medical demonstrated significant viability. For dobutamine echocardiography,
therapy for heart failure and coronary artery disease (602 patients) or ≥5 dysfunctional segments with evidence of contractile reserve were
optimal medical therapy plus CABG (610 patients). Only 601 of the required. For SPECT, ≥11 viable segments (irrespective of baseline con-
1212 patients underwent assessment of myocardial viability.82 Of these, tractility) had to be present. Using two different imaging methods that
298 received medical therapy plus CABG and 303 received medical had different definitions of viability further disturbs the homogeneity
therapy alone—yet the decision to perform viability testing was not of the two groups of patients defined as having viable myocardium or
randomized and treatment was not assigned in a randomized fashion absence of viability. Thus one would expect to find different patients
based on viability status. Over a median follow-up of 5.1 years, 178 of in groups defined by these criteria than in groups defined by traditional
487 (37%) patients with viable myocardium and 58 of 140 patients viability criteria. Again, this may have influenced the surprising results.
without viable myocardium (41%) died. The hazard ratio for death However, when the data of the STICH viability substudy were separately
CHAPTER 21  Myocardial Viability 279

Apical Midventricular Basal

NH3

FDG

CMR

FIG. 21.21  Three short-axis views (apical, equatorial, and basal) of a positron emission tomography viability
study with assessment of rest perfusion (NH3; top) and glucose metabolism (FDG; middle). Bottom, Cardio-
vascular magnetic resonance (CMR) images in corresponding slices show enhancement. Note that in seg-
ments with reduced perfusion and metabolism, there is an increased CMR signal. Because of better spatial
resolution in CMR, distinction between transmural, subendocardial, and papillary defects can be made. The
border between enhanced and normal areas is distinct. (From Klein C, Nekolla SG, Bengel FM, et al. Assess-
ment of myocardial viability with contrast-enhanced magnetic resonance imaging: comparison with positron
emission tomography. Circulation. 2002;105[2]:162–167.)

analyzed for the dobutamine echocardiography and SPECT cohorts, It has been debated whether different imaging techniques with higher
the results remained the same.84 Fourth, almost 20% of patients had sensitivities and specificities such as PET or CMR could have altered
LVEF >35% when images were reanalyzed at the core laboratory. A the results.83,86 For practical reasons, these techniques could not be
higher LVEF would reduce the potential for improvement with revas- employed in such a large multicenter study, which was performed in
cularization. However, EFs in most previous studies were higher than 127 sites in 26 countries. Moreover, it is likely that the definitions
in STICH.70,85 Fifth, there was a large imbalance in clinical variables in employed and the advances of medical therapy since the time of the
each of the viability groups, which led to equalization of results after early viability studies influenced the unexpected results more than the
statistical correction for these imbalances. Sixth, it is unclear whether choice of the imaging techniques. This is reflected in the considerably
patients receiving revascularization had complete revascularization of lower mortality rates of patients with viable myocardium treated medi-
viable tissue. cally in the STICH trial than in a previous metaanalysis.70 Thus the
280 SECTION II  Ischemic Heart Disease

1.0
Hazard ratio, 0.64 (95% Cl, 0.48−0.86)
0.9 P = .003
0.8

Probability of death
0.7
0.6
0.5
0.4 Without viability
0.3
With viability
0.2
0.1
0.0
0 1 2 3 4 5 6
Years since randomization
No. at risk
Without viability 114 99 85 80 63 36 16
With viability 487 432 409 371 294 188 102
FIG. 21.22  Kaplan-Meier analysis of the probability of death, according
to myocardial viability status. The comparison that is shown has not been
adjusted for other prognostic baseline variables. After adjustment for
such variables on multivariable analysis, the between-group difference
was not significant (P = .21). CI, Confidence interval. (From Bonow RO,
Maurer G, Lee KL, et al. Myocardial viability and survival in ischemic
left ventricular dysfunction. N Engl J Med. 2011;364:1617–1625, with
permission of the publisher.)

Without myocardial viability With myocardial viability


1.0 1.0
0.9 0.9
0.8 0.8
Probability of death

Probability of death

0.7 0.7
Medical therapy (33 deaths)
0.6 0.6
0.5 0.5
0.4 0.4 Medical therapy (95 deaths)
0.3 0.3
0.2 0.2
CABG (25 deaths) CABG (83 deaths)
0.1 0.1
0.0 0.0
0 1 2 3 4 5 6 0 1 2 3 4 5 6
Years since randomization Years since randomization
No. at risk No. at risk
Medical therapy 60 51 44 39 29 14 4 Medical therapy 243 219 206 179 146 94 51
CABG 54 48 41 41 34 22 12 CABG 244 213 203 192 148 94 51

A B

P value for
Subgroup No. Deaths Hazard ratio (95% Cl) interaction
Without viability 114 58 0.70 (0.41−1.18) 0.53
With viability 487 178 0.86 (0.64−1.16)

0.25 0.50 1.0 2.0

CABG Medical
better therapy
C better
FIG. 21.23  Kaplan-Meier analysis of the probability of death according to myocardial viability status and
treatment. At 5 years in the intention-to-treat analysis, the rates of death for patients without myocardial
viability were 41.5% in the group assigned to undergo coronary artery bypass grafting (CABG) and 55.8%
in the group assigned to receive medical therapy (A). Among patients with myocardial viability, the respective
rates were 31.2% and 35.4% (B). There was no significant interaction between viability status and treatment
assignment with respect to mortality (P = .53) (C). CI, Confidence interval. (From Bonow RO, Maurer G, Lee
KL, et al. Myocardial viability and survival in ischemic left ventricular dysfunction. N Engl J Med. 2011;364:1617–
1625, with permission of the publisher.)
CHAPTER 21  Myocardial Viability 281

Stress
Rest
LGE

FIG. 21.24  Adenosine stress perfusion images (top) show the presence of a subendocardial inducible defect
in the basal and mid-cavity inferior wall (arrows), not present at rest (middle). Late gadolinium enhancement
(LGE) images demonstrate a viable inferior wall with absent myocardial enhancement (no infarction) (bottom).
(From Bucciarelli-Ducci C, Auger D, Di Mario C, et al. CMR guidance for recanalization of coronary chronic
total occlusion. JACC Cardiovasc Imaging. 2016;9:547–556.)

results of STICH suggest that viability testing is not a prerequisite for preserved viable epicardial rim. On the other hand, myocardial necrosis is
decisions regarding medical versus surgical management in patients characterized by LGE (possibly with a low-intensity core region because
with severely reduced EFs. Imaging should be reserved for those patients of no reflow) of the infarct area after injection of gadolinium, reduced
in whom management decisions are difficult in view of age, comorbidi- wall thickness (in chronic infarcts) without a substantial viable rim, and
ties, or complex coronary anatomy, and in whom the demonstration the absence of a contractile reserve during dobutamine stimulation.
of substantial viability might tilt the scale toward revascularization.84 Evaluation of viable myocardium is most important in patients with
In contrast with the difficult clinical scenario of identifying viable severe LV dysfunction because these patients can gain most from revas-
myocardium in patients with severely reduced EFs, identification of cularization if substantial amounts of myocardium are present. Recent
myocardial ischemia and viability is a “must” in patients with chroni- data from the STICH trial questioned the value of viability imaging
cally occluded coronary arteries.87–89 Recently, Bucciarelli-Ducci et al.90 for improving outcome in these patients. Unfortunately, the informa-
demonstrated in 50 consecutive patients with chronic total occlusions tion available about the value of CMR techniques in identifying patients
(CTOs) that most patients with inducible perfusion deficits and myo- with global LV dysfunction who have a high likelihood of profiting
cardial viability (infarct transmurality <50% by LGE CMR) within from a coronary revascularization is still scarce. Low-dose dobutamine
the CTO arterial territory before percutaneous coronary intervention CMR may be less accurate than LGE CMR in this patient group. In
had complete or near-complete resolution of CTO-related perfusion patients with regional LV dysfunction, however, in whom the need for
defects (Fig. 21.24). Moreover, these patients selected on the basis of revascularization needs to be established, both dobutamine CMR and
ischemia and viability in the CTO-related myocardium as demonstrated LGE CMR are well validated and can be used clinically. Depiction of
by CMR had favorable reverse remodeling and improvements in the zones of acute necrosis by observing enhancement after gadolinium
quality of life. carries substantial promise for detailed studies of the effects of different
treatment strategies for acute myocardial infarction, as well as for treat-
ment in the early and late postinfarction phase. In contrast, CMR may
CONCLUSION not be able to hold its initial promise for identifying the area at risk
CMR provides a variety of methods of obtaining information on residual and distinguishing it from the area of infarction.
viability after myocardial infarction. Indirect signs of viability that can
be observed by CMR are any sign of wall thickening at rest (which
is detectable with high accuracy by cine CMR), wall thickening after
REFERENCES
stimulation by low-dose dobutamine, preserved wall thickness, and a A full reference list is available online at ExpertConsult.com
CHAPTER 21  Myocardial Viability 281.e1

REFERENCES 23. Kim RJ, Fieno DS, Parrish TB, et al. Relationship of MRI delayed contrast
enhancement to irreversible injury, infarct age, and contractile function.
1. Rahimtoola SH. The hibernating myocardium. Am Heart J. Circulation. 1999;100:1992–2002.
1989;117:211–221. 24. Yabe T, Mitsunami K, Inubushi T, Kinoshita M. Quantitative measurements
2. Popio KA, Gorlin R, Bechtel D, Levine JA. Postextrasystolic potentiation of cardiac phosphorus metabolites in coronary artery disease by 31P
as a predictor of potential myocardial viability: preoperative analyses magnetic resonance spectroscopy. Circulation. 1995;92:15–23.
compared with studies after coronary bypass surgery. Am J Cardiol. 25. Bottomley PA, Weiss RG. Non-invasive magnetic-resonance detection of
1977;39:944–953. creatine depletion in non-viable infarcted myocardium. Lancet.
3. Soman P, Lahiri A, Mieres JH, et al. Etiology and pathophysiology of 1998;351:714–718.
new-onset heart failure: evaluation by myocardial perfusion imaging. J 26. Choi SI, Jiang CZ, Lim KH, et al. Application of breath-hold
Nucl Cardiol. 2009;16:82–91. T2-weighted, first-pass perfusion and gadolinium-enhanced T1-weighted
4. Bengel FM, Higuchi T, Javadi MS, Lautamaki R. Cardiac positron MR imaging for assessment of myocardial viability in a pig model. J
emission tomography. J Am Coll Cardiol. 2009;54:1–15. Magn Reson Imaging. 2000;11:476–480.
5. McLean DS, Anadiotis AV, Lerakis S. Role of echocardiography in the 27. Fisher MR, McNamara MT, Higgins CB. Acute myocardial infarction: MR
assessment of myocardial viability. Am J Med Sci. 2009;337:349–354. evaluation in 29 patients. AJR Am J Roentgenol. 1987;148:247–251.
6. Mahrholdt H, Wagner A, Judd RM, Sechtem U. Assessment of myocardial 28. Abdel-Aty H, Boye P, Zagrosek A, et al. Diagnostic performance of
viability by cardiovascular magnetic resonance imaging. Eur Heart J. cardiovascular magnetic resonance in patients with suspected acute
2002;23:602–619. myocarditis: comparison of different approaches. J Am Coll Cardiol.
7. Grover S, Srinivasan G, Selvanayagam JB. Evaluation of myocardial 2005;45:1815–1822.
viability with cardiac magnetic resonance imaging. Prog Cardiovasc Dis. 29. Simonetti OP, Kim RJ, Fieno DS, et al. An improved MR imaging
2011;54:204–214. technique for the visualization of myocardial infarction. Radiology.
8. Allman KC. Noninvasive assessment myocardial viability: current status 2001;218:215–223.
and future directions. J Nucl Cardiol. 2013;20:618–637, quiz 638–619. 30. Saeed M, Lund G, Wendland MF, Bremerich J, Weinmann H, Higgins CB.
9. Jimenez Juan L, Crean AM, Wintersperger BJ. Late gadolinium Magnetic resonance characterization of the peri-infarction zone of
enhancement imaging in assessment of myocardial viability: techniques reperfused myocardial infarction with necrosis-specific and extracellular
and clinical applications. Radiol Clin North Am. 2015;53:397–411. nonspecific contrast media. Circulation. 2001;103:871–876.
10. Braunwald E, Kloner RA. The stunned myocardium: prolonged, 31. Amado LC, Gerber BL, Gupta SN, et al. Accurate and objective infarct
postischemic ventricular dysfunction. Circulation. 1982;66:1146–1149. sizing by contrast-enhanced magnetic resonance imaging in a canine
11. Heusch G, Schulz R. Hibernating myocardium: a review. J Mol Cell myocardial infarction model. J Am Coll Cardiol. 2004;44:2383–2389.
Cardiol. 1996;28:2359–2372. 32. Fieno DS, Kim RJ, Chen EL, Lomasney JW, Klocke FJ, Judd RM.
12. Bodi V, Sanchis J, Lopez-Lereu MP, et al. Evolution of 5 cardiovascular Contrast-enhanced magnetic resonance imaging of myocardium at risk:
magnetic resonance-derived viability indexes after reperfused myocardial distinction between reversible and irreversible injury throughout infarct
infarction. Am Heart J. 2007;153:649–655. healing. J Am Coll Cardiol. 2000;36:1985–1991.
13. Kim RJ, Shah DJ. Fundamental concepts in myocardial viability 33. Mahrholdt H, Wagner A, Holly TA, et al. Reproducibility of chronic
assessment revisited: when knowing how much is “alive” is not enough. infarct size measurement by contrast-enhanced magnetic resonance
Heart. 2004;90:137–140. imaging. Circulation. 2002;106:2322–2327.
14. Shah DJ, Kim HW, James O, et al. Prevalence of regional myocardial 34. Oshinski JN, Yang Z, Jones JR, Mata JF, French BA. Imaging time after
thinning and relationship with myocardial scarring in patients with Gd-DTPA injection is critical in using delayed enhancement to determine
coronary artery disease. JAMA. 2013;309:909–918. infarct size accurately with magnetic resonance imaging. Circulation.
15. Cigarroa CG, deFilippi CR, Brickner ME, Alvarez LG, Wait MA, Grayburn 2001;104:2838–2842.
PA. Dobutamine stress echocardiography identifies hibernating 35. Ito H, Maruyama A, Iwakura K, et al. Clinical implications of the ‘no
myocardium and predicts recovery of left ventricular function after reflow’ phenomenon. A predictor of complications and left ventricular
coronary revascularization. Circulation. 1993;88:430–436. remodeling in reperfused anterior wall myocardial infarction. Circulation.
16. Walls MC, Verhaert D, Min JK, Raman SV. Myocardial edema imaging 1996;93:223–228.
in acute coronary syndromes. J Magn Reson Imaging. 2011;34:1243– 36. Rochitte CE, Lima JA, Bluemke DA, et al. Magnitude and time course of
1250. microvascular obstruction and tissue injury after acute myocardial
17. Abdel-Aty H, Zagrosek A, Schulz-Menger J, et al. Delayed enhancement infarction. Circulation. 1998;98:1006–1014.
and T2-weighted cardiovascular magnetic resonance imaging differentiate 37. Gerber BL, Rochitte CE, Melin JA, et al. Microvascular obstruction and
acute from chronic myocardial infarction. Circulation. left ventricular remodeling early after acute myocardial infarction.
2004;109:2411–2416. Circulation. 2000;101:2734–2741.
18. Ambrosio G, Weisman HF, Mannisi JA, Becker LC. Progressive 38. Amado LC, Kraitchman DL, Gerber BL, et al. Reduction of “no-reflow”
impairment of regional myocardial perfusion after initial restoration of phenomenon by intra-aortic balloon counterpulsation in a randomized
postischemic blood flow. Circulation. 1989;80:1846–1861. magnetic resonance imaging experimental study. J Am Coll Cardiol.
19. Kim RJ, Chen EL, Lima JA, Judd RM. Myocardial Gd-DTPA kinetics 2004;43:1291–1298.
determine MRI contrast enhancement and reflect the extent and severity 39. Wu KC, Zerhouni EA, Judd RM, et al. Prognostic significance of
of myocardial injury after acute reperfused infarction. Circulation. microvascular obstruction by magnetic resonance imaging in patients
1996;94:3318–3326. with acute myocardial infarction. Circulation. 1998;97:765–772.
20. Lima JA, Judd RM, Bazille A, Schulman SP, Atalar E, Zerhouni EA. 40. Rogers WJ Jr, Kramer CM, Geskin G, et al. Early contrast-enhanced MRI
Regional heterogeneity of human myocardial infarcts demonstrated by predicts late functional recovery after reperfused myocardial infarction.
contrast-enhanced MRI. Potential mechanisms. Circulation. Circulation. 1999;99:744–750.
1995;92:1117–1125. 41. Gerber BL, Garot J, Bluemke DA, Wu KC, Lima JA. Accuracy of
21. Pereira RS, Prato FS, Sykes J, Wisenberg G. Assessment of myocardial contrast-enhanced magnetic resonance imaging in predicting
viability using MRI during a constant infusion of Gd-DTPA: further improvement of regional myocardial function in patients after acute
studies at early and late periods of reperfusion. Magn Reson Med. myocardial infarction. Circulation. 2002;106:1083–1089.
1999;42:60–68. 42. Beek AM, Kuhl HP, Bondarenko O, et al. Delayed contrast-enhanced
22. Rehwald WG, Fieno DS, Chen EL, Kim RJ, Judd RM. Myocardial magnetic resonance imaging for the prediction of regional functional
magnetic resonance imaging contrast agent concentrations after improvement after acute myocardial infarction. J Am Coll Cardiol.
reversible and irreversible ischemic injury. Circulation. 2002;105:224–229. 2003;42:895–901.
281.e2 SECTION II  Ischemic Heart Disease

43. Hillenbrand HB, Kim RJ, Parker MA, Fieno DS, Judd RM. Early perfusion imaging for detection of subendocardial myocardial infarcts: an
assessment of myocardial salvage by contrast-enhanced magnetic imaging study. Lancet. 2003;361:374–379.
resonance imaging. Circulation. 2000;102:1678–1683. 63. McCrohon JA, Moon JC, Prasad SK, et al. Differentiation of heart failure
44. Choi KM, Kim RJ, Gubernikoff G, Vargas JD, Parker M, Judd RM. related to dilated cardiomyopathy and coronary artery disease using
Transmural extent of acute myocardial infarction predicts long-term gadolinium-enhanced cardiovascular magnetic resonance. Circulation.
improvement in contractile function. Circulation. 2001;104:1101–1107. 2003;108:54–59.
45. Friedrich MG, Abdel-Aty H, Taylor A, Schulz-Menger J, Messroghli D, 64. Mahrholdt H, Wagner A, Deluigi CC, et al. Presentation, patterns of
Dietz R. The salvaged area at risk in reperfused acute myocardial myocardial damage, and clinical course of viral myocarditis. Circulation.
infarction as visualized by cardiovascular magnetic resonance. J Am Coll 2006;114:1581–1590.
Cardiol. 2008;51:1581–1587. 65. Karamitsos TD, Francis JM, Myerson S, Selvanayagam JB, Neubauer S.
46. Eitel I, Desch S, Fuernau G, et al. Prognostic significance and The role of cardiovascular magnetic resonance imaging in heart failure. J
determinants of myocardial salvage assessed by cardiovascular magnetic Am Coll Cardiol. 2009;54:1407–1424.
resonance in acute reperfused myocardial infarction. J Am Coll Cardiol. 66. Kim RJ, Wu E, Rafael A, et al. The use of contrast-enhanced magnetic
2010;55:2470–2479. resonance imaging to identify reversible myocardial dysfunction. N Engl J
47. Whalen DA Jr, Hamilton DG, Ganote CE, Jennings RB. Effect of a Med. 2000;343:1445–1453.
transient period of ischemia on myocardial cells. I. Effects on cell volume 67. Selvanayagam JB, Kardos A, Francis JM, et al. Value of delayed-
regulation. Am J Pathol. 1974;74:381–397. enhancement cardiovascular magnetic resonance imaging in predicting
48. Jennings RB, Schaper J, Hill ML, Steenbergen C Jr, Reimer KA. Effect of myocardial viability after surgical revascularization. Circulation.
reperfusion late in the phase of reversible ischemic injury. Changes in cell 2004;110:1535–1541.
volume, electrolytes, metabolites, and ultrastructure. Circ Res. 1985;56: 68. Pegg TJ, Selvanayagam JB, Jennifer J, et al. Prediction of global left
262–278. ventricular functional recovery in patients with heart failure undergoing
49. Kim HW, Van Assche L, Jennings RB, et al. Relationship of T2-weighted surgical revascularisation, based on late gadolinium enhancement
MRI myocardial hyperintensity and the ischemic area-at-risk. Circ Res. cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2010;
2015;117:254–265. 12:56.
50. Giri S, Chung YC, Merchant A, et al. T2 quantification for improved 69. Gerber BL, Rousseau MF, Ahn SA, et al. Prognostic value of myocardial
detection of myocardial edema. J Cardiovasc Magn Reson. 2009;11:56. viability by delayed-enhanced magnetic resonance in patients with
51. Mallory GK, White PD, Salcedo-Galger J. The speed of healing of coronary artery disease and low ejection fraction: impact of
myocardial infarction: a study of the pathologic anatomy in 72 cases. Am revascularization therapy. J Am Coll Cardiol. 2012;59:825–835.
Heart J. 1939;18:647–671. 70. Allman KC, Shaw LJ, Hachamovitch R, Udelson JE. Myocardial viability
52. Haendchen RV, Corday E, Torres M, Maurer G, Fishbein MC, Meerbaum testing and impact of revascularization on prognosis in patients with
S. Increased regional end-diastolic wall thickness early after reperfusion: a coronary artery disease and left ventricular dysfunction: a meta-analysis. J
sign of irreversibly damaged myocardium. J Am Coll Cardiol. Am Coll Cardiol. 2002;39:1151–1158.
1984;3:1444–1453. 71. Romero J, Xue X, Gonzalez W, Garcia MJ. CMR imaging assessing
53. Pirolo JS, Hutchins GM, Moore GW. Infarct expansion: pathologic viability in patients with chronic ventricular dysfunction due to coronary
analysis of 204 patients with a single myocardial infarct. J Am Coll artery disease: a meta-analysis of prospective trials. JACC Cardiovasc
Cardiol. 1986;7:349–354. Imaging. 2012;5:494–508.
54. Ito H, Yu H, Tomooka T, et al. Incidence and time course of left 72. Knuesel PR, Nanz D, Wyss C, et al. Characterization of dysfunctional
ventricular dilation in the early convalescent stage of reperfused anterior myocardium by positron emission tomography and magnetic resonance:
wall acute myocardial infarction. Am J Cardiol. 1994;73:539–543. relation to functional outcome after revascularization. Circulation.
55. Bellenger NG, Swinburn JM, Rajappan K, Lahiri A, Senior R, Pennell DJ. 2003;108:1095–1100.
Cardiac remodelling in the era of aggressive medical therapy: does it still 73. Ichikawa Y, Sakuma H, Suzawa N, et al. Late gadolinium-enhanced
exist? Int J Cardiol. 2002;83:217–225. magnetic resonance imaging in acute and chronic myocardial infarction.
56. Baer FM, Smolarz K, Jungehulsing M, et al. Chronic myocardial Improved prediction of regional myocardial contraction in the chronic
infarction: assessment of morphology, function, and perfusion by state by measuring thickness of nonenhanced myocardium. J Am Coll
gradient echo magnetic resonance imaging and 99mTc-methoxyisobutyl- Cardiol. 2005;45:901–909.
isonitrile SPECT. Am Heart J. 1992;123:636–645. 74. Ansari M, Araoz PA, Gerard SK, et al. Comparison of late enhancement
57. Baer FM, Voth E, Schneider CA, Theissen P, Schicha H, Sechtem U. cardiovascular magnetic resonance and thallium SPECT in patients with
Comparison of low-dose dobutamine-gradient-echo magnetic resonance coronary disease and left ventricular dysfunction. J Cardiovasc Magn
imaging and positron emission tomography with [18F] Reson. 2004;6:549–556.
fluorodeoxyglucose in patients with chronic coronary artery disease. A 75. Klein C, Nekolla SG, Bengel FM, et al. Assessment of myocardial viability
functional and morphological approach to the detection of residual with contrast-enhanced magnetic resonance imaging: comparison with
myocardial viability. Circulation. 1995;91:1006–1015. positron emission tomography. Circulation. 2002;105:162–167.
58. Baer FM, Theissen P, Schneider CA, et al. Dobutamine magnetic 76. Wellnhofer E, Olariu A, Klein C, et al. Magnetic resonance low-dose
resonance imaging predicts contractile recovery of chronically dobutamine test is superior to SCAR quantification for the prediction of
dysfunctional myocardium after successful revascularization. J Am Coll functional recovery. Circulation. 2004;109:2172–2174.
Cardiol. 1998;31:1040–1048. 77. Gunning MG, Anagnostopoulos C, Knight CJ, et al. Comparison of
59. Trent RJ, Waiter GD, Hillis GS, McKiddie FI, Redpath TW, Walton S. 201Tl, 99mTc-tetrofosmin, and dobutamine magnetic resonance imaging
Dobutamine magnetic resonance imaging as a predictor of myocardial for identifying hibernating myocardium. Circulation. 1998;98:1869–1874.
functional recovery after revascularisation. Heart. 2000;83:40–46. 78. Bonow RO. Identification of viable myocardium. Circulation. 1996;94:
60. Ramani K, Judd RM, Holly TA, et al. Contrast magnetic resonance 2674–2680.
imaging in the assessment of myocardial viability in patients with stable 79. Schvartzman PR, Srichai MB, Grimm RA, et al. Nonstress delayed-
coronary artery disease and left ventricular dysfunction. Circulation. enhancement magnetic resonance imaging of the myocardium predicts
1998;98:2687–2694. improvement of function after revascularization for chronic ischemic
61. Wu E, Judd RM, Vargas JD, Klocke FJ, Bonow RO, Kim RJ. Visualisation heart disease with left ventricular dysfunction. Am Heart J.
of presence, location, and transmural extent of healed Q-wave and 2003;146:535–541.
non-Q-wave myocardial infarction. Lancet. 2001;357:21–28. 80. Kim RJ, Manning WJ. Viability assessment by delayed enhancement
62. Wagner A, Mahrholdt H, Holly TA, et al. Contrast-enhanced MRI and cardiovascular magnetic resonance: will low-dose dobutamine dull the
routine single photon emission computed tomography (SPECT) shine? Circulation. 2004;109:2476–2479.
CHAPTER 21  Myocardial Viability 281.e3

81. Kaandorp TA, Bax JJ, Schuijf JD, et al. Head-to-head comparison between 87. Strauss BH, Shuvy M, Wijeysundera HC. Revascularization of chronic
contrast-enhanced magnetic resonance imaging and dobutamine total occlusions: time to reconsider? J Am Coll Cardiol.
magnetic resonance imaging in men with ischemic cardiomyopathy. Am J 2014;64:1281–1289.
Cardiol. 2004;93:1461–1464. 88. Baks T, van Geuns RJ, Duncker DJ, et al. Prediction of left ventricular
82. Bonow RO, Maurer G, Lee KL, et al. Myocardial viability and survival in function after drug-eluting stent implantation for chronic total coronary
ischemic left ventricular dysfunction. N Engl J Med. 2011;364:1617–1625. occlusions. J Am Coll Cardiol. 2006;47:721–725.
83. Srichai MB, Jaber WA. Viability by MRI or PET would have changed the 89. Cheng AS, Selvanayagam JB, Jerosch-Herold M, et al. Percutaneous
results of the STICH trial. Prog Cardiovasc Dis. 2013;55:487–493. treatment of chronic total coronary occlusions improves regional
84. Bonow RO, Holly TA. Myocardial viability testing: still viable after stich? J hyperemic myocardial blood flow and contractility: insights from
Nucl Cardiol. 2011;18:991–994. quantitative cardiovascular magnetic resonance imaging. JACC Cardiovasc
85. Schinkel AF, Bax JJ, Poldermans D, Elhendy A, Ferrari R, Rahimtoola SH. Interv. 2008;1:44–53.
Hibernating myocardium: diagnosis and patient outcomes. Curr Probl 90. Bucciarelli-Ducci C, Auger D, Di Mario C, et al. CMR Guidance for
Cardiol. 2007;32:375–410. Recanalization of Coronary Chronic Total Occlusion. JACC Cardiovasc
86. Asrani NS, Chareonthaitawee P, Pellikka PA. Viability by MRI or PET Imaging. 2016;9:547–556.
would not have changed the results of the STICH trial. Prog Cardiovasc
Dis. 2013;55:494–497.
22 
Cardiovascular Magnetic Resonance Tagging
for Assessment of Left Ventricular
Diastolic Function
El-Sayed H. Ibrahim and Matthias Stuber

Left ventricular (LV) diastolic function has been recognized as an from end-systolic volume or associated strains.6 In normal subjects,
important factor in the pathophysiology of many common cardio- the circumferential-longitudinal shear strain decreases before the axial
vascular diseases. Dilated and hypertrophic cardiomyopathy (HCM), strain.7 Furthermore, the early-diastolic filling efficiency can be aug-
coronary artery disease (CAD), and systemic hypertension are all associ- mented during exercise stress in an effort to maintain stroke volume
ated with abnormal LV filling dynamics. Diastolic dysfunction has also despite shortened diastole.8 Studying early-diastolic strain patterns
been increasingly appreciated as a major cause of heart failure (HF), through myocardial strain analysis allows for comparing normal sub-
especially in the elderly. Although invasive hemodynamic measures/ jects with those with diastolic dysfunction. Therefore clinicians would
assessment of diastole are considered the gold standard, echocardio- be able to identify patients with regional diastolic dysfunction that
graphic methods, including tissue Doppler imaging, have gained greater places them at the risk of HF and sudden cardiac death following
use in the clinical assessment of LV diastolic function because of their pathologic conditions such as myocardial infarction (MI), and aortic
noninvasive acquisition, which greatly facilitates serial assessments. In stenosis (AS).
cardiovascular magnetic resonance (CMR) imaging, with the advent
of parallel imaging, three-dimensional (3D) data collection, spiral and Assessment of Cardiac Rotation/Motion:
balanced steady-state free precession (bSSFP) imaging, and higher mag- Noncardiovascular Magnetic Resonance Methods
netic field strength, the prolonged tag persistence permits easier access With echocardiographic imaging, the myocardium has relatively poor
to diastole. Together with rapid state-of-the-art software analysis tools, internal structure because of the absence of structural landmarks, making
quantification of LV diastolic wall motion can now easily be performed limited quantification of parameters, such as rotation, stress, and strain.9,10
using CMR. Several invasive methods have been reported during diastole to examine
cardiac motion. One approach is the surgical/invasive implantation of
CARDIAC MOTION tantalum markers into the midwall of the myocardium.11 In combina-
tion with x-ray angiography, the motion of these markers can then be
Systolic and Diastolic Heart Motion recorded with high temporal and spatial resolution. Using such an
During systole, the heart performs a complex motion analogous to approach, alterations in diastolic untwisting have been observed in
“wringing” a towel, and as a result, the base and apex rotate in opposite patients who have undergone heart transplantation shortly before rejec-
directions. There is counterclockwise rotation at the apex and clockwise tion.12 Although this method is very powerful, it is invasive, requires
rotation at the base.1 In parallel, the atrioventricular valvular plane ionizing radiation, and is inappropriate for routine and repeated clinical
(basal left ventricle and right ventricle) descends toward the apex. The use. Alternative angiographic “markers,” such as tracking of the bifurca-
lateral free wall of the right ventricle performs a more pronounced tions of the coronary arteries,13 suffer from the limited number of
long-axis contraction than the lateral wall of the left ventricle.2 During landmarks and their irregular geometric distribution. Furthermore, they
isovolumic relaxation, the myofibrils return to their resting state from only provide motion information about the epicardial layers of the
the contracted state. This process is accompanied by a rapid untwisting myocardium.
at the apex, whereas the volume and cavity shape of the heart remain
nearly unchanged. This rapid untwisting typically lasts less than 75 ms Assessment of Cardiac Rotation/Motion: Cardiovascular
and precedes the early passive filling phase of the ventricles. During Magnetic Resonance Methods
this filling phase, practically no rotational components can be seen at Similar to echocardiography, conventional cine CMR images do not
the apex of the healthy heart.3 provide information about the internal structure of the myocardium.
Understanding different diastolic strain components and their However, a CMR myocardial tagging technique, spatial modulation of
intrinsic mechanisms during different pathologic conditions allows magnetization (SPAMM), originally proposed by Axel15 and further
for better understanding of diastolic dysfunction.4 Isovolumic relax- developed and refined by others, offers the opportunity to assess strain
ation is associated with changes in principal strains and untwisting, noninvasively. With these methods, the magnetization of the muscle
which are all related to an apparent increase in the LV volume, likely tissue is spatially modulated, or “tagged,” by the application of a specific
reflecting expansion of the LV myocardium.5 The diastolic shear strain time series of radiofrequency (RF) pulses and magnetic field gradients.
rates are linearly related to the corresponding preceding systolic shear The tagging is typically applied immediately after the R-wave of the
strain components. Nevertheless, the torsional recoil is uncoupled electrocardiogram (ECG). Images are then acquired during successive

282
CHAPTER 22  CMR Tagging for Assessment of Left Ventricular Diastolic Function 283

heart phases in which the tags may be identified as dark lines or grids. percentage of the available longitudinal magnetization is used at later
Because these tags are spatially fixed with respect to the muscle tissue heart phases.
at the time of the tag’s application, local myocardial motion can be
derived from the translation, rotation, and distortion of the tags on the Complementary Spatial Modulation of Magnetization
myocardium. However, because of the relaxation effects, the tags fade With Slice-Following
rapidly and cannot be reliably detected after end systole (approximately Because of through-plane cardiac motion, two-dimensional (2D) cine
300 ms after tag application). This is a serious drawback for the quan- imaging of the heart may not show the same myocardial tissue throughout
tification of systolic and diastolic dynamics of the heart wall. Another the cardiac cycle; rather, the imaging plane shows whatever tissue lies
limitation is that this approach does not compensate for through-plane inside it at the time of data acquisition. This could lead to inaccurate
motion. These limitations have been resolved through the development assessment of myocardial motion; for example, an apparent myocardial
of the complementary SPAMM (CSPAMM) technique,3,16 on which we thickening in a basal short-axis slice could be in fact caused by myocar-
will focus in the remainder of the chapter. dial basal displacement toward the apex. Slice-following CSPAMM3 was
developed as an improvement of the original CSPAMM technique to
COMPLEMENTARY SPATIAL MODULATION OF resolve the through-plane motion problem. The technique is based on
MAGNETIZATION: TECHNICAL DEVELOPMENTS implementing slice-selective tagging instead of the nonselective tagging
used in conventional CSPAMM. A thin slice of interest is tagged by
Complementary Spatial Modulation of switching one (or both) of the tagging RF pulses with a slice-selective
Magnetization Tagging17 pulse, which has the effect of confining the tagging pattern inside the
One limitation of SPAMM tagging is the fading of the tagging pattern slice of interest (whose slice thickness = Δz, as shown in Fig. 22.2). Later,
through the cardiac cycle as a result of longitudinal magnetization during the data acquisition part of the sequence, a thicker slice (whose
relaxation. The loss of tagging contrast toward the end of the cardiac slice thickness of Δs encompasses the thin tagged slice) is excited. The
cycle results in unrecognizable tagging pattern, which precludes the excited slice should be thick enough to accommodate the thin tagged
analysis of diastolic heart phases. It was not until 1993 when Fischer slice despite its displacement in the through-plane (z) direction. Because
et al.16 introduced the CSPAMM sequence to resolve this problem (Fig. nontagged magnetization is eliminated in CSPAMM, the only source of
22.1). To grasp the idea behind CSPAMM, it is necessary to understand signal in the resulting image comes from the initially tagged slice, regard-
the magnetization evolution with time in a tagging sequence. Imme- less of its displacement in the through-plane direction. This ensures that
diately after applying the tagging module of the pule sequence, the the same myocardial tissue is imaged during the whole cardiac cycle,
whole magnetization is tagged or modulated (90-degree RF pulses are and that apparent motion illusions are eliminated. The choice of the
assumed here) and stored in the longitudinal position. With time, the imaging slice thickness has to be carefully considered to ensure inclusion
magnetization experiences longitudinal relaxation, trying to reach the of the tagged slice throughout the cardiac cycle, and at the same time
equilibrium state. This has two effects on the stored tagging pattern: to avoid unnecessary thickness that would only add noise to the image.
(1) introducing a growing nontagged magnetization offset (we call it Typically, double oblique short-axis sections of the myocardium
here the DC component, borrowing the term direct current [DC] from are tagged with a slice thickness of 6 to 8 mm. Subsequently, 16 to 20
electrical engineering); and (2) reducing the magnitude of the tagged sequential heart phase images are acquired with a temporal resolu-
component (the peak-to-peak difference of the sinusoidal tagged mag- tion (Δt) of about 35 ms. With this high temporal resolution, rapid
netization). Thus, during the imaging stage, the excited magnetization motion components, such as diastolic untwisting, can be identified
has two components: tagged and DC, with the DC overhead impairing readily. Because the ratio of wanted to unwanted signal components
the visibility of the (already fading) tagged component. It should be must be optimized, the thickness of the imaged volume (Δs; see Fig.
noted that the multiple applications of RF pulses during the imaging 22.2) must be reduced to a minimum. Therefore it depends on the
(data acquisition) part of the sequence also contributes to reducing level of the tagged slice with respect to its level on the long axis. For
the tagged magnetization component (each RF pulse consumes part basal LV images, where a long-axis contraction of more than 20 mm
of the tagged magnetization stored in the longitudinal direction). The may be expected for the lateral right ventricular (RV) free wall,2 a slice
solution provided by CSPAMM consisted of two parts: eliminating the thickness of 30 mm is typically chosen. For midventricle slices, a slice
nontagged (DC) magnetization and enhancing the fading tagged mag- thickness of 25 mm is appropriate, and at the apex, a slice thickness
netization. To eliminate the nontagged magnetization, two consecutive of 20 mm is used because of reduced through-plane motion. For sup-
SPAMM scans are acquired with exactly the same parameters, except pression of breathing-induced motion artifacts, a repetitive breath-
for the polarity of one of the tagging RF pulses. The 90-degree/90- holding scheme3,16 or single breath-hold techniques18 can be applied.
degree tagging RF pulses in the first scan modulate the magnetization Furthermore, for high-resolution CSPAMM tagging, the two SPAMM
with a positive sinusoidal pattern, whereas the 90-degree/−90-degree acquisitions could be acquired with a 90-degree phase shift between
RF pulses in the second scan result in a negative sinusoidal pattern. them before constructing the CSPAMM images.19
Note that the DC magnetization component is the same in both scans Considering the location of the relevant tagging information in
at corresponding time points. Therefore, the overhead DC magnetiza- k-space, a reduced k-space acquisition scheme can be applied.3,16 Hereby,
tion can be simply eliminated by subtracting the images in the first two sets of orthogonally line tagged images are acquired. Subsequent
scan from the corresponding images (at the same heart phases) in combination of these acquisitions results in grid-tagged images (Figs.
the second scan. This subtraction has also the effect of improving the 22.3 and 22.4). With this method, acquisition time is significantly reduced
image’s signal-to-noise ratio (SNR) by 40% as two acquisitions with and image resolution perpendicular to the line tags is not affected.
independent noise terms are added together. To resolve the second Fig. 22.3 shows 20 heart phase images with a temporal resolution of
problem of fading tagging contrast, the concept of “ramped flip angle” 37 ms acquired at an apical slice of a healthy subject.18 The grid struc-
was introduced. Basically, during the imaging stage, the flip angles of the ture remains visible, with a high contrast up to the last acquisition in
RF pulses determine how much magnetization is tipped into the trans- late diastole (>700 ms). No fading of the tags is seen in the images.
verse plane for data acquisition. Thus increasing the flip angles through Therefore the method is well suited for the quantification of diastolic
the cardiac cycle compensates for the fading tagging contrast as a larger heart wall motion.
284 SECTION II  Ischemic Heart Disease

Tagging Imaging

90° ± 90°
QRS
Ramped flip angles
RF
Crusher
Modulation
X Grad. RO
RV

Y Grad. PE LV

Z Grad. SS

A Time B

ML 1st scan ML 2nd scan

Mo Mo
AC

x x

ML ML
Time Time
Mo Mo
AC
DC

+ x x

ML CSPAMM ML x (−1)
Mo

+
AC

x x

−Mo
C
FIG. 22.1  (A) Complementary spatial modulation of magnetization (CSPAMM) pulse sequence. The sequence
runs two SPAMM sequences, with the polarity of the second tagging radiofrequency (RF) pulse changed in
the second SPAMM acquisition. Notice also the ramped flip angles of the imaging RF pulses to compensate
for fading tagging. (B) Example of a grid-tagged CSPAMM image. Notice that nontagged tissues appear black
because of the elimination of the offset DC signal. (C) The concept of magnetization subtraction in CSPAMM.
Two scans are acquired as shown in the pulse sequence, which results in positive and negative sinusoidal
tagging patterns from the first and second scans, respectively. With time, the tagging patterns experience
longitudinal relaxation, trying to reach equilibrium magnetization (Mo). Magnetization relaxation has two effects
on the tagging pattern: the peak-to-peak (AC) magnitude is decreased and the tagging pattern has nonzero
average (DC) value. However, the DC component is the same in both scans. Thus, at any time point, when
two corresponding images (obtained from the first and second acquisitions at the same time point) are
subtracted, the DC component cancels out and the peak-to-peak tagging magnitude doubles. Grad, Gradient;
LV, left ventricular; RV, right ventricular. (From Ibrahim el-SH. Myocardial tagging by cardiovascular magnetic
resonance: evolution of techniques—pulse sequences, analysis algorithms, and applications. J Cardiovasc
Magn Reson. 2011;13:36.)

Data Acquisition Trajectory in Complementary Spatial T1 can only be increased by going to a higher magnetic field strength,
Modulation of Magnetization and the number of RF excitations can be reduced by using alternative
Although traditionally segmented k-space gradient recalled echo (GRE) imaging sequences with fewer RF excitations. Hereby, echo planar imaging
techniques are used for signal readout, both T1 relaxation and serial (EPI)18,20 (see Fig. 22.3) and, more recently, spiral imaging21 proved to
RF excitations for imaging are responsible for the fading of the tags. be very valuable alternatives (see Fig. 22.4).
CHAPTER 22  CMR Tagging for Assessment of Left Ventricular Diastolic Function 285

Spiral imaging provides a time-efficient k-space coverage, which imaging at a very high temporal resolution. Finally, the short echo time
makes it a very powerful alternative to EPI for signal readout of the (TE) of spiral acquisition allows for minimizing flow and motion arti-
tagged myocardium. Because a small number of excitation RF pulses facts.23 Ryf et al.21 combined CSPAMM with interleaved spiral imaging,
is needed in spiral imaging, the tagging pattern persists longer, leading which allowed for acquiring high spatial resolution (4-mm tag separa-
to improved tagging contrast.22 Furthermore, spiral imaging allows for tion) or high temporal resolution (77 frames per second) grid-tagged
efficient coverage of the k-space in a short amount of time, leading to images in a single breath-hold (see Fig. 22.4).
Together with EPI and spiral readouts, the use of bSSFP was exploited
by Herzka et al.,24 who found that bSSFP leads to an improved tagging
contrast compared with the more conventional GRE imaging. In another
study, Zwanenburg et al.25 combined CSPAMM with bSSFP imaging
for a single breath-hold scan. In this approach, the steady-state mag-
Δz
netization is stored in the longitudinal direction using an α/2 flip-back
RF pulse immediately before tagging preparation. Imaging proceeds
normally, although excitation is achieved using a series of the linearly
Δs increasing start-up angles (LISA) technique to minimize off-resonance
artifacts. Ibrahim et al.26 developed a technique for improving the
CSPAMM tagging contrast in bSSFP cine images by optimizing the RF
excitation angles to compensate for tags fading during the cardiac cycle,
FIG. 22.2  Slice-following principle. An initially tagged planar slice of the similar to Fischer’s approach for the gradient echo CSPAMM-tagged
thickness dz translates and distorts during the cardiac cycle. A volume cine images.16
of the thickness ds is imaged multiple times during the cardiac cycle. With the advent of parallel and 3D imaging, 3D assessment of myo-
This volume must encompass the potential extent of the motion of the cardial motion based on 3D CSPAMM lattice tagging was reported by
tagged, thin slice. Ryf et al.27 Fig. 22.5 shows an isosurface-rendered image based on 3D

t = 35 ms t = 72 ms t = 109 ms t = 146 ms t = 183 ms t = 220 ms

t = 257 ms t = 294 ms t = 331 ms t = 368 ms t = 405 ms t = 442 ms

t = 479 ms t = 516 ms t = 553 ms t = 590 ms t = 627 ms t = 664 ms

t = 701 ms t = 738 ms
FIG. 22.3  Twenty apical left ventricular short-axis images in a healthy adult subject acquired with comple-
mentary spatial modulation of magnetization tagging. The systolic images include frames 1 to 9 (331 ms
after the R-wave of the electrocardiogram), and the diastolic images are shown in frames 10 to 20 (368 to
738 ms). The temporal resolution in these images is 37 ms, and two line-tagged acquisitions were combined
to create a grid-tagged image off-line. During systole, a counterclockwise apical rotation is seen, followed
by a rapid clockwise untwisting in frames 10 to 15 (368 to 553 ms) during early diastole. This precedes the
filling phase (frames 16 to 20; 590 to 738 ms) of the ventricles.
286 SECTION II  Ischemic Heart Disease

17 ms 332 ms 542 ms

17 ms 332 ms 542 ms

24 ms 339 ms 549 ms

FIG. 22.4  Spiral complementary spatial modulation of magnetization tagging using a spectral-spatial excita-
tion for fat suppression in each cine frame. Midventricular short-axis and long-axis views of a line-tagged
and a grid-tagged myocardium in a healthy subject. The tagline distance is 4 mm, and the temporal resolution
is 35 ms. The time after the R-wave is indicated on the images. On the grid-tagged, short-axis view, 50
tagline intersections can be observed.

CSPAMM imaging. Simultaneously, the same authors proposed an persistence.30 By reconstructing only magnitude images, MICSR yields
extension to a previously reported analysis procedure28 that enables tags with zero mean sinusoidal profiles, which obviates the need for
relatively time-efficient analysis and quantification of both systolic and acquiring phase calibration data or applying phase correction algorithms.
diastolic motion of the heart.29 However, the acquisition times were Based on the MICSR technique, the same authors developed a method
lengthy and only practical in coached breathing patterns in well-trained for trinary display of the resulting images, which emphasized the tagging
subjects. persistence and presented a novel way for visualizing myocardial defor-
mation.30,31 MICSR was shown to have the following advantages: (1) a
Complementary Spatial Modulation of Magnetization true sinusoidal tagging profile is created as opposed to rectified sinusoids;
Sequence Development Approaches (2) a peak MICSR contrast is obtained between 200 and 500 ms after
Magnitude Reconstruction the R-wave, corresponding to the period in the cardiac cycle with the
The magnitude image CSPAMM reconstruction (MICSR) technique largest myocardial deformation (late systole to early diastole); (3) the
has been developed for improving CSPAMM tagging contrast and tagging contrast is higher and persists for a longer period of time than
CHAPTER 22  CMR Tagging for Assessment of Left Ventricular Diastolic Function 287

FIG. 22.5  Isosurface rendering of three-dimensional complementary FIG. 22.6  End-diastolic apical complementary spatial modulation of
spatial modulation of magnetization. A volume of 128 × 128 mm2 is magnetization acquired in a healthy adult subject. The grid-tagged image
displayed. Tag spacing is 10 mm. (From Ryf S, Spiegel MA, Gerber M, is overlain with the corresponding local trajectories. The arrows start at
Boesiger P. Myocardial tagging with 3D-CSPAMM. J Magn Reson Imaging. the beginning of systole and at end diastole.
2002;16:320–325, with permission.)

Orthogonal Complementary Spatial Modulation


in conventional CSPAMM; and (4) the images are optimized for proc- of Magnetization
essing with harmonic phase magnetic resonance (HARP) analysis.28 Orthogonal CSPAMM has been recently introduced to reduce the scan
time in half while maintaining the CSPAMM advantage of removing
Real-Part Reconstruction the central nontagged (DC) signal peak in k-space.36 Because CSPAMM
Kuijer et al.32 presented a technique for improving the tagging contrast is based on acquiring two separate SPAMM images (which are 180
without increasing the scan time through real-part data reconstruction degrees out of phase with each other) for producing a line-tagged image,
using an internal phase reference from the tagged images. The tagging then four separate SPAMM acquisitions would be required for produc-
efficiency of the proposed technique was similar to that of real-part ing a 2D grid-tagged CSPAMM image. The orthogonal CSPAMM
reconstruction with a separate reference scan. However, compared with sequence is based on rotating the tag orientation in the second SPAMM
magnitude image subtraction, the tagging contrast-to-noise ratio was acquisition by 90 degrees relative to the first one, such that when the
improved by 70% without increasing the scan time. two images are subtracted to produce the final CSPAMM image, the
DC signal peak is canceled (as in conventional CSPAMM), while a 2D
Fat Suppression in Complementary Spatial Modulation grid-tagging is obtained, thus producing a CSPAMM grid-tagged image
of Magnetization from only two SPAMM acquisitions.
An efficient fat suppression method (inherent fat cancelation) has been
developed for CSPAMM imaging.33 The inherent fat cancelation method Tagging Analysis
makes use of the two acquisitions required for CSPAMM imaging by For the extraction of motion data from the tagged time series of images,
allowing for complementary and in-phase modulations of the water sophisticated image analysis tools are needed. Although the identifica-
and fat spins, respectively; therefore, in the final CSPAMM image, the tion of tags was very labor intensive and took hours in the early years,
tagging contrast of water is doubled, while the tagging signal from fat sophisticated algorithms that reduce quantification of tagged time series
is canceled. Compared with the spectral-spatial selective fat suppres- of cardiac images to seconds are now readily available,28 and variants
sion,34 the inherent fat cancelation method provides fat-suppressed have been successfully implemented.29 Analysis of tagged images involves
CSPAMM images with high temporal resolution and short echo time the identification of the tags in each heart phase image. Using automated
without increasing the scan time. or semiautomated algorithms, the tags may be identified with an accu-
racy that exceeds the image resolution.37 If the grid intersection points
Off-Resonance Insensitive–Complementary Spatial are identified for all heart phase images, local trajectories on the myo-
Modulation of Magnetization cardium (Fig. 22.6) are defined, and motion-specific parameters (e.g.,
Recently, CSPAMM has been combined with Fourier analysis of stimu- strain, rotation, rotation velocity, radial or circumferential shortening,
lated echoes (FAST) for quantification of LV systolic and diastolic func- or shear between epicardial and endocardial muscle layers of the myo-
tion.35 The developed technique provides a semiautomatic means for cardium) can be derived. Using this approach, studies of healthy subjects
quick and quantitative assessment of LV systolic and diastolic twist and and patients with MI,38 HCM,39 and AS with pathologically hypertro-
torsion. The developed technique requires a short scan time and provides phied hearts, as well as athletes with physiologic hypertrophy, were
quantitative assessment of systolic and diastolic LV twist, torsion, twist- evaluated for apical untwisting during diastole.40 Untwisting velocity,
ing rates, time-to-peak twist, duration of untwisting, and ratio of rapid time-to-peak untwisting velocity, and early diastolic strain can be deter-
untwist to peak twist. mined as indices of diastolic function. Time-to-peak untwisting velocity
288 SECTION II  Ischemic Heart Disease

(untwisting time; Table 22.1) is defined as the time delay between the heart function. Other CSPAMM applications include CAD, ischemic
point in time of minimum inner cavity area and the maximum untwist- heart disease, MI, HCM, interventricular synchrony, valvular diseases,
ing velocity (Fig. 22.7). postsurgical cardiac function evaluation, and congenital heart disease.
In this section, we focus on applications related to studying diastolic
CLINICAL APPLICATIONS cardiac function.
Beyond the technical developments addressed earlier in this chapter,
CSPAMM has been implemented in a large number of clinical appli- Ventricular Rotation
cations. CSPAMM reproducibility has been studied in healthy sub- As mentioned previously, at the apex of the healthy heart, a counter-
jects, where the results showed good intraobserver, interobserver, and clockwise rotation during systole can be seen (see Fig. 22.3, phases 1
interstudy reproducibility for measuring LV circumferential strain and to 9; see Fig. 22.7, phase 1). This systolic rotation is followed by a rapid
twist, and less reproducibility for measuring radial strain.41 Studies untwisting during isovolumic relaxation (see Fig. 22.3, phases 10 to 15
for measuring regional LV function are promising, as are those for and early diastole; see Fig. 22.7, phase 2). This untwisting phase is
examining the pattern of RV contractility. Heart rotation and torsion typically followed by the filling phase of the ventricles, during which
are equally interesting areas of study, as well as the effect of aging on little rotational component is seen (see Fig. 22.7, phase 3).3,42 An identi-
cal separation of early diastolic apical untwisting and the filling phase
of the ventricles is seen in highly competitive athletes with physiologi-
cally hypertrophied hearts. Neither the apical peak rotation angle nor
TABLE 22.1  Peak Rotation at the Apex, diastolic untwisting time is changed in the hypertrophied hearts of
Maximum Rotation Velocity During Diastolic athletes compared with healthy control subjects (see Table 22.1). However,
Untwisting and Untwisting Time for 12 among patients with LV hypertrophy, as a result of pressure overload/
Aortic Stenosis Patients, 12 Athletes, and AS, a completely different apical rotation pattern is seen (see Fig. 22.7).
11 Healthy Volunteersa The end-systolic peak rotation is significantly increased (P < .01) in
comparison with the athletes or healthy subjects, and no separation of
Untwisting untwisting and filling can be seen during diastole. Untwisting and filling
Peak Rotation Velocity Untwisting occur simultaneously, and the point in time of maximum untwisting
Patients (Degrees) (Degrees/s) Time (% ES) velocity is significantly delayed in these patients (P < .01; see Fig. 22.7).
Aortic stenosis 12 ± 5 80 ± 29 32 ± 6 Pressure overload LV hypertrophy results in the addition of new
Athletes 6±2 56 ± 8 17 ± 8 sarcomeres in parallel.43 Furthermore, an increase in the amount of col-
Volunteers 7±2 55 ± 17 16 ± 8 lagen, with a consequently increased elastic stiffness, has been reported.44
a
This rearranged fiber architecture, together with the increased stiffness
The untwisting time is related to the duration of systole. Data are
of the muscle tissue, may explain the alterations of the diastolic rota-
expressed as mean ± 1 standard deviation.
ES, End systole.
tion pattern with a prolonged and delayed apical untwisting in these
Modified from Stuber M, Scheidegger MB, Fischer SE, et al. patients. The prolongation of untwisting results in an overlap with
Alterations in the local myocardial motion pattern in patients suffering early diastolic filling and presumably impedes normal filling. Thus, a
from pressure overload due to aortic stenosis. Circulation. prolongation of early diastolic untwisting may be responsible for the
1999;100:361–368. occurrence of diastolic dysfunction in these patients. In patients with

14
Apical rotation angle (degrees)

12
10 100
1
Rotation (x100% ES)

8 80
6
60
4 2
2 40
0
20
–2
Systole Diastole 3
–4 0
0 20 40 60 80 100 120 140 160 180 200 0 10 20 30 40 50 60 70 80 90 100
A Time (% ES) B Apical area reduction (x100% ES)

Aortic stenosis Healthy Rowers


FIG. 22.7  (A) Cross-sectional apical rotation velocity of athletes, healthy adults, and patients with aortic
stenosis (AS). The values are mean ± 1 standard deviation. The time axis is normalized to the end-systolic
point of the cardiac cycle. The time point of maximum diastolic untwisting velocity (arrows) is delayed in the
patients with AS compared with the athletes or the healthy control subjects. Apical rotation velocity is identi-
cal in athletes and control subjects. (B) Left ventricular (LV) rotation-area loop (apical plane) in healthy control
subjects, rowers, and patients with AS. The loop is separated in isovolumic contraction (1), ejection (2),
isovolumic relaxation (3), and LV filling. Both rotation and area change of the inner lumen at the apex are
related to their maximum values (100%). ES, End systole.
CHAPTER 22  CMR Tagging for Assessment of Left Ventricular Diastolic Function 289

MI, a prolonged untwisting phase with an overlap of diastolic untwist- or athletes who have a similar degree of hypertrophy as the patients.
ing and filling has also been observed45 and end-systolic apical peak In another study by Nagel et al.,50 the authors used CSPAMM to study
rotation is usually severely reduced. cardiac torsion as well as systolic and diastolic wall motion in 12 healthy
In 2011, Russel et al.46 used CSPAMM to study ventricular torsion controls and 13 patients with severe AS. The results showed that during
in patients with recessive mutation causing familial HCM. The results systole, the normal LV undergoes a clockwise rotation (when viewed
showed that the mutation carriers have normal wall thickness, but from the apex) of −4.4 ± 1.6 degrees at the base and a counterclockwise
increased LV torsion with respect to controls, which suggests that healthy rotation of +6.8 ± 2.5 degrees at the apex. In patients with AS, however,
carriers may be targets for clinical intervention at a preclinical stage to this rotation was found to be reduced at the base and increased at the
prevent the onset of future dysfunction. apex. Furthermore, the overall rotation velocity is decreased and maximal
torsion is increased in AS. Recently, CSPAMM has been implemented
Ventricular Hypertrophy for studying changes in myocardial strain following transcatheter aortic
CMR assessment of diastolic function is beginning to be studied in valve implantation (TAVI).51 TAVI resulted in improved mid-LV cir-
broader populations. HCM has been studied using CSPAMM tagging.39 cumferential strain and decreased myocardial twist. However, although
CSPAMM was combined with the cardiac phase to order reconstruction systolic strain rate increased following TAVI, there was no significant
(CAPTOR) technique47 for quantifying regional indices of myocardial change in diastolic strain rate.
function throughout the cardiac cycle. The results showed distinct dif-
ferences in strain in HCM compared with controls (Fig. 22.8). Specifi- Diastolic Heart Failure
cally, although early systolic shortening was similar, the total systolic CMR tagging has been used to illustrate changes in regional myocardial
shortening was significantly reduced in the septal and inferior regions. function in HF patients with preserved ejection fraction (HFpEF). In
Furthermore, early diastolic strain rate was reduced in all regions and one study, LV function was examined in eight patients with HFpEF
no period of diastasis was observed in HCM. Therefore, the ventricular and eight normal subjects.52 The HFpEF patients showed decreased LV
response to atrial systole could be seen as strain exceeding the end- filling rate, reversed early-to-atrial filling ratio (E/A), decreased peak
diastolic reference. strain, and early diastolic strain rate, compared with the normal subjects.
A population-based study, the multiethnic subclinical atherosclerosis In another study, 218 patients with LV hypertrophy and normal systolic
(MESA) study, showed evidence of diastolic dysfunction in asymptomatic function were studied using CMR tagging.48 The study showed that HF
individuals with LV hypertrophy.48 Despite similar regional systolic strain progression begins as a regional myocardial dysfunction, which is affected
and strain rate among those with and without LV hypertrophy, the by patterns of hypertrophy. The results revealed a direct relationship
regional diastolic strain rate was significantly reduced among those between regional diastolic dysfunction and increased LV mass. Although
with LV hypertrophy. A provocative study in a small group suggested patients with LV hypertrophy were found to have less global systolic
that diastolic untwisting dysfunction could be identified by SPAMM function than that in normal subjects, systolic strain was not affected.
after a sedentary rest in previously active adult subjects.49 However, diastolic strain rate was significantly reduced with hypertrophy
(1.5 ± 1.1 per second) compared with a group without hypertrophy
Aortic Stenosis (2.2 ± 1.1 per second, P < .001), regardless of age or gender.
Among some of the early CSPAMM studies is one that studied the
effect of AS on alterations in local myocardial motion patterns.40 This Limitations
study found that diastolic apical untwisting in patients with pressure The additional value of CMR tagging applied for the quantification of
overload as a result of AS is delayed compared with healthy controls diastolic function remains to be more fully investigated compared with

Normal subject Hypertrophic cardiomyopathy


Early Mid Atrial Early Mid Atrial
Systole Systole
0.05 diastole diastole systole 0.05 diastole diastole systole
Circumferential strain

0.00 0.00
E D E
D
−0.05 −0.05 A B
C
A B C
−0.10 −0.10

−0.15 −0.15

−0.20 −0.20
0 500 1000 0 500 1000
Cardiac duration (ms) Cardiac duration (ms)
FIG. 22.8  Left ventricular (LV) midwall circumferential strain curve in the inferior wall in a normal subject
(left) and a hypertrophic cardiomyopathy patient (right). Indices of cardiac function are shown on the curves:
A, systolic strain rate; B, total systolic strain; C, early-diastolic strain rate; D, mid-diastolic strain rate; and E,
lengthening subsequent to atrial systole. Measures B and E were used to calculate the percent lengthening
subsequent to atrial systole, defined as E/B. (From Ennis DB, Epstein FH, Kellman P, et al. Assessment of
regional systolic and diastolic dysfunction in familial hypertrophic cardiomyopathy using MR tagging. Magn
Reson Med. 2003;50:638–642, with permission.)
290 SECTION II  Ischemic Heart Disease

gold standard techniques. Currently, the CMR technique is not widely the cardiac cycle, CSPAMM is well suited for the characterization of
accessible to clinical cardiologists and the CSPAMM sequence is not the diastolic portion of the cardiac cycle. Moreover, by the application
currently available on all vendors’ systems. Furthermore, because of the slice-following procedure, the effects of through-plane motion
CSPAMM is based on a subtraction technique, scanning time is doubled can be suppressed and the same tissue elements can be tracked. Because
and susceptibility to diaphragmatic drift or misregistration in serial of the relatively high temporal resolution of the data, rapid cardiac
breath-holds must be considered. However, with the availability of motion components, such as apical diastolic untwisting, can be recorded.
parallel imaging, EPI, or spiral imaging, the number of RF excitations The current data derived from CSPAMM myocardial tagging suggest
for imaging can be minimized. As an alternative to CSPAMM, more that alterations in the diastolic phase of the cardiac cycle can be recorded
conventional tagging12,53 in combination with bSSFP may support an readily. With the advent of parallel imaging, higher magnetic field
improved tagging contrast-to-noise ratio, as does 3 T imaging because strength, and more advanced imaging sequences, persistence of tags
of an increase in T1 (from ∼850 ms at 1.5 T to ∼1200 ms at 3 T for can be prolonged, enabling access to diastolic motion of the left ven-
myocardium and from ∼1200 ms at 1.5 T to ∼1650 ms for blood at tricle. Although CMR myocardial tagging requires off-line computer
3 T). For these reasons, access to early diastolic myocardial motion may analysis, image postprocessing time is now a matter of seconds and
be feasible, even without the availability of CSPAMM. Nevertheless, the technique may offer a new tool for the evaluation of diastolic wall
the investigation of heart wall motion is still the subject of basic research, relaxation in healthy and diseased states. Therefore CSPAMM, with
and appropriate parameters for the quantification of diastolic dysfunc- its inherent ability to visualize both systolic and diastolic myocardial
tion and threshold values for normal subjects remain to be fully defined. motion with or without true myocardial motion tracking, will continue
to play a very important role in advancing regional heart function
evaluation with CMR.
CONCLUSION
The CSPAMM myocardial tagging technique is a noninvasive method
for the quantification of local heart wall motion. Because of the sup-
REFERENCES
pressed fading of the tags and the accessibility to the diastolic phase of A full reference list is available online at ExpertConsult.com
CHAPTER 22  CMR Tagging for Assessment of Left Ventricular Diastolic Function 290.e1

REFERENCES 24. Herzka DA, Guttman MA, McVeigh ER. Myocardial tagging with SSFP.
Magn Reson Med. 2003;49:329–340.
1. Maier SE, Fischer SE, McKinnon GC, Hess OM, Krayenbuehl HP, 25. Zwanenburg JJ, Kuijer JP, Marcus JT, Heethaar RM. Steady-state free
Boesiger P. Evaluation of left ventricular segmental wall motion in precession with myocardial tagging: CSPAMM in a single breathhold.
hypertrophic cardiomyopathy with myocardial tagging. Circulation. Magn Reson Med. 2003;49:722–730.
1992;86:1919–1928. 26. Ibrahim E-SH, Stuber M, Schär M, Osman NF. Improved myocardial
2. Rogers WJ Jr, Shapiro EP, Weiss JL, et al. Quantification of and correction tagging contrast in cine balanced SSFP images. J Magn Reson Imaging.
for left ventricular systolic long-axis shortening by magnetic resonance 2006;24:1159–1167.
tissue tagging and slice isolation. Circulation. 1991;84:721–731. 27. Ryf S, Spiegel MA, Gerber M, Boesiger P. Myocardial tagging with
3. Fischer SE, McKinnon GC, Scheidegger MB, Prins W, Meier D, Boesiger 3D-CSPAMM. J Magn Reson Imaging. 2002;16:320–325.
P. True myocardial motion tracking. Magn Reson Med. 1994;31:401–413. 28. Osman NF, Kerwin WS, McVeigh ER, Prince JL. Cardiac motion tracking
4. Rosen BD, Gerber BL, Edvardsen T, et al. Late systolic onset of regional using CINE harmonic phase (HARP) magnetic resonance imaging. Magn
LV relaxation demonstrated in three-dimensional space by MRI tissue Reson Med. 1999;42:1048–1060.
tagging. Am J Physiol Heart Circ Physiol. 2004;287:H1740–H1746. 29. Ryf S, Tsao J, Schwitter J, Stuessi A, Boesiger P. Peak-combination HARP:
5. Cheng-Baron J, Chow K, Khoo NS, et al. Measurements of changes in left a method to correct for phase errors in HARP. J Magn Reson Imaging.
ventricular volume, strain, and twist during isovolumic relaxation using 2004;20:874–880.
MRI. Am J Physiol Heart Circ Physiol. 2010;298:H1908–H1918. 30. NessAiver M, Prince JL. Magnitude image CSPAMM reconstruction
6. Thompson RB, Paterson I, Chow K, et al. Characterization of the (MICSR). Magn Reson Med. 2003;50:331–342.
relationship between systolic shear strain and early diastolic shear strain 31. NessAiver M, Prince JL. Visualization of myocardial motion using MICSR
rates: insights into torsional recoil. Am J Physiol Heart Circ Physiol. trinary checkerboard display. Inf Process Med Imaging. 2003;18:573–585.
2010;299:H898–H907. 32. Kuijer JP, Zwanenburg JJ, Marcus JT, Hofman MB. CSPAMM real-part
7. Kuijer JP, Marcus JT, Gotte MJ, van Rossum AC, Heethaar RM. reconstruction using an internal phase reference. Proc Intl Soc Magn
Three-dimensional myocardial strains at end-systole and during diastole Reson Med. 2005.
in the left ventricle of normal humans. J Cardiovasc Magn Reson. 33. Fahmy AS, Basha TA, Osman NF. Inherent fat cancellation in
2002;4:341–351. complementary spatial modulation of magnetization. Magn Reson Med.
8. Sampath S, Derbyshire JA, Ledesma-Carbayo MJ, McVeigh ER. Imaging 2009;61:234–238.
left ventricular tissue mechanics and hemodynamics during supine 34. Delfaut EM, Beltran J, Johnson G, Rousseau J, Marchandise X, Cotten A.
bicycle exercise using a combined tagging and phase-contrast MRI pulse Fat suppression in MR imaging: techniques and pitfalls. Radiographics.
sequence. Magn Reson Med. 2011;65:51–59. 1999;19:373–382.
9. D’Hooge J, Heimdal A, Jamal F, et al. Regional strain and strain rate 35. Reyhan M, Ennis DB. Quantitative assessment of systolic and diastolic
measurements by cardiac ultrasound: principles, implementation and left ventricular twist using Fourier analysis of stimulated echoes (FAST)
limitations. Eur J Echocardiogr. 2000;1:154–170. and CSPAMM. J Magn Reson Imaging. 2013;37:678–683.
10. Gilman G, Khandheria BK, Hagen ME, Abraham TP, Seward JB, 36. Wang H, Kadbi M, Kotys M, et al. Orthogonal CSPAMM (OCSPAMM)
Belohlavek M. Strain rate and strain: a step-by-step approach to image MR tagging for imaging ventricular wall motion. Conf Proc IEEE Eng
and data acquisition. J Am Soc Echocardiogr. 2004;17:1011–1020. Med Biol Soc. 2011;2011:535–538.
11. Ingels NB Jr, Daughters GT 2nd, Stinson EB, Alderman EL. Measurement 37. Atalar E, McVeigh ER. Optimization of tag thickness for measuring
of midwall myocardial dynamics in intact man by radiography of position with magnetic resonance imaging. IEEE Trans Med Imaging.
surgically implanted markers. Circulation. 1975;52:859–867. 1994;13:152–160.
12. Yun KL, Niczyporuk MA, Daughters GT 2nd, et al. Alterations in left 38. Nagel E, Stuber M, Lakatos M, Scheidegger MB, Boesiger P, Hess OM.
ventricular diastolic twist mechanics during acute human cardiac Cardiac rotation and relaxation after anterolateral myocardial infarction.
allograft rejection. Circulation. 1991;83:962–973. Coron Artery Dis. 2000;11:261–267.
13. Potel MJ, Rubin JM, MacKay SA, Aisen AM, Al-Sadir J, Sayre RE. 39. Ennis DB, Epstein FH, Kellman P, Fananapazir L, McVeigh ER, Arai AE.
Methods for evaluating cardiac wall motion in three dimensions using Assessment of regional systolic and diastolic dysfunction in familial
bifurcation points of the coronary arterial tree. Invest Radiol. hypertrophic cardiomyopathy using MR tagging. Magn Reson Med.
1983;18:47–57. 2003;50:638–642.
14. Deleted in review. 40. Stuber M, Scheidegger MB, Fischer SE, et al. Alterations in the local
15. Axel L, Dougherty L. MR imaging of motion with spatial modulation of myocardial motion pattern in patients suffering from pressure overload
magnetization. Radiology. 1989;171:841–845. due to aortic stenosis. Circulation. 1999;100:361–368.
16. Fischer SE, McKinnon GC, Maier SE, Boesiger P. Improved myocardial 41. Swoboda PP, Larghat A, Zaman A, et al. Reproducibility of myocardial
tagging contrast. Magn Reson Med. 1993;30:191–200. strain and left ventricular twist measured using complementary spatial
17. Ibrahim el SH. Myocardial tagging by cardiovascular magnetic resonance: modulation of magnetization. J Magn Reson Imaging. 2014;39:887–894.
evolution of techniques—pulse sequences, analysis algorithms, and 42. Rademakers FE, Buchalter MB, Rogers WJ, et al. Dissociation between left
applications. J Cardiovasc Magn Reson. 2011;13:36. ventricular untwisting and filling. Accentuation by catecholamines.
18. Stuber M, Spiegel MA, Fischer SE, et al. Single breath-hold slice-following Circulation. 1992;85:1572–1581.
CSPAMM myocardial tagging. MAGMA. 1999;9:85–91. 43. Grossman W, Jones D, McLaurin LP. Wall stress and patterns of
19. Stuber M, Fischer SE, Scheidegger MB, Boesiger P. Toward high- hypertrophy in the human left ventricle. J Clin Invest. 1975;56:56–64.
resolution myocardial tagging. Magn Reson Med. 1999;41:639–643. 44. Hess OM, Lavelle JF, Sasayama S, Kemper WS, Ross J. Diastolic
20. Ryf S, Schwitter J, Spiegel MA, et al. Accelerated tagging for the myocardial wall stiffness of the left ventricle in chronic pressure overload.
assessment of left ventricular myocardial contraction under physical Eur Heart J. 1982;3:315–324.
stress. J Cardiovasc Magn Reson. 2005;7:693–703. 45. Matter C, Mandinov L, Kaufmann P, Nagel E, Boesiger P, Hess OM.
21. Ryf S, Kissinger KV, Spiegel MA, et al. Spiral MR myocardial tagging. [Function of the residual myocardium after infarct and prognostic
Magn Reson Med. 2004;51:237–242. significance]. Z Kardiol. 1997;86:684–690.
22. Taylor AM, Keegan J, Jhooti P, Gatehouse PD, Firmin DN, Pennell DJ. A 46. Russel IK, Brouwer WP, Germans T, et al. Increased left ventricular
comparison between segmented k-space FLASH and interleaved spiral torsion in hypertrophic cardiomyopathy mutation carriers with normal
MR coronary angiography sequences. J Magn Reson Imaging. wall thickness. J Cardiovasc Magn Reson. 2011;13:3.
2000;11:394–400. 47. Feinstein JA, Epstein FH, Arai AE, et al. Using cardiac phase to order
23. Meyer CH, Pauly JM, Macovski A, Nishimura DG. Simultaneous spatial reconstruction (CAPTOR): a method to improve diastolic images. J Magn
and spectral selective excitation. Magn Reson Med. 1990;15:287–304. Reson Imaging. 1997;7:794–798.
290.e2 SECTION II  Ischemic Heart Disease

48. Edvardsen T, Rosen BD, Pan L, et al. Regional diastolic dysfunction in 52. Ibrahim el SH, Miller AB, White RD. The relationship between aortic
individuals with left ventricular hypertrophy measured by tagged stiffness and E/A filling ratio and myocardial strain in the context of left
magnetic resonance imaging—the Multi-Ethnic Study of Atherosclerosis ventricular diastolic dysfunction in heart failure with normal ejection
(MESA). Am Heart J. 2006;151:109–114. fraction: insights from magnetic resonance imaging. Magn Reson Imaging.
49. Dorfman TA, Rosen BD, Perhonen MA, et al. Diastolic suction is 2011;29:1222–1234.
impaired by bed rest: MRI tagging studies of diastolic untwisting. J Appl 53. Axel L, Dougherty L. Heart wall motion: improved method of spatial
Physiol. 2008;104:1037–1044. modulation of magnetization for MR imaging. Radiology.
50. Nagel E, Stuber M, Burkhard B, et al. Cardiac rotation and relaxation in 1989;172:349–350.
patients with aortic valve stenosis. Eur Heart J. 2000;21:582–589.
51. Uddin A, Fairbairn TA, Swoboda PP, et al. CMR evaluation of change in
myocardial strain following transcatheter aortic valve implantation. J
Cardiovasc Magn Reson. 2014;16(suppl 1):P260.
23 
Magnetic Resonance Imaging of Coronary
Arteries: Technique
Mehmet Akçakaya, Claudia Prieto, René M. Botnar, and Reza Nezafat

Despite significant efforts in prevention and treatment, coronary artery contrast agents)17-19 and thrombus/hemorrhage20,21 visualization. The
disease (CAD) remains the leading cause of death in the United States, combination of these techniques could add invaluable prognostic
accounting for one in every seven deaths.1 Each year nearly 700,000 information for patients at risk or with known CAD. Thus techni-
Americans are estimated to have a new myocardial infarction (MI), cal developments are being investigated to allow coronary MRI to
and nearly 325,000 to have a recurrent infarction. Furthermore, an achieve similar diagnostic lumen accuracy as the current clinical
additional estimated 165,000 will have their first silent MI.1 The current gold standard, as well as plaque characterization. Main technical
clinical “gold standard” for the diagnosis of significant (≥50% diameter challenges include suboptimal spatial resolution (as a result of long
stenosis) CAD is catheter-based invasive x-ray angiography. More than acquisition times required), and coronary motion suppression with
a million catheter-based x-ray coronary angiograms are performed unpredictable scan times (depending largely on the breathing pattern
annually in the United States,1 with a higher volume in Europe. However, of the subject). In this chapter, we will review the technical imaging
these invasive tests have a relatively low yield, with less than 40% of strategies for MRI of coronary arteries, coronary vessel walls, and
patients referred for x-ray coronary angiography having obstructive coronary veins. The clinical results of coronary MRI are addressed
CAD,2 unnecessarily exposing these patients to the potential risks and in Chapter 24.
complications of an invasive test that includes ionizing radiation and
iodinated contrast.3,4 To relieve symptoms or decrease pharmaceutical
use, percutaneous coronary intervention in single vessel disease is com-
CORONARY MAGNETIC RESONANCE IMAGING
monly performed, but the greatest impact on mortality occurs with The early approaches to coronary artery MRI were based on two-
mechanical intervention among patients with left main (LM) and mul- dimensional (2D) breath-hold electrocardiogram (ECG)-triggered
tivessel CAD. Thus alternative noninvasive imaging modalities, which segmented sequences.22,23 Subsequently, three-dimensional (3D) free-
allow direct visualization of the proximal and mid native coronary breathing approaches have replaced 2D breath-hold approaches, enabling
vessels for the accurate identification/exclusion of LM/multivessel CAD, greater anatomical coverage and higher signal-to-noise ratio (SNR).
are desirable. Three-dimensional coronary MRI can be acquired using a targeted
Cardiovascular magnetic resonance (CMR) imaging is a very or whole-heart coverage of the coronary anatomies. In the targeted
promising noninvasive tool for comprehensive early risk assessment, technique,24 a double-oblique 3D volume aligned along the major axis
guidance of therapy, and treatment monitoring of CAD. CMR is of the left or right coronary artery (RCA) is acquired.25-27 For the visu-
considered the gold standard for the assessment of cardiac anatomy, alization of the RCA, the imaging plane passing through the proximal,
biventricular systolic function, myocardial viability (late gadolinium mid, and distal coordinates of the RCA is identified and the targeted
enhancement [LGE]) and rest/stress myocardial perfusion.5-8 Clini- 3D coronary sequence is acquired in this orientation, typically with a
cal research studies also have demonstrated its usefulness for quan- 30-mm slab with 20 slices, using a segmented acquisition.26,27 For the
titative myocardial tissue characterization (T 1 and T2 relaxation imaging of the left main (LM), left anterior descending (LAD), and
time mapping), and its ability to differentiate between healthy and left circumflex (LCX) coronary arteries, a 3D volume is interactively
diseased tissue.9,10 Coronary magnetic resonance imaging (MRI) is prescribed in the axial plane centered about the LM coronary artery
a noninvasive diagnosis alternative to catheter-based x-ray angiog- (Fig. 23.1).27,28 In the whole-heart coronary MRI technique,29-43 an axial
raphy among patients with suspected anomalous coronary artery (or coronal) 3D volume encompassing the entire heart is sampled in
disease11 and coronary artery aneurysms.12 Although coronary a single acquisition, in a manner analogous to coronary MDCT. This
multi-detector computed tomography (MDCT) offers superior iso- facilitates the imaging setup via simpler slab prescription, and provides
tropic spatial resolution and more rapid imaging, coronary MRI has a more complete anatomical coverage, positioned ~1 cm above the
advantages over MDCT in several respects, including the absence of LM and extending to the inferior cardiac border. However, based on
ionizing radiation or iodinated contrast, which facilitates follow-up single-center trials to date, it has not been shown to be superior to the
scanning, as well as smaller artifacts related to epicardial calcium. targeted approach for CAD assessment (Table 23.1).
Because of the advantages of coronary MRI and its diagnostic accu- Targeted thin-slab 3D acquisitions have been acquired using both
racy, coronary MRI is recommended and deemed appropriate in gradient recalled echo (GRE) and balanced steady-state free precession
patients suspected of anomalous coronary artery disease by both the (bSSFP) sequences.44 A thin-slab 3D targeted acquisition with a GRE
American College of Cardiology and American Heart Association.13,14 sequence results in more homogenous blood pool signal, but is heavily
However, CMR assessment of coronary lumen integrity and plaque dependent on the inflow of unsaturated protons.45 Saturation effects will
burden/activity remains challenging. Nevertheless, CMR has shown cause a local signal loss if coronary artery flow is slow or stagnant. This
great potential for coronary lumen,15,16 plaque (with and without signal loss is often relatively exaggerated, as compared with the lumen

291
292 SECTION II  Ischemic Heart Disease

1.5 T, SSFP appears to be the sequence of choice as a result of its higher


blood–myocardium contrast and superior inflow properties.45
Even with these technical advances, clinical acceptance of coronary
MRI remains challenging because of coronary artery motion, long scan
times, limited spatial resolution, suboptimal SNR and blood–myocardium
contrast-to-noise-ratio (CNR). The technical challenges in coronary
artery MRI are different from other CMR acquisitions as a result of
unique issues including the coronary artery: (1) small caliber (3–6 mm
diameter), (2) high level of tortuosity, (3) near-constant motion during
LAD both the respiratory and the cardiac cycles, and (4) surrounding signal
from adjacent epicardial fat and myocardium.
LM
Cardiac-Induced Motion
Bulk epicardial coronary artery motion is a major impediment to coro-
LCX nary CMR, and it can be separated into motion related to direct cardiac
contraction/relaxation during the cardiac cycle and motion attributed
to superimposed diaphragmatic and chest wall movement during res-
piration. The magnitude of motion from each component may greatly
exceed the coronary artery diameter, leading to blurring artifacts in the
FIG. 23.1  Reformatted coronary artery magnetic resonance image of
absence of motion–suppressive methods.
the left coronary system acquired using a targeted free-breathing acquisi- To compensate for bulk cardiac motion, accurate external ECG
tion with real-time navigator gating and tracking in a healthy adult subject. synchronization with QRS detection is required, and vector ECG
The transverse acquisition displays the left main (LM), left anterior approaches are preferred.48 Coronary artery motion during the cardiac
descending (LAD), and the left circumflex (LCX) coronary arteries. The cycle has been characterized using both catheter based x-ray angiog-
in-plane spatial resolution is 0.7 × 1.0 mm2. raphy49,50 and CMR.51-53 Both the proximal/mid RCA and the LAD display
a triphasic pattern, with the magnitude of in-plane motion nearly twice
as great for the RCA. Coronary artery motion is minimal during iso-
volumic relaxation, approximately 350 to 400 ms after the R wave, and
TABLE 23.1 Single-Center again at mid diastole (immediately before atrial systole). The duration
Echocardiogram-Triggered, Free-Breathing, of LAD diastasis is longer than that of the RCA, and it begins earlier
Targeted Three-Dimensional and Whole- in the cardiac cycle.54 The duration of the mid diastolic diastasis period
Heart Coronary Magnetic Resonance Imaging is inversely related to the heart rate and dictates the preferred coronary
With and Without Contrast Agents artery data acquisition interval.
As compared with MDCT in which the acquisition is constrained
Single-
by gantry rotation, for coronary artery MRI the acquisition interval is
Center/ No. of
adapted to the heart rate/diastasis interval using a patient-specific dias-
Study Multicenter Patients Sensitivity Specificity
tasis period. This can be readily identified by the acquisition of high
Noncontrast 3D Targeted Coronary MRI temporal resolution cine dataset orthogonal to the long axis of the
Kim24 Multicenter 109 88%–98% 32%–52% proximal/mid RCA and of the LAD. Semiautomated tools to identify
Bunce126 Single-center 46 50%–89% 72%–100% the optimal data acquisition window have also been proposed.55,56 For
Sommer127 Single-center 107 74%–88% 63%–91% patients with a heart rate of 60 to 70 beats per minute, a coronary
Bogaert128 Single-center 21 85%–92% 50%–83% artery MRI acquisition duration of ~80 ms during each cardiac cycle
results in improved image quality.28 The duration must be further
Noncontrast 3D Whole-Heart Coronary MRI abbreviated (e.g., <50 ms) at higher heart rates, whereas with brady-
Jahnke129 Single-center 21 79% 91% cardia, the acquisition interval can be expanded to 120 ms or longer.
Sakuma41 Single-center 39 82% 91% The use of patient-specific acquisition windows serves to reduce overall
Sakuma40 Single-center 131 82% 90% scan time.57,58 Image degradation can be caused by sinus arrhythmia,
Pouleur130 Single-center 77 100% 72% leading to heart rate variability, which is common especially in younger
Kato15 Multicenter 138 88% 72% adults.59 An adaptive real-time arrhythmia rejection algorithm can correct
for heart rate variability and improves coronary artery MRI quality.56
Contrast Enhanced 3D Whole-Heart Coronary MRI
Yang124 Single-center 62 94% 82% Respiratory-Induced Motion
Yang131 Multicenter 272 91% 80%
The second major challenge for coronary artery MRI is compensation
3D, Three-dimensional; MRI, magnetic resonance imaging. for bulk respiratory motion. With inspiration, the diaphragm may
descend up to 30 mm and the chest wall expands, resulting in an inferior
displacement and anterior rotation of the heart.60 Several approaches
stenosis. Compared with GRE sequences, bSSFP provides intrinsically have been proposed to minimize respiratory motion artifacts, including
higher SNR because of its balanced gradients and improved blood- sustained end-expiratory breath-holding, chest wall bellows, respiratory
myocardium contrast attributed to its T1/T2 weighting,46 with reduced navigators, fat navigators, and self-gating methods.
sensitivity to inflow effects. Both GRE and bSSFP have been used for Prolonged (15–20 seconds) end-expiratory breath-holds were used
targeted 3D coronary MRI, where both have shown similar diagnostic to suppress respiratory motion in initial 2D coronary artery MRI
accuracy for CAD.46,47 For whole-heart noncontrast coronary MRI at methods.61 Breath-holding offers the advantage of relative ease of
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 293

implementation in compliant subjects, but it limits the temporal acqui- to appropriately adjust spatial coordinates.72 This technique allows for
sition window, image spatial resolution, and anatomic coverage. Addi- the use of wider gating windows and increased navigator efficiency,
tionally, many patients are unable to adequately sustain a breath-hold. leading to shorter scan times. Real-time tracking implementations with
Furthermore, slice registration errors (attributed to variability in end- a 5-mm diaphragmatic gating window are often used with a navigator
expiratory diaphragmatic position) are very common as is diaphragmatic efficiency approaching 50%.69 Coronary artery MRI with real-time
drift during the breath-hold61-64 and may occur in up to half of patients.54 navigator tracking has been shown to minimize registration errors (as
Supplemental oxygen and hyperventilation (separately or in combina- compared with breath-holding) while maintaining or improving the
tion) can be used to prolong the breath-hold duration,63,64 but these image quality.69,73 It should also be noted that the quality of coronary
methods may not be appropriate for all patients, and both diaphragmatic artery MRI is improved by using consistent ECG timing, as well as
drift and slice registration errors persist.64 respiratory suppression methodology for both the coronary localizing/
Diaphragmatic respiratory navigators, first proposed by Ehman65 motion scout images and for the coronary artery MRI acquisitions.74
for abdominal MRI, enable free-breathing acquisitions without the A number of refinements to the navigator method have been pro-
stringent time constraints and patient cooperation requirements imposed posed. Although a “fixed” superior–inferior correction factor of 0.6
by multiple breath-holds, and thus offer superior spatial resolution (with no left–right or anterior–posterior correction)72,75 is commonly
opportunities. Although the specifics of navigator implementation vary used, significant individual variability has been observed.62 A subject-
among CMR vendors, in the ideal implementation, the navigator can specific tracking factor has been advocated and shown to improve the
be positioned at any interface that accurately reflects respiratory motion, quality of coronary images when the subject-specific tracking factor
including the dome of the right hemidiaphragm (Fig. 23.2),66,67 the left differs from 0.6.76 The use of multiple navigator locations, use of leading
hemidiaphragm, the anterior chest wall, the anterior free wall of the and trailing navigators, and use of navigators that provide guidance
left ventricle,67,68 or even through the coronary artery of interest. The for affine transformations (i.e., 3D translations and rotations) of the
navigator should not cause an image artifact and should be temporally slice prescription for each heart beat have been proposed.37,77-79 The
located immediately preceding the imaging portion of the sequence affine transformation permits use of larger navigator windows, and
with data accepted (used for image reconstruction) only when the hence higher navigator efficiency. It has also been proposed that the
navigator indicates that the “interface” (e.g., diaphragm position) falls heart itself be tracked,34,80-82 such as with methods that track the epi-
within a user-defined window. The dome of the right hemidiaphragm cardial fat to detect the heart position.83-86 Navigator gating with fixed
has become the preferred location26,68 because of the simplicity and scan efficiency has also been studied, which results in imaging at a fixed
ease in set-up, where the motion of the right hemidiaphragm in the scan time based only on heart rate and acquisition duration.87 Novel
superior-inferior direction can be tracked. From CMR studies of cardiac k-space trajectories and various image reconstruction based methods,
border position during the respiratory cycle, it was observed that the such as cross correlation of low resolution images, have also been pro-
ratio between cardiac and diaphragmatic displacement is ~0.6 for the posed for respiratory motion compensation.42,88-90
RCA and ~0.7 for the left coronary artery at end-expiration,60 although Self-navigation methods have also been proposed to derive the
there is variability among subjects62,69,70 and position (e.g., supine vs. respiratory-induced motion of the heart from the acquired data itself
prone imaging).71 This rule-of-thumb offers the opportunity for pro- without the need of either a one-dimensional (1D) navigator echo or
spective navigator gating with real-time tracking,67,69 in which the posi- a heart-diaphragm tracking factor. Respiratory-induced displacements
tion of the interface (diaphragm) is determined, and the slice position of the heart can be directly estimated from the repetitive acquisition
coordinates can then be shifted in real time (before the data collection) of the central k-space point91 or the central k-space line,92-95corresponding

Position of interface during


each RR interval
20 mm
Lung
80 mm
5 mm

RHD Nav Liver

A B

C
FIG. 23.2  Positioning and utility of the respiratory navigator. Coronal (A) and axial (B) thoracic images for
positioning the navigator at the dome of the right hemidiaphragm (RHD Nav). (C) Respiratory motion of the
lung-diaphragm interface is recorded using a two-dimensional selective navigator with the lung (superior)
and liver (inferior) interface. In this example, the maximum excursion between end-inspiration and end-
expiration is ~11 mm. The position of the lung-liver interface at each RR interval is indicated by the broken
line in the middle of (C). Data are only accepted if the lung-liver interface is within the acceptance window
of 5 mm. Data acquired with the navigator outside of the window are rejected. Accepted data are indicated
by the broken green line at the bottom of (C).
294 SECTION II  Ischemic Heart Disease

to zero-dimensional or 1D projections of the field of view. Similar to


the 1D navigator echo approaches, self-navigation methods typically Image Quality Assessment
perform motion correction only in the foot-head translational motion. Epicardial fat and the myocardium surround the coronary arteries.
Another drawback of the self-navigation approach is that the inclusion Thus CNR can be improved by suppressing the fat and myocardium
of static structures, such as the chest wall, can degrade motion estima- signals surrounding the coronary arteries. Frequency (spectrally) selec-
tion and correction. tive prepulses are applied to saturate signal from fat tissue, thereby
To overcome these problems and account for more complex motion, allowing visualization of the underlying coronary arteries.61,104 To dif-
several 2D and 3D image-based navigator (so-called iNAV) approaches ferentiate myocardium and the coronary lumen, endogenous contrast
have been proposed for coronary artery MRI96-103 (Fig. 23.3). In these preparation techniques are commonly used.28,104-107 Two methods that
approaches a low-resolution 2D or 3D image is acquired in every heart- can enhance the contrast between the coronary lumen and underlying
beat before (or after) the coronary MRI data acquisition. The main myocardium are T2 preparation prepulses28,105,106,108 and magnetization
advantage of this approach is that the moving heart can be spatially transfer contrast (MTC).104,107 The former is often used for coronary
isolated from surrounding static tissues and the respiratory-induced artery MRI because it also suppresses deoxygenated venous blood,
cardiac motion can be directly estimated via (rigid or affine) image- whereas the latter is used for coronary vein CMR.109
registration of iNAVs at different respiratory positions. Self-navigation The limited SNR in coronary artery MRI, along with constraints
and image-based navigator methods can be used to gate the acquisition on acquisition duration, restricts the spatial resolution in the acquisi-
and correct for motion within a small gating window, as described for tion. Spatial resolution requirements for clinical coronary artery MRI
1D navigator echoes. Furthermore, because these methods directly track depend on whether the goal is to identify the origin and proximal
heart motion/position, a much larger gating window can be used, or course of the coronary artery (e.g., issues of anomalous coronary disease)
it can be removed entirely, thereby increasing the scan efficiency to, or or to identify focal stenoses in the proximal and middle segments.
close to, 100%. These promising approaches may lead to shorter and The SNR of coronary MRI can be enhanced by higher B0 field
predictable scan times. strength,110 larger 3D spatial coverage,43 vasodilator administration, and

Gated and tracked Nonlinear motion correction Translational correction No motion correction

LAD

RCA Ao

LAD

Ao
RCA

FIG. 23.3  Nearly 100% scan efficiency nonrigid motion correction coronary artery magnetic resonance image.
This approach is based on beat-to-beat translation correction followed by respiratory bin-to-bin nonlinear
motion correction. Reformatted images are shown for conventional navigator gated and tracked acquisition,
nonrigid motion correction, translation correction only, and no motion correction for two different subjects.
Blurring present in the no motion correction images is reduced with translation correction and sharpness
further increased with the nonlinear motion correction approach (boxes). The distal portions of both the right
coronary artery (RCA) and the left anterior descending (LAD) coronary artery are particularly affected by
motion (arrows). The nonlinear motion correction approach has similar image quality to the conventional
navigator gated and tracked acquisition; however, the latter requires significantly longer scan times (~2 to
3 times longer). Ao, Aorta.
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 295

contrast agents based on gadolinium chelates. The intrinsically higher in coronary artery MRI is the administration of vasodilators because
SNR associated with higher magnetic field strengths may be advanta- the increased coronary blood flow secondary to vasodilatation reduces
geous for noncontrast coronary MRI. However, additional considerations, the inflow saturation effects.113,114 Fig. 23.5 demonstrates the impact of
such as higher B1 and B0 inhomogeneity and higher specific absorption sublingual isosorbide dinitrate administration on 3D targeted coronary
rate, affect certain aspects of coronary MRI, such as the diminished artery MRI up to 30 minutes after drug administration, in terms of
utility of bSSFP sequences at 3 T. Hence, GRE sequences, which are subjective image quality and objective SNR and vessel sharpness.
less sensitive to field inhomogeneity, as well as localized shimming111 The administration of exogenous gadolinium contrast agents (both
and contrast preparation techniques that deal with B1 inhomogene- extracellular30,115,116 and intravascular117-122) that shorten the T1 relaxation
ities108,112 have been advocated. time provides an alternative flow-independent approach to improve
The increased coverage of whole-heart coronary MRI can potentially SNR and CNR. Because conventional extracellular contrast agents (e.g.,
improve the SNR, but this also increases the scan time. Thus the SNR gain gadopentetate dimeglumine [Gd-DTPA]) diffuse rapidly into the inter-
is often counteracted by the need for accelerated imaging to reduce scan stitial space, early contrast-enhanced coronary artery MRI studies focused
time, which carries an SNR penalty. Furthermore, whole-heart imaging on breath-hold coronary artery MRI to take advantage of the first passage
suffers from saturation effects of the inflowing blood magnetization.45 of these agents.116 However, both the breath-hold and first-pass aspects
Despite these issues, excellent image quality of whole-heart coronary of such approaches limit the spatial resolution and are unsuitable for
MRI has been shown in several studies,40,43 and an example from a single- whole-heart coronary acquisitions.123 Following the availability of a
center study40 is depicted in Fig. 23.4. Another technique to improve SNR high relaxivity extracellular contrast agent, gadobenate dimeglumine

A B C
FIG. 23.4.  Whole-heart coronary artery magnetic resonance image. (A) A stenosis in the left anterior descending
(LAD) coronary artery is visualized using a curved multiplanar reconstruction (arrow). (B) A three-dimensional
view of LAD with stenosis is depicted in the volume-rendered image. (C) X-ray coronary angiography con-
firms proximal LAD stenosis (arrowhead). (Modified from Sakuma H, Ichikawa Y, Chino S, et al. Detection
of coronary artery stenosis with whole-heart coronary magnetic resonance angiography. J Am Coll Cardiol.
2006;48:1946–1950.)

Pre 1 min 7 min 13 min 22 min


Subject C (2.5 mg)

*3.3 *3.8 *4.0 *4.0 *4.0


#39 #43 #43 #46 #49
~3161 ~3527 ~3864 ~4396 ~3890
Pre 1 min 8 min 13 min 18 min
Subject D (5 mg)

*3.0 *3.1 *3.6 *3.2 *3.6


#36 #38 #43 #45 #40
~1791 ~2002 ~2143 ~2175 ~2186
FIG. 23.5  Reformatted images from a targeted three-dimensional (3D) coronary magnetic resonance image
of the right coronary artery (RCA). Images were acquired before and after sublingual isosorbide dinitrate
administration on two healthy subjects using a 3D free-breathing balanced steady-state free precession fol-
lowing 2.5 mg (top row) or 5 mg doses (bottom row) as a function of time. Improved RCA vasodilation and
signal enhancement can be observed in all images after isosorbide dinitrate (arrows in top and bottom row).
The enhanced signal-to-noise ratio following isosorbide dinitrate also allows for improved visualization of the
distal segments.
296 SECTION II  Ischemic Heart Disease

Ao RCA
LAD LAD
RCA
Ao
LAD
LCX
LCX

OM1
RCA
OM2 A B
A B
FIG. 23.7  Whole-heart balanced steady-state free precession coronary
magnetic resonance image acquired with a bolus injection of gadobenate
dimeglumine. (A) Three-dimensional volume rendering of the acquisition
volume. (B) Corresponding reformatted whole-heart image. All three
major coronary arteries and distal branches are clearly depicted. LAD,
Left anterior descending coronary artery; LCX, left circumflex coronary
LAD RCA
LM artery; RCA, right coronary artery.

LCX

Furthermore, the bolus contrast injection method is advantageous in


OM1 multiple ways because it simplifies the initiation time of coronary MRI
C OM2 D acquisition compared with slow infusion, and it is compatible with late
gadolinium enhancement imaging, which enables the assessment of
coronary artery stenosis and myocardium viability using a single bolus
LM LAD contrast injection. Many patients with CAD also have renal dysfunction.
The use of gadolinium contrast coronary MRI must consider the patient’s
OM1 renal function for issues related to nephrogenic systemic fibrosis and
RCA
long-term retained gadolinium.

LCX
OM2
CORONARY MAGNETIC RESONANCE IMAGING—
ADVANCED METHODS
The sensitivity and specificity of coronary MRI for detection of CAD
remain moderate, based on single-center40,41,124,126-130 (Table 23.1) and
multicenter15,24,131 studies (see Chapter 24), despite the tremendous
E F technical improvements in the last two decades. Coronary motion, SNR
and CNR remain as major impediments to coronary MRI, and these
FIG. 23.6  Contrast-enhanced whole-heart three-dimensional coronary issues need to be addressed before clinical prime time for coronary
magnetic resonance image with a slow infusion of gadobenate dimeglu- MRI. To overcome some of these hurdles, several CMR centers continue
mine contrast agent in a patient with atypical chest pain. (A, B) Contrast- with the development and implementation of novel approaches, includ-
enhanced whole-heart maximum intensity projection images show a
ing non-Cartesian acquisitions, accelerated imaging techniques, coronary
significant stenosis in the proximal left circumflex (LCX) coronary artery
and a nonsignificant stenosis in the middle right coronary artery (RCA;
vein MRI, and higher field imaging.
arrows), respectively. (C, D) The volume-rendered images have the same Non-Cartesian acquisitions provide efficient k-space traversals that
findings in LCX and RCA (arrows). These were consistent with the lead to incoherent or less visually significant artifacts. Thus alternative
findings (arrows) of conventional coronary angiography (E, F). Ao, Aorta; non-Cartesian k-space acquisitions, including spiral and radial coro-
LM, left main; OM, obtuse marginal artery. (Modified from Yang Q, Li nary MRI, have received attention. The use of spiral coronary artery
K, Liu X, et al. Contrast-enhanced whole-heart coronary magnetic reso- MRI was first reported more than 2 decades ago.132 Spiral acquisitions
nance angiography at 3.0-T: a comparative study with X-ray angiography are advantageous to Cartesian acquisitions in several respects, includ-
in a single center. J Am Coll Cardiol. 2009;54:69–76.) ing a more efficient filling of k-space, enhanced SNR,46,133 and favor-
able flow properties. However, drawbacks of spiral trajectories include
increased sensitivity to magnetic field inhomogeneity and longer image
(Gd-BOPTA; MultiHance; Bracco Imaging SpA, Milan, Italy), improved reconstruction. Interleaved spiral imaging is typically used because of
whole-heart coronary artery MRI at 3 T was shown to be feasible using reduced artifacts,132-135 although a single-shot k-space trajectory can also
a T1-weighted inversion recovery (IR) GRE sequence with a slow infu- be employed. Both breath-hold and free-breathing/navigator-gated 2D
sion of Gd-BOPTA.30 An example of a contrast-enhanced whole-heart acquisitions can be performed with spiral coronary artery.46,117,133,135 Com-
coronary artery MR image from a CAD patient and the corresponding pared with conventional Cartesian approaches, single spiral acquisitions
x-ray angiogram is shown in Fig. 23.6, demonstrating agreement between (per RR interval) afford a near 3-fold improvement in SNR.46,133 Hence,
two modalities in detecting significant stenosis.124 A bolus infusion of acquiring two spirals during each RR interval will halve the acquisition
Gd-BOPTA for coronary MRI has also been reported,125 and an example time, while maintaining superior SNR (vs. Cartesian acquisition) and
depicted in Fig. 23.7 shows a clear visualization of the three major CNR. Variable density spirals have also shown benefit.90 Radial trajec-
coronary vessels in the reformatted and 3D-volume rendered images. tories also enable more rapid acquisitions, while decreasing sensitivity
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 297

to motion. Data in healthy subjects appear promising46,136-138 and may with non-Cartesian imaging, such as with spiral acquisitions, to enable
be particularly beneficial for coronary wall imaging.82,139,140 whole-heart acquisitions in a single prolonged breath-hold89 or with
Parallel imaging techniques such as generalized autocalibrating par- 3D radial trajectories.151
tially parallel acquisition (GRAPPA)141 or sensitivity encoding (SENSE)142 Coronary MRI at high fields has been an active area of research as
are the most commonly used clinical acceleration technique for coronary a result of potential benefits in SNR and CNR. SNR is directly related
artery MRI.30,122,124,125 Resultant acceleration rates of up to 2-fold while to field strength (B0), and thus 3 T imaging would offer the opportu-
using 5 to 16 element cardiac-coil arrays, and up to 4-fold acceleration nity to double SNR compared with 1.5 T systems.152 Although the vast
rate using 32-channel coils have been achieved.143,144 Currently, parallel majority of coronary artery MRI investigations have been performed
imaging is considered the state-of-the-art accelerated imaging technique on 1.5 T, clinical 3 T systems are commonly available and becoming
for whole-heart coronary artery MRI, and is commonly used for clinical the platform of choice for CMR. Technical challenges to 3 T include
imaging. increased susceptibility artifacts, field inhomogeneities,108 reduced
In addition to the non-Cartesian trajectories145 described previously, T2*,153,154 increased specific absorption rate (SAR), T1 prolongation,
compressed sensing (CS) has emerged as a robust alternative accelera- and the amplified magnetohydrodynamic effect.48 At 3 T, free-breathing
tion technique that exploits the sparsity of the image in a transform navigator and breath-hold 3D coronary artery MRI studies in healthy
domain.146,147 CS also requires an incoherent undersampling pattern, volunteers have demonstrated >50% improvement in SNR with impres-
which can be achieved by random undersampling of k-space data in sive image quality using segmented k-space gradient echo or SSFP,155
the ky-kz plane for 3D Cartesian acquisitions. In high-resolution coronary as well as spiral and contrast enhanced methods.156-158 Coronary MRI
MRI, an advanced CS-based reconstruction strategy was shown to at 3 T using SSFP sequences is challenging because of increased field
provide reconstructions with reduced blurring compared with conven- inhomogeneity and high SAR; thus GRE sequences have become widely
tional CS techniques148 and was successfully used in contrast-enhanced used for coronary CMR at 3 T. To reduce the impact of B1 inhomogene-
whole-heart coronary MRI.149 More recently, for highly-accelerated ity at the high field strengths, improved preparation sequences such as
sub-millimeter resolution whole-heart coronary MRI, CS was shown adiabatic T2 magnetization preparation108,112 and adiabatic fat saturation
to outperform parallel imaging at 6-fold accelerated imaging in a head- have also been used. Fig. 23.9 shows example coronary MRI acquired at
to-head comparison150 (Fig. 23.8). CS can also be used in conjunction 3 T using improved T2 magnetization preparation, which suppresses the

LOST (R6) SENSE (R6)

LM LM

FIG. 23.8  Example images from two separate highly-


accelerated sub-millimeter resolution whole-heart coronary
artery magnetic resonance images. An example coronal
slice (top) containing a cross section of the left main (LM)
shows that sensitivity encoding (SENSE) images, acquired
with 6-fold uniform undersampling (right), suffers from
LCX LCX
noise amplification. In contrast, the LM is clearly visualized
using an advanced compressed sensing–based technique
(low-dimensional-structure self-learning and thresholding
RCA RCA [LOST]), acquired using 6-fold random undersampling (left).
In the reformatted coronal images (bottom), the proximal
left circumflex (LCX) coronary artery cannot be tracked
because of the high noise level in the SENSE reconstruc-
tion, but right coronary artery (RCA) and LCX branches
are visualized with the LOST technique.

FIG. 23.9  Example reformatted three-dimensional coronary


magnetic resonance image of the right coronary artery at 3 T
(A) acquired with no T2-preparation; (B) with T2-preparation;
and (C) with adiabatic T2-preparation. Arrows in B point to the
artifacts resulting from T2-preparation sequence. The banding
C artifacts are suppressed in C, where the homogeneity of the
A B
signal is also improved.
298 SECTION II  Ischemic Heart Disease

banding artifact resulting from conventional T2 magnetization prepa- One limitation of black-blood approaches for coronary vessel wall
ration. Coronary MRI at even higher field strengths, such as 7 T,159 is is that these techniques rely on blood flow, which may be insufficient
even more challenging. Several technical issues, including coil design, in patients with very low coronary flow or heavily calcified coronary
motion compensation, and B0 and B1 field inhomogeneity, need to be plaque. Furthermore, these acquisitions are limited in coverage to 2D
addressed before clinical evaluation is possible. Finally, simultaneous acquisitions or targeted 3D scans. Because the coronary arteries have
black-blood LGE methods and bright-blood coronary MRI imaging
are being explored as a means of improving scan efficiency for the
comprehensive assessment of patients with known or suspected CAD.159a

Coronary Artery Wall Imaging


As with coronary artery lumen MRI, the first in vivo MRI demonstrating
the coronary vessel wall were obtained using 2D fat-saturated fast-spin
echo techniques.139,160 A double inversion recovery (IR) preparation
pulse was applied to obtain black-blood images and improved contrast
RCA
between the blood and the vessel wall.161 Further developments of the RCA wall
technique include the combination of the double IR preparation with
fast-gradient echo readout techniques,162 including spiral163 and radial140 A CMRA B In-plane vessel wall
acquisition trajectories. Examples of in-plane and cross-sectional coro-
FIG. 23.10  (A) Coronary artery magnetic resonance image and (B) three-
nary vessel wall images with double IR preparation and radial and dimensional (3D) in-plane coronary vessel wall imaging using a local
spiral acquisitions are shown in Fig. 23.10 and Fig. 23.11, respectively. inversion preparation pulse and a 3D stack-of-stars (radial) scan with an
Clinical studies demonstrated outward positive remodelling with lumen in-plane spatial resolution of 0.9 mm × 0.9 mm and a slice thickness of
preservation in patients with established CAD and increased vessel wall 2.4 mm. CMRA, Coronary magnetic resonance angiography; RCA, right
thickness in patients with type 1 diabetes and renal dysfunction.18,164 coronary artery.

Slice 2 Slice 3

RCA

Slice 4 Slice 5 Slice 6

FIG. 23.11  Three-dimensional (3D) cross-sectional coronary vessel wall imaging performed at 3 T using a
local inversion recovery preparation pulse and a 3D stack-of-spirals scan with in-plane spatial resolution of
0.6 mm × 0.6 mm and a slice thickness of 3 mm. To acquire cross-sectional slices of the vessel wall, the
3D imaging volume (6 slices) was positioned perpendicular to a relatively linear portion of the proximal/mid
right coronary artery (RCA). (Modified from Peel SA, Hussain T, Schaeffter T, et al. Cross-sectional and in-
plane coronary vessel wall imaging using a local inversion prepulse and spiral read-out: a comparison between
1.5 and 3 Tesla. J Magn Reson Imaging. 2012;35:969–975.)
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 299

CS
CS

PostV LatV

RCA
RCA PostV
A B

CS
CS LatV
LatV

PostV
RCA PostV
C D
FIG. 23.12  (A–D) Example coronary vein cardiovascular magnetic resonance acquired using magnetization
transfer contrast gradient recalled echo during the systolic rest period, depicting the variations in the coronary
venous anatomy in four healthy adult subjects. Clear variations in the branching point, angle, and diameter
of different tributaries of coronary sinus are observed, highlighting the potential for noninvasive assessment
of the coronary venous anatomy in cardiac resynchronization therapy. For example, subject A has no visible
lateral vein (LatV). CS, Compressed sensing; PostV, posterior vein; RCA, right coronary artery.

a tortuous course, complicated scan planning is required. A 3D flow branches adjacent to an appropriate LV wall and the great variabil-
independent approach for vessel wall imaging was proposed based on ity in coronary vein anatomy.175 Ideally coronary venous morphology
an interleaved acquisition and subtraction of bSSFP data with and should be assessed noninvasively before CRT procedure to determine
without a T2 preparation pulse.165 This allows for nulling of arterial whether epicardial or transvenous lead placement would be more
blood signal while maintaining myocardial and vessel wall signal. appropriate.
Although this approach provides simultaneously coronary lumen and The technical challenges of coronary vein CMR are similar to coro-
vessel wall images, the required subtraction is particularly sensitive to nary artery MRI, and techniques developed for coronary artery MRI
respiratory motion corruption. Further recent improvements include are widely applicable. Notable differences in coronary vein CMR include
the combination of this technique with an image-navigator based non- the MTC methods and optimal time window for imaging within the
rigid motion correction approach.166 cardiac cycle, as well as more modest spatial resolution requirements
because information regarding vein anatomy and vessel size are desired,
Coronary Vein Cardiovascular Magnetic Resonance but not focal stenoses. MTC has been used in coronary vein imaging,
For several interventional cardiac procedures, including epicardial radio- which is different than the T2 magnetization preparation commonly
frequency ablation,167,168 retrograde perfusion therapy in high-risk or used in coronary artery MRI, for both targeted109 and whole-heart177
complicated coronary angioplasty,169 arrhythmia assessment,170,171 stem approaches. Fig. 23.12 shows an example of coronary vein CMR using
cell delivery,172 coronary artery bypass surgery,173 and cardiac resyn- a targeted approach with a MTC sequence. Contrast in coronary vein
chronization therapy (CRT),174,175 there has been increased interest in CMR can be improved by other means, including the use of intravascular
imaging the coronary vein anatomy. In CRT, simultaneous pacing of contrast agents such as gadocoletic acid trisodium salt178 or the use of
the right ventricle and left ventricle (LV), or pacing the LV alone, results high relaxivity extracellular contrast agents such as Gd-BOPTA.125 For
in hemodynamic improvement and restoration of a more physiologi- the optimal window of imaging, whereas coronary artery MRI is com-
cal contraction pattern.176 One of the technical difficulties of CRTs is monly performed during the mid diastolic quiescent period, coronary
achieving effective, safe, and permanent pacing of the LV. Transve- vein CMR is acquired in the end-systolic quiescent period as it coincides
nous coronary sinus pacing is the most common technique because with the maximum size of the coronary veins.109
it has the least procedural risk, but it is associated with long proce-
dure times, extensive radiation exposure from fluoroscopy, implanta-
tion failure, and LV lead dislodgment. Two of the major difficulties
REFERENCES
of the transvenous approach are the small number of coronary vein A full reference list is available online at ExpertConsult.com
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 299.e1

REFERENCES acute myocardial infarction. JACC Cardiovasc Imaging.


2009;2(5):580–588.
1. Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart disease and stroke 18. Kim WY, Astrup AS, Stuber M, et al. Subclinical coronary and aortic
statistics—2017 update: a report from the American Heart Association. atherosclerosis detected by magnetic resonance imaging in type 1
Circulation. 2017;135(10):e146–e603. diabetes with and without diabetic nephropathy. Circulation.
2. Patel MR, Peterson ED, Dai D, et al. Low diagnostic yield of elective 2007;115(2):228–235.
coronary angiography. N Engl J Med. 2010;362(10):886–895. 19. Yeon SB, Sabir A, Clouse M, et al. Delayed-enhancement cardiovascular
3. Johnson LW, Lozner EC, Johnson S, et al. Coronary arteriography magnetic resonance coronary artery wall imaging: comparison with
1984-1987: a report of the Registry of the Society for Cardiac multislice computed tomography and quantitative coronary
Angiography and Interventions. I. Results and complications. Cathet angiography. J Am Coll Cardiol. 2007;50(5):441–447.
Cardiovasc Diagn. 1989;17(1):5–10. 20. Jansen CH, Perera D, Makowski MR, et al. Detection of intracoronary
4. Davidson CJ, Mark DB, Pieper KS, et al. Thrombotic and cardiovascular thrombus by magnetic resonance imaging in patients with acute
complications related to nonionic contrast media during cardiac myocardial infarction. Circulation. 2011;124(4):416–424.
catheterization: analysis of 8,517 patients. Am J Cardiol. 1990;65(22): 21. Noguchi T, Kawasaki T, Tanaka A, et al. High-intensity signals in
1481–1484. coronary plaques on noncontrast T1-weighted magnetic resonance
5. Bandettini WP, Arai AE. Advances in clinical applications of imaging as a novel determinant of coronary events. J Am Coll Cardiol.
cardiovascular magnetic resonance imaging. Heart. 2014;63(10):989–999.
2008;94(11):1485–1495. 22. Manning WJ, Edelman RR. Magnetic resonance coronary angiography.
6. Gore TB, Rollings RC, Gore AW 3rd. The many facets of cardiovascular Magn Reson Q. 1993;9(3):131–151.
magnetic resonance imaging: review of background, clinical utility, and 23. Manning WJ, Li W, Edelman RR. A preliminary report comparing
increasing use in the community hospital. South Med J. magnetic resonance coronary angiography with conventional
2009;102(7):719–724. angiography. NEJM. 1993;328(12):828.
7. Lockie T, Nagel E, Redwood S, Plein S. Use of cardiovascular magnetic 24. Kim WY, Danias PG, Stuber M, et al. Coronary magnetic resonance
resonance imaging in acute coronary syndromes. Circulation. 2009; angiography for the detection of coronary stenoses. N Engl J Med.
119(12):1671–1681. 2001;345(26):1863–1869.
8. Fratz S, Chung T, Greil GF, et al. Guidelines and protocols for 25. Stuber M, Botnar RM, Danias PG, Kissinger KV, Manning WJ.
cardiovascular magnetic resonance in children and adults with Breathhold three-dimensional coronary magnetic resonance
congenital heart disease: SCMR expert consensus group on congenital angiography using real-time navigator technology. J Cardiovasc Magn
heart disease. J Cardiovasc Magn Reson. 2013;15:51. Reson. 1999;1(3):233–238.
9. Ferreira VM, Piechnik SK, Robson MD, Neubauer S, Karamitsos TD. 26. Stuber M, Botnar RM, Danias PG, Kissinger KV, Manning WJ.
Myocardial tissue characterization by magnetic resonance imaging: Submillimeter three-dimensional coronary MR angiography with
novel applications of T1 and T2 mapping. J Thorac Imaging. 2014; real-time navigator correction: comparison of navigator locations.
29(3):147–154. Radiology. 1999;212(2):579–587.
10. Salerno M, Kramer CM. Advances in parametric mapping with CMR 27. Stuber M, Botnar RM, Danias PG, et al. Double-oblique free-breathing
imaging. JACC Cardiovasc Imaging. 2013;6(7):806–822. high resolution three-dimensional coronary magnetic resonance
11. McConnell MV, Ganz P, Selwyn AP, Li W, Edelman RR, Manning WJ. angiography. J Am Coll Cardiol. 1999;34(2):524–531.
Identification of anomalous coronary arteries and their anatomic course 28. Botnar RM, Stuber M, Danias PG, Kissinger KV, Manning WJ. Improved
by magnetic resonance coronary angiography. Circulation. 1995;92: coronary artery definition with T2-weighted, free-breathing, three-
3158–3162. dimensional coronary MRA. Circulation. 1999;99(24):3139–3148.
12. Greil GF, Stuber M, Botnar RM, et al. Coronary magnetic resonance 29. Bhat H, Zuehlsdorff S, Bi X, Li D. Whole-heart contrast-enhanced
angiography in adolescents and young adults with Kawasaki disease. coronary magnetic resonance angiography using gradient echo
Circulation. 2002;105:908–911. interleaved EPI. Magn Reson Med. 2009;61(6):1388–1395.
13. Hendel RC, Patel MR, Kramer CM, et al. ACCF/ACR/SCCT/SCMR/ 30. Bi X, Carr JC, Li D. Whole-heart coronary magnetic resonance
ASNC/NASCI/SCAI/SIR 2006 appropriateness criteria for cardiac angiography at 3 Tesla in 5 minutes with slow infusion of Gd-BOPTA,
computed tomography and cardiac magnetic resonance imaging: a a high-relaxivity clinical contrast agent. Magn Reson Med. 2007;58(1):
report of the American College of Cardiology Foundation Quality 1–7.
Strategic Directions Committee Appropriateness Criteria Working 31. Bi X, Deshpande V, Carr J, Li D. Coronary artery magnetic resonance
Group, American College of Radiology, Society of Cardiovascular angiography (MRA): a comparison between the whole-heart and
Computed Tomography, Society for Cardiovascular Magnetic volume-targeted methods using a T2-prepared SSFP sequence.
Resonance, American Society of Nuclear Cardiology, North American J Cardiovasc Magn Reson. 2006;8(5):703–707.
Society for Cardiac Imaging, Society for Cardiovascular Angiography 32. Kim YJ, Seo JS, Choi BW, Choe KO, Jang Y, Ko YG. Feasibility and
and Interventions, and Society of Interventional Radiology. J Am Coll diagnostic accuracy of whole heart coronary MR angiography using
Cardiol. 2006;48(7):1475–1497. free-breathing 3D balanced turbo-field-echo with SENSE and the
14. Hundley WG, Bluemke DA, Finn JP, et al. ACCF/ACR/AHA/NASCI/ half-fourier acquisition technique. Korean J Radiol. 2006;7(4):235–242.
SCMR 2010 Expert Consensus Document on Cardiovascular Magnetic 33. Lai P, Bi X, Jerecic R, Li D. A respiratory self-gating technique with
Resonance. A Report of the American College of Cardiology Foundation 3D-translation compensation for free-breathing whole-heart coronary
Task Force on Expert Consensus Documents. Circulation. 2010;121(22): MRA. Magn Reson Med. 2009.
2462–2508. 34. Lai P, Larson AC, Bi X, Jerecic R, Li D. A dual-projection respiratory
15. Kato S, Kitagawa K, Ishida N, et al. Assessment of coronary artery self-gating technique for whole-heart coronary MRA. J Magn Reson
disease using magnetic resonance coronary angiography: a national Imaging. 2008;28(3):612–620.
multicenter trial. J Am Coll Cardiol. 2010;56(12):983–991. 35. Liu X, Bi X, Huang J, Jerecic R, Carr J, Li D. Contrast-enhanced
16. Yoon YE, Kitagawa K, Kato S, et al. Prognostic value of coronary whole-heart coronary magnetic resonance angiography at 3.0 T:
magnetic resonance angiography for prediction of cardiac events in comparison with steady-state free precession technique at 1.5 T. Invest
patients with suspected coronary artery disease. J Am Coll Cardiol. Radiol. 2008;43(9):663–668.
2012;60(22):2316–2322. 36. Maintz D, Ozgun M, Hoffmeier A, et al. Whole-heart coronary magnetic
17. Ibrahim T, Makowski MR, Jankauskas A, et al. Serial contrast-enhanced resonance angiography: value for the detection of coronary artery
cardiac magnetic resonance imaging demonstrates regression of stenoses in comparison to multislice computed tomography
hyperenhancement within the coronary artery wall in patients after angiography. Acta Radiol. 2007;48(9):967–973.
299.e2 SECTION II  Ischemic Heart Disease

37. Nehrke K, Bornert P, Mazurkewitz P, Winkelmann R, Grasslin I. 58. Plein S, Jones TR, Ridgway JP, Sivananthan MU. Three-dimensional
Free-breathing whole-heart coronary MR angiography on a clinical coronary MR angiography performed with subject-specific cardiac
scanner in four minutes. J Magn Reson Imaging. 2006;23(5):752–756. acquisition windows and motion-adapted respiratory gating. AJR Am J
38. Niendorf T, Hardy CJ, Giaquinto RO, et al. Toward single breath-hold Roentgenol. 2003;180(2):505–512.
whole-heart coverage coronary MRA using highly accelerated parallel 59. Tangcharoen T, Jahnke C, Koehler U, et al. Impact of heart rate variability
imaging with a 32-channel MR system. Magn Reson Med. in patients with normal sinus rhythm on image quality in coronary
2006;56(1):167–176. magnetic angiography. J Magn Reson Imaging. 2008;28(1):74–79.
39. Okada T, Kanao S, Ninomiya A, et al. Whole-heart coronary magnetic 60. Wang Y, Riederer SJ, Ehman RL. Respiratory motion of the heart:
resonance angiography with parallel imaging: Comparison of kinematics and the implications for the spatial resolution in coronary
acceleration in one-dimension vs. two-dimensions. Eur J Radiol. 2008. imaging. Magn Reson Med. 1995;33(5):713–719.
40. Sakuma H, Ichikawa Y, Chino S, Hirano T, Makino K, Takeda K. 61. Edelman RR, Manning WJ, Burstein D, Paulin S. Coronary arteries:
Detection of coronary artery stenosis with whole-heart coronary breath-hold MR angiography. Radiology. 1991;181(3):641–643.
magnetic resonance angiography. J Am Coll Cardiol. 2006;48(10): 62. Taylor AM, Jhooti P, Wiesmann F, Keegan J, Firmin DN, Pennell DJ. MR
1946–1950. navigator-echo monitoring of temporal changes in diaphragm position:
41. Sakuma H, Ichikawa Y, Suzawa N, et al. Assessment of coronary arteries implications for MR coronary angiography. J Magn Reson Imaging.
with total study time of less than 30 minutes by using whole-heart 1997;7(4):629–636.
coronary MR angiography. Radiology. 2005;237(1):316–321. 63. Holland AE, Goldfarb JW, Edelman RR. Diaphragmatic and cardiac
42. Stehning C, Bornert P, Nehrke K, Eggers H, Stuber M. Free-breathing motion during suspended breathing: preliminary experience and
whole-heart coronary MRA with 3D radial SSFP and self-navigated implications for breath-hold MR imaging. Radiology. 1998;209(2):
image reconstruction. Magn Reson Med. 2005;54(2). 483–489.
43. Weber OM, Martin AJ, Higgins CB. Whole-heart steady-state free 64. Danias PG, Stuber M, Botnar RM, Kissinger KV, Chuang ML, Manning
precession coronary artery magnetic resonance angiography. Magn WJ. Navigator assessment of breath-hold duration: impact of
Reson Med. 2003;50(6):1223–1228. supplemental oxygen and hyperventilation. AJR Am J Roentgenol.
44. Deshpande VS, Shea SM, Laub G, Simonetti OP, Finn JP, Li D. 3D 1998;171(2):395–397.
magnetization-prepared true-FISP: a new technique for imaging 65. Ehman RL, Felmlee JP. Adaptive technique for high-definition MR
coronary arteries. Magn Reson Med. 2001;46(3):494–502. imaging of moving structures. Radiology. 1989;173(1):255–263.
45. Nezafat R, Herzka D, Stehning C, Peters DC, Nehrke K, Manning WJ. 66. Oshinski JN, Hofland L, Mukundan S Jr, Dixon WT, Parks WJ, Pettigrew
Inflow quantification in three-dimensional cardiovascular MR imaging. RI. Two-dimensional coronary MR angiography without breath holding.
J Magn Reson Imaging. 2008;28(5):1273–1279. Radiology. 1996;201(3):737–743.
46. Maintz D, Aepfelbacher FC, Kissinger KV, et al. Coronary MR 67. Sachs TS, Meyer CH, Hu BS, Kohli J, Nishimura DG, Macovski A.
angiography: comparison of quantitative and qualitative data from four Real-time motion detection in spiral MRI using navigators. Magn Reson
techniques. AJR Am J Roentgenol. 2004;182(2):515–521. Med. 1994;32(5):639–645.
47. Ozgun M, Hoffmeier A, Kouwenhoven M, et al. Comparison of 3D 68. McConnell MV, Khasgiwala VC, Savord BJ, et al. Comparison of
segmented gradient-echo and steady-state free precession coronary MRI respiratory suppression methods and navigator locations for MR
sequences in patients with coronary artery disease. AJR Am J Roentgenol. coronary angiography. AJR Am J Roentgenol. 1997;168(5):1369–1375.
2005;185(1):103–109. 69. Danias PG, McConnell MV, Khasgiwala VC, Chuang ML, Edelman RR,
48. Polson MJ, Barker AT, Gardiner S. The effect of rapid rise-time magnetic Manning WJ. Prospective navigator correction of image position for
fields on the ECG of the rat. Clin Phys Physiol Meas. 1982;3(3): coronary MR angiography. Radiology. 1997;203(3):733–736.
231–234. 70. Nagel E, Bornstedt A, Schnackenburg B, Hug J, Oswald H, Fleck E.
49. Paulin S. Coronary angiography. A technical, anatomic and clinical Optimization of realtime adaptive navigator correction for 3D magnetic
study. Acta Radiol Diagn (Stockh). 1964;54(suppl 233):L+. resonance coronary angiography. Magn Reson Med. 1999;42(2):408–411.
50. Wang Y, Vidan E, Bergman GW. Cardiac motion of coronary arteries: 71. Huber S, Bornstedt A, Schnackenburg B, Paetsch I, Fleck E, Nagel E. The
variability in the rest period and implications for coronary MR impact of different positions and thoracial restrains on respiratory
angiography. Radiology. 1999;213(3):751–758. induced cardiac motion. J Cardiovasc Magn Reson. 2006;8(3):483–488.
51. Kaji S, Yang PC, Kerr AB, et al. Rapid evaluation of left ventricular 72. McConnell MV, Khasgiwala VC, Savord BJ, et al. Prospective adaptive
volume and mass without breath-holding using real-time interactive navigator correction for breath-hold MR coronary angiography. Magn
cardiac magnetic resonance imaging system. J Am Coll Cardiol. Reson Med. 1997;37(1):148–152.
2001;38(2):527–533. 73. Stuber M, Botnar RM, Danias PG, Kissinger KV, Manning WJ.
52. Hofman MB, Wickline SA, Lorenz CH. Quantification of in-plane Submillimeter three-dimensional coronary MR angiography with
motion of the coronary arteries during the cardiac cycle: implications real-time navigator correction: comparison of navigator locations.
for acquisition window duration for MR flow quantification. J Magn Radiology. 1999;212(2):579–587.
Reson Imaging. 1998;8(3):568–576. 74. Spuentrup E, Stuber M, Botnar RM, Manning WJ. The impact of
53. Kim WY, Stuber M, Kissinger KV, Andersen NT, Manning WJ, Botnar navigator timing parameters and navigator spatial resolution on 3D
RM. Impact of bulk cardiac motion on right coronary MR angiography coronary magnetic resonance angiography. J Magn Reson Imaging.
and vessel wall imaging. J Magn Reson Imaging. 2001;14(4):383–390. 2001;14(3):311–318.
54. Jahnke C, Paetsch I, Achenbach S, et al. Coronary MR imaging: 75. Danias PG, Stuber M, Botnar RM, Kissinger KV, Edelman RR, Manning
breath-hold capability and patterns, coronary artery rest periods, and WJ. Relationship between motion of coronary arteries and diaphragm
beta-blocker use. Radiology. 2006;239(1):71–78. during free breathing: lessons from real-time MR imaging. AJR Am J
55. Jahnke C, Paetsch I, Nehrke K, et al. A new approach for rapid Roentgenol. 1999;172(4):1061–1065.
assessment of the cardiac rest period for coronary MRA. J Cardiovasc 76. Moghari MH, Hu P, Kissinger KV, et al. Subject-specific estimation of
Magn Reson. 2005;7(2):395–399. respiratory navigator tracking factor for free-breathing cardiovascular
56. Leiner T, Katsimaglis G, Yeh EN, et al. Correction for heart rate MR. Magn Reson Med. 2012;67(6):1665–1672.
variability improves coronary magnetic resonance angiography. J Magn 77. McConnell M, Khasgiwala V, Savord B, et al. Comparison of respiratory
Reson Imaging. 2005;22(4):577–582. suppression methods and navigator locations for MR coronary
57. Jahnke CPI, Gebker R, Schnackenburg B, et al. Comparison of angiography. AJR Am J Roentgenol. 1997;168(5):1369.
individually adapted breath-hold and free-breathing coronary MRA 78. Nezafat R, Stehning C, Gharib AM, et al. Improved Spatial-Temporal
using steady state free precession [abstr]. J Cardiovasc Magn Reson. Resolution MR Coronary Blood Flow Imaging at 3T. J Cardiovasc Magn
2004;6:166–167. Reson. 2005;7(1):199.
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 299.e3

79. Nehrke K, Bornert P. Prospective correction of affine motion for arbitrary 101. Moghari MH, Annese D, Geva T, Powell AJ. Three-dimensional heart
MR sequences on a clinical scanner. Magn Reson Med. 2005;54(5). locator and compressed sensing for whole-heart MR angiography. Magn
80. Lai P, Larson AC, Park J, Carr JC, Li D. Respiratory self-gated four- Reson Med. 2016;75(5):2086–2093.
dimensional coronary MR angiography: a feasibility study. Magn Reson 102. Moghari MH, Roujol S, Henningsson M, et al. Three-dimensional heart
Med. 2008;59(6):1378–1385. locator for whole-heart coronary magnetic resonance angiography.
81. Larson AC, Kellman P, Arai A, et al. Preliminary investigation of Magn Reson Med. 2014;71(6):2118–2126.
respiratory self-gating for free-breathing segmented cine MRI. Magn 103. Wu HH, Gurney PT, Hu BS, Nishimura DG, McConnell MV. Free-
Reson Med. 2005;53(1). breathing multiphase whole-heart coronary MR angiography using
82. Park J, Larson AC, Zhang Q, Simonetti O, Li D. 4D radial coronary image-based navigators and three-dimensional cones imaging. Magn
artery imaging within a single breath-hold: cine angiography with Reson Med. 2013;69(4):1083–1093.
phase-sensitive fat suppression (CAPS). Magn Reson Med. 104. Li D, Paschal CB, Haacke EM, Adler LP. Coronary arteries: three-
2005;54(4):833–840. dimensional MR imaging with fat saturation and magnetization transfer
83. Keegan J, Gatehouse P, Yang G, Firmin D. Non-model based correction contrast. Radiology. 1993;187(2):401–406.
of respiratory motion using beat-to-beat 3D spiral fat-selective imaging. 105. Brittain JH, Hu BS, Wright GA, Meyer CH, Macovski A, Nishimura DG.
J Magn Reson Imaging. 2006;367. Coronary angiography with magnetization-prepared T2 contrast. Magn
84. Nguyen T, Spincemaille P, Cham M, Weinsaft J, Prince M, Wang Y. Reson Med. 1995;33(5):689–696.
Free-breathing 3D steady-state free precession coronary magnetic 106. Shea SM, Deshpande VS, Chung YC, Li D. Three-dimensional true-FISP
resonance angiography: Comparison of diaphragm and cardiac fat imaging of the coronary arteries: improved contrast with
navigators. J Magn Reson Imaging. 2008;28(2):509. T2-preparation. J Magn Reson Imaging. 2002;15(5):597–602.
85. Nguyen T, Spincemaille P, Prince M, Wang Y. 2128 Free-breathing 107. Balaban RS, Ceckler TL. Magnetization transfer contrast in magnetic
steady-state free precession 3D coronary MRA: comparison of resonance imaging. Magn Reson Q. 1992;8(2):116–137.
diaphragm and cardiac fat navigator techniques. J Cardiovasc Magn 108. Nezafat R, Stuber M, Ouwerkerk R, Gharib AM, Desai MY, Pettigrew RI.
Reson. 2008;10(suppl 1):A397. B1-insensitive T2 preparation for improved coronary magnetic
86. Nguyen T, Spincemaille P, Cham M, Weinsaft J, Prince M, Wang Y. resonance angiography at 3 T. Magn Reson Med. 2006;55(4):858–864.
Free-breathing 3-dimensional steady-state free precession coronary 109. Nezafat R, Han Y, Peters DC, et al. Coronary magnetic resonance vein
magnetic resonance angiography: comparison of four navigator gating imaging: imaging contrast, sequence, and timing. Magn Reson Med.
techniques. Magn Reson Imaging. 2009;27(6):807–814. 2007;58(6):1196–1206.
87. Moghari MH, Chan RH, Hong SN, et al. Free-breathing cardiac MR 110. Stuber M, Botnar RM, Fischer SE, et al. Preliminary report on in vivo
with a fixed navigator efficiency using adaptive gating window size. coronary MRA at 3 Tesla in humans. Magn Reson Med.
Magn Reson Med. 2012;68(6):1866–1875. 2002;48(3):425–429.
88. Gurney PT, Hargreaves BA, Nishimura DG. Design and analysis of a 111. Schar M, Kozerke S, Fischer Se Boesiger P. Cardiac SSFP imaging at 3
practical 3D cones trajectory. Magn Reson Med. 2006;55(3):575–582. Tesla. Magn Reson Med. 2004;51(4):799–806.
89. Santos JM, Cunningham CH, Lustig M, et al. Single breath-hold 112. Nezafat R, Ouwerkerk R, Derbyshire AJ, Stuber M, McVeigh ER.
whole-heart MRA using variable-density spirals at 3T. Magn Reson Med. Spectrally selective B1-insensitive T2 magnetization preparation
2006;55(2):371–379. sequence. Magn Reson Med. 2009;61(6):1326–1335.
90. Sussman M, Stainsby J, Robert N, Merchant N, Wright G. Variable- 113. Terashima M, Meyer CH, Keeffe BG, et al. Noninvasive assessment of
density adaptive imaging for high-resolution coronary artery MRI. coronary vasodilation using magnetic resonance angiography. J Am Coll
Magn Reson Med. 2002;48(5):753–764. Cardiol. 2005;45(1):104–110.
91. Buehrer M, Curcic J, Boesiger P, Kozerke S. Prospective self-gating for 114. Hu P, Chuang ML, Ngo LH, et al. Coronary MR imaging: effect of
simultaneous compensation of cardiac and respiratory motion. Magn timing and dose of isosorbide dinitrate administration. Radiology.
Reson Med. 2008;60(3):683–690. 2010;254(2):401–409.
92. Kim WS, Mun CW, Kim DJ, Cho ZH. Extraction of cardiac and 115. Zheng J, Bae KT, Woodard PK, Haacke EM, Li D. Efficacy of slow
respiratory motion cycles by use of projection data and its applications infusion of gadolinium contrast agent in three-dimensional MR
to NMR imaging. Magn Reson Med. 1990;13(1):25–37. coronary artery imaging. J Magn Reson Imaging. 1999;10(5):800–805.
93. Lai P, Bi X, Jerecic R, Li D. A respiratory self-gating technique with 116. Goldfarb JW, Edelman RR. Coronary arteries: breath-hold, gadolinium-
3D-translation compensation for free-breathing whole-heart coronary enhanced, three-dimensional MR angiography. Radiology.
MRA. Magn Reson Med. 2009;62(3):731–738. 1998;206(3):830–834.
94. Piccini D, Littmann A, Nielles-Vallespin S, Zenge MO. Respiratory 117. Knuesel PR, Nanz D, Wolfensberger U, et al. Multislice breath-hold
self-navigation for whole-heart bright-blood coronary MRI: methods spiral magnetic resonance coronary angiography in patients with
for robust isolation and automatic segmentation of the blood pool. coronary artery disease: effect of intravascular contrast medium. J Magn
Magn Reson Med. 2012;68(2):571–579. Reson Imaging. 2002;16(6):660–667.
95. Stehning C, Bornert P, Nehrke K, Eggers H, Stuber M. Free-breathing 118. Herborn CU, Barkhausen J, Paetsch I, et al. Coronary arteries:
whole-heart coronary MRA with 3D radial SSFP and self-navigated contrast-enhanced MR imaging with SH L 643A—experience in 12
image reconstruction. Magn Reson Med. 2005;54(2):476–480. volunteers. Radiology. 2003;229(1):217–223.
96. Addy NO, Ingle RR, Luo J, et al. 3D image-based navigators for coronary 119. Prompona M, Cyran C, Nikolaou K, Bauner K, Reiser M, Huber A.
MR angiography. Magn Reson Med. 2016. Contrast-enhanced whole-heart MR coronary angiography at 3.0 T
97. Henningsson M, Koken P, Stehning C, Razavi R, Prieto C, Botnar RM. using the intravascular contrast agent gadofosveset. Invest Radiol.
Whole-heart coronary MR angiography with 2D self-navigated image 2009;44(7):369–374.
reconstruction. Magn Reson Med. 2012;67(2):437–445. 120. Kelle S, Thouet T, Tangcharoen T, et al. Whole-heart coronary magnetic
98. Henningsson M, Smink J, Razavi R, Botnar RM. Prospective respiratory resonance angiography with MS-325 (Gadofosveset). Med Sci Monit.
motion correction for coronary MR angiography using a 2D image 2007;13(11):CR469–CR474.
navigator. Magn Reson Med. 2013;69(2):486–494. 121. de Haen C, Anelli PL, Lorusso V, et al. Gadocoletic acid trisodium
99. Kawaji K, Spincemaille P, Nguyen TD, et al. Direct coronary motion salt (b22956/1): a new blood pool magnetic resonance contrast agent
extraction from a 2D fat image navigator for prospectively gated with application in coronary angiography. Invest Radiol. 2006;41(3):
coronary MR angiography. Magn Reson Med. 2014;71(2):599–607. 279–291.
100. Luo J, Addy NO, Ingle RR, et al. Nonrigid motion correction with 3D 122. Tang L, Merkle N, Schar M, et al. Volume-targeted and whole-heart
image-based navigators for coronary MR angiography. Magn Reson Med. coronary magnetic resonance angiography using an intravascular
2017;77(5):1884–1893. contrast agent. J Magn Reson Imaging. 2009;30(5):1191–1196.
299.e4 SECTION II  Ischemic Heart Disease

123. Weber OM, Martin AJ, Higgins CB. Whole-heart steady-state free 32-channel cardiac coils: initial single-center experience. Radiology.
precession coronary artery magnetic resonance angiography. Magn 2011;259(2):384–392.
Reson Med. 2003;50(6):1223–1228. 144. Niendorf T, Hardy CJ, Giaquinto RO, et al. Toward single breath-hold
124. Yang Q, Li K, Liu X, et al. Contrast-enhanced whole-heart coronary whole-heart coverage coronary MRA using highly accelerated parallel
magnetic resonance angiography at 3.0-T: a comparative study with imaging with a 32-channel MR system. Magn Reson Med. 2006;56(1):
X-ray angiography in a single center. J Am Coll Cardiol. 2009; 167–176.
54(1):69–76. 145. Bhat H, Yang Q, Zuehlsdorff S, Li K, Li D. Contrast-enhanced
125. Hu P, Chan J, Ngo LH, et al. Contrast-enhanced whole-heart coronary whole-heart coronary magnetic resonance angiography at 3 T with
MRI with bolus infusion of gadobenate dimeglumine at 1.5 T. Magn radial EPI. Magn Reson Med. 2011;66(1):82–91.
Reson Med. 2011;65(2):392–398. 146. Block KT, Uecker M, Frahm J. Undersampled radial MRI with multiple
126. Bunce NH, Jhooti P, Keegan J, et al. Evaluation of free-breathing coils. Iterative image reconstruction using a total variation constraint.
three-dimensional magnetic resonance coronary angiography with Magn Reson Med. 2007;57(6):1086–1098.
hybrid ordered phase encoding (HOPE) for the detection of proximal 147. Lustig M, Donoho DL, Pauly JM. Sparse MRI: The application of
coronary artery stenosis. J Magn Reson Imaging. 2001;14(6):677–684. compressed sensing for rapid MR imaging. Magn Reson Med.
127. Sommer T, Hofer U, Hackenbroch M, et al. Submillimeter 3D coronary 2007;58(6):1182–1195.
MR angiography with real-time navigator correction in 107 patients 148. Akcakaya M, Basha TA, Goddu B, et al. Low-dimensional-structure
with suspected coronary artery disease. Rofo. 2002;174(4):459–466. self-learning and thresholding: Regularization beyond compressed
128. Bogaert J, Kuzo R, Dymarkowski S, Beckers R, Piessens J, Rademakers sensing for MRI Reconstruction. Magn Reson Med. 2011;66(3):
FE. Coronary artery imaging with real-time navigator three-dimensional 756–767.
turbo-field-echo MR coronary angiography: initial experience. 149. Akcakaya M, Basha TA, Chan RH, et al. Accelerated contrast-enhanced
Radiology. 2003;226(3):707–716. whole-heart coronary MRI using low-dimensional-structure self-
129. Jahnke C, Paetsch I, Schnackenburg B, et al. Coronary MR angiography learning and thresholding. Magn Reson Med. 2012;67(5):1434–1443.
with steady-state free precession: individually adapted breath-hold 150. Akcakaya M, Basha TA, Chan RH, Manning WJ, Nezafat R. Accelerated
technique versus free-breathing technique. Radiology. 2004;232(3): isotropic sub-millimeter whole-heart coronary MRI: Compressed
669–676. sensing versus parallel imaging. Magn Reson Med. 2014;71(2):815–822.
130. Pouleur AC, le Polain de Waroux JB, Kefer J, Pasquet A, Vanoverschelde 151. Nam S, Akçakaya M, Basha TA, et al. Compressed sensing reconstruction
JL, Gerber BL. Direct comparison of whole-heart navigator-gated for whole-heart imaging with 3D radial trajectories: A graphics
magnetic resonance coronary angiography and 40- and 64-slice processing unit implementation. Magn Reson Med. 2013;69(1):91–102.
multidetector row computed tomography to detect the coronary artery 152. Wen H, Denison TJ, Singerman RW, Balaban RS. The intrinsic
stenosis in patients scheduled for conventional coronary angiography. signal-to-noise ratio in human cardiac imaging at 1.5, 3, and 4 T.
Circ Cardiovasc Imaging. 2008;1(2):114–121. J Magn Reson. 1997;125(1):65–71.
131. Yang Q, Li K, Sun B, et al. Assessment of coronary artery disease using 153. Noeske R, Seifert F, Rhein KH, Rinneberg H. Human cardiac imaging at
3.0 T magnetic resonance coronary angiography: a national multicenter 3 T using phased array coils. Magn Reson Med. 2000;44(6):978–982.
trial. J Cardiovasc Magn Reson. 2013;15(suppl 1):E5. 154. Atalay MK, Poncelet BP, Kantor HL, Brady TJ, Weisskoff RM. Cardiac
132. Meyer CH, Hu BS, Nishimura DG, Macovski A. Fast spiral coronary susceptibility artifacts arising from the heart-lung interface. Magn Reson
artery imaging. Magn Reson Med. 1992;28(2):202–213. Med. 2001;45(2):341–345.
133. Bornert P, Stuber M, Botnar RM, et al. Direct comparison of 3D spiral 155. Bi X, Deshpande V, Simonetti O, Laub G, Li D. Three-dimensional
vs. Cartesian gradient-echo coronary magnetic resonance angiography. breathhold SSFP coronary MRA: a comparison between 1.5T and 3.0T.
Magn Reson Med. 2001;46(4):789–794. J Magn Reson Imaging. 2005;22(2):206–212.
134. Thedens DR, Irarrazaval P, Sachs TS, Meyer CH, Nishimura DG. Fast 156. Stuber M, Botnar RM, Fischer SE, et al. Preliminary report on in vivo
magnetic resonance coronary angiography with a three-dimensional coronary MRA at 3 Tesla in humans. Magn Reson Med.
stack of spirals trajectory. Magn Reson Med. 1999;41(6):1170– 2002;48(3):425–429.
1179. 157. Steen HLJ, Giannitsis E, Katus HA, Stuber M. Comparison of high field
135. Bornert P, Aldefeld B, Nehrke K. Improved 3D spiral imaging for magnetic resonance imaging at 1.5T and 3T for coronary artery MRA
coronary MR angiography. Magn Reson Med. 2001;45(1):172–175. [abstr]. J Cardiovasc Magn Reson. 2004;6:342–344.
136. Leiner TYE, Katsimaglis G, Kissinger KV, et al. Comparison of Cartesian 158. Bi XDV, Simonetti O, Laub G, Li D. Three-dimensional breath-hold
and Radial Balanced GTFE coronary MRA [abstr]. J Cardiovasc Magn coronary MRA: a comparison between 1.5T and 3.0T [abstr].
Reson. 2004;6:75. J Cardiovasc Magn Reson. 2004;6:335–336.
137. Weber OM, Pujadas S, Martin AJ, Higgins CB. Free-breathing, 159. van Elderen SG, Versluis MJ, Webb AG, et al. Initial results on in vivo
three-dimensional coronary artery magnetic resonance angiography: human coronary MR angiography at 7 T. Magn Reson Med.
comparison of sequences. J Magn Reson Imaging. 2004;20(3):395–402. 2009;62(6):1379–1384.
138. Nam S, Akcakaya M, Basha TA, et al. Compressed sensing reconstruction 159a.  Ginami G, Neji R, Rashid I, et al. 3D whole-heart phase sensitive
for whole heart imaging with 3D radial trajectories: a GPU inversion recovery CMR for simultaneous black-blood late gadolinium
implementation. Magn Reson Med. 2013;69(1):91–102. enhancement and bright-blood coronary CMR angiography.
139. Botnar RM, Stuber M, Kissinger KV, Kim WY, Spuentrup E, Manning J Cardiovasc Magn Reson. 2017;19:94.
WJ. Noninvasive coronary vessel wall and plaque imaging with magnetic 160. Fayad ZA, Fuster V, Fallon JT, et al. Noninvasive in vivo human coronary
resonance imaging. Circulation. 2000;102(21):2582–2587. artery lumen and wall imaging using black-blood magnetic resonance
140. Katoh M, Spuentrup E, Buecker A, Manning WJ, Gunther RW, Botnar imaging. Circulation. 2000;102(5):506–510.
RM. MR coronary vessel wall imaging: comparison between radial and 161. Edelman RR, Chien D, Kim D. Fast selective black blood MR imaging.
spiral k-space sampling. J Magn Reson Imaging. 2006;23(5):757–762. Radiology. 1991;181(3):655–660.
141. Griswold MA, Jakob PM, Heidemann RM, et al. Generalized 162. Botnar RM, Stuber M, Lamerichs R, et al. Initial experiences with in
autocalibrating partially parallel acquisitions (GRAPPA). Magn Reson vivo right coronary artery human MR vessel wall imaging at 3 tesla.
Med. 2002;47(6):1202–1210. J Cardiovasc Magn Reson. 2003;5(4):589–594.
142. Pruessmann KP, Weiger M, Bornert P, Boesiger P. Advances in sensitivity 163. Botnar RM, Kim WY, Bornert P, Stuber M, Spuentrup E, Manning WJ.
encoding with arbitrary k-space trajectories. Magn Reson Med. 2001; 3D coronary vessel wall imaging utilizing a local inversion technique
46(4):638–651. with spiral image acquisition. Magn Reson Med. 2001;46(5):848–854.
143. Nagata M, Kato S, Kitagawa K, et al. Diagnostic accuracy of 1.5-T 164. Kim WY, Stuber M, Bornert P, Kissinger KV, Manning WJ, Botnar RM.
unenhanced whole-heart coronary MR angiography performed with Three-dimensional black-blood cardiac magnetic resonance coronary
CHAPTER 23  Magnetic Resonance Imaging of Coronary Arteries: Technique 299.e5

vessel wall imaging detects positive arterial remodeling in patients with 172. Thompson CA, Nasseri BA, Makower J, et al. Percutaneous transvenous
nonsignificant coronary artery disease. Circulation. 2002;106(3):296–299. cellular cardiomyoplasty. A novel nonsurgical approach for myocardial
165. Andia ME, Henningsson M, Hussain T, et al. Flow-independent 3D cell transplantation. J Am Coll Cardiol. 2003;41(11):1964–1971.
whole-heart vessel wall imaging using an interleaved T2-preparation 173. Oesterle SN, Reifart N, Hauptmann E, Hayase M, Yeung AC.
acquisition. Magn Reson Med. 2013;69(1):150–157. Percutaneous in situ coronary venous arterialization: report of the first
166. Cruz G, Atkinson D, Henningsson M, Botnar RM, Prieto C. Highly human catheter-based coronary artery bypass. Circulation. 2001;103(21):
efficient nonrigid motion-corrected 3D whole-heart coronary vessel wall 2539–2543.
imaging. Magn Reson Med. 2016. 174. de Paola AA, Melo WD, Tavora MZ, Martinez EE. Angiographic and
167. Kusano KF, Morita H, Fujimoto Y, Hirose E, Ohe T. Catheter ablation of electrophysiological substrates for ventricular tachycardia mapping
an epicardial accessory pathway via the middle cardiac vein guided by through the coronary veins 1. Heart. 1998;79(1):59–63.
monophasic action potential recordings. Europace. 2001;3(2):164–167. 175. Singh JP, Houser S, Heist EK, Ruskin JN. The coronary venous anatomy:
168. Haissaguerre M, Gaita F, Fischer B, Egloff P, Lemetayer P, Warin JF. a segmental approach to aid cardiac resynchronization therapy. J Am
Radiofrequency catheter ablation of left lateral accessory pathways via Coll Cardiol. 2005;46(1):68–74.
the coronary sinus. Circulation. 1992;86(5):1464–1468. 176. Abraham WT, Fisher WG, Smith AL, et al. Cardiac resynchronization in
169. Kar S, Nordlander R. Coronary veins: an alternate route to ischemic chronic heart failure. N Engl J Med. 2002;346(24):1845–1853.
myocardium. Heart Lung. 1992;21(2):148–157. 177. Stoeck CT, Han Y, Peters DC, et al. Whole heart magnetization-prepared
170. Stellbrink C, Diem B, Schauerte P, Ziegert K, Hanrath P. Transcoronary steady-state free precession coronary vein MRI. J Magn Reson Imaging.
venous radiofrequency catheter ablation of ventricular tachycardia. 2009;29(6):1293–1299.
J Cardiovasc Electrophysiol. 1997;8(8):916–921. 178. Rasche V, Binner L, Cavagna F, et al. Whole-heart coronary vein
171. Butter C, Meisel E, Engelmann L, et al. Human experience with imaging: a comparison between non-contrast-agent-and contrast-agent-
transvenous biventricular defibrillation using an electrode in a left enhanced visualization of the coronary venous system. Magn Reson Med.
ventricular vein. Pacing Clin Electrophysiol. 2002;25(3):324–331. 2007;57(6):1019–1026.
24 
Coronary Artery Imaging: Clinical Results
Shiro Nakamori, Hajime Sakuma, and Warren J. Manning

Chapter 23 reviewed the technical issues and solutions for coronary segmented k-space gradient echo coronary artery CMR,5–7 although
artery cardiovascular magnetic resonance (CMR) imaging. This chapter most centers now use targeted 3D8–11 or whole heart11–16 free-breathing
reviews the clinical data comparing coronary artery CMR with invasive navigator coronary artery CMR because of the superior reconstruction
x-ray coronary angiography for identification of anomalous coronary capabilities afforded by 3D datasets, with similar results. The ability to
artery disease (CAD), characterization of coronary artery aneurysms, acquire these data using CMR without the use of ionizing radiation is
detection of native vessel disease, and assessment of coronary artery likely to be of particular benefit in the generally younger population.17
bypass graft integrity. It also describes studies comparing CMR with In addition, noncontrast coronary artery CMR is likely preferred to
coronary artery multidetector computed tomography (MDCT), the avoid potential long-term issues related to retention of gadolinium.18
other principal noninvasive modality for imaging the coronary arteries. There have been at least eight published series5–10,12,14,16 of patients
The majority of data represent single-center experience, with quantita- who underwent a comparison of coronary artery CMR at 1.5 T or 3 T
tive x-ray coronary angiography used as the reference standard for most with x-ray angiography for suspected anomalous CAD. These studies
of the larger single-center and few multicenter studies. have uniformly reported excellent accuracy, including several instances
in which coronary artery CMR was determined to be superior to x-ray
IDENTIFICATION OF ANOMALOUS angiography (Table 24.1). Data also suggest coronary artery CMR evi-
dence of a coronary anomaly carries important prognostic information11
CORONARY ARTERIES with the identification of the anomalous segment originating from the
As discussed in Chapter 23, using current whole-heart imaging with opposite sinus of Valsalva (see Fig. 24.1) as conveying an adverse prog-
or without gadolinium contrast, the native proximal coronary arter- nosis. As a result, CMR is considered a class I indication for suspected
ies can be reliably visualized in nearly all subjects. Although unusual anomalous CAD.19 At experienced CMR centers, clinical coronary artery
(<1% of the general population1,2) and most often benign, congenital CMR is the preferred test for patients in whom anomalous disease is
coronary anomalies in which the anomalous segment courses anterior suspected, those with known anomalous disease that must be further
to the aorta and posterior to the pulmonary artery are well-recognized clarified, and those with coronary anomalies associated with other
causes of myocardial ischemia and sudden cardiac death in children cardiac anomalies (e.g., tetralogy of Fallot). Although MDCT has also
and young adults.3,4 These adverse events commonly occur during or been shown to be efficacious for this indication,20–22 coronary artery
immediately after intense exercise and are believed to be related to CMR is often preferred because there is no need for ionizing radiation
compression of the anomalous segment, vessel kinking, or coexistent or intravenous contrast.
eccentric ostial stenoses.3
The ability of coronary artery CMR to reliably identify the major CORONARY ARTERY ANEURYSMS AND
coronary arteries and their relationship to the ascending aorta and
pulmonary artery immediately provides for its application for the iden-
KAWASAKI DISEASE
tification and characterization of anomalous CAD. The spatial resolution Coronary artery aneurysms are relatively uncommon, but have received
requirements for identifying anomalous coronary vessels are less stringent increasing attention because of their common occurrence in pediatric
than for defining native vessel stenoses, allowing for lower resolution and young adult patients with a history of mucocutaneous lymph node
and faster CMR imaging. syndrome (Kawasaki disease), a generalized vasculitis of unknown etiol-
Projection x-ray angiography had traditionally been the imaging ogy usually occurring in children age >5 years. The prevalence of Kawasaki
test of choice for the diagnosis and characterization of these anomalies. disease is highest among children of East Asian countries, with the
However, the presence of an anomalous vessel is sometimes suspected greatest prevalence in Japan.23 Infants and children with this syndrome
only after the procedure, particularly in a situation in which there was may show evidence of myocarditis or pericarditis, with nearly 20%
unsuccessful engagement of a coronary artery. In addition, the uncom- having coronary artery aneurysms. These aneurysms (Fig. 24.2) are the
mon use of a pulmonary artery catheter has made characterization of source of both short-term and long-term morbidity and mortality.24
the anterior or posterior trajectory of the anomalous vessels more dif- Fortunately, one-half of the children with coronary aneurysms during
ficult to appreciate on projection x-ray angiography. the acute phase of the disease have a normal-appearing coronary lumen
Coronary artery CMR has several advantages compared with both on catheter-based x-ray angiography 2 years later.24,25 Transthoracic
coronary MDCT and x-ray angiography in the diagnosis of these coronary echocardiography is often adequate for diagnosing and following these
anomalies. In addition to being noninvasive and not requiring ionizing proximal and mid-vessel aneurysms in very young children, but this
radiation or iodinated contrast agents, coronary artery CMR provides modality is deficient after adolescence and in obese children. These
a definitive three-dimensional (3D) “road map” of the mediastinum young adults are therefore often referred for serial catheter-based x-ray
(Fig. 24.1). Early studies applied two-dimensional (2D) breath-hold coronary angiography, with the accumulation of significant radiation

300
CHAPTER 24  Coronary Artery Imaging: Clinical Results 301

PA
Ao

LA

FIG. 24.1  Noncontrast whole heart 3 T coronary artery cardiovascular magnetic resonance imaging in a
patient with an anomalous left coronary artery (arrow) from the right sinus of Valsalva. This is the malignant
form of anomalous disease in which the anomalous segment courses between the anterior right ventricular
outflow tract/pulmonary artery (PA) and posterior aorta (Ao). LA, Left atrium.

TABLE 24.1  Coronary Artery


Cardiovascular Magnetic Resonance for
Anomalous Coronary Artery Disease
Correctly Classified
Investigator Patients (N) Vessels
McConnell et al.5 15 14 (93%)
Post et al.6 19 19 (100%)a Ao
Vliegen et al.7 12 11 (92%)b
Taylor et al.8 25 24 (96%)
Bunce et al.9 26 26 (100%)c
Razmi et al.10 12 12 (100%)
Tangcharoen et al.15 46 with congenital 46 (100%)
disease
Piccini16 16 15 (94%)
a
Including 3 patients originally misclassified by x-ray angiography. RV
b
Including 5 patients unable to be classified by x-ray angiography.
c
Including 11 patients unable to be classified by x-ray angiography.

exposure over time. Coronary artery CMR data from two series of
adolescents and young adults with coronary artery aneurysms have FIG. 24.2  Noncontrast whole heart coronary artery cardiovascular mag-
confirmed the high accuracy of coronary artery CMR for both the netic resonance in a child with Kawasaki disease and serial coronary
identification and the characterization (diameter and length [Fig. 24.3]) artery aneurysms (arrows) involving the right coronary artery. Ao, Aorta;
of these aneurysms26–30 with addition of cine ventricular function and RV, right ventricle.
late gadolinium enhancement (LGE) for a comprehensive cardiac assess-
ment.28 Although not specifically examined in long-term follow-up
studies, these data suggest that coronary aneurysms can now be effectively As discussed in earlier chapters, noncontrast gradient echo sequences
followed with serial CMR studies, including vessel wall inflammation.29,30 show rapidly moving laminar blood flow as bright, whereas areas of
Similar data have been reported for ectatic coronary vessels.31 stagnant flow or focal turbulence appear dark because of local satura-
tion (stagnant flow) or dephasing (turbulence). Areas of focal stenoses
appear as varying severity of “signal voids” in the coronary artery CMR,
NATIVE VESSEL CORONARY ARTERY STENOSES with the severity of signal loss related to the angiographic stenosis.32
Data support a broad clinical role for coronary artery CMR in the Because of time constraints of breath-hold, 2D breath-hold coronary
assessment of suspected anomalous CAD (and coronary artery bypass artery CMR has relatively limited in-plane spatial resolution, but the
graft patency, which is addressed later), but data are not yet sufficient technique has successfully shown proximal coronary stenoses in several
to support the use of clinical coronary artery CMR for routine identi- clinical studies (Table 24.2).32–35 When reported, the distance from the
fication of coronary artery stenoses among patients presenting with vessel origin to the focal stenosis on coronary artery CMR correlates
chest pain or for screening, even in high-risk patients. However, data closely with x-ray angiography findings.32 However, there have been
suggest a role for coronary artery CMR among patients for the dis- wide variations in the reported sensitivity and specificity of 2D coronary
crimination of ischemic versus nonischemic cardiomyopathy. artery CMR, much of which may be attributable to technical and
302 SECTION II  Ischemic Heart Disease

10

XRA (mm) 8

2
2 4 6 8 10
A MRA (mm)

18

16

14
XRA (mm)

12

10

6
6 8 10 12 14 16 18
B MRA (mm)
FIG. 24.3  Comparison of x-ray angiography and noncontrast two-dimensional breath-hold coronary cardio-
vascular magnetic resonance (A) aneurysm diameter and (B) aneurysm length in patients with Kawasaki
disease. MRA, Magnetic resonance angiography; XRA, x-ray angiography. (Modified from Mavrogeni S,
Papadopoulos G, Douskou M, et al. Magnetic resonance angiography is equivalent to x-ray coronary angiog-
raphy for the evaluation of coronary arteries in Kawasaki disease. J Am Coll Cardiol. 2004;43:649–652.)

TABLE 24.2  Two-Dimensional Breath-Hold Coronary Artery Cardiovascular Magnetic


Resonance for Identification of Focal Coronary Stenoses 50% or More in Diameter
Investigator Subjects (N) Vessels (N, %) Sensitivity Specificity
33
Manning et al. 39 52 (35%) 90% (71%–100%) 92% (78%–100%)
Duerinckx and Urman34 20 27 (34%) 63% (0%–73%) 56% (37%–82%)
Pennell et al.32 39 55 (35%) 85% (75%–100%) —
Post et al.35 35 35 (28%) 63% (0%–100%) 89% (73%–96%)
CHAPTER 24  Coronary Artery Imaging: Clinical Results 303

A B
FIG. 24.4  (A) Noncontrast three-dimensional coronary magnetic resonance angiography in a patient with a
significant stenosis (arrow) of the left anterior descending coronary artery with (B) comparative projection
x-ray angiogram. (From Yoon YE, Kitagawa K, Kato S, et al. Prognostic value of coronary magnetic resonance
angiography for prediction of cardiac events in patients with suspected coronary artery disease. J Am Coll
Cardiol. 2012;60:2316–2322.)

methodologic issues, including the wide variation in patient selection, suggested that gradient echo approaches had higher sensitivity, whereas
presence of arrhythmias, prevalence of disease, the wide range of varia- bSSFP approaches had better specificity.45
tion in technical implementation (CMR vendor, echo time, receiver Increasing studies have examined the role of contrast-enhanced
coils, timing of acquisition, acquisition duration, and breath-hold coronary artery CMR for the detection of disease. Paetsch and associ-
maneuvers), and the need for 20 to 40 breath-holds to complete a study. ates59 compared noncontrast 3D-targeted coronary artery CMR with an
Over the past decade, 2D coronary artery CMR has been supplanted investigational intravascular agent using an inversion recovery prepulse.
by free-breathing 3D gradient echo or balanced steady-state free preces- Superior specificity was found with the addition of contrast. Yang and
sion (bSSFP) coronary artery CMR with targeted 3D or whole heart coworkers using gadobenate dimeglumine (Gd-BOPTA, MultiHance,
approaches with or without contrast agents at 1.5 T and, increasingly, Bracco, Milan, Italy) with a 32-channel receiver coil at 3 T in 62 patients
3 T (see Chapter 23). The free-breathing, whole heart approach has and found an overall patient, vessel, and segment accuracy of 92%, 94%,
higher patient acceptance, improved signal-to-noise ratio, and datasets and 95%, respectively.60 In another study of 110 patients, they found a
that facilitate multiplanar reconstructions for improved visualization patient-based sensitivity and specificity of 96% and 87%, respectively.62
of the coronary arteries. As with noncontrast 2D gradient echo methods, A large number of the whole heart clinical studies have been led by
a focal stenosis or turbulent flow with 3D gradient echo imaging appears groups led by Drs. Hajime Sakuma and Debiao Li, including multicenter
as a signal void or marked narrowing of the lumen along the course trials in Japan61 and China,62a respectively (see Table 24.4). As expected,
of the vessel, with less dependence on blood flow characteristics (laminar multicenter coronary artery CMR data are not as favorable as those
vs. turbulent) with bSSFP imaging. Data from numerous single-center reported in single centers, but multicenter coronary artery CMR and
sites have been published using modern prospective navigator gating multicenter coronary MDCT data appear similar (discussed later).
with real-time correction (Table 24.3).36–47 Although assessment of the Available data suggest that 3D gradient echo coronary artery CMR
sensitivity of coronary artery stenosis was found to be similar for both findings at 1.5 T and 3 T are similar.47 A multicenter 3 T whole heart
source and projection images,48 our strong preference is to make diag- contrast-enhanced coronary artery CMR study by Yang et al. in 278
noses through review of the source images. We then use the projection patients obtained diagnostic quality CMR images in 86% of patients
images to convey our findings visually to the referring physician. An with a sensitivity and specific of 91% and 80%, respectively.62a
international, multicenter, free-breathing, noncontrast, 3D targeted A systematic review and metaanalysis of coronary artery CMR63
coronary artery CMR study of 109 patients without previous x-ray involving 24 reports and 1638 patients showed a pooled sensitivity of
angiography using common hardware and software showed high sen- 89% (95% CI 63%–79%) and specificity of 72% (95% CI 63%–79%).
sitivity (although only modest specificity) and negative predictive value The sensitivity of contrast-enhanced coronary artery CMR was higher
of coronary artery CMR for the identification of coronary disease (>50% (95% vs. 87% for noncontrast), and comparable with coronary MDCT.
diameter stenosis by quantitative coronary angiography; Table 24.4).49 The specificity of 3 T coronary artery CMR was superior (83% vs. 68%
The sensitivity and negative predictive value were particularly high for for 1.5 T) as well as for whole heart acquisitions (78% vs. 57% for
the identification of left main or multivessel disease. Accordingly, coro- targeted acquisitions).
nary artery CMR was especially valuable for patients with dilated car-
diomyopathy in the absence of clinical myocardial infarction. In our Comparison of Coronary Artery CMR and Other
experience, coronary artery CMR is highly accurate and superior to CMR Components
LGE methods for determining the etiology (ischemic vs. nonischemic) The multicenter Clinical Evaluation of Magnetic Resonance Imaging in
of cardiomyopathy. Coronary Heart Disease (CE-MARC) study offered the unique oppor-
Data are increasingly available on whole heart bSSFP coronary artery tunity to examine different components of the CMR examination (left
CMR methods (Fig. 24.4) with clinical results at least as accurate as ventricular function, myocardial perfusion, LGE, coronary artery imaging)
those obtained with targeted acquisition free-breathing methods (see for the detection of CAD (Table 24.5)56 and suggests that coronary artery
Table 24.3).50–58 A comparative study of whole heart gradient echo and CMR image acquisition had the lowest technical success and offered no
whole heart bSSFP coronary artery CMR by Ozgun and colleagues additional benefit for the detection of angiographic disease. A single center
304 SECTION II  Ischemic Heart Disease

TABLE 24.3  Free-Breathing, Three-Dimensional, Gradient Echo Coronary Magnetic Resonance


Imaging Using Prospective Navigators for Identification of Focal Coronary Stenoses 50% or
More in Diameter
Investigator Subjects (N) Technique Sensitivity Specificity
Prospective Navigators With Real-Time Correction Targeted Three-Dimensional Coronary Artery CMR
Regenfus et al.36 50 TFE 94% 57%
Bunce et al.37 34 TFE 88% 72%
Moustapha et al.38 25 TFE 92% 55%
90% (proximal) 92% (proximal)
Sommer et al.39 112 TFE 74% 63%
88% (good quality) 91% (good quality)
Bogaert et al.40 19 TFE 85%–92% 50%–83%
Plein et al.41 10 TFE 75% 85%
Ozgun et al.42 14 TFE 91% 57%
bSSFP 76% 85%
Dewey et al.43 15a bSSFP 86% 98%
Maintz et al.44 12 TFE 92% 67%
bSSFP 81% 82%
Ozgun et al.45 20 bSSFP 82% 82%
Jahnke et al.46 21 bSSFP 79% 91%
Paetsch et al.59 18 bSSFP 83% 77%
18 Contrast 86% 95%
Sommer et al.47 18 TFE 82% 88%

Prospective Navigator With Real-Time Correction Whole Heart Steady-State Free Precession Coronary Artery CMR
Sakuma et al.50 39 bSSFP 82% 91%
Jahnke et al.51 55 bSSFP 78% 91%
Sakuma et al.52 113 bSSFP 82% 90%
Nagata et al.53 62 32-channel SSFP 83% 93%
Klein et al.54 46 bSSFP 91% 54%
Pouleur et al.55 77 bSSFP 100% 72%
Greenwood et al.56 598 bSSFP 72% (M)/67% (F) 90% (M)/88% (F)
Nagata et al.57 67 bSSFP 87% 86%
Heer et al.58 59 bSSFP 86.7% 79.3%

1.5 T Self-Gated Postcontrast Whole Heart Coronary Artery CMR


Piccini et al.16 40 Postcontrast bSSFP 71% 63%

3 T Whole Heart Coronary Artery CMR


Sommer et al.47 18 TFE 82% 89%
Yang et al.60 69 Contrast IR-GRE 94% 82%
a
Based on 60% of patients with good free breathing coronary magnetic resonance images.
bSSFP, Balanced steady-state free precession; CMR, cardiovascular magnetic resonance; IR-GRE, inversion recovery gradient echo; TFE, turbo
field echo.

TABLE 24.4  Multicenter Coronary Artery Cardiovascular Magnetic Resonance Trials


Author Field Strength Patients (N) Sensitivity Specificity PPV NPV
49
Kim et al. 1.5 T 109 93% 42% 70% 81%
Kato et al.61 1.5 T 138 88% 72% 71% 88%
Yang et al.62a 3T 272 91% 80%

NPV, Negative predictive value; PPV, positive predictive value.

study by Heer et al.58 found that the combination of coronary artery CMR Coronary Artery Cardiovascular Magnetic Resonance
with adenosine stress perfusion improved overall accuracy from 78% to and Prognosis
92% for the detection of angiographically significant CAD. A 3T study Increasingly, validation of cardiac imaging is focused on the impact on
by Zhang et al. involving 46 patients found the addition of contrast- outcomes. There are limited data on the prognostic valve of coronary
enhanced coronary artery CMR resulted in higher sensitivity (100% artery CMR. Yoon and colleagues65 studied 207 consecutive patients
vs. 76.5%) and accuracy (89% vs. 74%) with no change in specificity.64 without prior known CAD using whole heart bSSFP free-breathing
CHAPTER 24  Coronary Artery Imaging: Clinical Results 305

coronary artery CMR at 1.5 T. Follow-up was available for a median


TABLE 24.5  Comparison of Different
of 25 months and 41% had CAD on coronary artery CMR (Fig. 24.5).
Components of the Cardiovascular Magnetic The presence of a stenosis on coronary artery CMR had a univariate
Resonance Imaging Examination for hazard ratio of 20.8 (multivariate 18)/12% risk of cardiac death (1.2%),
Detection of Significant Coronary Artery unstable angina (4.8%), or late revascularization (8.3%) whereas the
Disease absence of a stenosis was associated with late revascularization in <1%
CMR Component Sex (N) Sensitivity Specificity of subjects.
CE-MARC Trial; Modified From Greenwood et al. 56
COMPARISON OF CORONARY ARTERY
LV function 393M 49.2% 92.9%
235F 39.6% 95.1% CARDIOVASCULAR MAGNETIC RESONANCE WITH
Perfusion 388M 75.4% 92.9% MULTIDETECTOR COMPUTED TOMOGRAPHY
229F 81.1% 89.2%
Several studies have directly compared MDCT with coronary artery
LGE 392M 41.2% 94.4%
CMR for the detection of CAD (Table 24.6). Overall, these studies have
235F 34.0% 97.8%
suggested similar accuracy when interpretable segments were considered
Coronary artery 349M 72.3% 90.3%
and superiority of 64-slice MDCT for patients with lower calcium scores
208F 66.7% 87.7%
with “intention to diagnose” analyses because of the superior ability of
Heer et al.58 MDCT to acquire interpretable images. The first direct study by Gerber
Coronary artery 59 86.7% 79.3% and colleagues66 compared free-breathing, targeted noncontrast 3D
Coronary artery with 59 95.7% 88.9% coronary artery CMR with four-slice MDCT and showed slight supe-
perfusion riority of coronary artery CMR for overall accuracy. A follow-up study
of 52 patients performed by the same group with 16-slice MDCT showed
Gotschy et al.91 equivalence with visual analysis.67 Another 16-slice MDCT comparative
Coronary artery 11 75% 79% study by Dewey and colleagues showed superiority of MDCT68 for
Coronary artery with 11 94% 82% sensitivity, but the coronary artery CMR technique used free breathing
perfusion and LGE or an inferior multiple-breath-hold approach. Interestingly, coronary
artery CMR sensitivity (74%) was improved compared with an earlier
CE-MARC, Clinical Evaluation of Magnetic Resonance Imaging in report from this group,43 suggesting a learning curve for coronary artery
Coronary Heart Disease; CMR, cardiovascular magnetic resonance; CMR. Patients expressed a preference for MDCT,68 with the advantage
LGE, late gadolinium enhancement; LV, left ventricle.
of very rapid and simplified protocols at the expense of substantial

Severe cardiac events All cardiac events


Significant stenosis (−) Significant stenosis (−)
1.0 1.0
Proportion of event-free survival

0.8 Significant stenosis (+) 0.8

P = .003 Significant stenosis (+)


0.6 0.6
P < .001

0.4 0.4

0.2 0.2

0.0 0.0

0 1 2 3 4 0 1 2 3 4
Follow-up time (years) Follow-up time (years)
No. at risk
Significant stenosis (−) 123 101 68 19 123 101 68 19
A Significant stenosis (+) 84 49 34 11 B 84 45 29 7
FIG. 24.5  Kaplan-Meier event free survival curves based on noncontrast three-dimensional whole-heart
coronary artery cardiovascular magnetic resonance for (A) severe cardiac events and (B) all cardiac events.
(Modified from Yoon YE, Kitagawa K, Kato S, et al. Prognostic value of coronary magnetic resonance angi-
ography for prediction of cardiac events in patients with suspected coronary artery disease. J Am Coll Cardiol.
2012;60:2316–2322.)
306 SECTION II  Ischemic Heart Disease

TABLE 24.6  Comparative Studies of Coronary Artery Cardiovascular Magnetic Resonance


and Multidetector Computed Tomographya
Investigator CMR/CCT Method Sensitivity Specificity PPV NPV Accuracy
66 b
Gerber et al. CMR:FB-SSFP 62% 84% 49% 90% 80%c
4-slice MDCT 79%c 71% 40% 93% 73%
Kefer et al.67 CMR:FB-SSFP 75% 77% 42% 93% 77%
16-slice MDCT 82% 79% 46% 95% 80%
Dewey et al.68 CMR-BH/FB-SSFP 74%b 75% 95% 84% —
16-slice CT 92% 79% 95% 90% —
Maintz et al.69 CMR-FB-SSFP 84% 95% 77% 95% 93%
16-slice MDCT 82% 88% 68% 94% 87%
Pouleur et al.55 CMR-FB SSFP 100% 72%c 50%c 100% 78%c
40/64-slice MDCT 94% 88% 70% 98% 90%
Liu et al.74 CMR-FB-SSFP 75% 81% — — —
64-slice MDCT 75% 48%c — — —
a
Reported values are from patient-based analyses for the detection of coronary artery disease.
b
P < .001.
c
P < .05.
BH, Breath-hold; CMR, cardiovascular magnetic resonance; FB, free breathing; MDCT, multidetector computed tomography; NPV, negative
predictive value; PPV, positive predictive value; SSFP, steady-state free precession.

LAD

LAD
LAD
AO

AO
A B C
FIG. 24.6  (A) Extensive epicardial coronary calcium of the left anterior descending (LAD) coronary artery as
visualized by multidetector computed tomography (MDCT). (B) Coronary artery cardiovascular magnetic reso-
nance image. (C) X-ray coronary angiography. The proximal calcium deposit (arrowhead) obscures the lumen
on MDCT but is seen on cardiovascular magnetic resonance as patent. The more distal calcium deposit
(arrow) is identified as a stenosis on cardiovascular magnetic resonance image and the corresponding x-ray
angiogram. AO, Aorta. (Modified from Liu X, Zhao X, Huang J, et al. Comparison of 3D free-breathing coronary
MR angiography and 64-MDCT angiography for detection of coronary stenosis in patients with high calcium
scores. Am J Roentgenol. 2007;189:1326–1332.)

radiation exposure and need for iodinated contrast. A third comparative appearance on imaging (frequently referred to as “blooming”) that
study by Maintz and associates69 of coronary artery CMR and a 16-slice interferes with the accurate determination of coronary artery steno-
MDCT showed superiority of coronary artery MDCT image quality sis. Several trials using MDCT have excluded patients with substantial
and disease analysis on a coronary segment basis, but showed equiva- epicardial calcium71 or have shown marked reduction in specificity for
lence when only evaluable segments were included. Finally, a comparison patients with Agatston calcium scores >400.72,73 A comparative study of
of whole heart coronary artery CMR with 40-slice or 64-slice MDCT coronary artery CMR and 64-slice MDCT in patients with high calcium
by Pouleur and coworkers55 showed very high sensitivity of coronary scores showed superiority of coronary artery CMR74 (Fig. 24.6).
artery CMR, but overall superiority of coronary artery MDCT. This is
likely related to their “intention to diagnose” analysis and the classifica- CORONARY ARTERY CARDIOVASCULAR
tion of uninterpretable segments as “diseased” in a population with a MAGNETIC RESONANCE FOR CORONARY ARTERY
low (22%) prevalence of CAD. Uninterpretable segments were more
frequent for coronary artery CMR.
BYPASS GRAFT ASSESSMENT
Epicardial calcium is associated with aging and with the development Assessment of coronary artery bypass graft patency is a common clinical
of coronary atherosclerosis. It is a marker for increased risk of adverse issue.75,76 Compared with the native coronary arteries, reverse saphe-
events associated with CAD.70 The presence of epicardial calcium is a nous vein and internal mammary artery grafts are relatively easy to
well-recognized limitation of coronary artery MDCT, with an exaggerated image because of their relatively minimal motion during the cardiac
CHAPTER 24  Coronary Artery Imaging: Clinical Results 307

FIG. 24.7  Transverse fast spin echo images in a patient with previous coronary artery bypass grafting.
(A) The proximal graft is identified (arrow), with subsequent inferior sections (B) and (C) showing a patent
graft (arrows).

TABLE 24.7  Sensitivity, Specificity, and Accuracy of Coronary Artery Cardiovascular


Magnetic Resonance for Assessment of Coronary Artery Bypass Graft Patency
Investigator Technique Grafts (N) Patency Sensitivity Specificity Accuracy
77
White et al. 2D spin echo 72 69% 86% 59% 78%
Rubinstein et al.82 2D spin echo 47 62% 90% 72% 83%
Jenkins et al.83 2D spin echo 41 63% 89% 73% 83%
Galjee et al.79 2D spin echo 98 74% 98% 85% 89%
White et al.78 2D GRE 28 50% 93% 86% 89%
Aurigemma et al.80 2D GRE 45 73% 88% 100% 91%
Galjee et al.79 2D GRE 98 74% 98% 88% 96%
66% (SVG) 92% 85% 89%
Molinari et al.84 3D GRE 51 76.5% 91% 97% 96%
Engelmann et al.81 CE-3D GRE 96 SVG 66% 92% 85% 89%
37 IMA 100% 100% 100%
Wintersperger et al.85 CE-3D GRE 39 87% 97% 100% 97%
Vrachliotis et al.86 CE-3D GRE 45 67% 93% 97% 95%

2D, Two-dimensional; 3D, three-dimensional; CE, contrast-enhanced; GRE, gradient recalled echo; IMA, internal mammary artery graft; SVG,
saphenous vein graft.

and respiratory cycle and the larger lumen of reverse saphenous vein
TABLE 24.8  Diagnostic Accuracy of
grafts. Furthermore, their predictable and less convoluted course has
allowed imaging of bypass grafts, even with less sophisticated CMR
Submillimeter Coronary Artery
methods. Cardiovascular Magnetic Resonance for
With schematic knowledge of the origin and touchdown site of each Saphenous Vein Graft Disease
graft, conventional free-breathing electrocardiographic (ECG)-triggered Sensitivity Specificity
2D spin echo and noncontrast 2D gradient echo coronary artery CMR
Graft occlusion 83% (36%–100%) 100% (92%–100%)
in the transverse plane have both been used to reliably assess bypass
Graft stenosis ≥50% 82% (57%–96%) 88% (72%–97%)
graft patency (Fig. 24.7 and Table 24.7).77–86 Patency is determined by
Graft stenosis ≥70% 73% (39%–94%) 80% (64%–91%)
visualizing a patent graft lumen in at least two contiguous transverse
images along its expected course (presenting as a signal void for spin Modified from Langerak SE, Vliegan HW, de Roos A, et al. Defection
echo techniques and bright signal for gradient echo approaches). If of vein graft disease using high resolution magnetic resonance
signal consistent with flow is identified in the area of the graft lumen, angiography. Circulation. 2002;105:328–333.
it is very likely to be patent. If a patent lumen is seen at only one level
(e.g., for spin echo techniques, a signal void is seen at only one level),
a graft is considered indeterminate. If a patent graft lumen is not seen Limitations of coronary artery CMR bypass graft assessment include
at any level, the graft is very likely occluded. Combining spin echo and difficulties related to local signal loss and artifact as a result of implanted
gradient echo imaging in the same patient does not appear to improve metallic objects (hemostatic clips, ostial stainless-steel graft markers,
accuracy.79 A 3D noncontrast84 and contrast-enhanced coronary artery sternal wires, coexistent prosthetic valves and supporting struts or rings,
CMR has also been described for the assessment of graft patency,85,86 and graft stents; Fig. 24.8). The inability to identify severely diseased
with slightly improved results (see Table 24.7). The accuracy of ECG- yet patent grafts is also a hindrance to clinical utility and acceptance.
triggered bSSFP sequences appears to be similar to that of spin echo Langerak and colleagues88 found free-breathing navigator 3D gradi-
and gradient echo approaches.87 ent echo coronary artery CMR to be accurate for the assessment of
308 SECTION II  Ischemic Heart Disease

SVG
marker

Ao

A B C
FIG. 24.8  (A) Posterior-anterior chest radiograph in a patient with coronary artery bypass grafts. Note the
sternal wires (thick arrow) and the coronary artery bypass markers (thin arrow). (B) Transverse coronary
cardiovascular magnetic resonance image in the same patient. Note the large local artifacts (signal voids)
related to sternal wires (thick arrow) and bypass graft markers (thin arrow). (C) Barium and tantalum markers
(arrow) result in the smallest artifacts. The size of the artifacts is reduced somewhat with spin echo and
black-blood cardiovascular magnetic resonance. Ao, Aorta; SVG, saphenous vein graft.

saphenous vein graft stenoses, with very good agreement with quantita- strongly suggest the absence of left main or multivessel disease and also
tive x-ray angiography for the assessment of both graft occlusion and conveys an excellent prognosis. Overall, data suggest that successful
graft stenoses (Table 24.8). This group has also advocated assessment coronary artery imaging is accomplished more often with MDCT, but
of rest and adenosine stress coronary artery flow assessment using the when successfully obtained, accuracy is similar for CMR and MDCT.
phase velocity CMR technique,89,90 suggesting superior results for flow CMR is superior in patients with prominent epicardial calcium and has
assessment. the advantage of avoiding ionizing radiation and iodinated contrast.
Technical and methodologic advances in motion suppression, along
with improved surface coil design, faster acquisitions, and intravascular
CONCLUSION contrast agents continue to advance the field (see Chapter 23). Impor-
Over the last two decades, coronary artery CMR has been transformed tantly, future studies comparing coronary artery CMR with fractional
from a scientific curiosity to a clinically useful imaging tool in selected flow reserve and assessing outcomes will be needed for coronary magnetic
populations. Uses include the identification and characterization of resonance imaging to successfully transition to the clinical arena for
anomalous coronary arteries, characterization of aneurysms, and the assessment of patients with suspected CAD.
assessment of coronary artery bypass graft patency. Coronary artery
CMR also appears to be of clinical value for the assessment of native
vessel-integrity in selected patients, especially those with suspected left
REFERENCES
main or multivessel disease. Normal findings on coronary artery CMR A full reference list is available online at ExpertConsult.com
CHAPTER 24  Coronary Artery Imaging: Clinical Results 308.e1

REFERENCES Task Force on Expert Consensus Documents. J Am Coll Cardiol.


2010;55(23):2614–2662.
1. Engel HJ, Torres C, Page HL. Major variations in anatomical origin of the 20. Schmid M, Achenbach S, Ludwig J, et al. Visualization of coronary artery
coronary arteries: angiographic observations in 4,250 patients without anomalies by contrast-enhanced multidetector row spiral computed
associated congenital heart disease. Cathet Cardiovasc Diagn. tomography. Int J Cardiol. 2006;111(3):430–435.
1975;1(2):157–169. 21. Deibler AR, Kuzo RS, Vohringer M, et al. Imaging of congenital coronary
2. Chaitman BR, Lesperance J, Saltiel J, Bourassa MG. Clinical, anomalies with multislice computed tomography. Mayo Clin Proc.
angiographic, and hemodynamic findings in patients with anomalous 2004;79(8):1017–1023.
origin of the coronary arteries. Circulation. 1976;53(1):122–131. 22. Budoff MJ, Ahmed V, Gul KM, et al. Coronary anomalies by cardiac
3. Cheitlin MD, De Castro CM, McAllister HA. Sudden death as a computed tomographic angiography. Clin Cardiol. 2006;29:
complication of anomalous left coronary origin from the anterior sinus 489–493.
of Valsalva, a not-so-minor congenital anomaly. Circulation. 1974;50(4): 23. Makino N, Nakamura Y, Yashiro M, et al. Descriptive epidemiology of
780–787. Kawasaki disease in Japan, 2011-2012: from the results of the 22nd
4. Maron BJ, Doerer JJ, Haas TS, et al. Sudden deaths in young competitive nationwide survey. J Epidemiol. 2015;25:239–245.
athletes. Analysis of 1866 deaths in the United States, 1980-2006. 24. Akagi T, Rose V, Benson LN, et al. Outcome of coronary artery
Circulation. 2009;119:1085–1092. aneurysms after Kawasaki disease. J Pediatr. 1992;121(5 Pt 1):689–694.
5. McConnell MV, Ganz P, Selwyn A, et al. Identification of anomalous 25. Kato H, Ichinose E, Yoshioka F, et al. Fate of coronary aneurysms in
coronary arteries and their anatomic course by magnetic resonance Kawasaki disease: serial coronary angiography and long-term follow-up
coronary angiography. Circulation. 1995;92(11):3158–3162. study. Am J Cardiol. 1982;49(7):1758–1766.
6. Post JC, van Rossum AC, Bronzwaer JG, et al. Magnetic resonance 26. Greil GF, Stuber M, Botnar RM, et al. Coronary magnetic resonance
angiography of anomalous coronary arteries: a new gold standard angiography in adolescents and young adults with Kawasaki disease.
for delineating the proximal course? Circulation. 1995;92(11): Circulation. 2002;105(8):908–911.
3163–3171. 27. Mavrogeni S, Papadopoulos G, Douskou M, et al. Magnetic resonance
7. Vliegen HW, Doornbos J, de Roos A, et al. Value of fast gradient echo angiography is equivalent to X-ray coronary angiography for the
magnetic resonance angiography as an adjunct to coronary arteriography evaluation of coronary arteries in Kawasaki disease. J Am Coll Cardiol.
in detecting and confirming the course of clinically significant coronary 2004;43(4):649–652.
artery anomalies. Am J Cardiol. 1997;79(6):773–776. 28. Mavrogeni S, Papadopoulos G, Douskou M, et al. Magnetic resonance
8. Taylor AM, Thorne SA, Rubens MB, et al. Coronary artery imaging in angiography, function and viability evaluation in patients with Kawasaki
grown up congenital heart disease: complementary role of magnetic disease. J Cardiovasc Magn Reson. 2006;8(4):493–498.
resonance and x-ray coronary angiography. Circulation. 29. Greil GF, Seeger A, Miller S, et al. Coronary magnetic resonance
2000;101(14):1670–1678. angiography and vessel wall imaging in children with Kawasaki disease.
9. Bunce NH, Lorenz CH, Keegan J, et al. Coronary artery anomalies: Pediatr Radiol. 2007;27(7):666–673.
assessment with free-breathing three-dimensional coronary MR 30. Mavrogeni S, Bratis K, Karanasios E, et al. CMR evaluation of cardiac
angiography. Radiology. 2003;227(1):201–208. involvement during the convalescence of Kawasaki disease. JACC
10. Razmi RM, Chun W, Rathi VK, Pohost GM. Coronary magnetic Cardiovasc Imaging. 2011;4(10):1140–1141.
resonance angiography (CMRA): the gold standard for determining the 31. Mavrogeni S, Papadakis E, Foussas S, et al. Correlation between magnetic
proximal course of anomalous coronary arteries [abstract]. J Am Coll resonance angiography (MRA) and quantitative coronary angiography
Cardiol. 2001;37:380. (QCA) in ectatic coronary vessels. J Cardiovasc Magn Reson.
11. Ripley DP, Saha A, Teis A, et al. The distribution and prognosis of 2004;6(1):17–23.
anomalous coronary arteries identified by cardiovascular magnetic 32. Pennell DJ, Bogren HG, Keegan J, et al. Assessment of coronary artery
resonance: 15 year experience from two tertiary centres. J Cardiovasc stenosis by magnetic resonance imaging. Heart. 1996;75(2):127–133.
Magn Reson. 2014;16:34. 33. Manning WJ, Li W, Edelman RR. A preliminary report comparing
12. Setser RM, Arruda J, Weaver JA, et al. Exclusion of coronary artery magnetic resonance coronary angiography with conventional
anomalies in patients with congenital cardiovascular abnormalities using angiography. N Engl J Med. 1993;328(12):828–832.
whole heart approach [abstract]. J Cardiovasc Magn Reson. 34. Duerinckx AJ, Urman MK. Two-dimensional coronary MR angiography:
2007;9(2):128–129. analysis of initial clinical results. Radiology. 1994;193(3):731–738.
13. Su JT, Chung T, Muthupillai R, et al. Usefulness of real-time navigator 35. Post JC, van Rossum AC, Hofman MB, et al. Clinical utility of two-
magnetic resonance imaging for evaluating coronary artery origins in dimensional magnetic resonance angiography in detecting coronary
pediatric patients. Am J Cardiol. 2005;95(5):679–682. artery disease. Eur Heart J. 1997;18(3):426–433.
14. Gharib AM, Ho VB, Rosing DR, et al. Feasibility of using free-breathing 36. Regenfus M, Ropers D, Achenbach S, et al. Noninvasive detection of
3D whole-heart coronary MR angiography at 3T to assess coronary coronary artery stenosis using contrast-enhanced three-dimensional
artery anomalies and variants [abstract]. J Cardiovasc Magn Reson. breath-hold magnetic resonance coronary angiography. J Am Coll Cardiol.
2007;9(2):129–130. 2000;36:44–50.
15. Tangcharoen T, Bell A, Hegde S, et al. Detection of coronary artery 37. Bunce NRS, Jhooti P, Lorenz C, Pennell D. The assessment of coronary
anomalies in infants and young children with congenital heart disease by artery disease by combined magnetic resonance coronary arteriography
using MR imaging. Radiology. 2011;259(1):240–247. and perfusion. [abstract]. J Cardiovasc Magn Reson. 2001;3(1):118.
16. Piccini D, Monney P, Sierro C, et al. Respiratory self-navigated post- 38. Moustapha AI, Muthupillai R, Wilson J, Flamm SD. Coronary magnetic
contrast whole-heart coronary MR angiography: initial experience in resonance angiography using a free breathing, T2 weighted, three-
patients. Radiology. 2014;270(2):378–386. dimensional gradient echo sequence with navigator respiratory and ECG
17. Einstein AJ, Henzlova MJ, Rajagopalan S. Estimating risk of cancer gating can be used to detect coronary artery disease [abstract]. J Am Coll
associated with radiation exposure from 64-slice computed tomography Cardiol. 2001;37(suppl):380.
coronary angiography. JAMA. 2007;298:317–323. 39. Sommer T, Hofer U, Meyer C, et al. Submillimeter 3D coronary MR
18. Radbruch A, Weberling LD, Kleislich PJ, et al. Gadolinium retention in angiography with real-time navigator correction in 112 patients with
the dentate nucleus and globus pallidus is dependent on the class of suspected coronary artery disease [abstract]. J Cardiovasc Magn Reson.
contrast agent. Radiology. 2015;275:783–791. 2002;4(1):28.
19. Hundley WG, Bluemke DA, Finn JP, et al. ACCF/ACR/AHA/NASACI/ 40. Bogaert J, Kuzo R, Dymarkowski S, et al. Coronary artery imaging with
SCMR 2010 expert consensus document on cardiovascular magnetic real-time navigator three-dimensional turbo-field-echo MR coronary
resonance: a report of the American College of Cardiology Foundation angiography: initial experience. Radiology. 2003;226(3):707–716.
308.e2 SECTION II  Ischemic Heart Disease

41. Plein S, Ridgway JP, Jones TR, et al. Coronary artery disease: assessment 60. Yang Q, Li K, Bi X, et al. Contrast-enhanced whole heart coronary
with a comprehensive MR imaging protocol—initial results. Radiology. magnetic resonance angiography at 3T: a comparative study with x-ray
2002;225(1):300–307. angiography in a single center. J Am Coll Cardiol. 2009;54:69–76.
42. Ozgun M, Quante M, Kouwenhoven M, et al. Comparison of spoiled TFE 61. Kato S, Kitagawa K, Ishida N, et al. Assessment of coronary artery disease
and balanced TFE coronary MR angiography [abstract]. J Cardiovasc using magnetic resonance coronary angiography: a national multicenter
Magn Reson. 2004;6(1):268–269. trial. J Am Coll Cardiol. 2010;56:983–991.
43. Dewey M, Teige F, Schnapauff D, et al. Combination of free-breathing 62. Yang Q, Li K, Liu X, et al. 3.0T whole-heart coronary magnetic resonance
and breathhold steady-state free precession magnetic resonance angiography performed with 32-channel cardiac coils: a single center
angiography for detection of coronary artery stenoses. J Magn Reson experience. Circ Cardiovasc Imaging. 2012;5(5):573–579.
Imaging. 2006;23(5):674–681. 62a.  Yang Q, Li K, Sun B, et al. Assessment of coronary artery disease using
44. Maintz D, Aepfelbacher FC, Kissinger KV, et al. Coronary MR 3.0 T magnetic resonance coronary angiography: a national multicenter
angiography: comparison of quantitative and qualitative data from four trial. J Cardiovasc Magn Reson. 2013;15(Suppl1):E5.
techniques. AJR Am J Roentgenol. 2004;182(2):515–521. 63. Di Leo G, Fisci E, Seechi F, et al. Diagnostic accuracy of magnetic
45. Ozgun M, Hoffmeier A, Kouwenhoven M, et al. Comparison of 3D resonance angiography for detection of coronary artery disease: a
segmented gradient-echo and steady-state free precession coronary MRI systemic review and meta-analysis. Eur Radiol. 2016;26:3706–3718.
sequences in patients with coronary artery disease. AJR Am J Roentgenol. 64. Zhang L, Song X, Dong L, et al. Additive value of 3T cardiovascular
2005;185(1):103–109. magnetic resonance coronary angiography for detecting coronary artery
46. Jahnke C, Paetsch I, Schnackenburg B, et al. Coronary MR angiography disease. J Cardiovasc Magn Reson. 2018;20. In press.
with steady-state free precession: individually adapted breathhold 65. Yoon YE, Kitagawa K, Kato S, et al. Prognostic value of coronary
technique versus free-breathing technique. Radiology. magnetic resonance angiography for prediction of cardiac events in
2004;232(3):669–676. patients with suspected coronary artery disease. J Am Coll Cardiol.
47. Sommer T, Hackenbroch M, Hofer U, et al. Coronary MR angiography at 2012;60:2316–2322.
3.0T versus that at 1.5 T: initial results in patients suspected of having 66. Gerber BL, Coche E, Pasquet A, et al. Coronary artery stenosis: direct
coronary artery disease. Radiology. 2005;234:718–725. comparison of four section multi-detector row CT and three-dimensional
48. van Geuns RJ, de Bruin HG, Rensing BJ, et al. Magnetic resonance navigator MR imaging for detection-initial results. Radiology.
imaging of the coronary arteries: clinical results from three dimensional 2005;234(1):98–108.
evaluation of a respiratory gated technique. Heart. 1999;82(4):515–519. 67. Kefer J, Coche E, Legros G, et al. Head-to-head comparison of three-
49. Kim WY, Danias PG, Stuber M, et al. Coronary magnetic resonance dimensional navigator-gated magnetic resonance imaging and 16-slice
angiography for the detection of coronary stenoses. N Engl J Med. computed tomography to detect coronary artery stenosis in patients.
2001;345(26):1863–1869. J Am Coll Cardiol. 2005;46(1):92–100.
50. Sakuma H, Ichikawa Y, Suzawa N, et al. Assessment of coronary arteries 68. Dewey M, Teige F, Schnapauff D, et al. Noninvasive detection of coronary
with total study time of less than 30 minutes by using whole-heart artery stenoses with multislice computed tomography or magnetic
coronary MR angiography. Radiology. 2005;237(1):316–321. resonance imaging. Ann Intern Med. 2006;145(6):407–415.
51. Jahnke C, Paetsch I, Nehrke K, et al. Rapid and complete coronary 69. Maintz D, Ozgun M, Hoffmeier A, et al. Whole heart coronary MR
arterial tree visualization with magnetic resonance imaging: feasibility angiography: value for the detection of coronary artery stenosis in
and diagnostic performance. Eur Heart J. 2005;26(21):2313–2319. comparison to MSCT-angiography. Acta Radiol. 2007;48(9):967–973.
52. Sakuma H, Ichikawa Y, Chino S, et al. Detection of coronary artery 70. Detrano R, Guerci AD, Carr JJ, et al. Coronary calcium as a predictor of
stenosis with whole-heart coronary magnetic resonance angiography. coronary events in four racial or ethnic groups. N Engl J Med.
J Am Coll Cardiol. 2006;48(10):1946–1950. 2008;358:1336–1345.
53. Nagata M, Sakuma H, Ishida N, et al. Whole heart coronary magnetic 71. Miller JM, Rochitte CE, Dewey M, et al. Diagnostic performance of
resonance angiography using 32-channel cardiac coils and high parallel coronary angiography by 64-row CT. N Engl J Med. 2008;359:2324–
imaging factor [abstract]. Circulation. 2008;118(18):S778. 2336.
54. Klein C, Gebker R, Kokocinski T, et al. Combined magnetic resonance 72. Budoff MJ, Dowe D, Jolis JG, et al. Diagnostic performance of
coronary artery imaging, myocardial perfusion, and late gadolinium 64-multidetector row coronary computed tomographic angiography for
enhancement in patients with suspected coronary artery disease. evaluation of coronary artery stenosis in individuals without known
J Cardiovasc Magn Reson. 2008;10:45. coronary artery disease. J Am Coll Cardiol. 2008;52:1724–1732.
55. Pouleur AC, de Waroux JBP, Kefer J, et al. Direct comparison of 73. Raff GL, Gallagher MJ, O’Neill WW, Goldstein JA. Diagnostic accuracy of
whole-heart navigator-gated magnetic resonance coronary angiography non-invasive coronary angiography using 64-slice spiral computed
and 40- and 64-slice multidetector row computed tomography to detect tomography. J Am Coll Cardiol. 2005;46:552–557.
the coronary artery stenosis in patients scheduled for conventional 74. Liu X, Zhao X, Huang J, et al. Comparison of 3D free-breathing coronary
coronary angiography. Circ Cardiovasc Imaging. 2008;1:114–121. MR angiography and 64-MDCT angiography for detection of coronary
56. Greenwood JP, Motwani M, Maredia N, et al. Comparison of stenosis in patients with high calcium scores. AJR Am J Roentgenol.
cardiovascular magnetic resonance and single-photon emission computed 2007;189:1326–1332.
tomography in women with suspected coronary artery disease from the 75. Shroyer AL, Grover FL, Hattler B, et al. On-pump versus off-pump
Clinical Evaluation of Magnetic Resonance Imaging in Coronary Heart coronary-artery bypass surgery. N Engl J Med. 2009;361:1827–1837.
Disease (CE-MARC) trial. Circulation. 2014;129:1129–1138. 76. Lopez RD, Hafley GE, Allen KB, et al. Endoscopic versus open vein-graft
57. Nagata M, Kato S, Kitagawa K, et al. Diagnostic accuracy of 1.5-T harvesting in coronary-artery bypass surgery. N Engl J Med.
unenhanced whole-heart coronary MR angiography performed with 2009;361:235–244.
32-channel cardiac coils: initial single-center experience. Radiology. 77. White RD, Caputo GR, Mark AS, et al. Coronary artery bypass graft
2011;259:384–392. patency: noninvasive evaluation with MR imaging. Radiology.
58. Heer T, Reiter S, Hofling B, Pilz G. Diagnostic performance of non- 1987;164(3):681–686.
contrast-enhanced whole-heart magnetic resonance coronary 78. White RD, Pflugfelder PW, Lipton MJ, Higgins CB. Coronary artery
angiography in combination with adenosine stress perfection cardiac bypass grafts: evaluation of patency with cine MR imaging. AJR Am J
magnetic resonance imaging. Am Heart J. 2013;166(6):999–1009. Roentgenol. 1988;150(6):1271–1274.
59. Paetsch I, Jahnke C, Barkhausen J, et al. Detection of coronary stenoses 79. Galjee MA, van Rossum AC, Doesburg T, et al. Value of magnetic
with contrast enhanced, three-dimensional free breathing coronary MR resonance imaging in assessing patency and function of coronary artery
angiography using the gadolinium-based intravascular contrast agent bypass grafts: an angiographically controlled study. Circulation.
gadocoletic acid (B-22956). J Cardiovasc Magn Reson. 2006;8(3):509–516. 1996;93(4):660–666.
CHAPTER 24  Coronary Artery Imaging: Clinical Results 308.e3

80. Aurigemma GP, Reichek N, Axel L, et al. Noninvasive determination of 86. Vrachliotis TG, Bis KG, Aliabadi D, et al. Contrast-enhanced breath-hold
coronary artery bypass graft patency by cine magnetic resonance MR angiography for evaluating patency of coronary artery bypass grafts.
imaging. Circulation. 1989;80(6):1595–1602. AJR Am J Roentgenol. 1997;168(4):1073–1080.
81. Engelmann MG, Knez A, von Smekal A, et al. Non-invasive coronary 87. Bunce NH, Lorenz CH, John AS, et al. Coronary artery bypass graft
bypass graft imaging after multivessel revascularisation. Int J Cardiol. patency: assessment with true ast imaging with steady-state precession
2000;76(1):65–74. versus gadolinium-enhanced MR angiography. Radiology. 2003;227(2):
82. Rubinstein RI, Askenase AD, Thickman D, et al. Magnetic resonance 440–446.
imaging to evaluate patency of aortocoronary bypass grafts. Circulation. 88. Langerak SE, Vliegan HW, de Roos A, et al. Detection of vein graft
1987;76(4):786–791. disease using high resolution magnetic resonance angiography.
83. Jenkins JP, Love HG, Foster CJ, et al. Detection of coronary artery bypass Circulation. 2002;105(3):328–333.
graft patency as assessed by magnetic resonance imaging. Br J Radiol. 89. Langerak SE, Kunz P, Vliegen HW, et al. MR flow mapping in coronary
1988;61(721):2–4. artery bypass grafts: a validation study with Doppler flow measurements.
84. Molinari G, Sardanelli F, Zandrino F, et al. Value of navigator echo Radiology. 2002;222(1):127–135.
magnetic resonance angiography in detecting occlusion/patency of 90. Langerak SE, Vliegen HW, Jukema JW, et al. Value of magnetic resonance
arterial and venous, single and sequential coronary bypass grafts. Int J imaging for the noninvasive detection of stenosis in coronary artery
Card Imaging. 2000;16(3):149–160. bypass grafts and recipient coronary arteries. Circulation.
85. Wintersperger BJ, Engelmann MG, Von Smekal A, et al. Patency of 2003;107(11):1502–1508.
coronary bypass grafts: assessment with breath-hold contrast-enhanced 91. Gotschy A, Wissmann L, Goolaub D, et al. Fusion of 3D-CMR-perfusion
MR angiography–value of a non-electrocardiographically triggered with 3D-MR-coronary angiography. J Cardiovasc Magn Reson. 2015;
technique. Radiology. 1998;208(2):345–351. 17(suppl 1):88.
25 
Coronary Artery and Sinus Velocity and Flow
Jennifer Keegan and Dudley J. Pennell

A coronary stenosis may be observed during cardiovascular magnetic generate results. This chapter reviews the progress made to date and
resonance (CMR) as an area of signal loss caused by turbulent flow, discusses potential future improvements.
and although both the degree and the extent of signal loss are indicative
of the severity of the stenosis, accurate quantification is not possible.1 INDIRECT ASSESSMENT OF TOTAL CORONARY
However, both phasic coronary artery blood flow and flow velocity
may be affected by the presence of stenosis, and the ratio of coronary
FLOW AND FLOW RESERVE
artery flow under maximal vasodilation to coronary artery flow at rest Two indirect approaches for assessing total coronary flow and flow
(coronary flow reserve) is a good indicator of the physiologic signifi- reserve by CMR have been reported, the first from velocity mapping
cance to the myocardium. This makes the measurement of coronary of cardiac venous outflow and the second from velocity mapping in
flow and velocity valuable. Measurements of instantaneous and mean the aortic root.
coronary flow parameters are most commonly made using an intra-
coronary Doppler flow wire, positioned in the arterial lumen during Coronary Sinus Flow
x-ray contrast angiography. However, this is an invasive procedure, Velocity mapping of coronary venous outflow is less problematic than
with a small but definite risk of complications, and the radiation dose velocity mapping in the coronary arteries because the coronary sinus
to the patient is relatively high for a diagnostic test. The repeated use has a much larger diameter (typically 7–10 mm), and also because
of such a technique to monitor disease progression or regression in effects of signal loss are unlikely because flow is less susceptible to
response to drug therapy or lifestyle changes is therefore not acceptable. turbulence. In the human heart, coronary sinus flow almost equals total
In addition, the presence of the Doppler wire may itself affect flow,2 as coronary flow because approximately 96% of left ventricular (LV) venous
may the stress to the patient resulting from the invasive nature of the blood flow drains into the right atrium via the coronary sinus.5 van
procedure. Other techniques capable of assessing mean flow parameters Rossum and colleagues were the first to show that blood flow in the
include continuous thermodilution and positron emission tomogra- coronary sinus could be measured using CMR.6 This feasibility study
phy (PET), the former also requiring placement under x-ray contrast assessed the ability of cine CMR velocity mapping to measure phasic
angiography, with the concomitant disadvantages, and the latter being and mean coronary venous outflow in the distal coronary sinus of 24
relatively unavailable and expensive. healthy subjects. The flow profiles were generally biphasic and primarily
CMR has the ability to quantify blood flow noninvasively and has diastolic, with 37% of subjects showing some reverse flow immediately
the potential to be a useful noninvasive alternative to the intracoronary after the R-wave. The mean volumetric flow over the cardiac cycle was
technique. Since the 1980s, it has been used extensively for the assess- 144 ± 62 mL/min, similar to values reported in normal subjects using
ment of phasic blood flow velocity and flow in a wide range of cardio- continuous thermodilution (122 mL/min).7 Although phasic blood flow
vascular applications,3,4 and it has the potential to be implemented in in the sinus may have been expected to be predominantly systolic, as
coronary flow studies. One of the main problems relating specifically in other venous structures, the authors suggested that the thin, compli-
to applying the technique to the coronary arteries is their small size ant walls of the sinus, together with its drainage into the right atrium,
(typically <5 mm in diameter), which, for current levels of in-plane where pressure varies considerably, may be responsible for the pre-
resolution, results in only a few pixels across the vessel. This has impli- dominantly diastolic flow profile. These findings have been supported
cations for the accurate measurement of both vessel cross-sectional by other independent studies, both directly, using an ultrasonic transit
area and blood flow velocity. Vessel tortuosity is a further problem, time technique to measure phasic volumetric coronary sinus flow in
giving rise to difficulties in accurately aligning the vessel so that flow conscious dogs,8 and indirectly, by observing areas of signal void, caused
is truly through-plane or in-plane, as required. This is exacerbated by by accelerating and turbulent flow near the entrance of the coronary
the movement of the arteries with the cardiac and respiratory cycles. sinus in the right atrium in early diastole, in conventional cine CMR
In addition, the temporal resolution of the velocity encoding sequence in healthy subjects.9
must be sufficiently good to minimize the blurring of the vessel as a Fig. 25.1 shows the typical image plane orientation for coronary sinus
result of motion within the period of acquisition and to resolve the flow measurements together with a typical coronary sinus flow curve.
phasic velocity profile. The low peak flow velocities in normal arteries The imaging time for the feasibility study was typically 4 minutes,
at rest (typically <25 cm/s) present a further challenge and require and this may be inappropriate when assessing sinus flow under phar-
highly sensitive velocity windows, whereas in the presence of stenoses, macologically induced maximal vasodilation. Kawada and associates10
high velocities are present, together with complex flow, which may lead assessed the possibility of using a segmented k-space11 gradient echo
to signal loss. The combination of these problems is formidable, and CMR approach to the acquisition so that all data could be acquired in
it was only in the last 10 to 15 years that CMR techniques started to <25 seconds. This had the further advantage that the full acquisition

309
310 SECTION II  Ischemic Heart Disease

Coronary sinus Coronary sinus

A B C D Coronary sinus E
FIG. 25.1  Coronary sinus flow measurement. The coronary sinus is identified in the atrioventricular groove,
on the basal slice of a short-axis stack (A). The plane for flow measurement is prescribed parallel to the
long-axis of the heart on the 4-chamber view and perpendicular to the direction of flow in the coronary sinus,
approximately 0.5 cm from the ostium (green line, A and B). The proximal coronary sinus is seen in cross-
section on the phase contrast images (C and D). Flow versus time curves are generated by drawing a region
of interest around the coronary sinus to calculate through-plane flow (E). (From Dandekar VK, Baumi MA,
Ertel AW, et al. Assessment of global myocardial perfusion using cardiovascular magnetic resonance of coro-
nary sinus flow at 3 Tesla. J Cardiovasc Magn Reson. 2014;16:24–33.)

could be performed in a single breath-hold, thereby eliminating the investigate the role of aminophylline reversal.18 It has also been used
effects of respiratory motion. In a study of 9 healthy subjects and 29 to show reduced coronary flow reserve in patients with surgically repaired
patients with hypertrophic cardiomyopathy (HCM), data were acquired tetralogy of Fallot, compared with healthy volunteers (1.19 ± 0.34 vs.
from an oblique coronal plane. Four reference and four velocity encoded 2.00 ± 0.43, P = .002)19 and in two similar studies (one at 1.5 T20 and
views were acquired per data segment, giving a segment duration of one at 3 T)21 to show that, whereas smokers and nonsmokers have
120 ms, but the temporal resolution was effectively improved by view similar baseline myocardial blood flows, smokers have a significantly
sharing,12 a technique whereby data are generated at intermediate time reduced response to cold pressor testing (Fig. 25.2). A spiral k-space
points from the preceding and following data segments. Hence, depend- coverage phase velocity mapping sequence at 3 T has allowed the acqui-
ing on the RR interval, velocity maps were obtained at up to 14 phases sition of data with a spatial resolution of 0.8 mm × 0.8 mm in an 11
in the cardiac cycle. The authors observed the same biphasic velocity to 15 s breath-hold with a temporal resolution of 60 to 69 ms and has
and flow profiles6 in both healthy subjects and patients with HCM, been used to show myocardial blood flow increases in response to cold
with no significant difference noted between the baseline myocardial pressor testing in asymptomatic women.22 A validation of the breath-
blood flow (coronary blood flow per unit mass of myocardium) in the hold approach was reported by Koskenvuo and associates, who compared
two groups (0.74 mL/min per gram vs. 0.62 mL/min per gram). However, myocardial blood flow measured by CMR with that measured by PET
after intravenous administration of 0.56 mg/kg dipyridamole, the increase in both healthy subjects23 and patients with coronary artery disease
in myocardial blood flow in patients with HCM (0.62 mL/min per (CAD).24 Good correlations were reported for myocardial blood flow
gram to 1.03 mL/min per gram) was less than that in the healthy subjects in both subject groups (0.82 and 0.80, respectively), whereas for coronary
(0.74 to 2.14 mL/min per gram), resulting in significantly different flow reserve, the correlations were 0.76 and 0.5, respectively.
coronary flow reserves for the two groups (1.72 ± 0.49 vs. 3.01 ± 0.75, One issue with the breath-hold approach to measuring coronary
respectively, P < .01). The authors concluded that, in healthy subjects, sinus flow is that breath-holding changes intrathoracic pressure and
myocardial blood flow and coronary flow reserve, as measured by breath- affects cardiac output and venous blood flow.25 Schwitter and cowork-
hold phase velocity mapping, were similar to those found by other ers26 performed a validation of nonbreath-hold velocity mapping against
techniques; in addition, the CMR technique was able to distinguish PET. Although taking longer to acquire (typically 4 minutes), the
between healthy subjects and patients with HCM. A more recent study nonbreath-hold technique has the advantages of higher spatial resolu-
in young HCM patients (22.3 ± 6.4 years) and age-matched subjects tion (0.8 × 0.8 mm) and improved SNR through the acquisition of
at risk of HCM has similarly shown that HCM patients (but not subjects multiple averages. Furthermore, the high temporal resolution that is
at risk of HCM) have reduced myocardial blood flow response to achievable is better suited for resolving the highly pulsatile flow profile
adenosine-induced hyperemia, even in the absence of diastolic dysfunc- and for minimizing the blurring effects of the extensive in-plane motion
tion or left ventricular outflow tract obstruction.13 The breath-hold of the coronary sinus during the cardiac cycle. Unlike the initial study
approach has also been used by Kennedy and colleagues14 to show of van Rossum and coworkers,6 this free breathing study used retrospec-
significant differences in the coronary flow reserve of healthy subjects tive electrocardiogram (ECG) gating,27 which enables data acquisition
(4.59 ± 0.58) and heart transplant patients with mild (2.15 ± 0.44, P < throughout the entire cardiac cycle, and respiratory ordered phase
.05) and severe (2.21 ± 0.59, P < .05) coronary disease, as determined encoding,28 which reduces the effects of respiratory motion. Scans were
by posttransplant coronary angiography. It has also been used to show performed both before and after the administration of 0.56 mg/kg
reduced coronary flow reserve in patients with high serum eicosapen- dipyridamole in 16 healthy subjects and 10 orthotopic heart transplant
taenoic acid (EPA) compared with those with low EPA,15 and in those recipients and the results were compared with PET data. The mean
with heart failure with preserved ejection fraction compared with those difference between coronary flow reserve measured by PET and CMR
with hypertensive left ventricular hypertrophy and controls.16 At 3 T, was 2.2%, with limits of agreement of −27.2% and 31.6%. Correlation
the increased signal-to-noise ratio (SNR) available generally allows between CMR-measured myocardial blood flow (coronary sinus flow
imaging of coronary sinus flow with higher spatial resolution or, par- divided by myocardial mass) and PET was good (r = 0.93), although
ticularly when used in combination with parallel imaging techniques, the slope was considerably less than unity (0.73). This underestimation
reduced breath-hold duration.17 Imaging at 3 T has been used to deter- of blood flow measured by CMR results from the fact that a variable
mine the feasibility of flow reserve quantification using regadenoson, part of the inferior and inferior-septal myocardium is drained by the
a relatively new selective adenosine A2A receptor agonist and to middle cardiac vein, which either enters the coronary sinus just before
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 311

0.7 80
P = .005 P = .003
0.6 70

0.5 60

0.4 50

Percentage change in MBF (%)


0.3 40
∆MBF (mL/min/g)

0.2 30

0.1 20

0 10

−0.1 0

−0.2 −10

−0.3 −20

−0.4 −30
Nonsmokers Smokers Nonsmokers Smokers
FIG. 25.2  Individual myocardial blood flow (MBF) Δ values and percent change of MBF in nonsmokers and
smokers. The mean ΔMBF during the continuous performance test (CPT) in nonsmokers was 0.26 ± 0.18 mL/
min per gram. The mean ΔMBF during the CPT in smokers was 0.02 ± 0.20 mL/min per gram and was
significantly lower than in nonsmokers (P = .005). The mean percent change of MBF in smokers was signifi-
cantly lower than in nonsmokers (3.6 ± 19.7 vs. 30.7 ± 21.3%, P = .003). (From Ichikawa Y, Kitgawa K, Kato
S. Altered coronary endothelial function in young smokers detected by magnetic resonance assessment of
myocardial blood flow during the cold pressor test. Int J Cardiovasc Imaging. 2014;30[suppl 1]:73–80.)

its orifice or empties directly into the right atrium.29 If coronary sinus been recently confirmed by Aras and colleagues34 (coronary flow reserve
flow is divided instead by the mass of drained myocardium (as measured 1.45 vs. 3 in patients with chronic heart failure and healthy subjects,
from a stack of short axis images), the correlation remains good (r = respectively, P < .001). Similarly, Watzinger and coworkers35 have shown
0.95), whereas the slope approaches unity (1.05). Fig. 25.3 shows a significantly reduced flow reserve in patients with idiopathic cardio-
regression plot of myocardial blood flow (sinus blood flow per unit myopathy compared with healthy subjects (2.19 ± 0.77 vs. 3.51 ± 1.29,
mass of drained myocardium) measured by CMR and PET, with baseline P < .05). In all three studies, there were no significant differences in
flows normalized for rate-pressure product, together with a Bland- baseline flow between healthy subjects and those with disease (0.52 mL/
Altman plot. The mean differences between PET and CMR were 3.4% min per gram vs. 0.46 mL/min per gram, P = not significant [NS]; 0.83
± 12.8% at resting baseline and 3.9% ± 20.2% during hyperemia and ± 0.26 mL/min per gram vs. 0.85 ± 0.30 mL/min per gram, P = NS;
were not significant. The authors have subsequently used this technique and 0.55 ± 0.19 mL/min per gram vs. 0.48 ± 0.07 mL/min per gram,
to show that administration of 17β-estradiol over 3 months without P = NS).
progestin coadministration does not improve coronary flow reserve in These studies provide useful results, but have a number of important
postmenopausal women.30 A similar nonbreath-hold technique was limitations. Cardiac and respiratory motion results in blurring of the
used by Moro and colleagues at 3 T31 to show that women have a higher sinus, and the small number of pixels covering the sinus (typically five
increase in myocardial blood flow in response to cold pressor testing across the diameter in diastole) results in considerable partial volume
than men (0.73 ± 0.43 mL/g per minute vs. 0.22 ± 0.19 mL/g per minute, averaging in edge pixels, which is problematic for the accurate assess-
P = .0012). ment of sinus cross-sectional area and for the determination of mean
A validation of nonbreath-hold CMR-measured coronary sinus flow sinus flow velocity. As discussed, drainage of a variable part of the
has also been performed against flow probes in dogs by Lund and inferior and inferior-septal myocardium by the middle cardiac vein28
associates.32 In this study, the correlation between coronary blood flow results in underestimates of myocardial blood flow measured, and
(measured as the sum of left anterior descending [LAD] and circumflex although this estimate is an indicator of total coronary blood flow, no
[LCX] coronary flow) with flow probes against coronary sinus flow regional assessment of either flow or flow reserve is possible with this
measured with CMR was 0.98, with a nonsignificant mean difference technique.
of 3.1 ± 8.5 mL/min. Coronary sinus blood flow per unit mass of myo-
cardium was 0.40 ± 0.09 mL/min per gram (CMR) compared with 0.44 Total Coronary Flow Reserve From Measurements in
± 0.08 mL/min per gram (flow probes) and also was not significant. the Aortic Root
The authors went on to study patients with chronic heart failure and In 1993, it was proposed that coronary flow reserve might be derived
showed that coronary flow reserve was significantly reduced compared from flow measurements made in the ascending aorta, which is less
with that in healthy subjects (2.3 ± 0.9 vs. 4.2 ± 1.5, P = .01).33 This has susceptible to cardiac and respiratory motion and partial volume effects
312 SECTION II  Ischemic Heart Disease

6 r = 0.95 80

MBF (CMR-PET) (% of mean MBF)


P < .0001 60
5 Mean + 2 SD
40
CS flow/LVMdrain (mL/min/g)

20
4 Mean
0

3 −20

−40 Mean − 2 SD
2 −60

−80
1 0 1 2 3 4 5
B Mean MBF (mL/min/g)
y = 1.02x + 0.06
0 Baseline Hyperemia
0 1 2 3 4 5 6
A MBF PET (mL/min/g)
FIG. 25.3  (A) Correlation between estimate of myocardial blood flow (MBF) (mL/min per gram) derived from
positron emission tomography (PET) (x axis) and magnetic resonance (MR) (y axis) measurements. For
compressed sensing (CS) flow divided by drained myocardium, correlation with PET data is high and the
slope of the regression line approximates unity (dashed line). (B) Comparison of myocardial blood flow as
measured by PET and MR (coronary sinus flow divided by drained myocardium mass) with baseline flow
normalized for rate-pressure product (Bland-Altman analysis). Mean differences between PET and MR mea-
surements during baseline (3.4% ± 12.8%) and hyperemia (3.9% ± 20.2%) were not significantly different.
CMR, Cardiovascular magnetic resonance; LVM, left ventricular mass; SD, standard deviation. (From Schwitter
J, DeMarco T, Kneifel S et al. Magnetic resonance-based assessment of global coronary flow and flow
reserve and its relation to left ventricular functional parameters. Circulation. 2000;101:2696–2709.)

than the coronary arteries.36 Coronary diastolic flow, which represents


the bulk of coronary flow, can be estimated as the retrograde flow in Direct Assessment of Coronary Artery Velocity
the ascending aorta during systole and diastole minus the antegrade The problems associated with the assessment of flow velocity and coro-
flow during diastole. A variable velocity encoding window was imple- nary artery flow were discussed earlier and preclude the application of
mented to maintain the accuracy of velocity measurements during standard CMR techniques. As for coronary imaging, the major break-
periods of both high flow in systole (window = 200 cm/s) and low flow through for coronary flow techniques was the shortening of sequence
in diastole (window = 30 cm/s).37 Although it was suggested that the duration to the extent that data could be acquired over a single breath-
assessment of absolute diastolic coronary flow with this technique is hold, effectively freezing respiratory motion and eliminating the resulting
inaccurate because of known errors associated with the velocity mapping artifact. Further advances with navigator echo monitoring of the dia-
technique, it was argued that these errors should be the same both phragm position have allowed data to be acquired over multiple repro-
pre-vasodilation and postvasodilation and therefore should be eliminated ducible breath-holds or during free breathing, enabling increased spatial
from the assessment of diastolic coronary flow reserve, defined in this and temporal resolution and data averaging (resulting in higher SNRs).
instance as the difference (rather than the ratio) between the two mea- The following sections describe the approaches used to assess coronary
surements. In seven patients with abnormal findings on myocardial flow velocity and flow.
perfusion scintigraphy, the diastolic coronary flow reserve was −50 ±
76 mL/min compared with 260 ± 66 mL/min in eight healthy subjects. Bolus Tagging
The principle of this technique was later refined by taking into account The first report detailing the imaging of coronary artery flow was made
the motion of the coronary arteries during the cardiac cycle.38 A model in 1991 in rat and mice hearts with bolus tracking.40 With this technique,
was developed describing the flow through five transverse parallel aortic based on one previously implemented in vivo in the aorta and the
slices extending from the base of the aortic valve to above the level of carotid arteries,41 a section of blood above the coronary ostia on the
the coronary ostia. This was then solved mathematically, and in five aortic root is tagged by the application of a slice-selective presaturation
healthy subjects, it was shown that the standard error in the measure- radiofrequency pulse, and imaging is performed after a delay. During
ment of total coronary artery flow was approximately 90 mL/min, or this delay period, the tagged volume of blood washes into the coronary
30% of total coronary artery flow. The error in coronary flow reserve artery tree, where it is seen as a signal void. The mean velocity of the
is higher because the errors in baseline and maximal vasodilation flow tagged blood can be calculated from the degree of tag movement and
are additive. In addition, the assumption that flow is predominantly the wash-in delay time. The technique has also been developed for
diastolic, although true in healthy subjects, may not hold in the pres- multibolus tracking using stimulated echoes,42 and its application was
ence of disease, introducing further unknown errors into the technique. demonstrated in a 3-mm diameter tube with laminar flow and in an
A more recent attempt at measuring coronary artery blood flow in this isovolumetric perfused rat heart. Using this approach, an image of
way likewise found that the technique was compromised by poor repro- multiple boluses (typically three), each with a different wash-in time,
ducibility, although significant changes in coronary blood flow with can be obtained simultaneously to show the coronary artery tree. Again,
hormone replacement therapy were observed.39 the extent of the arterial pathway seen depends on the flow velocities
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 313

in the tagged volumes and on the wash-in delay times, with short delays planar time-of-flight coronary flow velocity reserve has been measured
required for visualization of the proximal portions and longer delays using this technique in healthy subjects (N = 10) after the infusion of
for the mid- and distal portions. By tracking multiple boluses simul- 0.56 mg/kg dipyridamole.44 In these subjects, peak diastolic velocity
taneously in this way, this technique essentially results in images of was observed to increase from 22 ± 7 cm/s to 90 ± 40 cm/s, resulting
blood flow. in a coronary flow velocity reserve of 3.9 ± 1.5, with velocity returning
to baseline (23 ± 5 cm/s) after the administration of aminophylline.
Echo Planar Time-of-Flight Technique In this study, the authors acquired low-resolution, single-shot coro-
Echo planar techniques are an attractive option for coronary artery nary images rather than using a segmented approach to build up higher-
investigations because of their fast imaging times, which reduce the resolution images over a number of cardiac cycles. This was prompted
effects of both cardiac and respiratory motion. In 1993, the first report by their observation of considerable variability in the beat-to-beat
detailing the use of an echo planar single-shot time-of-flight technique position of the LAD during both long and short breath-hold periods.
for assessing coronary artery flow velocity in 11 healthy subjects was This usually corresponded to a downward drift in vessel position as
reported.43 In this study, short axis cardiac slices were acquired with an the breath-hold continued and could be as much as 6 mm (i.e., greater
in-plane pixel size of 1.5 × 3 mm, each taking approximately 95 ms. than the vessel diameter). It was unclear, however, how much of this
Before the 90-degree slice selection radio frequency pulse, another movement was caused by poor breath-holding and how much was the
90-degree pulse was applied to saturate a thick band centered on the result of beat-to-beat variations in cardiac contraction. Regardless of
imaging slice. If increasing time delays are programmed between the the cause, it would introduce blurring, which is largely avoided in a
saturation and slice select pulses, the signal intensity in the vessel changes single-shot image. Beat-to-beat variations in flow, as opposed to spatial
as a result of blood wash-in through the slice. The rate at which it position, still have an effect on measured velocity because individual
changes can be used to calculate the blood velocity at the time of imaging. images must be acquired with different saturation delays in consecutive
The average velocity profile for the study group of 11 subjects is shown cardiac cycles to generate the wash-in curve. Therefore measured veloc-
in Fig. 25.4 and illustrates the expected peak flow in early diastole. ity is affected by changes over the breath-hold period. The importance
Using this technique, a separate breath-hold was required for velocity of this study lies in its pioneering and largely successful approach to a
assessment at each time point in the cardiac cycle and it was not pos- previously unsolved problem. It has largely been superseded by phase
sible to image at time points of <200 ms from the R-wave because this velocity mapping approaches, as discussed later, which are more robust
time period is required for the application of saturation delays. This and have higher availability.
was not considered a problem for normal subjects because left coronary
flow is predominantly diastolic. A further restriction of the technique Gradient Echo Phase Velocity Mapping
is that the long echo time of the sequence (echo time [TE] 28 ms) may Breath-Hold Techniques
lead to signal loss at sites of turbulent flow. At such sites, there may Using a velocity encoded segmented k-space gradient echo technique,
also be a breakdown in the assumption of laminar flow required for velocity maps may be acquired in a single breath-hold, thereby elimi-
the velocity calculations from the wash-in data. The authors reported nating respiratory motion artifact. To accomplish this, the segment
that in nine subjects who were imaged during continuous hand and duration is typically on the order of 100 ms, and to minimize blurring
lower extremity exercise, eight showed an increase in diastolic velocity as a result of cardiac motion, the acquisition is best performed in mid
(increase over exercise period = 52 ± 24%). Isometric exercise does not diastole, when the heart is relatively stationary. In addition to limiting
produce a maximal physiologic stress response, and more recently, echo the temporal resolution of the sequence, the need to perform data
acquisition within a single breath-hold also limits the number of phase
encoding steps that can be acquired, which in turn limits the spatial
resolution and the SNR in the resulting images.
40 The first diastolic through-plane coronary artery phase velocity maps
were reported by Edelman and coworkers in 1993.45 These were acquired
at 1.5 T using a fat-suppressed sequence consisting of a reference veloc-
30 ity compensated gradient waveform followed by a sensitized (velocity
window = 150 cm/s) gradient waveform, each repeated four times in
Vmax (cm/s)

a cardiac cycle. TE was 8 ms and repetition time (TR) was 15 ms, result-
ing in a segment duration of 120 ms. The sequence was validated against
20
a Doppler flow meter in vitro using both constant and mildly pulsatile
flow in an 8-mm diameter tube and in vivo against a standard non-
segmented velocity mapping sequence in the descending aorta of three
10 healthy volunteers. Velocity maps were acquired over 24 cardiac cycles
with in-plane pixel dimensions of 1.4 × 0.8 mm. In healthy subjects,
mean flow velocities at rest in the mid portion of the right and left
0 anterior descending coronary arteries were 9.9 cm/s and 20.5 cm/s,
0 100 200 300 400 500 600 700 800 900 respectively. These values are lower than those found in Doppler flow
studies, a finding that is to be expected because the small number of
Cardiac gate delay (ms)
pixels across the vessel results in partial volume averaging of the velocity
RR
interval profile. In four subjects who received intravenous administration of
FIG. 25.4  Time-of-flight coronary flow velocity profile in 11 healthy adenosine, velocities increased by at least a factor of four, suggesting
subjects. For each subject and for each gating delay, a series of images that CMR has the potential to assess the hemodynamic significance of
with different saturation delays was acquired. (From Poncelet BP, Weiss- a stenosis by measuring the flow response to vasodilation.
koff RM, Weeden VJ, et al. Time of flight quantification of coronary flow Another approach to quantifying the severity of stenosis is to perform
with echo-planar MRI. Magn Reson Med. 1993;30:447–457.) in-plane coronary artery velocity mapping with a view to measuring
314 SECTION II  Ischemic Heart Disease

increased velocity at the site of lumen narrowing. In-plane coronary occurring secondary to breath-holding, such as increases in intrathoracic
artery velocity mapping was first performed in healthy volunteers by pressure and heart rate, may themselves alter the blood flow being
Keegan and associates,46 using a sequence with TE of 10 ms, TR of measured. A navigator echo approach under either prospective or ret-
20 ms, and segment duration of 160 ms, which effectively limited acqui- rospective control would enable data to be acquired during free breathing
sitions to the period of relative cardiac diastasis in early diastole. The and would avoid these problems. Furthermore, temporal resolution of
in-plane resolution was 1.6 × 0.8 mm, and data were acquired over the sequence could be improved by reducing the segment duration,
breath-holds of 24 to 32 cardiac cycles. The velocity sensitivity used albeit at the expense of prolonged scan time.
was 50 cm/s and was achieved by phase map subtraction of two images, The influence of temporal resolution was studied by Hofman and
one sensitized so that flow velocities of 100 cm/s gave a phase shift of colleagues,47 who compared the use of a segmented breath-hold tech-
+2pi radians and the other sensitized so that flow velocities of 100 cm/s nique (segment duration of 126 ms) with a retrospective respiratory
gave a phase shift of −2pi radians. This was shown to result in fewer gated technique48 (reference and velocity sensitized view pair duration
blood flow artifacts than subtracting an image sensitized to flow veloci- of 32 ms) for the assessment of flow velocity, vessel cross-sectional area,
ties of 50 cm/s from a reference nonsensitized image. Through-plane and volume flow in the right coronary arteries of six healthy subjects.
and in-plane velocities measured with this technique were validated in Eight data averages were acquired, and the data were reconstructed
vitro against a standard nonsegmented velocity mapping sequence in with retrospective gating. The residual displacement of diaphragm posi-
a 5.6 mm diameter tube with pulsatile flow having a maximum velocity tions in the reconstructed data was 3.9 mm. In-plane spatial resolution
of 30 cm/s and a maximum rate of change of velocity of 126 cm/s2, was 0.8 × 1.6 mm, and velocity sensitivity was ±25 cm/s. Vessel regions
comparable with the values expected in normal human coronary arteries of interest were obtained semiautomatically from the magnitude images
at rest. Phantom work was also carried out to show the ability of the of the velocity compensated dataset by means of a seed growing algo-
technique to measure a velocity increase at the sites of mild, moderate, rithm and a 50% threshold. Velocity profiles generated from the ret-
and severe area-reducing stenoses and hence to quantify severity. However, rospectively gated data for all six subjects (uncorrected for through-plane
although velocity increases at the sites of area-reducing stenoses have velocity of the vessel itself) are shown in Fig. 25.5A and show a sharp
been observed with this approach,3 the tortuous pathways and small peak in systole, a minimum at end-systole, and a second peak in early
caliber of the coronary vessels result in partial-volume-type effects being diastole. The in-plane displacements of the vessels are shown in Fig.
more problematic for in-plane than for through-plane velocity mapping. 25.5B and typically show peaks in systole and early diastole, with minimal
Furthermore, in-plane studies require a high degree of reproducibility displacement in mid to end-diastole. The authors have since reported
in the breath-holding position, which is difficult to achieve without a similar pattern of movement for the LAD.49 During times of peak
techniques such as navigator echo monitoring. vessel displacement, motion blurring of the artery occurs and the breath-
hold segmented images are consequently of poorer quality than those
Navigator Techniques acquired with the retrospective respiratory gated short segment duration
The studies discussed earlier used breath-holding as a means of respira- sequence (Fig. 25.6B). In diastole, however, when in-plane displacement
tory motion control, limiting the sequence parameters to allow the of the vessel is low, the two techniques generate images of comparable
entire dataset to be acquired within the duration of a single breath-hold quality (see Fig. 25.6A). This results in the breath-hold segmented k-space
and requiring a high degree of patient cooperation, which is not always gradient echo sequence overestimating the instantaneous vessel cross-
forthcoming. In addition, intervariations and intravariations in the sectional area by as much as a factor of four, with an average increase
breath-hold position may be problematic, and hemodynamic changes in the time-averaged cross-sectional area of 90%. Based on this work,

30 5

25
4
20
Displacement (mm)

3
Velocity (cm/s)

15

10 2

5
1
0

–5 0
0 200 400 600 800 1000 0 200 400 600 800 1000

A Time (ms) B Time (ms)

FIG. 25.5  Through-plane cross-sectional averaged velocity (A) and in-plane displacement (B) in the right
coronary artery as a function of time in the cardiac cycle and as measured on respiratory gated acquisition
(oversampling factor = 8) in six subjects. Different symbols represent different subjects. (From Hofman MBM,
van Rossum AC, Sprenger M, et al. Assessment of flow in the right human coronary artery by magnetic
resonance phase contrast velocity measurement: effects of cardiac and respiratory motion. Magn Reson
Med. 1996;35:521–531.)
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 315

Magnitude image Velocity map

RRG

Breath-hold

A
Magnitude image Velocity map

RRG

Breath-hold

B
FIG. 25.6  Through-plane magnitude images and velocity maps for retrospective respiratory gated (RRG)
acquisitions (top) and breath-hold acquisitions (bottom) acquired (A) in mid diastole (gating delay of 760 ms)
and (B) in early systole (gating delay of 90 ms). In mid diastole, when in-plane displacement is low, good-
quality images are obtained with both techniques. In early systole, however, when in-plane displacement is
high, the long acquisition window of the breath-hold sequence results in considerable blurring of the artery
(arrows). (From Hofman MBM, van Rossum AC, Sprenger M, et al. Assessment of flow in the right human
coronary artery by magnetic resonance phase contrast velocity measurement: effects of cardiac and respira-
tory motion. Magn Reson Med. 1996;35:521–531.)

the authors suggest that, for acceptable levels of blurring throughout of interest determined. Both CMR techniques were found to significantly
the cardiac cycle, the acquisition window for studies of the left and underestimate flow velocity, although the correlations with invasive
right coronary arteries (RCAs) should be <90 ms and <25 ms, respec- measurements were strong (r = 0.70 and r = 0.86, respectively). The
tively. This is similar to the values derived by Marcus and coworkers,50 navigator echo controlled technique was significantly more accurate
who suggested an acquisition window of <58 ms and <23 ms for the than the breath-hold technique (P < .02), although this was at the
left and right arteries, respectively. expense of prolonged acquisition time. The underestimation of blood
A comparison between segmented k-space gradient echo phase veloc- flow velocity by both techniques is largely caused by partial volume
ity mapping and Doppler flow wire techniques has been performed in averaging of the spatial flow profile in the CMR studies, together with
26 angiographically normal coronary artery segments.51 Both breath- insufficient sampling of the temporal flow profile. The decreased accu-
holding and real-time slice-followed52 navigator echo controlled free racy of the breath-hold technique compared with the navigator echo-free
breathing CMR techniques (temporal resolution of 140 ms and 46 ms, breathing technique is likely to be caused by a combination of the
respectively) were performed within 24 hours of the invasive procedure longer acquisition window of the former, together with hemodynamic
and the maximal coronary flow velocities in a 2 × 2 mm pixel region changes resulting from the breath-holding procedure itself. Fig. 25.7
316 SECTION II  Ischemic Heart Disease

40 of rapid cardiac movement. This was due to misregistration of the


velocity encoded and reference datasets acquired consecutively in the
same cardiac cycle in the breath-hold acquisition, whereas in the free
Flow velocity (cm/s)

30
breathing acquisition, they were acquired at the same time point on
alternate cycles. In coronary blood flow studies, a region of adjacent
20
myocardium is generally used as a marker of through-plane motion
and is used to correct flow profiles. This is feasible for the left coronary
10
artery, but for the RCA, the adjacent myocardium is too thin to provide
a reliable correction. For this artery, epicardial fat surrounding the
0
artery has been used for through-plane correction, with the fat signal
0 200 400 600 provided from a fat-excitation image generated from a separate acquisi-
Time (ms) tion, or from one interleaved with the water-excitation image used for
imaging flow.58 Fig. 25.8 shows mean (± standard deviation [SD]) flow
FIG. 25.7  Original tracing of an invasively determined flow curve of the
left coronary artery compared with noninvasively determined flow curves profiles before and after correction in the left (N = 13) and right (N =
in the same patient. The solid line shows the real-time adaptive navigator 10) coronary arteries of healthy subjects. The corrected flow profiles
correction technique; the dotted line shows the breath-hold technique. bear strong resemblances to those found in normal arteries using Doppler
(From Nagel E, Bornstedt A, Hug J, et al. Noninvasive determination of flow wire.59
coronary blood flow velocity with magnetic resonance imaging: com- Brandts and colleagues developed a spiral phase velocity mapping
parison of breath-hold and navigator techniques with intravascular ultra- sequence at 3 T where the higher SNR allowed imaging with a spatial
sound. Magn Reson Med. 1999;41:544–549.) resolution of 0.8 mm × 8 mm, with a true temporal resolution of 33 ms
in a 24 cardiac cycle breath-hold.60 Using this technique, peak systolic
and peak diastolic velocities in repeated breath-holds in the right coro-
shows an example of the results obtained. Although still underestimat- nary arteries of healthy volunteers showed a high reproducibility (mean
ing flow velocities, the significantly improved accuracy of the navigator difference (±SD of paired differences) being 0 ± 16.5 mm/s and 10.5
echo-free breathing technique compared with the breath-hold technique ± 14.3 mm/s, respectively). A similarly high reproducibility in peak
provides a further step toward CMR assessment of coronary flow param- systolic and diastolic velocities in patients undergoing clinical coronary
eters, albeit at the expense of prolonged acquisition times. A navigator angiography for shortness of breath or chest pain was also shown by
gated phase velocity mapping sequence has also been developed at 3 T Keegan and associates (1.7 ± 15.8 mm/s and 2.5 ± 11.6 mm/s).61 In this
and validated in a realistic flow phantom (4 mm diameter, mean flow study, the spatial resolution (1.4 mm × 1.4 mm) was sacrificed for a
velocity 15–20 cm/s) with simulated respiratory motion.53 Using this higher temporal resolution (19 ms) and shorter breath-hold duration
technique, measured velocities were within 15% of actual velocities in (17 cardiac cycles) which enabled temporal details in the coronary
vitro and subsequent in vivo studies in 9 healthy volunteers resulted artery velocity profiles to be resolved. Fig. 25.9 shows the high degree
in time-averaged flow velocities of 6.8 ± 4.3 cm/s in the left anterior of inter–breath-hold reproducibility achieved with this technique in 5
descending artery, 8.0 ± 3.8 cm/s in the left circumflex artery and 6.0 right and 10 left coronary arteries. In a direct comparison with gold-
± 1.0 cm/s in the right coronary artery. standard Doppler guidewire studies in 23 vessels, CMR assessment of
peak systolic velocity, peak diastolic velocity and mean velocity through
Interleaved Spiral Phase Velocity Mapping the cardiac cycle underestimated Doppler values but the correlation
Interleaved spiral imaging is an alternative technique for generating between them was moderate to good (R2 = 0.57, 0.64, and 0.79, respec-
high-resolution images of the coronary arteries,54 which, compared tively) (Fig. 25.10). In addition, CMR measures of the peak diastolic
with those acquired using segmented gradient recalled echo (GRE), velocity to the peak systolic velocity showed a strong linear relation-
have higher SNRs and better temporal resolution.55 This potentially ship with Doppler values (R2 = 0.71 and 0.93 for left and right arteries,
enables velocity mapping in both the left, and the more mobile right, respectively) with a slope close to unity (0.89 and 0.90 for right and
coronary arteries.56 The results of breath-hold spiral, free breathing left arteries, respectively). In individual vessels, plots of CMR measured
spiral, and breath-hold segmented GRE phase velocity mapping have velocity at all cardiac phases against corresponding Doppler velocities
been compared with those of free breathing GRE in healthy volunteers.57 showed a consistent linear relationship between the two, with high R2
Eight left and eight right coronary arteries were studied and flow veloc- values (0.79 ± 0.13).
ity profiles generated with each technique, with the free breathing GRE The advantages of high SNR, good temporal and spatial resolution,
acquisitions being used as a gold standard. Flow profiles generated with and ability to implement fat suppression give the spiral technique con-
the free breathing spiral sequence (100-cardiac-cycle acquisition, assum- siderable promise, but more research is needed to investigate how the
ing 40% respiratory efficiency) agreed closely with those generated with blurring of off-resonance material62 and flow-direction-sensitive
the free breathing GRE sequence, taking 10 times longer to acquire for “implosion/explosion” artifacts in regions of poor homogeneity63 affect
the same spatial resolution. By comparison, the breath-hold segmented the measurements.
GRE sequence did not resolve the sharp peaks in the temporal flow
profiles of the RCA and, for the group as a whole, significantly under- Coronary Flow, Coronary Flow Reserve and Coronary
estimated peak systolic (88 mm/s vs. 252 mm/s, P < .001), peak diastolic Flow Velocity Reserve
(114 mm/s vs. 153 mm/s, P < .01) and mean (56 mm/s vs. 93 mm/s, P The assessment of coronary flow, rather than flow velocity, is more
< .001) velocities. For the less mobile left artery, peak systolic, peak difficult because of partial volume effects at the vessel boundary result-
diastolic and mean velocities were also underestimated by the breath- ing in overestimations of the vessel cross-sectional area.
hold segmented GRE sequence, but this only reached statistical signifi-
cance for the peak systolic velocity (80 mm/s vs. 135 mm/s, P < .01). Validation and Feasibility Studies
The breath-hold spiral sequence agreed reasonably well with the free The first directly validated measurements of coronary artery flow and
breathing GRE sequence, although deviations were observed at times coronary flow reserve using segmented GRE phase velocity mapping
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 317

300 Uncorrected 250 Corrected

250
200
Mean velocity (mm/s)

Mean velocity (mm/s)


200
150
150
100
100
50
50

0 0
0 100 200 300 400 500 600 700 0 100 200 300 400 500 600 700
−50 −50
Time after R-wave (ms) Time after R-wave (ms)
Separate acquisitions, mean Separate acquisitions, mean
Separate acquisitions, ± SD Separate acquisitions, ± SD
Combined acquisitions, mean Combined acquisitions, mean

A Left coronary arteries


250 Uncorrected 250 Corrected

200 200
Mean velocity (mm/s)

Mean velocity (mm/s)


150 150

100 100

50 50

0 0
0 100 200 300 400 500 600 0 100 200 300 400 500 600 700
−50 −50
Time after R-wave (ms) Time after R-wave (ms)

Separate acquisitions, mean Separate acquisitions, mean


Separate acquisitions, ± SD Separate acquisitions, ± SD
Combined acquisitions, mean Combined acquisitions, mean

B Right coronary arteries


FIG. 25.8  Mean (± standard deviation [SD]) flow velocities as measured by the separate water-excitation
and fat-excitation acquisitions in 10 left (A) and 13 right (B) coronary arteries both before (left) and after (right)
correction for through-plane motion. Mean flow velocities determined from the combined water-excitation/
fat-excitation acquisition are superimposed (open circles). (From Keegan J, Gatehouse PD, Yang GZ, et al.
Spiral phase velocity mapping of left and right coronary artery blood flow: correction for through-plane motion
using selective fat-only excitation. J Magn Reson Imaging. 2004;20:953–960.)

were performed by Clarke and coworkers in dogs in 1995.64 Nonmag- map was calculated. In this study, the size of the region of interest was
netic perivascular ultrasound probes were placed around the isolated kept constant from frame to frame and only its position changed because
LAD and LCX in eight ventilated dogs. A subcritical stenosis was gener- the authors believed that the spatial resolution in the CMR image was
ated in the LAD by placement of a polycarbonate resin constrictor. insufficient to track phasic changes in the coronary arterial diameter.
Breath-holding was effectively achieved by turning the ventilator off. The results of a typical experiment are shown in Fig. 25.11A, and a
Cine phase velocity mapping was performed using a sequence with TE plot of CMR flow reserve versus that measured by ultrasound is shown
of 11 ms and TR of 19.1 ms, with two to three reference and velocity in Fig. 25.11B. For the LCX and LAD, mean CMR-measured flow reserves
sensitized view pairs per data segment. The segment duration was were 2.57 ± 0.92 and 1.38 ± 0.31, respectively (P = .011), compared
therefore 76 to 115 ms, allowing the acquisition of four to six image with ultrasound-measured values of 2.55 ± 0.77 and 1.42 ± 0.31, respec-
frames over the cardiac cycle. Images were acquired with an in-plane tively (P = .002). The mean difference between the two techniques was
pixel size of 0.70 to 0.94 mm2 over breath-holds of up to 40 seconds 0.01, with limits of agreement of +0.64 and −0.61. The authors concluded
duration. The velocity sensitivity was ±138 cm/s. Data were acquired that, although the breath-hold period in this study is not feasible for
both before and after the administration of adenosine. A region of the majority of patients and both temporal and spatial resolution are
interest was drawn around the artery of interest in the magnitude image, limited, the results of the phase velocity mapping technique agreed well
and the mean velocity in that region on the corresponding velocity with Doppler ultrasound, suggesting that it could be used for accurate,
318 SECTION II  Ischemic Heart Disease

mm/s 120 120 120 120 120

mm/s

mm/s

mm/s

mm/s
80 80 80 80 80

40 40 40 40 40
RCA
0 0 0 0 0
0 250 500 750 0 250 500 750 0 250 500 750 0 250 500 750 0 250 500
−40 −40 −40 −40 −40

−80 Time (ms) −80 Time (ms) −80 Time (ms) −80 Time (ms) −80 Time (ms)

320 320 320 320 320


240 240 240 240 240
mm/s

mm/s

mm/s

mm/s

mm/s
160 160 160 160 160
LAD 80 80 80 80 80
0 0 0 0 0
0 250 500 750 0 250 500 750 0 250 500 7501000 0 250 500 750 0 250 500
−80 −80 −80 −80 −80
−160 Time (ms) −160 Time (ms) −160 Time (ms) −160 Time (ms) −160 Time (ms)

320 320 320 320 320


240 240 240 240 240
mm/s

mm/s

mm/s
mm/s

mm/s

160 160 160 160 160


LAD 80 80 80 80 80
0 0 0 0 0
0 250 500 750 0 250 500 750 0 250 500 7501000 0 250 500 750 0 250 500 7501000
−80 −80 −80 −80 −80
−160 Time (ms) −160 Time (ms) −160 Time (ms) −160 Time (ms) −160 Time (ms)

Initial breath-hold Repeat breath-hold


FIG. 25.9  Cardiovascular magnetic resonance velocity time curves in repeated breath-holds (red and green)
in 5 right coronary arteries (top) and 10 left coronary arteries (middle and bottom). LAD, Left anterior descend-
ing artery; RCA, right coronary artery. (From Keegan J, Raphael CE, Parker K, et al. Validation of high temporal
resolution spiral phase velocity mapping of temporal patterns of left and right coronary artery blood flow
against Doppler guidewire. J Cardiovasc Magn Reson. 2015;17:85.)

200 400 200


2
R = 0.57 R2 = 0.64 R2 = 0.79 100
CMR velocity (mm/s)

CMR velocity (mm/s)

CMR velocity (mm/s)

150 300
0
−1000 −600 −200 200
100 200 −100

−200
50 100

−300
0 0
0 200 400 600 0 400 800 1200 −400
Doppler velocity (mm/s) Doppler velocity (mm/s) Doppler velocity (mm/s)
A mean velocity (MV) B peak diastolic velocity (PDV) C peak systolic velocity (PSV)
FIG. 25.10  Cardiovascular magnetic resonance (CMR) measurement of mean velocity (MV) (A), peak diastolic
velocity (PDV) (B), and peak systolic velocity (PSV) (C) against Doppler values after scaling to same RR interval
in 23 vessels. (From Keegan J, Raphael CE, Parker K, et al. Validation of high temporal resolution spiral phase
velocity mapping of temporal patterns of left and right coronary artery blood flow against Doppler guidewire.
J Cardiovasc Magn Reson. 2015;17:85.)
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 319

Ultrasound coronary flow reserve


3
Coronary artery flow (mL/min)

180

140 2

100
1
60

20 Adenosine

0 20 40 60 80 100 0 1 2 3 4
A Time (minutes) B MRI coronary flow reserve
FIG. 25.11  (A) The typical experiment took approximately 90 minutes, with ultrasound measurements of
flow in the left circumflex artery (blue line) and left anterior descending artery (red line) being recorded
throughout. Two baseline cine velocity cardiovascular magnetic resonance (CMR) sets were acquired in the
left anterior descending artery (red circle) and left circumflex artery (blue square). Magnetic resonance velocity
cine image sets were recorded in each vessel during adenosine infusion. (B) Scatterplot of linear regression
of the 16 CMR-estimated and ultrasound-measured coronary flow reserve measurements produced a regres-
sion line with a slope of 1.04, an intercept of 0.1, and a correlation coefficient of 0.94. MRI, Magnetic reso-
nance imaging. (From Clarke GD, Eckels R, Chaney C, et al. Measurement of absolute epicardial coronary
artery blood flow and flow reserve with breath-hold cine phase-contrast magnetic resonance imaging. Circula-
tion. 1995;91:2627–2634.)

noninvasive measurement of absolute flow and flow reserve in the major velocity. In this study, volume flow data were not calculated because
epicardial coronary arteries. the authors believed that the small number of pixels across the vessel
The same technique was used to measure flow in the LAD in 12 prevented the accurate assessment of cross-sectional area. The average
subjects both before and after the administration of adenosine before baseline peak diastolic flow velocity was 14.8 cm/s, increasing to 46.3 cm/s
assessment with an intracoronary Doppler flow wire during cardiac after dipyridamole and resulting in an average coronary flow velocity
catheterization.65 In this study, with an in-plane pixel size of 0.8 to reserve of 3.1 ± 0.6. Interstudy reproducibility (i.e., absolute difference
1 mm, an in-plane presaturation pulse was applied before each cine in two measurements divided by the mean of two measurements
acquisition, suppressing the signal from tissue in the slice and enabling expressed as a percentage) was 9.5% and 6.8% prepharmacologic and
inflowing blood to be visualized with improved contrast. Depending postpharmacologic stress, respectively, with comparable interobserver
on the heart rate, four to five cine phases were acquired per cardiac reproducibility. A similar study performed at the same center in 12
cycle, with the number of phase encoding steps reduced to ensure com- healthy subjects using sustained handgrip exercise to induce vasodilation
plete data acquisition in a breath-hold of up to 25 seconds. A respiratory likewise showed an increase in peak diastolic flow velocity from 21 to
gating belt was used to ensure the consistency of the breath-hold posi- 31 cm/s.69
tions. Excellent agreement was reported between CMR and ultrasound Further validation against Doppler flow wire was found by Shibata
coronary flow and flow reserve measurements, with the limits of agree- and colleagues70 in a group of 19 patients scheduled for x-ray coronary
ment 43 and −27 mL/min and 0.5 and −0.3, respectively. angiography. Flow velocities measured with CMR were found to be
Sakuma and associates used a similar technique to validate CMR- significantly and considerably less than those measured by Doppler
measured coronary flow against findings obtained with an open-chest flow wire (12.5 ± 4.9 cm/s vs. 32.4 ± 12.9 cm/s, P < .01), although
sonographic flowmeter in seven dogs66 under suspended mechanical measures of coronary flow velocity reserve showed good correlation (r
ventilation. Good correlation was found (r = 0.95), with a slope close = 0.9), with no statistically significant difference. Fig. 25.12 shows
to unity and low interobserver variability (8%). The same authors used examples of flow velocity curves at baseline and after dipyridamole
CMR velocity mapping to study the velocity profiles in the LAD of injection in a patient with a normal LAD and in a patient with a sig-
eight healthy subjects both before and after dipyridamole administra- nificant stenosis. The flow curves in the stenosed artery show a <2-fold
tion.67 However, in this study, the use of view sharing enabled the effec- increase in peak diastolic flow velocity with dipyridamole, resulting in
tive temporal resolution of the imaging sequence to be improved to reduced coronary flow velocity reserve. The correlation between coronary
64 ms, and 7 to 13 cine images were acquired in an end-expiratory flow velocity reserve measured by Doppler guidewire and that measured
breath-hold of 24 heartbeats. Correction of coronary blood flow velocity by CMR is shown in Fig. 25.13. Bedaux and associates71 similarly found
for the through-plane velocity of the vessel as a whole was also per- that the coronary flow velocity reserve measured by CMR was not
formed by assuming that the velocity of the vessel was the same as that significantly different from that measured by Doppler flow wire in both
in an area of adjacent myocardium. This averages to zero over the healthy and diseased arteries (2.7 ± 1.0 vs. 3.1 ± 0.6 and 1.5 ± 0.7 vs.
cardiac cycle as a whole and is therefore unimportant for the assessment 1.9 ± 0.7, respectively). Validation of CMR measurement of left anterior
of mean coronary blood flow parameters, but may vary from as much descending coronary artery flow velocity reserve has also been performed
as +20 cm/s in systole to −10 cm/s in diastole.68 Consequently, it has against myocardial perfusion reserve in the anterior myocardium using
strong implications for the assessment of instantaneous flow and flow PET and 15O-labeled water.72 In 10 healthy subjects, coronary blood
320 SECTION II  Ischemic Heart Disease

30 8

CFR by guidewire average peak velocity


25 With dipyridamole 7
Velocity (cm/s)

20 6

15 5

10 4 Y = −0.48 + 1.446*X
At baseline R = 0.913
5 3 n = 19

0 2
0 100 200 300 400 500 600
1
A Delay after ECG trigger (ms)
0
0 1 2 3 4 5 6 7 8
35
CFR by CMR diastolic peak velocity
30 With dipyridamole FIG. 25.13  Cardiovascular magnetic resonance imaging (CMR; x axis) and
25 Doppler flow wire (y axis) measurements of coronary flow reserve (CFR)
Velocity (cm/s)

in 19 patients. Each symbol represents the data from one subject. The
20 regression line and the equation are shown. (From Shibata M, Sakuma
15 H, Isaka N, et al. Assessment of coronary flow reserve with fast cine
At baseline phase contrast magnetic resonance imaging: comparison with measure-
10 ment by Doppler guidewire. J Magn Reson Imaging. 1999;10:563–568.)
5
0
measured with volume flow and peak velocity, the most likely being
0 100 200 300 400 500 600 700 800
that the calculations made from peak velocity measurements are based
B Delay after ECG trigger (ms)
on single-pixel values with higher statistical noise, which, in this case,
FIG. 25.12  (A) Flow velocity curves in a patient with a normal left were uncorrected for the through-plane movement of the vessel itself.
anterior descending artery measured by cardiovascular magnetic reso-
Simultaneous flow mapping in the LAD and great cardiac vein has
nance (CMR) at baseline and after dipyridamole injection. Diastolic peak
velocity showed a threefold increase after the administration of dipyri-
also been performed by careful selection of the imaging plane and has
damole. (B) Flow velocity curves in a patient with a significant stenosis been used for vessel identification.74 These vessels run closely parallel
in the left anterior descending artery measured by CMR at baseline and to each other in the interventricular sulcus and can be difficult to dis-
after dipyridamole injection. Diastolic peak velocity showed a <2-fold tinguish. The results of flow mapping showed the expected diastolic-
increase after the administration of dipyridamole. ECG, Electrocardiogram. predominant flow in the LAD, whereas flow in the great cardiac vein
(From Shibata M, Sakuma H, Isaka N, et al. Assessment of coronary is predominantly systolic. These characteristic flow profiles clearly identify
flow reserve with fast cine phase contrast magnetic resonance imaging: the vessels and may be helpful in cases in which early intersection of
comparison with measurement by Doppler guidewire. J Magn Reson the great cardiac vein and the LAD make the distinction difficult.
Imaging. 1999;10:563–568.) For assessment of volume flow rate, accurate delineation of the
cross-sectional area of the coronary artery is essential. The majority of
studies use phase difference processing of the data, with a magnitude
flow velocity reserve measured with CMR was 2.44 ± 1.44 compared threshold of 30%26 or 50%.75 An alternative approach would be to use
with 2.52 ± 0.84 when measured with PET (P = NS), with a correlation complex difference processing, which takes into account partial volume
between the two measurements of 0.79. averaging of edge pixels. Given the small diameter of the coronary
In a further feasibility study, coronary volume flow profiles were arteries and the relatively large pixel size, this may be expected to perform
measured in normal subjects, with and without pharmacologic stress.73 better than phase difference processing.76,77 A study comparing complex
Again, by using view sharing techniques, 7 to 13 images could be obtained difference and phase difference (30% magnitude threshold) CMR veloc-
per cardiac cycle over breath-holds of up to 20 seconds, depending on ity mapping techniques in humans showed that the phase difference
the RR interval. In this study, the in-plane resolution was 0.9 × 1.4 mm. technique overestimated volume flow rates by approximately 25%.78
Regions of interest were drawn around the vessel, using the magnitude This has been studied in more detail by Wedding and colleagues,79 who
image as a guide. Within this region, automated edge detection was compared CMR measurements of coronary flow and flow reserve with
used to determine the exact vessel area, using a magnitude threshold those obtained from an ultrasonic transit time probe in dogs when
of 35%, and the region of interest was adjusted (in both position and using phase-difference processing (both 30%26 and 50% thresholds)
size) for each frame in the cardiac cycle. Three acquisitions were per- and complex-difference processing (both with and without correction
formed before dipyridamole administration, and one was performed for in-plane motion).80 They observed that mean CMR flow measure-
after dipyridamole administration. The mean area covered by the LAD ments correlated well with ultrasound data for all techniques (r = 0.92
was 15.6 mm2 before dipyridamole administration, increasing to to 0.94), but both the complex difference technique and the 50% mag-
17.7 mm2 after dipyridamole administration. Peak velocity, mean coro- nitude threshold phase difference technique systematically underestimated
nary flow velocity, and mean flow all increased significantly after dipyri- flow by 25% to 30%. Against expectations, the phase difference technique
damole administration, with coronary flow reserve of 5.0 ± 2.6 compared with the 30% magnitude threshold provided the best agreement with
with coronary flow velocity reserve of 3.5 ± 1.3. There are a number ultrasound, similar to that reported in other studies,66,67 but it was very
of potential reasons for the discrepancy between the coronary reserves sensitive to vessels within the boundary identification and the number
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 321

of pixels within it. Possible reasons for the unexpectedly poorer per- identified as having coronary flow reserve of 1.7 or less by CMR. The
formance of the complex difference technique were motion blurring sensitivity and specificity of CMR coronary flow reserve of 1.7 or less
of the vessel and poor SNR. for the identification of stenosis severity of >70% were 100% and 83%,
respectively. The correlation between Doppler flow wire and CMR
Patient Studies measures of coronary flow reserve was 0.87, with CMR underestimating
Despite the plethora of validation and feasibility studies that have been flow reserve by approximately 35%. The authors concluded that CMR-
performed, relatively few clinical studies have been reported. based measurement of coronary flow reserve may prove useful in iden-
The first report of using CMR coronary artery velocity mapping tifying patients who are likely to obtain a survival benefit from coronary
for the functional assessment of proximal and middle LAD stenoses in artery bypass grafting, where benefits are particularly high if there is
humans was by Hundley and coworkers.81 Thirty-three patients who involvement of the proximal LAD. A further report by the same authors
were referred for cardiac catheterization because of chest pain were discussed the assessment of coronary arterial restenosis after successful
examined with breath-hold CMR velocity mapping both before and percutaneous coronary arterial stenosis.82 In this study, 17 patients with
during intravenous administration of 140 µg/kg per minute adenosine. recurrent chest pain more than 3 months after intervention were studied
Coronary flow and coronary flow reserve were measured and compared by CMR, and it was found that a coronary flow reserve value of 2 or
with the results of computer-assisted quantitative coronary angiography less was 100% and 82% sensitive and 89% and 100% specific for detect-
and, where possible, Doppler flow wire (N = 17). All patients with ing luminal diameter narrowing of 70% or greater and 50% or greater,
stenosis severity of >70% by quantitative coronary angiography were respectively. Figs. 25.14 and 25.15 show examples of data in patients

CMR Coronary Angiograms

LAD
LAD

X-ray Coronary Angiogram


Vein

LM Stent
LM
Stent
LAD
LCX

LCX
Velocity Maps

Rest Stress Stenosis severity by QCA = 35%

CMR coronary flow reserve = 3.1


FIG. 25.14  Tangential (top left) and cross-sectional (top right) coronary artery cardiovascular magnetic reso-
nance (CMR) of the left anterior descending coronary artery (LAD). On the far right is a selected image from
the patient’s contrast coronary angiogram. On CMR and the contrast coronary angiogram, the location of
the intracoronary stent is shown. The arrow on the CMR cross-sectional image shows the corresponding
cross-sectional view of the LAD imaged 5 to 10 mm distal to the intracoronary stent. From these cross-
sectional images, the region of interest (ROI) is selected for calculating the cross-sectional area of the vessel.
The ROI is then transferred to velocity maps, in which the intensity of the grayscale of each pixel within the
ROI represents velocity (darker indicates higher velocity). The bottom row of images shows corresponding
velocity maps (cross-sectional images of vessels) from the patient at rest and after pharmacologic stress
with adenosine. On the right velocity map, darker pixels within black circles indicate higher velocities after
adenosine infusion. This patient had coronary flow reserve of 3.1 and 35% stenosis by quantitative coronary
angiography (QCA). LCX, Left circumflex; LM, left main. (From Hundley WG, Hillis LD, Hamilton CA, et al.
Assessment of coronary arterial restenosis with phase-contrast magnetic resonance imaging measurements
of coronary flow reserve. Circulation. 2000;101:2375–2385.)
322 SECTION II  Ischemic Heart Disease

CMR Coronary Angiograms

LAD
LAD X-ray Coronary Angiogram

LAD Stent
LM

Stent

LM
LCX

LCX
Velocity Maps

Stenosis severity by QCA = 57%

Rest Stress

CMR coronary flow reserve = 0.8


FIG. 25.15  Tangential (top left) and cross-sectional (top right) coronary artery cardiovascular magnetic reso-
nance (CMR) of the left anterior descending (LAD) coronary artery. On the far right is a selected image from
the patient’s x-ray coronary angiogram. On tangential CMR and the x-ray coronary angiogram, the location
of the intracoronary stent is shown. On the right velocity map, persistent gray pixels after adenosine admin-
istration in the patient indicate impaired coronary flow reserve (value = 0.8) that corresponded to the presence
of 57% stenosis by quantitative coronary angiography. LCX, Left circumflex; LM, left main; QCA, quantitative
coronary angiography. (From Hundley WG, Hillis LD, Hamilton CA, et al. Assessment of coronary arterial
restenosis with phase-contrast magnetic resonance imaging measurements of coronary flow reserve. Circula-
tion. 2000;101:2375–2385.)

without and with (respectively) a significant stenosis, as determined 2.0


by x-ray coronary angiography.
Two further studies have been performed to assess the detection of
restenosis after coronary artery stent implantation. In the first, serial 1.5
changes in CMR coronary flow velocity reserve were evaluated in 10
1.25
CMR–CFVR

patients with coronary artery disease who had undergone successful


elective stent implantation of a lesion in the LAD.83 Flow velocities were
1.0
measured distal to the stent every 4 weeks for 6 months, with follow-up
angiography performed 6 months after the initial intervention. Patients
without restenosis at follow-up angiography (N = 7) had normal coro-
nary flow velocity reserve during the 6 months of follow-up, with values 0.5
of 2.31 ± 0.30 at 1 month and 2.52 ± 0.25 at 6 months after stent
implantation. However, those with restenosis (N = 3) showed a signifi-
cant decrease, with values of 2.26 ± 0.49 at 1 month and 1.52 ± 0.09 0
at 6 months (P < .05). In a larger study, 38 patients were studied 24 <50 50−74 ≥75
hours after stent implantation and again 3 months later.84 Results were Cross-sectional area reduction (%)
compared with x-ray coronary angiography, and in 18 cases, results FIG. 25.16  Scattergram of cardiovascular magnetic resonance (CMR)
were compared additionally with the results of Doppler flow measure- coronary flow velocity reserve (CFVR) for different severities of coro-
ments. CMR measurements could be made in 75% of the patients nary artery stenoses. (From Nagel E, Thouet T, Klein C, et al. Noninva-
studied. At follow-up, coronary flow velocity reserve was 1.78 ± 0.16 sive determination of coronary blood flow velocity with cardiovascular
in vessels without coronary stenosis, 1.46 ± 0.22 in vessels with 50% magnetic resonance in patients after stent deployment. Circulation.
2003;107:1738–1747.)
to 74% stenosis, and 1.10 ± 0.22 in those with 75% or greater stenosis
(P < .05 between all groups), as shown in Fig. 25.16. A threshold
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 323

coronary flow velocity reserve of 1.2 resulted in sensitivity of 83% and 5.1% vs. −8.7% ± 6.3%, respectively).87 In a subsequent study in patients
specificity of 94% for the detection of stenoses of >75%. with mild coronary artery disease,88 the mean stress-induced change
In a more recent study of patients with pulmonary hypertension, in cross-sectional area was lower in patients with mild disease than in
van Wolferen and coworkers85 showed that peak systolic right coronary healthy subjects (−2.2% ± 6.8% vs. 13.5% ± 12.8%, P < .0001) and in
artery flow was lower than peak diastolic flow (1.02 ± 0.62 mL/s vs. these patients, stress-induced changes in area correlated inversely with
2.99 ± 1.97 mL/s, P < .001), whereas in normal subjects peak systolic coronary wall thickness (R2 = .53). In subjects with mild coronary artery
and diastolic right coronary artery flows were similar (1.63 ± 0.58 mL/s disease, the percentage increase in coronary blood flow with stress was
vs. 1.72 ± 0.93 mL/s, P = NS). The RCA systolic-to-diastolic flow ratio significantly less than that in healthy subjects (−4.3 ± 13% vs. 37.6 ±
and the mean flow per gram of right ventricular tissue were inversely 31.7%, P < .0001). In these patients, the stress-induced changes in flow
related to the right ventricular mass (R2 = .37 and .53, respectively) and were inversely related to the coronary wall thickness (R2 = 0.34). This
to the RV pressure (R2 = .69 and .32, respectively). study suggests that CMR can measure local functional and anatomic
Coronary artery area and flow assessment in response to isometric changes in the coronary artery that are closely related to arterial remod-
hand-grip exercise has been used for the noninvasive assessment of eling in early atherosclerosis, whereas a subsequent study confirms that
endothelial-dependent coronary vasoreactivity (Fig. 25.17). Using an the response to isometric handgrip exercise is mediated by nitric oxide.89
interleaved spiral breath-hold approach at 3 T, it was shown that in The same approach has been used to demonstrate reduced systemic
healthy volunteers, both coronary cross-sectional area and coronary endothelial function in the internal mammary arteries in patients with
blood flow increase with stress (both P < .0001), whereas in patients coronary artery disease.90
with coronary artery disease, both coronary artery cross-sectional area
and blood flow decrease with stress (P = .02 and P = .01, respectively).
In patients with severe single-vessel disease who had two studies—one
CONCLUSION AND FUTURE DEVELOPMENTS
in the severely diseased vessel and one in the contralateral mildly-diseased A number of validation and feasibility studies have shown that CMR
vessel—regional differences in response to stress were demonstrated techniques have the potential to assess coronary artery flow velocity,
with area and blood flow in the severely diseased vessel decreasing with flow, flow reserve, and flow velocity reserve, with the most commonly
stress (P =.01 and P = .02, respectively), whereas that in the mildly- used technique being segmented GRE phase velocity mapping. Although
diseased vessel was unchanged (Fig. 25.18).86 Repeated stress acquisitions a number of validation studies have shown good correlation of CMR
(separated by a 10-minute rest period) in healthy volunteers and in data with Doppler flow wire and ultrasound transit time techniques, the
patients with coronary artery disease showed that changes in cross- robustness of the technique must be improved and larger-scale valida-
sectional areas and changes in coronary flow in response to stress were tion studies need to be performed. The use of the interleaved spiral
both highly reproducible (healthy subjects: 14.8% ± 3.3% vs. 17.8% ± technique, with its short readout window and high temporal resolu-
3.6% and 44.3% ± 8.3% vs. 44.8% ± 8.1%, respectively; subjects with tion, may prove to be a preferable technique and has been shown to
coronary artery disease: −6.4% ± 2.0% vs. −5.0% ± 2.4% and −9.7% ± closely relate to Doppler flow wire measuresd63 and to allow reproducible

FIG. 25.17  Typical anatomical and flow-velocity encoded coronary images using magnetic resonance imaging
at rest and with sequential isometric handgrip stresses in a healthy subject. In image (A) A scout scan
obtained parallel to the right coronary artery (RCA) is shown together with the location for cross-sectional
imaging (white line). The white arrows show the region (corresponding to the cross-sectional location from
A) that was selected for analysis at rest (B), during the first handgrip stress (C) and second handgrip stress
(D). The white arrow in E shows a cross-section of the RCA that was selected for analysis of coronary flow
velocity measures in the healthy volunteer. The signal intensity is proportional to flow velocity with a black
signal indicating high velocity down through the imaging plane. In the view of the RCA (white arrow) at
baseline (E) and during the first handgrip stress (F) and second handgrip stress (G) the change in luminal
coronary signal intensity (increased blackness) indicates a proportional change in through-plane coronary flow
velocity. (From Hays AG, Stuber M, Hirsch GA et al. Noninvasive detection of coronary endothelial response
to sequential handgrip exercise in coronary artery disease patients and healthy adults. PLoS One 2013;8:e58047.)
324 SECTION II  Ischemic Heart Disease

80 being perpendicular to the direction of flow. As has been noted, the


Healthy effects of partial volume errors could potentially be reduced by using
Mild CAD a complex difference, rather than phase difference, approach to the
60 Severe CAD generation of velocity maps.78
2. Temporal resolution: The temporal resolution of the sequence used
% Change with stress

40 should be adequate to resolve the phasic velocity profile of coronary


P = .03 blood flow. This ability is determined by the number of views acquired
NS per data segment in a segmented GRE acquisition. For accurate
P = .01 *
20 * assessment of time-averaged parameters, measurements should be
† made throughout the entire cardiac cycle, with the accuracy increas-
* † † ing as the number of cine frames increases.95 For segmented GRE
0
acquisitions, these points favor the acquisition of small numbers of
views per data segment, although this leads to long breath-hold
−20 periods or the need for navigator free breathing techniques. Alter-
Area Velocity Flow natively, more rapid and efficient methods of k-space coverage may
be used, such as interleaved rectilinear echo planar or spiral tech-
−40 niques. A further reason to reduce the duration of data acquisition
FIG. 25.18  Percent change (mean ± standard deviation) in coronary is to minimize blurring of the vessel as a result of movement in the
artery area, peak diastolic coronary flow velocity, and blood flow during acquisition window, which leads to overestimation of the vessel
isometric handgrip stress for healthy subjects (n = 20 [open bars]) and cross-sectional area through partial volume averaging of edge pixels
patients with single-vessel coronary artery disease (CAD) (n = 10). Brack- and underestimation of mean flow velocity.
ets show comparisons between arteries with mild CAD (hatched bars) 3. Velocity sensitivity: The maximum velocity in normal coronary arter-
versus severe CAD (solid bars) within the same patients. P < .005,
ies at rest is typically <25 cm/s. Therefore the window used for
P < .001 versus healthy subjects. (From Hays AG, Hirsch GA, Kelle
S, Gerstenblith G, Weiss RG, Stuber M. Noninvasive visualisation of
resting studies should be narrow, approximately 50 cm/s, to allow
coronary artery endothelial function in healthy subjects and in patients measurement of these low velocities with maximum accuracy but
with coronary artery disease. J Am Coll Cardiol. 2010;56:1657–1665.) without aliasing. For maximal vasodilation studies, the window
should be increased accordingly.
4. Through-plane movement of the vessel: As has been noted, through-
noninvasive assessment of local coronary vasoreactivity.88–91 To sum- plane movement of the vessel through the cardiac cycle can affect
marize the requirements, any technique must take into account the instantaneous measurement of blood flow velocity and volume flow
following factors: in that vessel considerably, although time-averaged measurements
1. Spatial resolution: For the assessment of volume flow, spatial resolu- throughout the cardiac cycle are unaffected because the through-
tion must be sufficiently good to measure the cross-sectional area plane movement of the vessel averages to zero.
throughout the cardiac cycle and to minimize partial volume averag- In addition, beat-to-beat variations in the position of the artery and
ing of flow velocities in edge pixels. These partial volume effects of flow through the artery inevitably influence the results, and these
become larger as the vessel becomes smaller and the relative number variations may be exacerbated by breath-holding, particularly for long
of pixels in the boundary increases. The degree to which measured periods. Therefore a free breathing technique using navigator echoes
velocities are affected depends strongly on the relative magnitude may be the best approach and would also allow higher spatial and
of the stationary material included in the boundary pixels. Several temporal resolution images, albeit with reduced scan efficiency and
studies have investigated these effects in small vessels. Hofman and consequently longer scan duration. As discussed earlier, such a technique
associates77 compared phase contrast measurements of time-averaged has been shown to significantly improve the accuracy of CMR velocity
volume flow in the femoral arteries of dogs with measurements assessment compared with long segment duration breath-holding
made by an ultrasonic transit time meter and found that the pro- studies.52 The long scan times could potentially be reduced by using a
portional difference between the techniques was 0.8% when the real-time phase encode reordering technique whereby the most signifi-
number of pixels across the vessel diameter was just three. Tang and cant central lines of k-space are acquired with the diaphragm position,
colleagues91 found that, for both in vitro and in vivo studies, volume as measured by a navigator echo, within a narrow range and the out-
flow accuracy increases with resolution, as expected, and errors are ermost lines acquired with the diaphragm position within a larger range.
<10% when the ratio of pixel size to vessel radius is <0.5. This was These techniques have already been successfully applied to coronary
also seen by Wolf and coworkers, who showed an error of 9% for artery CMR.96,97 Furthermore, real-time slice-following may also be
five pixels across the vessel diameter.92 Sondergaard and colleagues implemented and could potentially result in improved scan efficiency
showed <18% error in measured flow rate for four pixels across the by allowing the use of larger navigator windows without a reduction
vessel diameter in vitro.93 Arheden and associates94 studied pulsatile in image quality.
flow in tubes with various spatial resolutions (but with a minimum
of five pixels across the tube diameter) and found that overestima-
tion of the size of the region of interest resulted in flow errors of
REFERENCES
<20%, provided that the imaging plane was within 10 degrees of A full reference list is available online at ExpertConsult.com
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 324.e1

REFERENCES 21. Kato S, Kitigawa K, Yoon YE, et al. Detection of diminished response to
cold pressor test in smokers: assessment using phase-contrast cine
1. Pennell DJ, Bogren H, Keegan J, Firmin DN, Underwood SR. Assessment magnetic resonance imaging of the coronary sinus. Magn Reson Imaging.
of coronary artery stenosis by magnetic resonance imaging. Heart. 2014;32:217–223.
1996;75:127–133. 22. Maroules CD, Chang AY, Kontak A, Dimitrov I, Kotys M, Peshock RM.
2. Torii R, Wood NB, Hughes AD, et al. A computational study on the Measurement of coronary flow response to cold pressor stress in
influence of catheter-delivered intravascular probes on blood flow in a asymptomatic women with cardiovascular risk factors using spiral
coronary artery model. J Biomech. 2007;40:2501–2509. velocity-encoded cine MRI at 3 Tesla. Acta Radiol. 2010;51:420–426.
3. Mohiaddin RH, Pennell DJ. Magnetic resonance imaging of flow in the 23. Koskenvuo JW, Sakuma H, Niemi P, et al. Global myocardial blood flow
cardiovascular system. In: Reichek N, ed. Cardiology Clinics. Philadelphia: and global flow reserve measurements by MRI and PET are comparable.
WB Saunders; 1998:161–188. J Magn Reson Imag. 2001;13:361–366.
4. Gatehouse PD, Keegan J, Crowe LA, et al. Applications of phase-contrast 24. Koskenvuo JW, Hartiala JJ, Knuuti J, et al. Assessing coronary sinus blood
flow and velocity imaging in cardiovascular MRI. Eur Radiol. 2005;15: flow in patients with coronary artery disease: a comparison of phase-
2172–2184. contrast MR imaging with positron emission tomography. Am J
5. Hood WB. Regional drainage of the human heart. Br Heart J. 1968;30: Roentgenol. 2001;177:1161–1166.
105–109. 25. Ferrigno M, Hickey DD, Liner MH, Lundgren CEG. Cardiac performance
6. van Rossum AC, Visser FR, Hofman MBM, et al. Global left ventricular in humans during breath-holding. J Appl Physiol. 1986;60:1871–1877.
perfusion: noninvasive measurement with cine MR imaging and phase 26. Schwitter J, DeMarco T, Kneifel S, et al. Magnetic resonance-based
velocity mapping of coronary venous outflow. Radiology. assessment of global coronary flow and flow reserve and its relation to
1992;182:685–691. left ventricular functional parameters. Circulation. 2000;101:2696–2709.
7. Ganz W, Tamura K, Marcus HS, et al. Measurement of coronary sinus 27. Pelc NJ, Herfkens RJ, Shimakawa A, Enzmann DR. Phase contrast cine
blood flow by continuous thermodilution in man. Circulation. magnetic resonance imaging. Magn Reson Q. 1991;7:229–254.
1971;54:181–195. 28. Bailes DR, Gilderdale DJ, Bydder GM, Collins AG, Firmin DN.
8. Canty JM, Brooks A. Phasic volumetric coronary venous outflow patterns Respiratory ordered phase encoding (ROPE): a method for reducing
in conscious dogs. Am J Physiol. 1990;258:H1457H1463. respiratory motion artefacts in MR imaging. J Comput Assist Tomogr.
9. Mirowitz SA, Lee JKT, Guterrez FR, Brown JJ, Eilenberg SS. Normal 1985;9:835–838.
signal-void patterns in cardiac cine MR images. Radiology. 1990;176: 29. Waller BF, Schlant RC. Anatomy of the heart. In: Schlant RC, Alexander
49–55. RW, eds. The Heart, Arteries and Veins. 8th ed. New York: McGraw-Hill;
10. Kawada N, Sakuma H, Yamakado T, et al. Hypertrophic cardiomyopathy: 1994:59–111.
MR measurement of blood flow and vasodilator flow reserve in patients 30. Schwitter J, Kozerke S, Bremerich J, et al. Oral administration of
and healthy subjects. Radiology. 1999;211:129–135. 17β-estradiol over 3 months without progestin co-administration does
11. Edelman RR, Wallner B, Singer A, Atkinson DJ, Saini S. Segmented not improve coronary flow reserve in post-menopausal women: a
TurboFLASH: method for breath-holding MR imaging of the liver with randomised placebo-controlled cross-over CMR study. J Cardiovasc Magn
flexible contrast. Radiology. 1990;177:515–521. Reson. 2007;9:665–672.
12. Foo TK, Bernstein MA, Aisen AM. Improved ejection fraction and flow 31. Moro PJ, Flavian A, Jacquier A, et al. Gender differences in response to
velocity estimates with use of view sharing and uniform repetition time cold pressor test assessed with velocity-encoded cardiovascular magnetic
excitation with fast cardiac techniques. Radiology. 1995;195:471–478. resonance of the coronary sinus. J Cardiovasc Magn Reson. 2011;13:54–62.
13. Gyllenhammar T, Fernlund E, Jablonowski R, et al. Young patients with 32. Lund GK, Wendland MF, Shimakawa A, et al. Coronary sinus flow
hypertrophic cardiomyopathy, but not subjects at risk, show decreased measurement by means of velocity encoded cine MR imaging: validation
myocardial perfusion reserve quantified with CMR. Eur Heart J by using flow probes in dogs. Radiology. 2000;217:487–493.
Cardiovasc Imaging. 2014;15:1350–1357. 33. Lund GK, Watzinger N, Saeed M, et al. Chronic heart failure: global left
14. Kennedy K, Dick A, Drangova M, et al. Magnetic resonance ventricular perfusion and coronary flow reserve with velocity-encoded
measurements of coronary flow reserve in heart transplant recipients: an cine MR imaging: initial results. Radiology. 2003;227:209–215.
exploratory study of the relationship to coronary angiographic findings. 34. Aras A, Anik Y, Demirci A, et al. Magnetic resonance imaging
J Cardiovasc Magn Reson. 2007;9:701–707. measurement of left ventricular blood flow and coronary flow reserve in
15. Kato S, Fukui K, Kawaguchi J, et al. Relationship between coronary flow patients with chronic heart failure due to coronary artery disease. Acta
reserve evaluated by phase-contrast cine cardiovascular magnetic Radiol. 2007;48:1092–1110.
resonance and serum eicosapentaenoic acid. J Cardiovasc Magn Reson. 35. Watzinger N, Lund GK, Saeed M, et al. Myocardial blood flow in patients
2013;15:106–116. with dilated cardiomyopathy: quantitative assessment with velocity-
16. Kato S, Saito N, Kirigaya H, et al. Impairment of coronary flow reserve encoded cine magnetic resonance imaging of the coronary sinus. J Magn
evaluated by phase contrast cine-magnetic resonance imaging in patients Reson Imaging. 2005;21:347–353.
with heart failure and preserved ejection fraction. J Am Heart Assoc. 36. Bogren HG, Buonocore MH. Measurement of coronary artery flow
2016;5(2). reserve by magnetic resonance velocity mapping in the aorta. Lancet.
17. Bloch KM, Carlsson M, Arheden H, Stahlberg F. Quantifying coronary 1993;341:899–900.
sinus flow and global LV perfusion at 3T. BMC Med Imaging. 37. Buonocore MH. Blood flow measurement using variable velocity
2009;9:9–21. encoding in the RR interval. Magn Reson Med. 1993;29:790–795.
18. Dandekar VK, Baumi MA, Ertel AW, Dickens C, Gonzalez RC, Farzaneh- 38. Buonocore M. Estimation of total coronary artery flow using
Far A. Assessment of global myocardial perfusion using cardiovascular measurements of flow in the ascending aorta. Magn Reson Med.
magnetic resonance of coronary sinus flow at 3 Tesla. J Cardiovasc Magn 1994;32:602–611.
Reson. 2014;16:24–33. 39. Sorenson MB, Fritz-Hansen T, Jensen HH, Pedersen AT, Ottesen B.
19. Cuypers J, Leirgul E, Samnoy S, et al. Assessment of coronary flow reserve Measurement of aortic blood flow by magnetic resonance below and
in the coronary sinus by cine 3T-magnetic resonance imaging in young above the origin of the coronary arteries in postmenopausal hormone
adults after surgery for Tetralogy of Fallots. Pediatr Cardiol. 2012;33: replacement therapy. J Cardiovasc Magn Reson. 2004;6:637–644.
65–74. 40. Burstein D. MR imaging of coronary artery flow in isolated and in vivo
20. Ichikawa Y, Kitgawa K, Kato S, et al. Altered coronary endothelial hearts. J Magn Reson Imaging. 1991;1:337–346.
function in young smokers detected by magnetic resonance assessment of 41. Edelman R, Mattle HP, Keefield J, Silver MS. Quantification of blood flow
myocardial blood flow during the cold pressor test. Int J Cardiovasc with dynamic MR imaging and presaturation bolus tracking. Radiology.
Imaging. 2014;30(suppl 1):73–80. 1989;171:551–556.
324.e2 SECTION II  Ischemic Heart Disease

42. Chao H, Burstein D. Multibolus stimulated echo imaging of coronary 62. Yudilevich E, Stark H. Spiral sampling in magnetic resonance imaging:
artery flow. J Magn Reson Imaging. 1997;7:603–605. the effect of inhomogeneities. IEEE Trans Med Imaging.
43. Poncelet BP, Weisskoff RM, Weeden VJ, Brady TJ, Kantor H. Time of 1987;MI-6:337–345.
flight quantification of coronary flow with echo-planar MRI. Magn Reson 63. Gatehouse PD, Keegan J, Firmin DN. Flow distortion and signal loss in
Med. 1993;30:447–457. spiral imaging. Magn Reson Med. 1999;41:1023–1031.
44. Firmin DN, Poncelet BP. Echo-planar imaging of the heart. In: Schmitt F, 64. Clarke GD, Eckels R, Chaney C, et al. Measurement of absolute epicardial
Stehling MK, Turner R, eds. Echo-Planar Imaging: Theory, Technique and coronary artery blood flow and flow reserve with breath-hold cine
Application. Berlin: Springer-Verlag; 1998:389–418. phase-contrast magnetic resonance imaging. Circulation.
45. Edelman RR, Manning WJ, Gervino E, Li W. Flow velocity quantification 1995;91:2627–2634.
in human coronary arteries with fast breath-hold MR angiography. 65. Hundley GW, Lange RA, Clarke GD, et al. Assessment of coronary arterial
J Magn Reson Imaging. 1993;3:699–703. flow and flow reserve in humans with magnetic resonance imaging.
46. Keegan J, Firmin D, Gatehouse PD, Longmore D. The application of Circulation. 1996;93:1502–1508.
breath hold phase velocity mapping techniques to the measurement of 66. Sakuma H, Saeed M, Takeda K, et al. Quantification of coronary artery
coronary artery blood flow velocity: phantom data and initial in vivo volume flow rate using fast velocity encoded cine MR imaging. Am J
results. Magn Reson Med. 1994;31:526–536. Roentgenol. 1997;168:1363–1367.
47. Hofman MBM, van Rossum AC, Sprenger M, Westerhof N. Assessment 67. Sakuma H, Blake LM, Amidon TM, et al. Coronary flow reserve:
of flow in the right human coronary artery by magnetic resonance phase non-invasive measurement in humans with breath-hold velocity encoded
contrast velocity measurement: effects of cardiac and respiratory motion. cine MR imaging. Radiology. 1996;198:745–750.
Magn Reson Med. 1996;35:521–531. 68. Karwatowski SP, Mohiaddin RH, Yang GZ, et al. Noninvasive assessment
48. Lenz GW, Haacke EM, White RD, et al. Retrospective respiratory gating: a of regional left ventricular long axis motion using magnetic resonance
review of technical aspects and future directions. Magn Reson Imaging. velocity mapping in normal subjects. J Magn Reson Imaging. 1994;4:
1989;7:445–455. 151–155.
49. Hofman MBM, Wickline SA, Lorenz CH. Quantification of in-plane 69. Globits S, Sakuma H, Shimakawa A, et al. Measurement of coronary
motion of the coronary arteries during the cardiac cycle: implications for blood velocity during handgrip exercise using breath-hold velocity
acquisition window duration for MR flow quantification. J Magn Reson encoded cine magnetic resonance imaging. Am J Cardiol.
Imaging. 1998;8:568–576. 1997;79:234–237.
50. Marcus JT, Smeenk HG, Kuijer JP, van der Geest RJ, Heethaar RM, van 70. Shibata M, Sakuma H, Isaka N, et al. Assessment of coronary flow reserve
Rossum AC. Flow profiles in the left anterior descending and the right with fast cine phase contrast magnetic resonance imaging: comparison
coronary artery assessed by MR velocity quantification: effects of with measurement by Doppler guidewire. J Magn Reson Imaging.
through-plane and in-plane motion of the heart. J Comput Assist Tomogr. 1999;10:563–568.
1999;23:567–576. 71. Bedaux WL, Hofman MB, Cock CC, Stoel MG, Visser CA, van Rossum
51. Nagel E, Bornstedt A, Hug J, et al. Noninvasive determination of coronary AC. Magnetic resonance imaging versus Doppler guidewire in the
blood flow velocity with magnetic resonance imaging: comparison of assessment of coronary flow reserve in patients with coronary artery
breath-hold and navigator techniques with intravascular ultrasound. disease. Coron Artery Dis. 2002;13:365–372.
Magn Reson Med. 1999;41:544–549. 72. Sakuma H, Koskenvuo JW, Niemmi P, et al. Assessment of coronary flow
52. Danias PG, McConnell MV, Khasigawala VC, et al. Prospective navigator reserve using fast velocity-encoded cine MR imaging. Validation study
correction of image position for coronary MR angiography. Radiology. using positron emission tomography. Am J Roentgenol.
1997;203:733–736. 2000;175:1029–1038.
53. Johnson K, Sharma P, Oshinski J. Coronary artery flow measurement 73. Davis CP, Liu P, Hauser M, et al. Coronary flow and coronary flow
using navigator echo gated phase contrast magnetic resonance velocity reserve measurements in humans with breath-hold magnetic resonance
mapping at 3.0 T. J Biomech. 2008;41:595–602. phase contrast velocity mapping. Magn Reson Med. 1997;37:537–544.
54. Meyer CH, Hu BS, Nishimura DG, Macovski A. Fast spiral coronary 74. Bedaux WLF, Hofman MBM, Visser CA, van Rossum AC. Simultaneous
artery imaging. Magn Reson Med. 1992;28:202–213. non-invasive measurement of blood flow in the great cardiac vein and
55. Keegan J, Gatehouse PD, Taylor AM, et al. Coronary artery imaging in a left anterior descending artery. J Cardiovasc Magn Reson. 2001;3:227–235.
0.5 Tesla scanner: implementation of real-time navigator echo controlled 75. Hofman MBM, Visser FC, van Rossum AC, Vink GQ, Sprenger M,
segmented k-space FLASH and interleaved spiral sequences. Magn Reson Westerhof N. In vivo validation of magnetic resonance blood volume
Med. 1999;41:392–399. flow measurements with limited spatial resolution in small vessels. Magn
56. Keegan J, Gatehouse PD, Yang GZ, Firmin DN. Interleaved spiral Reson Med. 1995;33:778–784.
cine coronary artery velocity mapping. Magn Reson Med. 2000;43: 76. Polzin JA, Alley MT, Korosec FR, et al. A complex-difference phase-
787–792. contrast technique for measurement of volume flow rates. J Magn Reson
57. Keegan J, Gatehouse PD, Mohiaddin RH, Yang GZ, Firmin DN. Imag. 1995;5:129–137.
Comparison of spiral and FLASH phase velocity mapping, with and 77. Polzin JA, Korosec FR, Wedding KL, et al. Effects of through-plane
without breath-holding, for the assessment of left and right coronary myocardial motion on phase difference and complex difference
artery blood flow velocity. J Magn Reson Imaging. 2004;19:40–49. measurements of absolute coronary artery flow. J Magn Reson Imaging.
58. Keegan J, Gatehouse PD, Yang GZ, Firmin DN. Spiral phase velocity 1996;1:113–123.
mapping of left and right coronary artery blood flow: correction for 78. Grist TM, Polzin JA, Bianco JA, Foo TKF, Bernstein MA, Mistretta CM.
through-plane motion using selective fat-only excitation. J Magn Reson Measurement of coronary flow and flow reserve using magnetic
Imaging. 2004;20:953–960. resonance imaging. Cardiology. 1997;88:80–89.
59. Ofili EO, Labovitz AJ, Kern MJ. Coronary flow velocity dynamics in 79. Wedding KL, Grist TM, Folts JD, et al. Coronary flow and flow reserve in
normal and diseased arteries. Am J Cardiol. 1993;71:3D–9D. canines using MR phase difference and complex difference processing.
60. Brandts A, Roes SD, Doornbos J, et al. Right coronary artery flow velocity Magn Reson Med. 1998;40:656–665.
and volume assessment with spiral k-space sampled breathhold 80. Frayne R, Polzin JA, Mazaheri Y, Grist TM, Mistretta CA. Effect of and
velocity-encode MRI at 3 Tesla: accuracy and reproducibility. J Magn correction for in-plane myocardial motion on estimates of coronary-
Reson Imaging. 2010;31:1215–1223. volume flow rates. J Magn Reson Imaging. 1997;7:815–828.
61. Keegan J, Raphael CE, Parker K, et al. Validation of high temporal 81. Hundley GW, Hamilton CA, Clarke GD, et al. Visualization and
resolution spiral phase velocity mapping of temporal patterns of left and functional assessment of proximal and middle left anterior descending
right coronary artery blood flow against Doppler guidewire. J Cardiovasc coronary stenoses in humans with magnetic resonance imaging.
Magn Reson. 2015;17:85. Circulation. 1999;99:3248–3254.
CHAPTER 25  Coronary Artery and Sinus Velocity and Flow 324.e3

82. Hundley WG, Hillis LD, Hamilton CA, et al. Assessment of coronary oxide: a noninvasive MRI test of coronary endothelial function. Am J
arterial restenosis with phase-contrast magnetic resonance imaging Physiol Heart Circ Physiol. 2015;308:H1343–H1350.
measurements of coronary flow reserve. Circulation. 2000;101:2375– 90. Iantorno M, Hays AG, Schär M, et al. Simultaneous noninvasive
2385. assessment of systemic and coronary endothelial function. Circ
83. Saito Y, Sakuma H, Shibat M, et al. Assessment of coronary flow velocity Cardiovasc Imaging. 2016;9:e003954.
reserve using fast velocity-encoded cine MRI for non-invasive detection 91. Tang C, Blatter DD, Parker DL. Accuracy of phase contrast flow
of restenosis after coronary stent implantation. J Cardiovasc Magn Reson. measurements in the presence of partial-volume effects. J Magn Reson
2001;3:209–214. Imaging. 1993;3:377–385.
84. Nagel E, Thouet T, Klein C, et al. Noninvasive determination of coronary 92. Wolf RL, Ehman RL, Riederer SJ, Rossman PJ. Analysis of systematic and
blood flow velocity with cardiovascular magnetic resonance in patients random error in MR volumetric flow measurements. Magn Reson Med.
after stent deployment. Circulation. 2003;107:1738–1747. 1993;30:82–91.
85. van Wolferen SA, Marcus JT, Westerhof N, et al. Right coronary artery 93. Sondergaard L, Stahlberg F, Thomsen C, et al. Accuracy and precision of
flow impairment in patients with pulmonary hypertension. Eur Heart J. MR velocity mapping in measurement of stenotic cross-sectional area,
2008;29:120–127. flow rate and pressure gradient. J Magn Reson Imaging. 1993;3:433–437.
86. Hays AG, Hirsch GA, Kelle S, Gerstenblith G, Weiss RG, Stuber M. 94. Arheden H, Saeed M, Tornqvist E, et al. Accuracy of segmented MR
Noninvasive visualisation of coronary artery endothelial function in velocity mapping to measure small vessel pulsatile flow in a phantom
healthy subjects and in patients with coronary artery disease. J Am Coll simulating cardiac motion. J Magn Reson Imaging. 2001;13:722–728.
Cardiol. 2010;56:1657–1665. 95. Clarke GD, Hundley WG, McColl RW, et al. Velocity-encoded, phase-
87. Hays AG, Stuber M, Hirsch GA, et al. Non-invasive detection of coronary difference cine MRI measurements of coronary artery flow: dependence
endothelial response to sequential handgrip exercise in coronary artery of flow accuracy on the number of cine frames. J Magn Reson Imaging.
disease patients and healthy adults. PLoS One. 2013;8:e58047. 1996;6:733–742.
88. Hays AG, Kelle S, Hirsch GA, et al. Regional coronary endothelial 96. Jhooti P, Keegan J, Gatehouse PD, et al. 3D Coronary artery imaging with
function is closely related to local early coronary atherosclerosis in phase reordering for improved scan efficiency. Magn Reson Med.
patients with mild coronary artery disease: pilot study. Circ Cardiovasc 1999;41:555–562.
Imaging. 2012;5:341–348. 97. Weiger M, Bornert P, Proksa R, Schaffter T, Haase A. A motion-adapted
89. Hays AG, Iantorno M, Soleimanifard S, et al. Coronary vasomotor gating based on k-space weighting for reduction of respiratory motion
responses to isometric handgrip exercise are primarily mediated by nitric artefacts. Magn Reson Med. 1997;38:322–333.
26 
Coronary Artery Bypass Graft Imaging and
Assessment of Flow
Constantin B. Marcu and Albert C. van Rossum

Since the report published in 1968 by Favaloro about the use of saphe- physiologic response to grafting during the intraoperative phase of
nous veins to restore coronary artery blood flow in 171 patients,1 a large CABG and which may be useful in predicting subsequent graft failure.11
number of coronary artery bypass grafting (CABG) procedures have Cardiovascular magnetic resonance (CMR) and coronary computed
been performed worldwide. In the United States alone, 219,000 patients tomography (CCT) are techniques which allow the direct evaluation
underwent a total of 397,000 CABG procedures in 2010.2 of bypass grafts patency.12-17 A unique feature of CMR is that in addi-
The left internal mammary artery (IMA) is frequently used as an tion to standard anatomic imaging, blood flow velocity and volume
arterial conduit to the left anterior descending (LAD) and its diagonal can be quantified within the grafts. Thus the true physiologic status of
branches. Other arterial conduits include the right IMA placed to the the functional unit represented by a graft and its recipient vessel can
right coronary artery (RCA) or LAD, the right gastroepiploic artery be determined noninvasively.18
to the RCA, or the use of free radial artery grafts. Reversed saphe-
nous veins are generally used for grafting to distal branches of the CARDIOVASCULAR MAGNETIC RESONANCE OF
RCA and circumflex coronary artery (LCX) or to diagonal branches of
the LAD.
BYPASS GRAFTS
The long-term results of aortocoronary bypass surgery depend Over the years, several CMR techniques have been introduced to evalu-
largely on the maintenance of graft patency.3 About 25% of venous ate aortocoronary bypass grafts (Table 26.1).19 The assessment strategy
grafts occlude within 1 year of surgery, whereas during the following 5 may include either anatomic (angiographic) or hemodynamic (flow
years there is a 2% annual occlusion rate, which increases to 5% yearly volume, velocities, flow reserve) evaluation or a combination of these
thereafter. Thus 50% to 60% of venous grafts are occluded after 10 modalities. This is usually combined in clinical practice with evaluation
years and only half of the remaining patent ones have no evidence of of the myocardial function (cine CMR), tissue characterization (late
atherosclerosis.4,5 The mechanisms responsible for venous graft occlu- gadolinium enhancement imaging for presence of myocardial fibrosis/
sion are believed to be thrombosis in the early weeks after surgery, infarction), and ischemia detection (pharmacologic first pass gadolinium
followed by intimal hyperplasia during the first year and accelerated contrast myocardial perfusion).
atherosclerosis in the later stages.6 Atherosclerotic changes develop Whereas pulse sequences developed for imaging native coronary
comparatively in a smaller percentage of patients with IMA grafts. As arteries are also applied for imaging bypass grafts, CABG imaging is
a result, in situ arterial grafts occlude less frequently, up to 5% in the first associated with specific problems (different vessel anatomy and flow
year and 20% to 30% after 10 years, leading to an improved long-term patterns and the presence of metallic vascular clips, ostial markers and
survival.5,7 sternal wires).
These graft attrition statistics result in the need for accurate evalu- A majority of published clinical CMR studies addressed imaging
ation of bypass graft patency and function, which often is required the proximal portion of vein grafts. Proximal segments are less affected
several times during the lifetime of any given patient. by bulk cardiac motion compared with distal graft segments or native
coronary arteries, resulting in fewer motion artifacts, whereas the
IMAGING MODALITIES CAPABLE OF lack of direct contact with epicardial fat or myocardium results in
higher contrast to surrounding tissues. Unfortunately, graft stenosis
EVALUATING GRAFTS often occurs at the site of anastomosis with the native vessel where
Selective x-ray coronary angiography is the gold standard for assessment CMR encounters artifacts and resolution problems similar to those
of graft anatomy and has the added advantage of simultaneous visu- in imaging the native coronary arteries. Arterial grafts imaging poses
alization of the native coronary arteries. Use of a Doppler-tipped guide- additional problems because of vessel tortuosity and smaller caliber
wire during angiography provides hemodynamic information about as well as presence of metallic artifacts from hemostatic clips and
graft function by assessing flow pattern at rest and after pharmacologi- sternal wires.
cally induced hyperemia.8
Selective coronary angiography is, however, invasive, uses ionizing BYPASS GRAFT ANATOMIC
radiation, iodine contrast, and has a small risk of complications such IMAGING TECHNIQUES
as coronary artery dissection, arrhythmia or stroke.
Two-dimensional (2D) Doppler echocardiography is restricted to Conventional Spin Echo and Gradient Echo Imaging
evaluation of grafts placed on the LAD artery.9,10 The assessment of saphenous vein aortocoronary bypass graft patency
Near infrared fluorescence complex angiography and perfusion has been one of the early indications for CMR. More than two decades
analysis is a novel real-time imaging technology used to assess the ago several groups reported the feasibility of assessing graft patency using

325
326 SECTION II  Ischemic Heart Disease

TABLE 26.1  Detection of Bypass Graft Patency According to Different Cardiovascular


Magnetic Resonance Techniques
Reference Technique No. of Grafts Sensitivity (%) Specificity (%) Accuracy (%)
20
White et al. SE CMR 65 91 72 86
Rubinstein et al.21 SE CMR 44 92 85 89
Jenkins et al.22 SE CMR 60 90 90 90
Frija et al.23 SE CMR 52 98 78 94
White et al.24 Cine CMR 28 93 86 89
Aurigemma et al.25 Cine CMR 45 88 100 91
Galjee et al.26 SE CMR 98 98 85 96
Cine CMR 98 88 96
Combined 98 76 94
Kessler et al.33 3D navigator 19 87 100 89
Vrachliotis et al.38 3D CE MRA, ECG-triggered 44 93 97 95
Wintersperger et al.37 3D CE MRA Non–ECG triggered 76 95 81 92
Kalden et al.30 HASTE 59 95 93 95
3D CE MRA ECG-triggered 93 93 93
Bunce et al.40 SSFP 79 84 45 78
3D CE MRA ECG-triggered 85 73 84
Langerak et al.35 3D navigator Two observers 56 65–83 80–100 ~80

CE MRA, Contrast-enhanced magnetic resonance angiography; ECG, electrocardiogram; HASTE, half-Fourier acquisition single-shot turbo spin
echo; SE CMR, spin echo cardiovascular magnetic resonance; SSFP, steady state free precession.

conventional electrocardiogram (ECG)-triggered multislice spin-echo Another 2D breath-hold approach uses a multislice half-Fourier
techniques.20–23 On spin-echo images, patent grafts with good blood flow acquisition single shot turbo spin echo (HASTE) sequence. Seven
appear in consecutive imaging planes as conduits with a signal void. T2-weighted images are generated within a breath-hold of approximately
In contrast, stenotic grafts with slow flow or occluded grafts display 15 seconds, with a 1.3 × 1.4 mm in-plane resolution and 5 mm slice
intermediate signal intensity (Fig. 26.1). With selective x-ray angiog- thickness. Using this technique, Kalden and colleagues reported a sen-
raphy as the method of reference, the sensitivity of spin-echo CMR in sitivity and specificity of 95% and 93%, respectively, in predicting graft
predicting graft patency ranged from 90% to 98% with a specificity patency and good visualization of 83% of distal graft segments.30 Because
of 72% to 90%. the HASTE sequence is less susceptible to metallic artifacts compared
Using conventional gradient echo nonbreath-hold CMR, with a with gradient echo sequences, a remarkably high accuracy was found
relatively long echo time (TE) and repetition time (TR), the sensitivity in detecting arterial graft patency (sensitivity of 90% and specificity of
was in the same order of magnitude (88%–98%), with a somewhat 100%). However, these figures must be interpreted with caution because
higher specificity (86%–100%).24-26 On gradient echo images blood the pretest probability of occluded arterial grafts is generally low. In
flow within patent grafts appears bright (Fig. 26.2). On spin-echo images, that study, it is unclear how many of the 14 reported IMA grafts were
signal voids from metal clips, stents or calcifications, can falsely mimic occluded. In our personal experience, the HASTE sequence provides
graft patency. These artifacts are accentuated, thus easier to detect, on excellent visualization of coronary arteries and bypass grafts but rather
gradient echo compared with spin-echo images, which decreases the poor detection of disease (Fig. 26.4). Accordingly, Kalden and colleagues
number of false positive patent grafts (increases specificity). On the noted that only two out of eight hemodynamically significant graft
other hand, the number of false-positive occlusions might be expected stenoses were detected using HASTE sequence.
to increase (decreased sensitivity).
Three-Dimensional Respiratory-Gated Cardiovascular
Two-Dimensional Breath-Hold Cardiovascular Magnetic Resonance Angiography
Magnetic Resonance Angiography A three-dimensional (3D) dataset of truly contiguous slices may be
This technique, first described for imaging coronary arteries, can also be obtained with respiratory-gated gradient echo techniques. Gating to
applied for visualization of bypass grafts.27 During a breath-hold lasting the respiratory cycle is achieved by using navigators that monitor the
for 15 to 25 seconds, a segmented gradient echo image is acquired with diaphragm position.31 Magnetic resonance (MR) data acquired within
4 to 5 mm slice thickness and in-plane resolution of approximately 1 a preset acceptance window of the respiration-induced diaphragm
× 1.4 mm. The entire course of a bypass graft is covered by repeat- excursion are used for image reconstruction. The patient is allowed to
ing the sequence multiple times. The procedure may become rather breathe freely, at the expense of an increase in total imaging time. Several
time consuming when multiple grafts have to be evaluated. Also, a refinements of this gating procedure have been developed for imaging
high susceptibility to metallic materials used during surgery has been of the coronary arteries.32
reported, with significant effects on diagnostic accuracy.28 This tech- Kessler and colleagues used respiratory-gated 3D MR angiography
nique has been used for evaluation of sequential grafts (Fig. 26.3).29 (MRA) with navigator guiding and retrospective data processing for
High sensitivity and specificity were found for predicting patency of imaging of bypass grafts.33 In a relatively small number of 19 grafts, 13
grafts to branches of the RCA, but the accuracy decreased in distal graft out of 15 patent grafts and 4 out of 4 occluded grafts were correctly
segments to branches of the LAD and was poor in distal segments to classified. Another study by Molinari and colleagues including 18 patients
the LCX.29 (51 grafts) which compared navigator guided 3D MRA with coronary
CHAPTER 26  Coronary Artery Bypass Graft Imaging and Assessment of Flow 327

1
1

A B

1
2 2

C D
FIG. 26.1  Series of four transverse spin echo images of a sequential vein graft to the obtuse marginal branch
of the circumflex (CX) artery (1) and a second vein graft to the left anterior descending artery (LAD) (2).
Patent grafts show low signal intensity. (A) Most superior image demonstrating the CX graft originating from
the ascending aorta and overriding the main pulmonary artery. (B) More descending course of the CX graft
left of the pulmonary artery. (C) LAD graft originating from the more proximal part of the ascending aorta.
(D) Course of both grafts at the level of the left atrium.

angiography showed a sensitivity of 91%, the specificity of 97% for a total thickness of approximately 9 cm is imaged. Before acquiring the
CMR in detecting occlusion versus patency of grafts.34 3D volume slab, a single-slice 2D turbo gradient echo sequence is used
A study by Langerak and colleagues including 38 post-CABG patients to time the arrival of a contrast agent test bolus in the aorta. To maxi-
(56 venous grafts) who underwent conventional angiography for recur- mize the contrast-enhancing effect, acquisition of the central k-space
rent angina, assessed the accuracy of contemporary high-resolution lines of the 3D imaging data is set to coincide with peak contrast arrival.
respiratory-gated 3D MRA in detecting vein graft disease.35 The sequence This is achieved by introducing a time delay between contrast injection
used in the study included a T2-preparation prepulse (for muscle sup- and start of the imaging sequence (delay = arrival time − 1 2 or 1 3 of
pression), a fat suppression prepulse and had an acquisition window acquisition time). Depending on the field of view, matrix size, number
of 71 ms placed in mid diastole and at end expiration. A total of 50 of partitions and slab thickness a spatial in-plane resolution of 1 ×
out of 56 grafts were adequately visualized with a spatial resolution of 1.5 mm with 2- to 3-mm section thickness is achieved. Each partition
0.7 × 1 × 1.5 mm. The study reported sensitivities of 73% and 83%, of the 3D acquisition yields a “source” image. Studies are evaluated by
with specificities of 80% to 87% and 98% to 100% in detecting grafts reviewing the source images and after postprocessing techniques such
with ≥70% diameter stenosis and complete occlusion, respectively. as maximum intensity projection and planar or curved reformatting
(Figs. 26.5 and 26.6).19
Three-Dimensional Contrast-Enhanced Breath-Hold Evaluation of aortocoronary bypass grafts using 3D contrast-enhanced
Cardiovascular Magnetic Resonance Angiography MRA has been reported without and with ECG triggering.30,37,38 Sen-
Breath-hold contrast-enhanced MRA is another technique which was sitivity, specificity, and accuracy for predicting graft patency varied
first applied for aorta imaging.36 The T1-shortening effect of the between 93% and 95%, 81% and 97%, and 92% and 95%, respectively
gadolinium-contrast agent on blood signal allows the obtaining of high (Table 26.1). A lower specificity was found in the non–ECG triggered
vascular contrast when using short TR/TE gradient echo sequences. study.37 Theoretically, one would expect ECG-triggered acquisitions to
After intravenous injection of an interstitial gadolinium-contrast agent, be superior for visualizing the sites of graft insertion on native coronary
a 3D spoiled gradient echo sequence with short TR/TE (4.4/1.4 ms or arteries. Although Kalden and colleagues specifically addressed this issue,
even shorter) is applied. Within a breath-hold of approximately 30 they succeeded in pursuing distal anastomoses in only 64% of cases.30
seconds, a 3D volume slab composed of 24 to 32 contiguous slices with This might have been caused partly by the relatively long data collection
328 SECTION II  Ischemic Heart Disease

window of 560 ms within each cardiac cycle, leading to residual blur-


ring attributed to cardiac motion. The use of a shorter acquisition
window of 120 ms, used for imaging coronary arteries of healthy vol-
unteers,39 has not been implemented yet in imaging bypass grafts.
Bunce and colleagues compared 3D contrast-enhanced MRA with
LAD the recently developed multislice balanced steady-state free precession
RCA A P (bSSFP) sequence for the assessment of 56 venous and 23 arterial bypass
grafts patency in 25 patients.40 Steady-state free precession (SSFP) had
a temporal resolution of 336 ms, with a spatial resolution of 2.7 × 1.4
CX × 4.0 mm, and 3D contrast-enhanced MRA had a temporal resolution
of 440 ms with spatial resolution of 1.6 × 1.6 × 2.0 mm. The two methods
had similar accuracy (78% vs. 84%, P = nonsignificant) for detection
of coronary artery bypass graft patency, but there was a trend toward
more false-positive findings for occlusion and reduced visualization of
arterial grafts with SSFP angiography.40

IMAGING STRATEGY
The imaging strategy and image interpretation are facilitated when the
surgical report is known before the CMR scan with respect to the number
of grafts and insertion sites. Grafts descending to the perfusion area of
the LCX, including obtuse marginal branches, generally originate most
FIG. 26.2  Transverse image obtained with gradient echo technique.
superiorly from the ascending aorta (Fig. 26.7).27 The graft to the LAD
Cross sections of patent grafts to the right coronary artery (RCA), left
anterior descending coronary artery (LAD), and circumflex coronary artery system, including diagonal branches, originates inferiorly compared
(CX) are indicated and demonstrate a high signal intensity. A, Aorta; P, with most LCX grafts. Both types of grafts cross the pulmonary artery
pulmonary artery. (From van Rossum AC, Galjee MA, Post JC, Visser trunk in a left lateral and inferior course. Then the LCX graft proceeds
CA. A practical approach to CMR of coronary artery bypass graft patency posteriorly and inferiorly, whereas the LAD graft goes left and anteriorly.
and flow. Int J Cardiac Imaging. 1997;13:199–204, with permission from Grafts to the posterior descending artery of the RCA generally have the
Kluwer Academic Publishers.) lowest origin from the ascending aorta and run inferiorly on the lateral

A B

FIG. 26.3  (A–C) Segmented gradient echo tech-


nique, 1 image per breath-hold of 16 heartbeats.
Patient with sequential graft from the aorta to the
diagonal branch of the left anterior descending
coronary artery (LAD) and obtuse-marginal branch
of the left anterior descending coronary artery and
left internal thoracic artery graft to the LAD. The
tall arrow indicates bright segments of the sequen-
tial graft. The short arrow points at the dark signal
loss of the ITA clip artifact. (From van Rossum
AC, Bedaux WLF, Hofman MBM. Morphologic
evaluation of coronary artery bypass conduits. J
C
Magn Reson Imaging. 1999;10:734–740.)
CHAPTER 26  Coronary Artery Bypass Graft Imaging and Assessment of Flow 329

1 3

A B

2
C D
FIG. 26.4  Half-Fourier acquisition single shot turbo spin echo sequence, four out of five images obtained
in a single breath-hold (A, B, C, D). Patient with single vein graft to the right coronary artery (1), sequential
vein graft to the first diagonal branch of the left anterior descending coronary artery (LAD) and obtuse marginal
branch of the circumflex coronary artery (2), and the left internal thoracic artery to the LAD (3). In panels A
and B, the native left main coronary artery, the LAD, and the great cardiac vein are visualized. In panels C
and D, the right coronary artery is seen inferior to the graft in the right atrioventricular groove.

side of the right atrium. For assessment of patency and measurement and may be subject to misregistration due to inconsistent breath-holding.
of flow within a graft, CMR can best be performed at the proximal part Alternatively, 3D MRA techniques may be used to acquire larger imaging
of the graft. At this level, most grafts have a straight course, less motion, slabs covering the course of the grafts. The resolution is higher, and
and are easily distinguished from native coronary vessels. The surface the acquisition time is shorter. However, the reconstruction time is
coil must be centered at the mid sternum level, which is somewhat longer than in 2D techniques, and interpretation requires some form
more cranial compared with a standard cardiac study. of postprocessing. Notwithstanding these limitations, the 3D approaches
Since the widespread use of k-space segmented sequences and phased- are likely to become the first choice with improving technology.
array coils, most images can be obtained within a breath-hold. This
decreases respiratory motion artifacts and increases image quality. CARDIOVASCULAR MAGNETIC RESONANCE
Because the course of a graft is more or less predictable, the choice QUANTIFICATION OF GRAFT FLOW AND
of imaging planes can be fairly well standardized. An effective approach
is first to acquire a set of multislice transverse images covering the
FLOW RESERVE
ascending aorta and superior part of the heart. In our experience, one Flow velocity and volume flow in bypass grafts can be measured by
or two breath-hold multislice series using the HASTE sequence will be applying velocity-encoded phase-contrast cine CMR sequences, thus
most informative (see Fig. 26.4). The images just superior of the pul- allowing the assessment of graft function in addition to a morphologic
monary artery trunk will demonstrate in-plane views of proximal parts evaluation (Fig. 26.8).41 Galjee and colleagues demonstrated that adequate
of LCX and LAD grafts, whereas the images at a lower level show cross velocity profiles throughout the cardiac cycle could be obtained in 85%
sections of grafts to the left and right coronary arteries. Once the proxi- of angiographically patent vein grafts, using nonbreath-hold CMR
mal course of the grafts has been localized, one may proceed with velocity mapping.26 Graft flow velocity was characterized by a biphasic
high-resolution single or multislice 2D imaging, in orientations following pattern, with systolic and diastolic peaks (Fig. 26.9).26 Similar findings
the more distal course of the grafts. The advantages of this approach have been obtained by using invasive Doppler guidewire approaches
are the short image reconstruction time and the immediate availability and transthoracic Doppler echocardiography.8,9 However, there are dif-
of the images. The disadvantage is that it requires patient cooperation ferences in resting phasic flow patterns between in situ IMA conduits
330 SECTION II  Ischemic Heart Disease

FIG. 26.5  Gadolinium-enhanced three-dimensional breath-hold magnetic resonance angiography, electrocar-


diogram-triggered. The six images on the left are several of the source images, demonstrating the proximal
course of two sequential vein grafts to the left anterior descending coronary artery and circumflex coronary
artery perfusion territory, respectively. The right image is obtained through postprocessing, using a maximum-
intensity projection.

and saphenous vein grafts that may have implications in the develop- feasibility of measuring flow and flow velocity reserve in saphenous
ment of degenerative graft disease and long-term conduit patency.8 vein grafts and its potential to differentiate between patent and stenotic
Thoracic artery grafts show a gradual longitudinal transition from bypass grafts or runoff vessels.42-48 A report by Voigtländer and col-
predominantly systolic velocity proximally to predominantly diastolic leagues demonstrated a flow velocity reserve of 2.6 ± 1.5 versus 0.8 ±
velocity distally, whereas saphenous vein graft flow velocity patterns 0.4 (P < .005) and a flow reserve of 2.9 ± 1.9 versus 1.2 ± 0.5 (P < .05)
have a consistently diastolic predominance, both proximally and distally.8 in 21 grafts without stenoses compared with 6 grafts with >75% diameter
CMR assessed volume flow of grafts with multiple sequential target stenosis.43 In a study by Bedaux and colleagues of 21 post-CABG patients
vessel anastomoses significantly differed from single grafts.26 Sequential (40 grafts: 18 single and 22 sequential) who underwent selective angi-
venous grafts have higher volume flow and velocities compared with ography due to recurrent angina pectoris, the combination of a basal,
single grafts, probably caused by a lower downstream resistance in CMR-derived proximal segment graft flow volume of less than 20 mL/
sequential grafts. min or a flow reserve of less than 2 had a sensitivity of 78% and a
Volume flow and velocity can both be analyzed on CMR velocity specificity of 80% in detecting graft stenosis or runoff vessel disease.44
maps. In the case of volume flow, the mean velocity is multiplied with Owing to the limited number of patients, no differentiation in volume
the vessel area and all pixels over that area are included in the analysis. flow was possible between single and sequential grafts. A larger study
However, errors caused by partial volume effects at the lumen edges by Langerak and colleagues of 69 post-CABG patients (166 grafts: 81
may occur, and the analysis is more time consuming. Velocity analysis single vein, 44 sequential vein, and 41 arterial grafts) with recurrent
that measures velocity in the center of the lumen (over a 2 × 2 pixel angina who underwent CMR and selective angiography reported suc-
area) is not affected by partial volume effects and is less time consum- cessful baseline and adenosine stress CMR flow scans in 80% of the
ing. Head-to-head comparisons have shown similar diagnostic accuracies grafts.45 The majority of unsuccessful flow scans (24 out of 26 grafts)
in detection of significant (>70%) vein graft stenosis between volume were attributed to adenosine-related side effects. Peak velocities of CMR
flow and velocity analysis.42 velocity maps were analyzed for single and sequential vein grafts sepa-
Several studies, using breath-hold segmented k-space sequences at rately, and a model using receiver operating characteristic (ROC) curve
baseline and during pharmacologic stress, have demonstrated the analysis and logistic regression to detect stenosis of 50% or more or
CHAPTER 26  Coronary Artery Bypass Graft Imaging and Assessment of Flow 331

A B

Rest
8
Adenosine stress
6
Velocity (cm/s)

0
0 200 400 600 800
−2 Time (ms)

D E

10 Rest

8 Adenosine stress
Velocity (cm/s)

0
0 200 400 600 800
−2 Time (ms)
F
FIG. 26.6  Patient with single vein graft inserting into the left anterior descending coronary artery (LAD) and
sequential vein graft inserting on posterior descending and posterolateral branch of the right coronary artery.
(A) Curved planar reformat of gadolinium-enhanced three-dimensional magnetic resonance angiography of
graft inserting on the LAD. The dashed line indicates the orientation of the plane perpendicular to the proximal
graft segment, used for velocity-encoded cine cardiac magnetic resonance (CMR). (B) X-ray angiography of
distal graft segment demonstrating irregular aspect of luminal borders and tight insertion on LAD. (C) Cross-
sectional averaged velocities measured at multiple phases throughout the cardiac cycle using velocity-encoded
cine CMR. Calculated volume-flow was 38 mL/min at rest and 68 mL/min during adenosine stress, yielding
a flow reserve of 1.8. (D) Curved planar reformat of proximal part of the sequential graft. The dashed line
indicates the plane of CMR flow acquisition. (E) X-ray angiography reveals a tight stenosis (arrow) in the graft
segment between the two distal insertion sites. (F) Volume flow calculated from magnetic resonance flow
velocity measurements was 50 mL/min at rest and 51 mL/min during adenosine stress, yielding a flow
reserve of 1. (From van Rossum AC, Bedaux WLF, Hofman MBM. Morphologic evaluation of coronary artery
bypass conduits. J Magn Reson Imaging. 1999;10:734–740.)
332 SECTION II  Ischemic Heart Disease

70% or more in the graft or runoff vessel was developed. Optimal cutoff rate of <20 mL/min or a flow reserve <2) with the model described by
points to differentiate single vein grafts with or without stenosis of Langerak and colleagues.45 The two methods demonstrated equivalent
70% or more were 13.58 cm/s for stress average peak velocity (sensitiv- sensitivities (70% vs. 74%), whereas the model using ROC curve analysis
ity: 91%, specificity: 62%), 20.86 cm/s for stress diastolic peak velocity with logistic regression demonstrated a much higher specificity (68%
(sensitivity: 91%, specificity: 74%), and 1.43 for coronary velocity reserve vs. 30%) for the detection of significant (≥50%) venous graft or target
(sensitivity: 91%, specificity: 78%). Higher cutoff points for stress average vessel stenosis.46 CMR has been compared with single-photon emission
peak velocity (22.47 cm/s; sensitivity 82%, specificity 62%) and diastolic perfusion computed tomography (SPECT) for the evaluation of the
peak velocity (38.67 cm/s; sensitivity 86%, specificity 62%) were found hemodynamic significance of angiographic findings in bypass grafts.
to detect sequential vein grafts with 70% stenosis or more compared In a study including 25 postbypass patients, flow velocity CMR was
with single vein grafts.45 A retrospective study by Salm and colleagues46 successful in 46 out of 57 grafts and demonstrated results similar to
including 68 post-CABG patients (125 saphenous venous grafts) com- those of SPECT in 80% (κ = 0.61) of those grafts.47 The effects of
pared the method used by Bedaux and colleagues44 (CMR basal flow percutaneous angioplasty on saphenous vein grafts function can be
assessed by using flow CMR. In a small study of 15 venous grafts before
and at least 6 weeks after percutaneous angioplasty, CMR has demon-
strated a significant improvement in baseline average peak velocity
(before: 9.2 ± 6.6 cm/s; after: 12.9 ± 7.9 cm/s; P = .008) and stress
average peak velocity (before: 12.9 ± 6.3 cm/s; after: 27.1 ± 13.9 cm/s;
P < .001).48 However, there was no improvement in velocity reserve, a
result that is explained by persistent impairment of stress average peak
CX graft velocity and not by an increased baseline peak velocity after interven-
RCA graft LAD graft tion.48 By using nonbreath-hold and breath-hold techniques, functional
results were also obtained in IMA grafts, despite problems with imaging
artifacts attributed to metallic clips and sternal wires.49,50 The diastolic/
Transverse plane
systolic peak velocity ratio was found to be higher in IMA grafts than
in native IMA. Findings in IMA grafts by Kawada and colleagues in 18
patients without and 5 patients with 75% or greater stenosis, indicate
a higher sensitivity and specificity for detection of IMA graft stenosis
using measurements of the mean flow rate and diastolic/systolic peak
flow ratio at rest over using the flow reserve after administration of
Sagittal plane
dipyridamole.51 A study by Ishida and colleagues including 26 patients
FIG. 26.7  Typical course of vein grafts from ascending aorta to coronary
with IMA grafts (with CMR flow performed successfully in the middle
artery insertion sites, relative to sagittal and transverse imaging planes.
CX, Circumflex; LAD, left anterior descending coronary artery; RCA,
segment of 24 grafts) demonstrated similar sensitivities of 86% and
right coronary artery. (From Galjee MA, van Rossum AC, Doesburg T, specificities of 88% and 94% in detecting greater than 70% diameter
van Eenige MJ, Visser CA. Value of magnetic resonance imaging in stenosis for mean diastolic/systolic peak velocity ratio and mean baseline
assessing patency and function of coronary artery bypass grafts: an blood flow, respectively. No significant difference was observed in the
angiographically controlled study. Circulation. 1996;93:660–666.) flow reserve ratio between grafts with more than 70% stenosis and

CX

LAD

LA

A B
FIG. 26.8  Oblique-sagittal image obtained with velocity-encoded phase-contrast technique to measure flow.
(A) Magnitude reconstruction of magnetic resonance (MR) signal depicting cross sections of two grafts
anterior and superior of the main pulmonary artery. (B) Corresponding reconstruction of the phase of the
MR signal, where the brightness of each pixel is proportional to the flow velocity, and midgray equals zero
flow velocity. Within the grafts, the bright signal (white arrows) indicates high cross-sectional velocities. CX,
Circumflex; LA, left atrium; LAD, left anterior descending coronary artery; P, pulmonary artery. (From van
Rossum AC, Galjee MA, Post JC, Visser CA. A practical approach to CMR of coronary artery bypass graft
patency and flow. Int J Cardiac Imaging. 1997;13:199–204.)
CHAPTER 26  Coronary Artery Bypass Graft Imaging and Assessment of Flow 333

VOLUME FLOWS
Corrected for RR interval

Single graft Sequential graft to 1 region


3 3

2 2

1 1
Volume flow in mL/s

0 0
200 400 600 800 200 400 600 800

Sequential graft to 2 regions Sequential graft to 3 regions


3 3

2 2

1 1

0 0
200 400 600 800 200 400 600 800
Time in ms
FIG. 26.9  Plots of volume-flow patterns in single and sequential saphenous vein grafts, normalized for heart
rate. Typical patterns consist of a first flow peak in systole and a second peak in diastole. (From Galjee MA,
van Rossum AC, Doesburg T, van Eenige MJ, Visser CA. Value of magnetic resonance imaging in assessing
patency and function of coronary artery bypass grafts: an angiographically controlled study. Circulation.
1996;93:660–666.)

those without significant stenosis.52 Stauder and colleagues evaluated alternative to other methods, such as SPECT or Doppler wire, for func-
graft patency, flow, and flow reserve in 30 patients who underwent tional graft assessment.
minimally invasive CABG of IMA grafts at least 6 years before using a
CMR protocol of rest and dipyridamole stress phase-contrast technique
and both contrast enhanced and navigator guided 3D MRA.53 The study
LIMITATIONS
reported a sensitivity of 100% and specificity of 68% for identifying So far, CMR of coronary artery bypass grafts has been limited, owing
IMA graft stenosis in the case of contrast-enhanced MRA. Addition of to the constraints of spatial resolution, to demonstrating vessel patency
navigator-guided 3D MRA increased the specificity to 89%. Flow reserve versus total occlusion only. CMR measurements of blood flow at rest
measurements had 100% sensitivity with 95% specificity, whereas flow and under stress, with calculation of the diastolic/systolic flow ratio and
reserve ratios demonstrated suboptimal performance.53 the coronary flow reserve, have become helpful additions to anatomic
In another study, published by the same group, which included 45 imaging to determine the functional status of a diseased bypass graft
symptomatic patients who had undergone CABG at least 5 years before and its recipient coronary artery. Furthermore, most of the reported
(86 grafts, 46 arterial, 40 venous), contrast-enhanced MRA and rest and studies have been confined to imaging of proximal graft segments,
dipyridamole stress phase-contrast flow measurements of grafts were and only few data are available with respect to assessing patency of
compared with selective angiography or CCT as reference standards. segments beyond the first coronary anastomosis in sequential bypass
Sensitivity, specificity, and positive and negative predictive values for grafts. Imaging of the distal graft segments requires a higher spatial
stenosis in arterial grafts were 95.2%, 96.8%, 80% and 99.4%, respectively, resolution and signal-to-noise ratio (SNR) than those obtained with
and in venous grafts were 100%, 97.8%, 87.5% and 100%, respectively.54 currently available techniques. Another point of consideration is the
There is limited experience with evaluation of graft flow using 3 T problem associated with CMR of arterial grafts. IMA grafts are increas-
CMR.55,56 Patency and flow characteristics of arterial and venous grafts ingly used because of the improved long-term survival but have been
were assessed and measured using phase contrast velocity mapping at excluded from most CMR studies, owing to metallic clip and sternal
3 T within 1 week post-CABG and compared with intraoperative Doppler wires artifacts. Flow reserve measurements proximal to the clip arti-
flow measurements. The studies demonstrated that CMR flow measure- facts may be of diagnostic help or, alternatively, methods to obtain
ments of venous bypass grafts agreed more with Doppler data than hemostasis without use of ferromagnetic clips might be useful. The
arterial bypass grafts and confirmed patency in all patients.55,56 anatomic imaging of bypass grafts after percutaneous angioplasty with
Given available evidence it is reasonable to consider that CMR flow stent deployment is limited by stent-induced artifacts (most stents are
variables such as mean graft blood flow, diastolic/systolic flow profiles, made of surgical steel or tantalum, with the latter demonstrating less
and flow velocity reserve of bypass grafts are useful in the noninvasive signal void artifacts), although measurement of flow is possible in the
assessment of graft and runoff vessel disease. CMR could become an nonstented regions.48,57 Integration of miniature radiofrequency coils
334 SECTION II  Ischemic Heart Disease

into stents (transforming them into intravascular antennas coupled to stripping,61 by demonstrating patency rates without the need for
a surface coil) allows the visualization of stent lumen.58 CMR-lucent invasive angiography.
stents are also under development. Notwithstanding these indications, the majority of patients after
However, even a clear demonstration of graft-segment patency or CABG require evaluation with respect to percutaneous or surgical
narrowing will often not suffice for clinical decision making. In most revascularization. Unless CMR will be able to provide more detailed
patients, there is a need to also know the status of the native coronary information regarding the status of native coronary arteries, a wide
arteries. Narrowing of the recipient coronary artery may have developed application of the technique is unlikely to occur. Continuing improve-
beyond the anastomosis of a patent graft segment, and progression of ment in CMR of coronary arteries and bypass grafts may be expected
disease in other native coronary arteries must be excluded. Thus CMR with new developments in scanner hardware and software and the use
does not eliminate the need for conventional x-ray coronary angiography of contrast agents. Most recent clinical guidelines and consensus docu-
when a revascularization procedure is considered. ments give MRA a high grading for evaluation of coronary anomalies;
however, in the case of aortocoronary bypass grafts, MRA alone receives
class II indication or uncertain appropriateness.62-64 However, when
INDICATIONS combined with stress first pass gadolinium-contrast myocardial perfu-
A clinical indication for CMR of bypass grafts may therefore exist sion CMR, most experts consider it an appropriate indication for patients
only in patients in whom there is no immediate need to know the who have undergone CABG and have recurrent symptoms suggesting
status of the native coronary arteries. Such a category consists, for myocardial ischemia.
example, of patients with chest pain shortly after CABG surgery.
Also, late after CABG surgery, information about graft patency and
function might be helpful in deciding whether to postpone coronary
CONCLUSION
angiography in patients with ambiguous anginal complaints. Nonin- There is clear evidence that conventional spin echo and gradient
vasive monitoring of flow parameters may then be useful to detect echo CMR are capable of assessing patency of venous aortocoronary
a gradual increase of graft stenosis and decide to proceed with con- grafts. With recent advances in breath-hold 2D and contrast-enhanced
ventional angiography and percutaneous intervention, before the 3D techniques, the accuracy in detecting graft occlusion has further
onset of a total occlusion. Similarly, flow measurements could be used improved. Limitations are still present in evaluating distal graft seg-
after bypass graft percutaneous angioplasty for the early detection of ments and sequential grafts caused by insufficient spatial resolution,
restenosis. low SNRs and motion. Imaging of arterial grafts is complicated by
Furthermore, CMR can be used as a screening procedure before the sternal wire and metallic clip artifacts. Recently, significant pro-
angiography, providing a road map for the grafts to be visualized. This gress has been made in the assessment of graft flow patterns and flow
might considerably shorten the angiographic procedure with less expo- reserve using phase contrast velocity-encoded CMR both at 1.5 T and
sure to ionizing radiation and lower total iodine contrast dose. A useful 3 T. Clinically, these functional measurements may become useful for
indication also appears to be the assessment of the patency of grafts noninvasive monitoring of bypass graft narrowing progression before
that are not visualized during conventional angiography. Although this and even after percutaneous interventions.48 However, the majority of
often indicates a proximal occlusion of the graft, failure of the catheter patients undergo graft evaluation in preparation for revascularization
to find an aortic graft anastomosis may result in a false diagnosis of procedures. In these cases knowing which venous grafts are completely
graft occlusion. In cases of doubt, MRA will rapidly confirm or discard occluded, although helpful, is still not sufficient because information
this diagnosis. Also, when angiography has demonstrated a graft stenosis, on the status of the native coronary arteries is still required. A broader
it can be helpful for further management to assess the flow reserve of role for CMR in the evaluation of patients with aortocoronary bypass
a graft. There are also occasions when CMR is useful in visualizing and grafts may therefore be expected only after further improvement in
assisting with further surgical planning for complications of vein graft- CMR coronary angiography.
ing, such as aneurysm of the graft.59
Finally, vein graft imaging may have a role in research of bypass
techniques and has been used to demonstrate the efficacy of apro-
REFERENCES
tinin60 during surgery and in comparison of techniques for vein graft A full reference list is available online at ExpertConsult.com
CHAPTER 26  Coronary Artery Bypass Graft Imaging and Assessment of Flow 334.e1

REFERENCES 20. White RD, Caputo GR, Mark AS, Modin GW, Higgins CB. Coronary
artery bypass graft patency: noninvasive evaluation with MR imaging.
1. Favaloro RG. Saphenous vein autograft replacement of severe segmental Radiology. 1987;164(3):681–686.
coronary artery occlusion: operative technique. Ann Thorac Surg. 21. Rubinstein RI, Askenase AD, Thickman D, Feldman MS, Agarwal JB,
1968;5(4):334–339. Helfant RH. Magnetic resonance imaging to evaluate patency of
2. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke aortocoronary bypass grafts. Circulation. 1987;76(4):786–791.
statistics—2016 update: a report from the American Heart Association. 22. Jenkins JP, Love HG, Foster CJ, Isherwood I, Rowlands DJ. Detection of
Circulation. 2016;133(4):e38–e360. coronary artery bypass graft patency as assessed by magnetic resonance
3. Fitzgibbon GM, Kafka HP, Leach AJ, Keon WJ, Hooper GD, Burton JR. imaging. Br J Radiol. 1988;61:2–4.
Coronary bypass graft fate and patient outcome: angiographic follow-up 23. Frija G. A study of coronary artery bypass graft patency using MR
of 5,065 grafts related to survival and reoperation in 1,388 patients imaging. J Comput Assist Tomogr. 1989;13:226–232.
during 25 years. J Am Coll Cardiol. 1996;28(3):616–626. 24. White RD, Pflugfelder PW, Lipton MJ, Higgins CB. Coronary artery
4. Hess CN, Lopes RD, Gibson CM, et al. Saphenous vein graft failure after bypass grafts: evaluation of patency with cine MR imaging. AJR Am J
coronary artery bypass surgery: insights from PREVENT IV. Circulation. Roentgenol. 1988;150(6):1271–1274.
2014;130(17):1445–1451. 25. Aurigemma GP, Reichek N, Axel L, Schiebler M, Harris C, Kressel HY.
5. Cao C, Manganas C, Horton M, et al. Angiographic outcomes of radial Noninvasive determination of coronary artery bypass graft patency by
artery versus saphenous vein in coronary artery bypass graft surgery: a cine magnetic resonance imaging. Circulation. 1989;80(6):1595–1602.
meta-analysis of randomized controlled trials. J Thorac Cardiovasc Surg. 26. Galjee MA, van Rossum AC, Doesburg T, van Eenige MJ, Visser CA. Value
2013;146(2):255–261. of magnetic resonance imaging in assessing patency and function of
6. Harskamp RE, Lopes RD, Baisden CE, de Winter RJ, Alexander JH. coronary artery bypass grafts. An angiographically controlled study.
Saphenous vein graft failure after coronary artery bypass surgery: Circulation. 1996;93(4):660–666.
pathophysiology, management, and future directions. Ann Surg. 27. Manning WJ, Li W, Boyle NG, Edelman RR. Fat-suppressed breath-hold
2013;257(5):824–833. magnetic resonance coronary angiography. Circulation. 1993;87(1):
7. Cameron A, Davis KB, Green G, Schaff HV. Coronary bypass surgery 94–104.
with internal-thoracic-artery grafts—effects on survival over a 15-year 28. Hartnell GG. Segmented k-space magnetic resonance angiography for the
period. N Engl J Med. 1996;334(4):216–219. detection of coronary artery bypass graft patency. In: Book of Abstracts of
8. Bach RG, Kern MJ, Donohue TJ, Aguirre FV, Caracciolo EA. Comparison the 4th Meeting of the International Society of Magnetic Resonance in
of phasic blood flow velocity characteristics of arterial and venous Medicine (ISMRM). New York: ISMRM; 1996.
coronary artery bypass conduits. Circulation. 1993;88(5 Pt 2):II133–II140. 29. Post JC, van Rossum AC, Bronzwaer JGF, de Cock CC, Visser CA.
9. Fusejima K, Takahara Y, Sudo Y, Murayama H, Masuda Y, Inagaki Y. Magnetic resonance angiography of sequential aortocoronary bypass
Comparison of coronary hemodynamics in patients with internal grafts. Circulation. 1997;1:133.
mammary artery and saphenous vein coronary artery bypass grafts: a 30. Kalden P, Kreitner KF, Wittlinger T, et al. Assessment of coronary artery
noninvasive approach using combined two-dimensional and Doppler bypass grafts: value of different breath-hold MR imaging techniques. AJR
echocardiography. J Am Coll Cardiol. 1990;15:131–139. Am J Roentgenol. 1999;172(5):1359–1364.
10. Jones C. Does Doppler echography have a diagnostic role in patency 31. Hofman MBM, Paschal CB, Li D, Haacke EM, van Rossum AC, Sprenger
assessment of internal thoracic artery grafts? Eur J Cardiothorac Surg. M. CMR of coronary arteries, 2D breath-hold versus 3D respiratory-
2005;28:692–700. gated acquisition. J Comput Assist Tomogr. 1995;19:56–62.
11. Ferguson T Jr. Fractional flow reserve-guided coronary artery bypass 32. Danias PG, McConnell MV, Khasgiwala VC, Chuang ML, Edelman RR,
grafting: can intraoperative physiologic imaging guide decision making? Manning WJ. Prospective navigator correction of image position for
J Thorac Cardiovasc Surg. 2013;146:824–835. coronary MR angiography. Radiology. 1997;203(3):733–736.
12. Gabriel J. Should computed tomography angiography supersede invasive 33. Kessler W, Achenbach S, Moshage W, et al. Usefulness of respiratory gated
coronary angiography for the evaluation of graft patency following magnetic resonance coronary angiography in assessing narrowings > or =
coronary artery bypass graft surgery? Interact Cardiovasc Thorac Surg. 50% in diameter in native coronary arteries and in aortocoronary bypass
2015;21:231–239. conduits. Am J Cardiol. 1997;80(8):989–993.
13. Heye T, Kauczor HU, Szabo G, Hosch W. Computed tomography 34. Molinari G, Sardanelli F, Zandrino F, Balbi M, Masperone MA. Value of
angiography of coronary artery bypass grafts: robustness in emergency navigator echo magnetic resonance angiography in detecting occlusion/
and clinical routine settings. Acta Radiol. 2014;55(2):161–170. patency of arterial and venous, single and sequential coronary bypass
14. Weustink AC, Nieman K, Pugliese F, et al. Diagnostic accuracy of grafts. Int J Card Imaging. 2000;16(3):149–160.
computed tomography angiography in patients after bypass grafting: 35. Langerak SE. Detection of vein graft disease using high-resolution
comparison with invasive coronary angiography. JACC Cardiovasc magnetic resonance angiography. Circulation. 2002;105:328–333.
Imaging. 2009;2(7):816–824. 36. Prince MR, Narasimham DL, Stanley JC, et al. Breath-hold gadolinium-
15. de Graaf FR, van Velzen JE, Witkowska AJ, et al. Diagnostic performance enhanced MR angiography of the abdominal aorta and its major
of 320-slice multidetector computed tomography coronary angiography branches. Radiology. 1995;197(3):785–792.
in patients after coronary artery bypass grafting. Eur Radiol. 37. Wintersperger BJ, Engelmann MG, von Smekal A. Patency of coronary
2011;21(11):2285–2296. artery bypass grafts: assessment with breath-hold contrast-enhanced MR
16. Rau NH, Rau P, Chin AS, et al. Coronary artery bypass graft imaging angiography—value of a non-electrocardiographically triggered
with 256-slice MDCT: surgical concepts, current techniques, and technique. Radiology. 1998;208:345–351.
interpretation. Clin Imaging. 2014;38(5):571–579. 38. Vrachliotis TG, Bis KG, Aliabadi D, Shetty AN, Safian R, Simonetti O.
17. van Rossum AC, Galjee MA, Doesburg T, Hofman M, Valk J. The role of Contrast-enhanced breath-hold MR angiography for evaluating patency
magnetic resonance in the evaluation of functional results after CABG/ of coronary artery bypass grafts. AJR Am J Roentgenol.
PTCA. Int J Card Imaging. 1993;9(suppl 1):59–69. 1997;168(4):1073–1080.
18. Galjee MA, van Rossum AC, Doesburg T, Hofman MB, Falke TH, Visser 39. Goldfarb JW, Edelman RR. Coronary arteries: breath-hold, gadolinium-
CA. Quantification of coronary artery bypass graft flow by magnetic enhanced, three-dimensional MR angiography. Radiology.
resonance phase velocity mapping. Magn Reson Imaging. 1998;206(3):830–834.
1996;14(5):485–493. 40. Bunce NH, Lorenz CH, John AS, Lesser JR, Mohiaddin RH, Pennell DJ.
19. van Rossum AC, Bedaux WL, Hofman MB. Morphologic and functional Coronary artery bypass graft patency: assessment with true fast imaging
evaluation of coronary artery bypass conduits. J Magn Reson Imaging. with steady-state precession versus gadolinium-enhanced MR
1999;10(5):734–740. angiography. Radiology. 2003;227(2):440–446.
334.e2 SECTION II  Ischemic Heart Disease

41. van Rossum AC, Galjee MA, Post JC, Visser CA. A practical approach to 55. Hoppe H, Reineke D, Rosskopf AB, et al. Morphological and functional
CMR of coronary artery bypass graft patency and flow. Int J Card 3-Tesla magnetic resonance imaging of saphenous vein coronary artery
Imaging. 1997;13:199–204. bypass grafts. Interact Cardiovasc Thorac Surg. 2011;12(4):582–585.
42. Salm LP, Langerak SE, Vliegen HW. Blood flow in coronary artery bypass 56. Reineke D, Ith M, Goeber V, et al. Comparison of arterial and venous
vein grafts: volume versus velocity at cardiovascular MR imaging. coronary artery bypass flow measurements using 3-T magnetic resonance
Radiology. 2004;232(3):915–920. phase contrast imaging. Eur J Radiol. 2012;81:e502–e506.
43. Voigtländer T, Kreitner KF, Wittlinger T, Scharhag J, Abegunewardene N, 57. Nagel E, Thouet T, Klein C, et al. Noninvasive determination of coronary
Meyer J. MR measurement of flow reserve in coronary grafts [abstract]. blood flow velocity with cardiovascular magnetic resonance in patients
J Cardiovasc Magn Reson. 1999;1:275. after stent deployment. Circulation. 2003;107(13):1738–1743.
44. Bedaux WL, Hofman MB, Vyt SL, Bronzwaer JG, Visser CA, van Rossum 58. Quick HH, Ladd ME. [Interventional MRA: concepts for active
AC. Assessment of coronary artery bypass graft disease using visualization of catheters and stents]. Z Med Phys. 2003;13(3):188–192.
cardiovascular magnetic resonance determination of flow reserve. J Am 59. Warner OJ, Ohri SK, Pennell DJ, Smith PLC. Magnetic resonance
Coll Cardiol. 2002;40(10):1848–1855. coronary artery imaging for redo cardiac surgery. Ann Thorac Surg.
45. Langerak SE, Vliegen HW, Jukema JW, et al. Value of magnetic resonance 1996;62:1513–1516.
imaging for the noninvasive detection of stenosis in coronary artery 60. Bidstrup BP, Underwood SR, Sapsford RN. Effect of aprotinin (Trasylol)
bypass grafts and recipient coronary arteries. Circulation. on aorto-coronary bypass graft patency. J Thorac Cardiovasc Surg.
2003;107(11):1502–1508. 1993;105:147–153.
46. Salm LP, Vliegen HW, Langerak SE, et al. Evaluation of saphenous vein 61. O’Regan DJ, Borland JA, Chester AH, Pennell DJ, Yacoub M, Pepper JR.
coronary artery bypass graft flow by cardiovascular magnetic resonance. Assessment of human long saphenous vein function with minimally
J Cardiovasc Magn Reson. 2005;7(4):631–637. invasive harvesting with the Mayo stripper. Eur J Cardiothorac Surg.
47. Salm LP, Bax JJ, Vliegen HW, et al. Functional significance of stenoses in 1997;12(3):428–435.
coronary artery bypass grafts. Evaluation by single-photon emission 62. American College of Radiology, Society of Cardiovascular Computed
computed tomography perfusion imaging, cardiovascular magnetic Tomography, Society for Cardiovascular Magnetic Resonance, et al.
resonance, and angiography. J Am Coll Cardiol. 2004;44(9):1877–1882. ACCF/ACR/SCCT/SCMR/ASNC/NASCI/SCAI/SIR 2006 appropriateness
48. Langerak SE, Vliegen HW, Jukema JW, et al. Vein graft function criteria for cardiac computed tomography and cardiac magnetic
improvement after percutaneous intervention: evaluation with MR flow resonance imaging. A report of the American College of Cardiology
mapping. Radiology. 2003;228(3):834–841. Foundation Quality Strategic Directions Committee Appropriateness
49. Debatin JF, Strong JA, Sostman HD, et al. MR characterization of blood Criteria Working Group. J Am Coll Radiol. 2006;3(10):751–771.
flow in native and grafted internal mammary arteries. J Magn Reson 63. American College of Cardiology Foundation Task Force on Expert
Imaging. 1993;3(3):443–450. Consensus Documents, Hundley WG, Bluemke DA, et al. ACCF/ACR/
50. Sakuma H, Globits S, O’Sullivan M, et al. Breath-hold MR measurements AHA/NASCI/SCMR 2010 expert consensus document on cardiovascular
of blood flow velocity in internal mammary arteries and coronary artery magnetic resonance: a report of the American College of Cardiology
bypass grafts. J Magn Reson Imaging. 1996;6(1):219–222. Foundation Task Force on Expert Consensus Documents. J Am Coll
51. Kawada N, Sakuma H, Cruz BC, et al. Noninvasive detection of Cardiol. 2010;55(23):2614–2662.
significant stenosis in the coronary artery bypass grafts using fast 64. Wolk MJ, Bailey SR, Doherty JU, et al. ACCF/AHA/ASE/ASNC/HFSA/
velocity-encoded cine CMR [abstract]. J Cardiovasc Magn Reson. HRS/SCAI/SCCT/SCMR/STS 2013 multimodality appropriate use criteria
1999;1:261. for the detection and risk assessment of stable ischemic heart disease: a
52. Ishida N, Sakuma H, Cruz BP, et al. MR flow measurement in the report of the American College of Cardiology Foundation Appropriate
internal mammary artery-to-coronary artery bypass graft: comparison Use Criteria Task Force, American Heart Association, American Society
with graft stenosis at radiographic angiography. Radiology. of Echocardiography, American Society of Nuclear Cardiology, Heart
2001;220(2):441–447. Failure Society of America, Heart Rhythm Society, Society for
53. Stauder NI, Fenchel M, Stauder H, et al. Assessment of minimally Cardiovascular Angiography and Interventions, Society of Cardiovascular
invasive direct coronary artery bypass grafting of the left internal thoracic Computed Tomography, Society for Cardiovascular Magnetic Resonance,
artery by means of magnetic resonance imaging. J Thorac Cardiovasc and Society of Thoracic Surgeons. J Am Coll Cardiol. 2014;63(4):380–406.
Surg. 2005;129(3):607–614.
54. Stauder NI, Klumpp B, Stauder H, et al. Assessment of coronary artery
bypass grafts by magnetic resonance imaging. Br J Radiol.
2007;80(960):975–983.
27 
Atherosclerotic Plaque Imaging:
Aorta and Carotid
Marc R. Dweck, Philip M. Robson, James H.F. Rudd, and Zahi A. Fayad

Although death rates in industrialized countries have been consistently arterial media and synthesize the extracellular matrix components of
falling since the 1980s, atherosclerosis is now raging throughout the the cap, such as elastin and collagen. The cap also contains variable
developing world. As a consequence the complications of atherosclerosis numbers of inflammatory cells, most importantly macrophages. As the
have become the leading cause of mortality and morbidity worldwide. plaque enlarges, the affected artery grows outward (by expansion of
Fundamentally, atherosclerosis is an inflammatory disease, which affects the external elastic lamina) so that lumen diameter and therefore blood
medium and large arteries from the first decade of life until death. It flow is initially preserved; this is a process known as positive remodel-
has a predilection for certain arterial beds, with the carotid artery and ing.6 As the artery’s wall stress increases with outward remodeling,
aorta being the most common sites of plaque formation. In the carotid further expansion eventually becomes impossible and the plaque then
circulation, the end result can be an ischemic cerebral insult causing encroaches into the lumen. Ultimately this may cause angina by com-
either temporary (transient ischemic attack [TIA]) or permanent (cere- promising blood flow to the myocardium.
brovascular attack [CVA]) symptoms. If unchecked, aortic atherosclerosis Mature plaques may also become calcified, a process that prefer-
can predispose to dissection, intramural hemorrhage, aneurysm forma- entially affects the intima of the artery and is thought to occur as a
tion and downstream embolus. Importantly, because atherosclerosis is healing response to inflammation and cell death within the plaque.
a systemic disease, assessments of the carotid arteries and aorta also Although the early stages of microcalcification are associated with
provide surrogate information about disease burden and activity in the high-risk inflamed atheroma, the latter stages of macrocalcification
coronary circulation, providing prognostic information about the risk are by contrast associated with burnt-out stable disease. Very advanced
of myocardial infarction.1 plaques with a large necrotic core and associated hypoxia will also often
be perforated by new blood vessels under the influence of angiogenic
factors, a process called neovascularization, with similarities to that
PATHOBIOLOGY OF ATHEROSCLEROSIS which occurs within growing tumors. However, these small arteries
Atherosclerosis is characterized by the gradual accumulation of lipid, are structurally fragile and have a tendency to spontaneously hemor-
inflammatory cells, and connective tissue within the arterial wall. It is a rhage, which can destabilize the plaque leading to increased plaque
chronic, progressive disease with a very long asymptomatic/subclinical inflammation, rupture, and the precipitation of acute myocardial
phase. The first abnormality to develop is the fatty streak, caused by infarction.7
the collection of lipid and macrophages in the subendothelial space. Atherosclerotic plaques may remain quiescent for decades. However,
Fatty streaks develop primarily in regions of low wall shear stress, when they initiate clot formation in the vessel lumen, they can very
which results in endothelial dysfunction and the production of less quickly become life threatening. This occurs predominantly as a result
nitric oxide.2 The major atherogenic risk factors including smoking, of fibrous cap rupture, with consequent exposure of the thrombogenic
hypertension, and diabetes mellitus also all affect endothelial function,3 and tissue factor-rich lipid core to circulating blood. Less commonly,
impairing both its barrier function and secretory capacity. Ultimately there can be erosion of the endothelial cell layer overlying the fibrous
the blood vessel wall becomes more permeable to blood-derived lipids cap, which in the context of an advanced thrombogenic response can
and inflammatory cells in these regions, encouraging the early stages of also lead to intravascular thrombosis. It is estimated that endothelial
atherosclerosis. erosion accounts for ~30% of myocardial infarctions.8 Regardless of
Once within the subendothelial space, low-density lipoprotein (LDL) the mechanism, plaque disruption will lead to varying degrees of local
becomes oxidized and attracts monocytes by triggering the release of platelet activation and thrombus formation, depending on the charac-
monocyte chemoattractant protein-1 (MCP-1) from the overlying teristics of the plaque and the thrombogenicity of the blood. Importantly,
endothelial cells.4 Endothelial adhesion molecules, including vascular however, the majority of these plaque ruptures do not appear to result
cell adhesion molecule-1 (VCAM), intercellular adhesion molecule-1, in overt clinical events but instead remain subclinical and responsible
E-selectin, and P-selectin, facilitate the internalization of further mono- for abrupt plaque growth.9
cytes. Once they have escaped the blood pool, monocytes transform It has become clear that the cellular and extracellular composition
into macrophages and bind and internalize oxidative LDL via their of the plaque is the primary determinant of plaque stability.10 Lesions
scavenger receptors.5 Eventually, the subendothelial accumulation of with a large lipid core, thin fibrous cap, positive remodeling, angio-
modified LDL and macrophage-derived foam cells leads to the forma- genesis, microcalcification, and a preponderance of inflammatory cells
tion of the atheromatous lipid core. The thrombogenic components of compared with VSMCs are at the highest risk of rupture. Inflammatory
the lipid core become separated from blood in the lumen by the endo- cells, particularly macrophages, produce metalloproteinase enzymes,
thelialized fibrous cap, which consists predominantly of vascular smooth which break down matrix proteins, weakening the fibrous cap. In addi-
muscle cells (VSMCs) and connective tissue. VSMCs migrate from the tion, they secrete inflammatory cytokines, in particular interferon γ,

335
336 SECTION II  Ischemic Heart Disease

which inhibit VSMC proliferation and matrix production. Further- burden that compare favorably with histology19 and predict major adverse
more, VSMCs in the fibrous cap have a reduced proliferative capacity cardiovascular outcomes.20
and a propensity to apoptosis.11 Consequently, inflammation within Black-blood CMR also allows the early detection of subclinical ath-
the plaques promotes destruction of the fibrous cap, a propensity to erosclerotic disease. Indeed CMR is particularly attractive for this purpose,
plaque rupture, and subsequent thrombosis. However, this is balanced given the absence of ionizing radiation, and has already proved useful
by the action of VSMCs, which nourish and repair the cap, promot- in several large cohort studies. In asymptomatic subjects enrolled in
ing plaque stabilization. This dynamic balance between proinflamma- the Framingham Heart Study (FHS), FHS coronary risk score was
tory and healing responses across the vasculature ultimately governs strongly associated with asymptomatic aortic atherosclerosis detected
a patient’s risk of myocardial infarction or stroke. These processes are by CMR.16 The prevalence and extent of aortic atherosclerosis increased
independent of the degree of luminal stenosis. Consequently, positively with age. In a substudy of the Multiethnic Study of Atherosclerosis
remodeled and angiographically invisible plaques can rupture to pre- (MESA), CMR aortic wall thickness increased as a function of age, but
cipitate a fatal clinical event, while many large plaques that obstruct males and black participants had the greatest wall thickness.21 In another
flow and cause angina may be stable and not life threatening. There study of 102 patients undergoing x-ray coronary angiography, aortic
is therefore an urgent need for imaging techniques that can discrimi- atherosclerotic plaques were detected with a higher frequency in active
nate “stable” from potentially “unstable” lesions in clinical practice. smokers and in those with high levels of LDL-cholesterol, but the volume
This chapter will investigate the role that cardiovascular magnetic and area of aortic plaques correlated most strongly with age and the
resonance (CMR) imaging might play in this respect, with a focus on presence of hypertension. Interestingly, only atherosclerotic plaques
its ability to measure the total plaque burden, to investigate high-risk located in the thoracic aorta were found to be associated with coronary
plaque characteristics, and to determine disease activity with advanced artery disease (CAD).17 Taken together, these studies confirm the strong
molecular techniques. correlation between the presence of cardiovascular risk factors and the
incidence of aortic atherosclerosis. It should be noted, however, that
there are racial and population differences in the response to individual
IMAGING ATHEROSCLEROSIS risk factors, which presumably have a genetic basis.21
CMR can make use of differences in tissue relaxation times (T1 and Another benefit readily appreciated by pharmaceutical companies
T2) and proton density, intrinsic material properties that affect the and others wishing to study the effects of drugs on plaque progression
magnitude of the magnetic resonance (MR) signal, to generate soft- (and regression) is the low coefficient of variability of CMR plaque
tissue contrast, providing detailed information about atherosclerotic volume measurements.22,23 This translates directly into the requirement
plaque composition. This is readily feasible in the large and relatively for lower patient numbers for studies investigating the impact of phar-
immobile carotid arteries and thoracic aorta, with intensive research maceutical agents on plaque volume, because any true drug effect will
aimed at transferring these techniques into the more challenging coro- not be swamped by noise within the measuring technique. This was
nary vasculature. illustrated in two separate studies where high-dose statin regimens were
The aim of atherosclerotic plaque imaging is ultimately to identify shown to be superior to low-dose regimens in terms of atheroma burden
vulnerable patients at high risk of myocardial infarction or stroke before reduction, an effect that could be demonstrated with only 20 patients
symptoms and complications develop.12 Similarly, early identification per group.24,25
of disease might allow the implementation of pharmacologic treatment Subsequently, in the first double-blind, multicentric, dal-PLAQUE
to halt or even reverse the process of plaque progression and possibly (safety and efficacy of dalcetrapib on atherosclerotic disease using novel
prevent rupture (e.g., with statin therapy).13 Recently, rapid advances noninvasive multimodality imaging) study, 130 patients were randomly
in CMR hardware, software, and molecular imaging tracers have seen assigned to placebo (N = 66) or 24 months treatment with the choles-
CMR develop into a modality of considerable utility and versatility for terylester transfer protein (CETP) inhibitor dalcetrapib (N = 64). In
imaging large vessel atheroma. In particular, assessments of atheroscle- patients receiving dalcetrapib, the absolute change from baseline relative
rotic plaque burden offer powerful prediction of adverse cardiovascular to placebo in total vessel area was 4.01 mm2 (90% CI, 7.23 to 0.80;
events, whereas more advanced techniques aimed at evaluating high-risk nominal P = .04) over 24 months.26
plaque characteristics and disease activity hold promise in refining this
risk prediction even further. Each of these features will be discussed in
detail below.
PLAQUE CHARACTERISTICS
Plaques at risk of rupture have certain pathological characteristics,
including inflammation, positive remodeling, a large necrotic core,
ASSESSMENTS OF PLAQUE BURDEN angiogenesis, microcalcification, and a thin, fibrous cap. Each represents
Simple measures of the atherosclerotic plaque burden in different vas- a potential imaging target, and although not all of these plaques will
cular beds provide powerful prognostic information, presumably because progress to rupture and even less will cause events,27 emerging evidence
the more plaques a patient has the more likely one will rupture or erode suggests that identification of such plaques may identify patients at
and cause an event. CMR can effectively image atherosclerotic plaque increased cardiovascular risk.28 Again, CMR techniques aimed at resolv-
using high-resolution, black-blood, fast-spin echo sequences.14 Prepara- ing these characteristics are best suited to the carotid arteries.
tory pulses null signal in the blood pool and perivascular fat, improving
contrast between the plaques and vessel lumen, and surrounding tissue, Positive Remodeling
respectively. The atherosclerotic plaque burden can then be quantified Similar black-blood imaging techniques as described earlier can be used
using measurements of plaque thickness in two dimensions and plaque to identify positive remodeling. Indeed, identification of positive remod-
volume in three dimensions. In the aorta these measurements demon- eling in the aorta and carotids has been shown to identify patients with
strate a close correlation with transesophageal echocardiography15 and an increased risk of future adverse cardiovascular outcomes.20 Moreover,
with increasing numbers of cardiovascular risk factors.16,17 Similar positive remodeling is also detectable with CMR in individual coronary
approaches have been used to measure plaque thickness and the plaque arteries,29 although the relationship between this CMR finding and
volume in the carotid arteries,18 providing measures of atherosclerotic prognosis is yet to be established.30
CHAPTER 27  Atherosclerotic Plaque Imaging: Aorta and Carotid 337

the effect of lipid-lowering therapy on the lipid content of atherosclerotic


Multicontrast Magnetic Resonance plaque after just 1 year of treatment. Importantly, the observed reduc-
Multicontrast CMR of the plaque is based on successive high-resolution tion continued in to the second year of therapy and preceded any effects
black-blood fast-spin echo CMR sequences that null signal in the flowing on vessel wall area.34
blood and perivascular fat with preparatory pulses. Several sequences
with different weightings are generally acquired (e.g., T1 weighted, T2 Imaging Acute Thrombus and Plaque Hemorrhage
weighted, and proton density weighted [PDW]). Analysis of the various Histopathological studies38 suggest that intraplaque hemorrhage may
signal intensities in each of these sequences allows reasonable differ- play a role in triggering both plaque rupture and growth. A first study
entiation of plaque components. In particular, the lipid core, fibrous proved that multicontrast CMR could accurately image intraplaque
tissue, plaque hemorrhage, the fibrous cap, and the degree of calcifica- hemorrhage in carotid atheroma using T2*-weighted sequences.39
tion can all be identified and quantified according to their different However CMR can also distinguish between recent and remote hemor-
relaxation properties on CMR.31,32 In Fig. 27.1 an axial section through rhage on the basis of its methemoglobin content using noncontrast
the thorax reveals a complex aortic plaque. Fig. 27.2 demonstrates a T1-weighted gradient recalled echo sequences, incorporating fat and
carotid plaque imaged at 3 T, with corresponding T1, T2, and PDW blood suppression. Methemoglobin is an intermediate breakdown product
images displayed. A final example of multicontrast CMR showing dif- of hemoglobin formed 12 to 72 hours following hemorrhage. It therefore
ferent plaque components and automatic segmentation of these plaques represents a key component of acute thrombus and is associated with
using a k-means cluster algorithm is shown in Fig. 27.3.33 a short T1 and high signal on T1-weighted imaging. This property
The lipid rich necrotic core can be identified in the carotid arteries allows CMR to detect fresh thrombus using T1-weighted images in a
and aorta using multicontrast weightings plus postcontrast T1-weighted range of conditions including deep venous thrombosis and pulmonary
imaging, with advances in CMR acquisition protocols allowing acquisi- embolism. This approach has been used in atherosclerosis to visualize
tion times to be reduced many-fold.14 The necrotic core usually appears regions of both intraplaque hemorrhage and endothelial thrombus
isointense to hyperintense on time-of-flight angiographic and precontrast related to plaque rupture/erosion. Indeed, the culprit carotid plaques
T1-weighted images and has minimal contrast enhancement compared of patients who have suffered a recent stroke consistently demonstrate
with the surrounding tissue on postcontrast T1-weighted images.34 this high T1-weighted signal.40,41
The combination of these images can therefore be used to identify
and quantify the lipid necrotic core burden. Yuan et al. demonstrated Early Imaging of Aortic Stenosis and Calcification
that multicontrast CMR of human carotid arteries had sensitivity and Histologic validation of this approach has also been provided using
specificity values of 85% and 92%, respectively, for the identification the carotid model in 63 patients undergoing endarterectomy. This
of a lipid core.35,36 demonstrated that high-intensity plaques had negative and positive
Alterations in the lipid necrotic core burden with therapy have also predictive values of 70% and 93%, respectively, for the detection of
been demonstrated and may be more sensitive to treatment effects than complex carotid lesions with evidence of surface rupture and intralu-
simple plaque burden measurements. The ORION trial assessed the minal or intraplaque hemorrhage.42 Moreover, imaging of hemorrhage
impact of rosuvastatin on carotid plaque volume and composition in and thrombus in the coronary arteries, where the small size of the vessel
43 patients treated for 2 years. Although no change in the plaque volume wall makes multicontrast imaging challenging, are the targets of intense
was observed, a reduction in the percentage of lipid necrotic core was research43-46 (see Chapter 3).
demonstrated.37 A subsequent study by Zhao and colleagues confirmed Importantly, unenhanced T1-weighted CMR imaging also appears to
provide important prognostic information. In one study, the presence
of high-intensity plaques in the carotid arteries predicted cardiac events,
outperforming carotid intimal medial thickness and traditional cardio-
vascular risk factors.47 This finding was corroborated in a subsequent
meta-analysis of 8 studies incorporating data from 689 patients, which
demonstrated a 6-fold increase in cerebrovascular events in patients
with high-intensity carotid plaques.48 The approach is also applicable
to the coronary arteries, with high-intensity plaque again appearing to
identify patients at increased risk of future coronary events.49 Noncontrast
T1-weighted imaging therefore holds major promise as a prognostic
marker, and in studying the natural history of plaque hemorrhage and the
prevalence and importance of subclinical atherosclerotic plaque rupture.

Angiogenesis
Pathological studies first revealed the central role of plaque neovascu-
larization in contributing to atherosclerotic plaque vulnerability.50 The
presence of neovessels is strongly associated with plaque inflammation,
macrophage content, and the likelihood of rupture,51 presumably because
they provide an alternative route for monocyte entry into the plaque
and because of the propensity of these vessels to leak and rupture.
Gadolinium (Gd) chelates represent the most commonly used CMR
contrast agents for imaging new plaque vessels. These paramagnetic agents
FIG. 27.1  An example of multicontrast plaque imaging with cardiovas- increase the blood signal on CMR images after intravenous injection
cular magnetic resonance. This axial section through the thoracic aorta and are therefore good candidates for measuring plaque neovasculature.
demonstrates an eccentrically shaped plaque involving the lateral wall Such techniques have been used in oncology imaging to quantify new
of the aorta. vessels associated with tumors.52 Kerwin and colleagues demonstrated
338 SECTION II  Ischemic Heart Disease

T2W TSE: 0.4 × 0.4 × 2 mm3

PDW T1W T2W


FIG. 27.2  An example of multicontrast cardiovascular magnetic resonance plaque imaging at high field
strength (3 T). A nonstenotic plaque is well visualized in the left carotid artery. The fibrous cap and lipid core
can be seen on the high-magnification images. PDW, Proton density weighted; T1W, T1 weighted; T2W, T2
weighted.

a correlation between the increased signal intensity in carotid atheroma between the atherosclerotic fibrous cap, which enhances with Gd, and
as a result of Gd-enhancement and the extent of plaque angiogenesis the necrotic core that lacks its own vasculature and so appears hypoin-
on histology. However, this imaging modality is not specific for neovas- tense. As discussed earlier, this approach can be used to estimate the
cularization and is limited because Gd chelates rapidly distribute into size of the lipid-rich necrotic core. However, in addition it can be used
the extracellular space.53 Dynamic contrast-enhanced (CE) imaging of to assess the fibrous cap in greater detail with respect to both its thick-
Gd-enhancement enables tracer kinetic modeling to characterize the ness and integrity. Indeed, LGE has been used to successfully image
leakiness of the neovessels by measuring a parameter called Ktrans as regions of both carotid plaque rupture and ulceration62 that may or
a potential marker for plaque vulnerability.54 In one study, neovessel may not be clinically apparent.63
abundance was correlated histologically with Gd uptake parameters
evaluated by dynamic CE CMR in a rabbit model.55 Relatively high Three-Dimensional Plaque Imaging
reproducibility of dynamic CE imaging has been shown to allow small As the utilization of plaque imaging has advanced, so has development
sample sizes.56 Another dynamic CE-CMR study demonstrated a reduc- of three-dimensional (3D) imaging to assess complex plaque without
tion in plaque neovasculature with longer statin therapy.57 In the coro- partial volume and re-slicing effects.64 Under development are 3D-imaging
nary arteries, some efforts have been made to image angiogenesis by techniques to assess plaque burden and positive remodeling, replicate
evaluating late Gd enhancement (LGE) in the coronary artery wall.58,59 multicontrast image weighting, as well as techniques focused on single
Further improvements in the quantification of plaque angiogenesis may plaque components such as plaque hemorrhage.65
be possible with novel intravascular albumin-binding Gd compounds
that have longer half-lives in the circulation.60,61 MOLECULAR MAGNETIC RESONANCE IMAGING OF
Fibrous Cap ATHEROSCLEROSIS
Imaging late after Gd administration can also provide information about Traditional plaque imaging modalities such as computed tomography
different plaque characteristics. In particular it can help differentiate (CT) and multicontrast CMR exploit anatomical variations between
CHAPTER 27  Atherosclerotic Plaque Imaging: Aorta and Carotid 339

T1W PDW T2W

df L - lumen

nc - necrotic core df
L L
lf H - intraplaque
hemorrhage lf

fc - fibrocellular tissue H lf
H
nc fc
fc df - dense fibrous tissue nc

lf - loose fibrous tissue

RGB Cluster
FIG. 27.3  Multicontrast cardiovascular magnetic resonance images of the carotid artery. Top row shows
T1-weighted (T1W), proton density weighted (PDW) and T2-weighted (T2W) images of the same atheroscle-
rotic plaque. The bottom left panel shows the red-green-blue (RGB) color composite image obtained by
mapping the T1W image to the red, the PDW image to the green, and the T2W image to blue channel,
respectively. The bottom right panel shows the plaque segmented automatically into its various components
using an automated k-means cluster analysis algorithm. Intraplaque hemorrhage (H), necrotic core (nc), loose
(lf), dense fibers (df) and the fibrous cap (fc) can be clearly differentiated.

tissues to provide images. Molecular (or target-specific) imaging unites accumulate in macrophages present in atherosclerotic plaques. Macro-
vascular biology with imaging modalities such as CMR and positron phages play a pivotal role in the destabilization of atherosclerotic plaques
emission tomography (PET), allowing us to study the activity of by secreting large quantities of fibrous cap-degrading metalloproteinases
specific pathological processes noninvasively, and with high-spatial (MMPs), along with proinflammatory cytokines and tissue factor.67
resolution. Iron oxide contrast agents have superparamagnetic properties (i.e., they
As we have seen, multicontrast CMR can characterize the various decrease T2* relaxation time by generating heterogeneities in the local
plaque components of intermediate to advanced atherosclerosis magnetic field) and can be detected on CMR as signal voids on T2*-
by exploiting pathological changes in tissue properties that lead to weighted sequences. USPIO plaque imaging with CMR has been validated
modified endogenous CMR contrast. However, the use of exogenous against histopathology at timepoints out to 8 weeks in an animal model.
imaging tracers targeting specific pathological processes (usually Iron staining closely matched that of macrophage distribution within
because of labeling with either Gd or iron oxide) has the potential to the plaque, but interestingly only a subset of smaller sized macrophages
greatly improve the sensitivity of the technique by amplifying the CMR actively accumulated USPIO.68,69
contrast.66 In clinical studies, Kooi and colleagues studied 11 symptomatic
Later we will discuss a variety of molecular imaging tracers that patients scheduled for carotid endarterectomy with USPIO-enhanced
may provide improved CMR imaging of atherosclerotic plaque. We will CMR. They reported a 24% decrease in signal intensity on correspond-
also discuss the synergistic role of PET and the potential utility of novel ing T2*-weighted sequences, and histologically verified uptake of USPIO
hybrid PET/CMR imaging systems. in 75% of ruptured or rupture-prone lesions.70 Trivedi et al. expanded
on this work and demonstrated that the optimum time for imaging
Plaque Inflammation symptomatic carotid plaque was 24 and 36 hours after USPIO injec-
Resident plaque macrophages have been successfully imaged using tion.71,72 USPIO imaging is also being explored in abdominal aortic
ultrasmall superparamagnetic particles of iron oxide (USPIO). These aneurysm disease, on the basis that inflamed aneurysms are likely to
are removed from the circulation by the reticuloendothelial system and expand more quickly and are more likely to rupture.73
340 SECTION II  Ischemic Heart Disease

USPIO imaging has also been used clinically to assess the antiin- infarction, demonstrating that >50% of the culprit thrombi were at
flammatory effects of novel therapies. The ATHEROMA (Atorvastatin least days or weeks old.84 Presumably, microplaque rupture events with
Therapy: Effects on Reduction of Macrophage Activity) study random- thrombus formation often predate the catastrophic rupture event that
ized 47 patients with carotid stenosis >40% and increased USPIO uptake causes the clinical syndrome. The ability to detect these early warnings
on baseline scanning to either 10 mg or 80 mg atorvastatin therapy for might therefore allow intense therapy or device placement to avert
12 weeks. A significant decrease in USPIO carotid plaque uptake was symptoms.
observed in the high-dose cohort but not the low-dose group.74 Aside from the T1-weighted CMR imaging approach described earlier,
Macrophages within atherosclerotic plaques have also been targeted several molecular techniques have been taken to image thrombus with
for imaging with Gd-loaded immunomicelles. These agents, with diam- CMR. Yu et al. used a Gd-loaded nanoparticle coupled to a fibrin anti-
eters between 20 and 120 nm, are composed of phospholipids, a sur- body and tested this against in vitro thrombus.85 They demonstrated
factant, and an aliphatic chain with Gd-diethylenetriaminepentaacetic significant signal enhancement and confirmed tight binding of the
acid (Gd-DTPA) attached at the polar head group. The polar head antibody to the thrombus with scanning electron microscopy. A similar
group of the aliphatic chain can be attached to antibodies directly or method, but using a different nanoparticle construct, was employed
via a biotin-avidin bridge. Using this model, we have made micelles successfully by Winter.81
that have over 10,000 Gd ions on each micelle surface and the ability A fibrin-specific CMR contrast agent has also been designed. With
to specifically target the macrophage scavenger receptor. Initial work this agent, thrombus resulting from plaque rupture has been identified
demonstrated that these immunomicelles resulted in enhanced signal using CMR in a rabbit model. In the 25 arterial thrombi induced by
in murine atherosclerotic plaque.75 carotid crush injury, Botnar et al. demonstrated a sensitivity and speci-
ficity of 100% for in vivo thrombus detection.86 Sirol et al. used the
Endothelial Adhesion Molecules same fibrin-specific CMR contrast agent (EP-1242) in 12 guinea pigs
Expression of endothelial adhesion molecules such as VCAM and to demonstrate that the signal intensity of the thrombus was increased
selectins occurs early in the development of atherosclerosis, driving by over 4-fold compared with noncontrast images. The detection of
progressive macrophage recruitment in to the plaques. These are poten- thrombi improved from 42% pickup precontrast injection compared
tial imaging targets in cardiovascular disease76 and other conditions.77 with 100% detection after injection.87 Finally, another experimental
The ability to image VCAM was elegantly demonstrated by Kelly et al. fibrin-targeted peptide (EP-2104R) for thrombus detection allowed
They used a superparamagnetic fluorescent nanoparticle coupled to a discrimination between occlusive and nonocclusive thrombi, and track-
payload peptide that was internalized by endothelial cells expressing ing of intravascular thrombus as it aged and became more organized
VCAM. This was tested in ApoE-/- mice fed a high cholesterol diet by fibrous tissue infiltration88 (Fig. 27.4).
and was highly specific for VCAM-expressing cells, compared with
immunohistochemistry.78
CMR imaging of the selectin family of molecules has also been
attempted, at least in vitro. Kang et al. showed that a monoclonal anti-
body fragment tagged with iron oxide nanoparticles was specific for
human endothelial cells expressing E-selectin in culture, with an increased
binding of 200 times compared with controls.79 Intracellular adhesion A B C
molecule 1 (ICAM-1) receptors expressed on the cerebral arterial vas-
cular endothelium have been imaged with CMR using antibody-
conjugated paramagnetic liposomes.80 CMR provided sufficient signal T1W Non-CE CMR
enhancement to determine the areas of increased expression, and binding 3 T2W Non-CE CMR
was verified by fluorescent histopathology. 2.5 EP-2104R
Signal intensity

2
Angiogenesis
A novel agent targeting the integrin αvβ3 (specifically expressed on the 1.5
endothelial surface of neovasculature) has been developed to identify 1
regions in the vessel wall undergoing angiogenesis. Winter et al. dem-
0.5
onstrated in a rabbit model of atherosclerosis that regions of neovas-
cularization in plaques had a 47% increase in signal intensity on CMR 0
after the injection of αvβ3-targeted nanoparticles.81 Another epitope 0 1 2 3 4 6 8
that has been successfully exploited for imaging plaque angiogenesis is D Weeks after thrombus induction
fibronectin. The binding of an antifibronectin antibody was confirmed FIG. 27.4  Axial cardiovascular magnetic resonance (CMR) images of a
in ApoE-/- mice, both autoradiographically and by the use of a fluo- rabbit carotid artery one week after thrombus induction, imaged using
rescent probe, demonstrating specificity for the vasovasorum of the a double inversion recovery turbo-spin echo sequence. (A) T1-weighted
atheroscleorisc plaque.82 (T1W) and (B) T2-weighted (T2W) images were obtained without any
injection of contrast agent. White arrow indicates location of the throm-
Thrombus bus. (C) T1W images obtained 30 minutes after EP-2104R injection.
(D) Relative signal intensity changes (mean ± standard deviation) over
Intraluminal thrombosis represents the final step in the evolution of
time for T1W (red circles), T2W (blue squares) and after EP-2104R
vulnerable atherosclerotic plaque and is therefore a candidate target
injection (yellow triangles). This gadolinium-based, fibrin-targeted CMR
for novel-specific CMR contrast agents. Histologic studies have dem- contrast agent demonstrates significant enhancement of the thrombus
onstrated that superficial thrombus superimposed on a ruptured ath- compared with T1W images (P < .001). (Modified from Sirol M, Fuster
erosclerotic plaque characterizes those plaques at high risk of ischemic V, Badimon JJ, et al. Chronic thrombus detection with in vivo mag-
events.83 Moreover, a clinical study analyzed the histology of intra- netic resonance imaging and a fibrin-targeted contrast agent. Circulation.
coronary thrombus aspirated from 211 patients with acute myocardial 2005;112:1594–1600.)
CHAPTER 27  Atherosclerotic Plaque Imaging: Aorta and Carotid 341

ApoA-I Phospholipid

Cholesterol
ester B C

Triglyceride

Unesterified
cholesterol
Gd-DTPA-DMPE
A D E

FIG. 27.5  (A) This represents the reconstituted high density lipoprotein (HDL)-like cardiovascular magnetic
resonance contrast agent composed of an HDL-like particle and a phospholipid-based contrast agent (Gd-DTPA-
DMPE). Axial in vivo magnetic resonance images of the abdominal aorta in an 8-week-old mouse at 9.4 T
before (B), 1 hour (C), 24 hours (D), and 48 hours (E) after the injection of recombinant HDL-like nanoparticles.
The insets denote the magnification of the aortic region. ApoA-I, Apolipoprotein A1. (Modified from Frias JC,
Williams KJ, Fisher EA, Fayad ZA. Recombinant HDL-like nanoparticles: a specific contrast agent for MRI of
atherosclerotic plaques. J Am Chem Soc. 2004;126:16316–16317.)

27.5 demonstrates the enhancement of atherosclerotic plaques after the


Extracellular Matrix injection of recombinant HDL.
Other novel CMR contrast agents have been found to accumulate within
atherosclerotic plaques. For example, gadofluorine M is a lipophilic, HYBRID POSITRON EMISSION TOMOGRAPHY/
macrocyclic, water-soluble, Gd chelate complex with a perfluorinated
side chain. Both Sirol and Barkhausen separately demonstrated that
CARDIAC MAGNETIC RESONANCE IMAGING
gadofluorine M significantly increased signal intensity in rabbit aortic PET is a molecular imaging technique that is highly sensitive to the
plaques compared with controls. A strong correlation was found between activity of specific disease processes that can be targeted with specially
the intensity of CMR signal enhancement after the injection of gado- designed radiotracers. This sensitivity gives it major advantages over
fluorine and the presence of lipid-rich plaques on corresponding his- CMR-based molecular imaging techniques; however, PET remains limited
tologic sections.89,90 This suggests a high affinity of gadofluorine M for by its low spatial resolution. With the availability of PET/CMR systems
atherosclerotic plaque. There is now emerging data that gadofluorine the capability now exists to combine functional information from PET
M is restricted to the extracellular space of plaques and may interact with the anatomic detail and soft-tissue characterization of CMR, com-
with resident proteins in the extracellular matrix milieu. bining the major advantages of these two imaging approaches (Fig.
27.6). Once again the absence of radiation associated with MR means
High-Density Lipoprotein Imaging these hybrid systems offer considerable potential advantages to the more
Our group developed another type of imaging agent based on a recom- established and widely available PET/CT scanners.
binant high-density lipoprotein (HDL) molecule that incorporates Arterial inflammation is a key driver of atherosclerosis and, in par-
Gd-DTPA phospholipids.91 The natural role of HDL in the body is ticular, the precipitation of acute plaque rupture and adverse cardio-
to remove lipid from atherosclerotic plaque and return it to the liver vascular events. 18F-fluorodeoxyglucose (18F-FDG) is a glucose analog
(reverse cholesterol transport). Elevated levels of HDL are associated that identifies macrophages, which use substantially more glucose than
with a reduction in plaque rupture events, presumably because of this neighboring cell types in the vasculature,93 particularly in hypoxic condi-
protective effect.92 The recombinant HDL imaging agent has a small tions.94 18F-FDG uptake has been demonstrated to be increased in the
diameter (7–12 nm) allowing it to diffuse into atherosclerotic plaques; culprit carotid plaques poststroke/TIA compared with asymptomatic
however, because it uses endogenous transport molecules, it does not contralateral lesions95 and associated with cardiovascular risk factors.96
trigger any immune reaction. Atherosclerotic plaques demonstrated a Moreover early studies suggests that the detection of increased vascular
35% increase of CMR signal intensity 24 hours after the injection of 18F-FDG activity might identify patients with an adverse prognosis.97,98
these recombinant HDL particles in an ApoE knockout mouse model. Importantly the arterial 18F-FDG signal is modifiable with drug
Furthermore, fluorescent recombinant HDL colocalized with macro- therapy, so that this technique is being increasingly employed as an
phages present in atherosclerotic plaques with confocal microscopy. Fig. end-point in trials assessing the antiinflammatory effects of novel
342 SECTION II  Ischemic Heart Disease

CONCLUSION AND FUTURE DIRECTIONS


Thanks to the absence of ionizing radiation, CMR represents the imaging
technology of choice for the noninvasive high-spatial resolution detec-
tion and serial monitoring of atherosclerotic plaques in the carotid
arteries and aorta. High image quality and sensitivity to small changes
in plaque size mean that there is little variance between measurements,
permitting small sample sizes to be used in comparative studies. Thus
multicontrast CMR is ideally suited for use in evaluation of novel anti-
atheroma drugs.
The development of functional molecular imaging of atheroscle-
rosis may also help to reveal the key pathological steps that lead from
a stable atherosclerotic plaque to an acute ischemic event. However,
clinical studies have underscored the multiple locations of vulnerable
and ruptured atherosclerotic plaques and the diffuse inflammation of
the arterial tree in patients with acute ischemic events compared with
stable patients. Therefore the concept of detecting infrequent vulnerable
atherosclerotic plaques with imaging and treating them individually
has started to shift to a more global process of identifying vulnerable
patients at high risk of acute clinical events, irrespective of the arterial
location.110
In the future, CMR atherosclerosis imaging may help to focus
individual evaluation of cardiovascular risk and to optimize antiath-
FIG. 27.6  Hybrid positron emission tomography/cardiovascular mag- erosclerotic therapies with hybrid PET/CMR allowing advanced mul-
netic resonance image in the axial plane demonstrating increased 18F- tiparametric imaging.
fluorodeoxyglucose signal localizing to a region of carotid atheroma.

ACKNOWLEDGMENTS AND FUNDING


26,99,100
antiatherosclerosis drugs in the carotid arteries and thoracic aorta. M.R.D. is supported by the British Heart Foundation (FS/14/78/31020)
The effect of therapy can be detected as early as 3 to 6 months, which and is the recipient of the Sir Jules Thorn Biomedical Research Award
is well before changes can be observed using CMR alone. Widely avail- 2015. J.H.F.R. is part-supported by the NIHR Cambridge Biomedical
able 18F-FDG is limited by its lack of specificity, leading to interest in Research Centre, the British Heart Foundation, the EPSRC, and the
more specific PET markers of inflammation101,102 and tracers targeting Wellcome Trust. ZAF is part supported by NIH/NHLBI R01 HL071021.
other disease processes of interest such as angiogenesis, microcalcifica-
tion, and plaque hypoxia.103-107 With the advent of such novel tracers,
PET/CMR holds major potential as an imaging tool for the study of
REFERENCES
carotid atheroma108 and cardiovascular disease in general.109 A full reference list is available online at ExpertConsult.com
CHAPTER 27  Atherosclerotic Plaque Imaging: Aorta and Carotid 342.e1

REFERENCES 22. Sun J, Zhao X-Q, Balu N, et al. Carotid magnetic resonance imaging for
monitoring atherosclerotic plaque progression: a multicenter
1. Baber U, Mehran R, Sartori S, et al. Prevalence, impact, and predictive reproducibility study. Int J Cardiovasc Imaging. 2015;31:95–103.
value of detecting subclinical coronary and carotid atherosclerosis in 23. Saam T, Kerwin WS, Chu B, et al. Sample size calculation for clinical
asymptomatic adults: the BioImage study. J Am Coll Cardiol. 2015;65: trials using magnetic resonance imaging for the quantitative assessment
1065–1074. of carotid atherosclerosis. J Cardiovasc Magn Reson. 2005;7:799–808.
2. Ku DN, Giddens DP, Zarins CK, Glagov S. Pulsatile flow and 24. Yonemura A, Momiyama Y, Fayad ZA, et al. Effect of lipid-lowering
atherosclerosis in the human carotid bifurcation. Positive correlation therapy with atorvastatin on atherosclerotic aortic plaques detected by
between plaque location and low oscillating shear stress. Arteriosclerosis. noninvasive magnetic resonance imaging. J Am Coll Cardiol.
1985;5:293–302. 2005;45:733–742.
3. Cunningham KS, Gotlieb AI. The role of shear stress in the pathogenesis 25. Corti R, Fuster V, Fayad ZA, et al. Effects of aggressive versus
of atherosclerosis. Lab Invest. 2005;85:9–23. conventional lipid-lowering therapy by simvastatin on human
4. Cushing SD, Berliner JA, Valente AJ, et al. Minimally modified low atherosclerotic lesions: a prospective, randomized, double-blind trial
density lipoprotein induces monocyte chemotactic protein 1 in human with high-resolution magnetic resonance imaging. J Am Coll Cardiol.
endothelial cells and smooth muscle cells. Proc Natl Acad Sci USA. 2005;46:106–112.
1990;87:5134–5138. 26. Fayad ZA, Mani V, Woodward M, et al. Safety and efficacy of dalcetrapib
5. Hamilton JA, Myers D, Jessup W, et al. Oxidized LDL can induce on atherosclerotic disease using novel non-invasive multimodality
macrophage survival, DNA synthesis, and enhanced proliferative response imaging (dal-PLAQUE): a randomised clinical trial. Lancet.
to CSF-1 and GM-CSF. Arterioscler Thromb Vasc Biol. 1999;19:98–105. 2011;378:1547–1559.
6. Glagov S, Weisenberg E, Zarins CK, Stankunavicius R, Kolettis GJ. 27. Arbab-Zadeh A, Fuster V. The myth of the “vulnerable plaque”:
Compensatory enlargement of human atherosclerotic coronary arteries. transitioning from a focus on individual lesions to atherosclerotic
N Engl J Med. 1987;316:1371–1375. disease burden for coronary artery disease risk assessment. J Am Coll
7. Fuster V, Moreno PR, Fayad ZA, Corti R, Badimon JJ. Atherothrombosis Cardiol. 2015;65:846–855.
and high-risk plaque: part I: evolving concepts. J Am Coll Cardiol. 28. Motoyama S, Ito H, Sarai M, et al. Plaque characterization by coronary
2005;46:937–954. computed tomography angiography and the likelihood of acute
8. Farb A, Burke AP, Tang AL, et al. Coronary plaque erosion without coronary events in mid-term follow-up. J Am Coll Cardiol. 2015;
rupture into a lipid core. A frequent cause of coronary thrombosis in 66:337–346.
sudden coronary death. Circulation. 1996;93:1354–1363. 29. Kim WY, Stuber M, Börnert P, Kissinger KV, Manning WJ, Botnar RM.
9. Mann J, Davies MJ. Mechanisms of progression in native coronary Three-dimensional black-blood cardiac magnetic resonance coronary
artery disease: role of healed plaque disruption. Heart. 1999;82:265–268. vessel wall imaging detects positive arterial remodeling in patients with
10. Davies MJ. The pathophysiology of acute coronary syndromes. Heart. nonsignificant coronary artery disease. Circulation. 2002;106:296–299.
2000;83:361–366. 30. Miao C, Chen S, Macedo R, et al. Positive remodeling of the coronary
11. Bennett MR, Evan GI, Schwartz SM. Apoptosis of human vascular arteries detected by magnetic resonance imaging in an asymptomatic
smooth muscle cells derived from normal vessels and coronary population. J Am Coll Cardiol. 2009;53:1708–1715.
atherosclerotic plaques. J Clin Invest. 1995;95:2266–2274. 31. Fayad ZA, Fuster V. Characterization of atherosclerotic plaques by
12. Tarkin JM, Dweck MR, Evans NR, et al. Imaging atherosclerosis. Circ magnetic resonance imaging. Ann N Y Acad Sci. 2000;902:173–186.
Res. 2016;118:750–769. 32. Singh N, Moody AR, Roifman I, Bluemke DA, Zavodni AEH. Advanced
13. Cannon CP, Braunwald E, McCabe CH, et al. Intensive versus moderate MRI for carotid plaque imaging. Int J Cardiovasc Imaging. 2016;32:83–89.
lipid lowering with statins after acute coronary syndromes. N Engl J 33. Itskovich VV, Choudhury RP, Aguinaldo JGS, et al. Characterization of
Med. 2004;350:1495–1504. aortic root atherosclerosis in ApoE knockout mice: high-resolution in
14. Mani V, Itskovich VV, Szimtenings M, et al. Rapid extended coverage vivo and ex vivo MRM with histological correlation. Magn Reson Med.
simultaneous multisection black-blood vessel wall MR imaging. 2003;49:381–385.
Radiology. 2004;232:281–288. 34. Zhao X-Q, Dong L, Hatsukami T, et al. MR imaging of carotid plaque
15. Fayad ZA, Fuster V. Clinical imaging of the high-risk or vulnerable composition during lipid-lowering therapy a prospective assessment of
atherosclerotic plaque. Circ Res. 2001;89:305–316. effect and time course. JACC Cardiovasc Imaging. 2011;4:977–986.
16. Jaffer FA, O’Donnell CJ, Larson MG, et al. Age and sex distribution of 35. Yuan C, Mitsumori LM, Beach KW, Maravilla KR. Carotid
subclinical aortic atherosclerosis: a magnetic resonance imaging atherosclerotic plaque: noninvasive MR characterization and
examination of the Framingham Heart Study. Arterioscler Thromb Vasc identification of vulnerable lesions. Radiology. 2001;221:285–299.
Biol. 2002;22:849–854. 36. Yuan C, Mitsumori LM, Ferguson MS, et al. In vivo accuracy of
17. Taniguchi H, Momiyama Y, Fayad ZA, et al. In vivo magnetic resonance multispectral magnetic resonance imaging for identifying lipid-rich
evaluation of associations between aortic atherosclerosis and both risk necrotic cores and intraplaque hemorrhage in advanced human carotid
factors and coronary artery disease in patients referred for coronary plaques. Circulation. 2001;104:2051–2056.
angiography. Am Heart J. 2004;148:137–143. 37. Underhill HR, Yuan C, Zhao X-Q, et al. Effect of rosuvastatin therapy
18. Underhill HR, Kerwin WS, Hatsukami TS, Yuan C. Automated on carotid plaque morphology and composition in moderately
measurement of mean wall thickness in the common carotid artery by hypercholesterolemic patients: a high-resolution magnetic resonance
MRI: a comparison to intima-media thickness by B-mode ultrasound. imaging trial. Am Heart J. 2008;155:584.e1–584.e8.
J Magn Reson Imaging. 2006;24:379–387. 38. Kolodgie FD, Gold HK, Burke AP, et al. Intraplaque hemorrhage and
19. Cai J-M, Hatsukami TS, Ferguson MS, Small R, Polissar NL, Yuan C. progression of coronary atheroma. N Engl J Med. 2003;349:2316–2325.
Classification of human carotid atherosclerotic lesions with in vivo 39. Chu B, Kampschulte A, Ferguson MS, et al. Hemorrhage in the
multicontrast magnetic resonance imaging. Circulation. 2002;106: atherosclerotic carotid plaque: a high-resolution MRI study. Stroke.
1368–1373. 2004;35:1079–1084.
20. Mani V, Muntner P, Gidding SS, et al. Cardiovascular magnetic 40. Moody AR, Allder S, Lennox G, Gladman J, Fentem P. Direct magnetic
resonance parameters of atherosclerotic plaque burden improve resonance imaging of carotid artery thrombus in acute stroke. Lancet.
discrimination of prior major adverse cardiovascular events. 1999;353:122–123.
J Cardiovasc Magn Reson. 2009;11:10. 41. Murphy RE. Prevalence of complicated carotid atheroma as detected by
21. Li AE, Kamel I, Rando F, et al. Using MRI to assess aortic wall thickness magnetic resonance direct thrombus imaging in patients with suspected
in the multiethnic study of atherosclerosis: distribution by race, sex, and carotid artery stenosis and previous acute cerebral ischemia. Circulation.
age. AJR Am J Roentgenol. 2004;182:593–597. 2003;107:3053–3058.
342.e2 SECTION II  Ischemic Heart Disease

42. Moody AR, Murphy RE, Morgan PS, et al. Characterization of 61. Kelle S, Thouet T, Tangcharoen T, et al. Whole-heart coronary magnetic
complicated carotid plaque with magnetic resonance direct thrombus resonance angiography with MS-325 (Gadofosveset). Med Sci Monit.
imaging in patients with cerebral ischemia. Circulation. 2007;13:CR469–CR474.
2003;107:3047–3052. 62. Hatsukami TS, Ross R, Polissar NL, Yuan C. Visualization of fibrous cap
43. Jansen CHP, Perera D, Makowski MR, et al. Detection of intracoronary thickness and rupture in human atherosclerotic carotid plaque in vivo
thrombus by magnetic resonance imaging in patients with acute with high-resolution magnetic resonance imaging. Circulation.
myocardial infarction. Circulation. 2011;124:416–424. 2000;102:959–964.
44. Ehara S, Hasegawa T, Nakata S, et al. Hyperintense plaque identified by 63. Lovett JK, Gallagher PJ, Hands LJ, Walton J, Rothwell PM. Histological
magnetic resonance imaging relates to intracoronary thrombus as correlates of carotid plaque surface morphology on lumen contrast
detected by optical coherence tomography in patients with angina imaging. Circulation. 2004;110:2190–2197.
pectoris. Eur Heart J Cardiovasc Imaging. 2012;13:394–399. 64. Balu N, Yarnykh VL, Chu B, Wang J, Hatsukami T, Yuan C. Carotid
45. Kawasaki T, Koga S, Koga N, et al. Characterization of hyperintense plaque assessment using fast 3D isotropic resolution black-blood MRI.
plaque with noncontrast T1-weighted cardiac magnetic resonance Magn Reson Med. 2011;65:627–637.
coronary plaque imaging. JACC Cardiovasc Imaging. 2009;2:720–728. 65. Wang J, Börnert P, Zhao H, et al. Simultaneous noncontrast angiography
46. Noguchi T, Kawasaki T, Tanaka A, et al. High-intensity signals in and intraplaque hemorrhage (SNAP) imaging for carotid atherosclerotic
coronary plaques on noncontrast T1-weighted magnetic resonance disease evaluation. Magn Reson Med. 2013;69:337–345.
imaging as a novel determinant of coronary events. J Am Coll Cardiol. 66. Choudhury RP, Fuster V, Fayad ZA. Molecular, cellular and functional
2014;63:989–999. imaging of atherothrombosis. Nat Rev Drug Discov. 2004;3:913–925.
47. Noguchi T, Yamada N, Higashi M, Goto Y, Naito H. High-intensity 67. Libby P. Mechanisms of acute coronary syndromes and their
signals in carotid plaques on T1-weighted magnetic resonance imaging implications for therapy. N Engl J Med. 2013;368:2004–2013.
predict coronary events in patients with coronary artery disease. J Am 68. Hyafil F, Laissy J-P, Mazighi M, et al. Ferumoxtran-10-enhanced MRI of
Coll Cardiol. 2011;58:416–422. the hypercholesterolemic rabbit aorta: relationship between signal loss
48. Saam T, Hetterich H, Hoffmann V, et al. Meta-analysis and systematic and macrophage infiltration. Arterioscler Thromb Vasc Biol.
review of the predictive value of carotid plaque hemorrhage on 2006;26:176–181.
cerebrovascular events by magnetic resonance imaging. J Am Coll 69. Yancy AD, Olzinski AR, Hu TC-C, et al. Differential uptake of
Cardiol. 2013;62:1081–1091. ferumoxtran-10 and ferumoxytol, ultrasmall superparamagnetic iron
49. Noguchi T, Kawasaki T, Tanaka A, et al. High-intensity signals in oxide contrast agents in rabbit: critical determinants of atherosclerotic
coronary plaques on noncontrast T1-weighted magnetic resonance plaque labeling. J Magn Reson Imaging. 2005;21:432–442.
imaging as a novel determinant of coronary events. J Am Coll Cardiol. 70. Kooi ME, Cappendijk VC, Cleutjens KBJM, et al. Accumulation of
2014;63:989–999. ultrasmall superparamagnetic particles of iron oxide in human
50. Kockx MM, Cromheeke KM, Knaapen MWM, et al. Phagocytosis and atherosclerotic plaques can be detected by in vivo magnetic resonance
macrophage activation associated with hemorrhagic microvessels in imaging. Circulation. 2003;107:2453–2458.
human atherosclerosis. Arterioscler Thromb Vasc Biol. 2003;23: 71. Trivedi RA, Mallawarachi C, U-King-Im J-M, et al. Identifying inflamed
440–446. carotid plaques using in vivo USPIO-enhanced MR imaging to label
51. Kumamoto M, Nakashima Y, Sueishi K. Intimal neovascularization in plaque macrophages. Arterioscler Thromb Vasc Biol. 2006;26:1601–1606.
human coronary atherosclerosis: its origin and pathophysiological 72. Trivedi RA, U-King-Im J-M, Graves MJ, et al. In vivo detection of
significance. Hum Pathol. 1995;26:450–456. macrophages in human carotid atheroma: temporal dependence of
52. Jacobs MA, Barker PB, Argani P, Ouwerkerk R, Bhujwalla ZM, Bluemke ultrasmall superparamagnetic particles of iron oxide-enhanced MRI.
DA. Combined dynamic contrast enhanced breast MR and proton Stroke. 2004;35:1631–1635.
spectroscopic imaging: a feasibility study. J Magn Reson Imaging. 73. Richards JMJ, Semple SI, Macgillivray TJ, et al. Abdominal aortic
2005;21:23–28. aneurysm growth predicted by uptake of ultrasmall superparamagnetic
53. Kerwin W, Hooker A, Spilker M, et al. Quantitative magnetic resonance particles of iron oxide: a pilot study. Circ Cardiovasc Imaging.
imaging analysis of neovasculature volume in carotid atherosclerotic 2011;4:274–281.
plaque. Circulation. 2003;107:851–856. 74. Tang TY, Howarth SPS, Miller SR, et al. The ATHEROMA (Atorvastatin
54. Sourbron SP, Buckley DL. Classic models for dynamic contrast- Therapy: Effects on Reduction of Macrophage Activity) study.
enhanced MRI. NMR Biomed. 2013;26:1004–1027. Evaluation using ultrasmall superparamagnetic iron oxide-enhanced
55. Calcagno C, Cornily J-C, Hyafil F, et al. Detection of neovessels in magnetic resonance imaging in carotid disease. J Am Coll Cardiol.
atherosclerotic plaques of rabbits using dynamic contrast enhanced 2009;53:2039–2050.
MRI and 18F-FDG PET. Arterioscler Thromb Vasc Biol. 2008;28:1311– 75. Lipinski MJ, Fuster V, Fisher EA, Fayad ZA. Technology insight: targeting
1317. of biological molecules for evaluation of high-risk atherosclerotic
56. Chen H, Sun J, Kerwin WS, et al. Scan-rescan reproducibility of plaques with magnetic resonance imaging. Nat Clin Pract Cardiovasc
quantitative assessment of inflammatory carotid atherosclerotic plaque Med. 2004;1:48–55.
using dynamic contrast-enhanced 3T CMR in a multi-center study. J 76. McAteer MA, Schneider JE, Ali ZA, et al. Magnetic resonance imaging of
Cardiovasc Magn Reson. 2014;16:51. endothelial adhesion molecules in mouse atherosclerosis using
57. O’Brien KD, Hippe DS, Chen H, et al. Longer duration of statin therapy dual-targeted microparticles of iron oxide. Arterioscler Thromb Vasc Biol.
is associated with decreased carotid plaque vascularity by magnetic 2008;28:77–83.
resonance imaging. Atherosclerosis. 2016;245:74–81. 77. McAteer MA, Sibson NR, zur Muhlen von C, et al. In vivo magnetic
58. Yeon SB, Sabir A, Clouse M, et al. Delayed-enhancement cardiovascular resonance imaging of acute brain inflammation using microparticles of
magnetic resonance coronary artery wall imaging: comparison with iron oxide. Nat Med. 2007;13:1253–1258.
multislice computed tomography and quantitative coronary 78. Kelly KA, Allport JR, Tsourkas A, Shinde-Patil VR, Josephson L,
angiography. J Am Coll Cardiol. 2007;50:441–447. Weissleder R. Detection of vascular adhesion molecule-1 expression
59. Ibrahim T, Makowski MR, Jankauskas A, et al. Serial contrast-enhanced using a novel multimodal nanoparticle. Circ Res. 2005;96:327–336.
cardiac magnetic resonance imaging demonstrates regression of 79. Kang HW, Josephson L, Petrovsky A, Weissleder R, Bogdanov A.
hyperenhancement within the coronary artery wall in patients after Magnetic resonance imaging of inducible E-selectin expression in
acute myocardial infarction. JACC Cardiovasc Imaging. 2009;2:580–588. human endothelial cell culture. Bioconjug Chem. 2002;13:122–127.
60. Tang L, Merkle N, Schär M, et al. Volume-targeted and whole-heart 80. Sipkins DA, Gijbels K, Tropper FD, Bednarski M, Li KC, Steinman L.
coronary magnetic resonance angiography using an intravascular ICAM-1 expression in autoimmune encephalitis visualized using
contrast agent. J Magn Reson Imaging. 2009;30:1191–1196. magnetic resonance imaging. J Neuroimmunol. 2000;104:1–9.
CHAPTER 27  Atherosclerotic Plaque Imaging: Aorta and Carotid 342.e3

81. Winter PM, Caruthers SD, Yu X, et al. Improved molecular imaging 96. Tahara N, Kai H, Yamagishi S-I, et al. Vascular inflammation evaluated
contrast agent for detection of human thrombus. Magn Reson Med. by [18F]-fluorodeoxyglucose positron emission tomography is associated
2003;50:411–416. with the metabolic syndrome. J Am Coll Cardiol. 2007;49:1533–1539.
82. Matter CM, Schuler PK, Alessi P, et al. Molecular imaging of 97. Marnane M, Merwick A, Sheehan OC, et al. Carotid plaque
atherosclerotic plaques using a human antibody against the extra- inflammation on 18F-fluorodeoxyglucose positron emission tomography
domain B of fibronectin. Circ Res. 2004;95:1225–1233. predicts early stroke recurrence. Ann Neurol. 2012;71:709–718.
83. Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from 98. Figueroa AL, Abdelbaky A, Truong QA, et al. Measurement of arterial
sudden coronary death: a comprehensive morphological classification activity on routine FDG PET/CT images improves prediction of risk of
scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol. future CV events. JACC Cardiovasc Imaging. 2013;6:1250–1259.
2000;20:1262–1275. 99. Tahara N, Kai H, Ishibashi M, et al. Simvastatin attenuates plaque
84. Rittersma SZH, van der Wal AC, Koch KT, et al. Plaque instability inflammation: evaluation by fluorodeoxyglucose positron emission
frequently occurs days or weeks before occlusive coronary thrombosis: a tomography. J Am Coll Cardiol. 2006;48:1825–1831.
pathological thrombectomy study in primary percutaneous coronary 100. Tawakol A, Singh P, Rudd JHF, et al. Effect of treatment for 12 weeks
intervention. Circulation. 2005;111:1160–1165. with rilapladib, a lipoprotein-associated phospholipase A2 inhibitor, on
85. Yu X, Song SK, Chen J, et al. High-resolution MRI characterization of arterial inflammation as assessed with 18F-fluorodeoxyglucose-positron
human thrombus using a novel fibrin-targeted paramagnetic emission tomography imaging. J Am Coll Cardiol. 2014;63:86–88.
nanoparticle contrast agent. Magn Reson Med. 2000;44:867–872. 101. Tahara N, Mukherjee J, de Haas HJ, et al. 2-Deoxy-2-[(18)F]fluoro-D-
86. Botnar RM, Perez AS, Witte S, et al. In vivo molecular imaging of acute mannose positron emission tomography imaging in atherosclerosis. Nat
and subacute thrombosis using a fibrin-binding magnetic resonance Med. 2014;20:215–219.
imaging contrast agent. Circulation. 2004;109:2023–2029. 102. Rominger A, Saam T, Vogl E, et al. In vivo imaging of macrophage
87. Sirol M, Fuster V, Badimon JJ, et al. Chronic thrombus detection with in activity in the coronary arteries using 68Ga-DOTATATE PET/CT:
vivo magnetic resonance imaging and a fibrin-targeted contrast agent. correlation with coronary calcium burden and risk factors. J Nucl Med.
Circulation. 2005;112:1594–1600. 2010;51:193–197.
88. Sirol M, Aguinaldo JGS, Graham PB, et al. Fibrin-targeted contrast agent 103. Dweck MR, Jones C, Joshi NV, et al. Assessment of valvular calcification
for improvement of in vivo acute thrombus detection with magnetic and inflammation by positron emission tomography in patients with
resonance imaging. Atherosclerosis. 2005;182:79–85. aortic stenosis. Circulation. 2012;125:76–86.
89. Sirol M, Itskovich VV, Mani V, et al. Lipid-rich atherosclerotic plaques 104. Jenkins WSA, Vesey AT, Shah ASV, et al. Valvular (18)F-fluoride and (18)
detected by gadofluorine-enhanced in vivo magnetic resonance imaging. F-fluorodeoxyglucose uptake predict disease progression and clinical
Circulation. 2004;109:2890–2896. outcome in patients with aortic stenosis. J Am Coll Cardiol. 2015;66:
90. Barkhausen J, Ebert W, Heyer C, Debatin JF, Weinmann H-J. Detection 1200–1201.
of atherosclerotic plaque with Gadofluorine-enhanced magnetic 105. Irkle A, Vesey AT, Lewis DY, et al. Identifying active vascular
resonance imaging. Circulation. 2003;108:605–609. microcalcification by 18F-sodium fluoride positron emission
91. Frias JC, Williams KJ, Fisher EA, Fayad ZA. Recombinant HDL-like tomography. Nat Commun. 2015;6:1–11.
nanoparticles: a specific contrast agent for MRI of atherosclerotic 106. Dweck MR, Chow MWL, Joshi NV, et al. Coronary arterial 18F-sodium
plaques. J Am Chem Soc. 2004;126:16316–16317. fluoride uptake. J Am Coll Cardiol. 2012;59:1539–1548.
92. Wilson PWF, D’Agostino RB, Parise H, Sullivan L, Meigs JB. Metabolic 107. Joshi NV, Vesey AT, Williams MC, et al. 18F-fluoride positron emission
syndrome as a precursor of cardiovascular disease and type 2 diabetes tomography for identification of ruptured and high-risk coronary
mellitus. Circulation. 2005;112:3066–3072. atherosclerotic plaques: a prospective clinical trial. Lancet. 2014;383:
93. Weisdorf DJ, Craddock PR, Jacob HS. Granulocytes utilize different 705–713.
energy sources for movement and phagocytosis. Inflammation. 108. Vesey AT, Dweck MR, Fayad ZA. Utility of combining PET and MR
1982;6:245–256. imaging of carotid plaque. Neuroimaging Clin N Am. 2016;26:55–68.
94. Folco EJ, Sheikine Y, Rocha VZ, et al. Hypoxia but not inflammation 109. Abgral R, Dweck MR, Trivieri MG, et al. Clinical utility of combined
augments glucose uptake in human macrophages. J Am Coll Cardiol. FDG-PET/MR to assess myocardial disease. JACC Cardiovasc Imaging.
2011;58:603–614. 2017;10:594–597.
95. Rudd JHF. Imaging atherosclerotic plaque inflammation with 110. Naghavi M, Libby P, Falk E, et al. From vulnerable plaque to vulnerable
[18F]-fluorodeoxyglucose positron emission tomography. Circulation. patient: a call for new definitions and risk assessment strategies: part I.
2002;105:2708–2711. Circulation. 2003;108:1664–1672.
28 
Atherosclerotic Plaque Imaging: Coronaries
Imran Rashid, W. Yong Kim, Claudia Prieto, Marcus R. Makowski,
Warren J. Manning, and René M. Botnar

Coronary artery disease (CAD) is a leading cause of global mortality.1 CAD with a single imaging modality. Furthermore, evolving molecular
It results from atherosclerosis, which is a systemic and progressive disease imaging techniques may allow for the assessment of disease activity,
involving the intimal layer of large- and medium-sized arteries. Ath- delineation of vulnerable plaque phenotypes and identification of novel
erothrombosis, defined as atherosclerotic plaque disruption (predomi- therapeutic targets.
nantly plaque rupture) with superimposed thrombosis, can lead to
arterial occlusion and subsequent life-threatening conditions such as DIAGNOSTIC PERFORMANCE OF CORONARY
acute myocardial infarction (MI) or ischemic stroke. The concept of a
“vulnerable plaque” was first introduced to distinguish unstable, rupture-
MAGNETIC RESONANCE IMAGING
prone plaques from plaques with a stable phenotype. Analyses of human Studies have shown that coronary magnetic resonance angiography
coronary plaque have demonstrated that plaque rupture occurs at a (CMRA) can identify significant coronary artery plaque (>50% stenosis)
higher frequency than clinical events, suggesting that prothrombotic with a diagnostic accuracy comparable with MDCT.6–8 Multiple single
conditions are also necessary for plaque rupture to trigger acute coronary center studies have compared the accuracy of CMRA against invasive
syndromes.2,3 As such, effective risk stratification algorithms need to angiography culminating in a meta-analysis that showed a sensitiv-
include both local and systemic factors that confer increased risk of ity of 87% and specificity of 70% for detection of >50% stenoses.9–12
cardiovascular events. At present, clinical risk scoring systems are largely However, technical improvements in coil design, image acquisition/
based on the assessment of traditional risk factors with the addition of reconstruction, and motion compensation have allowed for a reduc-
noninvasive imaging or functional (“stress”) testing for investigation tion in total imaging time for whole heart CMR coronary angiography
of symptoms that are potentially ischemic in origin.4 A limitation of to approximately 5 minutes with improved diagnostic accuracy (see
this approach includes failure to detect specific features of coronary Chapter 24).8 Another study has demonstrated that noncontrast, free
plaque that confer increased risk of rupture. The identification of high- breathing, three-dimensional (3D) balanced steady-state free precession
risk patients might be further improved through the direct assessment (bSSFP) whole heart imaging has a sensitivity of 91% and a specific-
of coronary plaque burden, high-risk characteristics, or disease activity, ity of 86%, with an area under the receiver operator curve (ROC) of
allowing for targeted administration of therapies that are likely to result 0.92 when compared with quantitative invasive angiography for the
in prognostic benefits. detection of hemodynamically significant plaque.13 Furthermore, 3D
Subclinical atherosclerosis can precede the development of clinical cross-sectional imaging of the coronary vessel wall at 3 T has been able
disease by many years or even decades, providing an opportunity to to achieve an in-plane resolution of 0.5 × 0.5 mm, which is comparable
identify high-risk patients for targeted primary prevention therapies with MDCT (Fig. 28.1).
and ongoing clinical surveillance.5 At present, multidetector computed
tomography (MDCT) coronary angiography is often used for the assess- Technical Challenges
ment of low–intermediate risk patients with chest pain where its high Imaging of the coronary vessel wall and lumen is more challenging and
negative predictive value allows for the exclusion of coronary artery technically demanding than imaging of other vascular beds because of
disease in many individuals. MDCT also allows for the quantification the following14–16:
of the overall atherosclerotic plaque burden and the identification of 1. Motion: myocardial contraction/relaxation and respiration
some vulnerable plaque characteristics, through patterns of positive 2. Small and tortuous vessels
remodeling, low x-ray attenuation, and spotty calcification. Cardiovas- 3. Close proximity to epicardial fat, coronary blood, and myocardium
cular magnetic resonance (CMR) imaging is a noninvasive modality 4. Requirement for high spatial resolution
with excellent contrast of soft tissues and the blood/vessel wall inter-
face that has significant potential to assess coronary lumen integrity, CORONARY MAGNETIC RESONANCE
plaque burden, and composition without the requirement for ionizing
radiation. Advances in CMR data acquisition and gating techniques
IMAGING TECHNIQUES
have improved image quality, which to date has been constrained by In both noncontrast and contrast-enhanced CMR, the main limiting
limited spatial and temporal resolution. As such, CMR is emerging factor is imaging time. For coronary artery imaging, a compromise has
as a promising modality for coronary artery imaging where its selec- to be made between imaging time, the spatial resolution achieved, and
tive use in conjunction with myocardial perfusion imaging, viability, arterial coverage.
and ventricular function assessments, where CMR is the clinical refer- Because a high spatial resolution is paramount for diagnostic coro-
ence standard, may allow for the complete assessment of suspected nary artery imaging, longer scan times are necessary, which in turn

343
344 SECTION II  Ischemic Heart Disease

FIG. 28.1  Three-dimensional cross-sectional images of the right coronary artery in a healthy subject acquired
at 3 T with an in-plane spatial resolution of 0.5 × 0.5 mm and a slice thickness of 3 mm.

results in greater susceptibility to motion-related artifacts such as ghost- interleaved with the noncontrast CMR scan and used to compensate
ing and blurring.17 Adequately compensating for motion-induced artifacts for respiratory motion in the CMR acquisition (see Fig. 28.2). Uni-
is an unsolved problem and one of the technical challenges in CMR dimensional respiratory compensation typically involves positioning
that remains an active area of current research. the navigator on the dome of the right hemidiaphragm to measure
head–foot motion at the lung–liver interface to perform real-time gating
Motion Compensation Techniques and slice tracking.14 The use of navigator echoes that record and correct
Cardiac Motion Compensation for diaphragmatic motion29 allows for free breathing and eliminates
Cardiac motion compensation involves synchronizing image acquisition the time constraints of the breath hold approach, thereby allowing for
to the electrocardiogram (ECG) to allow for imaging during a motion- higher spatial resolution.30,31 The respiratory navigators can also be used
free period in the cardiac cycle.18 The preferred time is the mid diastolic to track the position of the moving artery by appropriately adjusting
coronary rest period (100–150 ms, approximately 10% of the cardiac the radiofrequency offset and the receiver phase and frequency of the
cycle), and the exact duration of this interval is heart rate dependent. CMR.32 Because a limited amount of the collected data are used to
ECG gating can be performed prospectively (image acquisition limited generate images, image efficiency is reduced and scan time is increased.
to mid diastole) or retrospectively, where data are acquired throughout However, advances in navigator tracking have allowed for larger respira-
the cardiac cycle but only data from mid diastole are used to generate tory gating windows and increased scan efficiency without compromising
images (Fig. 28.2). These measures are essential to avoid cardiac-motion scan quality.33 One such technique is the “affine” motion correction,
induced artifacts but reduce scan efficiency. which accurately approximates respiratory-induced coronary motion
and, through a 3D geometric transformation, incorporates translation
Respiratory Motion Compensation (head–foot movement), rotation, scaling, and shearing.34,35 Other limi-
During normal tidal breathing diaphragmatic excursion results in cardiac tations of diaphragmatic navigators are the need for a motion model
motion of up to 20 mm, where the cardiac displacement is many fold and the need for patient-specific tracking factors.36 Image-based motion
greater than coronary vessel thickness (0.5–2 mm).20 Although breath- correction with 2D or 3D navigators (iNav; see Fig. 28.2) increases
holding can be used to suspend respiratory motion for 10 to 20 seconds, motion tracking accuracy because it does not require a motion model
this limits the achievable signal-to-noise ratio (SNR) and thus spatial as a result of direct measurement of respiration-induced bulk motion
resolution. Three-dimensional coverage of such scans is limited to a of the heart.37,37a
thin slab, which reduces the clinical potential as imaging of the entire
proximal–mid coronary segments is required to identify prognostically Self-Navigation
significant disease. Thus coronary artery imaging is usually performed Self-navigation techniques have been developed to correct for respira-
during free breathing with respiratory navigation to compensate for tory motion, eliminating the requirement for diaphragmatic tracking.38
bulk respiratory motion. This is achieved using CMR navigators that Here, unidimensional self-navigation is used to track the superior–
monitor the position of the diaphragm with selective data acquisition inferior motion of the heart, with segmentation of 3D radial k-space
before image acquisition and restrict image reconstruction usually to data into different respiratory bins, with respiratory motion correc-
data acquired at or near end-expiration (see Chapter 7).21 The respira- tion through a retrospective affine transform enabling 1 mm3 isotropic
tory navigators can be unidimensional,22,23 two-dimensional (2D),24–26 resolution and whole heart coverage in 7 minutes.38 The development
or three-dimensional (3D),27,28 where real-time image acquisitions are of four-dimensional (4D) whole heart, self-navigated sequences for
CHAPTER 28  Atherosclerotic Plaque Imaging: Coronaries 345

Trigger delay

R-wave

Prepulses

NAV
Acquisition

IP
T2 prep dNAV SPGR
ECG
Fat sup iNAV bSSFP

MSDE TSE
FIG. 28.2  Coronary lumen sequence. Cardiac motion is compensated for by synchronizing image acquisition
with an electrocardiogram (ECG) and using a trigger delay from the R-wave to mid-diastole. For white blood
imaging, a T2 prep prepulse for suppression of signal from myocardium and coronary veins is applied fol-
lowed by a fat-selective radiofrequency prepulse (Fat sup) for fat suppression. Black-blood imaging can be
achieved with inversion preparation (IP), where an inversion delay is used to null signal from blood, or alter-
natively using flow sensitive prepulse (MSDE), where signal from flowing blood is destroyed using dephasing
gradients. The use of diaphragmatic navigators (dNAV) accounts for respiratory motion by limiting image
acquisition to end-expiration. Alternatively, image-based navigators (iNAV) use a low-resolution image imme-
diately before image acquisition to allow for translational foot-head and left-right motion correction. Coronary
images are typically acquired using either spoiled gradient echo (SPGR; preferred at 3 T) or balanced steady-
state free precession (bSSFP; preferred at 1.5 T) sequences for white blood imaging and turbo spin echo
(TSE) sequences for black-blood imaging.

both noncontrast and contrast-enhanced (CE) protocols has allowed T1-weighted (T1W) imaging sequence and therefore provides excellent
for simultaneous assessment of coronary artery anatomy and ventricular contrast in concert with T1-shortening contrast agents both at 1.5 T
function, with the potential for further reductions in scan time.39,40 and 3 T. T2-weighted and proton-density weighted (PDW) turbo spin-
Larger studies are currently awaited to demonstrate the efficacy of these echo sequences have a high SNR and have been used for black-blood
self-navigated and 4D imaging methods. vessel wall noncontrast CMR. A limitation of spin-echo techniques is
their dependence on blood flow for contrast generation, which can
Accelerated Imaging Techniques limit their use in patients where coronary blood flow is significantly
K-Space Filling reduced. Additional preparatory techniques such as T2 prepulses, fre-
K-space filling has a significant impact on image contrast and quality. quency selective fat suppression and magnetization transfer contrast
Cartesian sampling is most commonly used for CMR, but is also the can also be used to enhance contrast between the vessel lumen and the
most susceptible to ghosting artifacts. Radial sampling is well suited surrounding fat, myocardium, or venous blood (Fig. 28.3).47a
for motion compensation because the center of k-space (which contains
a substantial amount of structural information and image contrast) is NONCONTRAST MAGNETIC RESONANCE IMAGING
repeatedly measured and averaged, with the disadvantage of a lower
SNR.26 Spiral trajectories have been used in studies for both coronary
OF CORONARY ARTERY DISEASE
artery lumen41,42 and vessel wall CMR43,44 but are susceptible to off- Noncontrast CMR allows for the direct assessment of:
resonance artefacts that can lead to image blurring. Non-Cartesian image 1. Vascular remodeling
reconstruction is complex and computationally expensive, especially if 2. Coronary plaque burden
used in conjunction with parallel imaging or compressed sensing.45,45a 3. High-risk plaque characteristics
Furthermore, repeated measurement of the center of k-space makes
radial imaging less sensitive to motion but reduces the impact of pre- Vascular Remodeling and Plaque Burden
pulses and compromises imaging of tissues with short T1 times (e.g., Progressive increases in atherosclerotic plaque volume initially lead to
fat or gadolinium [Gd]). positive or Glagovian vascular remodeling, characterized by compensa-
tory enlargement of the outer vessel wall with relative luminal preserva-
Pulse Sequences tion that precludes these changes from being detected by invasive
CMRA requires rapid imaging sequences, such as spoiled gradient angiography. The total coronary plaque burden and presence of positive
recalled echo (GRE) or bSSFP. GRE benefits from administration of vascular remodeling have been shown to be strong predictors of car-
an intravenous contrast to generate bright blood angiograms, whereas diovascular events.48,49 Black-blood imaging can be used to quantify
bSSFP provides high SNR both with and without the use of contrast plaque burden and this has been demonstrated in patients with sub-
agents. However, bSSFP is susceptible to magnetic field inhomogeneity- clinical coronary artery disease and type 1 diabetes mellitus in the
induced black-band artifacts, which has largely restricted its use to field Multiethnic Study of Atherosclerosis (MESA) cohort (Fig. 28.4).50–52
strengths of ≤1.5 T. For imaging at higher field strengths, CE-GRE is These studies highlight the potential of noncontrast CMR to identify
favored because it is less susceptible to magnetic field inhomogene- asymptomatic patients with significant plaque burden in the absence
ities.46,47 GRE in combination with an inversion prepulse is a heavily of luminal stenosis that would otherwise not be detected with routine
346 SECTION II  Ischemic Heart Disease

LAD vessel wall

RCA vessel wall

A T2 prep ON B
FIG. 28.3  T2 prep and black-blood noncontrast coronary artery cardiovascular magnetic resonance imaging.
(A) Noncontrast bright-blood coronary lumen imaging with T2 prep. The application of T2 preparation pulses
results in significantly higher signal from arterial blood compared with venous blood or myocardium. The
right coronary artery (RCA), left main, and left anterior descending (LAD) coronary arteries are well seen.
(B) Black-blood coronary imaging of the same subject showing clear delineation of the coronary vessel wall
and lumen interface.

MPR WALL MRI

MRA
RCA1

RCA1
B C

RCA2
A RCA2

D E
FIG. 28.4  Cardiovascular magnetic resonance imaging (CMR) coronary angiography of nonocclusive plaque
with positive remodeling. (A) Magnetic resonance angiography (MRA) of the right coronary artery (RCA)
showing no significant stenosis in the proximal segment. Cross-sections of the lumen show a round lumen
proximally (B) and an oval lumen distally (D). Black-blood CMR at the same levels show normal wall in the
proximal segment (C) and eccentric plaque 5 mm distally (E). MPR, Multiplanar reconstruction; MRI, magnetic
resonance imaging. (Modified from Miao C, Chen S, Macedo R, et al. Positive remodeling of the coronary
arteries detected by magnetic resonance imaging in an asymptomatic population: MESA [Multi-Ethnic Study
of Atherosclerosis]. J Am Coll Cardiol. 2009;53[18]:1708–1715.)

functional assessment. At present, cross-sectional 2D black-blood imaging is a breakdown product of hemoglobin released 12 to 72 hours follow-
sequences with fat suppression techniques allow for the visualization ing hemorrhage and is a key component of both intraplaque hemorrhage
and quantification of vascular remodeling and plaque burden; however, and maturation of thrombus. Methemoglobin contains 5 unpaired
correlation with intravascular ultrasound (the reference standard) has electrons, leading to significant shortening of T1 relaxation times of
produced variable results.53,54 Challenges of noncontrast coronary vessel surrounding water molecules. The presence of HIP has been associated
wall imaging remain the relatively long scan time, complex planning with symptomatic cerebrovascular disease, with a positive predictive
procedures, and requirement of sufficient coronary flow to generate value of 93% for the identification of complex atherosclerotic lesions
black-blood images. Recent developments have focused on 3D whole- with evidence of cap disruption, intraluminal thrombosis, or intraplaque
heart coronary vessel wall imaging with either flow-sensitized or flow hemorrhage.59,60 Translation of T1W imaging of HIP has become feasible
independent T2 prepulses.55–56a for coronary imaging because of advanced motion compensation in
High-intensity plaque (HIP) on T1W imaging was first identified combination with T2 preparation, local inversion, or inversion recovery
in carotid arteries. The high-intensity signal is considered to represent 3D sequences. Because T1W imaging does not yield detailed anatomical
intraplaque hemorrhage57,58 and is associated with recent cerebrovascular images, coregistration with sequences that have superior soft-tissue
events.57–61 The signal is postulated to arise from methemoglobin, which contrast is required (Fig. 28.5).60a,64
CHAPTER 28  Atherosclerotic Plaque Imaging: Coronaries 347

shortening the T1 time of neighboring free water protons, leading to


a detectable signal on T1W (e.g., inversion recovery) sequences, where
it produces a positive (bright) signal. These agents are commonly used
for imaging the blood pool in contrast-enhanced CMR angiography.
Gd contrast agents rapidly redistribute with the surrounding tissue,
leaving a 10 to 15 minute window for intravascular (coronary) imaging.
They accumulate in both the inflamed vessel wall69,70 (increased perme-
ability) and in fibrotic atherosclerotic plaques (increased volume of
distribution)68 (Fig. 28.7). Compared with the previously described
PMR 1.91 double inversion recovery (IR) black-blood coronary vessel wall
approaches, nonselective CE-CMR is expected to be less flow sensitive
A B with shorter acquisition times. Assessment of plaque late gadolinium
enhancement reduces the imaging task to the presence or absence of
Noncontrast T1W1 CTA
contrast uptake in the plaque, with less stringent requirements on spatial
resolution than for previously described approaches. The technique
also has the potential to show various plaque characteristics such as
inflammation and neovascularization. In the context of acute MI, non-
targeted Gd results in increased signal in the atherosclerotic vessel wall
that diminishes on serial CE-CMR over the ensuing months, possibly
indicative of inflammatory regression.71
PMR 2.05 Alternative contrast agents have been designed to achieve longer cir-
culating times. Gadofosveset is an albumin-binding Gd compound that
is approved for clinical use, where its longer half-life allows for longer
periods of contrast-enhanced angiography. In addition, Gadofosveset
has been shown to allow for assessment of endothelial permeability
C D because blood albumin is more likely to penetrate areas of increased
Noncontrast T1W1 CTA endothelial gap junction width (i.e., increased permeability), which has
been shown to identify vulnerable coronary plaque in both preclinical
FIG. 28.5  Noncontrast T1-weighted (T1W1) images of high-intensity
plaques (HIPs; yellow arrowheads) in the proximal left anterior descend-
and clinical studies.71a,71b
ing coronary artery (A) and the right coronary artery (C). These HIPs
correspond to coronary plaques identified on computed tomographic
Targeted Contrast Agents for Biological
angiography (CTA) (yellow arrowheads on curved multiplanar reformation Characterization of Atherosclerosis
images; B and D). (Modified from Noguchi T, Kawasaki T, Tanaka A, Different CMR probes have been developed to study various biologi-
et al. High-intensity signals in coronary plaques on noncontrast T1-weighted cal processes by targeting a spectrum of molecular markers associated
magnetic resonance imaging as a novel determinant of coronary events. with atherosclerosis disease progression. The molecular probes generally
J Am Coll Cardiol. 2014;63[10]:989–999.) target changes in receptor expression, alterations in metabolic processes,
enzymatic activity, or connective tissue markers (see Chapter 3). The
molecular imaging agent is typically composed of a ligand, such as
From a coronary viewpoint, the presence of HIP on T1W imaging an antibody, a short peptide, or sugar molecule with a signal element
effectively identifies luminal thrombosis in patients following acute MI (Gd3+ or iron oxide), that is attached via a linker or spacer (Fig. 28.8).
with a sensitivity and specificity of >90% (Fig. 28.6).62–64 Multimodal The limitation of molecular CMR is the inherently low sensitivity of
imaging has demonstrated that HIP on T1W imaging correlates with CE-CMR requiring a relatively high target molecular concentration
positive vascular remodeling, low attenuation, and spotty calcification on (100 µM Gd) for sufficient signal amplification. Therefore larger molecu-
coronary CT angiography, which are all features of vulnerable plaque.63 lar imaging probes often include a carrier or nanoparticle (e.g., liposomes,
Furthermore, subsequent prospective studies have demonstrated that perfluorocarbon, or emulsions) that can be loaded with several to a
HIP with a plaque:myocardial signal ratio (PMR) >1.4 on noncontrast few thousand signal elements and ligands, thereby increasing signal
T1W imaging is a significant independent predictor of future coronary amplification and avidity to the target of interest. A disadvantage of
events in patients with CAD.64 The PMR cut off of 1.4 was determined these probes is slower clearance (often hepatic) and more complicated
by ROC curve analysis and has a sensitivity of 69.5% and specificity biodistribution that can require increased endothelial permeability to
of 82.3% for predicting future cardiac events64 and statin therapy has reach intraplaque targets.
been shown to reduce the PMR of HIP.64a Overall HIP analysis pro- Gadoflurine is a macrocyclic contrast agent with a high affinity for
vides prognostic and clinically useful information that is additive to the extracellular matrix. With both hydrophilic and hydrophobic moieties,
assessment of overall plaque burden for the improved identification of this agent has a significantly longer blood half-life compared with most
high-risk patients with CAD. clinically used molecular contrast agents.72 It has a high affinity for
lipid-rich plaque in a rabbit model of atherosclerosis but has also been
CONTRAST-ENHANCED MAGNETIC RESONANCE shown to colocalize with both collagenous and neovascularized plaque.73,74
Fibrin has been studied extensively as a molecular target for CE-CMR,
IMAGING OF CORONARY ARTERY DISEASE with successful imaging demonstrated in coronary arteries using fibrin-
CE-CMR using extracellular paramagnetic contrast agents has been specific contrast agents.75–77 In the clinical setting, targeted CE-CMR of
studied for the characterization of atherosclerotic plaque.65–68 Chelated fibrin has been shown to detect intracardiac, carotid, and aortic thrombi.
gadolinium (Gd3+) complexes are the most frequently used signal ele- Fibrin-targeting nanoparticles with >50,000 Gd per particle have also
ments in nontargeted and targeted CMR contrast agents. They act by been shown to visualize fibrin clots in an animal model in vivo.77
A B C

h h
D E 50 m F 20 m

FIG. 28.6  Noncontrast cardiovascular magnetic resonance images of a patient with acute inferior myocardial
infarction. T1-weighted images show increased in signal intensity (A) that colocalizes with the proximal right
coronary artery when T1-weighted images are fused with bright-blood imaging (B). Invasive x-ray coronary
angiography shows total occlusion of the proximal RCA (C) because of thrombus that was successfully
aspirated (D). Subsequent histology confirmed the presence of acute thrombus with abundant platelets and
erythrocytes that are the likely source of signal on T1–weighted images (E and F).

RCA RCA

A B

C D
FIG. 28.7  Contrast-enhanced coronary vessel wall imaging. (A) X-ray angiography of a patient with unstable
angina showing luminal irregularities (red arrows) of the mid right coronary artery (RCA). (B) Multiplanar
reformatted bright-blood imaging of the RCA and corresponding late gadolinium vessel wall enhancement
demonstrating increased contrast uptake (C; arrows), indicative of an increased atherosclerotic plaque burden
and possible inflammation. (D) Fusion of B and C. Areas of increased vessel wall uptake appear light orange
(arrows).
CHAPTER 28  Atherosclerotic Plaque Imaging: Coronaries 349

A B

Carrier Signal element


Cells Gd3+ chelates or iron oxide
Liposomes
Microbubbles
Perfluorocarbon emulsions
Ligand
Crosslinked iron oxide (CLIO)
Monoclonal antibodies or
fragments
Target Small peptides
Adhesion molecules
Intergins
Free water
Receptors
Fibrin

FIG. 28.8  Molecular cardiovascular magnetic resonance (CMR) imaging tracer characteristics. (A) CMR signal
elements (Gd3+ or iron oxide) are conjugated to ligands such as monoclonal antibodies, antibody fragments,
or small peptides that specifically bind to target sites. Gd3+-based contrast agents achieve tissue contrast
through shortening of T1-relaxation times of nearby protons; iron oxide promotes proton dephasing and can
be detected as negative signal on T2* imaging. (B) Carrier molecules bound to multiple target-specific ligands
and signal elements and are often used to maximize tracer concentration at target sites for enhanced tracer
localization.

Progressive atherosclerosis results in increased deposition of extra- and therefore are detected with greater sensitivity.83 In contrast to Gd3+
cellular matrix proteins in the neo-intima by smooth muscle cells and they are fairly large and typically have slower clearance (5–30 nm par-
macrophages.5 These include different types of collagen and elastin, ticles) and more complex biodistribution.
which represent the most abundant extracellular matrix proteins.78 An
elastin-specific low-molecular weight CMR contrast agent (ESMA) has HYBRID POSITRON EMISSION TOMOGRAPHY/
been shown to improve detection of atherosclerotic plaque and plaque
burden.79,80 This probe was shown to have favorable pharmacokinetic
CARDIOVASCULAR MAGNETIC RESONANCE
properties such as rapid biodistribution and fast clearance. In vivo Positron emission tomography (PET) is the most sensitive molecu-
measurements of plaque burden using ESMA in the ApoE-/- mouse lar imaging modality with a wide variety of radiotracers that can
model of atherosclerosis correlated with ex vivo histological assessment. be detected in the nano to picomolar range. The limitations of PET
Subsequent studies have similarly demonstrated good agreement between include the absence of anatomical information, relatively slow acqui-
imaging and histology in a swine model of coronary remodeling fol- sition times, and susceptibility to physiologic motion. As CMR has
lowing angioplasty and stent deployment79,81 (Fig. 28.9). superior soft-tissue contrast compared with computed tomography
Iron oxide particles differ from Gd-based contrast agents and to (CT), hybrid PET-MR platforms allow for the accurate in vivo localiza-
date they have not been used for characterization of coronary plaque. tion of radiotracers and acquisition of multiple CMR contrasts, with
However, iron oxide-based tracers have been used extensively for the the capacity to assess coronary intraplaque hemorrhage, thrombus,
characterization of carotid and aortic atherosclerosis. These particles and edema as well as myocardial perfusion and fibrosis. Furthermore,
shorten T1 relaxation times; however, phagocytosis and accumulation CMR can be performed simultaneously with PET allowing for CMR-
in macrophages leads to more pronounced shortening of T2 and T2*, based PET motion correction.83a As such, PET-MR harnesses the com-
where it yields negative contrast or signal void on T2 and T2*-weighted plementary strengths of these modalities for improved image fusion,
images.82 These particles exhibit higher relaxivity than Gd-based tracers workflow, and multiparametric data analysis.
350 SECTION II  Ischemic Heart Disease

A B C
Stent Stent
Stent

LAD LAD
LAD

RV AA
Stent

DA
MRA post BMS753951 Fusion

D E F A G
A
N
L
M M
A M
L L N

IEL
M
IEL

FIG. 28.9  Elastin imaging of coronary remodelling. (A) Comparison of coronary magnetic resonance angiog-
raphy (MRA) with (B) delayed enhancement MRI. (C) Fusion of A and B of stented and control coronary
vessel segments and corresponding histology (D–G). (B and C) Strong enhancement can be observed at the
stent location (dotted white arrow), whereas little to no enhancement is visible in the normal, noninjured
proximal segment of the left anterior descending (LAD) artery (solid white arrow). (D and E) Elastin von
Gieson stain of noninjured coronary vessel segment shows intact internal elastic lamina (IEL) and circular
arranged elastin fibers (black) in the media. (F and G) Elastin von Gieson stain of a stented vessel segment
demonstrates disruption of IEL and neointima formation, with diffuse elastin deposition (black dots). E and
G are magnifications of insets in D and F. A, Adventitia; AA, ascending aorta; BMS, bare metal stent; DA,
descending aorta; L, lumen; M, media; N, neointima. (From von Bary C, Makowski M, Preissel A, et al. MRI
of coronary wall remodeling in a swine model of coronary injury using an elastin-binding contrast agent. Circ
Cardiovasc Imaging. 2011;4:147–155.)

18
F-flurodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) conditions, release of nitric oxide by endothelial cells induces local
are PET radiotracers that have previously been used for plaque imaging. coronary vasodilation. Assessment of endothelial function requires
18
F-FDG is a glucose analogue that accumulates in areas of increased assessment of the vasodilatory and flow responses to endothelial
metabolic activity. Increased 18F-FDG signal has been demonstrated in stressors.87 Several studies have demonstrated that noncontrast CMR
carotid atherosclerotic plaque where the signal intensity correlates with can noninvasively assess coronary vasodilation pre- and poststimulus.89,90
areas of inflammatory cell infiltration.84 However, 18F-FDG imaging of An abnormal coronary vasodilatory response to isometric handgrip
coronary atherosclerosis is complicated by the relatively high meta- exercise has been shown to correlate with local plaque burden,91 and
bolic activity of the surrounding myocardium that can obscure signal the same technique can provide insight into systemic vascular endothelial
from the coronary vessel wall.85 More recently, imaging of vascular function via assessment of the internal mammary artery.92 Coronary
microcalcification using 18F-NaF has been shown to colocalize with artery distensibility,93 coronary flow and flow reserves can also be assessed
culprit plaques in postacute MI patients.86 Further studies will be noninvasively with noncontrast CMR.94,95 As such, in addition to the
required to assess the prognostic impact of 18F-NaF PET in ischemic quantification and characterization of atherosclerotic plaque, CMR has
heart disease. the capacity to assess the impact of disease on coronary physiology,
which is an important determinant of coronary ischemia.
ASSESSMENT OF ENDOTHELIAL FUNCTION BY
NONCONTRAST–CARDIOVASCULAR CONCLUSION
MAGNETIC RESONANCE Recent advances in CMRA have yielded improvements in spatial resolu-
Endothelial dysfunction is an early feature of subclinical CAD and can tion and imaging times, highlighting the potential of CMR for com-
be a predictor of adverse cardiovascular events.87,88 Under physiological prehensive coronary assessment without the need for ionizing radiation.
CHAPTER 28  Atherosclerotic Plaque Imaging: Coronaries 351

In addition to plaque burden, noncontrast CMR can provide important and feasibility of coronary magnetic resonance imaging continues to
information regarding plaque characteristics such as intraplaque hem- improve, and its eventual use in combination with existing CMR refer-
orrhage and thrombus formation, through the T1-shortening effect of ence standards such as myocardial perfusion, viability, and functional
methemoglobin that has proven prognostic utility. Furthermore, CE-CMR imaging raises the enticing prospect of comprehensive, single modality
and molecular imaging using hybrid PET-MR systems have the potential assessment of patients with ischemic heart disease.
to identify active cellular and molecular processes that drive athero-
sclerotic disease progression. These may allow for the precise risk strati-
fication of individuals with subclinical disease, in addition to the
REFERENCES
identification of novel therapeutic targets. Despite challenges, the quality A full reference list is available online at ExpertConsult.com
CHAPTER 28  Atherosclerotic Plaque Imaging: Coronaries 351.e1

REFERENCES 23. Stehning C, Börnert P, Nehrke K, Eggers H, Stuber M. Free-breathing


whole-heart coronary MRA with 3D radial SSFP and self-navigated
1. World Health Organization. Global Health Estimates: Deaths by Cause, image reconstruction. Magn Reson Med. 2005;54(2):476–480.
Age and Sex 2000–2012. 24. Henningsson M, Koken P, Stehning C, Razavi R, Prieto C, Botnar RM.
2. Tian J, Ren X, Vergallo R, et al. Distinct morphological features of Whole-heart coronary MR angiography with 2D self-navigated image
ruptured culprit plaque for acute coronary events compared to those reconstruction. Magn Reson Med. 2012;67(2):437–445.
with silent rupture and thin-cap fibroatheroma: a combined optical 25. Henningsson M, Mens G, Koken P, Smink J, Botnar RM. A new
coherence tomography and intravascular ultrasound study. J Am Coll framework for interleaved scanning in cardiovascular MR: application to
Cardiol. 2014;63(21):2209–2216. image-based respiratory motion correction in coronary MR angiography.
3. Cheruvu PK, Finn AV, Gardner C, et al. Frequency and distribution of Magn Reson Med. 2015;73(2):692–696.
thin-cap fibroatheroma and ruptured plaques in human coronary 26. Aitken AP, Henningsson M, Botnar RM, Schaeffter T, Prieto C. 100%
arteries: a pathologic study. J Am Coll Cardiol. 2007;50(10):940–949. Efficient three-dimensional coronary MR angiography with two-
4. Rodgers A, Ezzati M, Vander Hoorn S, et al. Distribution of major health dimensional beat-to-beat translational and bin-to-bin affine motion
risks: findings from the Global Burden of Disease study. PLoS Med. correction. Magn Reson Med. 2015;74(3):756–764.
2004;1:e27. 27. Henningsson M, Prieto C, Chiribiri A, Vaillant G, Razavi R, Botnar RM.
5. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating Whole-heart coronary MRA with 3D affine motion correction using 3D
the biology of atherosclerosis. Nature. 2011;473(7347):317–325. image-based navigation. Magn Reson Med. 2014;71(1):173–181.
6. Hamdan A, Asbach P, Wellnhofer E, et al. A prospective study for 28. Powell J, Prieto C, Henningsson M, Koken P, Botnar RM. CMRA with
comparison of MR and CT imaging for detection of coronary artery 100% navigator efficiency with 3D self-navigation and interleaved
stenosis. JACC Cardiovasc Imaging. 2011;4(1):50–61. scanning. J Cardiovasc Magn Reson. 2014;16(suppl 1):O8.
7. Yang Q, Li K, Liu X, et al. Contrast-enhanced whole-heart coronary 29. Ehman RL, Felmlee JP. Adaptive technique for high-definition MR
magnetic resonance angiography at 3.0-T: a comparative study with imaging of moving structures. Radiology. 1989;173(1):255–263.
X-ray angiography in a single center. J Am Coll Cardiol. 2009;54(1):69–76. 30. Li D, Kaushikkar S, Haacke EM, et al. Coronary arteries: three-
8. Nagata M, Kato S, Kitagawa K, et al. Diagnostic accuracy of 1.5-T dimensional MR imaging with retrospective respiratory gating. Radiology.
unenhanced whole-heart coronary MR angiography performed with 1996;201(3):857–863.
32-channel cardiac coils: initial single-center experience. Radiology. 31. Stuber M, Botnar RM, Danias PG, Kissinger KV, Manning WJ.
2011;259(2):384–392. Submillimeter three-dimensional coronary MR angiography with
9. Jahnke C, Paetsch I, Nehrke K, et al. Rapid and complete coronary real-time navigator correction: comparison of navigator locations.
arterial tree visualization with magnetic resonance imaging: feasibility Radiology. 1999;212(2):579–587.
and diagnostic performance. Eur Heart J. 2005;26(21):2313–2319. 32. McConnell MV, Khasgiwala VC, Savord BJ, et al. Prospective adaptive
10. Sakuma H, Ichikawa Y, Suzawa N, et al. Assessment of coronary arteries navigator correction for breath-hold MR coronary angiography. Magn
with total study time of less than 30 minutes by using whole-heart Reson Med. 1997;37(1):148–152.
coronary MR angiography. Radiology. 2005;237(1):316–321. 33. Kolbitsch C, Prieto C, Smink J, Schaeffter T. Highly efficient whole-heart
11. Sakuma H, Ichikawa Y, Chino S, Hirano T, Makino K, Takeda K. imaging using radial phase encoding-phase ordering with automatic
Detection of coronary artery stenosis with whole-heart coronary window selection. Magn Reson Med. 2011;66(4):1008–1018.
magnetic resonance angiography. J Am Coll Cardiol. 2006;48(10): 34. Nehrke K, Börnert P. Prospective correction of affine motion for arbitrary
1946–1950. MR sequences on a clinical scanner. Magn Reson Med.
12. Schuetz GM, Zacharopoulou NM, Schlattmann P, Dewey M. Meta-analysis: 2005;54(5):1130–1138.
noninvasive coronary angiography using computed tomography versus 35. Bhat H, Ge L, Nielles-Vallespin S, Zuehlsdorff S, Li D. 3D radial sampling
magnetic resonance imaging. Ann Intern Med. 2010;152(3):167–177. and 3D affine transform-based respiratory motion correction technique
13. Yonezawa M, Nagata M, Kitagawa K, et al. Quantitative analysis of 1.5-T for free-breathing whole-heart coronary MRA with 100% imaging
whole-heart coronary MR angiograms obtained with 32-channel cardiac efficiency. Magn Reson Med. 2011;65(5):1269–1277.
coils: a comparison with conventional quantitative coronary angiography. 36. Manke D, Nehrke K, Börnert P, Rösch P, Dössel O. Respiratory motion in
Radiology. 2014;271(2):356–364. coronary magnetic resonance angiography: a comparison of different
14. Kim WY, Danias PG, Stuber M, et al. Coronary magnetic resonance motion models. J Magn Reson Imaging. 2002;15(6):661–671.
angiography for the detection of coronary stenoses. N Engl J Med. 37. Fischer RW, Botnar RM, Nehrke K, Boesiger P, Manning WJ, Peters DC.
2001;345(26):1863–1869. Analysis of residual coronary artery motion for breath hold and
15. Sakuma H. Coronary CT versus MR angiography: the role of MR navigator approaches using real-time coronary MRI. Magn Reson Med.
angiography. Radiology. 2011;258(2):340–349. 2006;55(3):612–618.
16. Yang Q, Li K, Liu X, et al. 3.0T whole-heart coronary magnetic resonance 37a.  Henningsson M, Koken P, Stehning C, et al. Whole-heart coronary MR
angiography performed with 32-channel cardiac coils: a single-center angiography with 2D self-navigated image reconstruction. Magn Reson
experience. Circ Cardiovasc Imaging. 2012;5(5):573–579. Med. 2012;67(2):437–445.
17. Scott AD, Keegan J, Firmin DN. Motion in cardiovascular MR imaging. 38. Pang J, Bhat H, Sharif B, et al. Whole-heart coronary MRA with 100%
Radiology. 2009;250(2):331–351. respiratory gating efficiency: self-gated three-dimensional retrospective
18. Fischer SE, Wickline SA, Lorenz CH. Novel real-time R-wave detection image based motion correction (TRIM). Magn Reson Med.
algorithm based on the vectorcardiogram for accurate gated magnetic 2014;71(1):67–74.
resonance acquisitions. Magn Reson Med. 1999;42(2):361–370. 39. Coppo S, Piccini D, Bonanno G, et al. Free-running 4D whole-heart
19. Deleted in review. self-navigated golden angle MRI: initial results. Magn Reson Med.
20. Taylor AM, Jhooti P, Wiesmann F, Keegan J, Firmin DN, Pennell DJ. MR 2015;74(5):1306–1316.
navigator-echo monitoring of temporal changes in diaphragm position: 40. Pang J, Sharif B, Fan Z, et al. ECG and navigator-free four-dimensional
implications for MR coronary angiography. J Magn Reson Imaging. whole-heart coronary MRA for simultaneous, cardiac anatomy
1997;7(4):629–636. and function visualization. Magn Reson Med. 2014;72(5):1208–
21. Piccini D, Bonnano G, Ginami G, Littman A, Zenge MO, Stuber M. Is 1217.
there an optimal respiratory reference position for self-navigated whole 41. Meyer CH, Hu BS, Nishimura DG, Macovski A. Fast spiral coronary
heart coronary angiography? J Magn Reson Imaging. 2014;43(2):426–433. artery imaging. Magn Reson Med. 1992;28(2):202–213.
22. Li D, Kaushikkar S, Haacke EM, et al. Coronary arteries: three- 42. Börnert P, Stuber M, Botnar RM, Kissinger KV, Manning WJ.
dimensional MR imaging with retrospective respiratory gating. Radiology. Comparison of fat suppression strategies in 3D spiral coronary magnetic
1996;201:857–863. resonance angiography. J Magn Reson Imaging. 2002;15(4):462–466.
351.e2 SECTION II  Ischemic Heart Disease

43. Keegan J, Gatehouse PD, Yang GZ, Firmin DN. Non-model-based 60a.  Xie Y, Young-Jin K, Jianing P, et al. Coronary atherosclerosis
correction of respiratory motion using beat-to-beat 3D spiral fat-selective T1-weighted characterization with integrated anatomical reference. JACC
imaging. J Magn Reson Imaging. 2007;26(3):624–629. Cardiovasc Imaging. 2017;10(6):637–648.
44. Botnar RM, Kim WY, Börnert P, Stuber M, Spuentrup E, Manning WJ. 61. Takaya N, Yuan C, Chu B, et al. Association between carotid plaque
3D coronary vessel wall imaging utilizing a local inversion technique with characteristics and subsequent ischemic cerebrovascular events: a
spiral image acquisition. Magn Reson Med. 2001;46(5):848–854. prospective assessment with MRI—initial results. Stroke.
45. Pruessmann KP, Weiger M, Börnert P, Boesiger P. Advances in sensitivity 2006;37(3):818–823.
encoding with arbitrary k-space trajectories. Magn Reson Med. 62. Jansen CHP, Perera D, Makowski MR, et al. Detection of intracoronary
2001;46(4):638–651. thrombus by magnetic resonance imaging in patients with acute
45a.  Akcakaya M, Basha TA, Chan RH, et al. Accelerated isotropic sub- myocardial infarction. Circulation. 2011;124(4):416–424.
millimeter whole-heart coronary MRI: compressed sensing versus parallel 63. Kawasaki T, Koga S, Koga N, et al. Characterization of hyperintense
imaging. Magn Reson Med. 2014;71(2):815–822. plaque with noncontrast T1-weighted cardiac magnetic resonance
46. Frahm J, Haase A, Matthaei D. Rapid three-dimensional MR imaging coronary plaque imaging: comparison with multislice computed
using the FLASH technique. J Comput Assist Tomogr. 1986;10(2):363–368. tomography and intravascular ultrasound. JACC Cardiovasc Imaging.
47. Makowski MR, Wiethoff AJ, Uribe S, et al. Congenital heart disease: 2009;2(6):720–728.
cardiovascular MR imaging by using an intravascular blood pool contrast 64. Noguchi T, Kawasaki T, Tanaka A, et al. High-intensity signals in
agent. Radiology. 2011;260(3):680–688. coronary plaques on noncontrast T1-weighted magnetic resonance
47a.  Ginami G, Neji R, Rashid I, et al. 3D whole-heart phase sensitive imaging as a novel determinant of coronary events. J Am Coll Cardiol.
inversion recovery CMR for simultaneous black-blood late gadolinium 2014;63(10):989–999.
enhancement and bright-blood coronary CMR angiography. J Cardiovasc 64a.  Noguchi T, Tanaka A, Kawasaki T, et al. Effect of intensive statin therapy
Magn Reson. 2017;19(1):94. on coronary high-intensity plaques detected by noncontrast T1-weighted
48. Mani V, Muntner P, Gidding SS, et al. Cardiovascular magnetic resonance imaging. J Am Coll Cardiol. 2015;66(3):245–256.
parameters of atherosclerotic plaque burden improve discrimination of 65. Maintz D, Ozgun M, Hoffmeier A, et al. Selective coronary artery plaque
prior major adverse cardiovascular events. J Cardiovasc Magn Reson. visualization and differentiation by contrast-enhanced inversion prepared
2009;11:10. MRI. Eur Heart J. 2006;27(14):1732–1736.
49. Stone GW, Maehara A, Lansky AJ, et al. A prospective natural-history 66. Yuan C, Kerwin WS, Ferguson MS, et al. Contrast-enhanced high
study of coronary atherosclerosis. N Engl J Med. 2011;364(3):226–235. resolution MRI for atherosclerotic carotid artery tissue characterization.
50. Miao C, Chen S, Macedo R, et al. Positive remodeling of the coronary J Magn Reson Imaging. 2002;15(1):62–67.
arteries detected by magnetic resonance imaging in an asymptomatic 67. Wasserman BA, Smith WI, Trout HH 3rd, Cannon RO 3rd, Balaban AE,
population: MESA (Multi-Ethnic Study of Atherosclerosis). J Am Coll Arai AE. Carotid artery atherosclerosis: in vivo morphologic
Cardiol. 2009;53(18):1708–1715. characterization with gadolinium-enhanced double-oblique MR imaging
51. Gerretsen SC, Kooi ME, Kessels AG, et al. Visualization of coronary wall initial results. Radiology. 2002;223(2):566–573.
atherosclerosis in asymptomatic subjects and patients with coronary 68. Cai J, Hatsukami TS, Ferguson MS, et al. In vivo quantitative
artery disease using magnetic resonance imaging. PLoS One. 2010;5(9): measurement of intact fibrous cap and lipid-rich necrotic core size in
e12998. atherosclerotic carotid plaque: comparison of high-resolution, contrast-
52. Kim WY, Astrup AS, Stuber M, et al. Subclinical coronary and aortic enhanced magnetic resonance imaging and histology. Circulation.
atherosclerosis detected by magnetic resonance imaging in type 1 2005;112:3437–3444.
diabetes with and without diabetic nephropathy. Circulation. 69. Bley TA, Wieben O, Uhl M, Thiel J, Schmidt D, Langer M. High-
2007;115(2):228–235. resolution MRI in giant cell arteritis: imaging of the wall of the
53. He Y, Zhang Z, Dai Q, et al. Accuracy of MRI to identify the coronary superficial temporal artery. AJR Am J Roentgenol. 2005;184(1):
artery plaque: a comparative study with intravascular ultrasound. J Magn 283–287.
Reson Imaging. 2012;35(1):72–78. 70. Desai MY, Stone JH, Foo TK, Hellmann DB, Lima JA, Bluemke DA.
54. Gerretsen S, Kessels AG, Nelemans PJ, et al. Detection of coronary Delayed contrast-enhanced MRI of the aortic wall in Takayasu’s arteritis:
plaques using MR coronary vessel wall imaging: validation of findings initial experience. AJR Am J Roentgenol. 2005;184(5):1427–1431.
with intravascular ultrasound. Eur Radiol. 2013;23(1):115–124. 71. Ibrahim T, Makowski MR, Jankauskas A, et al. Serial contrast-enhanced
55. Andia ME, Henningsson M, Hussain T, Phinikaridou A, Protti A, Greil G. cardiac magnetic resonance imaging demonstrates regression of
Botnar RM. Flow-independent 3D whole-heart vessel wall imaging using hyperenhancement within the coronary artery wall in patients after acute
an interleaved T2-preparation acquisition. Magn Reson Med. 2013;69(1): myocardial infarction. JACC Cardiovasc Imaging. 2009;2(5):580–588.
150–157. 71a.  Phinikaridou A, Andia ME, Lavin B, et al. Increased vascular
56. Wang J, Yarnykh VL, Yuan C. Enhanced image quality in black-blood permeability measured with an albumin-binding magnetic resonance
MRI using the improved motion-sensitized driven-equilibrium (iMSDE) contrast agent is a surrogate marker of rupture-prone atherosclerotic
sequence. J Magn Reson Imaging. 2010;31(5):1256–1263. plaque. Circ Cardiovasc Imaging. 2016;9(12):e004910.
56a.  Cruz G, Atkinson D, Henningsson M, et al. Highly efficient nonrigid 71b.  Engel L-C, Landmesser U, Gigengack K, et al. Novel approach for in
motion-corrected 3D whole-heart coronary vessel wall imaging. Magn vivo detection of vulnerable coronary plaques using molecular 3-T
Reson Med. 2017;77(5):1894–1908. imaging with an albumin-binding probe. JACC Cardiovasc Imaging. 2018.
57. Moody AR. Magnetic resonance direct thrombus imaging. J Thromb In press.
Haemost. 2003;1(7):1403–1409. 72. Meding J, Urich M, Licha K, et al. Magnetic resonance imaging of
58. Moody AR, Allder S, Lennox G, Gladman J, Fentem P. Direct magnetic atherosclerosis by targeting extracellular matrix deposition with
resonance imaging of carotid artery thrombus in acute stroke. Lancet. Gadofluorine M. Contrast Media Mol Imaging. 2007;2(3):120–129.
1999;353(9147):122–123. 73. Sirol M, Itskovich VV, Mani V, et al. Lipid-rich atherosclerotic plaques
59. Moody AR, Murphy RE, Morgan PS, et al. Characterization of detected by gadofluorine-enhanced in vivo magnetic resonance imaging.
complicated carotid plaque with magnetic resonance direct thrombus Circulation. 2004;109(23):2890–2896.
imaging in patients with cerebral ischemia. Circulation. 2003;107(24): 74. Ronald JA, Chen Y, Belisle AJ, et al. Comparison of Gadofluorine-M and
3047–3052. Gd-DTPA for noninvasive staging of atherosclerotic plaque stability using
60. Murphy RE, Moody AR, Morgan PS, et al. Prevalence of complicated MRI. Circ Cardiovasc Imaging. 2009;2:226–234.
carotid atheroma as detected by magnetic resonance direct thrombus 75. Botnar RM, Perez AS, Witte S, et al. In vivo molecular imaging of acute
imaging in patients with suspected carotid artery stenosis and previous and subacute thrombosis using a fibrin-binding magnetic resonance
acute cerebral ischemia. Circulation. 2003;107(24):3053–3058. imaging contrast agent. Circulation. 2004;109(16):2023–2029.
CHAPTER 28  Atherosclerotic Plaque Imaging: Coronaries 351.e3

76. Spuentrup E, Botnar RM, Wiethoff A, et al. MR imaging of thrombi 86. Joshi NV, Vesey AT, Williams MC, et al. 18F-fluoride positron emission
using EP-2104R, a fibrin specific contrast agent: initial results in patients. tomography for identification of ruptured and high-risk coronary
Eur Radiol. 2008;18(9):1995–2005. atherosclerotic plaques: a prospective clinical trial. Lancet.
77. Flacke S, Fischer S, Scott MJ, et al. Novel MRI contrast agent for 2013;383(9918):705–713.
molecular imaging of fibrin: implications for detecting vulnerable 87. Deanfield JE, Halcox JP, Rabelink TJ. Endothelial function and dysfunction:
plaques. Circulation. 2001;104(11):1280–1285. testing and clinical relevance. Circulation. 2007;115(10):1285–1295.
78. Krettek A, Sukhova GK, Libby P. Elastogenesis in human arterial disease: 88. Anderson TJ, Meredith IT, Yeung AC, Frei B, Selwyn AP, Ganz P. The
a role for macrophages in disordered elastin synthesis. Arterioscler effect of cholesterol-lowering and antioxidant therapy on endothelium-
Thromb Vasc Biol. 2003;23(4):582–587. dependent coronary vasomotion. N Engl J Med. 1995;332(8):488–493.
79. Makowski MR, Wiethoff AJ, Blume U, et al. Assessment of atherosclerotic 89. Terashima M, Meyer CH, Keeffe BG, et al. Noninvasive assessment of
plaque burden with an elastin-specific magnetic resonance contrast agent. coronary vasodilation using magnetic resonance angiography. J Am Coll
Nat Med. 2011;17(3):383–388. Cardiol. 2005;45(1):104–110.
80. Makowski MR, Preissel A, von Bary C, et al. Three-dimensional imaging 90. Nguyen PK, Meyer C, Engvall J, Yang P, McConnell MV. Noninvasive
of the aortic vessel wall using an elastin-specific magnetic resonance assessment of coronary vasodilation using cardiovascular magnetic
contrast agent. Invest Radiol. 2012;47(7):438–444. resonance in patients at high risk for coronary artery disease. J Cardiovasc
81. von Bary C, Makowski M, Preissel A, et al. MRI of coronary wall Magn Reson. 2008;10:28.
remodeling in a swine model of coronary injury using an elastin-binding 91. Hays AG, Hirsch GA, Kelle S, Gerstenblith G, Weiss RG, Stuber M.
contrast agent. Circ Cardiovasc Imaging. 2011;4(2):147–155. Noninvasive visualization of coronary artery endothelial function in
82. Bulte JW, Kraitchman DL. Iron oxide MR contrast agents for molecular healthy subjects and in patients with coronary artery disease. J Am Coll
and cellular imaging. NMR Biomed. 2004;17(7):484–499. Cardiol. 2010;56(20):1657–1665.
83. Weissleder R, Elizondo G, Wittenberg J, Rabito CA, Bengele HH, 92. Iantorno M, Hays AG, Schar M, et al. Simultaneous noninvasive
Josephson L. Ultrasmall superparamagnetic iron oxide: characterization assessment of systemic and coronary endothelial function. Circ
of a new class of contrast agents for MR imaging. Radiology. 1990;175(2): Cardiovasc Imaging. 2016;9(3):e003954.
489–493. 93. Kelle S, Hays AG, Hirsch GA, et al. Coronary artery distensibility assessed
83a.  Munoz C, Neji R, Cruz G, et al. Motion-corrected simultaneous cardiac by 3.0 Tesla coronary magnetic resonance imaging in subjects with and
positron emission tomography and coronary MR angiography with high without coronary artery disease. Am J Cardiol. 2011;108(4):491–497.
acquisition efficiency. Magn Reson Med. 2018;79(1):339–350. 94. Hundley WG, Lange RA, Clarke GD, et al. Assessment of coronary
84. Rudd JHF, Warburton EA, Fryer TD, et al. Imaging atherosclerotic plaque arterial flow and flow reserve in humans with magnetic resonance
inflammation with 18F-fluorodeoxyglucose positron emission imaging. Circulation. 1996;93(8):1502–1508.
tomography. Circulation. 2002;105(23):2708–2711. 95. Globits S, Sakuma H, Shimakawa A, Foo TK, Higgins CB. Measurement
85. Rudd JH, Narula J, Strauss HW, et al. Imaging of atherosclerotic plaque of coronary blood flow velocity during handgrip exercise using
inflammation by fluorodeoxyglucose with positron emission tomography: breath-hold velocity encoded cine magnetic resonance imaging. Am J
ready for prime time? JACC. 2009;55(23):2527–2535. Cardiol. 1997;79(2):234–237.
29 
Assessment of the Biophysical Mechanical
Properties of the Arterial Wall
Raad H. Mohiaddin

Arteries are elastic tubes whose diameter varies with the pulsating in vivo, the vessels become more distended and less distensible. Indirect
pressure. In addition, they propagate the pulse created by ejection of measurements of pulmonary artery compliance have suggested that pul-
blood by the heart, at a velocity that is determined largely by the elastic monary arterial distensibility decreases with rising pulmonary artery
properties of the arterial wall. The vascular wall can be deformed by pressure.12,13 The ability of cardiovascular magnetic resonance (CMR)
pressure and shear stress forces exerted by the blood as well as the to image flow within any medium-sized vessel in any plane provides
tethering imposed by the surrounding tissues. Biophysical mechanical a unique opportunity to study the pulmonary arteries. Distensibility
properties of the arterial wall play an important role in the patho- and flow have already been assessed in patients with pulmonary arterial
genesis of cardiovascular diseases. Sclerosis (or stiffness), for example, hypertension.14,15
is an important aspect of atherosclerotic vascular disease that can be In this chapter, the clinical importance of arterial biophysical func-
demonstrated in experimental disease both in animals1 and in humans,2 tion and its assessment by CMR will be examined in detail as a comple-
and regression of the disease leads to reduced stiffness.3,4 Rupture of ment to Chapter 27.
atherosclerotic plaque (a common mechanism of myocardial infarc-
tion) and aortic dissection can be viewed as mechanical failures in the
diseased vessels. In addition, the effectiveness of interventional pro-
ARTERIAL STRUCTURE
cedures such as angioplasty is often achieved by causing mechanical A normal artery consists of three morphologically distinct layers. The
injury to the vessel wall, and the injury itself may lead to restenosis. A intima consists of a single continuous layer of endothelial cells bounded
number of common conditions are associated with changes in arte- peripherally by a fenestrated sheet of elastic fibers. The media consists
rial mechanical properties, although their importance is not always entirely of diagonally oriented smooth muscle cells, surrounded by
recognized. variable amounts of collagen, elastin, and proteoglycans. The adventitia
Systemic hypertension is almost always associated with altered consists predominantly of fibroblasts intermixed with smooth muscle
mechanical properties of the peripheral vasculature. Although it is not cells loosely arranged between bundles of collagen and proteoglycans.
clear which of the two is the inceptive event, one begets the other, Each structural component has its own characteristic properties. Smooth
fostering a vicious cycle. Compliance (the reciprocal of the resistance muscle is the physiologically active element, and by contracting or
offered to deformation) of the proximal aorta is reduced as a result. developing force, it can alter the diameter of the vessel or the tension
This causes waves reflected from the periphery to return prematurely in the wall. The other components are essentially passive in their mechani-
and coincide with the incident or forward-traveling wave produced by cal behavior. Elastin, which can be stretched to up to 300% of its length
the ventricle. This augments aortic systolic pressure, increases pulse at rest without rupturing,16 behaves mechanically more closely to a
pressure, and reduces diastolic pressure (because the reflected wave no linear elastic material such as rubber than other connective tissue com-
longer contributes to diastolic pressure). Decreased subendocardial/ ponents do. When elastin fibers are stretched and released, they return
subepicardial flow ratio has been demonstrated with reduced aortic promptly to their original state. Elastin fibers are important for main-
compliance.5 Thus decreased aortic distensibility may increase the risk taining normal pulsatile behavior, but they fracture at very low stresses
of subendocardial ischemia in the presence of coronary artery stenosis, and are probably much less important in determining the overall strength
left ventricular hypertrophy, or both. of the vessel wall. Collagen fibers, on the other hand, are much stiffer,
Aortic compliance represents an important determinant of left ven- but they are much stronger. The proportion of these components varies
tricular performance. Measurement of ventricular–vascular coupling from artery to artery. In the thoracic aorta, the elastin forms 60% of
takes into account the pulsatile load imposed on the left ventricle, as total fibrous element, and collagen forms 40%. The collagen proportion
well as the systemic vascular resistance.6 Metabolic disorders such as increases with increasing distance from the heart, reaching 30% elastin
Ehlers–Danlos and Marfan syndromes,7 diabetes mellitus,8 familial and 70% collagen in the extrathoracic vessels.17 The collagen-to-elastin
hypercholesterolemia,9 and growth hormone deficiency10 are also known ratio increases with age, which is one reason why vascular stiffness
to alter arterial compliance. increases with age. The human thoracic aorta is supplied by vasa vasorum
Similarly, the distensibility of the pulmonary arteries is reduced in and grows by increasing the number of lamellar units. The abdominal
pulmonary hypertension. Postmortem studies of externalized pulmonary aorta, in comparison, is avascular because it lacks vasa vasorum and
arterial strips have shown that the extensibility of the pulmonary trunk grows by increasing the thickness of each lamellar unit. The avascular
is decreased in pulmonary arterial hypertension.11 The wall of an artery thickness and the elevated tension per lamellar unit of the abdominal
becomes less extensible, the more it is stretched from its natural length. aorta predispose it to atherosclerosis.
An increased stretching of the circumference of the vessel will dimin- The distensibility of a blood vessel depends on the proportions and
ish the distensibility. When the pulmonary artery resistance increases interconnections of these materials and on the contractile state of the

352
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 353

vascular smooth muscle. Elasticity is a material’s ability to return to its abound. Although smooth muscle tone is not considered, in vitro human
original shape and dimensions after deformation, the deformation being arterial compliance has been measured from pressure–volume curves
proportional to the force applied. This proportionality was first described in postmortem arteries.20–23 In vivo estimation of arterial wall compli-
by Hooke in 1676 and is known as Hooke’s law. The point at which ance is more difficult, however, and has been performed by using indirect
Hooke’s law ceases to apply is known as the elastic limit, and when a and invasive techniques, including pulse wave velocity measurements
solid has been deformed beyond this point, it cannot regain its original in animals and in humans,24,25 the pressure–radius relationship using
form and acquires a permanent distortion. With larger loads still, the the Peterson transformer coil in animals,26 x-ray contrast angiography
yield point is reached when the deformation continues to increase in humans,27,28 and ultrasonography.29–31 The contributions of CMR to
without further load and usually rapidly leads to breakage. In purely the assessment of arterial wall mechanics are discussed in the following
elastic bodies, stress (the force per unit area that produces deformation) paragraphs. Other noninvasive measures are forced to rely on the assess-
produces its characteristic strain (the deformation of a stressed object) ment of accessible and often superficial vessels. Under the assumption
instantaneously, and strain vanishes immediately on removal of the that central and peripheral arteries behave in a similar fashion, the
stress. Some materials, however, require a finite time to reach the state properties of these arteries are then used as surrogates for those of
of deformation appropriate to the stress and a similar time to regain central arteries. There is, however, considerable heterogeneity between
their unstressed shape. Blood vessels typically exhibit such behavior, peripheral and central sites. CMR circumvents this problem by allowing
which is called viscoelasticity. the study of central arteries. Its ability to identify anatomic landmarks
suggests that reproducibility between studies should be improved, allow-
ing more effective follow-up.
DEFINITION OF VASCULAR WALL STIFFNESS
Vascular mechanics have been described by using different elastic moduli CARDIOVASCULAR MAGNETIC RESONANCE OF
and assumptions and for different purposes. Several approaches have
been described that use clinically available methods for in vivo char-
REGIONAL AORTIC COMPLIANCE
acterization of the stiffness of the vessel wall. The ability to measure CMR provides a direct, noninvasive way of studying local aortic com-
vascular stiffness has been greatly improved by the recent advances in pliance.32,33 High-resolution cine imaging or electrocardiogram-gated
imaging, such as high-frequency ultrasound and CMR. spin echo imaging in a plane perpendicular to the ascending and/or
The relationship between vascular wall deformation (strain) and descending aorta allows measurement of aortic cross-sectional area
the pressure exerted on the inner surface of the vascular wall (stress) is during systole and diastole. Measurement of regional aortic compliance
commonly used for the measurement of arterial wall biophysical proper- by CMR is calculated from the change in volume of an aortic segment
ties (elastic modulus). A plethora of terminology for the description of and from aortic pulse pressure estimated by a sphygmomanometer at
different elastic moduli, which can be confusing, has been described.18 the level of the brachial artery. The lumen of the aorta is outlined
The pressure–strain elastic modulus of the arterial wall (Ep) described by manually on the computer screen to measure the change in aortic area
Peterson and colleagues19 is commonly used. This elastic modulus that (ΔA) between diastole and systole. Regional aortic compliance (C)
applies to an open-ended vessel in the absence of reflection is defined (microL/mm Hg, m3/Nm−2) is calculated from the change in volume
as Ep = 2ΔP/(ΔV/V). This is the inverse of the fractional distensibility (ΔV = ΔA × slice thickness) of the aortic segment divided by the aortic
ΔV/V of the arterial lumen per unit pulse pressure ΔP. pulse pressure (ΔP) measured by a sphygmomanometer (Fig. 29.1).
Arterial compliance, C, which is defined as the change in volume ΔV Automatic measurement of aortic cross-sectional area is also possible.34
per unit change in pressure ΔP, has been also used in the literature. It Other indices of aortic stiffness that can be derived from these measure-
has been argued that this definition is appropriate to measurement of ments include distensibility, Peterson’s elastic modulus, and stiffness
ventricular compliance and not to the compliance of an open-ended index β ([systolic blood pressure/diastolic blood pressure]/area strain).
arterial segment. For the latter, the inverse of Peterson’s modulus was The accuracy of the indirect measurement of the pressure change
suggested 1/Ep. that is needed to compute compliance is limited because it ignores the
The average arterial compliance of a particular vessel pathway can changes in the pressure wave as it propagates through the arterial tree
also be determined by measuring the speed of propagation of the pulse (a process known as amplification). Further, it is important to obtain
in the vessel pathway. The velocity of such waves depends principally this pressure data on patients who are ideally lying in the magnetic
on the distensibility of the vessel wall. In real terms, this pulse is mea- resonance imaging (MRI) scanner using CMR-compatible apparatus.
surable by the disturbances in pressure, flow, or vessel diameter that it Despite the limitations of the pressure measurement, there is a good
causes. The propagation of flow waves has not been studied as extensively correlation between measurement of local aortic compliance and mea-
as that of pressure waves, partly because, unlike flow, accurate methods surement of global compliance from the speed of the propagation of
of pulsatile pressure measurements have been available for a long time the flow wave within the vessel.35
and partly because the distinction between flow wave velocity and blood In addition, noninvasive pressure wave analysis using arterial appla-
velocity has not always been clearly recognized. Blood velocity means nation tonometry has been shown to provide an accurate and repro-
the speed of an average drop of blood, whereas flow wave velocity ducible measurement of central vascular pressure measurements and
means the speed with which motion is transmitted. The wave velocity can be used to overcome this limitation.36
is usually much faster than that of the blood itself. Although CMR is
unable to assess pressure changes, alterations in the flow within (or CARDIOVASCULAR MAGNETIC RESONANCE OF
diameter of) a vessel can be measured accurately.
FLOW WAVE VELOCITY
Flow wave velocity is defined as the speed with which a flow wave
MEASUREMENT OF ARTERIAL WALL STIFFNESS propagates along a vessel and is regarded as the purest measure of
Arterial stiffness, which describes the resistance of arterial wall to defor- arterial stiffness. It is the quotient of distance traveled divided by the
mation, is difficult to measure because of the complex mechanical time taken for the flow wave to move between the two points and
behavior of arterial wall. As a result, a bewildering number of choices represents an average for that length of vessel (Figs. 29.2 and 29.3). The
354 SECTION II  Ischemic Heart Disease

A B

AA

DA

C D
FIG. 29.1  (A) Oblique sagittal image of the ascending aorta, arch and descending thoracic aorta showing
the sites where flow wave velocity and regional compliance are measured. The oblique transverse plane
shown in the top image is represented in diastole (B) and systole (C). This shows the change in area of a
22-year-old healthy subject. AA, Ascending aorta; DA, descending aorta.

approach is dependent on assessment of path length traveled and accu- the two points obtained from an oblique sagittal image. The distance
rate measurement of pulse arrival time. The latter requires recognition is determined manually on the computer screen by drawing a line in
of equivalent features or points on leading edges of the proximal and the center of the aorta joining the two points. In Fig. 29.4, the foot
distal flow waveforms (see Fig. 29.2), a process that is made complicated of the flow wave was defined by extrapolation of the rapid upstroke
by alterations that occur in flow wave morphology and magnitude as of the flow wave to the baseline as opposed to the midpoint of the
it progresses down the vessel. Unlike noninvasive measurements that upslope method used in Fig. 29.2.
rely on linear, transcutaneous measurements, CMR makes no assump- Others have used different magnetic resonance flow imaging tech-
tions about the shape of the artery and can accurately measure the path niques to assess arterial compliance. Tarnawski and colleagues37 used
length traveled. the comb-excited Fourier velocity-encoded method, previously reported
Mohiaddin and colleagues showed the feasibility of using CMR by Dumoulin and colleagues,38 to measure local arterial wave speed in
phase-shift velocity mapping to measure aortic flow wave velocity in the femoral artery in healthy men. In this method, simultaneous Fourier
humans.35 By taking advantage of the anatomy of the aorta, cine two- velocity-encoded data from multiple stations were acquired. The tech-
dimensional (2D) phase shift velocity maps were acquired with high nique employs a comb excitation radiofrequency pulse that excites an
temporal resolution in a single plane perpendicular to the ascending arbitrary number of slices. This causes the signals from the spin in a
and descending aorta, and the time taken for the flow wave to travel particular slice to appear at a position in the phase-encoding direction,
between the two points was measured (Fig. 29.4). Instantaneous flow which is the sum of the spin velocity and an offset arising from the
(in liters per second) in the mid-ascending aorta and mid-descending phase increment given to that excitation slice. Acquisition of spin veloc-
aorta was calculated by multiplying the aortic cross-sectional area and ity information occurs simultaneously for all slices, permitting the
the mean velocity within that area. Pulse wave velocity (PWV) was calculation of wave velocities arising from the pulsatile flow.
calculated in meters per second from the transit time (T) of the foot Hardy and colleagues39 studied aortic flow wave velocity using a
of the flow wave (see Fig. 29.4) and from the distance (D) between 2D CMR selective excitation pulse to repeatedly excite a cylinder of
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 355

Distance (mm)

1 2
A

1
1 2
MPA

2 2

RPA LPA
B

0.8

0.7

0.6
Mean velocity (m/s)

0.5

0.4

0.3 1 2

0.2

0.1

0
0 10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190 200 210

Time (ms)
C

PWV = transit time (AT2 − AT1)/distance


D
FIG. 29.2  Assessment of pulse wave velocity (PWV). (A) Phase velocity acquisition across a vessel is
undertaken at two points (1, 2), and the distance between the two is measured. (B) Examples of slice pre-
scription are given for the proximal pulmonary arteries and aorta; note that coverage of the ascending aorta
and descending aorta is enabled by a single slice. (C) Transit time is defined by the difference in arrival time
(AT) for the flow wave at both points, and is divided by the distance to give pulse wave velocity (D). LPA,
Left pulmonary artery; MPA, main pulmonary artery; RPA, right pulmonary artery.
356 SECTION II  Ischemic Heart Disease

Flow (L/s)
3

0
17 47 76 106 136 165 195 225 254 284
FIG. 29.3  Cine velocity mapping in a plane equivalent to that shown
in Fig. 29.1. The first frame was acquired 50 ms after the R-wave of A Time delay from R-wave (ms)

the electrocardiogram and represents the onset of left ventricular systole. Ascending aorta Descending aorta
The velocity maps indicate zero velocity as medium gray, caudal veloci-
ties in the descending aorta as light gray to white, and cranial velocities
as darker shades of gray to black, gray level intensity being proportional 4.5
to velocity.
4.0

3.5
magnetization in the aorta, with magnetization readout along the cyl-
inder axis each time. A toggled bipolar flow-encoding pulse was applied 3.0
before readout to produce a one-dimensional (1D) phase-contrast flow 2.5
Flow (L/s)

image. Cardiac gating and data interleaving were employed to improve


the effective time resolution to 2 ms. Wave velocities were determined 2.0
from the slope of the leading edge of flow measured on the resulting 1.5
M-mode velocity image. Aortic pulse wave velocity was also measured
by the same group, using a combination of cylinder of magnetization 1.0
with Fourier velocity encoding and readout gradients applied along the 0.5
cylinder axis (aorta) (Fig. 29.5),40 with the advantage of eliminating
partial volume effects that hindered their previous approach, but the 0.0
Fourier method has the drawback that it is no longer in real time and −0.5
errors occur, owing to accumulation of flow data over several (typi- 17 47 76 106 136 165 195 225 254 284
cally 16 to 32) cardiac cycles. For both methods, the interleaved repeats B Time delay from R-wave (ms)
needed to achieve high temporal resolution make them highly sensitive
to physiologic variability. Ascending aorta Descending aorta
In vitro experiments showed CMR measurements of pulse wave FIG. 29.4  The foot of the flow wave is defined by extrapolation of the
velocity in a tube phantom to be very reproducible and in good agree- rapid upstroke of the flow wave to the baseline, and this is performed
ment with pulse wave velocity measurements made with a pressure for flow in both the ascending aorta and descending aorta. The transit
catheter.41 These methods have not been widely applied in clinical time needed for the flow wave to propagate from a point in the mid-
research, nor has their in vivo reproducibility been proven yet. ascending aorta to a point in the mid-descending aorta can then be
measured. The distance around the arch from the plane in the ascending
aorta to the plane in the descending aorta can be measured from the
REFLECTED WAVES oblique sagittal image shown in Fig. 29.1, and the flow wave velocity
When the incident pulse wave from the ventricle reaches the periphery, is calculated by division of this distance by the propagation time. (A)
Transit time from a normal subject with good compliance. (B) Transit
it may be reflected and return toward the heart as a backward-running
time from an elderly patient with poor compliance. Note that the transit
wave. Reflected waves express properties of the peripheral circulation; time is significantly shorter in the patient with poor compliance. (From
if the peripheral resistance is elevated, reflected waves will be greater Mohiaddin RH, Firmin DN, Longmore DB. Age-related changes of human
in magnitude and will return sooner. Because its shape and magnitude aortic flow wave velocity measured noninvasively by magnetic resonance
will be defined by the complex interaction between forward incident imaging. J Applied Physiol. 1993;74:492–497.)
wave and backward reflected waves, the measured flow wave will also
be altered in pathologic conditions. As a result, the timing and location
of measurements become important considerations in the CMR assess- requirements become more demanding when the available length of
ment of arterial properties. vessel is limited (e.g., in the pulmonary arteries) or baseline data par-
If work is concerned only with the structure of proximal arter- ticularly noisy. Conversely, study of the influence of reflected waves on
ies, measurement should be made as far from the periphery and as measured flow waves might provide interesting insights into the nature
early in systole as possible to avoid the influence of reflections. These of the more distal vessels.
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 357

I Position S

21 25
Velocity

18 22 26

19 23 27

20 24 28

FOV 24 cm, 4.0 ms/frame, peak velocity 120 cm/s

FIG. 29.5  Fourier velocity measurements in a healthy subject. (A) In this method, the magnetic resonance
signal is obtained from a column aligned with the descending aorta. (B) The position along the column is
shown horizontally in each of the 12 cine frames. The velocity is shown by the vertical position of the signal.
FOV, Field-of-view; I, inferior; S, superior.

CLINICAL USE OF CARDIOVASCULAR MAGNETIC


RESONANCE FOR ASSESSING ARTERIAL
WALL STIFFNESS
100
Mohiaddin and colleagues were the first to use CMR for measurement
Compliance (mL/mm Hg)

of aortic compliance.42 They demonstrated that aortic compliance in 50


asymptomatic subjects falls with age and that patients with coronary
artery disease have abnormally low compliance (Fig. 29.6). The results
suggested a possible role for compliance in the assessment of cardio-
vascular fitness and the detection of coronary artery disease. Because 10
there is overlap between normal compliance and compliance in patients
with coronary artery disease above the age of 50 years, the test cannot 5
have perfect sensitivity and specificity. Below the age of 50 years, however,
there is much less overlap and the test is more specific. Abnormally low
aortic compliance has also been demonstrated in patients with aortic
coarctation43 and in patients with Marfan syndrome.44 The same group
also showed the feasibility of using CMR velocity mapping for measure- 20 40 60 80
ment of aortic flow wave velocity. Aortic flow wave velocity increased
Age
linearly with age, and there was a significant difference between the
youngest decade and the oldest decade studied. Flow wave velocity was Normals Athletes CAD
negatively correlated with regional ascending aortic compliance mea-
sured in the same subjects (Fig. 29.7). FIG. 29.6  Ascending aortic compliance displayed by using a logarithmic
scale and plotted against age in three groups: normals, athletes, and
In a study employing a single-slice phase-contrast acquisition of the
patients with coronary artery disease (CAD). The 95% confidence limits
ascending, arch, proximal aorta, and distal thoracic aorta, Rogers and are shown for the normals. The athletes’ compliance is abnormally high,
colleagues45 demonstrated an age-related increase in PWV among their and that in coronary disease patients is abnormally low. (From Mohiaddin
cohort. In addition, among the older patients, stiffness increased the RH, Underwood SR, Bogren HG, et al. Regional aortic compliance studied
more proximally the aorta was studied. The researchers ascribed these by magnetic resonance imaging: the effects of age, training, and coronary
changes to the disproportionate effect of elastin degradation with age artery disease. Br Heart J. 1989;62:90–96.)
358 SECTION II  Ischemic Heart Disease

8 which indicates that the lipid changes induced by fluvastatin (an increase
in high-density lipoprotein [HDL] level, decrease in very-low-density
lipoprotein level, and improvement in low-density:high-density lipo-
7 protein ratio) beneficially influenced vascular pathophysiology. In the
patients who were studied with carotid ultrasound means, carotid
Pulse wave velocity (m/s)

intimal–medial thickness decreased from 1.09 to 0.87 mm (P = .004),


6 corroborating these results.
Kupari and colleagues48 measured aortic elastic modulus by CMR
in asymptomatic subjects and correlated these measurements with
5 physical activity, ethanol consumption, systolic blood pressure, fasting
blood lipids, and serum insulin. They showed that the average value of
the ascending and descending aortic elastic modulus was associated
4
positively and statistically significantly with blood pressure, physical
inactivity, serum insulin, and HDL. The elastic modulus was associated
negatively with the ratio of low-density lipoprotein (LDL) cholesterol
to HDL cholesterol. No association between aortic elastic modulus and
3
either smoking or ethanol consumption was demonstrated in this study.
10 20 30 40 50 60 70
The same group demonstrated a higher aortic elastic modulus in patients
A Age (years)
with Marfan syndrome than in healthy subjects, indicating a relative
decrease in the distensibility of the thoracic aorta.49 Kupari and col-
8 leagues also demonstrated that aortic flow wave velocity was more
r = −0.77 reproducible (interobserver and intraobserver) than measurement of
Slope = −0.07 the pulsatile aortic area change or the elastic modulus. However, inter-
SEE = 0.88 study reproducibility has not been tested.50
Pulse wave velocity (m/s)

Adams and colleagues demonstrated abnormal aortic distensibility


7 and stiffness index in patients with Marfan syndrome using CMR.50
These findings were confirmed by Groenink and associates using a
CMR derived measure of distensibility and aortic PWV. Arrival time
was determined by the point at which flow had reached half its maximum
value.51 Beta-adrenergic blocking agents may reduce the rate of aortic
5 root dilation and the development of aortic complications in patients
with Marfan syndrome. This may be as a result of beta-blocker induced
changes in aortic stiffness. To investigate this, Groenink and colleagues
used CMR to measure aortic distensibility and aortic pulse wave velocity
to assess aortic stiffness in Marfan syndrome and healthy volunteers
before and after beta-blocker therapy.52 They showed that in both groups,
3
0 20 40 60 80 mean blood pressure decreased significantly but only the Marfan syn-
drome patients had a significant increase in aortic distensibility at
B Compliance (mL/mm Hg)
multiple levels and a significant decrease in pulse wave velocity after
FIG. 29.7  Flow wave velocity is directly proportional to age (A) and beta-blocker therapy. The same group53 sought to explain why some
inversely proportional to regional aortic compliance (B). SEE, Standard error
of these patients responded to beta-blockers whereas others did not.
of the estimate. (From Mohiaddin RH, Firmin DN, Longmore DB. Age-
related changes of human aortic flow wave velocity measured noninvasively
Cross-sectional assessment of aortic area was made at the level of the
by magnetic resonance imaging. J Applied Physiol. 1993;74:492–497.) pulmonary bifurcation by CMR and plotted against a surrogate for
central aortic pressures (using a finger arterial blood pressure–derived
brachial blood pressure). Using pressure-area lines, a subgroup with a
on the more proximal parts of the aorta, where the elastic to collagen transition point (a departure from a linear pressure-area relationship)
ratio is at its greatest. was identified in 6 out of 32 patients. It was suggested that this point
More recently, Voges and colleagues provided normal CMR values represented the recruitment of collagen to load-bearing elements. These
for cross-sectional areas, distensibility and pulse wave velocity of the patients demonstrated a trend toward reduced distensibility, although
thoracic aorta in children and young adolescents, which may serve as this relationship might have been stronger had the study group been
a reference for the detection of pathological changes of the aorta.46 larger. It was hypothesized that patients in whom the transition point
Regression of atheroma with reduction of cholesterol levels is rec- is seen at higher blood pressures might experience greater improvements
ognized to occur, but less is known about reversal of sclerosis. Nonin- in distensibility with beta-blockade than those whose transition points
vasive indices of sclerosis have largely been based on carotid ultrasound occurred at lower blood pressures.
measurements. Forbat and colleagues47 measured aortic compliance, Savolainen and colleagues54 used CMR and indirect brachial artery
coronary calcification, and carotid intimal–medial thickness during blood pressure measurements to assess aortic elastic modulus in patients
reduction of cholesterol level in hypercholesterolemic patients with with essential hypertension before therapy and after 3 weeks and 6
and without coronary artery disease. All received fluvastatin for 1 year. months of therapy with cilazapril (an angiotensin-converting enzyme
Aortic compliance was assessed by using CMR, and the coronary cal- inhibitor) or atenolol (a beta-1-adrenergic blocker). The authors con-
cification score was determined by electron beam computed tomography. cluded that 6 months of treatment with either cilazapril or atenolol
Carotid intimal–medial thickness was measured by carotid ultrasound. reduces the stiffness of the ascending aorta in essential hypertension.
The authors showed an improvement in aortic compliance over 1 year, No statistically significant differences between the effects of the two
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 359

drugs were observed. Honda and associates used CMR to measure Bogren and colleagues59 used CMR to study pulmonary artery
aortic distensibility in patients with systemic hypertension and dem- distensibility in healthy subjects (Fig. 29.8) and in patients with pul-
onstrated that the antihypertensive drugs nicardipine and alacepril have monary arterial hypertension (Fig. 29.9). The distensibility was found
a beneficial effect on aortic distensibility.55 Resnick and colleagues56 to be significantly lower in pulmonary arterial hypertension than in
assessed aortic distensibility, left ventricular mass index, abdominal fat normal subjects, but there was no age-related difference. The results
(subcutaneous and visceral), and free magnesium levels in the brain also demonstrated a small retrograde flow (2%) in the pulmonary trunk
and skeletal muscle by CMR. In patients with essential hypertension, of normal subjects close to the pulmonic valve. Antegrade plug flow
the following were concluded: Systolic hypertension and increased left occurred in most normal subjects but varied among individuals. There
ventricular mass index may result from arterial stiffness; arterial stiffness were also other variations in the flow pattern among normal individu-
may be one mechanism by which abdominal visceral fat contributes to als. All patients with pulmonary arterial hypertension had a markedly
cardiovascular risk; and decreased magnesium contributes to arterial irregular antegrade and retrograde flow and a large retrograde flow
stiffness in hypertension. (average 26%).
Chelsky and coworkers57 used CMR to measure aortic compliance in Changes in the cross-sectional area of the pulmonary artery in a
nine premenopausal women before and after menotropin therapy. They magnitude image from a phase velocity sequence have been measured
demonstrated that a short-term rise in estrogen induced by menotropin to derive pulmonary artery PWV60–62 values for normal subjects and
treatment was associated with an increase in aortic compliance. Aortic for patients with pulmonary hypertension.62 In the latter group, maximum
size was not significantly increased within this time frame. and minimum values for mean pulmonary artery pressure (mPAP)
Rider et al. demonstrated that HIV infection is an independent were predicted that “framed” the actual mPAP reliability.62
predictor of aortic stiffness as measured by CMR. However, because of Finally, Stefanides and colleagues have showed that aortic stiffness
the observational nature of this study, it is not possible to confirm has prognostic power in determining the likelihood of future cardiac
causality or mechanisms which might underlie the increase of aortic events (Fig. 29.10).63 This interesting study merits further examination
stiffness in patients with HIV.58 because it uses a simple marker of disseminated arterial disease that is

A B

C D
FIG. 29.8  Right ventricular outflow tract image (A) and oblique sagittal and transverse image (B) of the main
pulmonary artery, showing the sites where main pulmonary artery distensibility was measured (white lines).
In the bottom row, the change in the main pulmonary artery cross-sectional area between diastole (C) and
systole (D) demonstrated a large change in a 25-year-old healthy subject.
360 SECTION II  Ischemic Heart Disease

A B

C D
FIG. 29.9  Right ventricular outflow tract image (A) and oblique sagittal and transverse image (B) of the main
pulmonary artery, showing the sites where main pulmonary artery distensibility was measured (white lines).
In the bottom row, the change in the main pulmonary artery cross-sectional area between diastole (C) and
systole (D) demonstrated little change in a patient with pulmonary hypertension.

1.0 easy to measure in large populations. More recently, Maroules and


colleagues showed that in 2122 Dallas Heart Study participants without
0.9
Ratio free of all cardiac endpoints

cardiovascular disease, CMR measures of total arterial compliance and


0.8 aortic distensibility may be stronger predictors of nonfatal cardiac events,
0.7 whereas pulse wave velocity may be a stronger predictor of nonfatal
extracardiac vascular events (Fig. 29.11).64
0.6
0.5 ASSESSMENT OF ENDOTHELIAL FUNCTION
0.4 Brachial artery reactivity (BAR) testing has been proposed as a biomarker
0.3 of endothelial function.65 In the normal subject, release of a previously
occluded brachial blood pressure cuff results in postischemic hyperemia
0.2
(Fig. 29.12)66 and subsequent increased shear stress of the local arterial
0.1 wall. This, in turn, causes the release of endothelium-derived nitric
oxide, a potent vasodilator. The extent to which nitric oxide is released
0 10 20 30 40 50 60 (itself dependent on the amount produced and stored locally) can be
Months indirectly assessed by measuring the degree of vessel dilation resulting
FIG. 29.10  Relationship of aortic stiffness to occurrence of any cardiac from this provocation. Additionally, endothelium-independent function
end point, with the study population divided into terciles (P = .001). can be assessed by using dilation mediated by glyceryl trinitrate (GTN)
(From Stefanides C, Dernellis J, Tsiamis E, et al. Aortic stiffness as a (normally administered sublingually).
risk factor for recurrent acute coronary events in patients with ischaemic The noninvasive, uncomplicated, and well-tolerated nature of this
heart disease. Eur Heart J. 2000;21:390.) examination makes it an attractive endpoint in epidemiologic studies. This
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 361

20 20 20
TAC Quartile 1 AD Quartile 1 PWV Quartile 1
TAC Quartile 2 AD Quartile 2 PWV Quartile 2
of composite events (%)

of composite events (%)

of composite events (%)


Cumulative incidence

Cumulative incidence

Cumulative incidence
15 TAC Quartile 3 15 AD Quartile 3 15 PWV Quartile 3
TAC Quartile 4 AD Quartile 4 PWV Quartile 4
log-rank P < .0001 log-rank P < .0001 log-rank P < .0001
10 10 10

5 5 5

0 0 0
0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6 7 8
A Years B Years C Years
FIG. 29.11  Cumulative incidence curves for composite cardiovascular events based on quartile of (A) total
arterial compliance (TAC), (B) ascending aortic distensibility (AD), and (C) arch pulse wave velocity (PWV). (From
Maroules CD, Khera A, Ayers C, et al. Cardiovascular outcome associations among cardiovascular magnetic
resonance measures of arterial stiffness: the Dallas Heart Study. J Cardiovasc Magn Reson. 2014;16:33.)

800
700
Cuff release
600
500
Velocity (mm/s)

400
300
200
100
0
−100
−200
Cuff inflated
−300
Baseline 1-minute gap
Cardiac cycles (RR ≈ 900 ms)
FIG. 29.12  Mean velocity in real time shown graphically for 72 cardiac cycles. The first 5 cardiac cycles
are before cuff inflation. Imaging was suspended for the cuff inflation and a 5-minute delay. The next 10
cycles were acquired with the cuff inflated and show a shorter forward peak (i.e., reduced forward flow) in
the waveform, although the peak velocity does not reduce. The occlusion cuff was placed more than 10 cm
distal to the imaging plane. Note the increased reverse flow during the occlusion. After release of the cuff,
the forward peak is longer, and the reverse flow changes to forward flow. Both effects recover to baseline
after approximately 40 seconds. (From Mohiaddin RH, Gatehouse PD, Moon JCC, et al. Assessment of reac-
tive hyperemia using real time zonal echo planar flow imaging. J Cardiovasc Magn Reson. 2002;4:283–287.)

has been further strengthened as alterations in flow-mediated response were able to demonstrate impairment of flow-mediated dilation, whose
(by ultrasound) have been shown to precede clinical manifestations degree was inversely related to circulating levels of estradiol, in patients
of disease. These findings suggest a useful role for this biomarker as a who were given depot medroxyprogesterone acetate when compared
screening tool in the future, allowing risk stratification and intervention with controls. Weismann and colleagues69 showed that brachial artery
at an earlier stage than might previously have been thought possible. flow-mediated dilation was significantly reduced in smokers in com-
Although the peripheral location of the brachial artery allows ultra- parison with nonsmokers. As with Sorenson and colleagues’ work,
sound ready access to such a measure, CMR can also be employed in a impairment of dilation was endothelium-dependent only because the
similar fashion. In this way, BAR can be used as an adjunct, combining degree of GTN-mediated dilation was similar in the two groups. Con-
with more routine measures of ventricular and vascular function to versely, reduced endothelium-independent (but not endothelium-
provide a powerful means by which to evaluate patients in research dependent) function has been demonstrated in young elite rowers.70
(Fig. 29.13). CMR offers other advantages over ultrasonography for this
measure because it is more reproducible and less operator dependent.67
This has logistical and economic consequences for researchers under-
ARTERIAL WALL SHEAR STRESS
taking such work because it allows sample size to be reduced without The use of detailed anatomic model and boundary conditions, such
compromising the ability to identify statistically significant changes. as the inlet and outlet flow, captured in CMR are important for the
Also, CMR measures the true cross-sectional area, whereas ultrasound generation of a patient-specific computational fluid dynamic (CFD)
usually measures diameter only. In addition, assessment of reactive simulation. The CFD method allows the calculation of features and
hyperemic response using real-time CMR flow imaging has been shown properties such as wall shear stress (WSS) and mass transfer rate, which
to be associated with cardiovascular increased risk.68 are difficult to measure directly with imaging but are important to the
CMR has been used to demonstrate perturbations of endothelial understanding of basic hemodynamics. CFD is the technique of using
function in a variety of different scenarios. Sorenson and coworkers67 numerical methods to solve equations that govern the fluid flow. The basic
362 SECTION II  Ischemic Heart Disease

A B

C D
FIG. 29.13  Assessment of brachial artery reactivity. The top panel demonstrates the arrangement of the
surface coil and cuff on a subject’s right arm whose motion is restricted by sandbags. The transaxial plane
in which the brachial artery is at its most superficial (A) is used to prescribe a plane perpendicular to the
brachial artery (B). Arterial reactivity is then assessed through peripheral reactive hyperemia (C) and sublingual
glyceryl trinitrate (D). Arrows indicate the brachial artery position in the top panel and the cross section of
the brachial artery in A–D. (From Sorenson MB, Collins P, Ong PJL, et al. Long-term use of contraceptive
depot medroxyprogesterone acetate in young women impairs arterial endothelial function assessed by car-
diovascular magnetic resonance. Circulation. 2002;106:1646–1651.)

premise of CFD is to split the domain to be analyzed into small volumes flow velocities and pressure and therefore any derived quantities, such
or elements. For each of these, a set of partial differential equations is as shear stress. CFD has been extensively used to model physiologic flow
solved, which approximates a solution for the flow to achieve the basics and arterial WSS, particularly in the carotid bifurcation, in attempts
of conservation of mass, momentum, and energy for each volume or to better understand the mechanism of atherosclerosis in this region71
element. The three basic equations are solved simultaneously, with any and to give insight into the pathogenesis of arterial aneurysms.72 WSS
additional equations implemented in a particular model to obtain the is defined as the mechanical frictional force exerted on the vessel wall
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 363

by flowing blood and is the product of the blood viscosity with the change of aortic diameter.74 This is based on Laplace’s law for hollow
velocity gradient at the vessel wall. Low or oscillating arterial WSS is circular pipes, which states that the maximum stress within the arterial
correlated with atherosclerosis.73 wall is proportional to the radius. Works in literature suggest that the
In the aorta, once an aneurysm is formed, fluctuation in blood flow peak wall stress is a more reliable parameter for the assessment of the
within it can induce vibrations of the aneurysm wall that can contribute rupture risk of aortic aneurysms. Fillinger and colleagues reported that
to progression and eventual rupture. Volume flow measured by CMR the peak wall stress in aneurysms has a higher sensitivity and specificity
alone is not yet capable of sufficient spatial and temporal resolution for predicting the rupture than maximum diameter.75 CMR and CFD
for accurate measurement of WSS. Simulation with CFD provides more are promising tools for stress and velocity patterns simulation using
information than current diagnostic tools. In particular, it allows the patient-specific flow condition. Figs. 29.14 and 29.15 show how the
determination of the shear stress level, which in turn helps in identify- stress pattern and values are influenced by the shape of the aneurysm
ing regions that are susceptible to aneurysm formation, growth, and and the presence of intraluminal thrombus.
rupture. The only criterion that has been used so far for the selection CMR four-dimensional (4D) velocity mapping has been used to
of surgical patients has been the maximum diameter and the rate of study flow patterns in the heart and great vessels,76 and to derive WSS

A B

Smoothed
Effective
Stress
RST CALC
Time 1.000

64.50
57.00
49.50
42.00
34.50
27.00
19.50
12.00
4.50

C Stress pattern at peak systole (units: kPa)


FIG. 29.14  (A) Surface-rendered contrast-enhanced magnetic resonance angiography in the left anterior
oblique and right anterior oblique views in a patient with aortic arch aneurysm at the site of previous coarcta-
tion repair (left subclavian artery flap). (B) The aortic arch morphology was segmented from the cardiovascular
magnetic resonance (CMR) images, and the inflow–outflow flow conditions were extracted from CMR flow
mapping. (C) Stress pattern at peak systole (units: kilopascals [kPa]). Regions of high shear stress (arrowhead)
are found in the aortic arch and at the entry of the aneurysmal bulge in the first model, with low values only
in the distal region of the aneurysm.
364 SECTION II  Ischemic Heart Disease

A B

Smoothed
Effective
Stress
RST CALC
Time 1.000

64.50

57.00

49.50

42.00

34.50

27.00

19.50

12.00

4.50
C
FIG. 29.15  (A) Surface rendered contrast-enhanced magnetic resonance angiography in the coronal and
sagittal views in a patient with atherosclerotic aneurysm in the distal descending thoracic aorta. (B) The
aneurysm contains a large crescent-shape mural thrombus. (C) Stress pattern at peak systole (units: kilopas-
cals). Low values of shear stress throughout the aneurysm are noted where the thrombus appears to absorb
part of the pressure load resulting in low stress in the wall (arrowhead).

in the normal and dilated thoracic aorta.77 Increase in ascending aorta function, CMR is an ideal tool for its investigation, and further studies
diameter was found to significantly correlate with the presence and of the clinical role of parameters of sclerosis can be expected. The
strength of supraphysiologic helix and vortex formation in the ascend- capability of CMR to assess brachial artery reactivity and to measure
ing aorta, as well as with decrease in systolic regional WSS and increase WSS, in conjunction with CFD, will enhance further its role in the
in oscillatory shear index. studies of the biophysical properties of the arterial wall.

CONCLUSION ACKNOWLEDGMENTS
Atherosclerosis consists of two components, the most often discussed The author acknowledges the helpful contributions of Peter Gatehouse,
being atherosis, relating to plaque genesis and composition. Sclerosis PhD, and William Bradlow, MD, in the preparation of this manuscript.
is often the forgotten partner in clinical practice, but measurement of
vessel wall stiffening has now been shown to be useful both diagnosti-
cally and prognostically. Coupled with CMR’s powerful role in excluding
REFERENCES
secondary causes of hypertension and accurately assessing ventricular A full reference list is available online at ExpertConsult.com
CHAPTER 29  Assessment of the Biophysical Mechanical Properties of the Arterial Wall 364.e1

REFERENCES 25. Hallock P. Arterial elasticity in man in relation to age as evaluated by the
pulse wave velocity method. Arch Int Med. 1934;54:770–798.
1. Band W, Goedhard WJ, Knoop AA. Comparison of effects of high 26. Remington JW. Pressure-diameter relations of the in vivo aorta. Am J
cholesterol intake on viscoelastic properties of the thoracic aorta in rats Physiol. 1962;203:440–448.
and rabbits. Atherosclerosis. 1973;18:163–172. 27. Luchsinger PC, Sachs M, Patel D. Pressure-radius relationship in large
2. Banga I, Balo J. Elasticity of the vascular wall. 1. The elastic distensibility blood vessels of man. Circ Res. 1962;11:885–887.
of the human carotid as a function of age and arteriosclerosis. Acta 28. Stefanadis C, Stratos C, Boudoulas H, et al. Distensibility of the ascending
Physiol Acad Sci Hung. 1961;20–21:237–247. aorta: comparison of invasive and non-invasive techniques in healthy
3. Farrar DJ, Green HD, Wanger WD, Bond MG. Reduction in pulse wave men and in men with coronary artery disease. Eur Heart J.
velocity and improvement of aortic distensibility accompanying 1990;11:990–996.
regression of atherosclerosis in Rhesus monkey. Circ Res. 29. Gosling RG, King DH. Arterial assessment by Doppler-shift ultrasound.
1980;47:425–432. Proc Roy Soc Med. 1974;67:447–449.
4. Farrar DJ, Bond GM, Riley WA, Sawyer JK. Anatomic correlates of aortic 30. Kok WEM, Peters RJG, Prins MH, et al. Contribution of age and intimal
pulse wave velocity and carotid artery elasticity during atherosclerosis lesion morphology to coronary artery wall mechanics in coronary artery
progression and regression in monkeys. Circulation. 1991;83:1754–1763. disease. Clin Sci. 1995;89:239–246.
5. Ohtsuka S, Kakihana M, Watanabe H, et al. Chronically decreased aortic 31. Hanrath P, Heinzt B, Dahl V, et al. Evaluation of segmental elastic
distensibility causes deterioration of coronary perfusion during increased properties of the aorta in normotensive and medically treated patients by
left ventricular contraction. J Am Coll Cardiol. 1994;24:1406–1414. intravascular ultrasound. In: Boudoulas P, Toutouzas PK, Wooley C, eds.
6. Isnard RN, Pannier BM, Laurent S, et al. Pulsatile diameter and elastic Functional Abnormality of the Aorta. New York: Futura Publishing
modulus of the aortic arch in essential hypertension: a non-invasive Company; 1996:221–231.
study. J Am Coll Cardiol. 1989;13:399–405. 32. Mohiaddin RH, Underwood SR, Bogren HG, et al. Regional aortic
7. Handler CE, Child A, Light NM. Mitral valve prolapse, aortic compliance, compliance studied by magnetic resonance imaging: the effects of age,
and skin collagen in joint hypermotility syndrome. Br Heart J. training, and coronary artery disease. Br Heart J. 1989;62:90–96.
1985;54:501–508. 33. Chien D, Saloner D, Laub G, Anderson CM. High resolution cine MRI of
8. Lehman ED, Gosling RG, Sonksen PH. Arterial compliance in diabetes. vessel distension. J Comput Assist Tomogr. 1994;18:576–580.
Diabetes Care. 1986;9:27–31. 34. Rueckert D, Burger P, Yang GZ, Forbat SM, Mohiaddin RH. Automatic
9. Lehman ED, Watts GF, Gosling RG. Aortic distensibility and tracking of the aorta in cardiovascular MR images using deformable
hypercholesterolaemia. Lancet. 1992;340:1171–1172. models. IEEE Trans Med Imaging. 1997;16:581–590.
10. Lehman ED, Hopkins KD, Weissberger AJ, Gosling RG, Sonksen PH. 35. Mohiaddin RH, Firmin DN, Longmore DB. Age-related changes of
Aortic distensibility in growth hormone deficient adults. Lancet. human aortic flow wave velocity measured non-invasively by magnetic
1993;341:309. resonance imaging. J Appl Physiol. 1993;74:492–497.
11. Harris P, Heath D. The relation between structure and function in the 36. O’Rourke MF, Seward JB. Central arterial pressure and arterial pressure
blood vessels of the lung in pulmonary hypertension. In: Harris P, Heath pulse: new views entering the second century after Korotkov. Mayo Clin
D, eds. The Human Pulmonary Circulation. 3rd ed. London: Churchill Proc. 2006;81(8):1057–1068.
Livingstone; 1986:284–297. 37. Tarnawski M, Cybulski G, Doorly D, Dumoulin C, Darrow R, Caro C.
12. Reuben SR. Compliance of the human pulmonary arterial system in Noninvasive determination of local wavespeed and distensibility of the
disease. Circ Res. 1971;29:40–50. femoral artery by comb-excited Fourier velocity-encoded magnetic
13. Harris P, Heath D, Apostolopoulos A. Extensibility of the pulmonary resonance imaging: measurements on athletic and nonathletic human
trunk in heart disease. Br Heart J. 1965;27:660–666. subjects. Heart Vessels. 1994;9:194–201.
14. Paz R, Mohiaddin RH, Longmore DB. Magnetic resonance assessment of 38. Dumoulin CL, Doorly DJ, Caro CG. Quantitative measurement of
pulmonary trunk: anatomy, flow, pulsatility and distensibility. Eur Heart velocity at multiple positions using comb excitation and Fourier velocity
J. 1993;14:1524–1530. encoding. Magn Reson Med. 1993;29:44–52.
15. Mohiaddin RH, Paz R, Theodoropolus S, Firmin DN, Longmore DB, 39. Hardy CJ, Bolster BD, McVeigh ER, Adams WJ, Zerhouni EA. A
Yacoub MH. Magnetic resonance characterization of pulmonary arterial one-dimensional velocity technique for NMR measurement of aortic
blood flow following single lung transplantation. J Thorac Cardiovasc distensibility. Magn Reson Med. 1994;31:513–520.
Surg. 1991;101:1016–1023. 40. Hardy CJ, Bolster BD Jr, McVeigh ER, Iben IE, Zerhouni EA. Pencil
16. Mukherjee DP, Kagan HM, Jordan RE, Franzblau C. Effect of excitation with interleaved Fourier velocity encoding: NMR measurement
hydrophobic elastin ligands on the stress-strain properties of elastin of aortic distensibility. Magn Reson Med. 1996;35:814–819.
fibers. Connect Tissue Res. 1976;4:177. 41. Bolster BD Jr, Atalar E, Hardy CJ, McVeigh ER. Accuracy of arterial
17. Harkness ML, Harkness RD, McDonald DA. The collagen and elastin pulse-wave velocity measurement using MR. J Magn Reson Imaging.
content of the arterial wall in the dog. Proc Roy Soc [Biol]. 1957;146: 1998;8:878–888.
541–551. 42. Mohiaddin RH, Underwood SR, Bogren HG, et al. Regional aortic
18. Lee RT, Kamm RD. Vascular mechanics for the cardiologist. J Am Coll compliance studied by magnetic resonance imaging: the effects of age,
Cardiol. 1994;23:1289–1295. training, and coronary artery disease. Br Heart J. 1989;62:90–96.
19. Peterson LN, Jensen RE, Parnell R. Mechanical properties of arteries in 43. Rees RSO, Somerville J, Ward C, et al. Magnetic resonance imaging in late
vivo. Circ Res. 1968;8:622–639. post-operative assessment of coarctation of the aorta. Radiology.
20. Bergel DH. The dynamic elastic properties of the arterial wall. J Physiol. 1989;173:499–502.
1961;156:458–469. 44. Manzara CC, Mohiaddin RH, Pennell DJ, et al. Magnetic resonance
21. Hardung V. [Comparative studies on the dynamic elasticity and viscosity assessment of thoracic aorta in Marfan’s syndrome. American Heart
of blood vessels, rubber and synthetic elastomers. II.] Helv Physiol Acta. Association, Dallas. Circulation. 1990;82(suppl III):497 (Abstract).
1953;11:194–211 [in German]. 45. Rogers WJ, Hu YL, Coast D, et al. Age-associated changes in regional
22. Learoyd BM, Taylor MG. Alterations with age in the visco-elastic aortic pulse wave velocity. JACC. 2001;38(4):1123–1129.
properties of human arterial walls. Circ Res. 1966;18:278–292. 46. Voges I, Jerosch-Herold M, Hedderich J, et al. Normal values of aortic
23. Remington JW. Hysteresis loop, behaviour of the aorta and other dimensions, distensibility, and pulse wave velocity in children and young
extensible tissues. Am J Physiol. 1955;180:83–95. adults: a cross-sectional study. J Cardiovasc Magn Reson. 2012;14:77.
24. Bramwell JC, Hill AV, McSwiney BA. The velocity of the pulse wave in doi:10.1186/1532-429X-14-77.
man in relation to age as measured by hot-wire sphygmograph. Heart. 47. Forbat SM, Naoumova RP, Sidhu PS, et al. The effect of cholesterol
1923;10:233–255. reduction with fluvastatin on aortic compliance, coronary calcification
364.e2 SECTION II  Ischemic Heart Disease

and carotid intimal-medial thickness: a pilot study. J Cardiovasc Risk. 63. Stefanides C, Dernellis J, Tsiamis E, et al. Aortic stiffness as a risk factor
1998;5:1–10. for recurrent acute coronary events in patients with ischaemic heart
48. Kupari K, Hekali P, Keto P, et al. Relation of aortic stiffness to factors disease. Eur Heart J. 2000;21:390–396.
modifying the risk of atherosclerosis in healthy persons. Arterioscler 64. Maroules CD, Khera A, Ayers C, et al. Cardiovascular outcome
Thromb. 1994;14:386–394. associations among cardiovascular magnetic resonance measures of
49. Kupari K, Keto P, Hekali P, Poutanen V, Savolainen A, Standertskjold- arterial stiffness: the Dallas Heart Study. J Cardiovasc Magn Reson.
Nordenstam CG. Cine magnetic resonance imaging in the assessment of 2014;16:33.
aortic distensibility. In: Boudoulas P, Toutouzas PK, Wooley C, eds. 65. Deanfield J, Donald A, Ferri C, et al. Working Group on Endothelin and
Functional Abnormality of the Aorta. New York: Futura Publishing Endothelial Factors of the European Society of Hypertension. Endothelial
Company; 1996:247–268. function and dysfunction. Part II: association with cardiovascular risk
50. Adams JN, Brooks M, Redpath TW, et al. Aortic distensibility and factors and diseases. A statement by the Working Group on Endothelins
stiffness index measured by magnetic resonance imaging in patients with and Endothelial Factors of the European Society of Hypertension.
Marfan’s syndrome. Br Heart J. 1995;73:265–269. J Hypertens. 2005;23(2):233–246.
51. Groenink M, de Roos A, Mulder BJ, et al. Biophysical properties of the 66. Mohiaddin RH, Gatehouse PD, Moon JCC, et al. Assessment of reactive
normal-sized aorta in patients with Marfan syndrome: evaluation with hyperemia using real time zonal echo planar flow imaging. J Cardiovasc
MR flow mapping. Radiology. 2001;219:535–540. Magn Reson. 2002;4:283–287.
52. Groenink M, de Roos A, Mulder BJ, Spaan JA, van der Wall EE. Changes 67. Sorenson MB, Collins P, Ong PJL, et al. Long term use of contraceptive
in aortic distensibility and pulse wave velocity assessed with magnetic depot medroxyprogesterone acetate in young women impairs arterial
resonance imaging following beta-blocker therapy in the Marfan endothelial function assessed by cardiovascular magnetic resonance.
syndrome. Am J Cardiol. 1998;82:203–208. Circulation. 2002;106:1646–1651.
53. Nollen GJ, Westerhof BE, Groenink M, Osnabrugge A, Van der Wall EE, 68. Schwitter J, Oelhafen M, Wyss BM, et al. 2D spatially selective real time
Mulder BJM. Aortic pressure-area relation in Marfan patients with and magnetic resonance imaging for the assessment of microvasculature
without B blocking agents: a new invasive approach. Heart. 2004;90: function and its relation to the cardiovascular risk profile. J Cardiovasc
314–318. Magn Reson. 2006;8:759–769.
54. Savolainen A, Keto P, Poutanen VP, et al. Effects of angiotensin-converting 69. Wiesmann F, Petersen SE, Leeson PM, et al. Global impairment of
enzyme inhibition versus beta-adrenergic blockade on aortic stiffness in brachial, carotid, and aortic vascular function in young smokers: direct
essential hypertension. J Cardiovasc Pharmacol. 1996;27:99–104. quantification by high-resolution magnetic resonance imaging. J Am Coll
55. Honda T, Hamada M, Shigematsu Y, Matsumoto Y, Matsuoka H, Hiwada Cardiol. 2004;44(10):2056–2064.
K. Effect of antihypertensive therapy on aortic distensibility in patients 70. Petersen SE, Wiesmann F, Hudsmith LE, et al. Functional and structural
with essential hypertension: comparison with trichlormethiazide, vascular remodeling in elite rowers assessed by cardiovascular magnetic
nicardipine and alacepril. Cardiovasc Drugs Ther. 1999;13:339–346. resonance. J Am Coll Cardiol. 2006;48(4):790–797.
56. Resnick LM, Militianu D, Cunnings AJ, Pipe JG, Evelhoch JL, Soulen RL. 71. Milner JS, Moore JA, Rutt BK, Steinman DA. Hemodynamics of human
Direct magnetic resonance determination of aortic distensibility in carotid artery bifurcations: computational studies with models
essential hypertension: relation to age, abdominal visceral fat, and in situ reconstructed from magnetic resonance imaging of normal subjects.
intracellular free magnesium. Hypertension. 1997;30(3 Pt 2):654–659. J Vasc Surg. 1998;28(1):143–156.
57. Chelsky R, Wilson RA, Morton MJ, et al. Alteration of ascending thoracic 72. Borghi A, Wood NB, Mohiaddin RH, Xu XY. 3D geometric
aorta compliance after treatment with menotropin. Am J Obstet Gynecol. reconstruction of thoracic aortic aneurysms. Biomed Eng Online.
1997;176:1255–1259. 2006;5:59.
58. Rider OJ, Asaad M, Ntusi N, et al. HIV is an independent predictor of 73. Malek AM, Alper SL, Izumo S. Hemodynamic shear stress and its role in
aortic stiffness. J Cardiovasc Magn Reson. 2014;16:57. atherosclerosis. JAMA. 1999;282:2035–2042.
59. Bogren HG, Klipstein RH, Mohiaddin RH, et al. Pulmonary artery 74. Elefteriades JA. Natural history of thoracic aortic aneurysms: indications
distensibility and blood flow patterns: a magnetic resonance study of for surgery and surgical versus nonsurgical risks. Ann Thorac Surg.
normal subjects and of patients with pulmonary arterial hypertension. 2002;74:S1877–S1880.
Am Heart J. 1989;118:990–999. 75. Fillinger MF, Marra PS, Raghavan ML, Kennedy EF. Prediction of rupture
60. Laffon E, Bernard V, Montaudon M, Marthan R, Barat JL, Laurent F. risk in abdominal aortic aneurysm during observation: wall stress versus
Tuning of pulmonary arterial circulation evidenced by MR phase diameter. J Vasc Surg. 2003;37(4):724–732.
mapping in healthy volunteers. J Appl Physiol. 2001;90(2):469–474. 76. Markl M, Kilner PJ, Ebbers T. Comprehensive 4D velocity mapping of the
61. Laffon E, Laurent F, Bernard V, De Boucaud L, Ducassou D, Marthan R. heart and great vessels by cardiovascular magnetic resonance. J
Noninvasive assessment of pulmonary arterial hypertension by MR Cardiovasc Magn Reson. 2011;13:7–10. doi:10.1186/1532-429X-13-7.
phase-mapping method. J Appl Physiol. 2001;90(6):2197–2202. 77. Bürk J, Blanke P, Stankovic Z, et al. Evaluation of 3D blood flow patterns
62. Laffon E, Vallet C, Bernard V, et al. A computed method for noninvasive and wall shear stress in the normal and dilated thoracic aorta using
MRI assessment of pulmonary arterial hypertension. J Appl Physiol. flow-sensitive 4D CMR. J Cardiovasc Magn Reson. 2012;14:84.
2004;96(2):463–468. doi:10.1186/1532-429X-14-84.
SECTION III  Functional Disease

30 
Valvular Heart Disease
Philip J. Kilner and Raad H. Mohiaddin

As an imaging modality, cardiovascular magnetic resonance (CMR) • The accuracy of phase velocity mapping cannot be taken for granted.7
offers unrivaled versatility and freedom of anatomic access. In relation For jet velocity mapping, the dimensions of the voxels can be large
to heart valve disease, its relative strengths include the following: in relation to the size and shape of a narrow or fragmented jet,
• Depiction by cine imaging of valve movements and jet flow in planes, leading to possible inaccuracies because of signal loss, partial volume
or stacks of planes, of any orientation. averaging, and other artifacts. For the measurement of volume flow,
• Measurement of right as well as left ventricular volumes and mass particularly for the calculation of regurgitant fractions, surprisingly
by multislice cine imaging. large inaccuracies have been found to occur as CMR systems have
• Measurement of volume flow and regurgitant fraction (pulmonary been “improved” to allow more rapid acquisition in the last 10 years,
and aortic, at least) by phase contrast velocity mapping. because of eddy currents and concomitant gradients.7,8 The severity
• Assessment of the context and consequences of heart valve disease of the problem can vary considerably according to the hardware
using the wide fields of view, multiple image slices, and the versatility and software that are used and can change between the upgrades
of tissue characterization available to magnetic resonance. of a system. The uncertainties do not end there, however. The sequence
So although CMR is generally regarded as a second-line imaging parameters and imaging plane that are needed have to be chosen
modality after echocardiography for the assessment of heart valve disease,1 appropriately relative to the characteristics of flow under investiga-
it can have important contributions to make toward decision making tion if artifacts are to be minimized.
in regard to the timing and nature of surgical intervention, particularly • Measurements of biventricular volume and function are not nec-
in cases in which there have been inconclusive or conflicting findings, essarily as reliable and straightforward as has often been implied,
perhaps owing to inadequate echocardiographic access, or in which particularly if there is arrhythmia or the right ventricle is struc-
heart valve dysfunction is one aspect of more complex congenital or turally abnormal owing to congenital heart disease or previous
acquired pathology.2 And potentially, at least, CMR offers several pos- surgery, and volume analysis remains time consuming and to some
sible methods for the measurement of valve regurgitation,3–6 although extent subjective. The methods of acquisition and analysis need to
work is still needed to optimize and standardize acquisition protocols be specified appropriately for comparisons over time or between
and to fully validate the techniques used. patients.
If CMR is to become established as a reliable second-line modality, • In general, the unrivaled versatility of CMR is a strength, but it is
several potential weaknesses or pitfalls need to be recognized and avoided also a challenge. There is an ongoing dilemma between continuing
or corrected: development on the one hand and standardization on the other.
• The slice thickness (typically 6 mm) and the dimensions of voxels With these cautionary remarks in mind, the aim of this chapter
(typically about 6 × 1.5 × 1.2 mm3) of cine and velocity map acquisi- is to describe some important underlying principles and to guide
tions need to be borne in mind. users to the CMR techniques that we and others have found the most
• The images are not usually acquired in real time but are reconstructed informative.
from data gathered over several heart cycles during a breath-hold. Stenotic and regurgitant lesions of each heart valve are considered
For these reasons, valve leaflets are not always well seen, especially later, and an overview of measures of the severity of valve dysfunction
if there is arrhythmia; nor is CMR effective for visualizing the smaller, is given in Table 30.1, which is adapted from the 2006 American College
more mobile vegetations of endocarditis, although it can be useful of Cardiology/American Heart Association Guidelines for the Manage-
for identifying an abscess or false aneurysm. ment of Patients with Valvular Heart Disease.1 It is important to realize
• It is important to attempt to depict valve and jet structure by cine that right-sided valve lesions, particularly pulmonary or tricuspid regur-
imaging in several planes and orientations, not just one; appearances gitation, differ from their counterparts on the left. Although echocar-
and, potentially, interpretation can differ considerably between images diography has become well established in the assessment of valvular
of a particular valve acquired in different planes (Fig. 30.1). Con- lesions of the left heart, CMR, with phase velocity mapping, has particular
tiguous stacks of cine images are valuable for covering all parts of strengths in relation to the assessment of valvular and perivalvular
the mitral or tricuspid valves. lesions of the right heart.

365
366 SECTION III  Functional Disease

FIG. 30.1  Top left, A systolic frame of a steady-state free precession (SSFP) cine acquisition shows the
bright core and dark edges of the jet through the slit-like orifice of a stenosed bicuspid aortic valve. Top right,
A phase contrast velocity map, encoded through-plane, shows the dark jet, recording a peak velocity of
4.2 m/s. Bottom left, SSFP cine imaging aligned with the jet direction and orientated at right angles to the
slit shows the narrower dimension of the jet, which appears bright and clearly delineated (arrow). Bottom
right, The jet appears broad, diffuse, and dark in the SSFP cine, which is aligned with the length of the slit,
its voxels spanning the width of the narrow jet (arrows).

TABLE 30.1  Classification of the Severity of Heart Valve Disease in Adults, Based Partly on
the 2006 ACC/AHA Guidelines,1 Which Refer Mainly to Echocardiographic Indices
Aortic Stenosis
Mild Moderate Severe
Peak jet velocity (m/s) <3 3–4 >4
Orifice area (cm2) >1.5 1.0–1.5 <1.0
Orifice area index (cm2/m2) <0.6
Additional features LV hypertrophy, poststenotic dilatation of the ascending aorta
Subaortic stenosis and coarctation should be considered and excluded.

Pulmonary Stenosis
Mild Moderate Severe
Peak jet velocity (m/s) <3 3–4 >4.0
Valve orifice area (cm2) <1.0
Additional features RV hypertrophy, poststenotic dilatation of the MPA and LPA
Subpulmonary and pulmonary artery stenosis should be excluded.

Mitral Stenosis
Mild Moderate Severe
Peak jet velocity <1.2 1.2–2.2 >2.2
Valve orifice area (cm2) >1.5 1–1.5 <1.0
Additional features Possible evidence of pulmonary hypertension
CHAPTER 30  Valvular Heart Disease 367

TABLE 30.1  Classification of the Severity of Heart Valve Disease in Adults, Based Partly on
the 2006 ACC/AHA Guidelines, Which Refer Mainly to Echocardiographic Indices—Cont’d
Tricuspid Stenosis
Valve orifice area (cm2) <1.0

Aortic Regurgitation
Mild Moderate Severe
Regurgitant volume (mL/beat) <30 30–60 >60
Regurgitant fraction <30% 30%–50% >50% (CMR flow measurements tend to underestimate AR,
unless corrected for aortic root motion)
Regurgitant orifice area (cm2) <0.1 0.1–0.3 >0.3

Pulmonary Regurgitation (Assuming Near Normal Pulmonary Resistance)


“Free” or “Almost Free” (But May Be Well
Mild Moderate Tolerated)
Regurgitant jet or stream width Narrow, <2 mm Moderate 2–5 mm Unobstructed reversed stream, >6 mm across
Valve leaflet appearances Mobile, coapting Partly coapting Ineffective leaflets with wide failure of coaptation
Regurgitant volume (mL/beat) <30 30–40 >40
Regurgitant fraction <25% 20%–35% >35%, but modified by up- and downstream factors
Additional features Free PR occurs mainly in the first half of diastole, typically
followed by late diastolic forward flow if the right ventricle
is full and conduit-like when the atrium contracts

Mitral Regurgitation
Mild Moderate Severe
Regurgitant jet width Narrow, <1.0 mm 1.0–2 mm >2 mm, with extensive jet or swirling LA flow
No visible jet core Narrow core Bright jet core
>2 mm width
Regurgitant volume (mL/beat) <30 30–60 >60
Regurgitant fraction <30% 30%–50% >50%
Regurgitant orifice area (cm2) <0.2 0.2–0.4 >0.4
Additional features Dilated left atrium and pulmonary veins
Systolic flow reversal in pulmonary veins

Tricuspid Regurgitation (Assuming Near-Normal Pulmonary Resistance)


Mild Moderate Severe
Regurgitant jet width Narrow, <2 mm 2–6 mm >6 × 6 mm, measured on a through-plane velocity map
Additional features Dilated right atrium and caval veins

Measurements by CMR should generally be comparable with those by echo. They can be more accurate than echo where measurements of
flow volume and/or ventricular volume are used to quantify regurgitation, particularly pulmonary regurgitation. But CMR might not measure the
peak velocity of a narrow or fragmented jet as accurately as Doppler echo. These jet velocities depend on the rate of flow as well as the
amount of area reduction. Direct planimetry may be feasible by CMR, but only in cases in which the jet core, immediately downstream of an
orifice, is coherent and of suitable size, shape, and location relative to the image voxels for clear delineation.
ACC/AHA, American College of Cardiology/American Heart Association; AR, aortic regurgitation; CMR, cardiovascular magnetic resonance; LA,
left atrial; LPA, left pulmonary artery; LV, left ventricular; MPA, main pulmonary artery; RV, right ventricular.

BASIC PRINCIPLES rather than across the length of the voxels and will therefore be seen
most clearly when the structure passes perpendicularly through plane
Slice Thickness and the Visualization of of the image. This is one reason why valve leaflets are sometimes, but
Thin Structures not always, visible in cine acquisitions. Awareness of this can help with
The visualization by CMR of heart valves and flow through them is the choice of imaging plane; for example, the leaflets of a closed mitral
still done mainly by imaging in slices, and the thickness of a CMR slice, valve, particularly if it prolapses to the plane of the annulus or beyond,
for example 6 mm, is generally considerably greater than that of pixel are likely to be seen clearly in certain long-axis but not the short-axis
dimensions, which might be 1 to 1.6 mm. This means that the image slices. And a flat jet through a slit-like orifice will be seen most clearly
is effectively made up of a layer of relatively long, narrow voxels, as if the image plane cuts perpendicular to the line of the slit, although
shown in Fig. 30.2. A thin structure such as a valve leaflet or narrow the jet will appear narrower in this orientation than if depicted, less
jet will be visualized more clearly if it is aligned parallel with the voxels clearly, in a plane aligned with the length of the slit (see Fig. 30.1).
368 SECTION III  Functional Disease

6 mm

FIG. 30.2  Illustration of the importance of the orientation of a cardiac magnetic resonance image slice,
consisting of long, narrow voxels, with respect to a thin structure. The boundaries of the structure will be
depicted clearly only if it passes nearly perpendicular to the image slice. (Courtesy Robert Merrifield.)

Upstream convergence zone

Coherent jet core

Parajet shear layer Turbulence swept


downstream

FIG. 30.3  The structure of jet flow as visualized using a suspension of metal pigment in water flowing
continuously, left to right, in an open channel indented by a “stenosis.” In this case, the flow upstream and
in the core of the jet is laminar. Turbulence generated in the parajet shear layer is swept downstream, dis-
sipating the kinetic energy of the jet. Jet flow through heart valves, particularly through severely stenosed
or regurgitant orifices, is likely to be more complex in structure than this, with possible splaying and frag-
mentation of the jet core and possible asymmetric attachment to one wall or valve leaflet.

Visualization and Planimetry of Jets by Cine Imaging region of shear and turbulence (Fig. 30.3). The shear layer is the region
Stenosis or regurgitation of a diseased heart valve is associated with with the most extreme spatial gradients of velocity, and this is where
jet formation and consequent dissipation of energy through shear and much of the turbulence of a jet originates and where there is likely to
turbulence. Appreciation of the structure of jet flow is fundamental to be most loss of signal in cine acquisitions and potential inaccuracies in
the interpretation of jet appearances on cine images. Jet flow is associ- phase velocity maps. It is the friction associated with shear and turbu-
ated with an upstream region of convergence of streamlines where flow lence that dissipates fluid kinetic energy as heat, the fluid becoming less
accelerates into the orifice; then, in most situations, there is a relatively intensely turbulent as it loses kinetic energy and is swept downstream.
stable, high-velocity jet core, typically extending two to three orifice Each of the characteristic zones of a jet may be identifiable on cine
diameters beyond the orifice and, lateral and distal to the core, a parajet images, but the appearances depend not only on jet shape and structure
CHAPTER 30  Valvular Heart Disease 369

but also on the nature of the acquisition―the slice thickness, pixel SSFP imaging. For this reason, GRE cine imaging may be more suitable
dimensions and orientation, sequence design, and echo time9―and, than SSFP for the direct measurements of orifice area by planimetry,11–15
importantly, on the location of the image slice relative to the jet. The as long as the echo time is sufficiently short for the high-velocity core
relationships between flow features and the recovery of magnetic reso- of the jet, directed through the image plane, to appear bright. More
nance signal are not straightforward. On the whole, coherent flow of work is needed to determine optimal imaging parameters and, if SSFP
the jet core is associated with local brightening of the signal on cine imaging is used, exactly where the jet should be outlined relative to
images. Acquisitions with longer echo times will be more subject to the dark shear layer.
signal loss from regions of high shear and turbulence, whereas sequences If planimetry is attempted, it is advisable to acquire a stack of
with shorter echo times tend to be less susceptible to signal loss caused slightly overlapping slices, shifting gradually from upstream to imme-
by turbulence. diately downstream of the orifice in, say, 3-mm steps, to ensure that
Steady-state free precession (SSFP) imaging has become widely used the minimum orifice/jet area is visualized. This approach is likely to
in CMR because of the good contrast it gives between blood and adja- be accurate only when the orifice is relatively simple in shape and
cent tissues. Its properties also make it suitable for the delineation of the jet core is coherent. It is not likely to be accurate if the orifice is
jets, but appearances differ somewhat from those of gradient echo irregularly deformed or is a narrow slit, giving rise to a fragmented,
imaging. The echo times used for SSFP imaging are usually very short splayed, or fan-like jet.7
(e.g., 2 ms), thus minimizing signal loss because of the higher orders
of motion associated with turbulence. But without velocity compensa- Phase Contrast Velocity Mapping
tion, there is marked dephasing and consequent loss of signal in voxels The principles of phase contrast velocity mapping are explained in
that contain a wide range of velocities simultaneously, which means Chapter 6. In brief, one or more of the directional components of veloc-
those that lie in the shear layer at the edge of a jet. SSFP sequences are ity are encoded in the phase of the signal in each voxel of the image for
also susceptible to slight inhomogeneities of the magnetic field, and each phase of the cardiac cycle.16 Velocity can be encoded horizontally
there is more marked loss of signal in regions of flow disturbance if or vertically (in the X- or Y-direction) in the image plane, but more
the magnetic field is slightly off shim. This can affect the appearance often, and generally more reliably, the through-plane Z-component is
of flow in the vicinity of ferromagnetic prosthetic material such as that encoded, this being in the direction of the slice select gradient. As with
in the rings of mechanical heart valves. Under optimal conditions, Doppler flow measurement, phase contrast velocity mapping is repre-
however, typical appearances of a jet on SSFP imaging include a rela- sented by shifts of phase and so is subject to aliasing. To avoid this, an
tively bright jet core, clearly delineated on either side by dark lines that appropriate velocity-encoding range (Venc) should be chosen before
represent the shear layers (see Fig. 30.1 and Fig. 30.4). But this is the acquiring velocity data in a particular cavity or vessel. An appropriate
case only if there is a coherent core and the image plane is aligned with Venc exceeds the peak expected velocity by 20% to 50%, meaning that
it. The same jet may appear dark, with no bright core, if the imaging 180 degrees of phase contrast corresponds to 120% to 150% of the
plane is displaced slightly to one side, tangential to the shear layer. For expected peak velocity. If the Venc is too low, set below the level of
this reason, careful alignment of planes, sequentially cross-cutting one the peak velocity, the result is aliasing, in which case the acquisition
cine acquisition with another and then another, is important in the should be repeated with the Venc increased appropriately. Too high a
assessment of heart valve disease by CMR. Each jet should be imaged Venc, however, reduces the sensitivity of velocity mapping by reduc-
in at least three planes (see Fig. 30.1), preferably six or more, before ing the amount of velocity-related phase shifts relative to background
the operator can be reasonably confident that the size, shape, and func- noise or other artifact-related phase shifts. Velocity mapping is used
tional significance of a jet have been adequately depicted. clinically for jet velocity measurements and for volume flow measure-
Gradient recalled echo (GRE) cine imaging using segmented k-space, ments. Each can be subject to artifacts, generally of distinct kinds in
also known as fast low-angle shot (FLASH)10 imaging, is generally veloc- each case, and these can give misleading clinical results if they are not
ity compensated and less subject to signal loss in the shear layer than is recognized and, if possible, corrected.

RVOT

LV
RV
RV

FIG. 30.4  Double-chambered right ventricle (RV), or subinfundibular stenosis, shown by steady-state free
precession cine imaging in oblique coronal (left) and short-axis (right) planes, with the systolic jet from the
hypertrophied lower part of the RV into the outflow tract arrowed. The pulmonary valve was not stenosed
in this patient. LV, Left ventricle; RVOT, right ventricular outflow tract.
370 SECTION III  Functional Disease

curved or dilated,20 but measurement of one directional component of


Jet Velocity Mapping for the Assessment of Stenoses velocity is adequate for most clinical applications.
In jet flow, measurement of the peak velocity of the jet and the rates Through-plane flow measurements are subject to particular kinds
of change of the peak velocity through time or use of the velocity time of inaccuracy, however, especially when used for regurgitant or shunt
integral17 may contribute to the assessment of the severity of stenosis flow rather than total flow measurements.8 Background phase offset
or possibly regurgitation. Recovery of signal is a prerequisite for calcula- errors can cause slight inaccuracies of velocity measurement across the
tion of velocity, so it is necessary for voxels to lie sufficiently within the whole field of view and through the whole cardiac cycle, typically increas-
coherent core of a jet to measure its velocity accurately. If a jet is narrow ing with distance from the center of the magnet. These errors have
and fragmented, this condition may be difficult to fulfill because there been identified as being mainly a result of concomitant Maxwell gra-
may not be a core of sufficient dimensions. Initial breath-hold cine dients, which can be calculated and corrected by using appropriate
acquisitions should be used, as described earlier, to decide whether it software, and to eddy currents in hardware components, which are
is worth trying to measure jet velocity by CMR. These, possibly followed harder to predict and eliminate.7 The result can be an upward or down-
by a preliminary in-plane breath-hold velocity map, can then be used ward shift of the baseline of the flow curve, which can significantly
to locate a through-plane breath-hold velocity map to measure the affect the calculation of a regurgitant fraction. The amount of the shift
velocities of the core of the jet. Breath-hold sequences are recommended of the flow curve depends not only on the underlying cause of the
because their echo time will be short and so will minimize several artifact but also on the Venc that is chosen (the higher the Venc, the
potential artifacts,8 and any respiratory motion of the jet will also be greater the error), and on the cross-sectional area of the vessel measured
avoided. What may be harder to avoid is movement of the valve related (the larger the area, the greater the error). A further source of inaccuracy
to cardiac motion. The plane chosen should therefore be located at the can be movement of the plane of a regurgitant valve itself relative to
phase of peak flow, or, better still, a moving plane is located by using the plane of acquisition. Because such movements, for example, of the
a motion-tracking method of acquisition, if available.18 In spite of these aortic valve in diastole or of the mitral valve in systole, displace a volume
precautions, velocity maps, particularly of a narrow jet, may give mis- of blood in the direction opposite that of the regurgitant flow, they
leading values. Of particular concern, voxels in shear layer at the edge cause underestimation of the regurgitation measured, the error increas-
of a jet are subject to signal loss and possible phase errors caused by ing with the amount of valve movement and the cross section of the
abrupt deceleration. Apparent velocities of edge pixels should therefore flow area measured. It is an error that can be overcome by appropriately
be treated with suspicion if they differ from those in the core of the implemented motion tracking,18 although this is not yet available on
jet, and they may degrade attempts to measure volume flow at or down- most systems. Both manufacturers and users need to be aware of these
stream of a narrow orifice. These problems explain why CMR velocity potential sources of inaccuracy and work together toward the optimiza-
mapping is generally less suitable than Doppler echocardiography for tion of systems and sequences.
the assessment of right ventricular (RV) or pulmonary artery pressures
by measurement of the velocities of relatively narrow tricuspid or pul- Fourier Cardiovascular Magnetic Resonance
monary regurgitant jets. Velocity Traces
In echocardiography, it became routine for jet velocity measurements Another type of velocity acquisition that is worth mentioning, although
to be converted to estimated differences of pressure, often referred to not widely implemented, is CMR Fourier velocity mapping. In this
as gradients, in units (mm Hg) that were familiar from the use of technique, the spatial phase-encoding gradient pulses are replaced by
catheterization. This conversion relies on assumptions that may not velocity phase-encoding bipolar gradient waveforms. The resulting
always be applicable (e.g., that all of the jet or stream reaches a uniform dataset has only a single spatial dimension (in the readout direction)
peak velocity and that virtually all of its kinetic energy is dissipated in and measures one component of velocity in the direction of velocity-
turbulence). We recommend that peak velocities measured at specific encoding gradient.21,22 Like M-mode Doppler, this approach could be
locations, by either CMR or Doppler echocardiography, are quoted as adapted to real-time acquisition and display of the time course of flow
such without conversion to presumed differences of pressure. through a particular valve or orifice (Fig. 30.5). Arguably, it should be
The velocity of a jet through an orifice is, of course, affected by the made available for routine CMR investigation―for example, to assist
rate of flow as well as by the degree of narrowing, so a low rate of in the evaluation of congenital and valvular heart diseases as well in
ejection, for example, because of ventricular dysfunction, will reduce the diagnosis of constrictive pericardial disease and diastolic ventricular
the degree of elevation of velocity through a stenosed outflow valve. dysfunction―although the technique has yet to be optimized and proven
in a wide spectrum of clinical use.
Regurgitant Flow Measurements
Phase contrast velocity mapping is able to measure velocities in pixels STENOTIC HEART VALVE AND OUTFLOW
throughout the plane of acquisition, which can be acquired in any TRACT LESIONS
chosen orientation. An advantage of this is that the volume of flow
passing through an image plane can be calculated. This is done by Aortic Valve Stenosis
multiplying the cross-sectional area of a vessel, for example, the aorta Stenosis at the level of the aortic valve can be a result of congenital
or pulmonary trunk, by the mean through-plane component of velocity malformation of the valve, rheumatic heart disease or degenerative
in that area for each phase of the cardiac cycle, after delineating the disease in older patients. On the whole, echocardiographic assessment
area of the vessel in every frame of the acquisition. In this way, CMR is reliable, but CMR may be called upon when there is limited echo-
is able to measure the volumes of forward or reversed flow in a large cardiographic access, when there are conflicting findings, in complex
vessel. Furthermore, CMR velocity mapping is the only imaging tech- disease with stenosis at more than one level, or for the assessment of
nique that has the potential to acquire comprehensive information (i.e., left ventricular (LV) volume and mass. Ranges of measurements reflect-
three-dimensional [3D], three directionally encoded, time-resolved ing different severities of aortic stenosis (AS) and other valve lesions1
acquisition) and is well suited for studying spatial and temporal pat- are shown in Table 30.1.
terns of flow in the human cardiovascular system.19 This can be of Several publications on CMR quantification of AS have used pla-
interest where flow is multidirectional in cavities or in vessels that are nimetry of the orifice, or rather the jet, to make direct estimates of
CHAPTER 30  Valvular Heart Disease 371

Velo m/s

S 4.00
3.50
3.00
2.50
2.00
1.50
1.00
0.50
0.00 S I
-0.50
I -1.00
-1.50
FIG. 30.5  Left, Systolic frame from cine steady-state free precession acquired in the left ventricular outflow
tract (LVOT) in a patient with hypertrophic obstructive cardiomyopathy and severe aortic valve stenosis.
Position of the cylindrical volume excitation along the LVOT, with Fourier velocity encoding along the long
axis of the cylinder. Right, A systolic frame from the cine Fourier velocity acquisition (Velo m/s) showing the
blood velocity spectrum or distribution (vertical axis) at each position along the cylinder (horizontal axis). The
bright line at 0 m/s is from stationary tissue. Notice the two forward peaks (arrows) from the subvalvular
(open arrow) and valvular stenoses (solid arrow). I, Inferior; S, superior.

orifice area.12–15 The results seem to have been encouraging, although jet is fragmented. Multiple in-plane cine views together with assessment
the spatial resolution, meaning slice thickness as well as pixel dimen- of the impact of the lesion of ventricular mass and function are par-
sions, of cine acquisitions may not be adequate when it comes to the ticularly important in such cases and, in combination, should make
delineation of narrow, splayed, or fragmented jets. In our experience, the severity of the stenosis apparent.
there is a proportion of orifices and jets that cannot be adequately Cine imaging should show the location and direction of the jet core,
defined by CMR planimetry. and the orientation of the slit, if applicable. In-plane breath-hold veloc-
By echocardiography, direct planimetry on two-dimensional (2D) ity mapping with the direction of velocity encoding aligned with that
images, peak stenotic jet velocity and mean jet velocity are possible of the jet can be helpful to visualize the jet, but it is likely to underes-
approaches to the assessment of the severity of stenosis, although pla- timate peak velocity if the jet is narrow. The above acquisitions should
nimetry may be unsatisfactory in calcified or deformed valves, and then be used to align a velocity-mapping slice to transect the jet core,
simple jet velocities depend on flow rate as well as stenotic severity. with velocity of an appropriate range (Venc) encoded through the image
Probably the most widely accepted echocardiographic approach uses plane. The velocity of the jet should be analyzed with caution in the
the continuity equation to calculate the area of the valve orifice.23,24 resulting image, particularly if the stenosis is severe. Signal loss can
Using this, the volume flow through the left ventricular outflow tract allow background noise or other artifacts to contribute spurious phase
(LVOT) is calculated by multiplying the LVOT velocity time integral shifts, particularly in the shear region at the edge of a jet, so peak jet
(VTI) by the LVOT cross-sectional area (typically calculated echocar- velocity should be measured in a region of good signal recovery within
diographically from its diameter) to give outflow volume, then dividing the core of the jet.
this volume by the VTI of the jet at the stenosis, which gives a measure-
ment representing the stenotic valve area. Comparable approaches using Emerging Role of Cardiovascular Magnetic Resonance
CMR measurements have been described.17,25 for Selecting Anatomically Suitable Patients for
A stenosed bicuspid aortic valve generally has a slit-like orifice (see Transcatheter Aortic Valve Implantation
Fig. 30.1), whereas degenerate or calcified valves can have irregular or Transcatheter aortic valve implantation (TAVI) is an effective alternative
three-pointed orifices, and the considerations described earlier regarding to surgical aortic valve replacement in patients with high surgical risk
jet shape and the locations of image slices are important. Cine imaging or who are inoperable.26,27 Cardiac imaging plays an essential role for
should aim to depict the jet in several orientations, which also help to assessment of severity of aortic calve stenosis and for selecting anatomi-
determine the best strategy for jet velocity mapping and planimetry of cally suitable patients for this procedure. A number of imaging modalities
the valve.11–15 Both of these should be attempted; unfortunately, each are often used for this purpose, including echocardiography, cardiac
is less likely to be accurate if the orifice is small and irregular and the computed tomography (CCT) and CMR. Each imaging method has
jet is fragmented. its advantages and limitations.28 The use of CMR in patients undergo-
For direct planimetry of the valve orifice, high-resolution breath- ing, or being considered for, TAVI is likely to expand. CMR provides
hold FLASH or SSFP imaging in a stack of about five slices, shifting several important pieces of structural information before consideration
3 mm at a time from immediately upstream to just downstream of the for TAVI; in addition to accurate quantification of ventricular volumes
orifice, should be acquired. The best cine, which is probably the one and function, aortic valve planimetry can be performed to accurately
that shows the smallest, most clearly delineated cross section of the jet, determine valve area, and late gadolinium enhancement (LGE) can
should then be used for direct planimetry. The jet velocity and orifice show areas of myocardial scarring post TAVI.29 Furthermore, work from
area might not be accurately measurable in severe stenosis or when the our institution has shown that CMR can provide accurate aortic annulus
372 SECTION III  Functional Disease

measurements that, in terms of predicting the presence and severity of to the septum by the low pressure of the jet, a fluid dynamic phenom-
aortic regurgitation (AR) after TAVI, are comparable with those obtained enon known as the Venturi effect, causing systolic anterior motion of
using CT (Figs. 30.6 and 30.7).30 Also, in cases where CT angiography the mitral valve, with probable vibration of the leaflet. Although vibra-
is not available or is deemed inappropriate, magnetic resonance angi- tion is rapid and unlikely to be visible directly, it may explain why
ography (MRA) of the peripheral vasculature can be performed to velocity mapping tends to be unsuccessful when this type of stenosis
assess access routes (Fig. 30.8). Following TAVI, CMR can be used to is significant, whereas Doppler echocardiography is probably more reli-
provide an accurate estimate of ventricular function, and a recent pub- able. Such stenoses tend to become very much more severe on exertion,
lication has also shown the accuracy of CMR to detect and quantify owing to enhancement of the Venturi effect with rapid ejection, and
paravalvular leak.31 Ultimately, however, unless any postprocedural may be associated with induced mitral regurgitation. Because admin-
CMR-specific measurements can be shown to lead to treatment changes istration of pharmacologic stress can be risky in such cases, with the
affecting prognosis, it will remain highly debatable whether it is worth patient lying in the bore of a magnet, echocardiography with exercise
the time and expense to perform CMR on a routine basis after TAVI; stress is a more appropriate approach to the assessment of severity.
echocardiography may be sufficient for the majority of cases. But in CMR is nevertheless valuable, giving good views of all parts of the
the longer term, a role may arise for percutaneous valve interventions ventricle and of any associated pathology of the LVOT.
such as TAVI to be completely MRI guided; real-time transarterial It is important to realize that LVOT and right ventricular outflow
implantation of the Medtronic CoreValve (using a modified, MRI- tract (RVOT) stenosis can be at more than one level and possibly at
compatible delivery device) has already been described in a swine model.32 three distinct levels, including the valve, in a single patient. The second
type of subaortic stenosis is caused by a subvalvular ridge or membrane,
Subaortic Stenosis usually only a few millimeters below the level of the valve. It is advisable
There are important anatomic differences between the left and the right here, as elsewhere, to acquire cine images of jet flow in several orienta-
ventricular outflow tracts resulting in distinct types of subvalvular ste- tions, especially as it can be hard to distinguish subvalvular from valvular
notic lesion. Two types of subaortic stenosis need to be distinguished. stenosis. In-plane velocity mapping can help to establish the level of
One that is typical of hypertrophic cardiomyopathy is caused by nar- flow acceleration, followed by velocity mapping through two planes,
rowing in the proximal part of the outflow tract between the hyper- each clearly labeled, immediately below and above the level of the valve.
trophied basal part of the septum and the septal leaflet of the mitral If subvalvular stenosis is present, the aortic valve leaflets, even if normal,
valve. Because the valve leaflet is mobile, its tip can be drawn still closer cannot be expected to open fully, owing to the Venturi effect, so

15 15 15
10 10 10

Difference (mm)
Difference (mm)

Difference (mm)

CMR - CCT
CMR - CCT

CMR - CCT

5 5 5
0 0 0
20 30 40 20 30 40 20 30 40
-5 -5 -5
-10 -10 -10
-15 -15 -15
A CMR - CCT B CMR - CCT C CMR - CCT
Average (mm) Average (mm) Average (mm)
FIG. 30.6  Bland-Altman plots demonstrating close agreement between cardiovascular magnetic resonance
(CMR)– and cardiac computed tomography (CCT)–derived aortic valve annulus diameters. The largest annulus
diameter (A), smallest annulus diameter (B), and average annulus diameter (C) show similar agreement.
Upper and lower dotted lines denote the 95% limits of agreement; the middle solid line is the bias. (From
Jabbour A, Ismail TF, Moat N, et al. Multimodality imaging in transcatheter aortic valve implantation and
postprocedural aortic regurgitation. J Am Coll Cardiol. 2011;58[21]:2165–2173, with permission.)

15 15 15
10 10 10
Difference (mm)
Difference (mm)

Difference (mm)

CMR - TTE
CMR - CCT

CMR - TTE

5 5 5
0 0 0
20 30 40 20 30 40 20 30 40
-5 -5 -5
-10 -10 -10
-15 -15 -15
A CMR - CCT B CMR - TTE C CMR - TTE
Average (mm) Average (mm) Average (mm)
FIG. 30.7  Bland-Altman plots demonstrating closer agreement and lower bias for cardiovascular magnetic
resonance (CMR)– and cardiac computed tomography (CCT)–derived aortic valve annulus diameters (A)
compared with CMR–transthoracic echocardiography (TTE) (B) and CMR–CCT (C). Upper and lower dotted
lines are the 95% limits of agreement; the middle solid line is the bias. (From Jabbour A, Ismail TF, Moat
N, et al. Multimodality imaging in transcatheter aortic valve implantation and postprocedural aortic regurgita-
tion. J Am Coll Cardiol. 2011;58[21]:2165–2173.)
CHAPTER 30  Valvular Heart Disease 373

C E
FIG. 30.8  Selected frames from cine acquisition in the left ventricular outflow tract (A, B) and short-axis
view of the aortic valve (C) in a patient with severe aortic stenosis and moderate regurgitation under con-
sideration for transcatheter aortic valve implantation. Contrast-enhanced magnetic resonance angiography
of the subclavian arteries (D) and the femoroiliac arteries and abdominal aorta (E) depict good vascular access.

assessment of the aortic valve has to remain cautious when subvalvular probably translates into peak velocity measurements of the order of 3
stenosis is present unless it is clear that there is additional, more severe to 4 m/s by CMR jet velocity mapping, but as was outlined earlier, jet
stenosis at the level of the valve itself. velocity mapping can be subject to errors, particularly if acquisitions
are not perfectly located. The strength of CMR in this setting is in the
Pulmonary and Other Stenoses of the Right Ventricular evaluation of associated pathophysiology, particularly of RV hypertrophy
Outflow Tract and dysfunction, and possible additional levels of stenosis, including
Stenosis of the pulmonary valve generally presents as a congenital con- the branch pulmonary arteries. RV to pulmonary artery conduits are
dition early in life, either isolated or associated with other abnormalities also readily evaluated by CMR, although ferromagnetic valve rings or
of the RVOT and pulmonary arteries, as in tetralogy of Fallot. Trans- sternal wires may cause localized artifacts.
catheter or surgical relief of pulmonary stenosis is typically recommended Isolated pulmonary valve stenosis can also present later in life,
when there are peak-to-peak transpulmonary catheter pressure differ- although a more common progressive lesion in the adult popula-
ences of 40 mm Hg, or 30 mm Hg in symptomatic patients. This tion is double-chambered right ventricle, which can also be called
374 SECTION III  Functional Disease

subinfundibular stenosis33 (see Fig. 30.4). Double-chambered right and tricuspid valve planes in long-axis acquisitions.37 The ventricle
ventricle is a diagnosis that can be misinterpreted on echocardiography with the leaking valve will have excess stroke volume that represents
as a ventricular septal defect, which frequently coexists, or as stenosis at the volume of leakage. The accuracy of this approach is dependent on
infundibular or pulmonary valve level. The distinction is important and the image quality and accuracy of planimetry, which, if done manually,
is usually clear on CMR imaging because subinfundibular stenosis is takes considerable postprocessing time. Automated methods have still
treatable, if necessary, by transatrial surgical resection of the obstructing to be developed and validated. Hypertrophy and deformity of the right
muscle bands without the need for pulmonary valve replacement. It ventricle, for example, in congenital heart disease or after surgery for
is important to use the excellent access provided by CMR to visualize it, present particular problems. Standardized approaches need to be
the level and nature of any RVOT stenosis as clearly as possible. RV to taken to the inclusion or exclusion of myocardial trabeculations and any
pulmonary artery conduits and the function of their valves can also be aneurysmal regions of the RVOT.38 If appropriately performed, CMR
evaluated very effectively by CMR cine imaging and velocity mapping. measurements of right and LV volumes should have greater accuracy
and reproducibility than those attempted by any other technique.39
Mitral and Tricuspid Valve Stenosis Where more than one valve is regurgitant, ventricular volume mea-
Methods of assessment of stenosis are fairly similar for the mitral and surement can be combined with aortic or pulmonary phase contrast
tricuspid valves. Both can usually be adequately assessed by 2D and flow measurements to calculate regurgitation of the inflow valve. The
Doppler echocardiography, and more recently by 3D echocardiogra- systolic flow measured in the great vessel by velocity mapping is sub-
phy, which can contribute useful morphologic information. The cross- tracted from the stroke volume of the associated ventricle (e.g., mitral
sectional shape and area of the stenotic orifice can generally be seen and regurgitant volume equals the LV stroke volume minus systolic aortic
measured on CMR short-axis cine acquisitions and phase velocity maps, flow).4 But this depends on the accuracy of techniques used, which may
oriented to transect the jet at right angles at or very close to the orifice need to be validated for a particular CMR and image-processing system.
on the downstream side. Motion tracking should be used if available.
Planimetry of the stenotic mitral valves has been reported by using Aortic Regurgitation
SSFP cine imaging34 and has been found to yield orifice areas slightly Uncomplicated AR can be relatively easily measured by through-plane
larger than those measured echocardiographically (e.g., 1.65 vs. 1.5 cm2). velocity mapping, the velocity-mapping plane being located to transect
Not only the area and velocity of the jet but also the time course the aortic root just above valve level, at about the sinotubular junction
of jet flow are important, with loss of the normal biphasic mitral flow (Fig. 30.9). If the valve is unstenosed, a Venc of 2 m/s should be adequate,
pattern and prolonged elevation of velocity as stenosis increases in and it is important not to set the Venc too high to maintain the sensi-
severity. The slope of the decline in the maximum velocity has been tivity of the measurement. For this reason, mixed stenotic and regurgitant
used to estimate the pressure half-time (the time it takes until the initial valve disease presents a problem, requiring a higher Venc in the systolic
pressure difference across the orifice is halved).35 The time curve of phase at least. If possible, the higher Venc should be used for the systolic
pulmonary venous flow, with marked atrial systolic reversal, can also be frames only, and a lower, more sensitive Venc should be used for diastole,
informative.36 An important contribution of CMR is in the assessment when the reversed flow back toward the regurgitant valve will be of
of associated pathophysiology such as the degree of atrial dilatation, and low velocity. Flow measurement through a plane located higher across
possible changes of the pulmonary veins, pulmonary arteries, and RV the ascending aorta is also possible but will be more subject to under-
if pulmonary hypertension has developed secondary to mitral stenosis. estimation of regurgitant flow for the reason explained below.
An important issue is the volume change of the section of aortic
VALVULAR REGURGITATION root beneath the velocity mapping plane through the course of the
cycle. This volume change is related to ventricular contraction and the
General Principles compliance of the aorta, the aortic root being pulled downward and
In regurgitant heart valve disease, the optimal time for surgical inter- expanding in systole and returning, piston-like, through the plane of
vention depends on a balanced assessment of when valve dysfunction acquisition in the diastolic phase when the regurgitation is being mea-
is sufficiently severe to warrant the short- and long-term risks of surgery sured. This upward movement of the root accounts for apparent slight
but before the heart muscle sustains irreversible damage. The form, diastolic forward flow by velocity mapping in a healthy individual with
structure, and mobility of regurgitant valve leaflets, particularly mitral a competent valve, and it causes underestimation of the regurgitant
and tricuspid, should be characterized with a view to surgical repair. volume or fraction in almost all cases with AR. The error is small if the
Owing to the slice thickness of acquisitions, this may not be easy by root is small and immobile—for example, after previous surgery—but
CMR, and 2D echocardiography and 3D echocardiography are probably can be significant if the root is dilated and ventricular function is good.
preferable in this respect.2 The orientations of CMR slices need to be The appropriate way to avoid this underestimation is to use motion
optimized in relation to individual valve structures. tracking for the velocity acquisition.18 If this is not available, approximate
The need for intervention is determined partly by the severity of calculation of the change of volume of the root from its cross-sectional
the symptoms and objective clinical indices such as oxygen consump- area times the distance of valve plane movement may be attempted to
tion and exercise capacity, but the measurements of regurgitant volume give an idea of how much the underestimation of regurgitant flow is
or regurgitant fraction and ventricular function are important. Mea- likely to be.
surement of valvular regurgitation is clinically important but is difficult Timing of surgical aortic valve replacement in patients with signifi-
to quantify with current echocardiographic and angiographic techniques. cant aortic valve regurgitation is crucial. Current criteria for surgery
Several CMR methods are available to assess the severity of valvular in patients with significant AR concentrate on symptoms and dilatation/
regurgitation. impairment of the left ventricle. This is problematic because prognosis
Regurgitation of any single heart valve, if isolated and uncomplicated is already reduced by this stage, and earlier identification of patients
by shunting, may be calculated by comparison of the LV and RV stroke for surgery would be preferable and could be beneficial. Myerson et al.40
volumes measured by planimetry from a standard multislice short-axis showed the clinical utility of CMR flow quantification of aortic valve
volume acquisition, preferably using software that can combine short-axis regurgitation to predict future need for surgery in asymptomatic patients.
data with information on the locations and movements of the mitral In this study 113 patients with echocardiographic moderate or severe
CHAPTER 30  Valvular Heart Disease 375

500

400

300

200

100

0
0 200 400 600 800 1000
−100

−200

FIG. 30.9  Moderate aortic regurgitation shown by steady-state free precession cine imaging (top) and phase
contrast velocity mapping (bottom left and right). With respect to the oblique coronal left ventricular outflow
tract image (top left), the black lines mark the plane chosen for imaging the valve leaflets, and the white
lines show the plane chosen for through-plane flow measurement, close to the level of the sinotubular junc-
tion. The flow curve shows systolic forward and diastolic reversed flow through the velocity mapping plane,
recording a regurgitant fraction of 32% in this case. This is likely to be an underestimate, probably by at
least 5%, owing to upward diastolic movement of the dilated aortic root relative to the plane of velocity
acquisition.

AR were monitored for up to 9 years following a CMR scan, and the systemic side of the heart. Regurgitant fractions of 35% to 45% are, in
progression to symptoms or other indications for surgery were moni- our experience, typical of free pulmonary regurgitation. Higher values
tored. AR quantification identified outcome with high accuracy with may be attributable to additional factors such as an unusually capacitant
regurgitant fraction 33% progressed to surgery (mostly within 3 years) right ventricle on the upstream side45 and, on the downstream side,
(Fig. 30.10). This could provide a new paradigm for the timing of large and compliant pulmonary arteries, raised pulmonary vascular
surgical intervention but requires confirmation in a clinical trial. resistance at branch arterial or microvascular level, or combinations of
these.45 In other words, the pulmonary regurgitant fraction, above a
Pulmonary Regurgitation level of about 30%, reflects other aspects of right-sided pathophysiol-
Although significant pulmonary regurgitation is relatively uncommon ogy, which should be assessed, as much as dysfunction of the valve
as an isolated or acquired lesion, it is commonly found as a residual itself. We suggest use of the terms free or almost free rather than severe
lesion following repair of tetralogy of Fallot41,42 or valvotomy for con- pulmonary stenosis when there is not an effective valve and a regurgitant
genital pulmonary stenosis. Pulmonary regurgitation, even if free, fraction below 50%. If the pulmonary regurgitant fraction exceeds 50%,
meaning that there is no effective valve function, is usually well tolerated however, it should be called severe, and the contributory factors should
for decades, but late morbidity and mortality can ensue, related to be considered: for example, unusually compliant pulmonary arteries
eventual deterioration of RV function and the onset of potentially fatal and/or elevated pulmonary vascular resistance.
arrhythmias.43,44 Although uncontained by effective valve action, the For pulmonary flow measurement, a suitable velocity-mapping plane
pulmonary regurgitant volumes and regurgitant fractions can remain transects the pulmonary trunk, or the ventriculopulmonary conduit,
surprisingly low and considerably less than would be expected on the above expected valve level and proximal to the bifurcation (Fig. 30.11).
376 SECTION III  Functional Disease

1.0 Regurgitant fraction ≤33% 1.0 LVEDV ≤246 mL


0.91

Survival without surgery


Survival without surgery

0.8 0.8 0.77

0.6 0.6 P < .0001


P < .0001

0.4 0.4 LVEDV >246 mL


Regurgitant fraction > 33%
0.2 0.2
0.15

A 0.0 0 B 0.0
0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6 7 8
No. remaining: No. remaining: Years
RF ≤33% 72 57 42 33 19 11 6 1 1 LVEDV ≤246 mL 60 50 38 29 16 10 6 1 1
RF >33% 41 18 11 7 3 3 1 0 0 LVEDV >246 mL 53 26 15 11 6 4 2 0 0

1.0 CMR regurgitant fraction ≤33% 1.0 1.00


0.95
0.90
Survival without surgery

Survival without surgery


0.8 P < .0001
0.8
P < .0001 for regurgitant fraction
0.6 P = .59 for difference 0.6 0.64
between centers
Other centers Regurgitant fraction ≤33%
0.4 CMR regurgitant 0.4 Regurgitant fraction >33%
Oxford
fraction >33%
0.2 0.2 LVEDV ≤246 mL
LVEDV >246 mL
0.0 0 0.0 0
0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6 7 8
C Years D Years
FIG. 30.10  Kaplan-Meier graphs for survival without surgery in 113 asymptomatic subjects with at least
moderate aortic regurgitation initially treated conservatively and followed for up to 9 years. (A) Stratified by
the highest aortic regurgitant fraction (RF) measured by cardiovascular magnetic resonance (CMR) during
follow-up ≤33% (N = 74) and >33% (N = 39), with the time of the scan with highest RF used as the baseline.
(B) Similar graph stratified by CMR-derived left ventricular end-diastolic volume index (LVEDV) ≤246 mL
(N = 60) and >246 mL (N = 53). (C) Same graph as in part A, stratified by both highest CMR RF and CMR
center. (D) Similar graph stratified by both highest CMR RF and LVEDV. (From Myerson SG, d’Arcy J, Mohi-
addin R, et al. Aortic regurgitation quantification using cardiocascular magnetic resonance: association with
clinical outcome. Circulation. 2012;126:1452–1460.)

Viewing of RVOT cines in sagittal and oblique cross-cut orientations orthogonal to the central part of the valve, progressing down from
should help to determine the most appropriate level in a particular the superior commissure through each scallop of the valve (A1 to P1,
patient. A Venc of 2 m/s is usually sufficient to measure systolic and A2 to P2, A3 to P3) so that any regurgitant jet, prolapse, or tethering of
diastolic velocities, but a higher Venc will be needed if there is a degree part of the valve can be visualized and located with a view to possible
of stenosis. surgical repair.
Several approaches to the quantification of mitral regurgitation have
Mitral Regurgitation been described,4–6 and each can be subject to errors. If the other three
Mitral regurgitation is probably the most difficult valve lesion to assess heart valves are demonstrably competent, then:
by CMR. If the regurgitant orifice is discrete, rounded, and in the severe
range, it may be possible to measure the cross-sectional area of the Mitral regurgitant volume = the stroke volume difference
jet by planimetry (Fig. 30.12). But the orifice is often extended and (LVSV − RVSV )
narrow, perhaps with more than one defect, and prolapse of a leaflet Mitral regurgitant fraction = mitral regurgitant volume LVSV
can cause asymmetric attachment of a fan-like jet to one leaflet or Eq. 30.1
wall of the atrium, which can be very difficult to evaluate visually
(Fig. 30.13). The thickness of CMR slices also makes the assessment But accurate calculation of RV as well as LV stroke volume is based on
of valve structure problematic in some cases, and the comments above four sets of relatively time-consuming multislice area measurements.
about cine acquisition in multiple slices are important. Visualization An alternative, more widely applicable approach is to calculate:
of the dynamic morphology of the valve and its suspensory apparatus
is needed with a view to repair.46 To achieve this, a contiguous stack of Mitral regurgitant volume = LV stroke volume − LV
5- or 6-mm thick cine slices is recommended to give comprehensive outflow volume (measured as systolic aortic forward
coverage of all scallops of the mitral valve, starting at the anterolat- flow, by mapping velocities through a plane
eral commissure and continuing down to the inferior commissure. transecting the aortic root above th
he aortic valve)
The stack is oriented parallel to an oblique LVOT long-axis plane, Eq. 30.2
CHAPTER 30  Valvular Heart Disease 377

PA

RV

30
Late diastolic
20 forward flow
Systolic
10
Pulmonary flow

forward flow

0
0 100 200 300 400 500 600 700 800 900 1000
Diastolic
-10 Time (ms)
reversal

-20

-30

FIG. 30.11  Free pulmonary regurgitation, late after repair of tetralogy of Fallot, shown by using steady-state
free precession cine imaging (top left) and through-plane velocity mapping (top right and below). There was
no evidence of effective valve action. The regurgitant fraction was 38%, which is typical of such cases.
Movement of the outflow tract is not a problem after this type of surgery. Notice the early diastolic reversal
of flow, when blood pours back to the right ventricle (RV) from the compliant pulmonary artery (PA), and the
late diastolic forward flow, when the full ventricle acts as an open conduit for flow returning from the sys-
temic veins, boosted by atrial systole.

FIG. 30.12  A central jet of mitral regurgitation depicted passing in-plane (left) and through-plane (white
arrows) relative to steady-state free precession cine acquisitions. In this case, the through-plane jet (black
arrow) shows a bright, coherent core that can be fairly clearly delineated, making planimetry feasible. But
this is not always the case; regurgitant jets may be narrow and fan-like or fragmented.
378 SECTION III  Functional Disease

Tricuspid Regurgitation
Mild tricuspid regurgitation is common in healthy individuals. More
severe tricuspid regurgitation can occur owing to congenital malforma-
tion of the valve, notably in Ebstein anomaly, or to acquired heart
disease, including any cause of dilation of the RV with secondary failure
of coaptation. In Ebstein anomaly, parts of the tricuspid valve, usually
the septal and mural leaflets, are displaced toward the apex with “atri-
alization” of the basal and inferior parts of the RV. This is commonly
associated with severe tricuspid regurgitation and, in some cases, an
atrial septal defect or pulmonary valve dysfunction. If the tricuspid
regurgitant jet is not visible in a standard four-chamber view, a contigu-
ous stack of transaxial cines can be acquired to visualize all parts of
the tricuspid valve and right ventricle and to locate the regurgitant
stream, which should then be cross-cut vertically and again horizontally
before velocity mapping of the jet. Transaxial cines are also usually
more suitable than short-axis cines for measuring RV volumes in most
cases of Ebstein anomaly.
FIG. 30.13  A mitral regurgitant jet (arrow) attaching asymmetrically to If pulmonary resistance is normal or nearly normal, the pressure
the posterolateral leaflet of the valve from the dominant left ventricle difference propelling the jet of tricuspid regurgitation is low in com-
in a patient with a ventricular septal defect and subaortic outlet right parison with regurgitant lesions of the left heart, and through-plane
ventricle. The patient, who had undergone a Fontan procedure, was
velocity mapping, encoded through a plane transecting the tricuspid
fluid loaded at the time, with a pleural effusion, which can be seen as
bright layers adjacent to the lung. The mitral regurgitation was less
regurgitant jet, is a very useful way of determining the size and shape
severe after diuresis. of the regurgitant orifice (Fig. 30.15). The Venc that is chosen is likely
to be in the range of 2 to 3 m/s, depending on relative RV and atrial
pressures. The relationships between orifice area and severity are not
simple, but tricuspid regurgitant orifice diameters of 6 mm are associ-
A third approach is to measure diastolic mitral inflow and systolic aortic ated with severe regurgitation. The driving difference of pressure is
outflow: clearly relevant, however. In the presence of pulmonary hypertension,
the assessment of tricuspid regurgitation becomes more like that of
Mitral regurgitant volume = diastolic mitral inflow mitral regurgitation.
Eq. 30.3
− systolic aortic outflow
CARDIOVASCULAR MAGNETIC RESONANCE IN
But owing to systolic displacement of the mitral valve, a moving slice
acquisition should be used for these flow measurements, particularly
PATIENTS WITH MECHANICAL HEART VALVES
the diastolic mitral inflow volume,18 and this has yet to be implemented The metal rings of prosthetic heart valves generally have only slight
on most commercial systems. ferromagnetic properties, so torque on the ring in the magnetic field
Through-plane velocity mapping is also recommended for further is small in comparison with the strength of anchorage necessary for
visualization (rather than quantification) of the regurgitant jet(s) on the valve to stay in place. It is therefore regarded as safe to image
the atrial side of the line of coaptation. patients with prosthetic heart valves,48 although variable amounts of
In severe mitral regurgitation, surgery is indicated if patients are image artifact are caused by metallic components, depending upon
symptomatic or LV dilatation/impairment developed but prognosis the nature of the alloy and the imaging sequence used. Lack of signal
is already reduced by this stage. Operating on asymptomatic patients in blood passing metallic components can exaggerate apparent dis-
has been advocated if valve repair is likely; however, identifying suit- turbances of flow, so caution is required in the assessment of possible
able patients for early surgery is difficult. Myerson and co-workers47 stenosis, regurgitation, or paravalvar leaks in this situation. A narrow
showed significant associations between quantification of mitral regur- regurgitant “wash jet” is normal through most prosthetic valves in the
gitation with CMR and outcome which was superior to CMR-derived closed position.
LV volume, and echocardiographic grading of regurgitation which Evaluation of prosthetic valve function by catheter and echocar-
might prove useful for identifying suitable patients for early mitral diography is also more difficult than with native valves, so CMR may
valve repair/replacement. Some 109 asymptomatic patients with echo- be called upon. Although velocity mapping is unlikely to be accurate
cardiographic moderate or severe mitral regurgitation had baseline in the immediate vicinity of a valve, volume flow, regurgitant fraction,
CMR scans and were followed-up for up to 8 years. CMR quanti- and ventricular function may be studied as for a native valve as long
fication accurately identified patients who progressed to symptoms as appropriate planes of imaging can be chosen outside the magnetic
or other indications for surgery; 91% of subjects with regurgitant field distortion caused by metal of the ring.
volume ≤55 mL survived to 5 years without surgery compared with
only 21% with regurgitant volume >55 mL (P < .0001). A similar Assessment of Interstitial Myocardial Fibrosis in
separation was observed for regurgitant fraction ≤40% and >40%. Valvular Heart Disease
CMR-derived end-diastolic volume index showed a weaker asso- Myocardial Late Gadolinium Enhancement
ciation with outcome (proportions surviving without surgery at 5 Aortic valve stenosis or regurgitation-related diffuse interstitial myo-
years, 90% for left ventricular end-diastolic volume index <100 mL/ cardial fibrosis in response to pressure and volume overload is well
m2 vs. 48% for ≥100 mL/m2) and added little to the discriminatory known and clinically important. LGE can be detected in patients with
(Fig. 30.14). AS (Fig. 30.16).49–51 Good correlation between LGE and histology for
CHAPTER 30  Valvular Heart Disease 379

1.0 Regurgitant volume ≤55 mL 1.0 Regurgitant fraction ≤40%


0.91 0.89

0.8 0.8
Regurgitant fraction 41%−50%
Survival without surgery

P < .0001
0.6 0.6 0.59

0.4 0.4
Regurgitant fraction >50%

0.2 0.21 0.2


Regurgitant volume ≤55 mL 0.16
P < .0001

0.0 0.0

A 0 1 2 3 4 5 B 0 1 2 3 4 5
Years Years
No. remaining: No. remaining:
RVol ≤55 mL 80 61 35 24 16 12 R fract ≤40% 67 52 30 21 14 11
RVol >55 mL 29 17 9 5 4 2 R fract 41%−50% 25 18 11 5 4 3
R fract > 50% 17 8 3 3 2 0

1.0 LVEDVi <100 mL/m2 1.0 Echo ERO <0.40 cm2


0.90
0.86
0.8 0.8
Survival without surgery

0.64
0.6 0.6
Echo ERO ≥0.40 cm2
0.48
0.4 LVEDVi ≥100 mL/m2 0.4

0.2 P < .0001 0.2 P = .36

0.0 0.0

0 1 2 3 4 5 0 1 2 3 4 5
C D Years
No. remaining: Years No. remaining:
LVEDVi <100 mL/m2 56 41 25 16 11 9 Echo ERO <0.40 cm2 28 19 7 4 3 3
LVEDVi ≥100 mL/m2 53 36 18 13 9 5 Echo ERO ≥0.40 cm2 25 19 8 5 4 3
FIG. 30.14  Kaplan-Meier graphs for survival without surgery in 109 asymptomatic subjects with at least
moderate mitral regurgitation initially treated conservatively and followed up for up to 8 years stratified by
cardiovascular magnetic resonance derived (A) mitral regurgitant volume (RVol), (B) mitral regurgitant fraction
(R fract), (C) left ventricular end-diastolic volume index (LVEDVi), and (D) echocardiographic effective regur-
gitant orifice area (Echo ERO) <0.40 and ≥0.40 cm2 (N = 53 for this group). (From Myerson SG, d’Arcy J,
Christiansen JP, et al. Determination of clinical outcome in mitral regurgitation with cardiovascular magnetic
resonance quantification. Circulation. 2016;133:2287–2296, with permission.)

the extent of interstitial myocardial fibrosis has been demonstrated,49 Myocardial T1 Mapping
and LGE directly related to increasing LVH50 and inversely associated More recently, myocardial T1 mapping has been used to assess diffuse
with the degree of functional recovery after aortic valve replacement myocardial fibrosis, which is associated with increased collagen content
(AVR),52 and all-cause mortality late after AVR.53,49 In addition, midwall and increased myocardial extracellular volume (ECV) fraction.55 T1
fibrosis has been shown to predict mortality, independent of ejection mapping is a novel technique that directly measures the T1 relaxation
fraction.54 LGE relies on the relative signal intensity of abnormal myo- time of the myocardium, allowing quantification on a standardized
cardium compared with the “normal-appearing” myocardium, which scale before (native) and after the administration of gadolinium contrast.
needs to be “nulled” by setting parameters during image acquisition There are multiple sequences and methods that can be used to quantify
(inversion time). This technique is therefore insensitive to more homo- T1 in patients and there are variations at different field strengths (please
geneous changes seen with interstitial fibrosis. refer to Chapter 32 for more details). Native T1 has been shown to be
FIG. 30.15  Severe tricuspid regurgitation. The regurgitant jet or stream may be visible on steady-state free
precession cine imaging (arrow), but it is much more clearly delineated by phase contrast velocity mapping,
encoded through a plane that transects the jet, as indicated by the white lines, with a Venc of 250 cm/s.
The velocity map shows the triangular cross section of the regurgitant jet, 10 to 20 mm wide in this case.

A1 A3

A2 A4
FIG. 30.16  Diastolic (A1) and systolic (A2) frames from complete cine steady-state free precession (SSFP)
acquisition in the ventricular outflow tract view in a patient with severe aortic stenosis. Aortic valve leaflets
are thick with systolic doming and systolic stenotic jet formation. (A3) Systolic frame from cine acquisition
in the short axis of the aortic valve depicting severely reduced aortic valve area (0.5 cm2). (A4) A systolic
velocity mapping frame in a plane corresponding to A3, recorded a peak velocity of 5.0 m/s. Concentric left
ventricular hypertrophy is obvious.
CHAPTER 30  Valvular Heart Disease 381

B1

B2
FIG. 30.16, cont’d  (B1) Diastolic frames from cine SSFP acquisition in the short-axis view of the left ventricle
from base to apex in the patient shown in Fig. 30.10A and (B2) the corresponding late gadolinium enhance-
ment images showing focci as well as diffuse myocardial enhancement patterns in keeping with focal and
diffuse fibrosis.

greater in patients with severe symptomatic AS compared with control Diffuse myocardial fibrosis has also been shown to be an independent
subjects, and is moderately correlated with fibrosis on histology.53 predictor of exercise capacity in severe AS before AVR. Six months after
However, there is marked overlap in native T1 values between asymp- AVR, diffuse fibrosis did not significantly decrease,57 as has been found
tomatic patients with AS and control subjects. T1 values correcting for previously on histology.58 T1 mapping therefore has the potential to
changes in the blood pool (partition coefficient and ECV methods) detect diffuse myocardial fibrosis before the onset of myocardial dys-
show better discrimination and excellent reproducibility between healthy function and might better identify asymptomatic patients at high risk,
volunteers and patients with AS (Fig. 30.17),56,57 although overlap remains. although further prognostic studies evaluating ECV are needed.
382 SECTION III  Functional Disease

SLP SLP SLA

A 10 P A 10 P A 10 P
R R R
S S S
0 0 0

1
1
1

: 0.158 cm2 / 1018.5 ± 38.8 median 102 : 0.250 cm2 / 1056.4 ± 32.2 median 105 : 0.323 cm2 / 1152.8 ± 60.8 median 115

IRA IRA IRA


FIG. 30.17  Native T1 maps obtained in the mid short-axis view of the left ventricle using Siemens 448B
modified Look-Locker inversion sequence on a 1.5 T Siemens Avanto scanner. Left image is from a healthy
volunteer (septal T1 = 1018.5 ms); middle image is from a patient with moderate aortic stenosis (septal T1
= 1056.4 ms); right image is from a patient with severe aortic stenosis (septal T1 = 1152.8 ms). (Courtesy
Dr. Vassiliou Vassilis, Royal Brompton Hospital, London.)

provide valuable prognostic information in patients with valvular heart


CONCLUSION
disease as well as for optimal timing of aortic valve replacement in
For investigation of valvular heart disease, the important strengths of asymptomatic patients.
CMR lie in visualization and quantification of regurgitation; the mea-
surement of ventricular volumes, function, and mass; and the investiga-
tion of associated congenital or acquired pathology. Freedom of access
ACKNOWLEDGMENTS
and free orientation of planes, together with ability to measure post- We wish to thank our colleagues at the CMR Unit of the Royal Brompton
stenotic jet velocities, also give CMR a role in cases such as calcified Hospital for their help and support and the British Heart Foundation
AS, in valvular dysfunction of the right heart including ventriculopul- and CORDA, the Heart Charity, for support and grants.
monary conduits, and in patients with chest deformity or lung abnor-
mality where ultrasonic access may be limited. In addition, CMR
techniques for assessment of focal and diffuse myocardial fibrosis in
REFERENCES
valvular heart disease are unique biomarkers that have the potential to A full reference list is available online at ExpertConsult.com
CHAPTER 30  Valvular Heart Disease 382.e1

REFERENCES comparison with Doppler echocardiography. J Magn Reson Imaging.


1997;7:657–663.
1. Bonow RO, Carabello BA, Chatterjee K, et al. ACC/AHA Guidelines for 22. Steeden JA, Jones A, Pandya B, Atkinson D, Taylor AM, Muthurangu V.
the management of patients with valvular heart disease. Circulation. High-resolution slice-selective fourier velocity encoding in congenital
2006;114:e84–e231. heart disease using spiral SENSE with velocity unwrap. Magn Reson Med.
2. Myerson SG. Heart valve disease: investigation by cardiovascular 2012;67:1538–1546.
magnetic resonance. J Cardiovasc Magn Reson. 2012;14:7. 23. Otto CM, Pearlman AS, Comess KA, et al. Determination of the stenotic
3. Wyttenbach R, Bremerich J, Saeed M, Higgins CB. Integrated MR aortic valve area in adults using Doppler echocardiography. J Am Coll
imaging approach to valvular heart disease. Cardiol Clin. 1998;16(2): Cardiol. 1986;7(3):509–517.
277–294. 24. Otto CM. Valvular aortic stenosis: disease severity and timing of
4. Kon MW, Myerson SG, Moat NE, Pennell DJ. Quantification of intervention. J Am Coll Cardiol. 2006;47(11):2141–2151.
regurgitant fraction in mitral regurgitation by cardiovascular magnetic 25. Caruthers SD, Lin SJ, Brown P, et al. Practical value of cardiac magnetic
resonance: comparison of techniques. J Heart Valve Dis. resonance imaging for clinical quantification of aortic valve stenosis:
2004;13(4):600–607. comparison with echocardiography. Circulation. 2003;108(18):2236–
5. Westenberg JJ, Doornbos J, Versteegh MI, et al. Accurate quantitation of 2243.
regurgitant volume with MRI in patients selected for mitral valve repair. 26. Smith CR, Leon MB, Mack MJ, et al. Transcatheter versus surgical
Eur J Cardiothorac Surg. 2005;27(3):462–466. aortic-valve replacement in high-risk patients. N Engl J Med.
6. Gelfand EV, Hughes S, Hauser TH, et al. Severity of mitral and aortic 2011;364:2187–2198.
regurgitation as assessed by cardiovascular magnetic resonance: 27. Leon MB, Smith CR, Mack M, et al. Transcatheter aortic-valve
optimizing correlation with Doppler echocardiography. J Cardiovasc implantation for aortic stenosis in patients who cannot undergo surgery.
Magn Reson. 2006;8(3):503–507. N Engl J Med. 2010;363(17):1597–1607.
7. Kilner P, Gatehouse P, Firmin D. Flow measurement by magnetic 28. Zamorano JL, Goncalves A, Lang R. Imaging to select and guide
resonance: a unique asset worth optimising. J Cardiovasc Magn Reson. transcatheter aortic valve implantation. Eur Heart J. 2014;35:1578–1587.
2007;9:723–728. 29. Ribeiro HB, Larose É, de la Paz Ricapito M, et al. Myocardial injury
8. Chernobelsky A, Shubayev O, Comeau CR, Wolff SD. Baseline correction following transcatheter aortic valve implantation: insights from
of phase contrast images improves quantification of blood flow in the delayed-enhancement cardiovascular magnetic resonance.
great vessels. J Cardiovasc Magn Reson. 2007;9:681–685. EuroIntervention. 2015;11(2):205–213.
9. Kilner PJ, Firmin DN, Rees RSO, et al. Valve and great vessel stenosis: 30. Jabbour A, Ismail TF, Moat N, et al. Multimodality imaging in
assessment with CMR jet velocity mapping. Radiology. 1991;178:229–235. transcatheter aortic valve implantation and post-procedural aortic
10. Edelman RR, Wallner B, Singer A, Atkinson DJ, Saini S. Segmented regurgitation. J Am Coll Cardiol. 2011;58(21):2165–2173.
TurboFLASH: method for breath-hold CMR imaging of the liver with 31. Crouch G, Tully PJ, Bennetts J, et al. Quantitative assessment of
flexible contrast. Radiology. 1990;177:515. paravalvular regurgitation following transcatheter aortic valve
11. Friedrich MG, Schulz-Menger J, Poetsch T, Pilz B, Uhlich F, Dietz R. replacement. J Cardiovasc Magn Reson. 2015;17(1):32.
Quantification of valvular aortic stenosis by magnetic resonance imaging. 32. Kahlert P, Parohl N, Albert J, et al. Real-time magnetic resonance
Am Heart J. 2002;144(2):329–334. imaging-guided transarterial aortic valve implantation: in vivo evaluation
12. John AS, Dill T, Brandt RR, et al. Magnetic resonance to assess the aortic in swine. J Am Coll Cardiol. 2012;59(2):192–193.
valve area in aortic stenosis: how does it compare to current diagnostic 33. Kilner PJ, Sievers B, Meyer GP, Ho SY. Double-chambered right ventricle
standards? J Am Coll Cardiol. 2003;42(3):519–526. or sub-infundibular stenosis assessed by cardiovascular magnetic
13. Kupfahl C, Honold M, Meinhardt G, et al. Evaluation of aortic stenosis resonance. J Cardiovasc Magn Reson. 2002;4(2):373–379.
by cardiovascular magnetic resonance imaging: comparison with 34. Djavidani B, Debl K, Lenhart M, et al. Planimetry of mitral valve stenosis
established routine clinical techniques. Heart. 2004;90(8):893–901. by magnetic resonance imaging. J Am Coll Cardiol. 2005;45(12):
14. Debl K, Djavidani B, Seitz J, et al. Planimetry of aortic valve area in aortic 2048–2053.
stenosis by magnetic resonance imaging. Invest Radiol. 35. Lin SJ, Brown PA, Watkins MP, et al. Quantification of stenotic mitral
2005;40(10):631–636. valve area with magnetic resonance imaging and comparison with
15. Reant P, Lederlin M, Lafitte S, et al. Absolute assessment of aortic valve Doppler ultrasound. J Am Coll Cardiol. 2004;44(1):133–137.
stenosis by planimetry using cardiovascular magnetic resonance imaging: 36. Mohiaddin RH, Amanuma M, Kilner PJ, Pennell DJ, Manzara C,
comparison with transoesophageal echocardiography, transthoracic Longmore DB. MR phase-shift velocity mapping of mitral and
echocardiography, and cardiac catheterisation. Eur J Radiol. 2006;59(2): pulmonary venous flow. J Comput Assist Tomogr. 1991;15(2):237–243.
276–283. 37. Maceira AM, Prasad SK, Khan M, Pennell DJ. Normalized left ventricular
16. Mohiaddin RH, Longmore DB. Functional aspects of cardiovascular systolic and diastolic function by steady state free precession
nuclear magnetic resonance imaging: techniques and application. cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2006;8(3):
Circulation. 1993;88:264–281. 417–426.
17. Yap SC, van Geuns RJ, Meijboom FJ, et al. A simplified continuity 38. Davlouros PA, Kilner PJ, Hornung TS, et al. Right ventricular function in
equation approach to the quantification of stenotic bicuspid aortic valves adults with repaired tetralogy of Fallot assessed with cardiovascular
using velocity-encoded cardiovascular magnetic resonance. J Cardiovasc magnetic resonance imaging: detrimental role of right ventricular
Magn Reson. 2007;9:899–906. outflow aneurysms or akinesia and adverse right-to-left ventricular
18. Kozerke S, Schwitter J, Pedersen EM, Boesiger P. Aortic and mitral interaction. J Am Coll Cardiol. 2002;40(11):2044–2052.
regurgitation: quantification using moving slice velocity mapping. J Magn 39. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ.
Reson Imaging. 2001;14(2):106–112. Interstudy reproducibility of right ventricular volumes, function, and
19. Weigang E, Kari FA, Beyersdorf F, et al. Flow-sensitive four-dimensional mass with cardiovascular magnetic resonance. Am Heart J. 2004;147(2):
magnetic resonance imaging: flow patterns in ascending aortic 218–223.
aneurysms. Eur J Cardiothorac Surg. 2008;34:11–16. 40. Myerson SG, d’Arcy J, Mohiaddin R, et al. Clinical outcome in aortic
20. Frydrychowicz A, Arnold R, Hirtler D, et al. Multidirectional flow analysis regurgitation with cardiovascular magnetic resonance. Circulation.
by cardiovascular magnetic resonance in aneurysm development 2012;126(12):1452–1460.
following repair of aortic coarctation. J Cardiovasc Magn Reson. 41. Rebergen SA, Chin JGL, Ottenkamp J, van der Wall EE, De Roos A.
2008;10:30. Pulmonary regurgitation in the late post-operative follow-up of tetralogy
21. Mohiaddin RH, Gatehouse PD, Henien M, Firmin DN. Cine magnetic of Fallot: volumetric quantification by nuclear magnetic resonance
resonance Fourier velocimetry of blood flow through cardiac valves: velocity mapping. Circulation. 1993;88:2257–2266.
382.e2 SECTION III  Functional Disease

42. Chaturvedi RR, Redington AN. Pulmonary regurgitation in congenital aortic stenosis and hypertrophic cardiomyopathy: visualisation of focal
heart disease. Heart. 2007;93:880–889. fibrosis. Heart. 2006;92:1447–1451.
43. Gatzoulis MA, Balaji S, Webber SA, et al. Risk factors for arrhythmia and 51. Weidemann F, Herrmann S, Stork S, et al. Impact of myocardial fibrosis
sudden cardiac death late after repair of tetralogy of Fallot: a multicentre in patients with symptomatic severe aortic stenosis. Circulation. 2009;
study. Lancet. 2000;356(9234):975–981. 120:577–584.
44. Shinebourne EA, Babu-Narayan SV, Carvalho JS. Tetralogy of Fallot: 52. Hein S, Arnon E, Kostin S, et al. Progression from compensated hypertrophy
from fetus to adult. Heart. 2006;92(9):1353–1359. to failure in the pressure-overloaded human heart: structural deterioration
45. Kilner PJ, Balossino R, Dubini G, et al. Pulmonary regurgitation: the and compensatory mechanisms. Circulation. 2003;107:984–991.
effects of varying pulmonary artery compliance, and of increased 53. Dweck MR, Joshi S, Murigu T, et al. Midwall fibrosis is an independent
resistance proximal or distal to the compliance. Int J Cardiol. 2009;133: predictor of mortality in patients with aortic stenosis. J Am Coll Cardiol.
157–166. 2011;58:1271–1279.
46. Chan KM, Wage R, Symmonds K, et al. Towards comprehensive 54. Bull S, White SK, Piechnik SK, et al. Human non-contrast T1 values and
assessment of mitral regurgitation using cardiovascular magnetic correlation with histology in diffuse fibrosis. Heart. 2013;99:932–937.
resonance. J Cardiovasc Magn Reson. 2008;10:61. 55. Kockova R, Kacer P, Pirk P, et al. Native T1 relaxation time and
47. Myerson SG, d’Arcy J, Christiansen JP, et al. Determination of clinical extracellular volume fraction as accurate markers of diffuse myocardial
outcome in mitral regurgitation with cardiovascular magnetic resonance fibrosis in heart valve disease—comparison with targeted left ventricular
quantitation. Circulation. 2016;133:2287–2296. myocardial biopsy. Circ J. 2016;80:1202–1209.
48. Shellock FG. Prosthetic heart valves and annuloplasty rings: assessment 56. Sado DM, Flett AS, Banypersad SM, et al. Cardiovascular magnetic
of magnetic field interactions, heating, and artifacts at 1.5 Tesla. resonance measurement of myocardial extracellular volume in health and
J Cardiovasc Magn Reson. 2001;3(4):317–324. disease. Heart. 2012;98:1436–1441.
49. Azevedo CF, Nigri M, Higuchi ML, et al. Prognostic significance of 57. Chin CW, Semple S, Malley T, et al. Optimization and comparison of
myocardial fibrosis quantification by histopathology and magnetic myocardial T1 techniques at 3T in patients with aortic stenosis. Eur Heart
resonance imaging in patients with severe aortic valve disease. J Am Coll J Cardiovasc Imaging. 2014;15:556–565.
Cardiol. 2010;56:278–287. 58. Flett AS, Sado DM, Quarta G, et al. Diffuse myocardial fibrosis in severe
50. Debl K, Djavidani B, Buchner S, et al. Delayed hyperenhancement in aortic stenosis: an equilibrium contrast cardiovascular magnetic
magnetic resonance imaging of left ventricular hypertrophy caused by resonance study. Eur Heart J Cardiovasc Imaging. 2012;13:819–826.
31 
Role of Cardiovascular Magnetic Resonance
in Dilated Cardiomyopathy
Brian P. Halliday, Upasana Tayal, and Sanjay Prasad

Dilated cardiomyopathy (DCM) is defined as a disease of the myocardium and <1% of unaffected controls.16,20 Mutations in the LMNA gene, which
characterized by left ventricular dilatation and systolic impairment that encodes the lamin A/C protein, part of the nuclear envelope, are also
cannot be exclusively explained by abnormal loading conditions (such associated with DCM.21 These mutations are characterized by a highly
as hypertension or valvular heart disease) or coronary artery disease.1,2 penetrant and malignant form of the disease that requires aggressive
The true prevalence is debated because of a lack of large contemporary treatment at an early stage given high rates of sudden death and advanced
population studies.3 The original Olmsted County study, performed heart failure.22,23 Aside from variants in LMNA, the identification of
between 1975 and 1984, estimated the prevalence to be in the region of DCM disease-associated mutations holds most significance for relatives
1 in 2700 individuals.4 However, the calculated prevalence of hypertrophic of the proband, enabling cascade screening and discharge from follow-
cardiomyopathy in the same study has since been shown to be a gross up if found to be mutation carrier negative. Cascade screening and the
underestimate, possibly explained by the fact that echocardiography was identification of mild disease in asymptomatic individuals enables early
still a developing technique.5 Recent reports have estimated the prevalence diagnosis, disease monitoring, and appropriate intervention. There is
to be closer to 1 in 400 people in the United States.3 Nevertheless, DCM also huge opportunity for targeted genetic-based interventions with
is a commonly encountered condition, representing the most frequent the development of gene editing techniques.
indication for cardiac transplantation and a common cause of heart failure The most common acquired causes of DCM include exposure to
and sudden cardiac death (SCD). Despite therapeutic advances, 3-year toxins such as excess alcohol and cardiotoxic chemotherapies.1,8,9 The
treated mortality rates are estimated to be 12% to 20%.6,7 Definitive early toxic effects of these agents appear to be dose-dependent and cardiac
investigation giving a prompt and accurate diagnosis is therefore essential dysfunction often resolves after toxin removal.24 A previous episode
for the expedient introduction of targeted therapy. We will discuss the of viral myocarditis is another common cause and is thought to
benefits of cardiovascular magnetic resonance (CMR) in the investiga- evolve into DCM in up to 30% of cases.25 Less common, noninfective
tion of DCM after a brief overview of our current understanding of autoimmune inflammatory conditions can also present with a DCM
the disease. phenotype. Given the heterogeneity of response to these environmen-
tal insults, it appears likely that individual susceptibility is linked to
genetic factors.
BACKGROUND Peripartum cardiomyopathy (PPCM) is a rarer form of the disease
DCM represents the final common phenotype of a diverse range of characterized by the development of a DCM in the last trimester of
genetic and acquired insults (Table 31.1).8,9 In 20% to 35% of cases, pregnancy or in the first 6 months postpartum. Many studies have linked
at least one first-degree relative is affected; in 20% to 40% of cases, a the development of this condition to an abnormally cleaved segment of
causative genetic mutation can be demonstrated with modern tech- prolactin which is toxic to cardiomyocytes.26–28 A strong genetic com-
niques.2,3,10–12 Mutations in over 50 genes have now been linked with ponent to the condition has, however, also been demonstrated.29–31 In
the disease, the majority of which occur in autosomal genes.12–14 In a one study, women with PPCM were found to have a similar burden of
small proportion of cases the mutations are associated with broader genetic variants, particularly in TTN (15% of the cohort), compared
syndromes, most commonly neuromuscular disorders such as Duchenne with a population with idiopathic DCM.29 Similarly, undiagnosed DCM
muscular dystrophy.15 Inheritance is generally autosomal dominant, has been found to be more common in first-degree relatives of patients
although often marked by reduced penetrance and variable expressiv- with PPCM.31 This neatly demonstrates that PPCM and DCM have a
ity.3 In contrast to hypertrophic cardiomyopathy, mutations associated common genetic etiology, which may be unmasked in the presence of
with DCM occur in a diverse range of genes responsible for many dif- specific environmental triggers.
ferent cellular functions. The most common mutations occur in genes In summary, DCM is the result of a complex interplay between
encoding sarcomeric proteins, with mutations found less commonly genetic abnormalities, such as variants in TTN, and environmental
in genes responsible for the function of ion channels, the cytoskeleton, factors including hemodynamic, inflammatory, and toxic insults. The
and the nuclear envelope (see Table 31.1). interaction between these factors may help to explain the variable pen-
Truncating variants of the large titin gene, TTN, are the most common etrance and reduced expressivity of mutations and the heterogeneous
mutations causing DCM.16 Titin is the largest protein in the human response of different individuals to environmental insults.
body, spanning the length of the sarcomere, and it acts to generate and
regulate contractile force.17–19 Advances in genetic sequencing have
enabled more detailed study of TTN mutations and their role in DCM
TREATMENT
and other cardiac pathology. Truncating variants of TTN have now The current treatment strategies for dilated cardiomyopathy focus on the
been identified in 25% cases of familial DCM, 18% of sporadic cases use of neurohormonal medical therapies and cardiac resynchronization

383
384 SECTION III  Functional Disease

fraction without the need for geometrical assumptions and is there-


TABLE 31.1 
fore considered the gold standard noninvasive technique (Fig. 31.1).45,46
Genetica Acquired Analysis of regional function and myocardial strain can also be assessed
Sarcomeric Toxins using myocardial tissue tagging and feature tracking.47,48 Analysis of
TTN (25%, titin) Chemotherapy (anthracyclines, longitudinal strain using myocardial feature tracking has been shown
MYH6 (4%), MYH7 (4%) (myosin monoclonal antibodies) to predict adverse outcomes, independently of other predictors such
heavy chain) Alcohol as LVEF and late gadolinium enhancement (LGE), in patients with
TNNC1 (<1%), TNNT2 (3%), Recreational drugs (cocaine, DCM.47 Analysis of wall thickening and end-systolic wall stress and
TNNI3 (<1%) (troponin) amphetamines, anabolic steroids) visualization of impaired fiber shortening can also be performed.45,49,50
MYPN (3%–4%, myopalladin) Right ventricular (RV) size and function is abnormal in between
Infectious Insults 30% and 60% of cases of DCM.51,52 This is thought to be secondary to
Nuclear Envelope Viral myocarditis (parvovirus, human intrinsic myocardial dysfunction and increased afterload related to a
LMNA (6%, lamin A/C) herpes virus, Coxsackie) rise in pulmonary vascular resistance. CMR provides the gold standard
HIV noninvasive assessment of both RV size and function because of its
Cytoskeleton Chagas disease (Trypanosoma cruzi) three-dimensional capabilities.53,54 Accurate assessment can be challeng-
DMD (N/A, dystrophin) ing using other forms of imaging, such as echocardiography, because of
DES (<1%, desmin) Peripartum its complex and variable shape. Reduced RV ejection fraction on CMR
Autoimmune is an independent predictor of all-cause mortality and adverse heart
Ion Channels Giant cell myocarditis failure outcomes.51 This supports a role for its use in prognostication
SCN5A (2%–3%, sodium channel Multisystem disease (systemic lupus, of patients with DCM.
protein type 5 subunit) Churg-Strauss) CMR also allows accurate quantification of left atrial (LA) volume
using the biplane area-length method (Fig. 31.2).55 This compares favor-
Mitochondrial Endocrine ably against other noninvasive imaging methods because of its excellent
TAZ (NA, tafazzin)b Pheochromocytoma endocardial border definition and multiplanar imaging ability, even in
Hypo- and hyperthyroidism the presence of atrial fibrillation.56–59 LA size is often increased in cases
Sarcoplasmic Reticulum
PLN (<1%, phospholamban) of DCM secondary to pressure overload from LV diastolic impairment,
Iron Overload
functional mitral regurgitation, and atrial fibrillation. It has been pro-
a
Gene listed followed by prevalence and protein encoded in posed that the degree of LA enlargement acts as a barometer of diastolic
brackets.2,3 dysfunction and, consequently, there has been interest in the use of LA
b
X-linked genes, remainder of genes are autosomal. size to predict heart failure outcomes.58 Indeed, it has been demonstrated
that indexed LA volume calculated using CMR independently predicts
cardiac transplant-free survival in DCM.60 A cutoff value of >72 mL/
therapy to treat and prevent heart failure, and implantable cardioverter m2 has been shown to predict a 3-fold increase in adverse outcomes
defibrillators (ICDs) to prevent sudden cardiac death from malignant in patients with DCM.60
ventricular arrhythmia.32–35 Evidence strongly supports the use of neu- Functional mitral regurgitation is also a common consequence of
rohormonal therapies such as beta-blockers and angiotensin-converting DCM secondary to mitral annular dilatation and leaflet tethering sec-
enzyme inhibitors in patients with reduced left ventricular ejection ondary to LV impairment. A long-axis cine stack and a short-axis cine
fraction (LVEF), and cardiac resynchronization therapy in patients with image across the valve allows accurate assessment of all parts of the
reduced LVEF and prolonged QRS duration.32–35 Data on the use of valve apparatus, including individual leaflet scallops, chordae, and papil-
primary prevention ICD therapy in DCM are, however, slightly less lary muscles.61 Mitral regurgitant volume can be calculated by estimating
convincing.36,37 Current guidelines recommend consideration of ICD the aortic forward flow volume, using phase contrast flow imaging, and
therapy in patients with severely depressed ejection fraction, although subtracting this from the total LV stroke volume.59,60 This method has
this single-criterion approach lacks sensitivity and specificity.32,34 In been validated against volumes calculated from echocardiographic indices
clinical trials investigating the benefit of ICD therapy in patients with and catheterization data with good intertechnique agreement.62,63 Once
severely reduced LVEF, only 5% of patients received appropriate therapies again, the accurate assessment of the degree of functional mitral regur-
per year.36–39 This demonstrates that the majority of patients currently gitation provides important prognostic information in DCM.64
receiving primary prevention devices on the basis of guidelines do not Given the accuracy in functional assessment, CMR has been proposed
gain benefit. Conversely, large population studies have shown that 70% as the method of choice for the follow-up of patients with DCM after
to 80% of SCD sufferers do not have a severely reduced LVEF.40,41 Addi- pharmacologic and surgical intervention.65,66 Given the favorable interob-
tional parameters, which identify patients at risk of SCD, will allow us server variability compared with other methods of assessment, the use
to improve the selection of patients for ICD therapy.42–44 We discuss of CMR in clinical trials can reduce the sample size required, reducing
the potential of CMR techniques in risk stratification. the overall cost and time needed to complete the research.67 Moreover,
it may be possible to avoid repeated studies using less precise methods,
CARDIOVASCULAR MAGNETIC RESONANCE which are inconsistent. More precise adjustment of therapy and reduc-
ASSESSMENT OF DILATED CARDIOMYOPATHY tion of admission for repeat studies are likely to overcome the additional
costs of a CMR study.
Morphology and Function
Diagnosis and subsequent management of patients with DCM relies Tissue Characterization
on detailed assessment of left ventricular morphology and function. Apart from accurate and reproducible assessment of cardiac morphol-
LVEF is currently the sole arbiter in the selection of patients for device ogy and function, the major advantage of CMR in the assessment of
therapy; therefore precise measurement is crucial.32,34 CMR allows accu- DCM is its ability to perform tissue characterization using LGE. This
rate and reproducible assessment of LV volumes, mass, and ejection allows the detection and quantification of myocardial fibrosis. We also
CHAPTER 31  Role of Cardiovascular Magnetic Resonance in Dilated Cardiomyopathy 385

A B

C D
FIG. 31.1  Steady-state free precession images demonstrating biventricular dilatation with severe left ven-
tricular systolic impairment, a small pericardial effusion, and right pleural effusion: (A) four chamber, end
diastole; (B) four chamber, end systole; (C) mid-ventricular short axis, end diastole; (D) mid-ventricular short
axis, endsystole.

A2ch
A4ch
L2ch
L4ch

A B
FIG. 31.2  Left atrial volume measurement using the biplane area-length method. The endocardial border is
traced in two-chamber (2Ch; A) and four-chamber views (4Ch; B) in end systole, excluding pulmonary veins
and left atrial appendage. The left atria length is measured from the midpoint of the mitral valve, perpendicu-
larly, to the top of the atrium. (From Gulati A, Ismail TF, Jabbour A, et al. Clinical utility and prognostic value
of left atrial volume assessment by cardiovascular magnetic resonance in non-ischaemic dilated cardiomy-
opathy. Eur J Heart Fail. 2013;15:660–670, with permission.)

discuss the potential role of the more recently developed T1 mapping fibrosis describes discrete areas of myocardial scarring resulting from
technique, the use of which is currently confined to research. myocyte cell death.68 Fibrosis is promoted through activation of the
renin–angiotensin–aldosterone and the beta-adrenergic axes, both con-
Fibrosis sequences of heart failure. Injurious stimuli and toxins also play an
One of the histologic hallmarks of DCM, common to all causes of heart important role by activating inflammatory cascades, leading to the
failure, is myocardial fibrosis.68–70 There are two main forms of fibrosis production of reactive oxygen species.71 These pathways result in activa-
detected histologically: interstitial fibrosis and replacement fibrosis (Fig. tion of myofibroblasts, with upregulation of transforming growth factor
31.3). Interstitial fibrosis describes an increase in the collagen volume beta, altered activity in matrix metalloproteinases, and, ultimately, the
fraction with expansion of the extracellular matrix, whereas replacement production of collagen.71,72
386 SECTION III  Functional Disease

Area of
detail

A B C

Area of
detail

D E F
FIG. 31.3  (A) Late gadolinium enhancement cardiovascular magnetic resonance (LGE CMR), demonstrating
extensive midwall enhancement (arrowhead). (B) Picrosirius red staining in the corresponding postmortem
macroscopic specimen, demonstrating bands of collagen (arrowheads) in the same distribution as the LGE.
(C) Replacement fibrosis (arrowheads) on microscopic examination. (D) LGE CMR, demonstrating absence
of LGE. (E) Picrosirius staining of the corresponding macroscopic postmortem specimen without staining
for collagen. (F) Small amounts of perivascular interstitial fibrosis (arrowhead) without replacement fibrosis.
(From Gulati A, Jabbour A, Ismail TF, et al. Association of fibrosis with mortality and sudden cardiac death
in patients with nonischemic dilated cardiomyopathy. JAMA. 2013;309:896–908, with permission.)

Fibrosis is associated with adverse ventricular remodeling and Late Gadolinium Enhancement Cardiovascular Magnetic
increased all-cause mortality.71 It has also been shown to provide Resonance and Midwall Replacement Fibrosis
an important substrate for the maintenance of re-entry ventricular LGE CMR identifies midwall enhancement in around 30% of patients
arrhythmias.73–77 An electroanatomical mapping study demonstrated that with DCM, typically occurring in a linear distribution in the septum
inducible ventricular arrhythmia and a history of sustained ventricular (see Fig. 31.3).7,83,84 Histologic validation has confirmed that this pattern
tachycardia (VT) only occurred in those patients with replacement of enhancement represents replacement fibrosis.7,84 The mass of fibrosis
fibrosis on LGE CMR.78 Additionally, in each of the cases with inducible can be quantified using semiautomated software and validated methods.
arrhythmia, its origin was mapped to the location of the LGE on CMR.78 The most commonly used methods are the full width at half maximum
Fibrosis has also been associated with fractionated electrograms, slowed (FWHM) and the >2 standard deviation (>2 SD) approach. 85 The
conduction, and conduction block during electrophysiology studies.76,79 FWHM method quantifies regions of myocardium with a signal intensity
Each of these abnormalities has been associated with an increased risk >50% of the maximally enhanced region, whereas the >2 SD approach
of sustained VT and ventricular fibrillation.76,79 The combination of includes regions with a signal intensity >2 SD above the signal intensity
fibrosis interspersed with areas of surviving myocardium has been cor- of a reference area of normal myocardium.
related with the generation of re-entry wavefronts.73,76,80 Moreover, the Given the suboptimal sensitivity and specificity of current LVEF-
targeting of isthmuses of surviving myocardium has been shown to based criteria for sudden death risk stratification, there has been interest
eliminate ventricular arrhythmia.81,82 The combination of fibrosis and in the incremental value of adding the presence and extent of mid-wall
areas of viable myocardium has therefore been proposed to act as a fibrosis, as determined by LGE CMR, to the model. The largest study
substrate for re-entry arrhythmia.73,76,80 All of these observations strongly to date reported all-cause mortality and SCD/aborted SCD rates of
support a role for fibrosis in the generation of ventricular arrhythmia. 27% and 30%, respectively, in patients with midwall fibrosis, compared
Noninvasive detection of fibrosis using CMR therefore provides an with 11% and 7% in patients without fibrosis.7 Multivariable analysis
opportunity to identify patients with DCM at high risk of sudden confirmed that the presence of midwall fibrosis was an independent
arrhythmic death. Treatments targeted at preventing or reversing fibrosis predictor of all-cause mortality and SCD/aborted SCD. The adjusted
also provide important future therapeutic possibilities. hazard ratio (HR) for the SCD composite endpoint was 4.6 in those
CHAPTER 31  Role of Cardiovascular Magnetic Resonance in Dilated Cardiomyopathy 387

patients with fibrosis (P < .001), whereas the adjusted HR for every 1% preadministration and postadministration of gadolinium contrast.
increment in the mass of fibrosis was 1.1 (95% confidence interval [CI] Using precontrast and postcontrast T1 values and hematocrit mea-
1.05–1.16; P < .001). The same study demonstrated that the addition surement, the extracellular volume (ECV) fraction can subsequently
of mid-wall fibrosis to an LVEF-based strategy improved overall risk be calculated.96 The extent of interstitial fibrosis has been shown to
stratification. This finding has been replicated in similar studies and a correlate well with ECV fraction and native T1 times.98 This has led
large meta-analysis.84–91 It has also been demonstrated that LGE quan- to the possibility of quantifying the degree of interstitial fibrosis in
tification by both the FWHM and 2 SD methods provides robust prog- DCM using a noninvasive technique. It has potential advances over
nostication for the prediction of adverse events.85 It has been demonstrated the qualitative LGE technique, which relies on subjective interpretation.
that an LGE extent of >6.1% by 2 SD and >4.4% by FWHM predicts Moreover, given the fact that the LGE technique relies on regional dif-
adverse events with the greatest sensitivity and specificity. ferences, there is the potential to miss diffuse myocardial fibrosis, which
Altogether, current evidence strongly supports a role for LGE CMR in can appear as “nulled” normal myocardium. T1 mapping also offers
improving the selection of patients who may benefit from ICD therapy, by opportunities for monitoring the response to therapies with potential
identifying those patients at highest risk of adverse arrhythmic outcomes. antifibrotic properties. Studies have reported elevated native T1 values
LGE CMR appears to add robust incremental value to the current LVEF- and ECV fractions in DCM, compared with controls.98–100 Elevation of
based approach. The added value of LGE mass quantification appears ECV in even mild phenotypes of DCM, compared with controls, has
less convincing, possibly because of interobserver and intraobserver been demonstrated.99,101 This raises the possibility of using T1 mapping
variability. Another explanation may be that, rather than the overall for the diagnosis of borderline cases.
mass of fibrosis, it is the size of the border between areas of fibrosis A large study on patients with DCM has demonstrated that native
and surviving myocardium that is most relevant to overall arrhythmic T1 values independently predicted all-cause mortality and a composite
risk. As discussed, isthmuses of surviving myocardium within fibrotic of adverse heart failure outcomes.102 However, despite the association
areas appear to play an important role in the generation of re-entry between outcome and native T1 values, it did not demonstrate an inde-
wavefronts.73,76,80–82 Therefore the size of the area where these two com- pendent association between ECV and outcomes. This may be explained
ponents interact may reflect arrhythmic risk more accurately. by heterogeneities in the methods of ECV calculation, including the
timing at which postcontrast images were taken and also the timing of
Confirming the Diagnosis hematocrit measurement relative to the study.103 This demonstrates the
The use of LGE CMR is also key to confirming the diagnosis of DCM need for standardized precise protocols for T1 mapping for future
in suspected cases. A study investigating the use of LGE CMR in patients research and clinical practice.
with a suspected diagnosis of DCM, based on angiographic and echo-
cardiographic findings, demonstrated that, in fact, 13% of patients had Perfusion Imaging
subendocardial enhancement indicating previous myocardial infarction.92 Studies have demonstrated a reduction in myocardial blood flow in
This demonstrates that coronary angiography misdiagnoses a propor- patients with DCM.104 Many theories have been proposed to explain
tion of patients as having a nonischemic etiology when in fact they this phenomenon despite visually normal epicardial coronary arteries.
have suffered previous asymptomatic infarction. It is recognized that These mainly focus on dysfunction of the microvessels or increased
around 25% of patients with myocardial infarction do not present to oxygen diffusion distance because of interstitial fibrosis.105,106 The degree
hospital. A proportion of these patients have unobstructed coronary of myocardial blood flow impairment has been strongly correlated with
vasculature on angiography, presumably with infarction secondary to prognosis.104,107 However, what has been less clear is whether abnormal
embolic phenomenon or plaque rupture with subsequent re-canalization perfusion is responsible for the adverse prognosis because of myocardial
(see Fig. 31.4E). This leads to the subsequent incorrect diagnosis of a oxygen deprivation or is merely a marker of disease severity. A study
nonischemic etiology. The correct diagnosis of this subgroup of patients, using blood oxygen level-dependent CMR, however, has improved our
mislabeled by coronary angiography, is crucial given the different clinical current understanding of the pathophysiologic relevance.108 This study
courses and management strategies with regards to ischemic and dilated assessed myocardial oxygenation and myocardial perfusion reserve
cardiomyopathies. Subsequent to this, LGE CMR has been shown to during adenosine stress in patients with DCM compared with controls.
be at least as sensitive and specific as invasive angiography in diagnosing Although myocardial perfusion reserve was reduced in patients with
the cause of LV impairment in patients presenting with heart failure.93 DCM, compared with controls, oxygenation during stress was not sig-
This demonstrated a 97% diagnostic accuracy for LGE CMR compared nificantly different. Moreover, resting cardiac energetics in patients with
with a 95% diagnostic accuracy for coronary angiography. This supports DCM were not affected by oxygen administration. This suggests that
the role of LGE CMR as the initial diagnostic test in patients presenting the perfusion deficit is not sufficient to cause a reduction in oxygenation
with heart failure and LV systolic impairment of uncertain etiology. during stress and also that the resting impairment in energy metabolism
Fig. 31.4 correlates the findings of LGE CMR and coronary angiography is not secondary to a myocardial oxygen deficit. This also indicates that
from the study, demonstrating the different possible combinations of the reduction in perfusion observed in DCM is merely a marker of
findings from CMR and angiography and the subsequent diagnoses. disease severity rather than a significant pathophysiologic driver.
LGE and T2* sequences also allow the diagnosis of other pathologies,
such as sarcoidosis and myocardial iron overload, which can occasion- Metabolic Imaging
ally present with a DCM phenotype, requiring different management Investigation of myocardial energetics in DCM is largely confined to
strategies, and which are not easily diagnosed using other imaging research. Magnetic resonance (MR) spectroscopy allows the level of
modalities.94,95 Sarcoidosis is discussed in further detail later. myocardial adenosine triphosphate (ATP) and phosphocreatine (PCr)
to be determined.109 Both compounds are involved in high-energy
RESEARCH TECHNIQUES metabolism and a reduced ratio of PCr:ATP indicates a myocardial
energy deficit. The use of MR spectroscopy therefore enables myocardial
T1 Mapping and Interstitial Fibrosis energetics to be assessed. In patients with DCM, the PCr:ATP ratio has
Recent advances in T1 mapping techniques now allow the direct quan- been shown to strongly correlate with outcomes and therapies such as
tification of T1 relaxation times of each myocardial voxel, enabling angiotensin-converting enzyme inhibitors have been linked with an
the construction of T1 maps.96,97 T1 mapping can be performed improvement in the ratio.109 Studies on exercise training have also
388 SECTION III  Functional Disease

True DCM

True CAD

DCM with
bystander
infarct

DCM with
bystander
CAD

Ischemic HF
with
unobstructed
coronary
arteries
E

Ischemic HF
without
infarction

F
FIG. 31.4  Late gadolinium enhancement cardiovascular magnetic resonance (LGE CMR) and correspond-
ing coronary angiography in patients presenting with heart failure (HF) and left ventricular dysfunction of
unknown cause. (A) True dilated cardiomyopathy (DCM) with unobstructed epicardial arteries and no LGE
on CMR. (B) True coronary artery disease (CAD) with a subendocardial inferolateral LGE, consistent with a
circumflex territory infarct and a tight proximal circumflex stenosis (arrows). (C) DCM with a subendocardial
LGE (arrow) representing a bystander infarct and unobstructed epicardial arteries. (D) DCM without sub-
endocardial LGE and bystander coincidental distal coronary disease (arrow). (E) Ischemic cardiomyopathy
with large apical infarct (arrow) on LGE and unobstructed coronary arteries. (F) Ischemic cardiomyopathy
with severe proximal CAD (arrows) but no infarction on LGE CMR. (From Assomull RG, Shakespeare C,
Kalra PR, Lloyd G, Gulati A, Strange J, et al. Role of cardiovascular magnetic resonance as a gatekeeper
to invasive coronary angiography in patients presenting with heart failure of unknown etiology. Circulation.
2011;124:1351–1360, with permission.)

confirmed that although training improves LV function, there is no and noncompact layers of myocardium (Fig. 31.5).112 It can occur as a
adverse effect on energetics with no change in PCr:ATP ratio.110,111 primary myocardial abnormality and also in the context of congenital
heart defects such as Ebstein anomaly and hypoplastic left heart syn-
LEFT VENTRICULAR NONCOMPACTION drome. Genetic abnormalities are thought to account for 30% to 50%
of cases, with sarcomeric mutations being the most common.113 Clinical
CARDIOMYOPATHY events include arrhythmia, ischemia, heart failure, and sudden death.
Left ventricular noncompaction cardiomyopathy (LVNC) is characterized The true prevalence of the disease is controversial but has been
by prominent trabeculations, deep intratrabecular recesses, and compact estimated at between 0.05% and 0.26%.114,115 Diagnostic criteria have
CHAPTER 31  Role of Cardiovascular Magnetic Resonance in Dilated Cardiomyopathy 389

FIG. 31.5  Cardiovascular magnetic resonance (CMR) in a patient with ventricular noncompaction. Steady-
state free precession (SSFP) images in a two-chamber view (top left panel), three-chamber view (bottom
left panel), four-chamber view (top right panel), and mid short axis (bottom right panel). All mid and apical
segments, except the septum, reveal a significant amount of noncompacted wall with a thin compact wall
(arrows). The left ventricular volume is increased, and its systolic function is substantially impaired.

been proposed on the basis of the ratio of noncompact myocardium Tissue Characterization
to compact myocardium. The most commonly used CMR criteria uses Similar to DCM, LGE is a common finding in LVNC, being found in
a ratio of greater than 2.3 : 1 in end diastole.116 Prominent trabecular up to 55% of patients.121 The most common pattern is septal midwall
patterns, meeting diagnostic criteria for LVNC, have, however, been enhancement followed by insertion point (20%), subendocardial (13%),
demonstrated in healthy populations exposed to hemodynamic loads and transmural (10%).121 In one study there was no relationship between
such as pregnant women and athletes.117,118 This demonstrates the dif- the pattern of noncompaction and the distribution of LGE. Although
ficulties in differentiating LVNC from normal variants. LGE corresponds to reductions in LVEF, there are no data correlating
LGE with adverse clinical outcomes yet.
Morphology and Function
A spectrum of LVNC phenotypes exist with varying morphologies.112
A dilated phenotype similar to DCM is the most common, character-
CARDIAC SARCOIDOSIS
ized by cavity dilatation, eccentric hypertrophy, and systolic dysfunction. The incidence of myocardial involvement in systemic sarcoid varies
Benign phenotypes with normal cavity size, mass, and systolic function with geography, with estimations of ~25% in the United States and
and, less commonly, hypertrophic phenotypes characterized by wall >50% in Japan.122–124 It accounts for between 13% and 25% of deaths
thickening, increased mass, and varying levels of systolic and diastolic because of the disease, primarily caused by sudden death and congestive
dysfunction are also recognized. heart failure.122–126 The histologic hallmark of the disease is the presence
Steady-state free precession (SSFP) cine imaging allows the easy of noncaseating granuloma which can be patchy and, consequently,
recognition of the morphologic abnormalities together with accurate missed with endomyocardial biopsy. Accurate diagnosis therefore requires
calculation of volumes, mass, function, and compact to noncompact a multimodality approach, including CMR.
myocardium ratios. A perpendicular orientation of the imaging plane to
the wall should be carefully ensured because tangential orientation may Morphology and Function
lead to overestimation of the relative extent of the noncompacted wall. Cardiac sarcoid can mimic a variety of phenotypes, most commonly
To visualize the trabecular meshwork without having partial volume DCM and less commonly hypertrophic and ischemic cardiomyopathies.
effects induced by intratrabecular blood, a high temporal and spatial It frequently produces LV dilatation with regional and global hypokinesia
resolution should be used. Although some trabeculation may be a normal and wall thinning. Granulomatous inflammation may also lead to an
variant, the recognition of additional abnormalities such as an abnor- increase in wall thickness, mimicking LV hypertrophy. SSFP sequences
mally thin compact myocardial layer with abnormal systolic motion allow the accurate and reproducible assessment of ventricular morphol-
is important. The calculation of trabeculated LV mass may also prove ogy and function.
a useful diagnostic technique.119 One study has demonstrated that a
trabeculated mass of >20% has a 94% sensitivity and specificity for Tissue Characterization
the diagnosis of LVNC. Studies have demonstrated the superiority of Together with accurate assessment of morphology and function, CMR
CMR imaging over two-dimensional echocardiography in the assess- allows the detection of underlying acute and chronic disease. Acute
ment of LVNC.120 CMR was shown to provide more complete evalua- inflammation and active disease can be identified using T2-weighted
tion of each myocardial segment and identified noncompacted layers triple inversion recovery spin echo sequences and early gadolinium-
more readily. enhanced images.127 LGE allows the assessment of scar and fibrosis and
390 SECTION III  Functional Disease

A B
FIG. 31.6  A case of biopsy-proven cardiac sarcoid with dense, almost transmural subepicardial late gadolinium
enhancement in a shepherd’s crook distribution (A) without corresponding signal intensity change of short
inversion time inversion recovery imaging (B) indicative of chronic disease.

in combination with T2-weighted imaging can differentiate active from In summary, CMR imaging allows rapid, high-resolution, noninvasive
chronic disease.94 Comparison with positron emission tomography– and reproducible assessment of the underlying pathologic changes and
computed tomography (PET–CT) demonstrates similar sensitivities functional consequences associated with cardiac sarcoid and is therefore
for both modalities but a higher specificity with CMR.94,128,129 considered the gold standard technique.
LGE occurs in a wide variety of patterns, including midwall, sub-
epicardial, subendocardial, and transmural (Fig. 31.6).94,125,130 A shepherd’s
crook pattern has been recognized with enhancement extending from
CONCLUSION
the anterior RV-free wall round to the anterior RV insertion point and In conclusion, the ability of CMR to provide accurate and robust assess-
the LV septum (see Fig. 31.6). A coronary distribution of enhancement ment of morphology and function, together with tissue characterization,
has been demonstrated in 48% of cases despite the absence of coronary in a single noninvasive test offers many advantages in the assessment
disease on angiography and demonstrates the ability of sarcoid to mas- of DCM. It enables accurate diagnosis, with exclusion of other ischemic
querade as other disease processes.94,125,130 The presence of LGE has been and nonischemic processes and provides the clinician with the ability
strongly correlated with adverse prognosis with a 30-fold increased risk to stratify risk and prognosis more accurately through the accurate
of death and aborted SCD.130,131 Studies have suggested that LGE imaging assessment of chamber size and function, and the detection and quan-
may also be used to assess the response to steroid therapy and also as tification of myocardial fibrosis.
a guide for endomyocardial biopsy.132 T1-mapping offers potential
advantages in the future for detecting diffuse processes missed by current
techniques, which rely on the comparison of signal intensity with a
REFERENCES
reference area of normal myocardium. A full reference list is available online at ExpertConsult.com
CHAPTER 31  Role of Cardiovascular Magnetic Resonance in Dilated Cardiomyopathy 390.e1

REFERENCES 21. Hegele R. LMNA mutation position predicts organ system involvement
in laminopathies. Clin Genet. 2005;68(1):31–34.
1. Elliott P, Andersson B, Arbustini E, et al. Classification of the 22. Pasotti M, Klersy C, Pilotto A, et al. Long-term outcome and risk
cardiomyopathies: a position statement from the European Society Of stratification in dilated cardiolaminopathies. J Am Coll Cardiol.
Cardiology Working Group on Myocardial and Pericardial Diseases. Eur 2008;52(15):1250–1260.
Heart J. 2008;29(2):270–276. 23. van Berlo JH, de Voogt WG, van der Kooi AJ, et al. Meta-analysis of
2. Pinto YM, Elliott PM, Arbustini E, et al. Proposal for a revised definition clinical characteristics of 299 carriers of LMNA gene mutations: do
of dilated cardiomyopathy, hypokinetic non-dilated cardiomyopathy, lamin A/C mutations portend a high risk of sudden death? J Mol Med.
and its implications for clinical practice: a position statement of the ESC 2005;83(1):79–83.
working group on myocardial and pericardial diseases. Eur Heart J. 24. George A, Figueredo VM. Alcoholic cardiomyopathy: a review. J Card
2016. Fail. 2011;17(10):844–849.
3. Hershberger RE, Hedges DJ, Morales A. Dilated cardiomyopathy: the 25. Caforio AL, Pankuweit S, Arbustini E, et al. Current state of knowledge
complexity of a diverse genetic architecture. Nat Rev Cardiol. on aetiology, diagnosis, management, and therapy of myocarditis: a
2013;10(9):531–547. position statement of the European Society of Cardiology Working
4. Codd MB, Sugrue DD, Gersh BJ, Melton LJ 3rd. Epidemiology of Group on Myocardial and Pericardial Diseases. Eur Heart J.
idiopathic dilated and hypertrophic cardiomyopathy. A population- 2013;34(33):2636–2648, 48a–48d.
based study in Olmsted County, Minnesota, 1975-1984. Circulation. 26. Hilfiker-Kleiner D, Kaminski K, Podewski E, et al. A cathepsin D-cleaved
1989;80(3):564–572. 16 kDa form of prolactin mediates postpartum cardiomyopathy. Cell.
5. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE. 2007;128(3):589–600.
Prevalence of hypertrophic cardiomyopathy in a general population of 27. Hilfiker-Kleiner D, Sliwa K. Pathophysiology and epidemiology of
young adults. Echocardiographic analysis of 4111 subjects in the peripartum cardiomyopathy. Nat Rev Cardiol. 2014;11(6):364–370.
CARDIA Study. Coronary Artery Risk Development in (Young) Adults. 28. Sliwa K, Blauwet L, Tibazarwa K, et al. Evaluation of bromocriptine in
Circulation. 1995;92(4):785–789. the treatment of acute severe peripartum cardiomyopathy: a proof-of-
6. Felker GM, Thompson RE, Hare JM, et al. Underlying causes and concept pilot study. Circulation. 2010;121(13):1465–1473.
long-term survival in patients with initially unexplained 29. Ware JS, Li J, Mazaika E, et al. Shared genetic predisposition in
cardiomyopathy. N Engl J Med. 2000;342(15):1077–1084. peripartum and dilated cardiomyopathies. N Engl J Med.
7. Gulati A, Jabbour A, Ismail TF, et al. Association of fibrosis with 2016;374(3):233–241.
mortality and sudden cardiac death in patients with nonischemic dilated 30. van Spaendonck-Zwarts KY, Posafalvi A, van den Berg MP, et al. Titin
cardiomyopathy. JAMA. 2013;309(9):896–908. gene mutations are common in families with both peripartum
8. Bender JR. Idiopathic dilated cardiomyopathy. An immunologic, genetic, cardiomyopathy and dilated cardiomyopathy. Eur Heart J.
or infectious disease, or all of the above? Circulation. 1991;83(2): 2014;35(32):2165–2173.
704–706. 31. van Spaendonck-Zwarts KY, van Tintelen JP, van Veldhuisen DJ, et al.
9. Kasper EK, Agema WR, Hutchins GM, Deckers JW, Hare JM, Baughman Peripartum cardiomyopathy as a part of familial dilated cardiomyopathy.
KL. The causes of dilated cardiomyopathy: a clinicopathologic review of Circulation. 2010;121(20):2169–2175.
673 consecutive patients. J Am Coll Cardiol. 1994;23(3):586–590. 32. Priori SG, Blomstrom-Lundqvist C, Mazzanti A, et al. 2015 ESC
10. Grunig E, Tasman JA, Kucherer H, Franz W, Kubler W, Katus HA. Guidelines for the management of patients with ventricular arrhythmias
Frequency and phenotypes of familial dilated cardiomyopathy. J Am Coll and the prevention of sudden cardiac death. The Task Force for the
Cardiol. 1998;31(1):186–194. Management of Patients with Ventricular Arrhythmias and the
11. Michels VV, Moll PP, Miller FA, et al. The frequency of familial dilated Prevention of Sudden Cardiac Death of the European Society of
cardiomyopathy in a series of patients with idiopathic dilated Cardiology (ESC). Endorsed by: Association for European Paediatric
cardiomyopathy. N Engl J Med. 1992;326(2):77–82. and Congenital Cardiology (AEPC). Europace. 2015;17(11):1601–
12. van Spaendonck-Zwarts KY, van Rijsingen IA, van den Berg MP, et al. 1687.
Genetic analysis in 418 index patients with idiopathic dilated 33. McMurray JJ, Adamopoulos S, Anker SD, et al. ESC Guidelines for the
cardiomyopathy: overview of 10 years’ experience. Eur J Heart Fail. diagnosis and treatment of acute and chronic heart failure 2012: The
2013;15(6):628–636. Task Force for the Diagnosis and Treatment of Acute and Chronic Heart
13. George AL Jr. Molecular and genetic basis of sudden cardiac death. Failure 2012 of the European Society of Cardiology. Developed in
J Clin Invest. 2013;123(1):75–83. collaboration with the Heart Failure Association (HFA) of the ESC. Eur
14. Millat G, Bouvagnet P, Chevalier P, et al. Clinical and mutational Heart J. 2012;33(14):1787–1847.
spectrum in a cohort of 105 unrelated patients with dilated 34. Russo AM, Stainback RF, Bailey SR, et al. ACCF/HRS/AHA/ASE/HFSA/
cardiomyopathy. Eur J Med Genet. 2011;54(6):e570–e575. SCAI/SCCT/SCMR 2013 appropriate use criteria for implantable
15. Limongelli G, D’Alessandro R, Maddaloni V, Rea A, Sarkozy A, McKenna cardioverter-defibrillators and cardiac resynchronization therapy: a
WJ. Skeletal muscle involvement in cardiomyopathies. J Cardiovasc Med. report of the American College of Cardiology Foundation appropriate
2013;14(12):837–861. use criteria task force, Heart Rhythm Society, American Heart
16. Herman DS, Lam L, Taylor MR, et al. Truncations of titin causing Association, American Society of Echocardiography, Heart Failure
dilated cardiomyopathy. N Engl J Med. 2012;366(7):619–628. Society of America, Society for Cardiovascular Angiography and
17. Horowits R, Kempner ES, Bisher ME, Podolsky RJ. A physiological role Interventions, Society of Cardiovascular Computed Tomography, and
for titin and nebulin in skeletal muscle. Nature. Society for Cardiovascular Magnetic Resonance. J Am Coll Cardiol.
1986;323(6084):160–164. 2013;61(12):1318–1368.
18. Liversage AD, Holmes D, Knight PJ, Tskhovrebova L, Trinick J. Titin and 35. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for
the sarcomere symmetry paradox. J Mol Biol. 2001;305(3):401–409. the management of heart failure: a report of the American College of
19. Muhle-Goll C, Habeck M, Cazorla O, Nilges M, Labeit S, Granzier H. Cardiology Foundation/American Heart Association Task Force on
Structural and functional studies of titin’s fn3 modules reveal conserved practice guidelines. Circulation. 2013;128(16):e240–e327.
surface patterns and binding to myosin S1–a possible role in the 36. Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable
Frank-Starling mechanism of the heart. J Mol Biol. 2001;313(2): cardioverter-defibrillator for congestive heart failure. N Engl J Med.
431–447. 2005;352(3):225–237.
20. Roberts AM, Ware JS, Herman DS, et al. Integrated allelic, 37. Kadish A, Dyer A, Daubert JP, et al. Prophylactic defibrillator
transcriptional, and phenomic dissection of the cardiac effects of titin implantation in patients with nonischemic dilated cardiomyopathy. N
truncations in health and disease. Sci Transl Med. 2015;7(270):270ra6. Engl J Med. 2004;350(21):2151–2158.
390.e2 SECTION III  Functional Disease

38. Bansch D, Antz M, Boczor S, et al. Primary prevention of sudden 54. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular
cardiac death in idiopathic dilated cardiomyopathy: the Cardiomyopathy systolic and diastolic function normalized to age, gender and body
Trial (CAT). Circulation. 2002;105(12):1453–1458. surface area from steady-state free precession cardiovascular magnetic
39. Strickberger SA, Hummel JD, Bartlett TG, et al. Amiodarone versus resonance. Eur Heart J. 2006;27(23):2879–2888.
implantable cardioverter-defibrillator: randomized trial in patients with 55. Lang RM, Bierig M, Devereux RB, et al. Recommendations for chamber
nonischemic dilated cardiomyopathy and asymptomatic nonsustained quantification: a report from the American Society of Echocardiography’s
ventricular tachycardia–AMIOVIRT. J Am Coll Cardiol. 2003;41(10): Guidelines and Standards Committee and the Chamber Quantification
1707–1712. Writing Group, developed in conjunction with the European Association
40. Gorgels AP, Gijsbers C, de Vreede-Swagemakers J, Lousberg A, Wellens of Echocardiography, a branch of the European Society of Cardiology.
HJ. Out-of-hospital cardiac arrest–the relevance of heart failure. The J Am Soc Echocardiogr. 2005;18(12):1440–1463.
Maastricht Circulatory Arrest Registry. Eur Heart J. 2003;24(13): 56. Anderson JL, Horne BD, Pennell DJ. Atrial dimensions in health and left
1204–1209. ventricular disease using cardiovascular magnetic resonance.
41. Stecker EC, Vickers C, Waltz J, et al. Population-based analysis of sudden J Cardiovasc Magn Reson. 2005;7(4):671–675.
cardiac death with and without left ventricular systolic dysfunction: 57. Whitlock M, Garg A, Gelow J, Jacobson T, Broberg C. Comparison of
two-year findings from the Oregon Sudden Unexpected Death Study. left and right atrial volume by echocardiography versus cardiac
J Am Coll Cardiol. 2006;47(6):1161–1166. magnetic resonance imaging using the area-length method. Am J
42. Fishman GI, Chugh SS, Dimarco JP, et al. Sudden cardiac death Cardiol. 2010;106(9):1345–1350.
prediction and prevention: report from a National Heart, Lung, and 58. Pritchett AM, Mahoney DW, Jacobsen SJ, Rodeheffer RJ, Karon BL,
Blood Institute and Heart Rhythm Society Workshop. Circulation. Redfield MM. Diastolic dysfunction and left atrial volume: a population-
2010;122(22):2335–2348. based study. J Am Coll Cardiol. 2005;45(1):87–92.
43. Goldberger JJ, Cain ME, Hohnloser SH, et al. American Heart Association/ 59. Therkelsen SK, Groenning BA, Svendsen JH, Jensen GB. Atrial and
American College of Cardiology Foundation/Heart Rhythm Society ventricular volume and function in persistent and permanent atrial
scientific statement on noninvasive risk stratification techniques for fibrillation, a magnetic resonance imaging study. J Cardiovasc Magn
identifying patients at risk for sudden cardiac death: a scientific statement Reson. 2005;7(2):465–473.
from the American Heart Association Council on Clinical Cardiology 60. Gulati A, Ismail TF, Jabbour A, et al. Clinical utility and prognostic value
Committee on Electrocardiography and Arrhythmias and Council on of left atrial volume assessment by cardiovascular magnetic resonance in
Epidemiology and Prevention. Heart Rhythm. 2008;5(10):e1–e21. non-ischaemic dilated cardiomyopathy. Eur J Heart Fail. 2013;15(6):
44. Zipes DP, Camm AJ, Borggrefe M, et al. ACC/AHA/ESC 2006 Guidelines 660–670.
for Management of Patients With Ventricular Arrhythmias and the 61. Chan KM, Wage R, Symmonds K, et al. Towards comprehensive
Prevention of Sudden Cardiac Death: a report of the American College assessment of mitral regurgitation using cardiovascular magnetic
of Cardiology/American Heart Association Task Force and the European resonance. J Cardiovasc Magn Reson. 2008;10:61.
Society of Cardiology Committee for Practice Guidelines (writing 62. Kizilbash AM, Hundley WG, Willett DL, Franco F, Peshock RM,
committee to develop Guidelines for Management of Patients With Grayburn PA. Comparison of quantitative Doppler with magnetic
Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death): resonance imaging for assessment of the severity of mitral regurgitation.
developed in collaboration with the European Heart Rhythm Am J Cardiol. 1998;81(6):792–795.
Association and the Heart Rhythm Society. Circulation. 2006;114(10): 63. Kon MW, Myerson SG, Moat NE, Pennell DJ. Quantification of
e385–e484. regurgitant fraction in mitral regurgitation by cardiovascular magnetic
45. Buser PT, Auffermann W, Holt WW, et al. Noninvasive evaluation of resonance: comparison of techniques. J Heart Valve Dis. 2004;
global left ventricular function with use of cine nuclear magnetic 13(4):600–607.
resonance. J Am Coll Cardiol. 1989;13(6):1294–1300. 64. Stolfo D, Merlo M, Pinamonti B, et al. Early improvement of functional
46. Maceira AM, Prasad SK, Khan M, Pennell DJ. Normalized left mitral regurgitation in patients with idiopathic dilated cardiomyopathy.
ventricular systolic and diastolic function by steady state free precession Am J Cardiol. 2015;115(8):1137–1143.
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 65. Bocchi EA, Bellotti G, Moreira LF, et al. Prognostic indicators of
2006;8(3):417–426. one-year outcome after cardiomyoplasty for idiopathic dilated
47. Buss SJ, Breuninger K, Lehrke S, et al. Assessment of myocardial cardiomyopathy. Am J Cardiol. 1994;73(8):604–608.
deformation with cardiac magnetic resonance strain imaging improves 66. Doherty NE 3rd, Seelos KC, Suzuki J, et al. Application of cine nuclear
risk stratification in patients with dilated cardiomyopathy. Eur Heart J magnetic resonance imaging for sequential evaluation of response to
Cardiovasc Imaging. 2015;16(3):307–315. angiotensin-converting enzyme inhibitor therapy in dilated
48. Shehata ML, Cheng S, Osman NF, Bluemke DA, Lima JA. Myocardial cardiomyopathy. J Am Coll Cardiol. 1992;19(6):1294–1302.
tissue tagging with cardiovascular magnetic resonance. J Cardiovasc 67. Bellenger NG, Davies LC, Francis JM, Coats AJ, Pennell DJ. Reduction in
Magn Reson. 2009;11:55. sample size for studies of remodeling in heart failure by the use of
49. MacGowan GA, Shapiro EP, Azhari H, et al. Noninvasive measurement cardiovascular magnetic resonance. J Cardiovasc Magn Reson.
of shortening in the fiber and cross-fiber directions in the normal 2000;2(4):271–278.
human left ventricle and in idiopathic dilated cardiomyopathy. 68. Beltrami CA, Finato N, Rocco M, et al. The cellular basis of dilated
Circulation. 1997;96(2):535–541. cardiomyopathy in humans. J Mol Cell Cardiol. 1995;27(1):291–305.
50. Delepine S, Furber AP, Beygui F, et al. 3-D MRI assessment of regional 69. Marijianowski MM, Teeling P, Mann J, Becker AE. Dilated
left ventricular systolic wall stress in patients with reperfused MI. Am J cardiomyopathy is associated with an increase in the type I/type III
Physiol Heart Circ Physiol. 2003;284(4):H1190–H1197. collagen ratio: a quantitative assessment. J Am Coll Cardiol.
51. Gulati A, Ismail TF, Jabbour A, et al. The prevalence and prognostic 1995;25(6):1263–1272.
significance of right ventricular systolic dysfunction in nonischemic 70. Gunja-Smith Z, Morales AR, Romanelli R, Woessner JF Jr. Remodeling
dilated cardiomyopathy. Circulation. 2013;128(15):1623–1633. of human myocardial collagen in idiopathic dilated cardiomyopathy.
52. La Vecchia L, Paccanaro M, Bonanno C, Varotto L, Ometto R, Vincenzi Role of metalloproteinases and pyridinoline cross-links. Am J Pathol.
M. Left ventricular versus biventricular dysfunction in idiopathic dilated 1996;148(5):1639–1648.
cardiomyopathy. Am J Cardiol. 1999;83(1):120–122, A9. 71. Mewton N, Liu CY, Croisille P, Bluemke D, Lima JA. Assessment of
53. Globits S, Pacher R, Frank H, et al. Comparative assessment of right myocardial fibrosis with cardiovascular magnetic resonance. J Am Coll
ventricular volumes and ejection fraction by thermodilution and Cardiol. 2011;57(8):891–903.
magnetic resonance imaging in dilated cardiomyopathy. Cardiology. 72. Souders CA, Bowers SL, Baudino TA. Cardiac fibroblast: the renaissance
1995;86(1):67–72. cell. Circ Res. 2009;105(12):1164–1176.
CHAPTER 31  Role of Cardiovascular Magnetic Resonance in Dilated Cardiomyopathy 390.e3

73. de Bakker JM, Coronel R, Tasseron S, et al. Ventricular tachycardia in systematic review and meta-analysis. Circ Cardiovasc Imaging.
the infarcted, Langendorff-perfused human heart: role of the 2014;7(2):250–258.
arrangement of surviving cardiac fibers. J Am Coll Cardiol. 92. McCrohon JA, Moon JC, Prasad SK, et al. Differentiation of heart failure
1990;15(7):1594–1607. related to dilated cardiomyopathy and coronary artery disease using
74. Spach MS, Boineau JP. Microfibrosis produces electrical load variations gadolinium-enhanced cardiovascular magnetic resonance. Circulation.
due to loss of side-to-side cell connections: a major mechanism of 2003;108(1):54–59.
structural heart disease arrhythmias. Pacing Clin Electrophysiol. 93. Assomull RG, Shakespeare C, Kalra PR, et al. Role of cardiovascular
1997;20(2 Pt 2):397–413. magnetic resonance as a gatekeeper to invasive coronary angiography in
75. Pogwizd SM, McKenzie JP, Cain ME. Mechanisms underlying patients presenting with heart failure of unknown etiology. Circulation.
spontaneous and induced ventricular arrhythmias in patients with 2011;124(12):1351–1360.
idiopathic dilated cardiomyopathy. Circulation. 1998;98(22):2404–2414. 94. Smedema JP, Snoep G, van Kroonenburgh MP, et al. Evaluation of the
76. Hsia HH, Marchlinski FE. Electrophysiology studies in patients with accuracy of gadolinium-enhanced cardiovascular magnetic resonance in
dilated cardiomyopathies. Card Electrophysiol Rev. 2002;6(4):472–481. the diagnosis of cardiac sarcoidosis. J Am Coll Cardiol.
77. John BT, Tamarappoo BK, Titus JL, Edwards WD, Shen WK, Chugh SS. 2005;45(10):1683–1690.
Global remodeling of the ventricular interstitium in idiopathic 95. Anderson LJ, Holden S, Davis B, et al. Cardiovascular T2-star (T2*)
myocardial fibrosis and sudden cardiac death. Heart Rhythm. magnetic resonance for the early diagnosis of myocardial iron overload.
2004;1(2):141–149. Eur Heart J. 2001;22(23):2171–2179.
78. Bogun FM, Desjardins B, Good E, et al. Delayed-enhanced magnetic 96. Flett AS, Hayward MP, Ashworth MT, et al. Equilibrium contrast
resonance imaging in nonischemic cardiomyopathy: utility for cardiovascular magnetic resonance for the measurement of diffuse
identifying the ventricular arrhythmia substrate. J Am Coll Cardiol. myocardial fibrosis: preliminary validation in humans. Circulation.
2009;53(13):1138–1145. 2010;122(2):138–144.
79. de Bakker JM, van Capelle FJ, Janse MJ, et al. Fractionated electrograms 97. Sado DM, Flett AS, Banypersad SM, et al. Cardiovascular magnetic
in dilated cardiomyopathy: origin and relation to abnormal conduction. resonance measurement of myocardial extracellular volume in health
J Am Coll Cardiol. 1996;27(5):1071–1078. and disease. Heart. 2012;98(19):1436–1441.
80. de Bakker JM, van Capelle FJ, Janse MJ, et al. Reentry as a cause of 98. Iles L, Pfluger H, Phrommintikul A, et al. Evaluation of diffuse
ventricular tachycardia in patients with chronic ischemic heart disease: myocardial fibrosis in heart failure with cardiac magnetic resonance
electrophysiologic and anatomic correlation. Circulation. 1988; contrast-enhanced T1 mapping. J Am Coll Cardiol.
77(3):589–606. 2008;52(19):1574–1580.
81. Delacretaz E, Stevenson WG. Catheter ablation of ventricular 99. aus dem Siepen F, Buss SJ, Messroghli D, et al. T1 mapping in dilated
tachycardia in patients with coronary heart disease: part I: mapping. cardiomyopathy with cardiac magnetic resonance: quantification of
Pacing Clin Electrophysiol. 2001;24(8 Pt 1):1261–1277. diffuse myocardial fibrosis and comparison with endomyocardial biopsy.
82. Estner HL, Zviman MM, Herzka D, et al. The critical isthmus sites of European heart journal cardiovascular Imaging. 2015;16(2):210–216.
ischemic ventricular tachycardia are in zones of tissue heterogeneity, 100. Dass S, Suttie JJ, Piechnik SK, et al. Myocardial tissue characterization
visualized by magnetic resonance imaging. Heart Rhythm. using magnetic resonance noncontrast t1 mapping in hypertrophic and
2011;8(12):1942–1949. dilated cardiomyopathy. Circ Cardiovasc Imaging. 2012;5(6):726–733.
83. Mahrholdt H, Wagner A, Judd RM, Sechtem U, Kim RJ. Delayed 101. Mordi I, Carrick D, Bezerra H, Tzemos N. T1 and T2 mapping for early
enhancement cardiovascular magnetic resonance assessment of diagnosis of dilated non-ischaemic cardiomyopathy in middle-aged
non-ischaemic cardiomyopathies. Eur Heart J. 2005;26(15):1461–1474. patients and differentiation from normal physiological adaptation. Eur
84. Assomull RG, Prasad SK, Lyne J, et al. Cardiovascular magnetic Heart J Cardiovasc Imaging. 2016;17(7):797–803.
resonance, fibrosis, and prognosis in dilated cardiomyopathy. J Am Coll 102. Puntmann VO, Carr-White G, Jabbour A, et al. T1-mapping and
Cardiol. 2006;48(10):1977–1985. outcome in nonischemic cardiomyopathy: all-cause mortality and heart
85. Neilan TG, Coelho-Filho OR, Danik SB, et al. CMR quantification of failure. JACC Cardiovasc Imaging. 2016;9(1):40–50.
myocardial scar provides additive prognostic information in 103. Chen Z, Sohal M, Voigt T, et al. Myocardial tissue characterization by
nonischemic cardiomyopathy. JACC Cardiovasc Imaging. cardiac magnetic resonance imaging using T1 mapping predicts
2013;6(9):944–954. ventricular arrhythmia in ischemic and non-ischemic cardiomyopathy
86. Lehrke S, Lossnitzer D, Schob M, et al. Use of cardiovascular magnetic patients with implantable cardioverter-defibrillators. Heart Rhythm.
resonance for risk stratification in chronic heart failure: prognostic value 2015;12(4):792–801.
of late gadolinium enhancement in patients with non-ischaemic dilated 104. Rigo F, Gherardi S, Galderisi M, et al. The prognostic impact of
cardiomyopathy. Heart. 2011;97(9):727–732. coronary flow-reserve assessed by Doppler echocardiography in
87. Gao P, Yee R, Gula L, et al. Prediction of arrhythmic events in ischemic non-ischaemic dilated cardiomyopathy. Eur Heart J.
and dilated cardiomyopathy patients referred for implantable cardiac 2006;27(11):1319–1323.
defibrillator: evaluation of multiple scar quantification measures for late 105. Nikolaidis LA, Doverspike A, Huerbin R, Hentosz T, Shannon RP.
gadolinium enhancement magnetic resonance imaging. Circ Cardiovasc Angiotensin-converting enzyme inhibitors improve coronary flow
Imaging. 2012;5(4):448–456. reserve in dilated cardiomyopathy by a bradykinin-mediated, nitric
88. Klem I, Weinsaft JW, Bahnson TD, et al. Assessment of myocardial oxide-dependent mechanism. Circulation. 2002;105(23):2785–
scarring improves risk stratification in patients evaluated for cardiac 2790.
defibrillator implantation. J Am Coll Cardiol. 2012;60(5):408–420. 106. Opherk D, Schwarz F, Mall G, Manthey J, Baller D, Kubler W. Coronary
89. Leyva F, Taylor RJ, Foley PW, et al. Left ventricular midwall fibrosis as a dilatory capacity in idiopathic dilated cardiomyopathy: analysis of 16
predictor of mortality and morbidity after cardiac resynchronization patients. Am J Cardiol. 1983;51(10):1657–1662.
therapy in patients with nonischemic cardiomyopathy. J Am Coll 107. Neglia D, Michelassi C, Trivieri MG, et al. Prognostic role of myocardial
Cardiol. 2012;60(17):1659–1667. blood flow impairment in idiopathic left ventricular dysfunction.
90. Muller KA, Muller I, Kramer U, et al. Prognostic value of contrast- Circulation. 2002;105(2):186–193.
enhanced cardiac magnetic resonance imaging in patients with newly 108. Dass S, Holloway CJ, Cochlin LE, et al. No evidence of myocardial
diagnosed non-ischemic cardiomyopathy: cohort study. PLoS One. oxygen deprivation in nonischemic heart failure. Circ Heart Fail.
2013;8(2):e57077. 2015;8(6):1088–1093.
91. Kuruvilla S, Adenaw N, Katwal AB, Lipinski MJ, Kramer CM, Salerno M. 109. Neubauer S, Horn M, Cramer M, et al. Myocardial phosphocreatine-to-
Late gadolinium enhancement on cardiac magnetic resonance predicts ATP ratio is a predictor of mortality in patients with dilated
adverse cardiovascular outcomes in nonischemic cardiomyopathy: a cardiomyopathy. Circulation. 1997;96(7):2190–2196.
390.e4 SECTION III  Functional Disease

110. Beer M, Wagner D, Myers J, et al. Effects of exercise training on resonance imaging with echocardiography. Arch Cardiovas Dis.
myocardial energy metabolism and ventricular function assessed by 2010;103(3):150–159.
quantitative phosphorus-31 magnetic resonance spectroscopy and 121. Nucifora G, Aquaro GD, Pingitore A, Masci PG, Lombardi M.
magnetic resonance imaging in dilated cardiomyopathy. J Am Coll Myocardial fibrosis in isolated left ventricular non-compaction and its
Cardiol. 2008;51(19):1883–1891. relation to disease severity. Eur J Heart Fail. 2011;13(2):170–176.
111. Holloway CJ, Dass S, Suttie JJ, et al. Exercise training in dilated 122. Patel MR, Cawley PJ, Heitner JF, et al. Detection of myocardial damage
cardiomyopathy improves rest and stress cardiac function without in patients with sarcoidosis. Circulation. 2009;120(20):1969–1977.
changes in cardiac high energy phosphate metabolism. Heart. 123. Sekiguchi M, Numao Y, Imai M, Furuie T, Mikami R. Clinical and
2012;98(14):1083–1090. histopathological profile of sarcoidosis of the heart and acute idiopathic
112. Towbin JA, Lorts A, Jefferies JL. Left ventricular non-compaction myocarditis. Concepts through a study employing endomyocardial
cardiomyopathy. Lancet. 2015;386(9995):813–825. biopsy. I. Sarcoidosis. Jpn Circ J. 1980;44(4):249–263.
113. Ichida F, Hamamichi Y, Miyawaki T, et al. Clinical features of isolated 124. Yoshida Y, Morimoto S, Hiramitsu S, Tsuboi N, Hirayama H, Itoh T.
noncompaction of the ventricular myocardium: long-term clinical Incidence of cardiac sarcoidosis in Japanese patients with high-degree
course, hemodynamic properties, and genetic background. J Am Coll atrioventricular block. Am Heart J. 1997;134(3):382–386.
Cardiol. 1999;34(1):233–240. 125. Chapelon-Abric C, de Zuttere D, Duhaut P, et al. Cardiac sarcoidosis: a
114. Ritter M, Oechslin E, Sutsch G, Attenhofer C, Schneider J, Jenni R. retrospective study of 41 cases. Medicine (Baltimore).
Isolated noncompaction of the myocardium in adults. Mayo Clin Proc. 2004;83(6):315–334.
1997;72(1):26–31. 126. Kim JS, Judson MA, Donnino R, et al. Cardiac sarcoidosis. Am Heart J.
115. Sandhu R, Finkelhor RS, Gunawardena DR, Bahler RC. Prevalence and 2009;157(1):9–21.
characteristics of left ventricular noncompaction in a community 127. Vignaux O, Dhote R, Duboc D, et al. Detection of myocardial
hospital cohort of patients with systolic dysfunction. Echocardiography. involvement in patients with sarcoidosis applying T2-weighted,
2008;25(1):8–12. contrast-enhanced, and cine magnetic resonance imaging: initial results
116. Petersen SE, Selvanayagam JB, Wiesmann F, et al. Left ventricular of a prospective study. J Comput Assist Tomogr. 2002;26(5):762–767.
non-compaction: insights from cardiovascular magnetic resonance 128. Tadamura E, Yamamuro M, Kubo S, et al. Effectiveness of delayed
imaging. J Am Coll Cardiol. 2005;46(1):101–105. enhanced MRI for identification of cardiac sarcoidosis: comparison with
117. Gati S, Papadakis M, Papamichael ND, et al. Reversible de novo left radionuclide imaging. AJR Am J Roentgenol. 2005;185(1):110–115.
ventricular trabeculations in pregnant women: implications for the 129. Ohira H, Tsujino I, Ishimaru S, et al. Myocardial imaging with
diagnosis of left ventricular noncompaction in low-risk populations. 18F-fluoro-2-deoxyglucose positron emission tomography and magnetic
Circulation. 2014;130(6):475–483. resonance imaging in sarcoidosis. Eur J Nucl Med Mol Imaging.
118. Peritz DC, Vaughn A, Ciocca M, Chung EH. Hypertrabeculation vs 2008;35(5):933–941.
left ventricular noncompaction on echocardiogram: a reason to 130. Vignaux O, Dhote R, Duboc D, et al. Clinical significance of myocardial
restrict athletic participation? JAMA Intern Med. 2014;174(8):1379– magnetic resonance abnormalities in patients with sarcoidosis: a 1-year
1382. follow-up study. Chest. 2002;122(6):1895–1901.
119. Jacquier A, Thuny F, Jop B, et al. Measurement of trabeculated left 131. Greulich S, Deluigi CC, Gloekler S, et al. CMR imaging predicts death
ventricular mass using cardiac magnetic resonance imaging in the and other adverse events in suspected cardiac sarcoidosis. JACC
diagnosis of left ventricular non-compaction. Eur Heart J. Cardiovasc Imaging. 2013;6(4):501–511.
2010;31(9):1098–1104. 132. Tadamura E, Yamamuro M, Kubo S, et al. Images in cardiovascular
120. Thuny F, Jacquier A, Jop B, et al. Assessment of left ventricular medicine. Multimodality imaging of cardiac sarcoidosis before and after
non-compaction in adults: side-by-side comparison of cardiac magnetic steroid therapy. Circulation. 2006;113(20):e771–e773.
32 
T1 and T2 Mapping and Extracellular Volume
in Cardiomyopathy
Thomas A. Treibel and James C. Moon

Cardiovascular magnetic resonance (CMR) exploits the inherent dif- fraction, determined by the picrosirius red method, and showed a sig-
ference between tissues in their configuration of atoms by generating nificant correlation between the two methods. In 2010, Flett et al.11
differing signals—the fundamental tissue properties T1, T2, and T2*. developed an at equilibrium extracellular volume technique and vali-
Whereas differences in these parameters had to be previously visualized dated it in patients with severe aortic stenosis and hypertrophic car-
by weighted sequences, they can now be measured in a single breath- diomyopathy (HCM). The ECV showed a high correlation with the
hold with T1, T2, or T2* displayed as pixel maps where each color-coded collagen volume fraction of biopsies obtained intraoperatively in this
pixel carries the absolute value. Furthermore, if T1 is measured before cohort. This work has been replicated in transplant hearts, shorter
and after contrast, the myocardial extracellular volume (ECV) is mapped, protocols, and with newer, faster T1 mapping sequences (Tables 32.1
representing the percentage of tissue that is extracellular water, a sur- and 32.2).12,13
rogate for the process holding water, be it fibrosis, amyloid, or edema.
In turn, T2 mapping is a highly attractive technique for characterization NATIVE T1, POSTCONTRAST T1, AND
of myocardial tissue in the disease state accompanied by inflammation. EXTRACELLULAR VOLUME
T1, T2, and ECV change in disease, each being differentially sensitive
to pathologic processes (Fig. 32.1). The technique potential is best con- Native T1
sidered in rare (infiltrations), common (edema), and ubiquitous (diffuse Native T1 measures the intrinsic signal from the combined cellular
fibrosis) disease processes. and interstitial compartments of the myocardium.14 The advantages
are that it does not require an exogenous gadolinium-based contrast
TISSUE CHARACTERIZATION: MOVING BEYOND agent. Native T1 relaxation time is prolonged with collagen (fibrosis),11
edema,15 and amyloid16 and is shortened with reduced fibrosis,17 iron,18
LATE GADOLINIUM ENHANCEMENT fat,19 and hemorrhage.20 Given that native T1 measures both intersti-
The key single technique that stimulated the greater adoption of CMR tium and myocyte T1, a signal from the interstitium alone is somewhat
into routine clinical practice was scar imaging. The late gadolinium diluted by the myocyte signal, so subtle differences (diffuse fibrosis) are
enhancement (LGE) technique was first used in infarction, but it has harder to detect. Moreover, capillary density, capillary vasodilatation,
proved to be reproducible1 and robust enough for use in a multicenter and “partial voluming” between blood pool and myocardium are also
clinical trial,2 and it has established itself as the gold standard method measured, potential biases if the signal sought is the matrix or myocyte
in both ischemic and nonischemic heart diseases, including cardiomy- compartments alone. Native T1 time is different with field strength and
opathy,3 myocarditis,4 aortic stenosis-induced pressure-overload hyper- sequence design21 and varies between scanners, making comparison of
trophy,5 and infiltrative diseases.6 However, LGE is a difference test native and postcontrast values between centers challenging. Currently,
between normal and abnormal myocardium, and therefore is not able several groups have developed T1 phantoms to facilitate multicenter trials
to characterize and quantify diffuse myocardial disease or inform on (Hypertrophic CardioMyopathy Registry [HCMR]22) or develop refer-
how nonscarred areas are adapting to the increased workload or whether ence standards (T1 mapping and ECV standardization in CMR [T1MES]
they are at risk of generating new scar. There are many pathways active program23).
in normal myocardium, and these change with different pathologic
processes. Each parameter may be differently sensitive to these. Multi- Postcontrast T1
parametric tissue characterization is therefore an attractive strategy for After administration of gadolinium, T1 is dominated by, and inversely
noninvasive “biopsy” and “whole heart” sampling, avoiding potential proportional to, the concentration of tissue gadolinium. Measuring T1
morbidity and mortality of actual biopsy.7 after contrast provides a value linked to the interstitium and has been
applied to patients with heart failure.24 Postcontrast T1 also varies with
HISTOLOGIC VALIDATION OF T1, T2, AND gadolinium dose, time post bolus, and importantly, patient-specific factors
such as heart rate, clearance rate, body composition, and hematocrit.
EXTRACELLULAR VOLUME
Extracellular tracers for measuring the interstitium were first described Extracellular Volume Fraction
in the 1960s.8 In the 1970s, Poole-Wilson et al.9 used 51Cr-EDTA in an If the change in T1 precontrast and postcontrast is measured in both
ex vivo heart model with photographic paper to measure the myo- blood and myocardium after sufficient equilibration of the contrast
cardial extracellular volume. Early in vitro work by Kehr et al.10 on distribution, the partition coefficient can be calculated. By adding in
human myocardium obtained postmortem compared T1 values, calcu- the blood compartment contrast volume of distribution (1 − hemato-
lated from the inversion recovery signal curves, with collagen volume crit), the myocardial ECV is derived (Fig. 32.2). ECV is a more stable

391
392 SECTION III  Functional Disease

Normal Infarction Diffuse fibrosis Myocarditis

A B C D

Disarray Fabry Amyloid Iron

E F G H
FIG. 32.1  Histopathological findings on myocardial biopsy. (A–H) Eight different pathologies that can poten-
tially be characterized by cardiovascular magnetic resonance multiparametric mapping and may allow us to
understand the underlying pathophysiologic processes of several diseases.

ECVblood= 1 - HCT Native T1 Postcontrast T1 ECV map

FIG. 32.2  Native T1, postcontrast T1, and extracellular volume (ECV). ECV maps derived by acquiring hema-
tocrit (HCT), native T1, and postcontrast T1 modified Look-Locker inversion recovery of the short axis of a
healthy volunteer.

TABLE 32.1  Histologic Validation Cohorts in T1 and T2 Mapping


Reference Year Population N Parameter Sequence
Iles et al.24 2008 DCM 25 Postcontrast T1 1.5 T; IR VAST
Flett et al.11 2010 AS/HCM 26 ECV 1.5 T; EQ-CMR; multibreath-hold FLASH IR
Sibley et al.100 2012 DCM/IHD/HCM/amyloid 47 Postcontrast T1 1.5 T, IR Look-Locker
Mascherbauer et al.88 2013 HFpEF 9 Postcontrast T1 1.5 T, IR FLASH
White et al.37 2013 AS 18 ECV 1.5 T; EQ-CMR; ShMOLLI
Miller et al.13 2013 DCM/IHD (transplant) 6 ECV 1.5 T, MOLLI
Bull et al.96 2013 AS 19 Native T1 1.5 T; ShMOLLI
Lee et al.106 2015 AS 20 Native T1 3 T, MOLLI
De Meester et al.107 2015 AS/AR/MR 31 T1 and ECV 3 T, MOLLI
Kammerlander et al.132 2016 Mixed HF 36 ECV 1.5 T, MOLLI
Lurz et al.53 2016 Myocarditis 129 T1/T2/ECV 1.5 T and 3 T, MOLLI

AR, Aortic regurgitation; AS, aortic stenosis; CMR, cardiovascular magnetic resonance; DCM, dilated cardiomyopathy; ECV, extracellular volume
fraction; EQ, equilibrium contrast; HCM, hypertrophic cardiomyopathy; HF, heart failure; HFpEF, heart failure with preserved ejection fraction;
IHD, ischemic heart disease; IR, inversion recovery; MOLLI, modified Look-Locker inversion recovery; MR, mitral regurgitation; ShMOLLI,
shortened modified Look-Locker inversion.
CHAPTER 32  T1 and T2 Mapping and Extracellular Volume in Cardiomyopathy 393

TABLE 32.2  Overview of T1, T2, and Extracellular Volume Fraction Mapping in Myocardial
Diseasea
Histological
Biological Process Characteristic Disease T1 Signal T2 Signal ECV Signal Validation Main Referencesa
Fibrosis
Focal Myocardial infarction, no Verhaert et al. 2011108;
hemorrhage Ugander et al. 2012109
Diffuse Aortic stenosis — — Flett et al., 201011; Bull et al.,
201396; Chin et al., 2014110;
Singh et al., 2015111
b
Systolic heart failure Iles et al., 200824; Su et al.,
2014113; Bohnen et al., 2015112
Diastolic heart failure Su et al., 2014113
Hypertrophic cardiomyopathy — Ho et al., 2013114;
Puntmann et al., 2013101
Nonischemic dilated Puntmann et al., 2013101;
cardiomyopathy Barison et al., 2015115
Mitochondrial Lee et al., 2014116
cardiomyopathy
Diabetes Wong et al., 2013117
Hypertensive heart disease — Kuruvilla et al., 201592;
Treibel et al., 201591
Obesity Shah et al., 2013118
Congenital heart disease Plymen et al., 2013119
Inflammatory Rheumatoid arthritis Ntusi et al., 201580
c
Systemic sclerosis Ntusi et al., 201479;
Barison et al., 2015120
Systemic lupus Puntmann et al., 201378;
erythematosus Zhang et al., 2015

Edema
Regional Acute myocarditis Ferreira et al., 201377;
Hinojar et al., 2015121
Takotsubo cardiomyopathy Thavendiranathan et al., 2012122
Global Acute myocarditis Ferreira et al., 201377;
Hinojar et al., 2015121
Antisynthetase syndrome Sado et al., 201682
Active systemic capillary Ertel et al., 2015123
leak syndrome
Acute cardiac allograft —/ —/ — Miller et al., 2014124
rejection

Infiltration
Amyloid AL amyloid Banypersad et al., 201564
Glycosphingolipid TTR amyloidosis Fontana et al., 2014125;
Anderson-Fabry disease — Messalli et al., ,2012; Thompson
et al. 2013126; Sado et al., 201419
Iron Thalassemia major Hanneman et al., 2015127
Sickle cell disease / Alam et al., 2015128
Hereditary hemochromatosis Alam et al., 2015128
Myocardial infarction, with Verhaert et al., 2011108;
hemorrhage Pedersen et al., 2012129
Toxins Uremia in chronic kidney Edwards et al., 2014130
disease
Anthracycline toxicity /— — /- Tham et al., 2013131
a
Reference list is nonexhaustive—several other references may exist that are not listed here.
b
If myocardial hemorrhage accompanies acute infarction.
c
In acute heart failure.
ECV, Extracellular volume; —, no significant change; ↑, significant increase; ↓, significant decrease; , unreported to date.
Modified from Captur G, Manisty C, Moon JC. Cardiac MRI evaluation of myocardial disease. Heart. 2016;102(18):1429--1435.
394 SECTION III  Functional Disease

LV Mass LV Mass
100 mL 150 mL

Matrix A B C
Cells Cells LVH
Cells
Cells

ECV 25% ECV 15% ECV 25% ECV 40%


Cells 75 mL Cells 127 mL Cells 112 mL Cells 90 mL
Matrix 25 mL Matrix 23 mL Matrix 38 mL Matrix 60 mL
FIG. 32.3  Extracellular volume (ECV) divides the myocardium into cell and matrix components. In diseases
with global homogeneous hypertrophy and extracellular matrix expansion, the ECV fraction may offer limited
information, because it just depicts the ratio between cell and matrix volumes. By multiplying the left ven-
tricular mass (LVM) with the global ECV, matrix volume (LVM * ECV) and cell volume (LVM * [1 − ECV]) can
be derived, respectively. LVH, Left ventricular hypertrophy.

and biologically significant biomarker, as well as a more robust parameter may offer more rapid multiparametric tissue characterization in the
than T1.25,26 future by providing myocardial T1, T2, and proton spin density in a
single breath-hold.41
Extracellular Volume Fraction Dichotomizing the For clinical use, these developments need to transition to standard-
Myocardium Into Cell and Matrix Components ized methodologies to diagnose disease; define mechanistic pathways
ECV divides the myocardium into two compartments (extracellular of disease affecting the interstitium, the myocyte, or both; change therapy;
and cellular) and, therefore allows noninvasive quantification of the and employ ECV as a surrogate endpoint in trials of drug development.
myocardial matrix volume and its counter-part, cell volume (Fig. 32.3). This is the aim underpinning the first T1 mapping consensus state-
The cell volume represents intact myocardial cellular components, ment.42 Our conceptual models are simple, but there is more going on;
proving a way to measure the myocyte volume (note that this also effects such as magnetization transfer, diffusion distance and time,
includes fibroblasts, blood cells, macrophages, etc.). How these change contrast mechanisms, transcytolemmal water exchange rate, flow, T2
in disease (e.g., left ventricular hypertrophy [LVH]) is important. For or T2* relaxation will require further investigation.43,44 Part of this
example, in transthyretin-related (TTR) hereditary amyloidosis and development requires global approaches. Quality control systems, com-
light-chain amyloidosis, both have a massive matrix increase, but TTR mercial sequences, megaregistries (e.g., Global CMR Registry, HCM
has more matrix and 20% higher cell volume suggesting compensatory Registry, UK Biobank) are in progress, and will provide high volumes
hypertrophy, which may permit more tolerance of the amyloid burden.27 of new insights in what is now the most active CMR research area.22,45
By modeling water exchange, there is also some evidence that the con-
trast kinetics could be used to obtain cell size-dependent parameters, Continuous Improvement Versus Stability
particularly if very high doses of gadolinium contrast are used.14 For a “feel” for the potential of mapping in different pathologic processes
compared with health, transform the absolute difference into the
T1 Mapping Evolution maximum possible signal-to-noise ratio (SNR) in standard deviation
The T1 mapping field is rapidly advancing to the point of widespread (SD) units in severe but not extreme disease (a measure of effect size).
clinical utility. The first to T1 mapping was Messroghli in 2004 with a Provided minimal systematic bias by disease-tracking confounders, such
pulse-sampling scheme known as MOLLI (modified Look-Locker inver- as heart rate or anemia, an SD change of 2 suggests the technique can
sion recovery),28 replacing previous multibreath-hold approaches. This detect between-group differences for biologic insights, >4 and it could
was refined, including new MOLLI variants, shortened modified Look- determine the choice of therapy in individuals, and >6 said therapy
Locker inversion recovery (ShMOLLI; a shortened variation with long could be monitored during treatment. A measured value consists of
T1 advantages28–30), saturation recovery variants such as saturation combined biologic and measurement variability. Considerable ongoing
recovery single-shot acquisition (SASHA; offering complete heart rate work is reducing the measurement variability, so the above SD changes
insensitivity30) or hybrid approaches (accelerated and navigator-gated are increasing with technical development.
Look-Locker imaging for cardiac T1 estimation [ANGIE], quantification
using an interleaved Look-Locker acquisition sequence with T2 prepa-
ration pulse [QALAS], SAturation Pulse Prepared Heart rate independent
T2 MAPPING AND MYOCARDIAL EDEMA
Inversion-REcovery sequence [SAPPHIRE]31–33). Incremental develop- Cardiovascular magnetic resonance can assess myocardial edema by
ments such as respiratory motion correction34 gradually increased early gadolinium enhancement (EGE), T2-weighted imaging, LGE, mul-
accuracy and precision.31,35 For ECV, contrast regimes were simplified tiparametric mapping (T1 precontrast, T1 postcontrast, T2 mapping),
from bolus followed by infusion or multi-timepoint sampling to a single and the hybrid novel positron emission tomography/magnetic reso-
precontrast and single postcontrast T1 map.36,37 Split contrast dose nance imaging (PET/MRI) technique for myocardial inflammation.
protocols suitable for stress perfusion imaging have been validated.38 T2-weighted CMR has become established for the detection of myocardial
ECV maps are now routine in some centers.39 ECV quantification is edema,46 has been a particularly valuable tool for the discrimination
less field and sequence sensitive than native T1 mapping but ECV stan- of acute from chronic myocardial infarction,47 and has been investi-
dardization is ongoing. Most recently, it was a found that a synthetic gated in health and disease.48–51 But as for T1, T2-weighted sequences
ECV can be automatically generated during scanning, in which the are prone artifacts, have low sensitivity,52 and require either reference
hematocrit of blood is inferred from the T1 of the blood pool (as the signal intensities in skeletal muscle (assuming that is normal) or are
relationship between hematocrit and R1 [1/Blood T1] is linear), remov- sensitive only to regional changes. As a result of different hardware or
ing the need for a blood test.40 Finally, magnetic resonance fingerprinting sequence parameters, reference values differ considerably. Furthermore,
CHAPTER 32  T1 and T2 Mapping and Extracellular Volume in Cardiomyopathy 395

local myocardial wall motion and heart rate influence local myocardial ECV was higher in women than in men (28.1 ± 2.8% vs. 25.8 ± 2.9%;
T2 values.51 Although T2 acquisition at 3 T has the potential benefit of P < .001). ECV correlated with age, however, with an R2 of 0.021 (P =
increased signal, it offered the same diagnostic accuracy as 1.5 T in the .012); that is, 98% of the variability of T1 was attributable to factors
MyoRacer study.53 Using a turbo gradient spin echo sequence at 1.5 T, other than age.
Bönner et al.54 showed that T2 mapping was highly reproducible, and A recent study investigating physiologic hypertrophy in an athlete’s
that female sex and aging were accompanied by increased myocardial heart using T1 mapping showed that hypertrophy was caused by an
T2 values, but found no influence of heart rate or regional strain. T2 expansion of the cellular compartment while the extracellular volume
mapping is therefore good for the characterization of myocardial tissue becomes relatively smaller, hinting that ECV quantification may have
in ischemia or inflammation, as described later. a future role in differentiating physiologic from pathologic left ventricular
hypertrophy (LVH), which is a major result if confirmed.17
T1 MAPPING IN HEALTH AND DISEASE
INFILTRATIVE CARDIOPATHIES: AMYLOID, IRON
T1 mapping appears robust in detecting disease characterized by high
T1 and ECV (Figs. 32.4, 32.5, and 32.6), such as amyloidosis,16 or marked
OVERLOAD, FABRY DISEASE
native T1 contrast diseases such as Fabry disease ([FD]; where fat storage Infiltrations (iron, FD, amyloid) are important because although rare,
makes native T1 fall)19; little biopsy histologic correlation is available expensive therapies are available that need noninvasive targeting and/
in these diseases, however. For smaller expansions, reference ranges are or development. Furthermore, they provide insights into more complex
needed. It is not exactly clear yet what these need to be. Gender appears and more common polygenic diseases because these diseases have very
to make a difference, but age has a much smaller or possibly no con- high signal changes on multiparametric mapping. The only pathology
tribution55 once partial volume issues are accounted for. Here, the ratio that causes T2* to fall is iron (−7 SD), although cobalt and chromium
of pathophysiologic signal-to-measurement error is a key determinant have been described in the liver57; T1 only falls substantially in two
of test performance. Liu et al.56 reported on T1 and ECV data using currently known diseases (see Fig. 32.4B and C), FD (−6 SD) and iron
older T1 techniques from 1231 participants aged 54 to 93 years of the (−15 SD), with one study suggesting a small fall in athletic adaption
Multi Ethnic Study of Atherosclerosis (MESA) study, where women (−1 SD). Amyloid native T1 elevation is marked (+8 SD; see Fig. 32.4D
had significantly higher native T1 and partition coefficient as well as and E), and ECV elevation in amyloidosis (amyloidosis―where the
lower postcontrast T1 times than men. When hematocrit was available, amyloid is the cause of disease rather than a bystander) is always above

3000
ms

A B C

300
D E F ms
FIG. 32.4  Native T1 in health and disease. Native T1 maps (shortened modified Look-Locker inversion) in
the basal short axis of: (A) a healthy volunteer―the myocardium appears homogenously green and the blood
is red; (B) severe iron overload―the myocardium appears blue as the T1 is low from iron; (C) Fabry disease―
the myocardium has a lower T1 (blue) because of intracellular lipid accumulation, except in the inferolateral
wall, which is red because of fibrosis; (D) transthyretin amyloidosis (ATTR)―the myocardium has a higher
T1 (red); (E) light chain (AL) amyloidosis―the myocardium has a very high T1 with less hypertrophy than
ATTR; and (F) hypertrophic cardiomyopathy―there is asymmetrical septal hypertrophy with right ventricular
insertion point scar (red).
396 SECTION III  Functional Disease

Healthy volunteer Myocarditis Aortic stenosis HCM ATTR amyloid


100%

0%

FIG. 32.5  Examples of extracellular volume (ECV) in health and disease. ECV maps (top row) and correspond-
ing late gadolinium enhancement (LGE, bottom row) in the short axis of a healthy volunteer: the myocardium
appears homogenously blue. Global myocarditis—note, because of the homogenous edema, there is no
focal LGE, but the ECV map shows diffusely elevated ECV. Aortic stenosis—the ECV maps depicts ECV
elevation corresponding to scar in the LGE image. Hypertrophic cardiomyopathy (HCM)—the ECV maps
shows ECV elevation corresponding to scar in the right ventricular insertion points in the LGE image. Trans-
thyretin amyloidosis (ATTR)—the blood:myocardial interface is lost in the ECV map because the ECV is as
high as in the blood pool (i.e., ~60%)—the LGE images show classical global enhancement.

High

Amyloidosis
Acute
infarction
Focal
fibrosis

Myocarditis
ECV/
Diffuse fibrosis
%

Fabry
Iron Normal

Athlete

Fat
Low

Low Native T1 (ms) High


FIG. 32.6  Native T1 versus extracellular volume (ECV) in different pathologies. (Modified from Martin Ugander
“Fibrosis and Edema” presentation, SCMR 2014.)

45%, a level seemingly impossible in other diffuse diseases (although Amyloidosis


more needs to be known about global myocarditis). Mapping in these Cardiac amyloidosis is a rare condition characterized by a progressive
diseases has important benefits: it may offer superior reproducibility infiltrative cardiomyopathy in which deposits of amyloid accumulate
(T1 in iron18), earlier disease detection (T1 is low in 50% of FD subjects in the ventricular myocardium, almost always of either immunoglobulin
without LVH19), or tracking change with therapy (amyloid58). light-chain (amyloid light-chain [AL]) or transthyretin (amyloid
CHAPTER 32  T1 and T2 Mapping and Extracellular Volume in Cardiomyopathy 397

transthyretin-related [ATTR]) type, the latter being wild type or mutant. T2* and was more reproducible than T2*. T2* mapping appears robust
Advances in echocardiography, bone tracer scintigraphy, and CMR mean and simplifies analysis,72 but performance in difficult cases remains
the noninvasive diagnosis of cardiac amyloidosis is maturing and biopsy undefined. Abdel-Gadir et al.73 performed 126 scans in two 12-hour
is no longer needed in many circumstances.59 Prognosis is often poor, days in Thailand, with an average scan time of 8.3 minutes for combined
and treatments are substantially influenced by cardiac involvement. T1 and T2* mapping that could be analyzed within 1 minute. Although
CMR with phase-sensitive LGE has characteristic findings in cardiac the combined T1 and T2* acquisition is similar in duration to a T2*
amyloidosis and provides incremental information on the outcome scan alone, further work is needed to establish the clinical value of such
even after adjustment for known prognostic factors; transmural involve- an approach.74 An ultrafast mapping approach is being pursued in 18
ment represents advanced cardiac amyloidosis with poor prognosis.60 countries and 28 sites globally.75
The marked interstitial expansion is also reflected on T1 and ECV
mapping: Native T1 is significantly elevated in both AL and ATTR
amyloidosis16,55,61—useful in a population with a high prevalence of
MYOCARDIAL EDEMA
renal failure. Karamitsos et al.16 studied 53 patients with AL amyloidosis Edema occurs in acute infarction and myocarditis (Fig. 32.7) but may
and compared native T1 and LGE to biomarker and echocardiographic be more ubiquitous—if we could detect it. It is also a therapeutic target.
criteria. They found that native T1 yielded a 92% accuracy for cardiac It can be intracellular or extracellular, and although it increases native
amyloidosis with possible or definite involvement and that it appeared T1 and ECV, T2 appears specifically sensitive with high elevations. Global
more sensitive than LGE. Similarly, Fontana et al.27 found that native inflammation by T2 can be found in acute heart failure, tracking his-
T1 has a similar diagnostic accuracy in TTR amyloidosis but with a tologic inflammation, and in connective tissue diseases. Whilst there is
lower maximal T1, which has led to the (as yet unproven) hypothesis increasing recognition of global myocarditis, LGE is likely only seeing
that the rapid deposition of amyloid fibrils in AL may be associated the tip of the iceberg.
with myocardial edema. T2 mapping data is currently awaited in cardiac
amyloid, although results from T2-weighted imaging have been con- Myocarditis
flicting, with one study showing no difference in T262 and another study The diagnosis of myocarditis still represents a diagnostic challenge due
showed a lower T2 ratio in cardiac amyloid, related to poorer survival to the variable clinical presentation, the invasive nature, and the limited
(although no differentiation between AL and ATTR was made).63 availability of the gold standard of endomyocardial biopsy (EMB). CMR
ECV is a more direct marker of extracellular expansion, has high offers the ability to make a morphologic, functional, and tissue assess-
diagnostic accuracy for the diagnosis of cardiac amyloidosis, and predicts ment in myocarditis, and this distinguishes it from other diagnostic
survival.64 ECV has therefore been incorporated as a surrogate endpoint modalities. However, it has lower sensitivity for pan-myocarditis or
in clinical trials in amyloidosis (ClinicalTrials.org: NCT01777243/ borderline cases.52 Traditionally, T2-weighted imaging has been used
NCT01981837).58 When confronted with a patient with LVH, a very to detect free water, and T1-weighted images precontrast and postcon-
elevated T1 or ECV helps distinguish amyloidosis from hypertension trast for hyperemia based on historic studies at 0.35 T.76 The Lake Louise
or HCM. criteria (LLC),52 consisting of late enhancement sequences, T2-weighted
edema images, and T1-weighted sequences before and after contrast
Fabry Disease injection (early enhancement), have been developed to standardize the
FD is a rare but treatable X-linked disorder resulting in sphingolipid diagnostic pathway, but they are now old, and new approaches are not
deposition in a number of different organs. Cardiac manifestations included. The LLC can be challenging in a heart failure population with
include LVH, arrhythmias, and valvular disease, and cardiovascular chronic significant overlap in symptoms and CMR findings. Multipa-
disease is now the primary cause of death in FD.65 Early treatment rametric mapping techniques and quantification of the ECV have been
with enzyme replacement therapy (ERT) may reverse or slow disease suggested to overcome some limitations of the LLC, creating high
progression. Native T1 mapping can identify patients early, poten- expectations about their diagnostic utility. A recent study comparing
tially helping target costly enzyme therapy. Two groups independently T2-weighted imaging, late gadolinium, and T1 mapping at 1.5 T in 50
demonstrated lower myocardial T1 values in FD.66,67 Furthermore, T1 patients with myocarditis showed that native T1 mapping detected
mapping can discriminate against other diseases with hypertrophy, with myocardial edema with higher sensitivity when compared with
no overlap in T1 values,66 and has excellent reproducibility.68 Pseudo- T2-weighted (note: nonquantitative) imaging77 and with a 91% diag-
normalization in T1 values (mixed extracellular fibrosis and lipid) and nostic accuracy when compared with LLC. In the recently published
subsequent prolongation in T1 (burnt-out fibrosis) may provide an MyoRacer Trial, Lurz et al.53 assessed the diagnostic performance of a
insight into disease progression. Furthermore, recent data by Nordin comprehensive CMR protocol against EMB, including the LLC, native
et al.69 comparing T1 and T2 mapping, LGE as well as high-sensitivity T1 mapping, quantification of ECV, and T2 mapping. In acute cases,
troponin T (hs-TnT) and N-terminal pro b-type natriuretic peptide native T1 yielded the best diagnostic performance, as defined by the
in FD with HCM, chronic myocardial infarction and healthy control, area under the curve (AUC) of receiver-operating curves (AUC 0.82)
showed elevated T2 values in areas of LGE in FD. This highly correlated followed by T2 (AUC 0.81), ECV (AUC 0.75), and LLC (AUC 0.56). In
with hs-TnT, suggesting that FD with LGE is a chronic inflammatory chronic cases, only T2 mapping has acceptable diagnostic performance
cardiomyopathy. in patients with chronic symptoms.

Cardiac Siderosis Connective Tissue Disease


The impact of T2* measurement is discussed in Chapter 33. CMR In addition to viral myocarditis, other systemic inflammatory disorders
linked to therapy has had a dramatic impact in reducing morbidity can affect the myocardium. T1 mapping and ECV detect myocardial
and mortality in cardiac iron loading, Cardiac iron loading has largely inflammation and fibrosis that in systemic lupus erythematosus, systemic
been the domain of T2* measurement sequences, which are histologi- sclerosis, and rheumatoid arthritis78–80 and may be able to guide treat-
cally validated,70 and can track iron levels with chelation. Cardiac sid- ment. T1 and ECV have been used to identify cardiac involvement in
erosis, however, prolongs all three CMR parameters: T1, T2, and T2*.71 other diseases such as systemic capillary leak81 and antisynthetase
Sado et al.18 showed in 88 patients that T1 mapping correlates well with syndrome—both are rare but potentially fatal.82
398 SECTION III  Functional Disease

A B

C D
FIG. 32.7  T2 mapping in health and disease. T2 mapping in a normal volunteer (A). High T2 value in patient
with myocarditis—here epicardial edema (B). Edema in acute myocardial infarction (C)—here patchy because
of microvascular obstruction—see late gadolinium enhancement (D). (From Maestrini A. New generation
cardiac parametric mapping: the clinical role of T1 and T2 mapping. Magnetom Flash. 5:2013 [Siemens];
https://www.healthcare.siemens.co.uk/magnetic-resonance-imaging/options-and-upgrades/clinical-applications/
myomaps/use.)

Sarcoid Myocarditis fibrosis and is a major therapeutic target. It is prognostic, and in


Sarcoidosis is a systemic granulomatous inflammatory disease with unselected patients it predicts outcomes as strongly as left ventricular
unknown etiology that can affect the heart either in isolation or as part ejection fraction11,86 in some cohorts―results that, if generalizable, place
of a multisystem disease. Typical cardiac manifestations include arrhyth- the measured ECV as a fundamental myocardial property.
mias, heart failure, and/or sudden cardiac death. Sarcoidosis causes LV From a trial perspective, therefore, T1 mapping is a useful sur-
impairment from scar. Acutely, this is inflammatory. CMR using the rogate for an established pharmacologic target. From a clinical stand-
LGE technique alone is unable to inform on whether there is disease point, ECV appears to be an important short-term prognostic marker,
activity that may potentially be treatable with immunosuppression. predicting mortality at 2 years and acute heart failure admissions in
18
F-Fluorodeoxyglucose positron emission tomography (FDG-PET) may diabetics.86,87 Mascherbauer et al.88 showed in a cohort of heart failure
be useful, but it is unlikely to be able to monitor the response to therapy with preserved ejection fraction that postcontrast T1 is associated
because of the radiation dose. Mapping may help. To date, exploration with prognosis. In an unselected cohort, ECV was associated with
has been limited, and further work is needed. hospitalization for heart failure, death, or both; was more strongly
associated with outcomes than nonischemic LGE; improved the clas-
sification of persons at risk; and improved model discrimination for
DIFFUSE FIBROSIS IN HEART FAILURE outcomes.89
Heart failure is a significant cost to health care services worldwide and
yet its pathophysiology remains unclear, in part because of inadequate DIFFUSE FIBROSIS IN PRESSURE
animal models but also because of the complexity of biochemical
processes that are involved. Nevertheless, extracellular matrix (ECM)
OVERLOAD HYPERTROPHY
expansion is an important element of adverse remodeling either reflect- Arterial hypertension and aortic stenosis (AS) are attractive diseases
ing end-stage disease or signaling further deterioration. Importantly, for T1 mapping because they are common, lead to diffuse myocardial
diffuse fibrosis can be reversed by current therapies,83,84 and new therapies fibrosis, and are amenable to treatment (e.g., antifibrotic antihyperten-
show promise.85 T1 and ECV rise in fibrosis, with ECV appearing the sives such as an angiotensin-converting enzyme [ACE] inhibitor or
better test (~ +3 SD vs. +4 SD change). The ECV is shown to track aortic valve replacement [AVR], respectively).
CHAPTER 32  T1 and T2 Mapping and Extracellular Volume in Cardiomyopathy 399

validated against normal populations.55,101,102 Partially voluming of blood


Arterial Hypertension is a particular challenge in this population where only 3 to 4 pixels may
Recent data exploring the utility of T1 mapping in arterial hypertension cover the thin ventricular wall and bundle branch block may add to
and hypertensive heart disease have emerged,90–92 but differences in this complication (a typical voxel size is ~ 2 × 2 × 8 mm3).
native T1 and ECV have been found to be small (with significant overlap
between hypertensive and control) and occurred only in those patients Hypertrophic Cardiomyopathy
with LVH. Our own group concluded erroneously that the changes T1 mapping with ECV measurement as a novel imaging biomarker of
were at the detectable limit, but we underappreciated that the ECV may diffuse myocardial fibrosis has been explored in HCM. HCM has also
be unchanged if fibrosis is accompanied by a proportional increase in been similarly validated for both ECV and native T1 against normal
cell hypertrophy (i.e., proportional increase in cell and matrix volume).91 populations.37,90,101 Puntmann et al.101 found that native T1 had a higher
discriminatory performance than ECV.103 This may hint that a high field
Aortic Stenosis strength or magnetization transfer weighting may improve native T1
The timing for surgery in asymptomatic severe AS remains unclear mapping for the detection of fibrosis, here in HCM. Others have shown
despite a number of studies in the last decade.93 AS is not just a disease that T1 mapping abnormalities are larger than the LGE extent.102 More
of the valve. Graham Steell94 noted in 1906 that it is the “reaction of work will be needed to understand these results. Wong et al.104 have
the heart muscle” that is the “unmixed evil.” Reflecting this, severe fibrosis observed a preliminary association between ECV and B-type natriuretic
by biopsy at the time of surgery was associated with worse outcome peptide (BNP), and Ho et al.105 reported that an ECV elevation is an
and more limited postoperative improvement in systolic and diastolic early phenomenon, occurring in preclinical HCM (sarcomeric HCM
function.95 Because fibrosis development seems to correlate with symp- mutation carriers without left ventricular hypertrophy). Finally, we await
toms and systolic function, ECV as an imaging surrogate has the potential the results of the large 40-center observational study of HCM, HCMR,
to provide a new marker to time surgery.11,96 Multiple studies are cur- which is currently well on the way to completing recruitment, organized
rently underway due to report in the next few years and should provide by Kramer and Neubauer to characterize prognostic markers in HCM,
prospective information on the utility of T1 mapping in AS (Clinical- including T1 mapping markers of diffuse fibrosis (ClinicalTrials.gov
Trials.org NCT01658345, NCT02174471, and NCT01755936). In severe identifier NCT01915615).22
AS patients, ECV has been found to be elevated, but, interestingly, LVH
regression 6 months after AVR was cellular rather than fibrosis regres-
sion.97 Recent data by our group (RELIEF-AS Study) found that at
CONCLUSION
1-year post-AVR, LV mass regressed by 20%, which was the result of a Multiparametric myocardial characterization with T1, T2, and ECV
17% reduction in fibrosis volume and a (higher) 23% reduction in cell has the potential to play a big role in a variety of clinical settings. The
volume. The greater cellular regression led to an increase in ECV; native ability to distinguish multiple new pathologic processes such as fat
myocardial T1 was unchanged. These data support the position that storage, inflammation, diffuse fibrosis, and amyloid is a major advance
human diffuse fibrosis is dynamic and that this is measurable by CMR, (see Fig. 32.6). This information is beginning to be understood and
a key biological result and proof-of-concept for drug development tar- incorporated into clinical practice. But it will take time to fully use
geting myocardial fibrosis.98 these techniques to develop new drugs and change patient care and
outcomes. Current challenges include restricted (although increas-
ing) accessibility, nonstandardized reference values for normal and
GENETIC CARDIOMYOPATHIES abnormal myocardium (addressed by current phantom studies), and
Cardiomyopathy encompasses a broad range of diseases from hyper- nonuniformity of technique amongst vendors. We anticipate it will
trophic, dilated to noncompaction. The phenotypic variety makes each be the gold standard for diagnosis and treatment monitoring in rare
difficult to characterize; myocardial fibrosis is often the common denomi- diseases, where there are high disease-related changes and clinical need,
nator, regardless of pathophysiology. This can be either reactive and and a drug development imperative will catalyze its standardization.
interstitial, such as present in idiopathic dilated cardiomyopathy (DCM), Subsequently, this should provide the development platform for more
or replacement fibrosis after myocyte necrosis, as in HCM.99 Previously, robust methods for diffuse fibrosis. Combined, the potential is of better
myocardial biopsy was the only means to establish fibrosis, but this mechanistic insights into disease processes. It should eventually lead
carries its own risk, is prone to sampling error, and does not reflect the to improved diagnostic pathways, prognostication, and monitoring
extent within the whole heart. of therapy.

Dilated Cardiomyopathy
REFERENCES
In DCM, prolonged native T1 has been histologically validated in small
numbers of patients100 and both native T1 and ECV are more extensively A full reference list is available online at ExpertConsult.com
CHAPTER 32  T1 and T2 Mapping and Extracellular Volume in Cardiomyopathy 399.e1

REFERENCES 21. Raman FS, Kawel-Boehm N, Gai N, et al. Modified Look-Locker


inversion recovery T1 mapping indices: assessment of accuracy and
1. Mahrholdt H, Wagner A, Holly TA, et al. Reproducibility of chronic reproducibility between magnetic resonance scanners. J Cardiovasc
infarct size measurement by contrast-enhanced magnetic resonance Magn Reson. 2013;15:64.
imaging. Circulation. 2002;106:2322–2327. 22. Kramer CM, Appelbaum E, Desai MY, et al. Hypertrophic
2. Kim RJ, Albert TS, Wible JH, et al. Performance of delayed-enhancement Cardiomyopathy Registry: the rationale and design of an international,
magnetic resonance imaging with gadoversetamide contrast for the observational study of hypertrophic cardiomyopathy. Am Heart J.
detection and assessment of myocardial infarction: an international, 2015;170:223–230.
multicenter, double-blinded, randomized trial. Circulation. 23. Captur G, Gatehouse P, Kellman P, et al. A T1 and ECV phantom for
2008;117:629–637. global T1 mapping quality assurance: the T1 mapping and ECV
3. Choudhury L, Mahrholdt H, Wagner A, et al. Myocardial scarring in standardisation in CMR (T1MES) program. J Cardiovasc Magn Reson.
asymptomatic or mildly symptomatic patients with hypertrophic 2016;18:1–3.
cardiomyopathy. J Am Coll Cardiol. 2002;40:2156–2164. 24. Iles L, Pfluger H, Phrommintikul A, et al. Evaluation of diffuse
4. Wagner A, Schulz-Menger J, Dietz R, Friedrich MG. Long-term myocardial fibrosis in heart failure with cardiac magnetic resonance
follow-up of patients paragraph sign with acute myocarditis by magnetic contrast-enhanced T1 mapping. J Am Coll Cardiol. 2008;52:1574–1580.
paragraph sign resonance imaging. MAGMA. 2003;16:17–20. 25. Ugander M, Oki AJ, Hsu LY, et al. Extracellular volume imaging by
5. Dweck MR, Joshi S, Murigu T, et al. Midwall fibrosis is an independent magnetic resonance imaging provides insights into overt and sub-
predictor of mortality in patients with aortic stenosis. J Am Coll Cardiol. clinical myocardial pathology. Eur Heart J. 2012;33:1268–1278.
2011;58:1271–1279. 26. Treibel TA, Fridman Y, Hackman B, et al. Extracellular volume associates
6. Maceira AM, Prasad SK, Hawkins PN, Roughton M, Pennell DJ. with outcomes more strongly than native or post-contrast myocardial
Cardiovascular magnetic resonance and prognosis in cardiac T1 (Abstract). Paper presented at: EuroCMR; 2016; Florence.
amyloidosis. J Cardiovasc Magn Reson. 2008;10:54. 27. Fontana M, Banypersad SM, Treibel TA, et al. Differential myocyte
7. Becker AE, Heijmans CD, Essed CE. Chronic non-ischaemic congestive responses in patients with cardiac transthyretin amyloidosis and
heart disease and endomyocardial biopsies. Worth the extra? Eur Heart J. light-chain amyloidosis: a cardiac MR imaging study. Radiology.
1991;12:218–223. 2015;277:388–397.
8. Brading AF, Jones AW. Distribution and kinetics of CoEDTA in smooth 28. Messroghli DR, Radjenovic A, Kozerke S, Higgins DM, Sivananthan MU,
muscle, and its use as an extracellular marker. J Physiol. Ridgway JP. Modified Look-Locker inversion recovery (MOLLI) for
1969;200:387–401. high-resolution T1 mapping of the heart. Magn Reson Med.
9. Poole-Wilson PA, Cameron IR. Intracellular pH and K+ of cardiac and 2004;52:141–146.
skeletal muscle in acidosis and alkalosis. Am J Physiol. 1975;229: 29. Piechnik SK, Ferreira VM, Dall’Armellina E, et al. Shortened modified
1305–1310. Look-Locker inversion recovery (ShMOLLI) for clinical myocardial
10. Kehr E, Sono M, Chugh SS, Jerosch-Herold M. Gadolinium-enhanced T1-mapping at 1.5 and 3 T within a 9 heartbeat breathhold. J Cardiovasc
magnetic resonance imaging for detection and quantification of fibrosis Magn Reson. 2010;12:69.
in human myocardium in vitro. Int J Cardiovasc Imaging. 2008;24:61–68. 30. Chow K, Flewitt JA, Green JD, Pagano JJ, Friedrich MG, Thompson RB.
11. Flett AS, Hayward MP, Ashworth MT, et al. Equilibrium contrast Saturation recovery single-shot acquisition (SASHA) for myocardial T
cardiovascular magnetic resonance for the measurement of diffuse mapping. Magn Reson Med. 2013.
myocardial fibrosis: preliminary validation in humans. Circulation. 31. Roujol S, Weingartner S, Foppa M, et al. Accuracy, precision, and
2010;122:138–144. reproducibility of four T1 mapping sequences: a head-to-head
12. White SK, Sado DM, Fontana M, et al. T1 mapping for myocardial comparison of MOLLI, ShMOLLI, SASHA, and SAPPHIRE. Radiology.
extracellular volume measurement by CMR: bolus only versus primed 2014;272:683–689.
infusion technique. JACC Cardiovasc Imaging. 2013. 32. Mehta BB, Chen X, Bilchick KC, Salerno M, Epstein FH. Accelerated and
13. Miller CA, Naish JH, Bishop P, et al. Comprehensive validation of navigator-gated Look-Locker imaging for cardiac T1 estimation
cardiovascular magnetic resonance techniques for the assessment of (ANGIE): development and application to T1 mapping of the right
myocardial extracellular volume. Circ Cardiovasc Imaging. 2013;6: ventricle. Magn Reson Med. 2015;73:150–160.
373–383. 33. Kvernby S, Warntjes MJ, Haraldsson H, Carlhall CJ, Engvall J, Ebbers T.
14. Coelho-Filho OR, Shah RV, Mitchell R, et al. Quantification of Simultaneous three-dimensional myocardial T1 and T2 mapping in one
cardiomyocyte hypertrophy by cardiac magnetic resonance: implications breath hold with 3D-QALAS. J Cardiovasc Magn Reson. 2014;16:102.
for early cardiac remodeling. Circulation. 2013;128:1225–1233. 34. Xue H, Greiser A, Zuehlsdorff S, et al. Phase-sensitive inversion recovery
15. Ugander M, Bagi PS, Oki AJ, et al. Myocardial edema as detected by for myocardial T1 mapping with motion correction and parametric
pre-contrast T1 and T2 CMR delineates area at risk associated with fitting. Magn Reson Med. 2013;69:1408–1420.
acute myocardial infarction. JACC Cardiovasc Imaging. 2012;5:596–603. 35. Kellman P, Hansen MS. T1-mapping in the heart: accuracy and
16. Karamitsos TD, Piechnik SK, Banypersad SM, et al. Noncontrast T1 precision. J Cardiovasc Magn Reson. 2014;16:2.
mapping for the diagnosis of cardiac amyloidosis. JACC Cardiovasc 36. Schelbert EB, Testa SM, Meier CG, et al. Myocardial extravascular
Imaging. 2013;6:488–497. extracellular volume fraction measurement by gadolinium
17. McDiarmid AK, Swoboda PP, Erhayiem B, et al. Athletic cardiac cardiovascular magnetic resonance in humans: slow infusion versus
adaptation in males is a consequence of elevated myocyte mass. Circ bolus. J Cardiovasc Magn Reson. 2011;13:16.
Cardiovasc Imaging. 2016;9. 37. White SK, Sado DM, Fontana M, et al. T1 mapping for myocardial
18. Sado DM, Maestrini V, Piechnik SK, et al. Noncontrast myocardial T extracellular volume measurement by CMR: bolus only versus primed
mapping using cardiovascular magnetic resonance for iron overload. infusion technique. JACC Cardiovasc Imaging. 2013;6:955–962.
J Magn Reson Imaging. 2014. 38. McDiarmid AK, Swoboda PP, Erhayiem B, et al. Single bolus versus split
19. Sado DM, White SK, Piechnik SK, et al. Identification and assessment of dose gadolinium administration in extra-cellular volume calculation at 3
Anderson-Fabry disease by cardiovascular magnetic resonance Tesla. J Cardiovasc Magn Reson. 2015;17:6.
noncontrast myocardial T1 mapping. Circ Cardiovasc Imaging. 39. Kellman P, Wilson JR, Xue H, Ugander M, Arai AE. Extracellular volume
2013;6:392–398. fraction mapping in the myocardium, part 1: evaluation of an
20. Dall’Armellina E, Piechnik SK, Ferreira VM, et al. Cardiovascular automated method. J Cardiovasc Magn Reson. 2012;14:63.
magnetic resonance by non contrast T1-mapping allows assessment of 40. Varga-Szemes A, van der Geest RJ, Spottiswoode BS, et al. Myocardial
severity of injury in acute myocardial infarction. J Cardiovasc Magn late gadolinium enhancement: accuracy of T1 mapping-based synthetic
Reson. 2012;14:15. inversion-recovery imaging. Radiology. 2016;278:374–382.
399.e2 SECTION III  Functional Disease

41. Hamilton JI, Jiang Y, Chen Y, et al. MR fingerprinting for rapid 61. Fontana M, White SK, Banypersad SM, et al. Comparison of T1
quantification of myocardial T, T, and proton spin density. Magn Reson mapping techniques for ECV quantification. Histological validation and
Med. 2016. reproducibility of ShMOLLI versus multibreath-hold T1 quantification
42. Moon JC, Messroghli DR, Kellman P, et al; Cardiovascular Magnetic equilibrium contrast CMR. J Cardiovasc Magn Reson. 2012;14:88.
Resonance Working Group of the European Society of C. Myocardial T1 62. Sparrow P, Amirabadi A, Sussman MS, Paul N, Merchant N.
mapping and extracellular volume quantification: a Society for Quantitative assessment of myocardial T2 relaxation times in cardiac
Cardiovascular Magnetic Resonance (SCMR) and CMR Working Group amyloidosis. J Magn Reson Imaging. 2009;30:942–946.
of the European Society of Cardiology consensus statement. J Cardiovasc 63. Wassmuth R, Abdel-Aty H, Bohl S, Schulz-Menger J. Prognostic impact
Magn Reson. 2013;15:92. of T2-weighted CMR imaging for cardiac amyloidosis. Eur Radiol.
43. Robson MD, Piechnik SK, Tunnicliffe EM, Neubauer S. T measurements 2011;21:1643–1650.
in the human myocardium: the effects of magnetization transfer on the 64. Banypersad SM, Fontana M, Maestrini V, et al. T1 mapping and survival
SASHA and MOLLI sequences. Magn Reson Med. 2013. in systemic light-chain amyloidosis. Eur Heart J. 2015;36:244–251.
44. Coelho-Filho OR, Mongeon FP, Mitchell R, et al. Role of 65. O’Mahony C, Elliott P. Anderson-Fabry disease and the heart. Prog
transcytolemmal water-exchange in magnetic resonance measurements Cardiovasc Dis. 2010;52:326–335.
of diffuse myocardial fibrosis in hypertensive heart disease. Circ 66. Sado DM, White SK, Piechnik SK, et al. Identification and assessment of
Cardiovasc Imaging. 2013;6:134–141. Anderson-Fabry disease by cardiovascular magnetic resonance
45. Petersen SE, Matthews PM, Bamberg F, et al. Imaging in population noncontrast myocardial T1 mapping. Circ Cardiovasc Imaging.
science: cardiovascular magnetic resonance in 100,000 participants of 2013;6:392–398.
UK Biobank—rationale, challenges and approaches. J Cardiovasc Magn 67. Thompson RB, Chow K, Khan A, et al. T(1) mapping with
Reson. 2013;15:46. cardiovascular MRI is highly sensitive for Fabry disease independent of
46. Scholz TD, Martins JB, Skorton DJ. NMR relaxation times in acute hypertrophy and sex. Circ Cardiovasc Imaging. 2013;6:637–645.
myocardial infarction: relative influence of changes in tissue water and 68. Pica S, Sado DM, Maestrini V, et al. Reproducibility of native myocardial
fat content. Magn Reson Med. 1992;23:89–95. T1 mapping in the assessment of Fabry disease and its role in early
47. Abdel-Aty H, Zagrosek A, Schulz-Menger J, et al. Delayed enhancement detection of cardiac involvement by cardiovascular magnetic resonance.
and T2-weighted cardiovascular magnetic resonance imaging J Cardiovasc Magn Reson. 2014;16:99.
differentiate acute from chronic myocardial infarction. Circulation. 69. Nordin S, Kozor R, Bulluck H, et al. Cardiac Fabry disease with late
2004;109:2411–2416. gadolinium enhancement is a chronic inflammatory cardiomyopathy
48. Thavendiranathan P, Walls M, Giri S, et al. Improved detection of evidence from multi parametric mapping by cardiovascular magnetic
myocardial involvement in acute inflammatory cardiomyopathies using resonance. J Am Coll Cardiol. 2016;68:1707–1708.
T2 mapping. Circ Cardiovasc Imaging. 2012;5:102–110. 70. Carpenter JP, He T, Kirk P, et al. On T2* magnetic resonance and cardiac
49. Giri S, Shah S, Xue H, et al. Myocardial T(2) mapping with respiratory iron. Circulation. 2011;123:1519–1528.
navigator and automatic nonrigid motion correction. Magn Reson Med. 71. Wood JC, Otto-Duessel M, Aguilar M, et al. Cardiac iron determines
2012;68:1570–1578. cardiac T2*, T2, and T1 in the gerbil model of iron cardiomyopathy.
50. van Heeswijk RB, Feliciano H, Bongard C, et al. Free-breathing 3 T Circulation. 2005;112:535–543.
magnetic resonance T2-mapping of the heart. JACC Cardiovasc Imaging. 72. Alam MH, He T, Auger D, et al. Validation of T2* in-line analysis for
2012;5:1231–1239. tissue iron quantification at 1.5 T. J Cardiovasc Magn Reson. 2016;18:23.
51. Wassmuth R, Prothmann M, Utz W, et al. Variability and homogeneity 73. Abdel-Gadir A, Vorasettakarnkij Y, Ngamkasem H, et al. Ultrafast CMR to
of cardiovascular magnetic resonance myocardial T2-mapping in deliver high volume screening of an at risk thalassemia population in the
volunteers compared to patients with edema. J Cardiovasc Magn Reson. developing world: preliminary results from the TIC-TOC study (Thailand
2013;15:27. and UK international collaboration in thalassaemia using an optimised
52. Friedrich MG, Sechtem U, Schulz-Menger J, et al; International ultrafast CMR protocol). J Cardiovasc Magn Reson. 2016;18:1–2.
Consensus Group on Cardiovascular Magnetic Resonance in 74. Alam MH, Auger D, Smith GC, et al. T1 at 1.5T and 3T compared with
Myocarditis. Cardiovascular magnetic resonance in myocarditis: a JACC conventional T2* at 1.5T for cardiac siderosis. J Cardiovasc Magn Reson.
white paper. J Am Coll Cardiol. 2009;53:1475–1487. 2015;17:102.
53. Lurz P, Luecke C, Eitel I, et al. Comprehensive cardiac magnetic 75. Fernandes JL, Siqueira MHA, Nobrega de Oliveira KT, et al. Use of an
resonance imaging in patients with suspected myocarditis: the accelerated protocol for rapid analysis of iron overload in the heart and
MyoRacer-Trial. J Am Coll Cardiol. 2016;67:1800–1811. liver: the All Iron Detected (AID) Multicenter Study. J Cardiovasc Magn
54. Bonner F, Janzarik N, Jacoby C, et al. Myocardial T2 mapping reveals Reson. 2015;17:O62.
age- and sex-related differences in volunteers. J Cardiovasc Magn Reson. 76. Higgins CB, Herfkens R, Lipton MJ, et al. Nuclear magnetic resonance
2015;17:9. imaging of acute myocardial infarction in dogs: alterations in magnetic
55. Sado DM, Flett AS, Banypersad SM, et al. Cardiovascular magnetic relaxation times. Am J Cardiol. 1983;52:184–188.
resonance measurement of myocardial extracellular volume in health 77. Ferreira VM, Piechnik SK, Dall’armellina E, et al. T Mapping for the
and disease. Heart. 2012;98:1436–1441. diagnosis of acute myocarditis using CMR: comparison to T-weighted
56. Liu CY, Liu YC, Wu C, et al. Evaluation of age related interstitial and late gadolinium enhanced imaging. JACC Cardiovasc Imaging. 2013.
myocardial fibrosis with cardiac magnetic resonance contrast-enhanced 78. Puntmann VO, D’Cruz D, Smith Z, et al. Native myocardial T1 mapping
T1 mapping in the multi-ethnic study of atherosclerosis (MESA). J Am by cardiovascular magnetic resonance imaging in subclinical
Coll Cardiol. 2013. cardiomyopathy in patients with systemic lupus erythematosus. Circ
57. Abdel-Gadir A, Berber R, Porter JB, et al. Detection of metallic cobalt Cardiovasc Imaging. 2013;6:295–301.
and chromium liver deposition following failed hip replacement using 79. Ntusi NA, Piechnik SK, Francis JM, et al. Subclinical myocardial
T2* and R2 magnetic resonance. J Cardiovasc Magn Reson. 2016;18:29. inflammation and diffuse fibrosis are common in systemic sclerosis—a
58. Richards DB, Cookson LM, Berges AC, et al. Therapeutic clearance of clinical study using myocardial T1-mapping and extracellular volume
amyloid by antibodies to serum amyloid P component. N Engl J Med. quantification. J Cardiovasc Magn Reson. 2014;16:21.
2015;373:1106–1114. 80. Ntusi NA, Piechnik SK, Francis JM, et al. Diffuse myocardial fibrosis and
59. Gillmore JD, Maurer MS, Falk RH, et al. Non-biopsy diagnosis of inflammation in rheumatoid arthritis: insights from CMR T1 mapping.
cardiac transthyretin amyloidosis. Circulation. 2016;133:2404–2412. JACC Cardiovasc Imaging. 2015;8:526–536.
60. Fontana M, Pica S, Reant P, et al. Prognostic value of late gadolinium 81. Kellman P, Wilson JR, Xue H, et al. Extracellular volume fraction
enhancement cardiovascular magnetic resonance in cardiac amyloidosis. mapping in the myocardium, part 2: initial clinical experience.
Circulation. 2015;132:1570–1579. J Cardiovasc Magn Reson. 2012;14:64.
CHAPTER 32  T1 and T2 Mapping and Extracellular Volume in Cardiomyopathy 399.e3

82. Sado DM, Kozor R, Corr L, Moon JC. Global myocardial edema in 102. Dass S, Suttie JJ, Piechnik SK, et al. Myocardial tissue characterization
antisynthetase syndrome detected by cardiovascular magnetic resonance using magnetic resonance noncontrast T1 mapping in hypertrophic and
mapping techniques. Circulation. 2016;133:e25–e26. dilated cardiomyopathy. Circ Cardiovasc Imaging. 2012;5:726–733.
83. Diez J, Querejeta R, Lopez B, Gonzalez A, Larman M, Martinez Ubago 103. Schelbert EB, Moon JC. Exploiting differences in myocardial
JL. Losartan-dependent regression of myocardial fibrosis is associated compartments with native T1 and extracellular volume fraction for the
with reduction of left ventricular chamber stiffness in hypertensive diagnosis of hypertrophic cardiomyopathy. Circ Cardiovasc Imaging.
patients. Circulation. 2002;105:2512–2517. 2015;8.
84. Izawa H, Murohara T, Nagata K, et al. Mineralocorticoid receptor 104. Wong TC, Chung J, Kellman P, Schelbert EB. Extracellular volume
antagonism ameliorates left ventricular diastolic dysfunction and fraction is associated with B-type natriuretic peptide in hypertrophic
myocardial fibrosis in mildly symptomatic patients with idiopathic cardiomyopathy. J Cardiovasc Magn Reson. 2014;16:1–2.
dilated cardiomyopathy: a pilot study. Circulation. 2005;112:2940–2945. 105. Ho CY, Abbasi SA, Neilan TG, et al. T1 measurements identify
85. Metra M, Cotter G, Davison BA, et al; RELAX-AHF Investigators. Effect extracellular volume expansion in hypertrophic cardiomyopathy
of serelaxin on cardiac, renal, and hepatic biomarkers in the Relaxin in sarcomere mutation carriers with and without left ventricular
Acute Heart Failure (RELAX-AHF) development program: correlation hypertrophy. Circ Cardiovasc Imaging. 2013;6:415–422.
with outcomes. J Am Coll Cardiol. 2013;61:196–206. 106. Lee SP, Lee W, Lee JM, et al. Assessment of diffuse myocardial fibrosis by
86. Wong TC, Piehler K, Meier CG, et al. Association between extracellular using MR imaging in asymptomatic patients with aortic stenosis.
matrix expansion quantified by cardiovascular magnetic resonance and Radiology. 2015;274:359–369.
short-term mortality. Circulation. 2012;126:1206–1216. 107. de Meester de Ravenstein C, Bouzin C, Lazam S, et al. Histological
87. Wong TC, Piehler KM, Kang IA, et al. Myocardial extracellular volume Validation of measurement of diffuse interstitial myocardial fibrosis by
fraction quantified by cardiovascular magnetic resonance is increased in myocardial extravascular volume fraction from modified Look-Locker
diabetes and associated with mortality and incident heart failure imaging (MOLLI) T1 mapping at 3 T. J Cardiovasc Magn Reson.
admission. Eur Heart J. 2014;35:657–664. 2015;17:48.
88. Mascherbauer J, Marzluf BA, Tufaro C, et al. Cardiac magnetic 108. Verhaert D, Thavendiranathan P, Giri S, et al. Direct T2 quantification of
resonance postcontrast T1 time is associated with outcome in patients myocardial edema in acute ischemic injury. JACC Cardiovasc Imaging.
with heart failure and preserved ejection fraction. Circ Cardiovasc 2011;4:269–278.
Imaging. 2013;6:1056–1065. 109. Ugander M, Bagi PS, Oki AJ, et al. Myocardial edema as detected by
89. Schelbert EB, Piehler KM, Zareba KM, et al. Myocardial fibrosis pre-contrast T1 and T2 CMR delineates area at risk associated with
quantified by extracellular volume is associated with subsequent acute myocardial infarction. JACC Cardiovasc Imaging. 2012;5:
hospitalization for heart failure, death, or both across the spectrum of 596–603.
ejection fraction and heart failure stage. J Am Heart Assoc. 2015;4. 110. Chin CW, Semple S, Malley T, et al. Optimization and comparison of
90. Hinojar R, Varma N, Child N, et al. T1 Mapping in discrimination of myocardial T1 techniques at 3T in patients with aortic stenosis. Eur
hypertrophic phenotypes: hypertensive heart disease and hypertrophic Heart J Cardiovasc Imaging. 2014;15:556–565.
cardiomyopathy: findings from the International T1 Multicenter 111. Singh A, Horsfield MA, Bekele S, Khan JN, Greiser A, McCann GP.
Cardiovascular Magnetic Resonance Study. Circ Cardiovasc Imaging. Myocardial T1 and extracellular volume fraction measurement in
2015;8. asymptomatic patients with aortic stenosis: reproducibility and
91. Treibel TA, Zemrak F, Sado DM, et al. Extracellular volume comparison with age-matched controls. Eur Heart J Cardiovasc Imaging.
quantification in isolated hypertension—changes at the detectable 2015;16:763–770.
limits? J Cardiovasc Magn Reson. 2015;17:74. 112. Bohnen S, Radunski UK, Lund GK, et al. Performance of t1 and t2
92. Kuruvilla S, Janardhanan R, Antkowiak P, et al. Increased extracellular mapping cardiovascular magnetic resonance to detect active myocarditis
volume and altered mechanics are associated with LVH in hypertensive in patients with recent-onset heart failure. Circ Cardiovasc Imaging.
heart disease, not hypertension alone. JACC Cardiovasc Imaging. 2015;8.
2015;8:172–180. 113. Su M-YM, Lin L-Y, Tseng Y-HE, et al. CMR-verified diffuse myocardial
93. Rosenhek R, Zilberszac R, Schemper M, et al. Natural history of very fibrosis is associated with diastolic dysfunction in HFpEF. JACC
severe aortic stenosis. Circulation. 2010;121:151–156. Cardiovasc Imaging. 2014;7:991–997.
94. Steell G. Text-book on Diseases of the Heart. Manchester: Manchester 114. Ho CY, Ho CY, Abbasi SA, et al. T1 Measurements identify extracellular
University Press; 1906. volume expansion in hypertrophic cardiomyopathy sarcomere mutation
95. Milano AD, Faggian G, Dodonov M, et al. Prognostic value of carriers with and without left ventricular hypertrophy. Circ Cardiovasc
myocardial fibrosis in patients with severe aortic valve stenosis. J Thorac Imaging. 2013;6:415–422.
Cardiovasc Surg. 2012;144:830–837. 115. Barison A, Del Torto A, Chiappino S, et al. Prognostic significance of
96. Bull S, White SK, Piechnik SK, et al. Human non-contrast T1 values and myocardial extracellular volume fraction in nonischaemic dilated
correlation with histology in diffuse fibrosis. Heart. 2013. cardiomyopathy. J Cardiovasc Med (Hagerstown). 2015;16:681–687.
97. Flett AS, Sado DM, Quarta G, et al. Diffuse myocardial fibrosis in severe 116. Lee KH, Park HS, Park CH, et al. Extracellular volume imaging and
aortic stenosis: an equilibrium contrast cardiovascular magnetic quantitative T2 mapping for the diagnosis of mitochondrial
resonance study. Eur Heart J Cardiovasc Imaging. 2012;13:819–826. cardiomyopathy. Circulation. 2014;130:1832–1834.
98. Treibel TA, Fontana M, Kozor R, et al. Diffuse myocardial fibrosis - a 117. Wong TC, Piehler KM, Kang IA, et al. Myocardial extracellular volume
therapeutic target? Proof of regression at 1-year following aortic valve fraction quantified by cardiovascular magnetic resonance is increased in
replacement: the RELIEF-AS study. J Cardiovasc Magn Reson. diabetes and associated with mortality and incident heart failure
2016;18:1–3. admission. Eur Heart J. 2013.
99. Mewton N, Liu CY, Croisille P, Bluemke D, Lima JA. Assessment of 118. Shah RV, Abbasi SA, Neilan TG, et al. Myocardial tissue remodeling in
myocardial fibrosis with cardiovascular magnetic resonance. J Am Coll adolescent obesity. J Am Heart Assoc. 2013;2:e000279.
Cardiol. 2011;57:891–903. 119. Plymen CM, Sado DM, Taylor AM, et al. Diffuse myocardial fibrosis in
100. Sibley CT, Noureldin RA, Gai N, et al. T1 Mapping in cardiomyopathy the systemic right ventricle of patients late after Mustard or Senning
at cardiac MR: comparison with endomyocardial biopsy. Radiology. surgery: an equilibrium contrast cardiovascular magnetic resonance
2012;265:724–732. study. Eur Heart J Cardiovasc Imaging. 2013;14:963–968.
101. Puntmann VO, Voigt T, Chen Z, et al. Native T1 mapping in 120. Barison A, Gargani L, De Marchi D, et al. Early myocardial and skeletal
differentiation of normal myocardium from diffuse disease in muscle interstitial remodelling in systemic sclerosis: insights from
hypertrophic and dilated cardiomyopathy. JACC Cardiovasc Imaging. extracellular volume quantification using cardiovascular magnetic
2013;6:475–484. resonance. Eur Heart J Cardiovasc Imaging. 2015;16:74–80.
399.e4 SECTION III  Functional Disease

121. Hinojar R, Foote L, Arroyo Ucar E, et al. Native T1 in discrimination of 128. Alam MH, Auger D, Smith GC, et al. T1 at 1.5T and 3T compared with
acute and convalescent stages in patients with clinical diagnosis of conventional T2* at 1.5T for cardiac siderosis. J Cardiovasc Magn Reson.
myocarditis: a proposed diagnostic algorithm using CMR. JACC 2015;17:102.
Cardiovasc Imaging. 2015;8:37–46. 129. Pedersen SF, Thrysoe SA, Robich MP, et al. Assessment of
122. Thavendiranathan P, Walls M, Giri S, et al. Improved detection of intramyocardial hemorrhage by T1-weighted cardiovascular magnetic
myocardial involvement in acute inflammatory cardiomyopathies using resonance in reperfused acute myocardial infarction. J Cardiovasc Magn
t2 mapping. Circ Cardiovasc Imaging. 2012;5. Reson. 2012;14:59.
123. Ertel A, Pratt D, Kellman P, et al. Increased myocardial extracellular 130. Edwards NC, Moody WE, Chue CD, Ferro CJ, Townend JN, Steeds RP.
volume in active idiopathic systemic capillary leak syndrome. Defining the natural history of uremic cardiomyopathy in chronic
J Cardiovasc Magn Reson. 2015;17:1–8. kidney disease: the role of cardiovascular magnetic resonance. JACC
124. Miller CA, Naish JH, Shaw SM, et al. Multiparametric cardiovascular Cardiovasc Imaging. 2014;7:703–714.
magnetic resonance surveillance of acute cardiac allograft rejection and 131. Tham EB, Haykowsky MJ, Chow K, et al. Diffuse myocardial fibrosis by
characterisation of transplantation-associated myocardial injury: a pilot T1-mapping in children with subclinical anthracycline cardiotoxicity:
study. J Cardiovasc Magn Reson. 2014;16:52. relationship to exercise capacity, cumulative dose and remodeling.
125. Fontana M, Banypersad SM, Treibel TA, et al. Native T1 mapping in J Cardiovasc Magn Reson. 2013;15:1–11.
transthyretin amyloidosis. JACC Cardiovasc Imaging. 2014;7:157–165. 132. Kammerlander AA, Marzluf BA, Zotter-Tufaro C, et al. T1 mapping by
126. Thompson RB, Chow K, Khan A, et al. T(1) mapping with CMR imaging: from histological validation to clinical implication. JACC
cardiovascular MRI is highly sensitive for Fabry disease independent of Cardiovasc Imaging. 2016;9:14–23.
hypertrophy and sex. Circ Cardiovasc Imaging. 2013;6:637–645.
127. Hanneman K, Nguyen ET, Thavendiranathan P, et al. Quantification of
myocardial extracellular volume fraction with cardiac MR imaging in
thalassemia major. Radiology. 2015;150341.
33 
Cardiac Iron Loading and Myocardial T2*
Mark A. Westwood and Dudley J. Pennell

CONDITIONS ASSOCIATED WITH CARDIAC to regular chelation therapy from a young age.6,8,9 Improving survival
with deferoxamine by later birth cohort has been confirmed in other
IRON LOADING
countries.9,10 In the current millennium, data using T2* from TM
There are several conditions that can potentially lead to cardiac iron patients across the world show that cardiac iron overload continues
loading with cardiac complications. Cardiac iron loading can occur via to be common, using definitions from T2* cardiovascular magnetic
two distinct mechanisms: first, primary disruption of iron regulation resonance (CMR) of severe cardiac iron loading of <10 ms, and mild-
(genetic hemochromatosis syndromes) where excess gastrointestinal to-moderate cardiac iron loading of 10 to 20 ms (Table 33.1). The
absorption is slow and symptoms often present in middle age; and, prevalence of cardiomyopathy is more difficult to measure. It can be
second, via transfusional iron overload where the iron accumulation estimated from either the prevalence of clinical heart failure or the pres-
rate is much faster and children can develop cardiac iron accumulation ence of detectable left ventricular (LV) dysfunction, with the latter being
within a few years of initiating regular transfusion therapy. Excess iron higher in any given population. Prevalence generally increases with age
cannot be naturally excreted in humans because of the lack of such a and decreases with a more recent year of birth. For example, in a cohort
regulatory excretory pathway. of patients born before 1976, 37% had heart disease as defined by need
The most common pathology leading to severe iron accumulation for inotropic or antiarrhythmic medications,6 but in a survey in 2004
is thalassemia major (TM). This is a genetic condition with severe the number of TM patients of all ages receiving cardiac medication
reduction or absent production of the beta-globin chain constituent was only 10%.
of hemoglobin A (HbA). Excess alpha-chains result from ineffective
erythropoiesis, and there is a life-threatening anemia by 2 years of age. MECHANISMS OF CARDIAC IRON ENTRY
Not only does this require treatment with regular, lifelong, repeated
blood transfusions, but it is compounded by a mild increase in gastro-
AND TOXICITY
intestinal iron uptake related to hepcidin suppression.1 TM occurs The mechanism of human cardiac iron loading is incompletely under-
predominantly where malaria is endemic because heterozygote genetic stood. In vitro and animal studies suggest cardiac entry of iron is
hemoglobin mutations confer resistance. In other parts of the world it mediated by calcium channels,11–14 and indeed nifedipine hinders iron
occurs primarily through immigration. uptake into cardiac cells.15 Accumulation of myocardial iron eventually
The detail of the hemoglobinopathy plays a role in the pattern and leads to increased levels of intracellular free iron. The toxicity of free
pathology of transfusion-dependent iron loading. For example, in the iron is mediated via several mechanisms (Fig. 33.1)16,17: (1) membrane
less severe condition of thalassemia intermedia, pulmonary hypertension damage caused by lipid peroxidation; (2) mitochondrial damage and
and thrombosis play a greater clinical role and iron loading occurs at perturbation of the respiratory enzyme chain18,19; (3) altered electri-
a later age.2,3 In sickle cell disease (SCD), the defining clinical features cal function, including ryanodine release channel interference20,21; (4)
include sickle cell crises (intermittent attacks of severe pain), pulmo- cardiac fibrosis, which was reported as prominent in early autopsy
nary hypertension, thrombosis, and stroke.4 Even where SCD patients studies22,23; and (5) changes in gene expression.24 Iron that is safely
are transfused to prevent cardiovascular complications, extrahepatic stored in ferritin or hemosiderin is nontoxic, yielding hearts with low
iron deposition is lower compared with other transfusional anemias. T2* and normal function. However, high iron stores predispose patients
Conversely, in Diamond-Blackfan anemia, where the intrinsic bone to development of cardiac dysfunction in the future. The result is that
marrow activity is low, patients are very prone to nonhepatic tissue the natural history and clinical course in untreated patients is one of a
iron accumulation. long asymptomatic phase of progressive myocardial iron accumulation
with sudden onset of either malignant arrhythmias or acutely impaired
PREVALENCE OF CARDIAC IRON OVERLOAD myocardial function in early adulthood. There was a significant mortal-
ity once symptomatic,5,25 and 5-year survival for patients presenting in
AND CARDIOMYOPATHY heart failure was only 48%.26 One factor behind this is that although
The introduction of chelation therapy in the 1960s with deferoxamine evidence suggested that intensive iron chelation therapy could completely
changed the natural progression of TM patients receiving regular trans- restore cardiac function in most patients with asymptomatic cardiac
fusions, where the most common cause of death was heart failure at dysfunction and even some clinical heart failure,27–29 there was a risk
a very young age.5 Despite initially promising data and an increased of relapse caused by a lag between the clearance of cardiac iron and
life expectancy with the introduction of deferoxamine, cardiac iron improvements in systolic function.27–29 Even in asymptomatic patients,
overload continued to dominate, accounting for 70% of deaths.6,7 treatment of severe cardiac siderosis by aggressive chelation therapy is
However, as shown in a UK cohort, by the year 2000 the median age associated with improvements in ventricular function.30 Improved life
at death was 35 years because by this time patients had been exposed expectancy for TM patients in the United Kingdom has resulted from

400
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 401

TABLE 33.1  Incidence of Cardiac Iron Overload From Published Data Around the World
Severe Mild-to-Moderate Normal
Country Sample Size T2* <10 ms T2* 10–20 ms T2* >20 ms
United Kingdom47 109 20% 43% 37%
Hong Kong145 180 26% 24% 50%
Turkey146 28 46% 39% 14%
Australia147 30 37% 27% 37%
Oman148 81 24% 22% 54%
United States of America149 141 13% 21% 66%
Italy150 167 13% (<8 ms) 52% (8–20 ms) 35%
Italy151 220 30% <20 ms 66%
Greece152 159 68% <20 ms 32%
Worldwide survey153 3445 20% 22% 58%
Egypt154 89 25% <20 ms 75%
Pakistan155 83 47% 16% 37%
Indonesia156 162 5% 10% 85%

Modified from Pennell DJ, Udelson JE, Arai AE, et al; American Heart Association Committee on Heart Failure and Transplantation of the
Council on Clinical Cardiology and Council on Cardiovascular Radiology and Imaging. Cardiovascular function and treatment in β-thalassemia
major: a consensus statement from the American Heart Association. Circulation. 2013;128:281–308.

Fecal

60% 90%
40%
Urine DFO DFX Antioxidants

DFP Fe LPI

ROS
Lipid
Fe LCI Fe OH• peroxidation

O2÷ Oxidation Base


H2O2 CO, methionine oxidation
O2 and tyrosine
ROI are normally converted
to water by resident Lysosomes
enzymes SOD and GPX
Mitochondrion Protein DNA

Endoplasmic reticulum
FIG. 33.1  Amelioration of free iron species (labile plasma iron [LPI] and labile cell iron [LCI]) by iron chela-
tors and antioxidants. LPI is penetrating through the cell membrane with a consequent accumulation of
LCI. Both LPI and LCI react with reactive oxygen intermediate (ROI) producing noxious reactive oxygen
species (ROS), for example, OH’ radicals, which are highly reactive and oxidize DNA, proteins, and lipid
components of the cell. Deferiprone (DFP) chelates LCI alone or in combination with LPI by deferoxamine
(DFO). Deferasirox (DFX) mainly removes LPI. (From Rachmilewitz EA, Giardina PJ. How I treat thalassemia.
Blood. 2011;118:3479–3488.)

the increasing availability of T2* CMR and earlier escalation of therapy.31 hypoparathyroidism. This suggests a common or similar iron uptake
Even with modern-day chelation regimens, it is important to avoid mechanism. In untreated cardiac iron accumulation, historical series also
stage IV heart failure in thalassemia as in-hospital mortality remains in showed more cardiac complications, such as pericarditis and myocarditis,
excess of 50%. that occurred in addition to heart failure and arrhythmias.5,32 Chelation
Of significant clinical interest is that cardiac iron loading is com- seems to have has altered this, with myocarditis and pericarditis now
monly associated with endocrinologic complications including hypo- being unusual. Chelation has also led to a significant reduction in the
thyroidism, diabetes, hypoadrenalism, growth hormone deficiency, and historical occurrence of dense replacement fibrosis of myocytes, even in
402 SECTION III  Functional Disease

those who die from heart failure, although occasional small patches of for cine imaging with end-expiratory breath-hold are acquired in the
fibrosis may be present.5,23,32–34 In the modern era of chelation therapy, four-chamber and two-chamber long-axis views along with contigu-
dilated cardiomyopathy (with restrictive features) and arrhythmia (pre- ous short-axis cines from the atrioventricular ring to the apex. The
dominantly atrial fibrillation) persist. Ventricular arrhythmias are more optimal end-diastolic volume (EDV) and end-systolic volume (ESV)
common in severe cardiac iron loading. Iron deposition throughout the are derived from LV volume/time curves generated from all frames of
myocardium in TM patients is nonuniform, being preferentially depos- all cines and should include systolic valve tracking to adjust for systolic
ited in the subepicardium, but no systematic variation occurs between valve descent. LV mass should be calculated using the epicardial and
myocardial regions such that iron deposition in the interventricular endocardial borders of the LV in the end-diastolic frame. Papillary
septum is highly representative of total cardiac iron. There are other muscles should be included in the LV mass and excluded from the LV
cardiac changes such as decreased left atrial (LA) function,35 impaired volumes. This method of volumetric assessment then has the benefit of
right ventricular (RV) function,36 impaired diastolic function,37–39 and having recognized normalized values for gender, body surface area, and
impaired endothelial function.40–42 age for both the LV,55 and the RV.56 These covariates have substantial
impact on the normal ranges. Even where this stringent analysis method
ASSESSMENT OF THE HEART IN is not possible, CMR is more reproducible than other techniques, par-
ticularly where patients can be followed over time to allow longitudinal
THALASSEMIA MAJOR analysis of data.57,58
New-onset electrocardiogram (ECG) abnormalities are usually evident Consequent to the low hemoglobin from anemia, an adaptive response
in TM patients with heart failure43 and may include supraventricular leads to increased cardiac output, usually through an increase in EDV,
arrhythmias, ECG findings suggesting right heart involvement, new- stroke volume, and heart rate. TM therefore represents a chronic, high-
onset T-wave inversion, and small QRS complexes. In patients without output state produced by volume-loaded ventricles (high preload) with
heart failure, an abnormal ECG was found in 46%.44 It is not known the ejection fraction increased caused by a decreased afterload and an
if progressive alterations in ECG tracings occur before heart failure increased preload. Because of this state, the appropriate normal ranges
develops, casting doubt over the use of serial ECG monitoring to assess to use are those derived from nonanemic TM patients because for both
disease progress. A chest x-ray may show cardiomegaly, signs of conges- the LV59 and the RV60 there are differences compared with standard
tive heart failure, and, on occasion, extramedullary hematopoiesis. B-type normal cohorts. Pediatric normal ranges are also available.61,62 The
natriuretic peptide is significantly increased in documented LV diastolic hyperdynamic circulation also leads to an increased LV mass. The same
dysfunction, which may be helpful.30,45,46 Exercise stress testing may be pattern is true for the RV indices, but the differences are not as marked.60
able to unmask subclinical LV dysfunction in TM, but results are influ- Using these adjusted “normal for TM” ranges may enhance diagnostic
enced by many other factors, and it seems to have questionable value. accuracy for detection of cardiomyopathy.
Before the development of CMR-derived T2*,47 cardiac function Whereas CMR offers several other techniques such as late gadolinium
was the key marker of disease progression despite a number of factors enhancement for the detection of myocardial fibrosis and myocarditis,
limiting its use. The limitations and reproducibility of echocardiography and myocardial perfusion imaging for coronary artery disease, these
around operator experience and acoustic windows are well documented48 are uncommon findings in this group of patients and routine cardiac
but should be tempered against the widespread, safe, economical, and assessment with these techniques is not advocated.63 Although pos-
routinely available nature of this test. Before the advent of three- sible, diastolic function using CMR is time consuming and more opera-
dimensional (3D) techniques, the need to make geometric assumptions tor dependent and in clinical practice is more readily assessed using
for the quantification of volumes and mass, which often break down echocardiography.64,65
in abnormal remodeled ventricles, led to significant interobserver vari-
ability.49 Despite this, impaired resting LV function by echocardiography CARDIOVASCULAR MAGNETIC RESONANCE OF
correlated with increased cardiac mortality over 12 years.50 Echocar-
diography provides less accurate quantification than CMR, and accuracy
CARDIAC IRON USING T2*
decreases with worsening LV function as geometric assumptions become In simple terms, radiofrequency (RF) pulses cause spinning protons to
an ever-greater approximation. Echocardiography is the preferred second- be deflected from their equilibrium state. Immediately after this RF
line technique after CMR. It is important that echo is done in experienced pulse, protons begin to return to equilibrium by two separate processes:
centers used to scanning large numbers of TM patients.51 namely, T1 recovery and T2 relaxation. Magnetic field inhomogeneity
Although radionuclide ventriculography during exercise can detect accelerates these processes whereby the protons return to equilibrium.
preclinical myocardial dysfunction in patients with iron overload,52 As myocardial stores increase in the heart, ferritin breakdown increases
concerns over radiation dose in young people, considerable intercenter into particulate hemosiderin, which is a form of ferrihydrite (hydrated
variation, and the availability of other techniques have limited the use iron oxide). The hemosiderin to ferritin ratio is significantly higher in
of this technique. Similarly, cardiac computed tomography (CT) can cardiac siderosis than in normal hearts.66 It is this particulate intracel-
assess cardiac function,53 and to some extent liver iron concentration,54 lular iron that markedly increases magnetic field inhomogeneity and
but use of this technique is also limited because of concerns over repeated thereby accelerates both T1 and T2 relaxation. Because the influence
ionizing radiation exposure. on these parameters as a result of particulate iron is much greater than
CMR is noninvasive, reliable, and free of ionizing radiation. Although normal field inhomogeneity, changes in these parameters become a
increasingly seen as a mainstream cardiology investigation, the widespread biomarker for myocardial iron deposition. Magnetic resonance (MR)
use of CMR is limited by the cost and claustrophobia, although the latter relaxometry is the use of specific MR sequences that repeatedly sample
is less of an issue with new-generation wide-bore scanners. The limitation image data every few milliseconds from which relaxation or recovery
with implanted cardiac devices is progressively being overcome. For the curves can be derived. These techniques do not require contrast media.
assessment of LV and RV function, the lack of geometric assumptions Given that it is increased field inhomogeneity which is affected by
optimizes accuracy and reproducibility. Indeed, CMR is widely accepted increased iron deposition, it follows that the sequence most sensitive
as the gold standard for all biventricular measurements and indices. to this parameter should be chosen. Gradient echo sequences are much
Routine use of steady-state free precession (SSFP)-based acquisitions more susceptible to field inhomogeneity than spin echo sequences and
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 403

B
FIG. 33.2  T2* multiecho images. (A) A single breath-hold was used and (B) multiple breath-holds were used
to acquire images of the myocardium with increasing echo time (TE). Note the better registration of the
position of the diaphragm with the breath-hold technique. The signal intensity of the myocardium falls as
the TE increases according to the myocardial T2*. (From Westwood M, Anderson LJ, Firmin DN, et al.
A single breath-hold multiecho T2* cardiovascular magnetic resonance technique for diagnosis of myocardial
iron overload. J Magn Reson Imaging. 2003;18:33–39.)

are particularly affected by particles of hemosiderin iron. Gradient echo at these low signal values is masked by these effects, this will affect T2*
sequences measure T2*, whereas spin echo sequences measure T1 and calculations and may lead to spuriously high values. The simplest way
T2. In addition, a T2* multiecho gradient echo can repeatedly sample to deal with these points at long TE, which are in effect just noise, is
image data at different echo times (TE) in a single 10-second to 15-second to delete them from the analysis and curve fitting, which is known as
breath-hold from which T2* can be derived, and such sequences are truncation (Fig. 33.4).75 To ensure the most robust analysis, dedicated
T1 independent (Fig. 33.2). A gating delay of 0 ms after the R-wave and validated analysis software is preferable.
ensures all images are obtained at the same point in the cardiac cycle T2* is highly robust and has a high interobserver and intraobserver
and removes any variability caused by changing heart rates. Such reproducibility as well as high reproducibility between different studies
sequences are well validated, initially with “bright-blood” images and performed a few hours or days apart and across different sites and
then subsequently with “black-blood” images, further refining repro- scanner platforms.76–78 Critically this means that T2* measurements at
ducibility and reducing artifacts.67–69 T2* and T2 are related by the 1.5 T are independent of the scanner, site, or country where they were
formula 1/T2* = 1/T2 + 1/T2′, where T2′ represents magnetic inho- obtained. Although it is also possible to derive T2* using a 3 T scanner,
mogeneity. There is no particular advantage to a multislice method, clinical experience is much more limited in TM, and there are potentially
which has been reported,70,71 because iron concentration in the septum more artifacts.79,80 Current recommendations are that that clinical T2*
is representative of mean total iron concentration.72–74 MR is performed at 1.5 T. Finally, in very simple terms, increasing
The method of calculation of T2* values from raw datasets may be cardiac iron will lead to a decrease in the measured cardiac T2* value.
prone to error if care is not taken. Susceptibility artifacts at tissue borders, Although T2* has become the established technique for the assess-
such as the lung myocardial interface and near the great cardiac veins ment of myocardial iron loading, previous work used the ratio of signal
in the anterior and posterior interventricular grooves, need to be mini- intensity between the myocardium and skeletal muscle on a T2-weighted
mized. For this reason, for routine analysis of T2*, a full-thickness spin echo image. Because T2-weighted images are less susceptible to
region of interest (ROI) in the interventricular septum should be chosen field inhomogeneity (indeed part of the refocusing of the RF pulse in
to measure mean signal intensity for each echo time, thereby avoiding a spin echo image is designed to mitigate effects attributed to field
the above areas. Care is needed to ensure that the blood-pool signal inhomogeneity), small changes in imaging parameters had a significant
and other artifacts are excluded from the ROI, which can be easier with effect on results, and reproducibility was poor. It is, however, possible
a black-blood image. Signal intensity is plotted against TE, and a mono- to construct multiecho spin echo sequences from which T2 can be
exponential decay curve is fitted to derive T2*, which represents the derived in a similar way to T2*. Although T2 does shorten with increas-
time taken for the signal intensity to decay to 37% of its initial value ing myocardial iron load, T2 assessment has not been validated to the
(Fig. 33.3). The signal intensity (SI) at each echo time is given by the same extent as T2*, and is not routinely used.
following equation (where SI0 represents the signal intensity at time Similarly, T1 can be used to assess myocardial iron. T1 is measured
zero and TE represents the echo time): using an inversion-recovery sequence with T1 representing the time
taken for the magnetization to recover to 63% of its equilibrium value
Signal intensity (SI) = SI0 ⋅ e− TE T 2* Eq. 33.1 following an 180-degree RF pulse and a series of inversion-recovery
images can be acquired with a range of inversion times. As with T2*
In severe myocardial iron loading, the myocardial signal obtained multiecho, a modified Look-Locker inversion (MOLLI) sequence can
from the images with longer echo times becomes predominantly back- produce a map of T1 values for the heart in a single breath-hold. T1
ground noise, motion, and blood-pool artifacts. Because the true decay mapping techniques are increasingly being used for clinical assessment
404 SECTION III  Functional Disease

120
y = 156.42e−0.1214x
100 T2* = 8.2 ms
R2 = 0.9942
80

Signal intensity
60

40

20

0
0 5 10 15 20
B TE (ms)
A
FIG. 33.3  (A) Image analysis software is used to measure the signal intensity of the interventricular septum.
The endocardial and epicardial contours are drawn, and the full thickness septal region of interest is shown
as the arc between the red and green radial lines, which is a region of interest that is not perturbed by
susceptibility effects from deoxygenated hemoglobin in the anterior and inferior cardiac veins, nor the lung
interface. (B) Decay curve and T2* calculation. The plot of septal signal intensity against echo time (TE) can
be fitted with an exponential curve yielding the decay constant T2*. (A, From He T, Zhang J, Carpenter JP,
et al. Automated truncation method for myocardial T2* measurement in thalassemia. J Magn Reson Imaging.
2013;37:479–483. B, From He T, Gatehouse PD, Kirk P, et al. Black-blood T2* technique for myocardial iron
measurement in thalassemia. J Magn Reson Imaging. 2007;25:1205–1209.)

120 Original data in a range of pathologies such as cardiac amyloidosis. For cardiac iron
assessment, T1 does not yet provide any additional clinical value to
Truncation model
T2*.81 T1 yields improved reproducibility at normal levels of myocardial
(R-square = 0.999)
iron loading,82 but this has no current clinical value in TM. Low native
Truncated points (noncontrast) T1 values are seen in two conditions affecting the heart:
Monoexponential model namely, myocardial siderosis and Anderson-Fabry disease. The normal
80
Signal intensity

(R-square = 0.921) range for T1 in noniron loaded hearts is approximately 800 to 1000 ms,
but is variable and can require different centers to perform T1 mapping
on normal subjects to establish local “normal” values, which is a sig-
nificant limitation when compared with T2* techniques.81

40 T2*, PREDICTION OF HEART FAILURE,


AND PROGNOSIS
Serum ferritin measurements are simple to undertake and are widely
available. Although trends in ferritin are useful in monitoring the direc-
tion of body iron loading, they do not fully predict cardiac iron loading
0 and, critically, a low ferritin measurement does not predict the future
0 5 10 15 20 25 absence of heart failure.6,50,83 In particular, a single ferritin measurement
TE (ms) can be misleading and does not reflect cardiac iron burden. There is a
FIG. 33.4  Truncation curve analysis. When myocardial T2* is very short, similar problem with liver iron, where single liver iron measurements
the myocardial signal may decay to near the level of noise by the third in patients on long-term iron chelation may be misleading because
or fourth echo time (TE). The later points in the graph simply measure they do not correlate with cardiac iron levels.27 Over time, poor liver
noise, but this has the unwanted effect of not allowing the exponential iron control probably increases the risk of cardiac iron loading.83,84
line-fit to drop to zero, which artificially increases the calculated T2* Similarly, noncompliance with chelation therapy leading to elevated
value, making it incorrect. This can easily be determined because the serum ferritin levels (and thereby elevated liver iron) is a major predic-
curve does not fit the first time points properly. Software tools are
tive factor for cardiac iron loading in patients.85 Therefore high liver
therefore used to truncate the signal values at long TE, so that the
exponential curve fits the first points correctly. This gives a significantly
iron levels per se may not be the best way to view the cardiac risk
lower T2* value, which is more accurate and in this example increases associated with liver iron loading, and although control of liver iron
the R-square of fit from 0.921 to 0.999. (From He T, Zhang J, Carpenter over time is likely to be important in prevention of cardiac iron accu-
JP, et al. Automated truncation method for myocardial T2* measurement mulation, a low liver iron does not guarantee absence of cardiac iron
in thalassemia. J Magn Reson Imaging. 2013;37:479–483.) loading.86 Early on in the development of myocardial T2* the differential
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 405

RV LV LV
RV

Liver
Liver
A B
FIG. 33.5  Disparate heart and liver loading. (A) A dark liver signal but normal myocardium, indicating iron
loading in the liver but not the heart. (B) The reverse is shown, with normal liver signal but dark myocardium
indicating cardiac iron loading in the absence of liver iron loading. The latter patient is at risk from heart
failure, but in previous decades before T2* cardiovascular magnetic resonance (CMR), it is the former patient
who would have received intensified iron chelation treatment. This disparate iron loading between organs
helps explain the high mortality burden from heart disease that occurred before T2* CMR was available to
directly identify patients at risk of heart failure. LV, Left ventricle; RV, right ventricle. (From Anderson LJ,
Holden S, Davis B, et al. Cardiovascular T2-star [T2*] magnetic resonance for the early diagnosis of myocardial
iron overload. Eur Heart J. 2001;22:2171–2179.)

organ iron loading became apparent and disparate loading between occurs at a late stage of the iron overload; hence, myocardial T2* is the
the liver and heart was revealed as the reason for the limitations of most appropriate biomarker to use to assess risk. In a cohort of 652
these biomarkers (Fig. 33.5). TM patients followed up for 1 year where 80 episodes of heart failure
An increased risk of clinical heart failure has been demonstrated occurred, the mean LVEF assessed by CMR within a median time of
for patients with a falling left ventricular ejection fraction (LVEF) or 158 days from heart failure event was 43.1%.90 In nearly all patients,
absolute values below the lower limit of the normal range.50,87 Limita- the myocardial T2* was <10 ms and the mean T2* was 6.7 ms. Fur-
tions of the use of LVEF are that it requires meticulous attention to thermore, the risk of developing heart failure within 1 year without
detail, but more importantly, changes in LVEF are a late event compared intensifying chelation therapy rises rapidly as the myocardial T2* pro-
with falling T2* levels <20 ms and with low-to-moderate levels of cardiac gressively falls below 10 ms. For severe myocardial iron loading, a myo-
iron, the change in LVEF is modest if at all present. Although absolute cardial T2* value <6 ms conferred a relative risk of 270 compared with
LVEF measurements vary between centers, volumetric analysis by CMR T2* values >10 ms for heart failure within 1 year. No additional prog-
is superior to echocardiography.57,58 There is a tipping point at which nostic information was provided from other biomarkers of cardiac iron
a marked deterioration in LVEF will occur with only a slight additional (serum ferritin or liver iron). Receiver operating characteristic curve
increase in cardiac iron loading. Because it is free iron that is toxic (but analysis shows that T2* is significantly superior in predicting heart
T2* measures storage iron in the form of hemosiderin) with intensive failure compared with serum ferritin and liver iron, and should therefore
chelation treatment the LVEF will improve rapidly. However, the storage always be used.90 From this, the currently accepted T2* “risk ranges”
iron loading of the heart will only have decreased marginally in this for cardiac complications are T2* of <10 ms, representing high risk, 10
time period. Prolonged periods of chelation are therefore required to to 20 ms intermediate, and >20 ms low risk.
remove all the stored iron in the heart. Although most focus has been Although myocardial T2* assesses the risk of heart failure, the key
on deteriorating LV function, myocardial iron deposition is also strongly next step is to change therapy to reduce that risk. Once a low T2* has
associated with RV dysfunction.88 been identified, it is necessary to intervene with intensive chelation
Cardiac T2* has been directly calibrated to human cardiac iron treatment to lower cardiac iron quickly. This greatly reduces the risk
concentration (Fig. 33.6).73,74 The practical and commonly used lower of developing heart failure, which is important because once heart
limit of normal is 20 ms, although it is not appropriate to consider failure is established, it can precipitate an irreversible spiral of progres-
cardiac iron as a dichotomous variable. Historical data suggest the upper sive deterioration in cardiac function, leading to death. Randomized
limit of normal for cardiac iron is approximately 0.5 mg/g dry weight.89 controlled trials indicate that combining deferiprone with deferoxamine
The probability of a reduced EF increases as cardiac iron increases,47 has the highest cardiac iron clearance rate and improves ventricular
and a cardiac T2* <10 ms predicts heart failure.90 In one study, 98% of function. Intervention with intensified chelation appears to be very
patients who developed heart failure had a myocardial T2* <10 ms and effective, as evidenced by a 71% reduction in cardiac deaths in the 5
patients with a cardiac T2* <6 ms had a 50% likelihood of develop- years after the introduction of myocardial T2* in the United Kingdom
ment of heart failure within 12 months (Fig. 33.7).90 A normal cardiac in 1999 (Fig. 33.8).91
T2* has a very high negative predictive value for exclusion of heart
failure for at least 12 months. T2* is the most significant predictor of HEART FAILURE CAUSED BY CARDIAC
the development of heart failure compared with liver iron or serum
ferritin.90
IRON LOADING
The high basal cardiac output in TM masks subtle but significant Because the historical death rate from heart failure was 50% within 1
alterations in systolic function and a significant deterioration in LVEF year, the current emphasis is to intervene early to prevent heart failure.5,25
406 SECTION III  Functional Disease

500 400

400
300

300

R2* (Hz)
R2* (Hz)

200

200

100
100

0 0
0 2 4 6 8 10 12 14 0 2 4 6 8 10
A Fe (mg/g) B Fe (mg/g)
600

500

400
Septal R2* (S−1)

300

200

100

0
0 100 200 300 400 500 600
C Whole heart R2* (S )−1

FIG. 33.6  Calibration of T2* to human iron. (A) The calibration of human R2* (equal to the reciprocal of T2*)
for >500 myocardial samples against iron concentration measured by emission spectroscopy. The relation is
[Fe] = 45 • T2*-1.22. (B) A similar graph but using the mean whole-heart iron R2* (N = 11) vs. iron concentration.
(C) The relation between whole-heart R2* and the septal R2* for each heart, confirming that using a septal
region of interest alone was highly representative of average whole-heart iron concentration. (From Carpen-
ter JP, He T, Kirk P, et al. On T2* magnetic resonance and cardiac iron. Circulation. 2011;123:1519–1528.)

In 52 patients with mean LVEF 36%, there was 48% survival after 5 patients having a T2* >10 ms (only 1 out of 80 episodes of heart failure
years with no change in iron chelator and use of cardiac medications.26 occurred in a trial over 7 years in patients with a myocardial T2*
Data to guide optimal management of iron chelator treatment are sparse >10 ms).90 Because the relative risk of overt heart failure is so great and
as there are few trials in acute heart failure. One follow-up study showed coupled with a poor prognosis,28 many centers treat patients with a
a mortality of only 1 in 7 patients in acute heart failure treated with myocardial T2* <6 ms and those with overt heart failure with the same
continuous uninterrupted intravenous deferoxamine for 12 months.27,92 intensified chelation regimens. Many symptoms that are common in
The only randomized controlled trial in acute heart failure in TM was anemia are also typically present in heart failure, which makes the
terminated early after only 23% recruitment.93 It is therefore very under- diagnosis of heart failure difficult on clinical grounds, and this further
powered and the results are difficult to interpret. It showed that the EF cements the rationale for this approach.94 Traditional signs of heart
after 1 year on combination treatment of deferoxamine with deferiprone failure may appear late and may further delay treatment.30,94 Sepsis may
(EF + 8.4%) was higher (but not significantly higher) than the standard precipitate sudden, acute heart failure, possibly by disrupting iron stores
of care of monotherapy with deferoxamine (EF + 4.1%). It is critical and is the second leading cause of death in TM patients with severe
to remember that even with a preserved LVEF, without escalation in cardiac iron loading.95,96
therapy, the prospective risk for developing heart failure in 1 year is Patients with moderately severe cardiac iron loading (T2* between
47% if cardiac T2* is <6 ms, with a relative risk of 270 compared with 6 and 10 ms) require intensified, but not necessarily maximal, chelation
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 407

Heart T2* (ms) Heart Failure Within 1 Year


0 10 20 30 40 50 60 70 80 100%
40 10 ms
Percentage of scans

35 90%
Patients without heart
30 failure within 1 year of scan
25 80%
20
15 70%
10
5 60%
0
0 50%
Percentage of scans

5
2500 µg/L
10 40%
15
20 30%
25
30 Patients who developed heart 20%
35 failure within 1 year of scan
40 10%
0 10 20 30 40 50 60 70 80 Heart T2* ms Liver T2* ms Ferritin µg/L
15 mg/g
A 6 ms 10 ms Heart T2* (ms) 0%
0% 20% 40% 60% 80% 100%
Proportion of patients suffering heart failure

B 1-Specificity
0.6
P < .001 <6 ms
0.5

0.4
6−8 ms
0.3

0.2 8−10 ms

0.1
10+ ms
0
0 60 120 180 240 300 360
C Follow-up time (days)
FIG. 33.7  T2* predicts heart failure and arrhythmia. (A) The top portion of the graph shows the distribution
of myocardial T2* in patients who had a baseline cardiovascular magnetic resonance (CMR) scan and did
not develop heart failure within 1 year. Note that the mean T2* was ~22 ms and there was a broad range
of values. The lower portion of the graph shows the distribution of patients who did develop heart failure
within 1 year of the baseline T2* CMR scan. Note the strong leftward shift of value, with a mean of ~7 ms.
Nearly all patients had a T2* value <10 ms, and therefore this threshold is used clinically to indicate “high-
risk” of cardiac events. (B) Receiver operating curve analysis of the relative power of cardiac T2*, serum
ferritin, and liver iron to predict future heart failure. Cardiac T2* greatly outperformed the conventional
measures. Note the relative strength of performance of conventional thresholds of serum ferritin of 2500 µg/L,
liver iron of 15 mg/g dry weight, and cardiac T2* of 10 ms. (C) Kaplan-Meier curves of the incremental likeli-
hood of developing heart failure according to the cardiac T2* value. Patients with a T2* <6 ms had an
approximately 50% likelihood of developing heart failure over 1 year. (From Kirk P, Roughton M, Porter JB,
et al. Cardiac T2* magnetic resonance for prediction of cardiac complications in thalassemia major. Circula-
tion. 2009;120:1961–1968.)

therapy. Patients with mild-to-moderate cardiac iron loading (T2* loading where more frequent assessment is required. More frequent
between 10 and 20 ms) can be managed more conservatively by modi- assessment is also required if there is an impaired LVEF.
fication of chelator dose, improving patient compliance, or alternative
chelating agents. Any reduction in cardiac function with the presence TREATMENT OF THALASSEMIA MAJOR AND
of any myocardial iron loading warrants escalation in chelation therapy,
even in asymptomatic cases.87 Patients with myocardial iron loading
IRON CHELATORS
where ventricular function measurements show a trend of deterioration Iron chelating agents have two functions: first, to tightly bind iron and
(even though the absolute values may remain in the normal range) prevent it acting as a catalyst for redox reactions; and second, to allow
require intensification of chelation. If this fails to stabilize the situation, iron to be transported and then excreted from the body. Hence, iron
then a change in iron chelator regime is required. In general, annual chelators should reduce tissue iron levels, prevent excessive organ iron
assessment of myocardial T2* is sufficient unless there is severe iron accumulation, and neutralize toxic labile iron pools. For a chelating
408 SECTION III  Functional Disease

50

45

40 Unknown

35 Other
Malignancy
30
Iron overload
25
Infection
20 BMT complication

15 Anemia

10

0
1950− 1955− 1960− 1965− 1970− 1975− 1980− 1985− 1990− 1995− 2000−
1954 1959 1964 1969 1974 1979 1984 1989 1994 1999 2003
FIG. 33.8  UK mortality graph in thalassemia major (TM). The vertical axis shows the number of deaths in
the United Kingdom in TM patients for each 5-year period from 1950 to after 2000. The portion of each
column in red is the deaths from cardiac iron overload. After the introduction of cardiac T2* cardiovascular
magnetic resonance in 1999, the mortality rate from cardiac iron overload fell by 71%. BMT, Bone marrow
transplantation. (From Modell B, Khan M, Darlison M, et al. Improved survival of thalassaemia major in the
UK and relation to T2* cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2008;10:42.)

TABLE 33.2  Main Features of Iron Chelators


FDA EU Typical Chronic
Drug Approved Approved Route Dosing Frequency Excretion Main Adverse Effects
Deferoxamine Yes Yes SC 20–50 mg/kg/d 8–14 h infusion for 60% urine Sensorineural deafness,
(IV in heart 5–7 days/week 40% feces visual disturbance,
failure) skeletal abnormality,
growth retardation
Deferiprone Yes Yes Oral 75–100 mg/kg/d ×3/d 75%–90% urine Agranulocytosis, GI
disturbance, arthropathy
Deferasirox Yes Yes Oral 20–40 mg/kg/d ×1/d ~90% feces Rash, GI disturbance, rise
in creatinine

EU, European Union; FDA, US Food and Drug Administration; GI, gastrointestinal; IV, intravenous; SC, subcutaneous.

agent, diffusion through biologic membranes and hence the absorption times daily orally because of its short half-life.100 The iron–chelator
from the gastrointestinal tract and the cellular penetration are governed complex is renally excreted.100 Minor side effects include gastrointestinal
not only by molecular size but also by lipophilicity and net molecular symptoms (nausea, vomiting, and abdominal pain) and a transient
charge.97 There are three commercially available iron chelators, each increase of liver enzymes. Agranulocytosis and neutropenia are more
with very different properties. serious side effects that can necessitate either temporary or permanent
The oldest iron chelator is deferoxamine, which was the first approved cessation of therapy.
for use in the 1960s. It is not absorbed effectively by the gastrointestinal Most recently, deferasirox has been developed. This is rapidly absorbed
tract. This property, coupled with a short plasma half-life,98 necessitates from the gastrointestinal tract and, because of a longer half-life, once
parenteral administration (usually subcutaneous) as a prolonged infu- daily administration is possible.101 The main route of excretion is fecal.102
sion or for severe iron loading a continuous infusion. Compliance is Minor side effects include transient gastrointestinal disturbances (nausea,
markedly adversely affected by the need for long parenteral administra- vomiting, diarrhea, and abdominal pain), diffuse maculopapular skin
tion. Other side effects include local infusion site reactions (induration, rash, and increased alanine aminotransferase (ALT) and serum creati-
erythema, swelling, and itching), ophthalmologic and audiologic com- nine.101 More severe side effects include Fanconi syndrome and auditory
plications, renal toxicity and even, in high doses, adult respiratory distress and ocular toxicities. The main features of the common iron chelators
syndrome.99 are summarized in Table 33.2.
The next chelator to be marketed was deferiprone. This is rapidly All three commonly used chelators remove cardiac iron if given in
absorbed from the upper gastrointestinal tract and is administered three adequate doses and there is good patient compliance. However, each
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 409

medication has advantages and disadvantages and optimal therapy must monotherapy.30,116,127–130 This accelerated cardiac iron clearance is associ-
be tailored to each patient. There are many reviews on this subject.103–105 ated with improved outcomes in severe cardiac iron loading.110 Com-
The following recommendations are specific to patients with detectable, bination therapy is used for moderate-to-severe cardiac iron loading
asymptomatic cardiac iron overload. There are several randomized trials or where there is LV impairment. Although it is possible to combine
of cardiac iron treatment using chelating agents. The introduction of deferiprone and deferasirox, currently there are relatively limited data
deferoxamine in the 1970s had a profound effect on reducing mortal- on the utility of this combination.131,132 Combination deferoxamine and
ity in TM,106 which was then followed by deferiprone, which has been deferasirox has also been tested.133 American Heart Association guidelines
associated with reduced cardiac mortality in the United Kingdom, Italy, on the heart in TM, the use of T2*, and the treatment of cardiac iron
Cyprus, and Hong Kong.91,107–111 The drastic improvement in cardiac loading have been published,134 and expert opinion135 and numerous
mortality is probably multifactorial, including early identification of national guidelines are in accord with these.136–141
patients at cardiac risk from cardiac T2*, improved compliance with
chelation therapy, and the use of specific agents alone or in combination.91
EMERGING CHALLENGES AND TREATMENTS
From a cardiac perspective, deferoxamine is only able to slowly
remove cardiac iron with rates varying from 1.1% to 2.2% per month As we now enter an age of much improved survival, and patients with
up to nearly 5% for constant 24/7 intravenous infusions.27,112 Long-term TM are entering their sixth decade of life and more, new challenges to
infusions are inconvenient and uncomfortable, leading to reduced patient cardiac health emerge. Diabetes caused by iron infiltration of the pan-
compliance. Although deferoxamine is the conventional chelating agent creas is a common finding in TM and is a risk factor for coronary artery
for TM, a change in therapy is probably warranted if compliance is disease. There is endothelial dysfunction even in the absence of iron
poor. Deferiprone monotherapy offers superior cardiac protection,113–115 loading. The prevalence of coronary artery disease in this population,
and improves survival compared with deferoxamine monotherapy.111 which will occur as they age, is unknown. Additionally, the risk of
In the only prospective randomized study comparing deferoxamine arrhythmias such as atrial fibrillation (AF) attributed to altered hemo-
and deferiprone monotherapy, deferiprone reduced cardiac iron at nearly dynamics in the left and right atrium is unknown but may also be
double the rate of deferoxamine.112 Deferiprone is therefore suitable higher than that in the general population. T1 mapping is able to assess
for patients with cardiac siderosis, either as monotherapy or in com- myocardial iron and to characterize diffuse fibrosis, which does still
bination with deferoxamine.116 It is noteworthy that the use of either occur with severe iron loading. However, these two processes have the
deferoxamine or deferasirox alone in mild-to-moderate cardiac iron opposite effect on T1 values, which may lead to challenges in interpreta-
loading does not improve LVEF,112,117–119 which is in contrast to deferi- tion of T1 values. The prognostic value of iron loading as assessed by
prone, which does improve LVEF.112,113,115,120 Deferasirox monotherapy T1 mapping is yet to be determined. Although recent trials on the use
can be used in patients with cardiac iron loading and preserved systolic of amlodipine to block cardiac iron uptake in TM are of great inter-
function with clearance rates similar to deferoxamine.121–123 Currently, est,142,143 much more work is needed to understand the prevention of
there are few data on the use of deferasirox in patients with severe heart failure. Finally, the ultimate goal would be to eradicate the defect
cardiac iron loading or a reduced LVEF. in hemoglobin synthesis and, with advances in gene therapy such as
There is evidence of a synergistic effect between deferiprone and CRISPR-Cas9 gene editing, so far demonstrated in stem cells from
deferoxamine,19,124 and these agents have been successfully combined patients with sickle cell anemia,144 clinical trials are being planned.
to improve cardiac and hepatic iron clearance.125,126 Combination therapy
can rapidly improve myocardial iron loading with an associated improve-
ment in LVEF and has been shown to lead to a near doubling in the
REFERENCES
rate of increase of myocardial T2* when compared with deferoxamine A full reference list is available online at ExpertConsult.com
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 409.e1

REFERENCES 24. Eaton JW, Qian M. Molecular bases of cellular iron toxicity. Free Radic
Biol Med. 2002;32:833–840.
1. Origa R, Galanello R, Ganz T, et al. Liver iron concentrations and 25. Engle MA. Cardiac involvement in Cooley’s anemia. Ann N Y Acad Sci.
urinary hepcidin in beta-thalassemia. Haematologica. 2007;92:583–588. 1964;119:698–702.
2. Aessopos A, Tsironi M, Andreopoulos A, Farmakis D. Heart disease in 26. Kremastinos DT, Tsetsos GA, Tsiapras DP, Karavolias GK, Ladis VA,
thalassemia intermedia. Hemoglobin. 2009;33(suppl 1):S170–S176. Kattamis CA. Heart failure in beta thalassemia: a 5-year follow-up study.
3. Musallam KM, Cappellini MD, Wood JC, Taher AT. Iron overload in Am J Med. 2001;111:349–354.
non-transfusion-dependent thalassemia: a clinical perspective. Blood 27. Anderson LJ, Westwood MA, Holden S, et al. Myocardial iron clearance
Rev. 2012;26(suppl 1):S16–S19. during reversal of siderotic cardiomyopathy with intravenous
4. Voskaridou E, Christoulas D, Terpos E. Sickle-cell disease and the heart: desferrioxamine: a prospective study using T2* cardiovascular magnetic
review of the current literature. Br J Haematol. 2012;157:664–673. resonance. Br J Haematol. 2004;127:348–355.
5. Engle MA, Erlandson M, Smith CH. Late cardiac complications of 28. Davis BA, Porter JB. Long-term outcome of continuous 24-hour
chronic, severe, refractory anemia with hemochromatosis. Circulation. deferoxamine infusion via indwelling intravenous catheters in high-risk
1964;30:698–705. beta-thalassemia. Blood. 2000;95:1229–1236.
6. Olivieri NF, Nathan DG, MacMillan JH, et al. Survival in medically 29. Miskin H, Yaniv I, Berant M, Hershko C, Tamary H. Reversal of cardiac
treated patients with homozygous beta-thalassemia. N Engl J Med. complications in thalassemia major by long-term intermittent daily
1994;331:574–578. intensive iron chelation. Eur J Haematol. 2003;70:398–403.
7. Charafeddine K, Isma’eel H, Charafeddine M, et al. Survival and 30. Tanner MA, Galanello R, Dessi C, et al. Combined chelation therapy in
complications of beta-thalassemia in Lebanon: a decade’s experience of thalassemia major for the treatment of severe myocardial siderosis with
centralized care. Acta Haematol. 2008;120:112–116. left ventricular dysfunction. J Cardiovasc Magn Reson. 2008;10:12.
8. Ehlers KH, Giardina PJ, Lesser ML, Engle MA, Hilgartner MW. 31. Thomas AS, Garbowski M, Ang AL, et al. A decade follow-up of a
Prolonged survival in patients with beta-thalassemia major treated with thalassemia major (TM) cohort monitored by cardiac magnetic
deferoxamine. J Pediatr. 1991;118:540–545. resonance imaging (CMR): Significant reduction in patients with
9. Zurlo MG, De Stefano P, Borgna-Pignatti C, et al. Survival and causes of cardiac iron and in total mortality. Blood (ASH Annual Meeting
death in thalassaemia major. Lancet. 1989;2:27–30. Abstracts). 2010;116:1011.
10. Ladis V, Chouliaras G, Berdoukas V, et al. Survival in a large cohort of 32. Buja LM, Roberts WC. Iron in the heart. Etiology and clinical
Greek patients with transfusion-dependent beta thalassaemia and significance. Am J Med. 1971;51:209–221.
mortality ratios compared to the general population. Eur J Haematol. 33. Pepe A, Positano V, Capra M, et al. Myocardial scarring by delayed
2011;86:332–338. enhancement cardiovascular magnetic resonance in thalassaemia major.
11. Ludwiczek S, Theurl I, Muckenthaler MU, et al. Ca2+ channel blockers Heart. 2009;95:1688–1693.
reverse iron overload by a new mechanism via divalent metal 34. Kirk P, Carpenter JP, Tanner MA, Pennell DJ. Low prevalence of fibrosis
transporter-1. Nat Med. 2007;13:448–454. in thalassemia major assessed by late gadolinium enhancement
12. Oudit GY, Sun H, Trivieri MG, et al. L-type Ca2+ channels provide a cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2011;13:8.
major pathway for iron entry into cardiomyocytes in iron-overload 35. Trikas A, Tentolouris K, Katsimaklis G, Antoniou J, Stefanadis C,
cardiomyopathy. Nat Med. 2003;9:1187–1194. Toutouzas P. Exercise capacity in patients with beta-thalassemia major:
13. Kumfu S, Chattipakorn S, Chinda K, Fucharoen S, Chattipakorn N. relation to left ventricular and atrial size and function. Am Heart J.
T-type calcium channel blockade improves survival and cardiovascular 1998;136:988–990.
function in thalassemic mice. Eur J Haematol. 2012;88:535–548. 36. Hamdy AM, Zein El-Abdin MY, Abdel-Hafez MA. Right ventricular
14. Kumfu S, Chattipakorn S, Fucharoen S, Chattipakorn N. Ferric iron function in patients with beta thalassemia: relation to serum ferritin
uptake into cardiomyocytes of β-thalassemic mice is not through level. Echocardiography. 2007;24:795–801.
calcium channels. Drug Chem Toxicol. 2013;36:329–334. 37. Vogel M, Anderson LJ, Holden S, Deanfield JE, Pennell DJ, Walker JM.
15. Tsushima RG, Wickenden AD, Bouchard RA, Oudit GY, Liu PP, Backx Tissue Doppler echocardiography in patients with thalassaemia detects
PH. Modulation of iron uptake in heart by L-type Ca2+ channel early myocardial dysfunction related to myocardial iron overload. Eur
modifiers: possible implications in iron overload. Circ Res. 1999;84: Heart J. 2003;24:113–119.
1302–1309. 38. Marcí M, Pitrolo L, Lo Pinto C, Sanfilippo N, Malizia R. Detection of
16. Murphy CJ, Oudit GY. Iron-overload cardiomyopathy: pathophysiology, early cardiac dysfunction in patients with Beta thalassemia by tissue
diagnosis, and treatment. J Card Fail. 2010;16:888–900. Doppler echocardiography. Echocardiography. 2011;28:175–180.
17. Hershko C. Pathogenesis and management of iron toxicity in 39. Westwood MA, Wonke B, Maceira AM, et al. Left ventricular diastolic
thalassemia. Ann N Y Acad Sci. 2010;1202:1–9. function compared with T2* cardiovascular magnetic resonance for
18. Link G, Pinson A, Hershko C. Ability of the orally effective iron early detection of myocardial iron overload in thalassemia major.
chelators dimethyl- and diethyl-hydroxypyrid-4-one and of J Magn Reson Imaging. 2005;22:229–233.
deferoxamine to restore sarcolemmal thiolic enzyme activity in 40. Cheung YF, Chan GC, Ha SY. Arterial stiffness and endothelial function in
iron-loaded heart cells. Blood. 1994;83:2692–2697. patients with beta-thalassemia major. Circulation. 2002;106:2561–2566.
19. Link G, Konjin AM, Breuer W, Cabantchik ZI, Hershko C. Exploring the 41. Cheung YF, Chan GCF, Ha SY. Effect of deferasirox (ICL670) on arterial
“iron shuttle” hypothesis in chelation therapy: effects of combined function in patients with beta-thalassaemia major. Br J Haematol.
deferoxamine and deferiprone treatment in hypertransfused rats with 2008;141:728–733.
labelled iron stores and in iron-loaded rat heart cells in culture. J Lab 42. Kukongviriyapan V, Somparn N, Senggunprai L, Prawan A,
Clin Med. 2001;138:130–138. Kukongviriyapan U, Jetsrisuparb A. Endothelial dysfunction and oxidant
20. Wood JC, Enriquez C, Ghugre N, et al. Physiology and pathophysiology of status in pediatric patients with hemoglobin E-beta thalassemia. Pediatr
iron cardiomyopathy in thalassemia. Ann N Y Acad Sci. 2005;1054:386–395. Cardiol. 2008;29:130–135.
21. Kim E, Giri SN, Pessah IN. Iron(II) is a modulator of ryanodine- 43. Mancuso L, Mancuso A, Bevacqua E, Rigano P. Electrocardiographic
sensitive calcium channels of cardiac muscle sarcoplasmic reticulum. abnormalities in thalassemia patients with heart failure. Cardiovasc
Toxicol Appl Pharmacol. 1995;130:57–66. Hematol Disord Drug Targets. 2009;9:29–35.
22. Buja LM, Roberts WC. Iron in the heart. Etiology and clinical 44. Ramazzotti A, Pepe A, Positano V, et al. Standardized T2* map of a
significance. Am J Med. 1971;51:209–221. normal human heart to correct T2* segmental artefacts; myocardial iron
23. Kirk P, Sheppard MN, Carpenter JP, et al. Post-mortem study of the overload and fibrosis in thalassemia intermedia versus thalassemia major
association between cardiac iron and fibrosis in transfusion dependent patients and electrocardiogram changes in thalassemia major patients.
anaemia. J Cardiovasc Magn Reson. 2017;19:36. Hemoglobin. 2008;32:97–107.
409.e2 SECTION III  Functional Disease

45. Kremastinos DT, Hamodraka E, Parissis J, Tsiapras D, Dima K, Maisel A. 63. Roghi A, Dellegrottaglie S, Pedrotti P, Pedretti S, Cassinerio E, Cappellini
Predictive value of B-type natriuretic peptides in detecting latent left MD. Unexpected myocarditis in thalassaemia major patient screened for
ventricular diastolic dysfunction in beta-thalassemia major. Am Heart J. iron load cardiomyopathy. BMJ Case Rep. 2009;2009.
2010;159:68–74. 64. Plein S, Schulz-Menger J, Almeida A, et al. Working Group on
46. Delaporta P, Kattamis A, Apostolakou F, et al. Correlation of Cardiovascular Magnetic Resonance, European Society of Cardiology.
NT-proBNP levels and cardiac iron concentration in patients with Training and accreditation in cardiovascular magnetic resonance in
transfusion-dependent thalassemia major. Blood Cells Mol Dis. Europe: a position statement of the working group on cardiovascular
2013;50:20–24. magnetic resonance of the European Society of Cardiology. Eur Heart J.
47. Anderson LJ, Holden S, Davis B, et al. Cardiovascular T2-star (T2*) 2011;32:793–798.
magnetic resonance for the early diagnosis of myocardial iron overload. 65. Kramer CM, Barkhausen J, Flamm SD, Kim RJ, Nagel E. Standardized
Eur Heart J. 2001;22:2171–2179. cardiovascular magnetic resonance imaging (CMR) protocols, Society
48. Bellenger NG, Burgess MI, Ray SG, et al. Comparison of left ventricular for Cardiovascular Magnetic Resonance: board of trustees task force on
ejection fraction and volumes in heart failure by echocardiography, standardized protocols. J Cardiovasc Magn Reson. 2008;10:35.
radionuclide ventriculography and cardiovascular magnetic resonance; 66. Fitchett DH, Coltart DJ, Littler WA, et al. Cardiac involvement in
are they interchangeable? Eur Heart J. 2000;21:1387–1396. secondary haemochromatosis: a catheter biopsy study and analysis of
49. Giakoumis A, Berdoukas V, Gotsis E, Aessopos A. Comparison of myocardium. Cardiovasc Res. 1980;14:719–724.
echocardiographic (US) volumetry with cardiac magnetic resonance 67. Westwood M, Anderson LJ, Firmin DN, et al. A single breath-hold
(CMR) imaging in transfusion dependent thalassemia major (TM). multiecho T2* cardiovascular magnetic resonance technique for
Cardiovasc Ultrasound. 2007;5:24. diagnosis of myocardial iron overload. J Magn Reson Imaging.
50. Hahalis G, Kourakli A, Gerasimidou I, et al. Cardiac mortality in 2003;18:33–39.
{beta}-thalassemia major: resting but not dobutamine stress 68. He T, Gatehouse PD, Kirk P, et al. Black-blood T2* technique for
echocardiography predicts mortality among initially cardiac disease-free myocardial iron measurement in thalassemia. J Magn Reson Imaging.
patients in a prospective 12-year study. Eur J Heart Fail. 2007;25:1205–1209.
2009;11:1178–1181. 69. Smith GC, Carpenter JP, He T, Alam MH, Firmin DN, Pennell DJ. Value
51. Nagueh SF, Appleton CP, Gillebert TC, et al. Recommendations for the of black blood T2* cardiovascular magnetic resonance. J Cardiovasc
evaluation of left ventricular diastolic function by echocardiography. Magn Reson. 2011;13:21.
J Am Soc Echocardiogr. 2009;22:107–133. 70. Pepe A, Positano V, Santarelli MF, et al. Multislice multiecho T2*
52. Leon MB, Borer JS, Bacharach SL, et al. Detection of early cardiac cardiovascular magnetic resonance for detection of the heterogeneous
dysfunction in patients with severe beta-thalassemia and chronic iron distribution of myocardial iron overload. J Magn Reson Imaging.
overload. N Engl J Med. 1979;301:1143–1148. 2006;23:662–668.
53. Maffei E, Messalli G, Martini C, et al. Left and right ventricle assessment 71. Pepe A, Positano V, Santarelli MF, et al. Multislice multiecho T2*
with cardiac CT: validation study vs. cardiac MR. Eur Radiol. cardiovascular magnetic resonance for detection of the heterogeneous
2012;22:1041–1049. distribution of myocardial iron overload. J Magn Reson Imaging.
54. Wood JC, Mo A, Gera A, Koh M, Coates T, Gilsanz V. Quantitative 2006;23:662–668.
computed tomography assessment of transfusional iron overload. Br J 72. Ghugre NR, Enriquez CM, Coates TD, Nelson MD, Wood JC. Improved
Haematol. 2011;153:780–785. R2* measurements in myocardial iron overload. J Magn Reson Imaging.
55. Maceira AM, Prasad SK, Khan M, Pennell DJ. Normalized left 2006;23:9–16.
ventricular systolic and diastolic function by steady state free precession 73. Ghugre NR, Enriquez CM, Gonzalez I, Nelson MD Jr, Coates TD, Wood
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. JC. MRI detects myocardial iron in the human heart. Magn Reson Med.
2006;8:417–426. 2006;56:681–686.
56. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular 74. Carpenter JP, He T, Kirk P, et al. On T2* magnetic resonance and cardiac
systolic and diastolic function normalized to age, gender and body iron. Circulation. 2011;123:1519–1528.
surface area from steady-state free precession cardiovascular magnetic 75. He T, Zhang J, Carpenter JP, et al. Automated truncation method for
resonance. Eur Heart J. 2006;27:2879–2888. myocardial T2* measurement in thalassemia. J Magn Reson Imaging.
57. Grothues F, Smith GC, Moon JC, et al. Comparison of interstudy 2013;37:479–483.
reproducibility of cardiovascular magnetic resonance with two- 76. Westwood M, Anderson LJ, Firmin DN, et al. Interscanner reproducibility
dimensional echocardiography in normal subjects and in patients with of cardiovascular magnetic resonance in the early diagnosis of myocardial
heart failure or left ventricular hypertrophy. Am J Cardiol. 2002;90: iron overload. J Magn Reson Imaging. 2003;18:616–620.
29–34. 77. Westwood MA, Firmin DN, Gildo M, et al. Intercentre reproducibility of
58. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ. magnetic resonance T2* measurements of myocardial iron in
Interstudy reproducibility of right ventricular volumes, function, and thalassaemia. Int J Cardiovasc Imaging. 2005;21:531–538.
mass with cardiovascular magnetic resonance. Am Heart J. 78. Kirk P, He T, Anderson LJ, et al. International reproducibility of single
2004;147:218–223. breathhold T2* MR for cardiac and liver iron assessment among five
59. Westwood MA, Anderson LJ, Maceira AM, et al. Normalized left thalassemia centers. J Magn Reson Imaging. 2010;32:315–319.
ventricular volumes and function in thalassemia major patients 79. Storey P, Thompson AA, Carqueville CL, Wood JC, de Freitas RA, Rigsby
with normal myocardial iron. J Magn Reson Imaging. 2007;25:1147– CK. R2* imaging of transfusional iron burden at 3T and comparison
1151. with 1.5T. J Magn Reson Imaging. 2007;25:540–547.
60. Carpenter JP, Alpendurada F, Deac M, et al. Right ventricular volumes 80. Meloni A, Positano V, Keilberg P, et al. Feasibility, reproducibility, and
and function in thalassemia major patients in the absence of myocardial reliability for the T*2 iron evaluation at 3 T in comparison with 1.5 T.
iron overload. J Cardiovasc Magn Reson. 2010;12:24. Magn Reson Med. 2012;68:543–551.
61. Fernandes JL, Silveira MA, Fertrin K, et al. Left and right ventricular 81. Alam MH, Auger D, Smith GC, et al. T1 at 1.5T and 3T compared with
function and volume assessment in young thalassemia major patients conventional T2* at 1.5T for cardiac siderosis. J Cardiovasc Magn Reson.
with no related myocardial iron overload. Ann Hematol. 2012;91: 2015;17:102.
1839–1844. 82. Sado DM, Maestrini V, Piechnik SK, et al. Noncontrast myocardial T1
62. Meloni A, Detterich J, Berdoukas V, et al. Comparison of biventricular mapping using cardiovascular magnetic resonance for iron overload.
dimensions and function between pediatric sickle-cell disease and J Magn Reson Imaging. 2015;41:1505–1511.
thalassemia major patients without cardiac iron. Am J Hematol. 83. Telfer PT, Prestcott E, Holden S, Walker M, Hoffbrand AV, Wonke B.
2013;88:213–218. Hepatic iron concentration combined with long-term monitoring of
CHAPTER 33  Cardiac Iron Loading and Myocardial T2* 409.e3

serum ferritin to predict complications of iron overload in thalassaemia 107. Piga A, Gaglioti C. Fogliacco E, Tricta F. Comparative effects of
major. Br J Haematol. 2000;110:971–977. deferiprone and deferoxamine on survival and cardiac disease in
84. Brittenham GM, Griffith PM, Nienhuis AW, et al. Efficacy of patients with thalassemia major: a retrospective analysis. Haematologica.
deferoxamine in preventing complications of iron overload in patients 2003;88:489–496.
with thalassemia major. N Engl J Med. 1994;331:567–573. 108. Ceci A, Baiardi P, Catapano M, et al. Risk factors for death in patients
85. Gabutti V, Piga A. Results of long-term iron-chelating therapy. Acta with beta-thalassemia major: results of a case-control study.
Haematol. 1996;95:26–36. Haematologica. 2006;91:1420–1421.
86. Noetzli LJ, Carson SM, Nord AS, Coates TD, Wood JC. Longitudinal 109. Maggio A, Vitrano A, Capra M, et al. Improving survival with
analysis of heart and liver iron in thalassemia major. Blood. 2008;112: deferiprone treatment in patients with thalassemia major: a prospective
2973–2978. multicenter randomised clinical trial under the auspices of the Italian
87. Davis BA, O’Sullivan C, Jarritt PH, Porter JB. Value of sequential Society for Thalassemia and Hemoglobinopathies. Blood Cells Mol Dis.
monitoring of left ventricular ejection fraction in the management of 2009;42:247–251.
thalassemia major. Blood. 2004;104:263–269. 110. Telfer PT, Warburton F, Christou S, et al. Improved survival in
88. Alpendurada F, Carpenter JP, Deac M, et al. Relation of myocardial T2* thalassemia major patients on switching from desferrioxamine to
to right ventricular function in thalassaemia major. Eur Heart J. combined chelation therapy with desferrioxamine and deferiprone.
2010;31:1648–1654. Haematologica. 2009;94:1777–1778.
89. Collins W, Taylor WH. Determination of iron in cardiac and liver tissues 111. Borgna-Pignatti C, Cappellini MD, De Stefano P, et al. Cardiac
by plasma emission spectroscopy. Ann Clin Biochem. 1987;24:483–487. morbidity and mortality in deferoxamine- or deferiprone-treated
90. Kirk P, Roughton M, Porter JB, et al. Cardiac T2* magnetic resonance patients with thalassemia major. Blood. 2006;107:3733–3737.
for prediction of cardiac complications in thalassemia major. 112. Pennell DJ, Berdoukas V, Karagiorga M, et al. Randomized controlled
Circulation. 2009;120:1961–1968. trial of deferiprone or deferoxamine in beta-thalassemia major patients
91. Modell B, Khan M, Darlison M, Westwood MA, Ingram D, Pennell DJ. with asymptomatic myocardial siderosis. Blood. 2006;107:3738–3744.
Improved survival of thalassaemia major in the UK and relation to T2* 113. Anderson LJ, Wonke B, Prescott E, Holden S, Walker JM, Pennell DJ.
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2008; Comparison of effects of oral deferiprone and subcutaneous
10:42. desferrioxamine on myocardial iron levels and ventricular function in
92. Porter J, Wood J, Olivieri N, et al. for the Thalassemia Clinical Research beta thalassemia. Lancet. 2002;360:516–520.
Network. Treatment of heart failure in adults with thalassaemia major: 114. Pepe A, Lombardi M, Positano V, et al. Evaluation of the efficacy of oral
response in patients randomised to deferoxamine with or without deferiprone in beta-thalassemia major by multislice multiecho T2*. Eur J
deferiprone. J Cardiovasc Magn Reson. 2013;15:38. Haematol. 2006;76:183–192.
93. Porter JB, Wood J, Olivieri N, et al. Treatment of heart failure in adults 115. Pepe A, Meloni A, Capra M, et al. Deferasirox, deferiprone and
with thalassemia major: response in patients randomised to desferrioxamine treatment in thalassemia major patients: cardiac iron
deferoxamine with or without deferiprone. J Cardiovasc Magn Reson. and function comparison determined by quantitative magnetic
2013;15:38. resonance imaging. Haematologica. 2011;96:41–47.
94. Forni GL, Derchi G. Typical manifestation of acute congestive heart 116. Tanner MA, Galanello R, Dessi C, et al. A randomized, placebo-
failure in patients with Thalassaemia major causing diagnostic delay in controlled, double-blind trial of the effect of combined therapy with
the emergency room. Eur J Heart Fail. 2003;5:607–608. deferoxamine and deferiprone on myocardial iron in thalassemia major
95. Rahav G, Volach V, Shapiro M, Rund D, Rachmilewitz EA, Goldfarb A. using cardiovascular magnetic resonance. Circulation. 2007;115:
Severe infections in thalassaemic patients: prevalence and predisposing 1876–1884.
factors. Br J Haematol. 2006;133:667–674. 117. Pennell DJ, Porter JB, Cappellini MD, et al. Efficacy of deferasirox in
96. Adamkiewicz TV, Berkovitch M, Krishnan C, Polsinelli C, Kermack D, reducing and preventing cardiac iron overload in beta-thalassemia.
Olivieri NF. Infection due to Yersinia enterocolitica in a series of patients Blood. 2010;115:2364–2371.
with beta-thalassemia: incidence and predisposing factors. Clin Infect 118. Pennell DJ, Porter JB, Cappellini MD, et al. Continued improvement in
Dis. 1998;27:1362–1366. myocardial T2* over two years of deferasirox therapy in beta-
97. Hider RC, Liu ZD. Emerging understanding of the advantage of small thalassemia major patients with cardiac iron overload. Haematologica.
molecules such as hydroxypyridinones in the treatment of iron overload. 2011;96:48–54.
Curr Med Chem. 2003;10:1051–1064. 119. Pennell DJ, Porter JB, Cappellini MD, et al. Deferasirox for up to 3 years
98. Lee P, Mohammed N, Marshall L, et al. Source Intravenous infusion leads to continued improvement of myocardial T2* in patients with
pharmacokinetics of desferrioxamine in thalassaemic patients. Drug β-thalassemia major. Haematologica. 2012;97:842–848.
Metab Dispos. 1993;21:640–644. 120. Maggio A, Vitrano A, Lucania G, et al. Long-term use of deferiprone
99. Freedman MH, Grisaru D, Oliveri N, MacLusky I, Thorner PS. significantly enhances left-ventricular ejection function in thalassemia
Pulmonary syndrome in patients with thalassemia major receiving major patients. Am J Hematol. 2012;87:732–733.
intravenous deferoxamine infusions. Am J Dis Child. 1990;144: 121. Wood JC, Glynos T, Thompson A, et al. Follow-up report on the 2-year
565–569. cardiac data from a deferasirox monotherapy trial. Am J Hematol.
100. Al-Refaie FN, Sheppard LN, Nortey P, Wonke B, Hoffbrand AV. 2010;85:818–819.
Pharmacokinetics of the oral iron chelator deferiprone (L1) in patients 122. Pathare A, Taher A, Daar S. Deferasirox (Exjade) significantly improves
with iron overload. Br J Haematol. 1995;89:403–408. cardiac T2* in heavily iron-overloaded patients with beta-thalassemia
101. Galanello R, Campus S, Origa R. Deferasirox: pharmacokinetics and major. Ann Hematol. 2010;89:405–409.
clinical experience. Expert Opin Drug Metab Toxicol. 2012;8:123–134. 123. Pennell DJ, Porter JB, Piga A, et al. CORDELIA Study Investigators. A
102. Nick H. Deferasirox (Exjade, ICL670) preclinical overview. Semin 1-year randomized controlled trial of deferasirox vs deferoxamine for
Hematol. 2007;44:S12–S15. myocardial iron removal in β-thalassemia major (CORDELIA). Blood.
103. Porter JB. Optimizing iron chelation strategies in beta-thalassaemia 2014;123:1447–1454.
major. Blood Rev. 2009;23(suppl 1):S3–S7. 124. Evans P, Kayyali R, Hider RC, Eccleston J, Porter JB. Mechanisms for the
104. Kwiatkowski JL. Oral iron chelators. Hematol Oncol Clin North Am. shuttling of plasma non-transferrin-bound iron (NTBI) onto
2010;24:229–248. deferoxamine by deferiprone. Transl Res. 2010;156:55–67.
105. Brittenham GM. Iron-chelating therapy for transfusional iron overload. 125. Wonke B, Wright C, Hoffbrand AV. Combined therapy with deferiprone
N Engl J Med. 2011;364:146–156. and desferrioxamine. Br J Haematol. 1998;103:361–364.
106. Modell B, Letsky E, Flynn D, Peto R, Weatherall DJ. Survival and 126. Kattamis A, Kassou C, Berdousi H, Ladis V, Papassotiriou I, Kattamis C.
desferrioxamine in thalassaemia major. Br Med J. 1982;284:1081–1084. Combined therapy with desferrioxamine and deferiprone in thalassemic
409.e4 SECTION III  Functional Disease

patients: effect on urinary iron excretion. Haematologica. 2003;88: Thalassemia Foundation of Canada, Ontario, Canada. 2009. http://www
1423–1425. .thalassemia.ca/wp-content/uploads/Thalassemia-Guidelines_LR.pdf.
127. Farmaki K, Tzoumari I, Pappa C, Chouliaras G, Berdoukas V. 142. Fernandes JL, Sampaio EF, Fertrin K, et al. Amlodipine reduces cardiac
Normalisation of total body iron load with very intensive combined iron overload in patients with thalassemia major: A pilot trial. Am J
chelation reverses cardiac and endocrine complications of thalassaemia Med. 2013;126:834–837.
major. Br J Haematol. 2010;148:466–475. 143. Fernandes JL, Loggetto SR, Veríssimo MP, et al. A randomized trial of
128. Jetsrisuparb A, Komvilaisak P, Wiangnon S, Jetsrisuparb C. Retrospective amlodipine in addition to standard chelation therapy in patients with
study on the combination of desferrioxamine and deferasirox for thalassemia major. Blood. 2016;128:1555–1561.
treatment of iron-overloaded thalassemic patients: first evidence of 144. DeWitt MA, Magis W, Bray NL, et al. Selection-free genome editing of
more than 2 years. J Pediatr Hematol Oncol. 2010;32:400–403. the sickle mutation in human adult hematopoietic stem/progenitor cells.
129. Farmaki K, Tzoumari I, Pappa C. Oral chelators in transfusion- Sci Transl Med. 2016;8:360ra134.
dependent thalassemia major patients may prevent or reverse iron 145. Au WY, Lam WW, Chu WW, et al. A cross-sectional magnetic resonance
overload complications. Blood Cells Mol Dis. 2011;47:33–40. imaging assessment of organ specific hemosiderosis in 180 thalassemia
130. Mourad FH, Hoffbrand AV, Sheikh-Taha M, Koussa S, Khoriaty AI, major patients in Hong Kong. Haematologica. 2008;93:784–786.
Taher A. Comparison between desferrioxamine and combined therapy 146. Aypar E, Alehan D, Hazirolan T, Gümrük F. The efficacy of tissue
with desferrioxamine and deferiprone in iron overloaded thalassaemia Doppler imaging in predicting myocardial iron load in patients with
patients. Br J Haematol. 2003;121:187–189. beta-thalassemia major: correlation with T2* cardiovascular magnetic
131. Song TS, Hsieh YW, Peng CT, et al. Combined versus monotherapy or resonance. Int J Cardiovasc Imaging. 2010;26:413–421.
concurrent therapy for treatment of thalassaemia. In Vivo. 2014;28: 147. Patton N, Brown G, Leung M, et al. Observational study of iron
645–649. overload as assessed by magnetic resonance imaging in an adult
132. Elalfy MS, Adly AM, Wali Y, Tony S, Samir A, Elhenawy YI. Efficacy and population of transfusion-dependent patients with beta thalassaemia:
safety of a novel combination of two oral chelators deferasirox/ significant association between low cardiac T2* < 10 ms and cardiac
deferiprone over deferoxamine/deferiprone in severely iron overloaded events. Intern Med J. 2010;40:419–426.
young beta thalassemia major patients. Eur J Haematol. 148. Daar S, Pathare AV, Jain R, Zadjali SA, Pennell DJ. T2* cardiovascular
2015;95:411–420. magnetic resonance in the management of thalassemia patients in
133. Aydinok Y, Kattamis A, Cappellini MD, et al. HYPERION Investigators. Oman. Haematologica. 2009;94:140–141.
Effects of deferasirox-deferoxamine on myocardial and liver iron in 149. Kwiatkowski JL, Kim HY, Thompson AA, et al. Thalassemia Clinical
patients with severe transfusional iron overload. Blood. Research Network. Chelation use and iron burden in North American
2015;125:3868–3877. and British thalassemia patients: a report from the Thalassemia
134. Pennell DJ, Udelson JE, Arai AE, et al. American Heart Association Longitudinal Cohort. Blood. 2012;119:2746–2753.
Committee on Heart Failure and Transplantation of the Council on 150. Tanner MA, Galanello R, Dessi C, et al. Myocardial iron loading in
Clinical Cardiology and Council on Cardiovascular Radiology and patients with thalassemia major on deferoxamine chelation. J Cardiovasc
Imaging. Cardiovascular function and treatment in β-thalassemia major: Magn Reson. 2006;8:543–547.
a consensus statement from the American Heart Association. 151. Positano V, Pepe A, Santarelli MF, et al. Multislice multiecho T2* cardiac
Circulation. 2013;128:281–308. magnetic resonance for the detection of heterogeneous myocardial iron
135. Rachmilewitz EA, Giardina PJ. How I treat thalassemia. Blood. distribution in thalassaemia patients. NMR Biomed. 2009;22:707–715.
2011;118:3479–3488. 152. Aessopos A, Fragodimitri C, Karabatsos F, et al. Cardiac magnetic
136. Angelucci E, Barosi G, Camaschella C, et al. Italian Society of resonance imaging R2* assessments and analysis of historical parameters
Hematology practice guidelines for the management of iron overload in in patients with transfusion-dependent thalassemia. Haematologica.
thalassemia major and related disorders. Haematologica. 2008;93: 2007;92:131–132.
741–752. 153. Carpenter JP, Roughton M, Pennell DJ. Myocardial Iron in Thalassemia
137. Ho PJ, Tay L, Lindeman R, Catley L, Bowden DK. Australian guidelines (MINT) Investigators. International survey of T2* cardiovascular
for the assessment of iron overload and iron chelation in transfusion- magnetic resonance in β-thalassemia major. Haematologica.
dependent thalassaemia major, sickle cell disease and other congenital 2013;98:1368–1374.
anaemias. Intern Med J. 2011;41:516–524. 154. El Beshlawy A, El Tagui M, Hamdy M, et al. Low prevalence of cardiac
138. Vichinsky E, Levine L. Standards of Care Guidelines for Thalassaemia. siderosis in heavily iron loaded Egyptian thalassemia major patients.
Oakland, CA: Children’s Hospital and Research Centre; 2012. http:// Ann Hematol. 2014;93:375–379.
thalassemia.com/documents/socguidelines2012.pdf. 155. Alvi N, Tipoo FA, Imran A, et al. Burden of cardiac siderosis in a
139. Cappellini MD, Cohen A, Porter J, Taher A, Viprakasit V. Guidelines for thalassemia-major endemic population: a preliminary report from
the Management of Transfusion Dependent Thalassaemia (TDT). 3rd ed. Pakistan. J Pediatr Hematol Oncol. 2016;38:378–383.
Thalassaemia International Federation; 2014. ISBN: 978-9963-717-06-4. 156. Wahidiyat PA, Liauw F, Sekarsari D, Putriasih SA, Berdoukas V, Pennell
140. UK Thalassaemia Society. Standards for the Clinical Care of Children and DJ. Evaluation of cardiac and hepatic iron overload in thalassemia major
Adults With Thalassaemia in the UK. 3rd ed. 2016. ISBN: patients with T2* magnetic resonance imaging. Hematology.
978-1-900254-20-5. 2017;22:501–507.
141. Sayani F, Warner M, Wu J, Wong-Rieger D, Humphreys K, Odame I.
Guidelines for the Clinical Care of Patients With Thalassaemia in Canada.
34 
Arrhythmogenic Right Ventricular
Cardiomyopathya
Anneline S.J.M. te Riele and David A. Bluemke

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a heritable RV myocardium, myocardial cell loss, and abundant subepicardial fat.
heart muscle disease characterized by fibrofatty replacement of, pre- The abnormal RV regions were traditionally thought to be located to
dominantly, the right ventricular (RV) myocardium, which predisposes the so-called triangle of dysplasia (i.e., RV inflow tract, RV apex, and
patients to potentially life-threatening arrhythmias and ventricular RV outflow tract [RVOT]).1 In subsequent years, several ARVC manu-
dysfunction.1 Affected patients typically present between the second scripts have focused on gross or histologic evidence of myocardial cell
and fourth decade of life with arrhythmias coming from the RV.2 ARVC loss with fibrofatty replacement.6–8 It is important to note that these
is an unusual condition, with an estimated prevalence of 1 in 1000 to observations were made in tertiary centers, without the advantage of
1 in 5000 Caucasian individuals.3,4 Approximately 60% of index cases genetic testing and without sensitive diagnostic criteria. Over the last
harbor mutations in genes encoding the cardiac desmosome, a structure decade, several collaborative efforts in the United States and Europe
that provides mechanical connection between cardiomyocytes.2,5 The have significantly enhanced our understanding of (the role of CMR
defective mechanical connections in ARVC may structurally be repre- in) ARVC diagnosis, clinical characteristics, and management. These
sented by global or regional contraction abnormalities, ventricular advancements are reviewed here.
enlargement, and/or fibrofatty replacement. As such, accurate evaluation
of cardiac morphology and function is essential for ARVC diagnosis DIAGNOSIS OF ARRHYTHMOGENIC RIGHT
and management.
ARVC constitutes a diagnostic challenge for the cardiovascular
VENTRICULAR CARDIOMYOPATHY
magnetic resonance (CMR) physician. The variation in shape and Diagnosis of ARVC is challenging because no single modality is suf-
contraction patterns of the normal RV complicates identification of ficiently specific to establish an ARVC diagnosis. Therefore multiple
early, subtle RV disease. In addition, many imaging centers have little sources of diagnostic information are combined in a complex set of
experience with evaluating ARVC, and gaining experience is difficult diagnostic criteria. Definite ARVC diagnosis is based on the presence
because of the low prevalence of disease. Missing a diagnosis of ARVC of major and minor criteria encompassing structural, histologic, elec-
may prove to be fatal, whereas overdiagnosis may lead to therapeutic trocardiographic, arrhythmic, and family history criteria (Box 34.1).9
repercussions in essentially healthy individuals. Despite these chal- These “Task Force” criteria (TFC) were first established in 1994 and
lenges, CMR is generally acknowledged to be the “best” noninvasive updated in 2010 to improve (1) the specificity of the TFC by includ-
test for ARVC diagnosis and to distinguish ARVC from other cardio- ing quantitative metrics for diagnosis; and (2) sensitivity of the TFC
myopathies. Thus the aim of this chapter is to review current knowl- in individuals who have a high likelihood of inherited/genetic disease.
edge of ARVC that is useful for CMR interpretation of individuals Structural criteria for ARVC diagnosis may be based on echocardiog-
suspected of this disease. Our emphasis will be on ARVC diagnosis and raphy, angiography, or CMR. However, echocardiographic evalua-
management, including diagnostic criteria, CMR acquisition/analysis, tion of the RV is difficult because of its complex geometry,10 and a
and common regional morphologic and functional abnormalities recent study has shown that echocardiographic evaluation of subtle
in ARVC. RV changes may be unreliable.11 In addition, angiography is invasive
and should not be employed as a first-line screening test for ARVC.
A HISTORICAL PERSPECTIVE ON In contrast, CMR allows for noninvasive multiplane morphologic and
ARRHYTHMOGENIC RIGHT VENTRICULAR functional evaluation, as well as tissue characterization in a single
investigation, and has emerged as the imaging modality of choice
CARDIOMYOPATHY in ARVC.12
The first comprehensive clinical description of ARVC dates back to 1982,
when Marcus and colleagues1 described a cohort of 24 patients with Cardiovascular Magnetic Resonance Task Force
arrhythmias originating from the RV and (right) heart failure. Using Criteria and Their Derivation
echocardiography, angiography, and histologic specimens obtained at The diagnostic TFC for CMR require the presence of both qualitative
surgery, they described ARVC as a regional RV disease with paper-thin findings (RV regional akinesia, dyskinesia, dyssynchronous contraction)
and quantitative metrics (decreased ejection fraction or increased indexed
RV end-diastolic volume) (see Box 34.1).9 Quantitative values for RV
a
Modified from Te Riele AS, Tandri H, Bluemke DA. Arrhythmogenic volume and function for TFC were derived from a comparison of ARVC
right ventricular cardiomyopathy/dysplasia (ARVC/D): cardiac magnetic probands with normal healthy volunteers that were included in the
resonance update. J Cardiovasc Magnet Reson. 2014;16:50. Multi-Ethnic Study of Atherosclerosis (MESA).13 To ascertain cutoff

410
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathy 411

BOX 34.1  Revised 2010 Task Force Criteria for Arrhythmogenic Right Ventricular
Cardiomyopathy
Global or Regional Dysfunction and Structural Alterations Minor
Major Inverted T waves in V1 and V2 in individuals >14 years of age (in the absence
Two-Dimensional Echocardiography Criteria of complete RBBB) or in V4, V5, and V6
Regional RV akinesia, dyskinesia, or aneurysm and one of the following measured Inverted T waves in leads V1, V2, V3, and V4 in individuals >14 years of age in
at end diastole: the presence of complete RBBB
• PLAX RVOT ≥32 mm (PLAX/BSA ≥19 mm/m2), or
• PSAX RVOT ≥36 mm (PSAX/BSA ≥21 mm/m2), or Depolarization/Conduction Abnormalities
• Fractional area change ≤33% Major
Epsilon wave (reproducible low-amplitude signals between end of QRS complex
Cardiovascular Magnetic Resonance Criteria to onset of T wave) in the right precordial leads (V1, V2, or V3)
Regional RV akinesia or dyskinesia or dyssynchronous RV contraction and one
of the following: Minor
• RV EDV/BSA ≥110 mL/m2 (male) or ≥100 mL/m2 (female), or Late potentials by SAECG in ≥1 of 3 parameters in the absence of a QRS duration
• RV ejection fraction ≤40% of ≥110 ms on standard ECG:
• Filtered QRS duration (fQRS) ≥114 ms
Right Ventricular Angiography Criteria • Duration of terminal QRS <40 µV ≥38 ms
Regional RV akinesia, dyskinesia, or aneurysm • Root-mean-square voltage of terminal 40 ms ≤20 µV
Terminal activation duration ≥55 ms measured from the nadir of the end of all
Minor depolarization deflections, including R′, in V1, V2, or V3 in absence of complete
Two-Dimensional Echocardiography Criteria RBBB
Regional RV akinesia or dyskinesia or dyssynchronous RV contraction and one
of the following measured at end diastole: Arrhythmias
• PLAX RVOT ≥29 to <32 mm (PLAX/BSA ≥16 to <19 mm/m2), or Major
• PSAX RVOT ≥32 to <36 mm (PSAX/BSA ≥18 to <21 mm/m2), or Nonsustained or sustained VT of LBBB morphology with superior axis
• Fractional area change >33% ≤40%
Minor
Cardiovascular Magnetic Resonance Criteria Nonsustained or sustained VT of RVOT configuration, LBBB morphology with
Regional RV akinesia or dyskinesia or dyssynchronous RV contraction and one inferior axis or of unknown axis
of the following: >500 PVCs per 24 hours on Holter monitoring
• RV EDV/BSA ≥100 to 110 mL/m2 (male) or ≥90 to 100 mL/m2 (female)
• RV ejection fraction >40 to ≤45% Family History
Major
Tissue Characterization of Wall ARVC in first-degree relative who meets Task Force Criteria
Major ARVC confirmed pathologically at autopsy or surgery in first-degree relative
Residual myocytes <60% by morphometric analysis (or <50% if estimated), with Identification of pathogenic mutation categorized as associated or probably
fibrous replacement of the RV free wall myocardium in ≥1 sample, with or associated with ARVC in the patient under evaluation
without fatty replacement of tissue on endomyocardial biopsy
Minor
Minor History of ARVC in first-degree relative in whom it is not possible to determine
Residual myocytes 60% to 75% by morphometric analysis (or 50% to 65% if whether the family member meets Task Force Criteria
estimated), with fibrous replacement of the RV free wall myocardium in ≥1 Premature sudden death (<35 years of age) due to suspected ARVC in first-degree
sample with or without fatty replacement of tissue on endomyocardial biopsy relative
ARVC confirmed pathologically or by current Task Force Criteria in second-degree
Repolarization Abnormalities relative
Major
Inverted T waves in right precordial leads (V1, V2, and V3) or beyond in individuals
>14 years of age (in the absence of complete RBBB)

ARVC, Arrhythmogenic right ventricular cardiomyopathy; BSA, body surface area; ECG, electrocardiogram; EDV, end-diastolic volume; LBBB, left
bundle branch block; PLAX, parasternal long axis; PSAX, parasternal short axis; PVCs, premature ventricular complexes; RBBB, right bundle
branch block; RV, right ventricular; RVOT, right ventricular outflow tract; SAECG, signal-averaged electrocardiogram; VT, ventricular tachycardia.
Modified from Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed
modification of the task force criteria. Circulation. 2010;121:1533–1541.

values, RV dimension and function from 462 normal MESA participants invariably result in lower sensitivity; this accounts for the major CMR
were compared with 44 probands in the North American ARVC registry.9 criteria sensitivity of 68% to 76%.14 Minor criteria (RV ejection fraction
Major criteria (RV ejection fraction ≤40% or indexed RV end-diastolic 40%–45% or indexed RV end-diastolic volume 100–110 mL/m2 for men
volume ≥110 mL/m2 for men and ≥100 mL/m2 for women) were chosen and 90–100 mL/m2 for women) had a higher sensitivity (79%–89%),
to achieve approximately 95% specificity. Cutoffs with high specificity but a consequently lower specificity (85%–97%).14
412 SECTION III  Functional Disease

example, the CMR cutoff values for RV size in ARVC (see Box 34.1)
Quantitative Evaluation of the Right Ventricle in the are ≥110 mL/m2 (male) or ≥100 mL/m2 (female) for major criteria.
Revised Task Force Criteria These values closely correspond to the 95th percentile confidence limits
Several studies have shown that including quantitative metrics as a of RV volumes for normal subjects less than 60 years of age.21,22 Thus
component to the CMR TFC increased its diagnostic value, mainly until further studies are available, we feel that the current RV metrics
through an increase in positive predictive value.15–17 However, the CMR in the revised TFC remain highly relevant. Important further develop-
physician should be aware of its limitations. First, although quantitative ments are necessary to improve the reproducibility of RV quantification
measures reduce subjectivity of CMR TFC, there is substantial inter- by CMR. When evaluating younger patients, CMR physicians should
reader variability for the RV. In our experience, two physician readers keep in mind that the size of the RV is expected to be ~10% larger in
had excellent agreement (within ~5% of reference values) only after a 20-year-old patient compared with a 40-year-old patient.
training on approximately 100 CMR cases.18 In clinical practice, we
expect it would be difficult to achieve reproducibility of less than 10% CARDIOVASCULAR MAGNETIC RESONANCE
for RV parameters. In addition, cutoffs for ARVC criteria were derived ACQUISITION FOR SUSPECTED ARRHYTHMOGENIC
from the MESA study that used the fast gradient recalled echo (FGRE)
technique, whereas a majority of study subjects in the US ARVC study
RIGHT VENTRICULAR CARDIOMYOPATHY
had steady-state free precession (SSFP) cine images. RV volumes by SSFP The CMR protocol that we recommend for evaluation of patients with
are at least 10% larger than those measured with the FGRE technique.19,20 possible ARVC is shown in Table 34.1. The protocol has been designed
SSFP images provide superior contrast between blood and endocardium to evaluate the RV for abnormalities in structure and tissue character-
at the endocardial border, with less blood flow dependence.19,20 Also, ization while enabling quantitative evaluation. For black-blood imaging,
the revised TFC used MESA subjects whose mean age was 60 years. fast spin echo, or turbo spin echo imaging sequences are ideal. The RV
ARVC subjects in the Task Force study were 20 to 30 years younger on free wall and RVOT are best evaluated in the axial black-blood images.
average. Since that time, Chahal et al.18 determined that, among MESA The stack of axial images should include the entire RV. This can be
participants aged 45 to 84 years old, RV end-diastolic volume decreased accomplished in 6 to 8 slices at intervals (slice thickness + gap) of about
4.6% per decade. A very similar percentage of approximately 4% decrease 1 cm. Most of the diagnostic information is obtained in the slices cen-
per decade was obtained by Maceira et al.21 using the SSFP technique tered on the middle of the RV. Obtaining an excessive number of image
in subjects aged 20 to 80 years old. Adjusting for body surface area did slices will adversely prolong the examination.
not remove the dependence of RV volume on age. For cine imaging, SSFP imaging is preferred at 1.5 T. Insufficient
Fortuitously, the issues of pulse sequence and older reference popu- information at 3 T is available to determine if SSFP or FGRE is superior.
lation in the MESA study approximately balance each other. As an Quantitative analysis of the RV and left ventricle (LV) is performed on

TABLE 34.1  Recommended Cardiovascular Magnetic Resonance Protocol for Suspected


Arrhythmogenic Right Ventricular Cardiomyopathy
Sequence Imaging Plane Parameters Comments
Double inversion recovery TSE/FSE (a) Axial: obtain ~6–8 images TR = 2 RR intervals, TE = 5 ms This sequence provides optimal tissue
(a) Axial: with and without fat centered on the left/right (minimum-full) (GE), TE = 30 ms characterization of the RV free wall.
suppression ventricle (Siemens), slice thickness = 5 mm, Prescribe from the pulmonary artery
(b) Short axis: without fat (b) Short axis: obtain ~6–8 interslice gap = 5 mm, and FOV = to the diaphragm. Fat suppression
suppression images centered on the left 28–34 cm improves reader confidence in
ventricle ETL = 16–24 diagnosis of RV fat infiltration.
SSFP bright-blood cine images Axial, four-chamber, and short TR/TE minimum, flip angle = 45–70 Axial images are best to assess RV
axis. RV three-chamber degrees, slice thickness = 8 mm, wall motion. RV quantitative
(optional) interslice gap = 2 mm analysis is performed on the
FOV = 36–40 cm, 16–20 views per short-axis cine images.
segment. Parallel imaging N = 2 is
desirable
Gadolinium is administered according to institutional protocol (usually 0.15–0.2 mmol/kg).
TI scout Four-chamber TI scout sequences or trial TI times to
suppress normal myocardium for
the right inversion time
Delayed gadolinium imaging Axial, short axis, four-chamber, TR/TE per manufacturer PSIR is more robust and independent
(fast gradient echo technique with and vertical long axis recommendations flip angle = 20–25 of TI time. Optimal for imaging
PSIR recommended) degrees, slice thickness = 8 mm, fibrosis. LV epicardial enhancement
interslice gap = 2 mm in the inferolateral wall has been
FOV = 36–40 cm. No parallel imaging. reported in classic ARVC and in left
Use PSIR, if available. dominant forms.

ARVC, Arrhythmogenic right ventricular cardiomyopathy, CMR, cardiovascular magnetic resonance; ETL, echo-train length; FOV, field of view;
FSE, fast spin echo; GE, General Electric; LV, left ventricle; PSIR, phase-sensitive inversion recovery, RV, right ventricle; SSFP, steady-state free
precession; TE, echo time; TI, inversion time; TR, repetition time; TSE, turbo spin echo.
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathy 413

short-axis images. Thus 10 to 12 slices encompassing the entire ven- Regional Systolic Wall Motion Abnormalities
tricular volume must be obtained. We prescribe these images beginning Most of our information about structural abnormalities in ARVC comes
approximately 1 cm above the valve plane and increment toward the from studies in subjects with a predominant RV phenotype. Besides
apex of the ventricles. Cine images should also be obtained in standard global reduction in RV function, more subtle regional disease of the
long-axis views of the LV. Some sites prefer to also acquire a vertical RV has been described in the literature using a variety of terms, includ-
long-axis view of the RV. Finally, we routinely obtain a stack of transaxial ing focal bulges, microaneurysms, segmental dilatation, and regional
images of the RV at the same slice positions as the black-blood images hypokinesis. In the current TFC, the terms “akinesia” (lack of motion)
described above. Given modern CMR scanners, the temporal resolution and “dyskinesia” (abnormal movement, instead of contracting in systole,
of cine images is typically around 40 ms. the myocardium bulges outward in systole), and “dyssynchronous”
Delayed gadolinium images are best obtained using phase-selective (regional peak contraction occurring at different times in adjacent
inversion recovery (PSIR), a sequence which does not depend upon myocardium) are used for all imaging modalities (CMR, echocardiog-
identifying the precise inversion time (TI).23 In patients with significant raphy, and angiography) to describe regional wall motion abnormalities
ventricular ectopy, a low dose of a beta-blocker (metoprolol 25–50 mg) in ARVC.9 Characteristic examples of RV wall motion abnormalities
is recommended for arrhythmia suppression during the CMR scan. are shown in Fig. 34.1. Microaneurysms are not explicitly described in
the revised TFC; overuse of this term was considered by the Task Force
CARDIOVASCULAR MAGNETIC RESONANCE members to be misinterpreted by CMR physicians, resulting in false-
FINDINGS ASSOCIATED WITH ARRHYTHMOGENIC positive diagnoses. However, microaneurysms are characterized by
regional akinesia/dyskinesia in the revised criteria.
RIGHT VENTRICULAR CARDIOMYOPATHY The location of regional wall motion abnormalities of the RV was
Use of CMR in ARVC was first described in 1987, when Casolo et al.8 not addressed in the revised TFC. We now recognize that the distal RV
demonstrated intramyocardial fat deposits using spin echo sequences (from the moderator band to the apex in long-axis views) shows highly
in an ARVC patient. Two years later, Wolf and colleagues24 described variable contraction patterns in the normal individual. In addition, a
“dysplastic lesions in the RV wall presenting as fat-like high signals” in recent study has shown that the RV apex is spared in early ARVC.30
7 ARVC patients. Over the years that followed, CMR findings associated Therefore we emphasize the significance of regional wall motion abnor-
with ARVC include RV wall thinning, RVOT enlargement, trabecular malities in the subtricuspid region in ARVC. An excellent example of
disarray, fibrofatty replacement, ventricular dilatation, and global systolic this is the “accordion sign” that represents a focal “crinkling” of the
dysfunction.25–29 However, our current emphasis is on regional wall myocardium31 (Fig. 34.2). In terms of TFC, the accordion sign is caused
motion abnormalities as the earliest and most reliable abnormalities by a small region of highly localized myocardium with dyssynchronous
identified by CMR. contraction.

A B

C D
FIG. 34.1  Four-chamber (A–B) and short-axis (C–D) bright-blood images in an arrhythmogenic right ventricular
cardiomyopathy subject with predominant right ventricular abnormalities. End-diastolic images are shown in
the left panels, end-systolic images in the right panels. Note subtricuspid dyskinesia in the end-systolic four-
chamber image (arrow), and right ventricular free wall aneurysms (i.e., both systolic and diastolic bulging) in
the short-axis image (arrows). Please also note subepicardial fatty infiltration in the apicolateral left ventricle
(arrowhead in B).
414 SECTION III  Functional Disease

RV RV

RA

LV LV
RA

A B
FIG. 34.2  Regional contraction abnormality in the subtricuspid region. End-diastolic (A) and end-systolic
(B) images show the so-called accordion sign (arrow) in an arrhythmogenic right ventricular cardiomyopathy
mutation carrier. Regional dyssynchronous contraction in the subtricuspid region is a readily recognized
qualitative pattern of abnormal right ventricular contraction. LV, Left ventricle; RA, right atrium; RV, right
ventricle.

A B
FIG. 34.3  Right ventricular late gadolinium enhancement (LGE) in arrhythmogenic right ventricular cardio-
myopathy. The short-axis image (A) shows LGE in the right ventricle as well as the left ventricle (black
arrows). The lateral wall of the left ventricle shows thinning because of fatty replacement (white arrow) that
was confirmed on T1-weighted images. The long axis view (B) shows diffuse LGE involving the free wall of
the right ventricle (black arrows).

provide guidance for electrophysiologic studies and endomyocardial


Late Gadolinium Enhancement biopsy. LGE CMR is also extremely useful when ARVC is excluded as
Myocardial late gadolinium enhancement (LGE) is a well-validated a result of other cardiomyopathy such as sarcoidosis. In our opinion,
technique for assessment of myocardial fibrosis, and has frequently the presence of LGE represents a late stage of ARVC; when LGE is
been associated with ARVC: RV LGE has been observed in up to 88% present, multiple other findings of advanced disease are typically present,
of ARVC patients,28,32 whereas LV LGE was reported in up to 61% of such as RV dilatation and dysfunction.28 Importantly, before LGE can
cases.28,33,34 A characteristic example is shown in Fig. 34.3. Given that be included in a future iteration of the TFC, more data regarding the
one of the pathologic hallmarks of ARVC is fibrofatty replacement of specific patterns of LGE that distinguish ARVC from other cardiomy-
the myocardium, it is important to note that LGE is not incorporated opathies are necessary. Also, improved methods to determine fibrosis
in the current diagnostic TFC. Although the Task Force did recognize in the thin RV wall are needed. Until such a method emerges, the
the presence of LGE in many patients with ARVC, several limitations presence of LGE should be considered as diagnostic confirmation, not
withheld its inclusion in the TFC. First, detection of LGE in the RV sole evidence of ARVC disease expression.
is greatly hampered by the thin RV wall. In ARVC, RV wall thinning
is pronounced, which makes the LGE technique less reliable than for Left Ventricular Involvement
the LV. Second, distinguishing fat from fibrosis by LGE sequences is The advent of genetic testing and use of sensitive TFC have significantly
challenging, which makes its interpretation highly subject to the CMR increased our awareness of the phenotypic spectrum of ARVC, which
physician’s experience. Last, LV LGE is nonspecific and has a wide dif- also includes nonclassical (e.g., left-dominant and biventricular) phe-
ferential diagnosis. LGE may be observed in many mimics of ARVC, notypes. As a result, we now know that some ARVC subjects have early
such as sarcoidosis, myocarditis, amyloidosis, and dilated cardiomy- and predominant LV involvement.34,35 LV involvement has even been
opathy (DCM). reported in 76% of ARVC subjects, of whom the majority had advanced
However, keeping these limitations in mind, LGE may be very useful disease.36 The disease is therefore increasingly being referred to as
in ARVC evaluation. Tandri et al.32 showed excellent correlation of RV “arrhythmogenic cardiomyopathy.”34,37
LGE with histopathology and inducible ventricular arrhythmias on In 2010, Jain led a study investigating LV regional dysfunction using
electrophysiologic study. Identification of LGE by CMR may therefore CMR tagging, and found that LV peak systolic strain was lower in ARVC
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathy 415

A B

C D
FIG. 34.4  Horizontal long-axis (A–B) bright-blood and late gadolinium enhancement images (C–D) in an
arrhythmogenic right ventricular subject with predominant left ventricular abnormalities. Note a dilated left
ventricle in the bright-blood images. Late enhancement is observed in a midmyocardial pattern in the basal
septum and basal lateral wall (arrows in C–D).

Misdiagnosis of Arrhythmogenic Right Ventricular


Cardiomyopathy Using Cardiovascular
Magnetic Resonance
Misdiagnosis of ARVC is a well-recognized problem. A prior study
has shown that more than 70% of patients who were referred to Johns
Hopkins Hospital from outside institutions with a diagnosis of ARVC
did not actually meet diagnostic TFC.41 In many cases, CMR misin-
terpretation is the cause of overdiagnosis in ARVC.41,42 It is important
to realize that, although CMR may be regarded as the standard of
reference for evaluation of RV morphology and function, the use of
CMR alone is not the “gold standard” for ARVC diagnosis. Rather,
the TFC prescribe the use of multiple diagnostic tests.9 Great caution
must be employed when the only abnormality in a presumed ARVC
patient is found on CMR because it is uncommon for ARVC patients
FIG. 34.5  Horizontal long-axis bright-blood image in an arrhythmogenic
to have a normal electrocardiogram (ECG) and Holter monitor but an
right ventricular cardiomyopathy patient revealing left ventricular lateral
wall fatty infiltration with myocardial wall thinning (arrowhead).
abnormal CMR.43,44
ARVC misdiagnosis may occur because of an over-reliance on a
single morphologic imaging finding, such as fatty replacement.41,42
subjects compared with controls.38 Sen-Chowdhry et al.34 recently pub- The focus on fatty replacement in ARVC evaluation is understand-
lished data supporting a genetic association between left-dominant able because fibrofatty replacement of the myocardium is a unique
ARVC and classical right-sided ARVC. In their study, the authors showed pathologic hallmark of ARVC. However, intramyocardial fat also occurs
that one-third of genotyped ARVC patients with a left-dominant phe- physiologically in older, obese patients (Fig. 34.6). In addition, mul-
notype have a pathogenic mutation in the ARVC-related desmosomal tiple reports have shown that CMR detection of intramyocardial fat
genes. Phenotypic variations of predominant RV and LV involvement was not reproducible even among experienced readers, constituting an
even coexisted in the same family. important cause of misdiagnosis.45–47 An apparently benign condition,
LV involvement in ARVC typically manifests as LGE, often involving lipomatous infiltration of the RV anterior wall shows abundant RV fat
the inferior and lateral walls without concomitant wall motion abnor- but does not demonstrate regional wall motion abnormality, which is
malities.29,39 Fig. 34.4 shows an example. Septal LGE is present in more otherwise required for ARVC diagnosis.46 In summary, replacement
than 50% of cases with left-dominant ARVC, in contrast with the right- of the myocardium by fat is not specific for ARVC in the absence of
dominant pattern in which septal involvement is unusual. In addition, functional abnormalities.48
LV fatty infiltration was shown to be a prevalent finding in ARVC, often Furthermore, normal variants may mimic ARVC. A comprehen-
involving the subepicardial lateral LV and resulting in myocardial wall sive description of ARVC mimics is available elsewhere.49 In short,
thinning (Fig. 34.5).29,40 Future studies are necessary to confirm these important normal variants previously mistaken for ARVC are pectus
data, and further our understanding of LV abnormalities in ARVC. excavatum,50 apical-lateral bulging of the RV free wall at the insertion
416 SECTION III  Functional Disease

of the moderator band,51 and the “butterfly apex,” a normal anatomic


variant of separate RV and LV apices causing the RV apex to look
dyskinetic.49 We have found the butterfly appearance of the apex to
be more common on horizontal long-axis views at inferior levels (Fig.
34.7). In addition, a prominent band of pericardial connective tissue
joining the RV free wall to the posterior sternum may lead to mis-
interpretation of RV wall motion; this “tethered” portion of the RV
free wall remains static in location and may be misinterpreted as RV
dyskinesia (Fig. 34.8).
In our experience, sarcoidosis is the most common mimic of ARVC,
but other disorders, such as DCM and myocarditis, may also have diag-
nostic overlap with ARVC.52,53 Table 34.2 shows a list of clinical features
and CMR findings of conditions potentially confused with ARVC.54
Compared with DCM, patients with (left-dominant) ARVC often have
significant ventricular arrhythmias, disproportionate to the morpho-
logic abnormalities and impaired LV systolic function. In addition,
inflammatory processes, such as (viral) myocarditis, may mimic ARVC.53
In myocarditis, T2-weighted imaging may detect tissue edema, which
is usually absent in ARVC. In addition, fast spin echo T1-weighted
images during the first minutes after contrast injection may be useful
FIG. 34.6  A 45-year-old female with abundant lipomatous replacement to detect myocardial hyperemia and muscular inflammation suggestive
of the anterior right ventricular wall by fat. This condition appears to be of myocarditis.53 In equivocal cases, invasive studies, such as electro-
associated with obesity and abundant pericardial and mediastinal fat. anatomic mapping and endomyocardial biopsy, may provide a more
Right ventricular wall motion was normal. definite diagnosis.

1 2

A B

3 4

C D
FIG. 34.7  Butterfly apex as a normal variation. Stack of horizontal long-axis views from inferior (A) to superior
(D) in a control subject. Note the appearance of a butterfly apex on inferior views (arrows in A–C). This
appearance is not seen on the more superior view (D). This is a normal variant.
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathy 417

A B
FIG. 34.8  Misdiagnosis of arrhythmogenic right ventricular cardiomyopathy: axial (A) and short-axis
(B) bright-blood images in a control subject. Note the “tethering” of the mid right ventricular free wall to the
anterior chest wall (arrows), giving the right ventricle a dyskinetic appearance.

TABLE 34.2  Clinical Findings of Conditions Considered in the Differential Diagnosis of


Arrhythmogenic Right Ventricular Cardiomyopathy
Condition Finding
Sarcoidosis Clinical findings: more common in women, African-Americans, and Northern European (Scandinavian) whites. Usually nonfamilial
disease pattern. May present with extracardiac manifestations (often pulmonary/mediastinal lymphadenopathy, but any organ
system may be involved). ECG may show PR interval prolongation and/or high-grade atrioventricular block.
CMR findings: pulmonary granulomas, mediastinal lymphadenopathy, decreased LV function/heart failure, LGE (nonischemic pattern)
in the interventricular septum.
Myocarditis Clinical findings: may present with a viral prodrome with fever, myalgia, respiratory and gastrointestinal symptoms, new-onset atrial
or ventricular arrhythmias, complete heart block, or acute myocardial infarction-like syndrome with chest pain, ST-T changes, and
elevated cardiac enzymes.
CMR findings: increased T2 signal due to tissue edema (acute phase), concomitant pericardial involvement, subepicardial patchy
myocardial LGE (nonischemic pattern).
DCM Clinical findings: variable. May be familial. Ventricular arrhythmias usually occur in the context of impaired LV systolic function and
morphologic abnormalities.
CMR findings: dilated LV with reduced function, midwall LGE enhancement in the septum.
Athlete’s heart Clinical findings: clinical history is indicative; subjects are typically engaged in intense and repetitive endurance type sports. Physical
examination may show low heart rate, especially in young athletes.
CMR findings: balanced dilatation of cardiac chambers, increased ventricular wall thickness (<15 mm), absence of regional ventricular
dysfunction or regional wall motion abnormalities; lack of LGE on CMR.
Idiopathic RVOT VT Clinical findings: benign, nonfamilial condition, only one VT morphology (LBBB with inferior axis), sinus rhythm ECG normal.
CMR findings: normal.
Brugada syndrome Clinical findings: ECG reveals RBBB and persistent ST segment elevation in the right precordial leads (spontaneous or after
provocative drug challenge). Arrhythmias often occur in a sedentary setting (after a meal or at rest due to high vagal tone).
Imaging findings: traditionally considered to be normal, although structural disease is increasingly reported.

CMR, Cardiovascular magnetic resonance imaging; DCM, dilated cardiomyopathy; ECG, electrocardiogram; LBBB, left bundle branch block; LGE,
late gadolinium enhancement; LV, left ventricular; PET, positron emission tomography; RBBB, right bundle branch block; RVOT, right ventricular
outflow tract; VT, ventricular tachycardia.
From Te Riele ASJM, Tandri H, Sanborn DM, Bluemke DA. Noninvasive multimodality imaging in arrhythmogenic right ventricular dysplasia/
cardiomyopathy. JACC Cardiovasc Imaging. 2015;8(5):597–611.

GENETIC BACKGROUND OF ARRHYTHMOGENIC most common gene involved is plakophilin-2, followed by desmoglein-2,
desmocollin-2, and desmoplakin.2,55 Prevalence of mutations is similar
RIGHT VENTRICULAR CARDIOMYOPATHY
in Europe, although desmoplakin mutations are more prevalent in the
Beginning with the seminal discovery of plakoglobin in 2000, the past United Kingdom and Italy.56,57 Desmosomes are complex multiprotein
decade has witnessed the identification of mutations in five genes encod- structures providing mechanical and electrical continuity to adjacent
ing the cardiac desmosome.5 In recent reports, desmosomal mutations cells. Mechanical uncoupling in ARVC is accompanied by cell death
are found in up to 60% of ARVC cases.2 A cartoon representation of and regional fibrosis, which causes the monomorphic arrhythmias typi-
the desmosome is shown in Fig. 34.9. Among US ARVC patients, the cally associated with ARVC.58 In addition, electrical uncoupling through
418 SECTION III  Functional Disease

Desmoplakin Desmocollin Plakoglobin

Keratin IF Desmoglein Plakophilin

FIG. 34.9  Cartoon representation of the desmosome. IF, Intermediate filament. (From Fuchs E, Raghavan
S. Getting under the skin of epidermal morphogenesis. Nat Rev Genet. 2002;3:199–209.)

gap junction remodeling and sodium channel dysfunction may lead to ARRHYTHMOGENIC RIGHT VENTRICULAR
significant activation delay, which increases the propensity to functional
CARDIOMYOPATHY MANAGEMENT
block and arrhythmia.58
ARVC is generally transmitted as an autosomal dominant trait with ARVC management is directed toward symptom reduction, delay of
incomplete penetrance and variable expressivity.59 However, it is impor- disease progression, and prevention of SCD. Because of a lack of ran-
tant to realize that 50% to 70% of mutation carriers will never develop domized trials comparing ARVC treatment options, management rec-
disease expression, and that severity of disease may vary greatly, even ommendations in ARVC are largely based on clinical judgments and
among members of the same family or those carrying the same muta- results from retrospective registry-based studies. Mainstay therapies
tion.5 In contrast, a negative genetic test result in a proband does not consist of conservative measures (exercise restriction), beta-blocking
exclude the possibility of disease, nor does it exclude the possibility of and antiarrhythmic agents, implantable cardioverter defibrillator (ICD)
a genetic process in the individual or family.59,60 Because of the com- implantation, and radiofrequency ablation of ventricular arrhythmias.
plexities associated with interpreting genetic test results in ARVC, includ- Evidence for a potential role of exercise in ARVC expression and
ing genetic counseling prior and subsequent to genetic testing has been disease progression is accumulating. Many ARVC patients are highly
strongly recommended.59 athletic and those who participate in competitive sports have a 5-fold
increased risk of arrhythmic death compared with nonathletes.63 Recently,
Cardiovascular Magnetic Resonance in Genotype– James64 led a study on the role of exercise in ARVC development, showing
Phenotype Associations that endurance exercise and frequent athletics increases the risk of
Over the last decade, several genotype–phenotype correlations in ARVC arrhythmias and heart failure in ARVC mutation carriers. A subsequent
have been proposed, but large-scale studies confirming these observa- study showed that a similar trend exists among affected patients without
tions are yet to come. Recently, patients with multiple desmosomal a pathogenic mutation65 and unaffected family members who are at
mutations were shown to have a more severe clinical course with more risk of developing disease.66 This vital piece of evidence highlights the
ventricular arrhythmias and more heart failure than subjects with a importance of exercise restriction in ARVC patients and those at risk
single mutation.55,56 In addition, individuals with a mutation in the of developing disease.
phospholamban or desmoplakin gene (especially when involving the Arrhythmia control in ARVC is often achieved by pharmacologic
C-terminus of desmoplakin) have significant left-dominant/biventricular treatment. Beta-blockers and class III antiarrhythmic drugs (e.g., sotalol,
disease expression and a high prevalence of heart failure.55,61,62 LV involve- amiodarone) have been shown to be successful in reducing the arrhyth-
ment in these patients often manifests as LGE in a LV circumferential, mia burden and the likelihood of ICD discharge.67–69 In addition, radio-
mid-myocardial pattern extending to the right side of the septum34,39 frequency ablation for ventricular arrhythmia in ARVC has gained
(see Fig. 34.4). This left-dominant ARVC pattern should not be confused enormous popularity over the last years. Although the results of endo-
with LV involvement that occurs in advanced stages of right-dominant cardial ablation have been moderate,70 good arrhythmia control (but
ARVC. These right-dominant ARVC subjects (often plakophilin-2 muta- not complete cure) has been obtained using epicardial ablation.71,72
tion carriers) commonly have focal LV disease involving the lateral LV This is understandable, given the primary (sub)epicardial location of
wall with only mild or moderate LV dysfunction.39 Large-scale studies the abnormal substrate in ARVC. CMR with LGE may be useful in the
from collaborative international registries are necessary to further unmask planning of these procedures by providing information on the presence
genotype–phenotype associations in ARVC. and distribution of ventricular scar.73
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathy 419

Role of Cardiovascular Magnetic Resonance in such, it will likely not be long until studies on T1 mapping in ARVC
Arrhythmic Risk Stratification will find their way into the literature.
Once the diagnosis of ARVC is established in a patient, the most impor- Current diagnostic TFC for CMR require the presence of a wall
tant decision is whether to implant an ICD for prevention of SCD. It motion abnormality, which is subjective and thereby “in the eye of
is now standard of care for ARVC subjects with prior sustained ven- the beholder.”14 CMR feature tracking is a novel method for objective
tricular arrhythmia to undergo placement of an ICD.74 Studies have quantification of RV regional wall motion abnormalities. It is able to
shown that these patients have a high incidence rate of appropriate reliably track ventricular motion throughout the cardiac cycle, thereby
ICD discharges of up to 70% during a mean follow-up of 3 to 5 years.74,75 obtaining quantitative data on regional peak strain and strain rate.83 Two
Unfortunately, guidelines for ICD implantation among subjects without recent studies have shown that feature tracking is a feasible and reliable
prior ventricular arrhythmia are more ambiguous. Recent reports suggest tool to distinguish overt ARVC from controls.84,85 By providing objective
an important role for CMR in risk stratification of these patients, because ascertainment of regional ventricular function, CMR feature tracking
a normal CMR has a high negative predictive value for sustained ven- may also reduce misdiagnosis of ARVC.85 In addition, it may ascertain
tricular arrhythmias among ARVC-associated mutation carriers.43,76 timing of contractions, which provides information on cardiac electri-
Conversely, the presence of multiple CMR abnormalities identified a cal activation. Of note, the standard deviation of time-to-peak strain, a
high-risk group of ARVC patients who may benefit from ICD implanta- measure of dyssynchrony, was recently shown to have incremental value
tion.77 Therefore CMR provides important prognostic information in over conventional cine CMR imaging in ARVC evaluation.86 Although
individuals under evaluation for ARVC. A subsequent study showed these results are promising, the feature tracking technique will require
that “electrical” abnormalities on electrocardiogram, Holter monitor, improved standardization before routine use in ARVC evaluation is
or signal-averaged electrocardiogram precede detectable CMR changes advocated.
in ARVC.44 This suggests a strategy to optimize the use of CMR to
detect ARVC-associated mutation carriers at significant risk of arrhyth-
mic events, while limiting its use among those who are unlikely to
CONCLUSION
derive benefit from its results. ARVC is a rare but important cause of sudden cardiac death in young
individuals. In spite of its low prevalence, ARVC accounts for a dispro-
portionately high percentage of referrals for CMR. Because of the inher-
FUTURE PERSPECTIVE ON ARRHYTHMOGENIC ent risk of potentially lethal arrhythmias, correct diagnosis and early
RIGHT VENTRICULAR CARDIOMYOPATHY detection of ARVC are essential. Clinical ARVC diagnosis is facilitated
EVALUATION USING CARDIOVASCULAR by a complex set of diagnostic criteria, which were first described in
1994 and updated in 2010 to increase sensitivity for early disease. As
MAGNETIC RESONANCE the noninvasive “gold standard” for RV evaluation, CMR plays an impor-
LGE depends on differences in signal intensity between regions of scar- tant role in clinical ARVC workup. Recent studies have shown that RV
ring and adjacent normal myocardium. In subjects with diffuse fibrosis, involvement in ARVC often manifests as regional wall motion abnor-
these differences in signal intensity are lacking, thus resulting in false- mality or global ventricular dysfunction, whereas LV involvement is
negative results. Measurements of myocardial relaxation times (T1 often observed as LGE and/or fatty infiltration without concomitant
mapping) with native or gadolinium-enhanced recovery sequences wall motion abnormalities. ARVC preferentially affects the basal RV
provide a quantitative measure of the extracellular volume fraction, and lateral LV, while sparing the RV apex. In addition to diagnostic
reflecting diffuse interstitial myocardial fibrosis.78,79 T1 mapping provides evaluation, CMR may also play a role in the prognostic workup of
a quantitative measure of interstitial fibrosis, which has been shown to ARVC by risk stratification for arrhythmic events. New techniques,
have good correlation with ex vivo fibrotic measurements.80 Studies on such as T1 mapping and CMR feature tracking, may prove useful in
T1 mapping have proven the technique to be useful in DCM,81 myo- detecting the early stages of disease.
carditis,82 and hypertrophic cardiomyopathy (HCM).81 A challenge of
T1 mapping is that the technique has low spatial resolution, thus limiting
application to the LV rather than the RV. New research pulse sequences
REFERENCES
have been developed to substantially improve spatial resolution. As A full reference list is available online at ExpertConsult.com
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathya 419.e1

REFERENCES patients suspected of ARVC/D. Int J Cardiovasc Imaging. 2012;28:


595–601.
1. Marcus FI, Fontaine GH, Guiraudon G, et al. Right ventricular dysplasia: 24. Wolf JE, Rose-Pittet L, Page E, et al. [Detection of parietal lesions using
a report of 24 adult cases. Circulation. 1982;65:384–398. magnetic resonance imaging in arrhythmogenic dysplasia of the right
2. Groeneweg JA, Bhonsale A, James CA, et al. Clinical presentation, ventricle]. Arch Mal Coeur Vaiss. 1989;82:1711–1717.
long-term follow-up, and outcomes of 1001 ARVD/C patients and family 25. te Riele AS, Tandri H, Bluemke DA. Arrhythmogenic right ventricular
members. Circ Cardiovasc Genet. 2015;8:437–446. cardiomyopathy: cardiovascular magnetic resonance update. J Cardiovasc
3. Peters S, Trummel M, Meyners W. Prevalence of right ventricular Magn Reson. 2014;14.
dysplasia-cardiomyopathy in a non-referral hospital. Int J Cardiol. 26. Jain A, Tandri H, Calkins H, Bluemke DA. Role of cardiovascular
2004;97:499–501. magnetic resonance imaging in arrhythmogenic right ventricular
4. Basso C, Corrado D, Thiene G. Cardiovascular causes of sudden death in dysplasia. J Cardiovasc Magn Reson. 2008;10:32.
young individuals including athletes. Cardiol Rev. 1999;7:127–135. 27. Tandri H, Friedrich MG, Calkins H, Bluemke DA. MRI of
5. Murray B. Arrhythmogenic right ventricular dysplasia/cardiomyopathy arrhythmogenic right ventricular cardiomyopathy/dysplasia. J Cardiovasc
(ARVD/D): a review of molecular and clinical literature. J Genet Couns. Magn Reson. 2004;6:557–563.
2012;21:494–504. 28. Rastegar N, Te Riele AS, James CA, et al. Fibrofatty changes: incidence at
6. Basso C, Thiene G, Corrado D, Angelini A, Nava A, Valente M. cardiac MR imaging in patients with arrhythmogenic right ventricular
Arrhythmogenic right ventricular cardiomyopathy. Dysplasia, dystrophy, dysplasia/cardiomyopathy. Radiology. 2016;150988.
or myocarditis? Circulation. 1996;94:983–991. 29. Rastegar N, Zimmerman SL, Te Riele AS, et al. Spectrum of biventricular
7. Blake LM, Scheinman MM, Higgins CB. Mr features of arrhythmogenic involvement on CMR among carriers of ARVD/D-associated mutations.
right ventricular dysplasia. AJR Am J Roentgenol. 1994;162:809–812. JACC Cardiovasc Imaging. 2015;8:863–864.
8. Casolo GC, Poggesi L, Boddi M, et al. ECG-gated magnetic resonance 30. Te Riele AS, James CA, Philips B, et al. Mutation-positive arrhythmogenic
imaging in right ventricular dysplasia. Am Heart J. 1987;113:1245–1248. right ventricular dysplasia/cardiomyopathy: the triangle of dysplasia
9. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic displaced. J Cardiovasc Electrophysiol. 2013;24:1311–1320.
right ventricular cardiomyopathy/dysplasia: proposed modification of the 31. Dalal D, Tandri H, Judge DP, et al. Morphologic variants of familial
task force criteria. Circulation. 2010;121:1533–1541. arrhythmogenic right ventricular dysplasia/cardiomyopathy a genetics-
10. Boxt LM. Radiology of the right ventricle. Radiol Clin North Am. magnetic resonance imaging correlation study. J Am Coll Cardiol.
1999;37:379–400. 2009;53:1289–1299.
11. Borgquist R, Haugaa KH, Gilljam T, et al. The diagnostic performance of 32. Tandri H, Saranathan M, Rodriguez ER, et al. Noninvasive detection of
imaging methods in ARVC using the 2010 task force criteria. Eur Heart J myocardial fibrosis in arrhythmogenic right ventricular cardiomyopathy
Cardiovasc Imaging. 2014;15:1219–1225. using delayed-enhancement magnetic resonance imaging. J Am Coll
12. Galea N, Carbone I, Cannata D, et al. Right ventricular cardiovascular Cardiol. 2005;45:98–103.
magnetic resonance imaging: normal anatomy and spectrum of 33. Marra MP, Leoni L, Bauce B, et al. Imaging study of ventricular scar in
pathological findings. Insights Imaging. 2013;4:213–223. arrhythmogenic right ventricular cardiomyopathy: comparison of 3D
13. Tandri H, Daya SK, Nasir K, et al. Normal reference values for the adult standard electroanatomical voltage mapping and contrast-enhanced
right ventricle by magnetic resonance imaging. Am J Cardiol. cardiac magnetic resonance. Circ Arrhythm Electrophysiol. 2012;5:
2006;98:1660–1664. 91–100.
14. Bluemke DA. ARVC: imaging diagnosis is still in the eye of the beholder. 34. Sen-Chowdhry S, Syrris P, Prasad SK, et al. Left-dominant
JACC Cardiovasc Imaging. 2011;4:288–291. arrhythmogenic cardiomyopathy: an under-recognized clinical entity. J
15. Vermes E, Strohm O, Otmani A, Childs H, Duff H, Friedrich MG. Impact Am Coll Cardiol. 2008;52:2175–2187.
of the revision of arrhythmogenic right ventricular cardiomyopathy/ 35. Sen-Chowdhry S, Syrris P, Ward D, Asimaki A, Sevdalis E, McKenna WJ.
dysplasia task force criteria on its prevalence by CMR criteria. JACC Clinical and genetic characterization of families with arrhythmogenic
Cardiovasc Imaging. 2011;4:282–287. right ventricular dysplasia/cardiomyopathy provides novel insights into
16. Femia G, Hsu C, Singarayar S, et al. Impact of new task force criteria in patterns of disease expression. Circulation. 2007;115:1710–1720.
the diagnosis of arrhythmogenic right ventricular cardiomyopathy. Int J 36. Lindstrom L, Nylander E, Larsson H, Wranne B. Left ventricular
Cardiol. 2013;171:179–183. involvement in arrhythmogenic right ventricular cardiomyopathy—a
17. Luijkx T, Velthuis BK, Prakken NH, et al. Impact of revised task force scintigraphic and echocardiographic study. Clin Physiol Funct Imaging.
criteria: distinguishing the athlete’s heart from ARVC/D using cardiac 2005;25:171–177.
magnetic resonance imaging. Eur J Prev Cardiol. 2012;19:885–891. 37. van der Zwaag PA, van Rijsingen IA, Asimaki A, et al. Phospholamban
18. Chahal H, Johnson C, Tandri H, et al. Relation of cardiovascular risk r14del mutation in patients diagnosed with dilated cardiomyopathy or
factors to right ventricular structure and function as determined by arrhythmogenic right ventricular cardiomyopathy: evidence supporting
magnetic resonance imaging (results from the multi-ethnic study of the concept of arrhythmogenic cardiomyopathy. Eur J Heart Fail.
atherosclerosis). Am J Cardiol. 2010;106:110–116. 2012;14:1199–1207.
19. Malayeri AA, Johnson WC, Macedo R, Bathon J, Lima JA, Bluemke DA. 38. Jain A, Shehata ML, Stuber M, et al. Prevalence of left ventricular regional
Cardiac cine MRI: quantification of the relationship between fast dysfunction in arrhythmogenic right ventricular dysplasia: a tagged MRI
gradient echo and steady-state free precession for determination of study. Circ Cardiovasc Imaging. 2010;3:290–297.
myocardial mass and volumes. J Magn Reson Imaging. 2008;28:60–66. 39. Te Riele AS, Bhonsale A, Burt JR, Zimmerman SL, Tandri H. Genotype-
20. Moon JC, Lorenz CH, Francis JM, Smith GC, Pennell DJ. Breath-hold specific pattern of lv involvement in ARVD/D. JACC Cardiovasc Imaging.
FLASH and FISP cardiovascular MR imaging: left ventricular volume 2012;5:849–851.
differences and reproducibility. Radiology. 2002;223:789–797. 40. Berte B, Denis A, Amraoui S, et al. Characterization of the left-sided
21. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular substrate in arrhythmogenic right ventricular cardiomyopathy. Circ
systolic and diastolic function normalized to age, gender and body Arrhythm Electrophysiol. 2015;8:1403–1412.
surface area from steady-state free precession cardiovascular magnetic 41. Bomma C, Rutberg J, Tandri H, et al. Misdiagnosis of arrhythmogenic
resonance. Eur Heart J. 2006;27:2879–2888. right ventricular dysplasia/cardiomyopathy. J Cardiovasc Electrophysiol.
22. Kawel-Boehm N, Maceira A, Valsangiacomo-Buechel ER, et al. Normal 2004;15:300–306.
values for cardiovascular magnetic resonance in adults and children. J 42. Looi KL, Edwards C, Hart H, Christiansen JP. Utility of cardiac magnetic
Cardiovasc Magn Reson. 2015;17:29. resonance in the evaluation of unselected patients with possible
23. Plaisier AS, Burgmans MC, Vonken EP, et al. Image quality assessment of arrhythmogenic right ventricular cardiomyopathy. Clin Med Insights
the right ventricle with three different delayed enhancement sequences in Cardiol. 2012;6:153–162.
419.e2 SECTION III  Functional Disease

43. te Riele AS, Bhonsale A, James CA, et al. Incremental value of cardiac desmosomal or nondesmosomal genotype? Heart Rhythm.
magnetic resonance imaging in arrhythmic risk stratification of 2013;10:548–559.
arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated 63. Corrado D, Basso C, Rizzoli G, Schiavon M, Thiene G. Does sports
desmosomal mutation carriers. J Am Coll Cardiol. 2013;62:1761–1769. activity enhance the risk of sudden death in adolescents and young
44. Te Riele AS, James CA, Rastegar N, et al. The yield of serial evaluation in adults? J Am Coll Cardiol. 2003;42:1959–1963.
at-risk family members of arrhythmogenic right ventricular dysplasia/ 64. James CA, Bhonsale A, Tichnell C, et al. Exercise increases age-related
cardiomyopathy patients. J Am Coll Cardiol. 2014;64:293–301. penetrance and arrhythmic risk in arrhythmogenic right ventricular
45. Tandri H, Castillo E, Ferrari VA, et al. Magnetic resonance imaging of dysplasia/cardiomyopathy-associated desmosomal mutation carriers. J
arrhythmogenic right ventricular dysplasia: sensitivity, specificity, and Am Coll Cardiol. 2013;62:1290–1297.
observer variability of fat detection versus functional analysis of the right 65. Sawant AC, Bhonsale A, Te Riele AS, et al. Exercise has a disproportionate
ventricle. J Am Coll Cardiol. 2006;48:2277–2284. role in the pathogenesis of arrhythmogenic right ventricular dysplasia/
46. Macedo R, Prakasa K, Tichnell C, et al. Marked lipomatous infiltration of cardiomyopathy in patients without desmosomal mutations. J Am Heart
the right ventricle: MRI findings in relation to arrhythmogenic right Assoc. 2014;3:e001471.
ventricular dysplasia. AJR Am J Roentgenol. 2007;188:W423–W427. 66. Sawant AC, Te Riele AS, Tichnell C, et al. Safety of American Heart
47. Bluemke DA, Krupinski EA, Ovitt T, et al. MR imaging of Association-recommended minimum exercise for desmosomal mutation
arrhythmogenic right ventricular cardiomyopathy: morphologic findings carriers. Heart Rhythm. 2016;13:199–207.
and interobserver reliability. Cardiology. 2003;99:153–162. 67. Wichter T, Paul TM, Eckardt L, et al. Arrhythmogenic right ventricular
48. Basso C, Thiene G. Adipositas cordis, fatty infiltration of the right cardiomyopathy. Antiarrhythmic drugs, catheter ablation, or ICD? Herz.
ventricle, and arrhythmogenic right ventricular cardiomyopathy. Just a 2005;30:91–101.
matter of fat? Cardiovasc Pathol. 2005;14:37–41. 68. Marcus GM, Glidden DV, Polonsky B, et al; Multidisciplinary Study of
49. Rastegar N, Burt JR, Corona-Villalobos CP, et al. Cardiac MR findings Right Ventricular Dysplasia Investigators. Efficacy of antiarrhythmic
and potential diagnostic pitfalls in patients evaluated for arrhythmogenic drugs in arrhythmogenic right ventricular cardiomyopathy: a report from
right ventricular cardiomyopathy. Radiographics. 2014;34:1553–1570. the North American ARVC Registry. J Am Coll Cardiol. 2009;54:609–615.
50. Oezcan S, Attenhofer Jost CH, Pfyffer M, et al. Pectus excavatum: 69. Wichter T, Borggrefe M, Haverkamp W, Chen X, Breithardt G. Efficacy of
echocardiography and cardiac MRI reveal frequent pericardial effusion antiarrhythmic drugs in patients with arrhythmogenic right ventricular
and right-sided heart anomalies. Eur Heart J Cardiovasc Imaging. disease. Results in patients with inducible and noninducible ventricular
2012;13:673–679. tachycardia. Circulation. 1992;86:29–37.
51. Sievers B, Addo M, Franken U, Trappe HJ. Right ventricular wall motion 70. Philips B, Madhavan S, James C, et al. Outcomes of catheter ablation of
abnormalities found in healthy subjects by cardiovascular magnetic ventricular tachycardia in arrhythmogenic right ventricular dysplasia/
resonance imaging and characterized with a new segmental model. J cardiomyopathy. Circ Arrhythm Electrophysiol. 2012;5:499–505.
Cardiovasc Magn Reson. 2004;6:601–608. 71. Philips B, te Riele AS, Sawant A, et al. Outcomes and ventricular
52. Steckman DA, Schneider PM, Schuller JL, et al. Utility of cardiac tachycardia recurrence characteristics after epicardial ablation of
magnetic resonance imaging to differentiate cardiac sarcoidosis from ventricular tachycardia in arrhythmogenic right ventricular dysplasia/
arrhythmogenic right ventricular cardiomyopathy. Am J Cardiol. cardiomyopathy. Heart Rhythm. 2015;12:716–725.
2012;110:575–579. 72. Santangeli P, Zado ES, Supple GE, et al. Long-term outcome with catheter
53. Pieroni M, Dello Russo A, Marzo F, et al. High prevalence of myocarditis ablation of ventricular tachycardia in patients with arrhythmogenic right
mimicking arrhythmogenic right ventricular cardiomyopathy differential ventricular cardiomyopathy. Circ Arrhythm Electrophysiol. 2015;8:1413–1421.
diagnosis by electroanatomic mapping-guided endomyocardial biopsy. J 73. Andreu D, Ortiz-Perez JT, Boussy T, et al. Usefulness of contrast-
Am Coll Cardiol. 2009;53:681–689. enhanced cardiac magnetic resonance in identifying the ventricular
54. Te Riele AS, Tandri H, Sanborn DM, Bluemke DA. Noninvasive arrhythmia substrate and the approach needed for ablation. Eur Heart J.
multimodality imaging in ARVD/D. JACC Cardiovasc Imaging. 2014;35:1316–1326.
2015;8:597–611. 74. Corrado D, Calkins H, Link MS, et al. Prophylactic implantable
55. Bhonsale A, Groeneweg JA, James CA, et al. Impact of genotype on defibrillator in patients with arrhythmogenic right ventricular
clinical course in arrhythmogenic right ventricular dysplasia/ cardiomyopathy/dysplasia and no prior ventricular fibrillation or
cardiomyopathy-associated mutation carriers. Eur Heart J. sustained ventricular tachycardia. Circulation. 2010;122:1144–1152.
2015;36:847–855. 75. Piccini JP, Dalal D, Roguin A, et al. Predictors of appropriate implantable
56. Rigato I, Bauce B, Rampazzo A, et al. Compound and digenic defibrillator therapies in patients with arrhythmogenic right ventricular
heterozygosity predicts lifetime arrhythmic outcome and sudden cardiac dysplasia. Heart Rhythm. 2005;2:1188–1194.
death in desmosomal gene-related arrhythmogenic right ventricular 76. Te Riele AS, James CA, Groeneweg JA, et al. Approach to family screening
cardiomyopathy. Circ Cardiovasc Genet. 2013;6:533–542. in ARVD/C. Eur Heart J. 2016;37(9):755–763.
57. Sen-Chowdhry S, Syrris P, Pantazis A, Quarta G, McKenna WJ, Chambers 77. Deac M, Alpendurada F, Fanaie F, et al. Prognostic value of cardiovascular
JC. Mutational heterogeneity, modifier genes, and environmental magnetic resonance in patients with suspected ARVC. Int J Cardiol.
influences contribute to phenotypic diversity of arrhythmogenic 2013;168:3514–3521.
cardiomyopathy. Circ Cardiovasc Genet. 2010;3:323–330. 78. Burt JR, Zimmerman SL, Kamel IR, Halushka M, Bluemke DA.
58. Delmar M. Desmosome-ion channel interactions and their possible role Myocardial T1 mapping: techniques and potential applications.
in arrhythmogenic cardiomyopathy. Pediatr Cardiol. 2012;33:975–979. Radiographics. 2014;34:377–395.
59. Ackerman MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus 79. Moon JC, Messroghli DR, Kellman P, et al. Myocardial T1 mapping and
statement on the state of genetic testing for the channelopathies and extracellular volume quantification: a Society for Cardiovascular
cardiomyopathies. Heart Rhythm. 2011;8:1308–1339. Magnetic Resonance (SCMR) and CMR Working Group of the European
60. Ashley EA, Hershberger RE, Caleshu C, et al; American Heart Association Society of Cardiology consensus statement. J Cardiovasc Magn Reson.
Advocacy Coordinating Committee. Genetics and cardiovascular disease: 2013;15:92.
a policy statement from the American Heart Association. Circulation. 80. Aus dem Siepen F, Buss SJ, Messroghli D, et al. T1 mapping in dilated
2012;126:142–157. cardiomyopathy with cardiac magnetic resonance: quantification of
61. Sen-Chowdhry S, Syrris P, McKenna WJ. Desmoplakin disease in diffuse myocardial fibrosis and comparison with endomyocardial biopsy.
arrhythmogenic right ventricular cardiomyopathy: early genotype- Eur Heart J Cardiovasc Imaging. 2015;16:210–216.
phenotype studies. Eur Heart J. 2005;26:1582–1584. 81. Dass S, Suttie JJ, Piechnik SK, et al. Myocardial tissue characterization
62. Groeneweg JA, van der Zwaag PA, Jongbloed JD, et al. Left-dominant using magnetic resonance noncontrast t1 mapping in hypertrophic and
arrhythmogenic cardiomyopathy in a large family: associated dilated cardiomyopathy. Circ Cardiovasc Imaging. 2012;5:726–733.
CHAPTER 34  Arrhythmogenic Right Ventricular Cardiomyopathya 419.e3

82. Ferreira VM, Piechnik SK, Dall’Armellina E, et al. T(1) mapping for the 85. Vigneault DM, Te Riele AS, James CA, et al. Right ventricular strain by
diagnosis of acute myocarditis using CMR: comparison to T2-weighted MR quantitatively identifies regional dysfunction in patients with
and late gadolinium enhanced imaging. JACC Cardiovasc Imaging. arrhythmogenic right ventricular cardiomyopathy. J Magn Reson Imaging.
2013;6:1048–1058. 2015.
83. Hor KN, Baumann R, Pedrizzetti G, et al. Magnetic resonance derived 86. Prati G, Vitrella G, Allocca G, et al. Right ventricular strain and
myocardial strain assessment using feature tracking. J Vis Exp. 2011;(48). dyssynchrony assessment in arrhythmogenic right ventricular
84. Heermann P, Hedderich DM, Paul M, et al. Biventricular myocardial cardiomyopathy: cardiac magnetic resonance feature-tracking study. Circ
strain analysis in patients with ARVC using CMR feature tracking. J Cardiovasc Imaging. 2015;8:e003647.
Cardiovasc Magn Reson. 2014;16:75.
35 
Myocarditis
Dominique Auger and Matthias G. Friedrich

Myocarditis (from Greek/Latin “inflammation of the heart muscle”) is For viral myocarditis, it is important to realize that CMR does not
commonly used for viral myocarditis, although there are numerous allow for verifying or excluding viral persistence or autoimmune processes
other heart conditions leading to myocardial inflammation, including because the inflammatory response on a tissue level is very similar. In
acute ischemic injury, infiltrative diseases, allergies, and toxic or mechani- patients with severe disease and CMR evidence for severe injury, an
cal injuries. Thus inflammatory tissue pathology is not specific to a endomyocardial biopsy may reveal giant-cell myocarditis, and immu-
viral etiology and any such tissue pathology visualized by cardiovascular nosuppressive therapy may be advised.5
magnetic resonance (CMR) by itself is not specific as well. Myocardial
inflammation is accompanied by hyperemia and leukocyte infiltration, Morphology and Function
as well as edema and necrosis, as typical features of reversible or irre- In cases of uncomplicated myocarditis, LV and right ventricu-
versible cellular injury. lar (RV) function may be entirely normal, even in the presence of
Patients with myocarditis may be asymptomatic or oligosymptomatic, myocardial tissue involvement/injury. In other cases, ventricular wall
although the frequency of electrocardiogram (ECG) changes in systemic motion abnormalities may be very subtle and often in the inferolat-
viral illness indicates a high rate of myocardial involvement in systemic eral region, which is notoriously difficult to assess by TTE. Because
viral disease.1 The clinical presentation of myocardial inflammation CMR allows for nonrestricted views and because of its very accurate
may vary from subtle illness to overt heart failure or sudden death. quantitative assessment of related parameters, it is better suited for
Symptomatic patients may present with chest pain (caused by inflam- understanding functional abnormalities. Moreover, accurate and repro-
matory neural stimulation itself), dyspnea, fatigue (both potentially ducible RV volumes and ejection quantification can be obtained.6
reflecting ventricular dysfunction with an absolute or relative decrease Today, balanced steady-state free precession (bSSFP) techniques
of cardiac output), palpitations (extrasystole), or malaise. offer the best contrast between blood and endocardium and thus are
In routine clinical practice, transthoracic echocardiography (TTE) recommended.7,8
is frequently used to assess left ventricular (LV) parameters. It is widely Under routine clinical circumstances, a biplanar approach (two-
available, noninvasive, fast, and straightforward to perform in most and four-chamber view)9 or multiplanar10 approach often are suffi-
patients. Its quantitative and diagnostic accuracy, however, suffers from cient, but most published data refer to the use of short-axis stacks,
a frequently poor ultrasound transmission of lungs and bones, angular covering the entire left and right ventricle from the mitral plane to the
errors of the imaging plane, difficulties in placement of the imaging apex.11 The slice thickness should be ≤10 mm; in case of circumscribed
plane, problems in identifying the endocardial border, and parallel shift or subtle global changes, it should be reduced adequately. Although
of the ultrasonic plane out of the targeted center. TTE does not provide including trabecular tissue may improve reproducibility, results for
relevant information on the myocardial tissue. LV volumes and mass are less accurate, especially in patients with LV
hypertrophy.
CARDIOVASCULAR MAGNETIC RESONANCE
Tissue Characterization
APPROACH TO THE PATIENT WITH MYOCARDITIS Several contrast-enhanced (CE) and noncontrast CMR approaches
CMR is regarded as the most useful noninvasive clinical tool to assess are available to assess the myocardial tissue. Currently, the most fre-
myocarditis,2–4 and its diagnostic accuracy may exceed that of other quently used techniques are imaging edema and necrosis (see Figs. 35.1
available invasive and noninvasive approaches. Therefore using proven and 35.2).
protocols, it should be a first-line diagnostic tool in centers with access
to CMR. This is because of the capabilities of CMR, which is the gold Edema: T2-Weighted Cardiovascular Magnetic Resonance
standard for noninvasively quantifying biventricular mass, volume, Noncontrast T2-weighted imaging using short inversion time (TI)
and function, and is especially useful for assessing tissue pathology inversion recovery techniques is a robust technique to visualize fluid
in vivo. As in other forms of cardiomyopathies, the spatial distribu- accumulation such as edema and effusion in inflammatory diseases
tion of tissue abnormalities is of special importance and, consequently, because of its specific properties.12 Patients with myocarditis typi-
the diagnosis of myocardial inflammation in CMR is mostly based cally show regional and/or global high signal intensity, which usually
on visualizing hyperemia, edema, necrosis or scar, and, as additional equals or exceeds that of skeletal muscle as an internal reference by
supporting features, pericardial effusion and ventricular dysfunction. a factor of 2 and is reported as the signal intensity (SI) ratio.13 An
If properly used, CMR can identify or rule out myocardial inflam- increased SI ratio in acute myocarditis is a good predictor of func-
mation, assess its acuity and extent/severity, and even give prognostic tional recovery.14 T2-weighted imaging of the heart is challenged by
information. an inherently low signal-to-noise ratio (SNR) and a susceptibility to

420
CHAPTER 35 Myocarditis 421

motion and arrhythmia-related artifacts. It is therefore important to generally do not involve subendocardial layers.24 It should be kept in
(1) use thicker slices than usual (12–15, or even 20 mm) to increase mind that the sensitivity of LGE to detect myocarditis is generally lower
SNR; (2) use a triple-inversion or double-inversion sequence and than that of T1/early enhancement and T2-weighted CMR,13,25 whereas
ensure an effective repetition time (TR) of at least 2000 ms to maxi- its regional distribution is very specific for nonischemic injury. Inter-
mize T2 sensitivity; and (3) avoid the use of surface coils to prevent estingly, necrosis/scars in myocarditis often affect lateral regions as a
overcalling in anterior segments and missing disease in inferolateral row of focal injuries (Fig. 35.1), sometimes, however, with transmural
segments. extent (Fig. 35.2).
Phase-sensitive inversion recovery (PSIR) LGE imaging usually
Hyperemia: Early Gadolinium Enhancement facilitates interpretation, as it is less sensitive to an inadequately chosen
Free breathing, black-blood, fast spin echo images acquired early inversion time.26
(for 2 minutes between 20 seconds and 3 minutes after injection)
after gadolinium (Gd) injection can be used to visualize myocar-
dial inflammation.15 The increased uptake is most likely caused by
a combination of increased inflow (inflammatory hyperemia), slow
interstitial washin/washout kinetics (capillary leakage and edema),
diffusion into cells through leaky membranes (necrosis), and an
increased volume of distribution (scar tissue). Long-term follow-
up revealed persistence of these changes in patients with clinical
and functional evidence for ongoing inflammation.16 An increased
Gd accumulation has also been described in patients with myocar-
dial inflammation due to Chagas myocarditis,17 sarcoidosis,18,19 and
after doxorubicin chemotherapy.20 In suspected chronic myocarditis,
early enhancement and edema ratio were found to be the most sensi-
tive CMR markers for immunohistologically defined inflammatory
activity.21

Necrosis and Scar: Late Gadolinium Enhancement


Late gadolinium enhancement (LGE) imaging is performed using an
inversion-recovery gradient echo sequence with inversion times which
“null” the signal of normal myocardial tissue, with images being obtained
≥10 minutes after injection. Bright areas indicate irreversible myocardial
injury,22 which can be acute (necrosis) or chronic (scar). Following the
success in visualizing myocardial scarring in coronary artery disease, LGE
has also been used to assess myocarditis with a very good agreement FIG. 35.1  Typical appearance of necrosis related to myocarditis in a
between CMR findings and histopathology.23 Importantly, the regional four-chamber view using late gadolinium enhancement imaging. There
distribution of myocarditis lesions differs starkly from that of ischemic are scattered, mostly subepicardial focal lesions in the septum and the
hyperenhancement, because—as opposed to myocardial infarcts—they anterolateral wall (arrows).

A B
FIG. 35.2  Cardiovascular magnetic resonance in a patient with acute myocarditis. (A) Water-sensitive
(T2-weighted) dark-blood image in a short-axis view, showing a bright signal intensity in the anterior septum
and inferoseptal regions (arrows). (B) Late gadolinium enhancement image with largely corresponding areas
of necrosis (arrows). As regions of the right ventricular (RV) free wall also appear bright, RV involvement is
also suggested.
422 SECTION III  Functional Disease

A Cine

B Dark-blood T2W

C T1 mapping

D LGE
FIG. 35.3  Comparison of cine (A), water-sensitive (B), T1 mapping (C), and late gadolinium enhancement
(LGE) (D) images in a patient with myocarditis. The affected regions appear with a higher signal in all images,
corresponding to a higher T1 in the maps. T2W, T2-weighted. (From Ferreira VM, Piechnik SK, Dall’armellina
E, et al. Native T1-mapping detects the location, extent and patterns of acute myocarditis without the need
for gadolinium contrast agents. J Cardiovasc Magn Reson. 2014;16:36.)

Novel Techniques: Mapping of Myocardial T1 and T2 Combined Protocols


The quantification of T1 and T2 relaxation times, also referred to as To overcome the limitations of available sequences, a combination of
parametric CMR mapping, has been proven to be a very useful option T2-weighted, T1-weighted early enhancement, and T1-weighted LGE
to evaluate global myocardial tissue pathology. Native (noncontrast) T1 techniques are generally recommended. In 2009, an expert group pro-
and T2, and postcontrast T1 have been established as a biomarker of posed the Lake Louise criteria (LLC), which combine the three CMR
myocardial edema and fibrosis.27–31 Native T1 is modulated by numerous approaches, with LV dysfunction and pericardial effusion serving as
factors, including edema, and patients with acute myocarditis consistently supporting criteria38 (Box 35.1). Such a combined approach provides
exhibit an increase of T1.32 Fig. 35.3 shows T1 maps in comparison to a higher diagnostic accuracy than each of the individual sequences. A
edema and necrosis. Its diagnostic use in myocarditis is increasing. It multiparametric CMR approach for myocarditis diagnosis is recom-
appears to be able to identify affected myocardium in areas without mended, using the LLC. These criteria combine imaging of (1) edema,
LGE.33 Furthermore, it may provide confirmatory information in acute (2) increased volume of distribution (hyperemia/capillary leak), and
settings.34 Native T1 has been shown to allow differentiation between (3) irreversible injury in a nonischemic regional distribution. If at least
acute pheochromocytoma myocarditis, cured (operated) pheochromo- two of the three tissue-based criteria are positive, myocarditis can be
cytoma, and hypertensive heart disease.35 Interesting data have been predicted or ruled out with a diagnostic accuracy between 59% and
recently provided on treated patients with HIV.36 A recent study provided 92%,34,38,39 in acute presentation typically around 85%.40 If only LGE
preliminary evidence that native T1 mapping may be able to not only imaging is performed, the diagnostic accuracy was found to be between
identify acute myocarditis but also convalescent myocarditis.37 48% and 89%,38–40 yet with very variable sensitivity values, ranging
In the future, relaxation times may be found useful as alternative from 44% to 100%.38 LGE also does not allow us to differentiate acute
markers for hyperemia and capillary leak (T1, extracellular volume from chronic or healed myocarditis.41 This combined protocol has been
[ECV]) and edema (T1, T2). successfully used to exclude or to establish the diagnosis of myocarditis
CHAPTER 35 Myocarditis 423

BOX 35.1  The Lake Louise Criteria for Cardiovascular Magnetic Resonance in Suspected
Acute Myocarditis
In the setting of clinically suspected myocarditis, CMR findings are consistent A repeat CMR study between 1 and 2 weeks after the initial CMR study is
with myocardial inflammation, if at least two of the following criteria are present: recommended if
1. Regional or global myocardial SI increase in T2-weighted imagesa • none of the criteria are present, but the onset of symptoms has been very
2. Increased global myocardial early gadolinium enhancement ratio between recent and there is strong clinical evidence for myocardial inflammation.
myocardium and skeletal muscle in gadolinium-enhanced T1-weighted imagesb • one of the criteria is present.
3. There is at least one focal lesion with nonischemic regional distribution in The presence of LV dysfunction or pericardial effusion provides additional,
IR-prepared gadolinium-enhanced T1-weighted images (LGE)c supportive evidence for myocarditis.
A CMR study is consistent with myocyte injury and/or scar caused by myocardial
inflammation if
• criterion 3 is present.
a
Images should be obtained using a body coil or a surface coil with an effective surface coil intensity correction algorithm; global SI increase has
to be quantified by an SI ratio of myocardium over skeletal muscle of ≥2.0. If the edema is more subendocardial or transmural in combination
with a co-localized ischemic (including the subendocardial layer) pattern of LGE, acute myocardial infarction is more likely and should be
reported.
b
Images should be obtained using a body coil or a surface coil with an effective surface coil intensity correction algorithm; a global SI
enhancement ratio of myocardium over skeletal muscle of ≥4.0 or an absolute myocardial enhancement of ≥45% is consistent with myocarditis.
c
Images should be obtained at least 5 minutes after gadolinium injection; foci typically exclude the subendocardial layer, are often multifocal, and
involve the subepicardium. If the LGE pattern clearly indicates myocardial infarction and is co-localized with a transmural regional edema, acute
myocardial infarction is more likely and should be reported.
CMR, Cardiovascular magnetic resonance; IR, inversion recovery; LGE, late gadolinium enhancement; LV, left ventricular; SI, signal intensity.
Modified from Friedrich MG, Sechtem U, Schulz-Menger J, et al; International Consensus Group on Cardiovascular Magnetic Resonance in
Myocarditis. Cardiovascular magnetic resonance in myocarditis: a JACC White Paper. J Am Coll Cardiol. 2009;53:1475–1487.

and is part of several current recommendations.8,42,43 A metaanalysis may indicate a less favorable prognosis.16 Thus a follow-up CMR,
of nine cohorts (614 patients) with suspected myocarditis showed the including assessment of LV volumes and function, is advisable with
Lake Louse Criteria had a sensitivity and specificity of 70% and 56%, the ITAMY (ITalian multicenter study on Acute MYocarditis) of 374
respectively.44 The addition of native T1 to this comprehensive protocol patients with acute myocarditis and preserved LV systolic function
may improve sensitivity.32,45 demonstrating that patients with LGE in the midwall of the ante-
rior septum had a worse prognosis compared with other patterns
of LGE.47
FOLLOW-UP
In uncomplicated myocarditis, the signal abnormalities in T1/early
enhancement as well as functional abnormalities (if present) typically
REFERENCES
disappear within the first few weeks,15,46 whereas their persistence A full reference list is available online at ExpertConsult.com
CHAPTER 35 Myocarditis 423.e1

REFERENCES 20. Wassmuth R, Lentzsch S, Erdbruegger U, et al. Subclinical cardiotoxic


effects of anthracyclines as assessed by magnetic resonance imaging—a
1. Akritidis N, Mastora M, Baxevanos G, Dimos G, Pappas G. pilot study. Am Heart J. 2001;141:1007–1013.
Electrocardiographic abnormalities in patients with novel H1N1 21. Gutberlet M, Spors B, Thoma T, et al. Suspected chronic myocarditis
influenza virus infection. Am J Cardiol. 2010;106:1517–1519. at cardiac MR: diagnostic accuracy and association with
2. Baughman KL. Diagnosis of myocarditis: death of Dallas criteria. immunohistologically detected inflammation and viral persistence.
Circulation. 2006;113:593–595. Radiology. 2008;246:401–409.
3. Liu PP, Yan AT. Cardiovascular magnetic resonance for the diagnosis of 22. Kim RJ, Fieno DS, Parrish TB, et al. Relationship of MRI delayed contrast
acute myocarditis prospects for detecting myocardial inflammation. J Am enhancement to irreversible injury, infarct age, and contractile function.
Coll Cardiol. 2005;45:1823–1825. Circulation. 1999;100:1992–2002.
4. Friedrich MG, Marcotte F. Cardiac magnetic resonance assessment of 23. Mahrholdt H, Goedecke C, Wagner A, et al. Cardiovascular magnetic
myocarditis. Circ Cardiovasc Imaging. 2013;6:833–839. resonance assessment of human myocarditis: a comparison to histology
5. Kandolin R, Lehtonen J, Salmenkivi K, Räisänen-Sokolowski A, Lommi J, and molecular pathology. Circulation. 2004;109:1250–1258.
Kupari M. Diagnosis, treatment, and outcome of giant-cell myocarditis 24. Laissy J-P, Hyafil F, Feldman LJ, et al. Differentiating acute myocardial
in the era of combined immunosuppression. Circ Heart Fail. 2013;6: infarction from myocarditis: diagnostic value of early- and delayed-
15–22. perfusion cardiac MR imaging. Radiology. 2005;237:75–82.
6. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ. 25. Röttgen R, Christiani R, Freyhardt P, et al. Magnetic resonance imaging
Interstudy reproducibility of right ventricular volumes, function, and findings in acute myocarditis and correlation with immunohistological
mass with cardiovascular magnetic resonance. Am Heart J. parameters. Eur Radiol. 2011;21:1259–1266.
2004;147:218–223. 26. Kellman P, Arai AE, McVeigh ER, Aletras AH. Phase-sensitive inversion
7. Kramer CM, Barkhausen J, Flamm SD, Kim RJ, Nagel E, Society for recovery for detecting myocardial infarction using gadolinium-delayed
Cardiovascular Magnetic Resonance Board of Trustees Task Force hyperenhancement. Magn Reson Med. 2002;47:372–383.
on Standardized Protocols. Standardized cardiovascular magnetic 27. Moon JC, Messroghli DR, Kellman P, et al. Myocardial T1 mapping and
resonance (CMR) protocols 2013 update. J Cardiovasc Magn Reson. extracellular volume quantification: a Society for Cardiovascular
2013;15:91. Magnetic Resonance (SCMR) and CMR Working Group of the European
8. Friedrich MG, Larose E, Patton D, et al; Canadian Society for CMR. Society of Cardiology consensus statement. J Cardiovasc Magn Reson.
Canadian Society for Cardiovascular Magnetic Resonance (CanSCMR) 2013;15:92.
recommendations for cardiovascular magnetic resonance image analysis 28. Ferreira VM, Piechnik SK, Dall’armellina E, et al. T1 mapping for the
and reporting. Can J Cardiol. 2013;29:260–265. diagnosis of acute myocarditis using CMR: comparison to T2-weighted
9. Friedrich MG, Schulz-Menger J, Strohm O, Dick AJ, Dietz R. The and late gadolinium enhanced imaging. JACC Cardiovasc Imaging.
diagnostic impact of 2D- versus 3D-left ventricular volumetry by MRI in 2013;6:1048–1058.
patients with suspected heart failure. MAGMA. 2000;11:16–19. 29. Radunski UK, Lund GK, Muellerleile K. CMR in patients with severe
10. Bloomer TN, Plein S, Radjenovic A, et al. Cine MRI using steady state myocarditis: diagnostic value of quantitative tissue markers including
free precession in the radial long axis orientation is a fast accurate extracellular volume imaging. JACC Cardiovasc Imaging. 2014;7:667–675.
method for obtaining volumetric data of the left ventricle. J Magn Reson 30. Bohnen S, Radunski UK, Lund GK, et al. Performance of T1 and T2
Imaging. 2001;14:685–692. mapping cardiovascular magnetic resonance to detect active myocarditis
11. Le Ven F, Bibeau K, De Larochellière E, et al. Cardiac morphology and in patients with recent-onset heart failure. Circ Cardiovasc Imaging.
function reference values derived from a large subset of healthy young 2015;8.
Caucasian adults by magnetic resonance imaging. Eur Heart J Cardiovasc 31. Bönner F, Spieker M, Haberkorn S, et al. Myocardial T2 mapping
Imaging. 2016;17:981–990. increases noninvasive diagnostic accuracy for biopsy-proven myocarditis.
12. O h-Ici D, Ridgway JP, Kuehne T, et al. Cardiovascular magnetic JACC Cardiovasc Imaging. 2016;9:1467–1469.
resonance of myocardial edema using a short inversion time inversion 32. Ferreira VM, Piechnik SK, Dall’armellina E, et al. Native T1-mapping
recovery (STIR) black-blood technique: diagnostic accuracy of visual and detects the location, extent and patterns of acute myocarditis without the
semi-quantitative assessment. J Cardiovasc Magn Reson. 2012;14:22. need for gadolinium contrast agents. J Cardiovasc Magn Reson.
13. Abdel-Aty H, Boyé P, Zagrosek A, et al. Diagnostic performance of 2014;16:36.
cardiovascular magnetic resonance in patients with suspected acute 33. Radunski UK, Lund GK, Säring D, et al. T1 and T2 mapping
myocarditis: comparison of different approaches. J Am Coll Cardiol. cardiovascular magnetic resonance imaging techniques reveal unapparent
2005;45:1815–1822. myocardial injury in patients with myocarditis. Clin Res Cardiol.
14. Vermes E, Childs H, Faris P, Friedrich MG. Predictive value of CMR 2016;1–8.
criteria for LV functional improvement in patients with acute 34. Lurz P, Luecke C, Eitel I, et al. Comprehensive cardiac magnetic
myocarditis. Eur Heart J Cardiovasc Imaging. 2014;15:1140–1144. resonance imaging in patients with suspected myocarditis. J Am Coll
15. Friedrich MG, Strohm O, Schulz-Menger J, Marciniak H, Luft FC, Dietz Cardiol. 2016;67:1800–1811.
R. Contrast media-enhanced magnetic resonance imaging visualizes 35. Ferreira VM, Marcelino M, Piechnik SK, et al. Pheochromocytoma is
myocardial changes in the course of viral myocarditis. Circulation. characterized by catecholamine-mediated myocarditis, focal and diffuse
1998;97:1802–1809. myocardial fibrosis, and myocardial dysfunction. J Am Coll Cardiol.
16. Wagner A, Schulz-Menger J, Dietz R, Friedrich MG. Long-term follow-up 2016;67:2364–2374.
of patients paragraph sign with acute myocarditis by magnetic paragraph 36. Ntusi N, O’Dwyer E, Dorrell L, et al. HIV-1–related cardiovascular
sign resonance imaging. MAGMA. 2003;16:17–20. disease is associated with chronic inflammation, frequent pericardial
17. Torreão JA, Ianni BM, Mady C, et al. Myocardial tissue characterization effusions, and probable myocardial edema. Circ Cardiovasc Imaging.
in Chagas’ heart disease by cardiovascular magnetic resonance. J 2016;9:e004430–9.
Cardiovasc Magn Reson. 2015;17:97. 37. Hinojar R, Foote L, Arroyo Ucar E, et al. Native T1 in discrimination of
18. Friedrich MG. Visualization of cardiac involvement in patients with acute and convalescent stages in patients with clinical diagnosis of
systemic sarcoidosis applying contrast-enhanced magnetic resonance myocarditis: a proposed diagnostic algorithm using CMR. JACC
imaging. MAGMA. 2000;11:82–83. Cardiovasc Imaging. 2015;8:37–46.
19. Vignaux O, Dhote R, Duboc D, et al. Detection of myocardial 38. Friedrich MG, Sechtem U, Schulz-Menger J, et al; International
involvement in patients with sarcoidosis applying T2-weighted, Consensus Group on Cardiovascular Magnetic Resonance in Myocarditis.
contrast-enhanced, and cine magnetic resonance imaging: initial results Cardiovascular magnetic resonance in myocarditis: a JACC White Paper. J
of a prospective study. J Comput Assist Tomogr. 2002;26:762–767. Am Coll Cardiol. 2009;53:1475–1487.
423.e2 SECTION III  Functional Disease

39. Luetkens JA, Homsi R, Sprinkart AM, et al. Incremental value of Society for Cardiovascular Magnetic Resonance (SCMR) Board of
quantitative CMR including parametric mapping for the diagnosis Trustees Task Force on Standardized Post Processing. J Cardiovasc Magn
of acute myocarditis. Eur Heart J Cardiovasc Imaging. 2016;17: Reson. 2013;15:35.
154–161. 44. Wei S, Fu J, Chen L, Yu S. Performance of cardiac magnetic resonance
40. Lurz P, Eitel I, Adam J, et al. Diagnostic performance of CMR imaging imaging for diagnosis of myocarditis compared with endomyocardial
compared with EMB in patients with suspected myocarditis. JACC biopsy: a meta-analysis. Med Sci Monit. 2017;23:3687–3696.
Cardiovasc Imaging. 2012;5:513–524. 45. Luetkens JA, Doerner J, Thomas DK, et al. Acute myocarditis:
41. Zagrosek A, Abdel-Aty H, Boyé P, et al. Cardiac magnetic resonance multiparametric cardiac MR imaging. Radiology. 2014;273:383–392.
monitors reversible and irreversible myocardial injury in myocarditis. 46. Mavrogeni S, Spargias C, Bratis C, et al. Myocarditis as a precipitating
JACC Cardiovasc Imaging. 2009;2:131–138. factor for heart failure: evaluation and 1-year follow-up using
42. Hundley WG, Bluemke D, Bogaert JG, et al. Society for Cardiovascular cardiovascular magnetic resonance and endomyocardial biopsy. Eur J
Magnetic Resonance guidelines for reporting cardiovascular magnetic Heart Failure. 2011;13:830–837.
resonance examinations. J Cardiovasc Magn Reson. 2009;11:5. 47. Aquaro GD, Perfetti M, Camastra G, et al. Cardiac MR with late
43. Schulz-Menger J, Bluemke DA, Bremerich J, et al. Standardized image gadolinium enhancement in acute myocarditis with preserved systolic
interpretation and post processing in cardiovascular magnetic resonance: function. ITAMY Study. J Am Coll Cardiol. 2017;70:1977–1987.
36 
Hypertrophic Cardiomyopathy
Ethan J. Rowin, Evan Appelbaum, and Martin S. Maron

Hypertrophic cardiomyopathy (HCM) is a common genetic cardiomy- In addition, the extent of LV wall thickness may similarly be under-
opathy, is present in potentially as many as 1 : 200 individuals in the estimated by TTE, with more accurate measurements made by CMR.13
general population, and is caused by over 1400 mutations in at least Discrepancy in wall thickness measurements between the two techniques
11 genes encoding proteins of the cardiac sarcomere.1,2 This genetic is often a result of underestimation of segmental areas of LV hypertrophy
diversity is largely responsible for the heterogeneous phenotypic expres- present in the aforementioned “echo blind” regions of the LV wall (see
sion associated with this disease, including the range in the pattern of Fig. 36.3). This observation can have direct management implications,
left ventricular (LV) hypertrophy (Fig. 36.1) and LV outflow obstruction because massive hypertrophy (wall thickness ≥30 mm) is an independent
due to mitral valve-septal contact.1–4 Although HCM is associated with risk factor for sudden death in HCM, and in some patients these extents
normal longevity in the vast majority of patients, a small but important of hypertrophy may only be recognized by CMR.13
subset of patients are at risk for a number of adverse clinical outcomes, In addition, overestimation of wall thickness can also occur if the
including development of advanced heart failure (HF) symptoms, atrial crista supraventricularis, a prominent right ventricular (RV) muscle
and ventricular arrhythmias, thromboembolic events, and sudden structure that originates in the RV apex and transects the cavity to inset
death.1,2,5 Indeed, HCM remains the most common cause of sudden adjacent to the ventricular septum, is included in the transdimensional
death in young people, including competitive athletes.6,7 measurement of ventricular basal septal thickness (Fig. 36.4B).14 With
Over the last 40 years, cardiovascular imaging has had a central role CMR, the crista can be reliably identified on the basal short-axis images
in the clinical evaluation of HCM patients, with two-dimensional (2D) and observed to separate from the septum in diastole, exposing the
transthoracic echocardiography (TTE) the primary imaging technique epicardial border of the interventricular septum and allowing for the
over this period. However, over the last two decades, cardiovascular epicardial border of ventricular septum to be clearly delineated.
magnetic resonance (CMR) has emerged with greater frequency in
clinical HCM practice as a technique well-suited to characterize the PHENOTYPE CHARACTERIZATION OF
HCM phenotype with noninvasive, nonionizing radiation, high spatial HYPERTROPHIC CARDIOMYOPATHY
resolution imaging, and complete tomographic reconstruction of the
entire cardiac chamber (without obliquity), unencumbered by a limited Patterns and Distribution of Left
acoustic window or interference from thoracic and pulmonary Ventricular Hypertrophy
parenchyma.8–10 In addition, contrast-enhanced CMR with late gado- The most common location of focal LV wall thickening in HCM involves
linium enhancement (LGE) has the capability of identifying areas of the basal anterior free wall in continuity with the anterior ventricular
focal fibrosis/scarring, information not obtainable with echocardiog- septum (see Fig. 36.1). This phenotype is present in three-quarters of
raphy.8 In this chapter, we discuss areas in which CMR has contributed HCM patients.3 The inferior septum at the mid-LV level is the next
to the routine clinical evaluation of HCM patients. most common region of increased wall thickness.3 A small subset (10%)
of HCM patients have LV hypertrophy, which is focal and limited involv-
ing only one or two LV segments, including the apex and the majority
DIAGNOSIS of these patients will have normal LV mass by CMR.15 These observa-
A diagnosis of HCM is made when unexplained LV hypertrophy tions underscores the principle that a diagnosis of HCM is compatible
(≥15 mm wall thickness; see Fig. 36.1) occurs in the absence of another with limited extent of wall thickening and that increased LV mass is
disease capable of producing the magnitude of increased wall thick- not a requirement for the clinical diagnosis of HCM.1 Of note, a sub-
ness.1,2 CMR, by virtue of its high spatial resolution and sharp con- group of HCM patients demonstrate a noncontiguous distribution of
trast between bright-blood and dark myocardium, allows for precise LV hypertrophy, a pattern in which hypertrophied segments are separated
and reliable noninvasive measurements of LV wall thickness and can (often abruptly) by segments of normal wall thickness.3
clarify the diagnosis when wall thickness measurements fall in the
nondiagnostic range (i.e., normal or borderline increased) by TTE Right Ventricle
(Fig. 36.2).3,9 In particular, focal areas of increased wall thickness may A number of CMR observations have demonstrated the presence of
not be well seen by TTE (“echo blind”) when confined to three specific morphologic RV abnormalities in HCM. A significant proportion of
regions of the LV chamber, including anterolateral wall, apex, and HCM patients demonstrate increased RV wall thickness by CMR (i.e.,
inferior septum (Fig. 36.3).11,12 For these reasons, when a diagnosis ≥8 mm), which is well suited to reliably visualize morphologic abnor-
of HCM is suspected based on clinical profile and TTE is normal/ malities of the RV.14 Areas of increased RV wall thickness are most
nondiagnostic, CMR should be routinely performed to clarify the diag- commonly seen near the junction of the insertion of the RV wall into
nosis (see Fig. 36.2).9 either the anterior or inferior septum; however, diffuse RV hypertrophy

424
CHAPTER 36  Hypertrophic Cardiomyopathy 425

* *

* LV
LV
RV LV
RV
*
RV

A B C

* FW

*
LV VS

RV
LA

LA
LA
D E F
FIG. 36.1  Cardiovascular magnetic resonance (CMR) end-diastolic images demonstrating the diverse patterns
of left ventricular (LV) hypertrophy in hypertrophic cardiomyopathy (HCM). (A–C) Short-axis CMR images
demonstrating (A) hypertrophy of the basal anterior free wall and a portion of the continuous anterior septum
(asterisks), representing the most common area of LV hypertrophy in HCM, (B) prominent hypertrophy in
the basal posterior ventricular septum (asterisk), (C) massive hypertrophy (wall thickness of 31 mm) con-
fined to the basal anterior free wall (asterisk). (D–F) Long axis four-chamber CMR images demonstrating
(D) hypertrophy localized to the LV apex (asterisks), (E) hypertrophy throughout the ventricular septum (VS)
but sparing the LV free wall (FW), (F) focal area of hypertrophy confined to the basal anterior septum (arrows).
LA, Left atrium; LV, left ventricle; RV, right ventricle.

can also occur (Fig. 36.4A). Although uncommon, RV LGE can also defibrillator (ICD) therapy, radiofrequency ablation for recurrent ven-
occur in HCM.14 Currently, the clinical significance of RV hypertrophy tricular tachycardia (VT), and prophylactic anticoagulation for stroke
or LGE, is uncertain. prevention.16

Left Ventricular Apical Aneurysm ASSESSMENT OF HYPERTROPHIC


Increasing penetration of CMR into routine cardiovascular practice
has resulted in more frequent identification of a subset of patients with
CARDIOMYOPATHY FAMILY MEMBERS
thin-walled, scarred LV apical aneurysms (Fig. 36.4C) often associated HCM is a genetic disorder; therefore screening of all first-degree rela-
with muscular mid-cavity obstruction. This important group of patients tives of HCM patients is indicated to identify those individuals with
had been underdiagnosed before the application of CMR to HCM, potentially unrecognized disease. Screening should begin at the onset
because small-to-moderately sized aneurysms may not be reliably identi- of adolescence, with repeat imaging performed annually (every 12 to
fied by TTE.16 LGE CMR has demonstrated that the aneurysm rim in 18 months) throughout adolescence, and then every 5 years until the
these patients is composed predominantly of fibrosis that extends from fourth decade of life, because delayed-onset hypertrophy can occur in
the aneurysm rim into the septum and free wall and represents a primary adulthood in a small number of patients.1,2 Although TTE has been
arrhythmogenic substrate for the generation of malignant ventricular the mainstay test used for longitudinal family screening, CMR has been
tachyarrhythmias. recognized to have an increasing role because it provides a more precise
This subgroup of HCM patients is considered high risk, with adverse delineation of LV hypertrophy and therefore more accurate diagnosis.17
event rates of 6% per year (over 3-fold greater than that of HCM cohorts In addition, CMR provides a benchmark for future studies to better
without aneurysms), a result largely of sudden death events and throm- define the potential progression of LV hypertrophy.
boembolic stroke (secondary to LV thrombus formation in the aneu- In addition, CMR can assess for a variety of morphologic abnormali-
rysmal cavity).16 Therefore identification of this unique phenotype raises ties that may be present in HCM family members who carry a disease-
important management considerations because these complications causing sarcomere mutation but without LV hypertrophy (i.e., genotype
are effectively treatable with primary prevention implantable cardioverter positive–phenotype negative; G+P−) patients, including myocardial crypts
426 SECTION III  Functional Disease

CMR

Uncertain HCM Diagnosis Confirmed HCM Diagnosis

Prognosis and Management


Family Excludec LV
assessment phenocopy noncompaction
vs. apical HCM

LVH in HTNd/ With gadolinium Heart failure and


No LVH but ambiguous or athletic contrast septal reduction therapies
crypt/LGE/ echo-blind hearte
elongated MV areas vs. HCM

Myectomy
No/minimal Extensive LV vs.
Genetic LGE LGEa apical ASA
testing for aneurysm
confirmation of (+LGE)
HCM diagnosis
Lower LVOT anatomy
risk
↑ Sudden death Develop
risk end-stage
(EF <50%)
Muscular Elongated Magnitude/
mid- MV In-homogenous
cavity septal thickness
Heart obstructionb
No conventional Arbitrator transplant
risk factors with other
risk factors

ICD for 1° prevention


FIG. 36.2  Flow diagram outlining the role of cardiovascular magnetic resonance (CMR) in the diagnosis and
management of patients within the broad hypertrophic cardiomyopathy (HCM) spectrum. a≥15% of total left
ventricular (LV) mass. bIncludes anomalous papillary muscle insertion directly into anterior mitral leaflet, and
abberant LV muscle bundles. cA variety of unique late gadolinium enhancement (LGE) patterns useful in
discerning infiltrative/metabolic diseases from HCM. dIdentification by CMR of a noncontiguous pattern of
LV hypertrophy (regions of increased LV wall thickening separated by segments of normal thickness) con-
sistent with genetic heart disease can discern HCM from increased wall thickness secondary to uncontrolled
systemic hypertension. eIn highly trained athletes with mildly increased LV wall thickness, the presence of
LGE and tagging may help differentiate HCM from physiologic “athletic heart.” Serial CMR studies are the
most reliable strategy for comparing LV wall thickness measurements before and after a period of decon-
ditioning to distinguish pathologic from physiologic hypertrophy. ASA, Alcohol septal ablation; EF, ejection
fraction; HTN, systemic hypertension; ICD, implantable cardioverter defibrillator; LVOT, left ventricular outflow
tract; MV, mitral valve. (From Maron MS, Maron BJ. Clinical impact of cardiovascular magnetic resonance
imaging in hypertrophic cardiomyopathy. Circulation. 2015;132:292–298.)

(i.e., narrow deep blood-filled invaginations within LV myocardium; DIFFERENTIATION OF OTHER ETIOLOGIES OF LEFT
Fig. 36.4F),18 elongated mitral valve leaflets,19 expanded extracellular VENTRICULAR HYPERTROPHY
space (with T1 mapping),20 and LGE.21 These observations suggest that
even in the absence of LV hypertrophy, the hearts of patients who are Athletic Heart
G+P- may be morphologically abnormal. Finally, the presence of one or LV hypertrophy associated with systemic training (i.e., athletic heart)
more of these structural abnormalities identified in a family member of may be difficult to differentiate from HCM.22 CMR can help distinguish
an HCM patient should prompt continued close surveillance with serial by identification of focal pattern of hypertrophy, a finding supportive
CMR for development of LV hypertrophy and conversion to clinical of a diagnosis of HCM. In addition, forced deconditioning of an athlete
disease and/or genetic testing to aid in confirming HCM diagnosis. may serve as a useful strategy to resolve diagnosis, with CMR well suited
CHAPTER 36  Hypertrophic Cardiomyopathy 427

Echo CMR hypertrophy in septum and lateral wall) is predominant, whereas LV


wall thickening in HCM is almost always asymmetric.3 Additionally, an
RV RV LV outflow obstruction because of typical systolic anterior motion of
VS * VS the mitral valve will also sway a diagnosis toward HCM. This finding
*
LV * LV is present in over one-third of HCM patients and very rarely seen in
hypertensive cardiomyopathy. CMR can also be helpful in the detection
A B
of changes in serial measurements of LV wall thickness after aggressive
treatment with antihypertensives, in which a regression of hypertrophy
*
* would favor a diagnosis of hypertensive cardiomyopathy.
LV LV
RV
C D Left Ventricular Noncompaction
Because of its superior spatial resolution with imaging of the LV apex,
CMR may also help to clarify (or alter) diagnosis by demonstrating
VS LV
the presence of prominent trabeculations consistent with a diagnosis
VS of left ventricular noncompaction (LVNC) in patients initially diag-
* LV nosed with apical HCM.24 LV trabeculations, associated with LVNC,
* when imaged by 2D TTE, may appear as apical hypertrophy, potentially
E F resulting in a misdiagnosis of apical HCM. This has important impli-
FIG. 36.3  Advantage of cardiovascular magnetic resonance (CMR) com- cations for management strategies because a diagnosis of LVNC has
pared with two-dimensional (2D) transthoracic echocardiography. (A) 2D significantly different treatment implications (including participation
transthoracic echocardiography (TTE). Anterolateral left ventricular free in competitive sports, anticoagulation, and risk stratification for sudden
wall is 18 mm; myocardial border and adjacent extracardiac structures cardiac death).
are not well defined (asterisks). (B) CMR in the same patient shows well
delineated border of anterolateral wall (arrowheads), which is massively Infiltrative Cardiomyopathy
thickened (35 mm), creating a sudden cardiac death (SCD) risk factor. (C) Infiltrative cardiomyopathies, including amyloidosis or glycogen/
2D TTE. Nondiagnostic left ventricle (LV). (D) CMR in the same patient; lysosomal storage diseases (such as Anderson-Fabry disease or Danon
segmental hypertrophy of left ventricular apex (asterisks); that is, apical
disease) can mimic HCM because they can produce increased wall
hypertrophic cardiomyopathy. (E) 2D TTE. Inferior septal (VS) thickness
is 21 mm (asterisk). (F) CMR in the same patient; massive hypertrophy
thickness as part of their phenotypic expression. Although these diseases
(41 mm) (asterisk), creating an SCD risk marker. RV, Right ventricle. may have noncardiac signs and symptoms, disease expression can be
(B, From Maron MS, Lesser JR, Maron BJ. Management implications confined only to the heart. The accurate differentiation of these “pho-
of massive left ventricular hypertrophy in hypertrophic cardiomyopathy tocopies” is critical because treatment strategies and prognosis differs
significantly underestimated by echocardiography but identified by car- compared with HCM. In amyloidosis, CMR identification of nearly
diovascular magnetic resonance. Am J Cardiol. 2010;105:1842–1843. D, identical wall thickness measurements in the septum and lateral wall
From Moon JC, Fisher NG, McKenna WJ, Pennell DJ. Detection of apical (“concentric”) combined with global subendocardial LGE is sugges-
hypertrophic cardiomyopathy by cardiovascular magnetic resonance in tive for cardiac amyloidosis and is not typical in HCM (Fig. 36.5).25
patients with nondiagnostic echocardiography. Heart. 2004;90:645–649.) Marked hypertrophy in the absence of electrocardiographic (ECG)
evidence for LV hypertrophy should also raise the clinician’s suspicion
for an infiltrative/amyloid cardiomyopathy. Suspicion for Anderson-
Fabry disease, an X-linked storage disease in which mutations in the
to compare maximum LV wall thickness measurements before and after α-galactosidase A gene lead to cellular accumulation of glycosphingo-
a period of systemic deconditioning.22 In this regard, a patient whose lipids in multiple organs including the heart, is potentially treatable with
wall thickness regresses more than 2 mm supports a diagnosis of athletic enzyme replacement therapy and can be raised by a concentric pattern of
heart, whereas LV hypertrophy that remains present despite decon- hypertrophy by CMR with LGE confined to the basal inferolateral wall26
ditioning supports a diagnosis of HCM. Myocardial tagging using spatial (see Fig. 36.5).
modulation of magnetization (SPAMM) may also be useful in discrimi- Danon disease, due to mutations in genes that encode the lysosomal
nating physiologic from pathologic hypertrophy.23 associated membrane protein 2 (LAMP2), leads to accumulation of
LGE CMR provides the opportunity to aid in the differentiation intracellular vacuoles and is a profound and accelerated disease process
given the ability to noninvasively provide tissue characterization by with rapid clinical deterioration leading commonly to advanced HF
means of identifying focal areas of replacement fibrosis and expanded and sudden death at a young age (commonly under 25 years of age).
extracellular space. Although LGE is present in about half of individuals CMR can be suggestive of diagnosis in setting of massive LV hypertrophy,
with HCM, LV remodeling associated with the athlete heart should not with extensive diffuse and often transmural LGE27 (see Fig. 36.5).
result in focal areas of myocardial scarring/fibrosis, especially in younger Although CMR findings may be suggestive of an infiltrative disease, no
individuals.22 Therefore in an athlete in whom suspicion has been raised CMR findings are pathognomonic and therefore confirmatory diagnosis
for HCM, the presence of LGE on contrast-enhanced CMR favors a with genetic testing or myocardial biopsy is ultimately required.
diagnosis of HCM. In contrast, the absence of LGE does not exclude
HCM because only half of HCM patients demonstrate LGE. LEFT VENTRICULAR OUTFLOW
Hypertensive Cardiomyopathy TRACT OBSTRUCTION
The differentiation of LV hypertrophy because of systemic hyperten- The most common mechanism of HF symptoms in HCM is mechanical
sion from HCM has historically been challenging. CMR can help in impedance to LV outflow due to systolic anterior motion (SAM) of the
differentiation by examining the pattern of hypertrophy. With long- mitral valve, with peak LV outflow tract (LVOT) gradient of ≥30 mm
standing systemic hypertension, concentric hypertrophy (near-identical Hg under resting conditions an independent determinant of progressive
428 SECTION III  Functional Disease

RV D

LV
VS P
RV
LV

RV
LA

A B C

Ao

LA
LA
VS

LV
LV
LV

D E F
FIG. 36.4  Cardiovascular magnetic resonance (CMR) end-diastolic images demonstrating diversity of the
phenotypic expression within hypertrophic cardiomyopathy (HCM). (A) Increased wall thickness in the superior
segment (arrowhead) and inferior segment (arrow) of the right ventricular (RV) free wall. (B) Prominent and
hypertrophied crista supraventricularis (outlined structure) and RV muscle structure. It is important not to
include this muscle structure in the measurement of ventricular septum because this can lead to overestima-
tion of maximal left ventricular (LV) wall thickness. (C) Medium-sized LV apical aneurysm (arrowheads) and
maximum wall thickness at midventricular level with muscular apposition of the ventricular septum and LV
free wall producing distinct proximal (P) and distal (D) chamber. (D) Elongation of the anterior mitral valve
leaflet (arrows), contributing to LV outflow obstruction in this patient. (E) Muscular midcavitary obstruction
attributable to the insertion of anomalous anterolateral papillary muscle directly into anterior leaflet (arrow),
contacting the midventricular septum in systole (arrowheads). (F) Asymptomatic genotype-positive, phenotype-
negative patient with three deep myocardial crypts in the basal inferior LV wall. Ao, Aorta; LA, left atrium;
LV, left ventricle; RV, right ventricle; VS, ventricular septum. (A, Modified from Maron MS, Hauser TH,
Dubrow E, et al. Right ventricular involvement in hypertrophic cardiomyopathy. Am J Cardiol. 2007;100:1293–
1298. D, Modified from Maron MS, Olivotto I, Harrigan C, et al. Mitral valve abnormalities identified by
cardiovascular magnetic resonance represent a primary phenotypic expression of hypertrophic cardiomyopathy.
Circulation. 2011;124:40–47; F, From Maron MS, Rowin EJ, Lin D, et al. Prevalence and clinical profile of
myocardial crypts in hypertrophic cardiomyopathy. Circ Cardiovasc Imaging. 2012;5:441–447.)

HF symptoms.4 Obstructive patients with advanced symptoms refrac-


tory to medical management become candidates for septal reduction Selection and Planning for Septal Reduction
interventions with surgical myectomy (or, alternatively, alcohol septal CMR has emerged to have an important role in the morphologic evalu-
ablation) for relief of LVOT gradients.1,2 ation of the LVOT and anomalies contributing to the mechanism of
Given the importance of LVOT obstruction in dictating manage- subaortic obstruction, including elongated mitral valve leaflet lengths19
ment strategies in HCM patients, accurate and reliable measurements of (Fig. 36.4D), anomalous insertion of the anterior papillary muscle
the pressure gradients remain critical. CMR allows for visualization of directed into the mitral leaflet28 (Fig. 36.4E), and muscle bundles that
SAM and septal contact, but it is limited in reliably measuring dynamic extend from the apex and attach into the basal anterior septum.29 The
LVOT pressure gradients in HCM.1,9 Therefore TTE, via continuous identification of these features may be missed by TTE, yet are important
wave Doppler, remains the preferred technique for assessing outflow because they potentially alter the septal reduction strategy in favor of
gradients.1 surgical myectomy, as a percutaneous approach using alcohol septal
CHAPTER 36  Hypertrophic Cardiomyopathy 429

*
LV
Amyloid
*
RV
Microvascular Microvascular
remodeling (“small dysfunction
A B vessel” disease)

Myocardial
ischemia
Anderson-Fabry LV
disease
RV

C D

Myocardial
fibrosis (LGE)

Danon FIG. 36.6  Proposed mechanism for late gadolinium enhancement (LGE)
LV in hypertrophic cardiomyopathy (HCM). Structurally abnormal intramural
disease
* coronary arteries are responsible for microvascular ischemia. Over time,
RV myocyte cell death and a process of repair with replacement fibrosis
E F can be imaged as LGE in HCM.

FIG. 36.5  Cardiovascular magnetic resonance (CMR) for differentiation


of etiology of left ventricular (LV) hypertrophy. (A) 64-year-old male with displaced accessory anterolateral or double bifid papillary muscles can
concentric LV hypertrophy (maximum LV wall thickness of 18 mm in tether the plan of the mitral valve toward the septum, contributing to
septum and 14 mm in lateral wall). (B) Late gadolinium enhancement the mechanism of obstruction.30,32 The surgeon can address these abnor-
(LGE) image in the same patient demonstrates early contrast washout malities by resecting these muscles to ensure a complete abolishment
with transmural LGE in septum and lateral wall (arrows) features con- of obstruction.
sistent with cardiac amyloid. The patient underwent cardiac biopsy
confirming the diagnosis of amyloid. (C) A 44-year-old female with con- PATHOPHYSIOLOGY AND CLINICAL PROFILE OF
centric LV hypertrophy (maximum wall thickness of 16 mm in septum
and 13 mm in the lateral wall). (D) LGE image in the same patient LATE GADOLINIUM ENHANCEMENT IN
demonstrates LGE confined to the basal inferolateral wall (arrows), a HYPERTROPHIC CARDIOMYOPATHY
location of LGE typical for Anderson-Fabry disease. The patient under-
went genetic testing, which demonstrated pathogenic mutation in the Histopathology
GLA gene, confirming the diagnosis of Anderson-Fabry disease. (E) A Areas of LGE in HCM are considered to represent the end result of a
21-year-old male demonstrated massive LV hypertrophy (wall thickness pathophysiologic cascade in which blunted myocardial blood flow due
of 32 mm) confined to the ventricular septum (asterisk). (F) LGE images to abnormal intramural coronary arteries results in myocyte cell death
in the same patient demonstrated transmural LGE throughout the anterior and reparative processes of fibrosis (imaged as LGE)33 (Fig. 36.6). This
and lateral walls (arrowheads) with mid-myocardial LGE throughout the
principle is supported by imaging studies in which myocardial blood
septum (arrow) in a pattern atypical for HCM. Genetic testing was sent
flow is decreased during stress in areas occupied by, and directly adjacent
and revealed a pathogenic mutation in the LAMP2 gene, consistent
with Danon cardiomyopathy. LV, Left ventricle; RV, right ventricle. to, regions of myocardial fibrosis as assessed by LGE.34
A number of histopathologic studies have identified LGE to represent
fibrosis.35,36 In ventricular septal tissue removed from HCM patients at
ablation is unable to intervene on these specific structural abnormali- the time of surgical myectomy, there is a stronger association between
ties9,30 (see Fig. 36.2). the extent of interstitial and replacement fibrosis assessed by histologic
The identification of these morphologic abnormalities also may examination and LGE seen on preoperative CMR studies.36 In addition,
alter preoperative surgical planning because they dictate a specific surgi- case reports of explanted HCM hearts have demonstrated strong cor-
cal strategy9,30 (see Fig. 36.2). Specifically, mitral valve leaflets are sub- relation between areas of LGE on pretransplant CMR and replacement
stantially increased in length in one-third of HCM patients and contribute fibrosis by histologic evaluation.35
to LV outflow obstruction, particularly when the leaflet length exceeds
2-fold the transverse dimension of the outflow tract at end systole.17,30 Clinical Profile
In obstructive HCM patients with this abnormality, surgical strategy Up to 60% of all HCM patients demonstrate some LGE, which, when
may be altered to perform extended myectomy trough and mitral valve present, occupies an average of 9% of total LV myocardial volume.37
repair (with plication) to eliminate SAM and the outflow gradient with Virtually any pattern, distribution, and location of LGE can be observed
less extensive septal hypertrophy (wall thickness ≤17 mm). CMR also in HCM, although in general the location of LGE does not correspond
allows for the reliable identification of anomalous and direct papillary to a coronary vascular distribution37–41 (Fig. 36.7). Most commonly,
muscle insertion into the anterior mitral leaflet, an uncommon but LGE is located in both the ventricular septum and the free wall (in
important mechanism of muscular obstruction,28 which dictates a spe- over 30% of patients), but less commonly can be confined to the apex
cific surgical approach with deep, extended muscular resection well and the areas of RV insertion into the interventricular septum.42,43 LGE
beyond the contact point of the mitral valve and ventricular septum can also occur in other myocardial structures outside of the LV wall,
as well as reduction in papillary muscle thickness.31 Similarly, apically including the RV wall, and papillary muscles.32 Transmural extent of
430 SECTION III  Functional Disease

LGE is not uncommon, occurring in one-half of patients.42 A significant of Cardiology (AHA/ACC) guidelines recommend the identification
but modest relationship exists between hypertrophy and LGE: patients of high-risk patients based on the presence of five noninvasive conven-
with LGE have greater maximal LV wall thickness and LV mass index tional risk factors,1 whereas more recently the European Society of
than patients without LGE. On an individual patient basis, there is a Cardiology (ESC) has promoted a risk score based on a mathematical
relationship between the segmental LV wall thickness and LGE.42,43 In formula taking into account numerous clinical variables.2 However,
contrast, prevalence and extent of LGE are no different across different current risk stratification strategies do not recognize all HCM patients
genders or across different age ranges, including in elderly (≥65 years at risk for sudden death, underscoring the need to identify additional
old) and younger (≤35 years old) HCM patients.37 markers of risk to more reliably identify high-risk patients, for whom
LGE has been promoted.37–41
LATE GADOLINIUM ENHANCEMENT Early cross-sectional studies demonstrated a strong association
AND PROGNOSIS between the presence of LGE and ventricular tachyarrhythmias on
ambulatory 24-hour Holter ECG in HCM, with LGE conferring up
Sudden Cardiac Death to a 7-fold increased risk for ambulatory nonsustained ventricular
HCM remains the most common cause of sudden death in young tachyarrhythmias compared with HCM patients without LGE44 (see
patients.1,2,7 Currently, American Heart Association/American College Fig. 36.7). These results suggested that ventricular tachyarrhythmias
emanate from regions of structurally abnormal myocardium, provid-
ing the rationale for a number of longitudinal LGE outcome studies.
14% When combining the data together in a pooled manner, the presence of
Sudden death events LGE is associated with an increased risk for sudden death.45 However,
12% Total mortality
LGE is common in HCM, with a prevalence of over 50% (and up to
70% in some select populations). Thus assessing LGE only in a binary
End-stage HCM
fashion (presence vs. absence) is not a practical strategy as a risk marker
Estimated 5-year event rate

10% because it would lead to over implantation of primary prevention


ICD devices.9
8% Three large studies have been completed evaluating the extent of
LGE and sudden death risk. When analyzed in a pooled manner, a
strong relationship is evident between the amount of LGE and the risk
6%
of a sudden death event45 (Table 36.1). The largest of these studies by
Chan et al.,37 a multicenter study employing a core laboratory analysis
4% of almost 1300 HCM patients followed for an average of 3 years after
CMR, demonstrated a statistically significant relationship between the
2% amount of LGE and sudden death events as well as total mortality
(see Fig. 36.7), with extensive LGE (≥15% of LV mass) associated with
a 2-fold greater risk of sudden death events compared with patients
0%
0% 5% 10% 15% 20% 25% 30% 35% 40%
with no LGE (Fig. 36.8), including in patients without any of the con-
ventional sudden death risk markers. In addition, in patients with ≥1
LGE by % left ventricular mass
conventional risk factor, the extent of LGE strengthened the current
FIG. 36.7  Predicted 5-year event rates relative to the percentage of
sudden death risk model by providing information that improved the
late gadolinium enhancement (LGE) for the risk of end-stage hypertrophic
cardiomyopathy (HCM) with systolic dysfunction, sudden death events,
ability to identify both high-risk and low-risk patients. On the other
and total mortality. (From Chan RH, Maron BJ, Olivotto I, et al. Prognostic hand, patients without LGE had a relatively benign course and were at
value of quantitative contrast-enhanced cardiovascular magnetic reso- low risk for adverse events37 (see Fig. 36.8). When data from this study
nance for the evaluation of sudden death risk in patients with hypertrophic were combined with Ismail et al.,41 the other large study that adjusted
cardiomyopathy. Circulation. 2014;130:484–513.) for other variables known to impact sudden death risk, the extent of

TABLE 36.1  Metaanalysis for Late Gadolinium Enhancement and Sudden Death
Unadjusted HR, Adjusted HR, per
Study per 10% LV Mass 95% CI P 10% LV Mass 95% CI P
SCD
Bruder et al.39 1.7 1.2–2.5 < .01 NA NA NA
Ismail et al.41 1.5 1.1–2.1 .007 1.2 0.8–1.7 .2
Chan et al.37 1.5 1.2–1.8 < .0001 1.4 1.1–1.9 .002
Pooled 1.5 1.3–1.8 < .0001 1.3 1.1–1.6 .005

HF Death
Ismail et al.41 1.5 1.0–2.1 .028 NA NA NA
Chan et al.37 1.7 1.1–2.7 .01 NA NA NA

CI, Confidence interval; HF, heart failure; HR, hazard ratio; LGE, late gadolinium enhancement; LV, left ventricular; NA, not available; SCD,
sudden cardiac death.
Modified from Weng Z, Yao J, Chan RH, et al. Prognostic value of LGE-CMR in HCM: a meta-analysis. JACC Cardiovasc Imaging.
2016;9:1392–1402.
CHAPTER 36  Hypertrophic Cardiomyopathy 431

30
LGE (+)
25 LGE (−)

% of HCM patients
20

with NSVT
LV 15

10

A 0
C Adabag et al.43 Rubinshtein et al.40

1.00

Freedom from sudden death


LGE absent
0.95 LGE ≤ 10%
LGE = 15%
RA
0.90 P = .008
LGE ≥ 20%

LA
0.85
B 0 1 2 3 4 5 6
D Follow-up (years)
FIG. 36.8  Relationship between late gadolinium enhancement (LGE) and sudden death risk in hypertrophic
cardiomyopathy (HCM). (A and B) LGE CMR images in two different HCM patients, each with extensive
LGE (arrows) throughout the ventricular septum. (C) Nonsustained ventricular tachycardia (NSVT) on 24-hour
Holter electrocardiogram (ECG) is 7-fold more common in HCM patients with LGE compared with those
without LGE. (D) Relation between extent of LGE and sudden death events in 1293 patients with HCM. LV,
Left ventricle; RV, right ventricle. (Modified from Chan RH, Maron BJ, Olivotto I, et al. Prognostic value of
quantitative contrast-enhanced cardiovascular magnetic resonance for the evaluation of sudden death risk
in patients with hypertrophic cardiomyopathy. Circulation 2014;130:484–513.)

LGE in the RV insertion area of the LV appears to be a reliable predictor


TABLE 36.2  Adjusted Hazard Ratios for
of adverse HCM-related events, including sudden cardiac death (SCD)46
Sudden Cardiac Death Using Data From (see Fig. 36.9). This is probably related to the morphologic observation
Chan et al.37 and Ismail et al.41 that the LGE in the RV insertion point is composed primarily of expanded
Pooled HRadjusted extracellular space with interstitial fibrosis embedded within disorderly
LGE % Point Estimate 95% CI arranged myocytes without replacement fibrosis.
0 1.0 — Based on these observations, it is now reasonable to consider incor-
1 1.03 1.01–1.05 porating the results of LGE CMR in the assessment of sudden death
10 1.36 1.10–1.69 risk in patients with HCM1,2,9,38–45 (Fig. 36.10). First, extensive (≥15%
15 1.59 1.15–2.20 LV volume) LGE identifies HCM patients at increased risk for sudden
20 1.86 1.21–2.86 death (even without a conventional risk factor) and could be deserving
30 2.5 1.33–4.83 of consideration for primary prevention ICD.1,9,37,45 In addition, extensive
40 3.4 1.46–8.16 LGE can act as an arbitrator to help resolve decision making regarding
ICD when sudden death risk remains ambiguous after standard risk
CI, Confidence interval; HR, hazard ratio; LGE, late gadolinium stratification because it can serve as an arbitrator toward ICD place-
enhancement. ment. In contrast, the absence of (or minimal) LGE is associated with
Modified from Weng Z, Yao J, Chan RH, et al. Prognostic value of lower risk for sudden death and should provide a measure of reassur-
LGE-CMR in HCM: a meta-analysis. JACC Cardiovasc Imaging. ance and can arbitrate away from device therapy in ambiguous risk
2016;9:1392–1402.
situations, potentially avoiding unnecessary ICD.37 Further insights into
the role of LGE in the management of at-risk HCM patients will emerge
LGE remained a strong independent predictor of sudden death events from the completion of the international multicenter Hypertrophic
(Table 36.2). Cardiomyopathy Registry (HCMR) study.47
It is not possible to make reliable associations between the pattern
of LGE and the outcome in HCM, a result of the enormous diversity Heart Failure
observed in the distribution of LGE in this disease. Indeed, the only A small subset of nonobstructed HCM patients are at risk for develop-
reliable LGE pattern seen in HCM is the relatively specific pattern of ing progressive HF symptoms refractory to pharmacologic treatment.
LGE confined to the RV insertion area (either anterior, inferior, or These patients constitute two distinct subgroups: patients with impaired
both)42–44 (Fig. 36.9). However, neither the presence nor the extent of LV systolic function and adverse LV remodeling associated with extensive
432 SECTION III  Functional Disease

of the brightest region of hyperenhancement).49,50 In an individual


patient with HCM there may be significant differences in amount of
LGE identified depending on which SD threshold is chosen, with 2 SDs
resulting in a 2-fold greater amount of LGE compared with 6 SDs.49
LV However, higher grayscale thresholds (6 SDs) and FWHM have
appeared to yield the closest approximation to the extent of LGE identi-
VS
fied visually and the most reproducible.36,48 In addition, higher grayscale
RV thresholding methods (such as 5 or 6 SDs) have been shown to provide
A B the best representation of total fibrosis burden as demonstrated by
histopathologic analysis of ventricular septal tissue removed in HCM
LGE confined patients undergoing surgical myectomy.36 Of note, grayscale threshold
1.0 P = .71
to insertion points can also be performed manually and adjusted to define areas of visually
0.9 identified LGE. This method was used to assess the extent of LGE in
Event-free survival

the Chan et al.37 multicenter LGE study in predicting sudden death


0.8 LGE - events because it correlates well to the amount LGE quantified using
high grayscale threshold cutoffs, has excellent reproducibility, and is
0.7 less time consuming to perform.
0.6 OTHER CARDIOVASCULAR MAGNETIC
0.5
RESONANCE TECHNIQUES IN HYPERTROPHIC
0 1 2 3 4 5 6 7 CARDIOMYOPATHY
C Follow-up (years)
Myocardial Perfusion
FIG. 36.9  Significance of right ventricular (RV) insertion point late gado-
linium enhancement (LGE) in hypertrophic cardiomyopathy (HCM). (A In HCM, the intramural coronary arterioles are structurally abnormal,
and B) The confluence of the RV wall and ventricular septum (RV inser- with small lumen resulting in blunted myocardial blood flow (MBF)
tion point) is a common area for LGE in HCM and can occur in either during stress (i.e., microvascular dysfunction). CMR perfusion sequences
the anterior, inferior (A) (arrow) or both (B) (thin arrow, anterior; thick can assess MBF at rest and during pharmacologic stress (typically
arrow, inferior) insertion points. (C) Kaplan-Meier event-free survival adenosine, regadenoson, or dipyridamole) with superior spatial resolu-
curves comparing 134 patients with HCM and LGE confined to the RV tion. Regional differences in MBF have been observed within LV seg-
insertion areas compared with 745 patients with HCM without LGE, ments, with MBF reduced in the subendocardial layer compared with
demonstrating no significant difference in HCM-related event-free sur- subepicardium, and the degree of abnormal perfusion related to the
vival. LV, Left ventricle; RV, right ventricle; VS, ventricular septum.
magnitude of wall thickness.51 In addition, MBF also appears reduced
(Modified from Chan RH, Maron BJ, Olivotto I, et al. Significance of late
gadolinium enhancement at right ventricular attachment to ventricular
in LV segments with LGE, suggesting that microvascular dysfunction
septum in patients with hypertrophic cardiomyopathy. Am J Cardiol. may be responsible for myocardial ischemia-mediated myocyte death,
2015;116:436–441.) and ultimately repair in the form of replacement fibrosis.51 However,
sufficient longitudinal follow-up studies with stress CMR in HCM have
not been completed. Therefore the clinical significance and management
implications of CMR MBF abnormalities remain uncertain.
LV fibrosis (ejection fraction [EF] <50%; end stage),48 and patients with
preserved LV systolic function with little or no remodeling.1,2 Extensive Myocardial Tagging
LGE has been observed as an emerging marker to prospectively identify Diastolic function is abnormal in nearly all HCM patients and is the
HCM patients with preserved systolic function who are at increased major etiology of HF symptoms in nonobstructive patients with pre-
risk for future development of end stage (see Fig. 36.7).37,41,45 Patients served LV systolic function. Proposed mechanisms for this dysfunction
who develop end stage are at increased risk for advanced HF symptoms in HCM include myocardial disarray, microvascular ischemia, interstitial
at some point in their clinical course, often necessitating consideration and replacement fibrosis, as well as LV hypertrophy. Myocardial tagging
for heart transplant.47 In support of this principle, pooled data for the SPAMM offers the opportunity to quantify diastolic function in CMR
two largest LGE follow-up studies in HCM demonstrated a significant by examining myocardial mechanical parameters, such as strain and
increase in risk for HF death with increasing amounts of LGE45 (see torsion, and has been demonstrated to be abnormal within HCM.52
Table 36.1). These abnormalities are correlated both to magnitude of LV hypertrophy
as well as areas of LGE in HCM. Of important note, these features can
QUANTIFICATION OF LATE be abnormal even in areas free of hypertrophy and without LGE, dem-
onstrating the multifactorial etiologies of abnormal diastolic function
GADOLINIUM ENHANCEMENT within HCM. Although of potential research interest, to date, tagging
Numerous techniques have been proposed to quantify the extent of has not been shown to be associated with clinical outcomes within this
LGE in HCM. At the time of this writing, there are no expert consensus disease and thereby has an uncertain role in clinical evaluation and
recommendations regarding the preferred strategy. The most widely management of HCM patients. As mentioned earlier, SPAMM may also
used approaches include semiautomated algorithms that identify high be useful in discriminating physiologic hypertrophy from aortic stenosis
signal intensity LGE pixels in the LV wall after applying a grayscale due to HCM.23
threshold a number of standard deviations (SDs) above the mean signal
intensity within a remote region containing normal “nulled” myocar- Native T1 Mapping
dium (i.e., 2, 4, 5, or 6 SDs) and the full width at half maximum (FWHM) Native T1 mapping and extracellular volume fraction (ECV) are novel,
method (pixels with a signal intensity >50% of the maximum intensity emerging CMR techniques that provide a noninvasive assessment of
CHAPTER 36  Hypertrophic Cardiomyopathy 433

2° prevention
Cardiac arrest/sustained VT

1° prevention
Family history of HCM SCD
ICD
Unexplained syncope
Multiple-repetitive NSVT (Holter) Highest
Abnormal exercise BP response
Massive LVH
LGE ≥ 15% LV

Intermediate

Potential arbitrators
End-stage phase
LV apical aneurysm
Marked LV outflow obstruction (rest)
Extensive delayed enhancement
Lowest
Modifiable
Intense competitive sports
CAD
Alcohol septal ablation (?)
LGE ≥ 15% LVa
FIG. 36.10  Risk stratification model currently used to identify hypertrophic cardiomyopathy (HCM) patients
at the highest sudden cardiac death (SCD) risk who may be candidates for implantable cardioverter defibril-
lators (ICDs) and SCD prevention. Primary risk markers and potential arbitrators appear in boxes at the left.
a
Extensive late gadolinium enhancement (LGE), while a primary risk marker, can also be used as an arbitrator
when conventional risk assessment is ambiguous. BP, Blood pressure; CAD, coronary artery disease; LV,
left ventricular; LVH, left ventricular hypertrophy; NSVT, nonsustained ventricular tachycardia; VT, ventricular
tachycardia.

expanded extracellular space within the myocardium, based on the mea- at risk for developing clinical disease with LV hypertrophy, although
surement of regional absolute T1 time of myocardial tissue, and which initial experience is encouraging.20,53 To date there has been no dem-
are rapidly evolving with the current emergence of multiple sequences onstrated link to T1 mapping and outcomes within HCM, but these
and imaging strategies.53 In this regard, the measurement of the T1 data are being gathered by the HCMR.47 Thus continued investigation
time before administration of any gadolinium contrast agent allows applying T1 mapping to HCM is necessary to determine if this tech-
for measurement of a native T1 value. In contrast, ECV is a technique nique has a role within risk stratification of HCM patients for sudden
that measures the proportion of the myocardium that is occupied by death or development of HF, and perhaps, more importantly, if this
the extracellular space and is a proposed marker of interstitial fibro- technique provides incremental value beyond LGE or as an alternative
sis, performed by measuring the change in relaxivity of myocardium to LGE for patients who are not candidates for gadolinium due to renal
and plasma before and at multiple time points after administration of dysfunction.
gadolinium. Within each of these strategies are multiple methods for
measuring absolute T1 values,54 making it difficult to correlate findings
between published studies. Despite these limitations, there remains much
CONCLUSION
interest applying T1 mapping within HCM because diffuse interstitial Over the last decade, CMR has become an important imaging technique
fibrosis is an important morphologic component of the disease process in the evaluation of HCM patients and is uniquely suited to characterize
that may not be reliably detected using LGE, which more reliably detects the incredibly diverse phenotypic expression associated with this complex
focal replacement fibrosis. In this regard there have been small-scale genetic heart disease. CMR provides information that can impact a
studies which have found significant correlations between ECV values variety of clinical management issues ranging from diagnosis and family
and collagen volume fraction quantified from histopathology obtained screening, to procedural planning for invasive septal reduction therapy.
from LV tissue obtained from HCM patients.55 Extensive LGE has emerged as a potential novel marker dictating the
The early clinical experience with T1 mapping in HCM has largely HCM clinical course along two divergent adverse pathways—risk for
been confined to differentiating HCM from other cardiovascular disease sudden death with preserved EF and for advanced HF with systolic
with LV hypertrophy and in the evaluation of HCM family members. dysfunction—and therefore may identify patients who may be candidates
In this regard, both ECV and native T1 values have been found to be for primary prevention ICD therapy as well as those at risk for future
significantly higher in HCM patients compared with patients with LV development of end-stage phase. Data regarding native T1 and ECV
hypertrophy secondary to systemic hypertension or cardiac amyloidosis. are expected in the coming years. These observations help justify an
However, overlap occurs between groups, potentially limiting utility important complementary role for CMR in the routine clinical assess-
of the technique for reliable diagnosis in these clinical scenarios.53 In ment of HCM patients.
contrast, encouraging observations have been demonstrated in reli-
ably differentiating HCM from Anderson-Fabry disease using native
T1 values.56 It remains uncertain if ECV (or native T1 values) can be
REFERENCES
used to identify HCM family members who are G+P- and therefore A full reference list is available online at ExpertConsult.com
CHAPTER 36  Hypertrophic Cardiomyopathy 433.e1

REFERENCES 21. Rowin EJ, Maron MS, Lesser JR, Maron BJ. CMR with late gadolinium
enhancement in genotype positive-phenotype negative hypertrophic
1. Gersh BJ, Maron BJ, Bonow RO, et al. 2011 ACCF/AHA guidelines for the cardiomyopathy. JACC Cardiol Imagin. 2012;5:119–122.
diagnosis and treatment of hypertrophic cardiomyopathy. J Am Coll 22. Caselli S, Maron MS, Urbano-Moral JA, Pandian NG, Maron BJ, Pelliccia
Cardiol. 2011;58:e212–e260. Circulation. 2011;124:2761-2796. A. Differentiating left ventricular hypertrophy in athletes from that in
2. Elliott PM, Anastasakis A, Borger MA, et al. 2014 ESC Guidelines on patients with hypertrophic cardiomyopathy. Am J Cardiol.
diagnosis and management of hypertrophic cardiomyopathy: the Task 2014;114:1383–1389.
Force for the Diagnosis and Management of Hypertrophic 23. Ennis DB, Epstein FH, Kellman P, Fananapazir L, McVeigh ER, Arai AE.
Cardiomyopathy of the European Society of Cardiology (ESC). Eur Heart Assessment of regional systolic and diastolic dysfunction in familial
J. 2014;35:2733–2779. hypertrophic cardiomyopathy using MR tagging. Magn Reson Med.
3. Maron MS, Maron BJ, Harrigan C, et al. Hypertrophic cardiomyopathy 2003;50:638–642.
phenotype revisited after 50 years with cardiovascular magnetic 24. Kelley-Hedgepeth A, Towbin JA, Maron MS. Images in cardiovascular
resonance. J Am Coll Cardiol. 2009;54:220–228. medicine. Overlapping phenotypes: left ventricular noncompaction and
4. Autore C, Bernabo P, Barilla CS, Bruzzi P, Spirito P. The prognostic hypertrophic cardiomyopathy. Circulation. 2009;119:e588–e589.
importance of left ventricular outflow obstruction in hypertrophic 25. Maceira AM, Joshi J, Prasad SK, et al. Cardiovascular magnetic resonance
cardiomyopathy varies in relation to the severity of symptoms. J Am Coll in cardiac amyloidosis. Circulation. 2005;111:186–193.
Cardiol. 2005;45:1076–1080. 26. Gange CA, Link MS, Maron MS. Utility of cardiovascular magnetic
5. Maron BJ, Rowin EJ, Casey SA, et al. How hypertrophic cardiomyopathy resonance in the diagnosis of Anderson-Fabry disease. Circulation.
became a contemporary treatable genetic disease with low mortality: 2009;120:e96–e97.
shaped by 50-years of clinical research and practice. JAMA Cardiol. 27. Arad M, Maron BJ, Gorham JM, et al. Glycogen storage diseases
2016;1:98. presenting as hypertrophic cardiomyopathy. N Engl J Med.
6. Basso C, Thiene G, Corrado D, Buja G, Melacini P, Nava A. Hypertrophic 2005;352:362–372.
cardiomyopathy and sudden death in the young: pathologic evidence of 28. Rowin EJ, Maron BJ, Lesser JR, Rastegar H, Maron MS. Papillary muscle
myocardial ischemia. Hum Pathol. 2000;31:988–998. insertion directly into the anterior mitral leaflet in hypertrophic
7. Maron BJ, Doerer JJ, Haas TS, Tierney DM, Mueller FO. Sudden deaths cardiomyopathy, its identification and cause of outflow obstruction by
in young competitive athletes: analysis of 1866 deaths in the united states, cardiac magnetic resonance imaging, and its surgical management. Am J
1980-2006. Circulation. 2009;119:1085–1092. Cardiol. 2013;111:1677–1689.
8. Pennell DJ. Cardiovascular magnetic resonance. Circulation. 29. Gruner C, Chan RH, Crean A, et al. Significance of left ventricular
2010;121:692–705. apical-basal muscle bundle identified by cardiovascular magnetic
9. Maron MS, Maron BJ. Clinical impact of cardiovascular magnetic resonance imaging in patients with hypertrophic cardiomyopathy. Eur
resonance imaging in hypertrophic cardiomyopathy. Circulation. Heart J. 2014;35:2706–2713.
2015;132(4):292–298. 30. Patel P, Dhillon A, Popovic ZB, et al. Left ventricular outflow tract
10. Noureldin R, Liu S, Nacif M, et al. The diagnosis of hypertrophic obstruction in hypertrophic cardiomyopathy patients without severe
cardiomyopathy by cardiovascular magnetic resonance. J Cardiac Magn septal hypertrophy: implications of mitral valve and papillary muscle
Reson. 2012;14:17. abnormalities assessed using cardiac magnetic resonance and
11. Rickers C, Wilke NM, Jerosch-Herold M, et al. Utility of cardiac magnetic echocardiography. Circ Cardiovasc Imaging. 2015;8:e003132.
resonance imaging in the diagnosis of hypertrophic cardiomyopathy. 31. Minakata K, Dearani JA, Nishimura RA, Maron BJ, Danielson GK.
Circulation. 2005;112:855–861. Extended septal myectomy for hypertrophic obstructive cardiomyopathy
12. Moon JC, Fisher NG, McKenna WJ, Pennell DJ. Detection of apical with anomalous mitral papillary muscles or chordae. J Thorac Cardiovasc
hypertrophic cardiomyopathy by cardiovascular magnetic resonance in Surg. 2004;127:481–489.
patients with nondiagnostic echocardiography. Heart. 2004;90:645–649. 32. Harrigan CJ, Appelbaum E, Maron BJ, et al. Significance of papillary
13. Maron MS, Lesser JR, Maron BJ. Management implications of massive muscle abnormalities identified by cardiovascular magnetic resonance in
left ventricular hypertrophy in hypertrophic cardiomyopathy significantly hypertrophic cardiomyopathy. Am J Cardiol. 2008;101:668–673.
underestimated by echocardiography but identified by cardiovascular 33. Maron MS, Olivotto I, Maron BJ, et al. The case for myocardial ischemia
magnetic resonance. Am J Cardiol. 2010;105:1842–1843. in hypertrophic cardiomyopathy. J Am Coll Cardiol. 2009;54:866–875.
14. Maron MS, Hauser TH, Dubrow E, et al. Right ventricular involvement 34. Knaapen P, van Dockum WG, Gotte MJ, et al. Regional heterogeneity of
in hypertrophic cardiomyopathy. Am J Cardiol. 2007;100:1293–1298. resting perfusion in hypertrophic cardiomyopathy is related to delayed
15. Olivotto I, Maron MS, Autore C, et al. Assessment and significance of left contrast enhancement but not to systolic function: a PET and MRI study.
ventricular mass by cardiovascular magnetic resonance in hypertrophic J Nucl Cardiol. 2006;13:660–667.
cardiomyopathy. J Am Coll Cardiol. 2008;52:559–566. 35. Moon JC, Reed E, Sheppard MN, et al. The histologic basis of late
16. Maron MS, Finley JJ, Bos JM, et al. Prevalence, clinical significance, and gadolinium enhancement cardiovascular magnetic resonance in
natural history of left ventricular apical aneurysms in hypertrophic hypertrophic cardiomyopathy. J Am Coll Cardiol. 2004;43:2260–2264.
cardiomyopathy. Circulation. 2008;118:1541–1549. 36. Moravsky G, Ofek E, Rakowski H, et al. Myocardial fibrosis in
17. Valente AM, Lakdawala NK, Powell AJ, et al. Comparison of hypertrophic cardiomyopathy: accurate reflection of histopathological
echocardiographic and cardiac magnetic resonace imaging in findings by CMR. JACC Cardiovasc Imaging. 2013;6:587–596.
hypertrophic cardiomyopathy sarcomere mutation carriers without left 37. Chan RH, Maron BJ, Olivotto I, et al. Prognostic value of quantitative
ventricular hypertrophy. Circ Cardiovasc Genet. 2013;6:230–237. contrast-enhanced cardiovascular magnetic resonance for the evaluation
18. Maron MS, Rowin EJ, Lin D, et al. Prevalence and clinical profile of of sudden death risk in patients with hypertrophic cardiomyopathy.
myocardial crypts in hypertrophic cardiomyopathy. Circ Cardiovasc Circulation. 2014;130:484–9513.
Imaging. 2012;5:441–447. 38. O’Hanlon R, Grasso A, Roughton M, et al. Prognostic significance of
19. Maron MS, Olivotto I, Harrigan C, et al. Mitral valve abnormalities myocardial fibrosis in hypertrophic cardiomyopathy. J Am Coll Cardiol.
identified by cardiovascular magnetic resonance represent a primary 2010;56:867–874.
phenotypic expression of hypertrophic cardiomyopathy. Circulation. 39. Bruder O, Wagner A, Jensen CJ, et al. Myocardial scar visualized by
2011;124:40–47. cardiovascular magnetic resonance imaging predicts major adverse events
20. Ho CY, Abbasi SA, Neilan TG, et al. T1 measurements identify in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol.
extracellular volume expansion in hypertrophic cardiomyopathy 2010;56:875–887.
sarcomere mutation carriers with and without left ventricular 40. Rubinshtein R, Glockner JF, Ommen SR, et al. Characteristics and clinical
hypertrophy. Circ Cardiovasc Imaging. 2013;6:415–422. significance of late gadolinium enhancement by contrast-enhanced
433.e2 SECTION III  Functional Disease

magnetic resonance imaging in patients with hypertrophic 50. Flett AS, Hasleton J, Cook C, et al. Evaluation of techniques for the
cardiomyopathy. Circ Heart Fail. 2010;3:51–58. quantification of myocardial scar of differing etiology using cardiac
41. Ismail TF, Jabbour A, Gulati A, et al. Role of late gadolinium magnetic resonance. JACC Cardiovasc Imaging. 2011;4:150–156.
enhancement cardiovascular magnetic resonance in the risk stratification 51. Petersen SE, Jersosch-Herold M, Hudsmith LE, et al. Evidence for
of hypertrophic cardiomyopathy. Heart. 2014;100:1851–1858. microvascular dysfunction in hypertrophic cardiomyopathy: new insights
42. Maron MS, Appelbaum E, Harrigan CJ, et al. Clinical profile and from multiparametric magnetic resonance imaging. Circulation.
significance of delayed enhancement in hypertrophic cardiomyopathy. 2007;115:2418–2425.
Circ Heart Fail. 2008;1:184–191. 52. Shehata M, Cheng S, Osman NF, Bluemke DA, Lima JAC. Myocardial
43. Moon JC, McKenna WJ. The emerging role of cardiovascular magnetic tissue tagging with cardiac magnetic resonance. J Cardiovasc Magn Reson.
resonance in refining the diagnosis of hypertrophic cardiomyopathy. Nat 2009;11:55.
Clin Pract Cardiovasc Med. 2009;6:166–167. 53. Hinojar R, Varma N, Child N, et al. T1 mapping in discrimination of
44. Adabag AS, Maron BJ, Appelbaum E, et al. Occurrence and frequency of hypertrophic phenotypes: hypertensive heart disease and hypertrophic
arrhythmias in hypertrophic cardiomyopathy in relation to delayed cardiomyopathy: findings from the International T1 Multicenter
enhancement on cardiovascular magnetic resonance. J Am Coll Cardiol. Cardiovascular Magnetic Resonance Study. Circ Cardiovasc Imaging.
2008;51:1369–1374. 2015;8:e004232.
45. Weng Z, Yao J, Chan RH, et al. Prognostic value of LGE-CMR in HCM: a 54. Messroghli DR, Moon JC, Ferreira VM, et al. Clinical recommendations
meta-analysis. JACC Cardiovasc Imaging. 2016;9:1392–1402. for cardiovascular magnetic resonance mapping of T1, T2, T2* and
46. Chan RH, Maron BJ, Olivotto I, et al. Significance of late gadolinium extracellular volume: a consensus statement by the Society for
enhancement at right ventricular attachement to ventricular septum in Cardiovascular Magnetic Resonance (SCMR) endorsed by the European
patients with hypertrohic cardiomyopathy. Am J Cardiol. Association for Cardiovascular Imaging (EACVI). J Cardiovasc Magn
2015;116:436–441. Reson. 2017;19(1):75.
47. Kramer CM, Appelbaum E, Desai MY, et al. Hypertrophic 55. Iles LM, Ellims AH, Llewellyn H, et al. Histological validation of cardiac
Cardiomyopathy Registry: the rationale and design of an international, magnetic resonance analysis of regional and diffuse interstitial fibrosis.
observational study of hypertrophic cardiomyopathy. Am Heart J. Eur Heart J Cardiovasc Imaging. 2015;16:14–22.
2015;170:223–230. 56. Sado DM, White SK, Piechnik SK, et al. Identification and assessment of
48. Harris KM, Spirito P, Maron MS, et al. Prevalence, clinical profile, and Anderson-Fabry disease by cardiovascular magnetic resonance
significance of left ventricular remodeling in the end-stage phase of noncontrast myocardial T1 mapping. Circ Cardiovasc Imaging.
hypertrophic cardiomyopathy. Circulation. 2006;114:216–225. 2013;6:392–398.
49. Harrigan CJ, Peters DC, Gibson CM, et al. Hypertrophic cardiomyopathy:
quantification of late gadolinium enhancement with contrast-enhanced
cardiovascular MR imaging. Radiology. 2011;258:128–133.
37 
Cardiovascular Magnetic Resonance Imaging
in the Evaluation of Cardiac Transplantation
Craig Ronald Butler, Daniel H. Kim, and Ian Paterson

Cardiac transplant recipients require lifelong surveillance for common pathologic expertise.7–10 Some reports suggest that 20% of acute rejec-
and life-threatening posttransplant complications such as acute cardiac tion episodes have a false-negative EMB and are treated on clinical
allograft rejection (ACAR) and coronary allograft vasculopathy (CAV). grounds alone.11,12
Imaging plays an important role in such screening initiatives because CMR has the potential to be a sensitive test to screen for acute
most posttransplant complications have long subclinical phases and rejection, using many of the same sequences that have been helpful in
produce nonspecific symptoms as a result of denervation of the trans- nontransplant inflammatory myopathies.
planted heart. Heart transplant recipients are also at risk for posttrans-
plant lymphoproliferative disease which makes radiation-based imaging T2 Cardiovascular Magnetic Resonance Imaging
tests particularly unappealing. T2-weighted CMR imaging can demonstrate myocardial edema and
Cardiovascular magnetic resonance (CMR) is well suited to offer has proven utility in assessing a variety of inflammatory cardiomyopa-
noninvasive and nonirradiating screening for common posttrans- thies, including myocarditis,13,14 takotsubo cardiomyopathy,15 and cardiac
plant complications, often without the need for contrast. Transtho- sarcoidosis.16 Increased myocardial edema accompanying acute rejection
racic echocardiography (TTE), endomyocardial biopsy (EMB), and has been recognized since the early 1980s and has been assessed both
invasive x-ray coronary angiography play key roles in current screen- semiquantitatively, using a signal intensity ratio, and quantitatively, via
ing for heart transplant complications,1 although CMR is poised to T2 relaxation times.
make an important contribution in this area. Multiparametric CMR
is particularly well suited for comprehensive graft surveillance, offer- Signal Intensity Ratio
ing the clinical gold standard evaluation of chamber size and func- Short-inversion time inversion recovery (STIR) techniques use a triple
tion as well as characterization of myocardial inflammation and inversion recovery pulse sequence with long echo times (TEs) to produce
perfusion. images in which myocardial signal intensity is proportional to water
This chapter outlines CMR techniques to noninvasively assess acute content. These T2-weighted (T2W) images can be analyzed semiquan-
transplant rejection, CAV, as well as novel and prognostically important titatively by comparing the signal intensity of the myocardium with
adverse remodeling of the cardiac allograft. the signal intensity of skeletal muscle within the field of view. Myocardial
edema is present when the myocardial signal intensity is more than
ACUTE CARDIAC ALLOGRAFT REJECTION twice that of skeletal muscle myocardium.13 Increased myocardial edema
by T2 signal intensity ratio is a key component of the Lake Louise
Background Criteria for myocarditis.14
ACAR is a T-lymphocyte-mediated process, eventually affecting host Aherne et al.17 was one of the first to demonstrate that increased
immune responses to the transplanted heart and causing diffuse T2 signal intensity in rejecting dog hearts was linearly correlated with
myocardial inflammation, myocytolysis, graft dysfunction, and even ex vivo myocardial water content and could be prevented with the
death.2 There are extensive pathologic similarities between acute rejec- immunosuppressant cyclosporine. Studies of signal intensity ratio in
tion and other forms of inflammatory cardiomyopathies such as viral human heart transplant recipients have had mixed results.18–22 Krieghoff
myocarditis, which are often indistinguishable on histopathology.3,4 et al.19 and Taylor et al.22 demonstrated that T2 signal intensity ratio
The incidence of acute rejection is highest in the first year, at approxi- increased from 1.7 to 2.1 during clinically significant rejection. Sensitiv-
mately 25%, and accounts for 8% of early posttransplant deaths.5 The ity and specificity of T2 signal intensity were modest, at 63% and 78%,
current gold standard for the detection and grading of acute rejec- respectively.19
tion is histopathologic assessment of EMB samples. The International In general, STIR-based T2 signal intensity ratio has not demonstrated
Society of Heart and Lung Transplantation (ISHLT) published stan- sufficient sensitivity in diagnosing ACAR to warrant its use as a stand-
dardized criteria to grade the severity of rejection6: grade 0R, no signs alone clinical diagnostic tool to detect rejection.22
of rejection; grade 1R, mild; grades 2R and 3R, moderate and severe,
respectively; and grades 2R and 3R, generally thought to be clinically T2 Relaxation Time
relevant. T2-weighted images acquired at multiple echo times allow the fitting
The clinical presentation of acute rejection ranges from asymp- of a decay time constant, or T2 relaxation time (ms), that is posi-
tomatic to cardiogenic shock and necessitates frequent screening with tively correlated with myocardial water content and can be displayed
as many as 13 EMBs in the first year alone. EMB is uncomfortable as a T2 map. T2 relaxation times vary with magnetic field strength
for heart transplant recipients, can damage the cardiac allograft, and and, generally, local calibration is necessary to establish normal ref-
diagnostic accuracy can be adversely affected by sampling error and erence ranges with elevated T2 defined as greater than mean plus 2

434
CHAPTER 37  CMR Imaging in the Evaluation of Cardiac Transplantation 435

standard deviations (2 SD). T2 relaxation time is the best studied is disagreement between CMR values and biopsy results, it is not always
CMR parameter to detect acute heart transplant rejection and, in obvious where the misclassification lies. Marie et al.35 demonstrated
our opinion, holds the most promise as a noninvasive alternative to that an elevated T2 relaxation time with negative EMB (i.e., false-positive
endomyocardial biopsy. CMR) was much more likely to be diagnosed with EMB-positive rejec-
Wisenberg et al.23 first evaluated T2 relaxation time during acute tion in the next 12 weeks. Bonnemains et al.40 confirmed this in the
rejection in human heart transplant recipients in the 1980s. They dem- largest case series published to date, showing that those with intermediate
onstrated that T2 relaxation times were universally increased during T2 relaxation times had significantly shorter time to the next biopsy-
the first month following transplantation, presumably because of peri- confirmed rejection episode.
operative edema caused by ischemia reperfusion injury and surgical It is important to note that published data to date have predomi-
insults to the cardiac graft.24–26 After 24 days, T2 relaxation time had nantly enrolled heart transplant recipients more than 1 year from their
very good agreement with EMB to detect acute rejection, with sensitivity transplantation. This is clinically relevant, because the incidence of ACAR
and specificity of 93% and 96%, respectively. is highest in the first few months after transplantation and declines
Over the subsequent three decades, Wisenberg’s findings have been steadily thereafter. Miller and colleagues41 prospectively studied heart
generally well validated in animal transplant models17,27–33 and human transplant recipients during their first 6 months after transplant and
transplant recipients.34–37 Two exceptions38,39 gated their T2 acquisition to found only a borderline association between T2 relaxation time and
systole, which probably exacerbated the motion artifact and low image acute rejection. Their results may in part be explained by the fact that
signal that complicates T2 imaging, particularly at lower field strength. T2 values in participants without rejection were persistently elevated for
Marie et al.35 published the largest prospective trial to date evaluating an extended period of time after transplantation and slowly declined,
the utility of CMR to diagnose rejection at 0.5 T and found that T2 although not to control levels even at 16 weeks. Mean T2 relaxation
relaxation times were elevated when significant rejection was present time of nonrejecting participants was 59 ms at 6 weeks, 57 ms at 16
(60 ms vs. 51 ms, P > .0001), with sensitivity of 89% and specificity weeks, and 56 ms at 20 weeks compared with healthy controls at 54 ms.
of 91%. To put this into context, other trials using contemporary CMR scan-
Work from our group confirmed very good accuracy of T2 relaxation ners have found nonrejecting T2 relaxation times in the range of 53 to
time at 1.5 T using a modified half-Fourier acquisition single-shot 57 ms34,37,40 and have also shown that heart transplant recipients have
turbo spin echo (HASTE) sequence, in which we found sensitivity of systematically higher T2 relaxation time than nontransplant controls.42
88% and a negative predictive value of 95%, to diagnose acute cel- The work by Miller et al.41 implies that T2 relaxation times remain
lular or antibody mediated rejection.34 Using a more contemporary elevated because of nonrejection-related causes for longer than the 4
T2-prepared balanced steady-state free precession (bSSFP) sequence, weeks suggested by Wisenberg in the 1980s.23 This discrepancy in the
Usman et al. 37 found increased T2 relaxation times with cellular and duration of peritransplant edema between Wisenberg and Miller may
antibody-mediated rejection (Fig. 37.1) that subsequently normalized be related to their use of widely differing CMR field strength (0.15 vs.
with antirejection therapy. 1.5 T), and therefore sensitivity to detect edema as well as significant
It is problematic to judge a new diagnostic test when the gold stan- differences in immunosuppression regimens between the 1980s and
dard test is acknowledged as being imperfect. Biopsy negative rejection contemporary practice. Interpretation of the Miller study is further
has an estimated incidence of between 10% and 20%; thus when there complicated by the inclusion of two heart transplant recipients with

70



60
∗ ∗ ∗∗

Average T2 relaxation time (ms)

50

40

30

20

10

0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31 33 35 37 39 41 43 45 47 49 51 53 55 57 59 61 63 65 67
Case number
FIG. 37.1  Sequentially ordered T2 relaxation time values of heart transplant recipients receiving screening
for rejection with both cardiovascular magnetic resonance and endomyocardial biopsy (EMB). *Significant
acute rejection diagnosed by EMB or clinical variables. (Modified from Usman AA, Taimen K, Wasielewski
M, et al. Cardiac magnetic resonance T2 mapping in the monitoring and follow-up of acute cardiac transplant
rejection: a pilot study. Circ Cardiovasc Imaging, 2012;5:782–790.)
436 SECTION III  Functional Disease

primary graft failure in whom T2 relaxation times were persistently analysis, RVEDVi was independently predictive of acute rejection (89 mL/
elevated despite normal EMB histology. m2 vs. 71 mL/m2, P = .0001). We have postulated that the thinner-walled
T2 relaxation times show very good agreement with EMB for detect- right ventricle may demonstrate dysfunction related to acute rejection
ing acute rejection, and may be able to prognosticate those at risk of earlier, or in a more pronounced way, than the thicker LV wall. The
impending rejection. There remains uncertainty as to the specificity of relationship between RV volume and acute rejection is intriguing, but
T2 relaxation for acute rejection in the early posttransplant period. requires further validation.
There is also uncertainty regarding utility of T2 relaxation time when LV morphology and function are generally not sufficiently sensitive
measured at 3 T. However, we are reassured by the consistent associa- for the more subtle, although still clinically significant, acute rejection
tion between T2 and acute rejection across the spectrum of platforms episodes seen on contemporary immunosuppression. RV volume as a
and CMR field strengths. potential marker of acute rejection warrants further investigation.

T1 Quantification T1 Contrast Imaging


T1 is the longitudinal relaxation time constant of the myocardium and Early Contrast Enhancement
is influenced by molecular changes in the myocardium, including tissue Intravenous administration of gadolinium (Gd) contrast increases
edema and pathologic expansion of the extracellular space. Native T1 myocardial signal intensity on early T1-weighted imaging that is pro-
mapping has been shown to outperform T2 signal intensity ratio in portional to myocardial blood flow. Comparison of precontrast myo-
the diagnosis of acute myocarditis.43 Increased T1 relaxation time cor- cardial signal intensity with myocardial signal intensity early following
relates to inflammatory infiltrates in animal models of rejection.44 Gd contrast injection generates a relative contrast enhancement ratio
Native T1 relaxation time has not been as extensively studied as T2 that is believed to reflect the hyperemia during acute inflammation. A
relaxation time, but has been shown to increase with acute rejection 4-fold increase in myocardial signal intensity has shown to be associated
in animal transplant models17,27,30 and human heart transplant recipi- with myocardial inflammation and is a component of the Lake Louise
ents.23,45 Miller et al. used a modified Look-Locker inversion recovery Criteria for the diagnosis of myocarditis.14
(MOLLI) sequence at 1.5 T to evaluate T1 maps in the early posttrans- Early work in animal models suggested that early enhancement
plant period and found that native T1 values decline significantly between increased with rejection and could identify different grades of rejec-
6 weeks and 20 weeks (1063 ± 40 vs. 1109 ± 53 ms, P = < .001) in tion.46,51 Relative enhancement has demonstrated inconsistent association
nonrejecting heart transplants. In this cohort only a trend toward with rejection in human heart transplant rejection.18 Mousseaux et al.39
decreased native myocardial T1 values was found with rejection. In showed that the relative enhancement increased in rejection compared
contrast, Reinhardt and Gutberlet45 found good diagnostic accuracy of with nonrejection, but could not discriminate between grades of rejection.
native T1 values to detect ACAR with an area under the curve of 0.78. Taylor et al.22 assessed early enhancement using a free breathing
Native T1 mapping sequences are sensitive to water and have the spin echo sequence and found a significant increase in those with sig-
potential to detect the presence of acute rejection, although more valida- nificant rejection compared with controls (ratio 4.1 vs. 2.8, P = .01)
tion is needed. As with T2 mapping, the relationship between native T1 that resolved with rejection treatment (4.1 vs. 2.9, P < .05). Relative
mapping and acute rejection in the early posttransplant period may be enhancement was also increased in heart transplant recipients with
confounded by perioperative edema, and requires further clarification. EMB-negative rejection (8.7 vs. 2.8, P = .02). The sensitivity and speci-
ficity of early enhancement to detect significant rejection was 82% and
Morphology and Function 79%, when using an enhancement ratio cutoff of 3.5. Krieghoff et al.19
The diffuse myocardial inflammation and attendant edema of acute found more modest accuracy of early enhancement to detect ACAR
rejection can produce increased myocardial wall thickness. CMR could with sensitivity and specificity of 63% and 70%, respectively.
detect changes in myocardial wall thickness with acute rejection but The role of early enhancement in diagnosing acute transplant rejec-
was not able to discriminate different rejection grades.17,27,31,46 In general, tion is limited by its inconsistent and moderate diagnostic accuracy. In
changes in wall thickness are too insensitive to be useful in diagnosing addition, serial screening for rejection with techniques that use contrast
ACAR.20,23 add cost and risk of nephrogenic systemic fibrosis (NSF), which is of
Left ventricular ejection fraction (LVEF) is frequently within normal particular relevance for heart transplant recipients with high rates of
range during even moderate severity rejection episodes (2R). Thus LVEF chronic renal insufficiency (see Chapter 3). Long-term retention of Gd
even by highly accurate CMR cannot reliably detect grade 2R rejection has also been described52,53 in patients with normal renal function and
and therefore is not a suitable metric for rejection screening.19,22,34,41 reminds us to be judicious in our use of Gd.
Serial LVEF evaluations to monitor for unexplained reductions in systolic
function may be a useful adjunct for the detection of rejection, but this Late Gadolinium Enhancement
has not been formally tested. Regional myocardial fibrosis can be visualized using T1-weighted phase-
A reduction in LV torsion has been correlated to acute rejection and sensitive inversion recovery (PSIR) images 6 to 15 minutes (i.e., “late”)
precedes overt systolic dysfunction.47,48 Miller et al.41 used myocardial after gadolinium contrast administration. Late gadolinium enhancement
tagging to show that myocardial strain and torsion were stable over the (LGE) is generally characterized as “infarct typical” when it is transmural
first 6 months posttransplant, and reduction in peak systolic circum- or subendocardial based, and “infarct atypical” when it is located in the
ferential strain was significantly associated with acute rejection. midwall or subepicardium. LGE has clinical utility in the diagnosis of
Diastolic function has been identified as an early sign of acute rejec- inflammatory myopathies: specifically, the presence of LGE is a diag-
tion that precedes systolic dysfunction in animal models46,49; however, nostic criterion for acute myocarditis (Lake Louise Criteria, Ferreira),
its utility in echocardiography surveillance studies is highly variable,50 and the absence of LGE is characteristic of takotsubo cardiomyopathy.54
and it is poorly studied in CMR. Heart transplant recipients have a high prevalence of LGE, although
Changes in right ventricular (RV) morphology and function during the presence of LGE does not correlate to the biopsy-proven acute
acute rejection have not been well studied. Work by our group found rejection.19,22,34,41 Taylor et al.22 found a correlation between the presence
that RV ejection fraction (RVEF) and RV end-diastolic volume index of LGE and heart transplant recipients with “presumed rejection” (defined
(RVEDVi) were both abnormal during rejection.34 In multivariable in this study as “an unexplained drop in LVEF of at least 20% in whom
CHAPTER 37  CMR Imaging in the Evaluation of Cardiac Transplantation 437

EMB is negative or not practical to obtain because of clinical instabil- CMR


1.0
ity”). These cases of presumed rejection probably included instances Angio
of antibody-mediated rejection, an important source of acute rejection
generally poorly characterized in the CMR literature.
LGE has not been shown to accurately identify biopsy-proven acute 0.8
rejection and as a contrast technique is not particularly well suited for
frequent serial screening of heart transplant recipients.
0.6

Sensitivity
CORONARY ALLOGRAFT VASCULOPATHY
Heart transplant recipients commonly develop a diffuse form of arte- 0.4
riopathy, termed coronary allograft vasculopathy, that is an important
cause of late graft loss and mortality.5 One in five heart transplant
recipients have observable CAV at 1 year post transplant, increasing to
0.2 CMR 0.89 (0.79−1.00)
one-half by 10 years posttransplant.55 CAV typically produces no symp-
toms until it is severe, at which time it commonly presents as arrhythmia, Angio 0.59 (0.42−0.77)
P = .01
sudden cardiac death, or heart failure. CAV affects both the epicardial
arteries and the microvasculature, both of which independently confer 0.0
a worse prognosis.56,57 0.0 0.2 0.4 0.6 0.8 1.0
Guidelines recommend screening for CAV with invasive x-ray coro- 1 - Specificity
nary angiography or intravascular ultrasound (IVUS) to detect early FIG. 37.2  Diagnostic accuracy of cardiovascular magnetic resonance
allograft vasculopathy and apply the, albeit limited, therapies shown (CMR) myocardial perfusion reserve to detect moderate coronary allograft
to reduce allograft vasculopathy progression.1 IVUS has superior sen- vasculopathy. (Modified from Miller CA, Sarma J, Naish JH, et al. Mul-
tiparametric cardiovascular magnetic resonance assessment of cardiac
sitivity to detect CAV over x-ray angiography; however, its use is limited
allograft vasculopathy. J Am Coll Cardiol, 2014;63:799–808.)
by expense and the need for specialized expertise. Coronary angiography
with contrast-enhanced computed tomography (CT) showed good
sensitivity to detect CAV58; however, its use is limited by radiation doses
Prediction of all cardiac events
and the need for iodinated contrast. CMR coronary angiography does
not have sufficient spatial resolution to sensitively rule out early coronary 100
allograft vasculopathy, although coronary gadolinium enhancement is
Event free survival (%)

90
a novel marker that may be useful to screen for CAV.59 MPRI > 1.3
All forms of coronary angiography, as well as IVUS, exclusively 80
evaluate the epicardial vessels and offer no practical insight into the MPRI ≤ 1.3
70
presence of CAV in the microvasculature. Assessing myocardial perfu-
sion has the advantage of being able to evaluate the presence of allograft 60
vasculopathy in both the epicardial and microvascular compartments. P < .001
50 X2 = 30.9
Nuclear perfusion modalities are disadvantaged compared with CMR HR = 0.03
by their need for radiation exposure. 40
CMR perfusion is used routinely in clinical practice with excellent 0 1 2 3 4 5 6
Time (years)
diagnostic accuracy and prognostic value in patients with suspected
FIG. 37.3  Association between myocardial perfusion reserve and car-
coronary artery disease.60–62 CMR-derived myocardial perfusion reserve
diovascular death and acute coronary syndrome. HR, Hazard ratio; MPRI,
(MPR) has shown particular utility in diagnosing CAV. MPR is a contrast-
myocardial perfusion reserve index. (Modified from Hofmann NP, Steuer
enhanced technique that compares contrast-enhanced myocardial per- C, Voss A, et al. Comprehensive bio-imaging using myocardial perfusion
fusion during hyperemia and at rest. Meuhling et al.63 were among the reserve index during cardiac magnetic resonance imaging and high-
first to show that reduced MPR was associated with CAV severity, and sensitive troponin T for the prediction of outcomes in heart transplant
an MPR >2.3 could dichotomously exclude CAV with a sensitivity and recipients. Am J Transplant. 2014;14:2607–2616.)
specificity of 100% and 85%, respectively.
Exemplary work by Miller et al.64 compared the diagnostic accuracy
of CMR-derived MPR to detect CAV against invasive CAV assessment MPR has significant diagnostic and prognostic potential in heart
in 48 heart transplant recipients. Multivariable analysis found that MPR transplant recipients with CAV; however, its routine clinical use is ham-
was the only variable that independently predicted CAV in both the pered by complicated and time-consuming postprocessing. MPR is a
epicardial and microvascular compartments. An MPR of >1.94 had a Gd contrast-enhanced technique which is not ideal for serial evaluation,
sensitivity of 88% and specificity of 85%, respectively, to detect moder- although assessments for CAV are typically far less frequent (i.e., annual
ate allograft vasculopathy and outperformed invasive x-ray angiography, or semiannual) than assessments for acute rejection. MPR evaluation
fractional flow reserve, and invasively measured index of microvascular is also not suitable for heart transplant recipients with advanced kidney
resistance (Fig. 37.2). disease. Efforts are needed to facilitate automated MPR calculation to
Hofmann et al.65 studied 108 heart transplant recipients and con- move this technique into the clinical mainstream.
firmed that MPR worsened with the degree of CAV observed on invasive
x-ray angiography, and that change in MPR over time mirrored pro-
gression of CAV. This cohort was followed for a mean of 4.2 years and
ALLOGRAFT REMODELING AND PROGNOSIS
demonstrated that MPR independently predicted cardiac death and The variables that influence long-term ventricular remodeling of the
acute coronary syndrome (Fig. 37.3). cardiac allograft are not well understood but include transplant rejection
438 SECTION III  Functional Disease

episodes, CAV, chronic immunosuppressive therapy, and traditional Epstein-Barr virus and cytomegalovirus infection and found no asso-
risk factors for remodeling such as hypertension and diabetes melli- ciation with LGE prevalence.76
tus.66 Most heart transplant recipients are currently followed with serial LGE confers worse prognosis in many myocardial pathologies, includ-
TTE, which is less sensitive than CMR to detect changes in LV mass ing dilated cardiomyopathy,77,78 sarcoidosis,79 myocarditis,80 hypertrophic
or volume.67 CMR can also provide unique insight into allograft tissue cardiomyopathy,81,82 and myocardial infarction.83,84 Heart transplant
characterization that may have important prognostic and ultimately recipients with LGE have reduced LV and RV ejection fractions and
therapeutic implications. have higher rates of cardiovascular death and hospitalization (Fig. 37.4)
A minority of adult heart transplanted allografts will survive past as well as all-cause mortality.70,71
15 years, but those that do tend to have normal LV size and LVEF.68 The PSIR techniques currently used to quantify infarct atypical LGE
However, the ISHLT registry has not identified any noninvasive imaging in heart transplant recipients only measure regional fibrosis, in what
variables that predict short-term or long-term grafts loss,5,69 despite is very likely a diffuse pathology. Therefore LGE probably underestimates
ongoing recommendation for annual echocardiography.1,50 the diffuse fibrotic changes that occur in the cardiac allograft over time
Longitudinal evaluation of heart transplant recipients with CMR and with repeated posttransplant complications. T1-derived estimates
has identified three morphologic parameters independently associated of extracellular volume (ECV) are well suited to provide a much more
with adverse outcome: LV mass, LGE, and RV volume.70–72 sensitive measure of myocardial fibrosis in heart transplant recipients.
Patel and colleagues72 evaluated 140 heart transplant recipients with There is very limited data regarding T1-derived ECV in heart transplant
CMR and identified concentric LV hypertrophy as independently pre- recipients, although Miller et al.41 showed ECV declined during the
dictive of shorter time to cardiovascular and all-cause mortality on study period independent of any identifiable clinical event (ECV 30%
multivariable survival analysis. In this population, LV hypertrophy was at 6 weeks vs. 28% at 20 weeks, P = .001).
more common in heart transplant recipients with longer time since LGE is a novel marker of prognosis in heart transplant recipients
transplant, diabetes mellitus, and in those with a history of rejection. and continued efforts are needed to explore the likely multifactorial
Increased LV mass may result from increased collagen deposition in pathogenesis. Furthermore, newer techniques such as T1-derived ECV
the extracellular matrix and myocyte hypertrophy in response to repeated are probably much more sensitive to fibrotic changes in the cardiac
immune injury to the graft over time or during repeated rejection allograft and may well predict adverse outcome and remodeling. Lon-
episodes.73,74 gitudinal evaluation of native T1 mapping in heart transplant recipients
LGE is prevalent in heart transplant populations in both infarct with correlation to clinical and outcome variables are needed.
typical and infarct atypical forms. Infarct typical LGE has a prevalence Lastly, RV remodeling posttransplant is poorly understood, probably
ranging from 5% to 37%41,75 and correlates with degree of CAV.71,75 because the bulk of heart transplant imaging literature to date has used
Infarct atypical LGE has a prevalence ranging from 40% to 51%, although TTE, which has difficulty quantifying RV parameters. RV size and func-
the etiology remains elusive.70,71,75 Miller et al.41 found infarct atypical tion are readily quantified by CMR and its thin walls may demonstrate
enhancement in only 1 of 22 participants (5%) within their first 20 myopathy earlier compared with the thicker LV wall. RV size and per-
weeks posttransplantation, suggesting that donor variables and peri- formance has been identified as an important prognostic marker in
operative injury may not be major contributors. Infarct atypical LGE heart failure85–87 as well as LV assist devices.88 Work by our group has
is associated with older graft age,70 inconsistently associated with shown that RVEDVi independently predicts cardiovascular death or
CAV,70,71,75 and has not shown association with acute or previously hospitalization in heart transplant recipients, even when controlling
reported rejection.19,22,70,71 Our group recently evaluated posttransplant for clinical variables.70 The natural history of RV size and function post

Kaplan-Meier survival curves:


cardiovascular death or hospitalization
1.00 0.75
Event free survival
0.25 0.50
0.00

0 2 4 6 8
Follow-up time
−Myocardial scar +Myocardial scar

FIG. 37.4  Kaplan-Meier survival curve for cardiovascular death or hospitalization with the presence of late
gadolinium enhancement. (Modified from Butler CR, Kim DH, Chow K, et al. Cardiovascular MRI predicts
5-year adverse clinical outcome in heart transplant recipients. Am J Transplant. 2014;14:2055–2061.)
CHAPTER 37  CMR Imaging in the Evaluation of Cardiac Transplantation 439

transplantation warrants further study, as it may identify novel markers of myocardial fibrosis post transplantation, as well as improving
of cardiac allograft health. our understanding of changes in ECV over time and in response to
posttransplant complications. Lastly, the inclusion of CMR in serial
transplant evaluations can provide early identification of grafts with
CONCLUSION high-risk features for poor outcome such as concentric LV hyper-
CMR can provide multiparametric assessment of cardiac allografts trophy, development or progression of myocardial fibrosis, or RV
with particular utility in the diagnosis of acute heart transplant rejec- enlargement that may prompt closer follow-up or ultimately inform
tion, perfusion abnormalities related to CAV, as well as characteriza- therapeutic trials.
tion of prognostically important adverse allograft remodeling. Work
remains to demonstrate the safety of CMR-based acute rejection
screening strategies compared with EMB alone in a prospective ran-
REFERENCES
domized trial format. Insight is also needed into the etiopathology A full reference list is available online at ExpertConsult.com
CHAPTER 37  CMR Imaging in the Evaluation of Cardiac Transplantation 439.e1

REFERENCES 23. Wisenberg G, et al. Diagnostic applicability of magnetic resonance


imaging in assessing human cardiac allograft rejection. Am J Cardiol.
1. Costanzo MR, et al. The International Society of Heart and Lung 1987;60(1):130–136.
Transplantation Guidelines for the care of heart transplant recipients. 24. Albers J, et al. 3D evaluation of myocardial edema: experimental study on
J Heart Lung Transplant. 2010;29(8):914–956. 22 pigs using magnetic resonance and tissue analysis. Thorac Cardiovasc
2. Davis MK, et al. State of the art: cardiac transplantation. Trends Surg. 2001;49(4):199–203.
Cardiovasc Med. 2014;24(8):341–349. 25. Pereira NL, et al. Myocardial mechanisms causing heart failure early after
3. de Souza MM, et al. Comparative histopathology of endomyocardial cardiac transplantation. Transplant Proc. 2006;38(9):2999–3003.
biopsies in chagasic and non-chagasic heart transplant recipients. J Heart 26. Vahl CF, et al. Heterogeneity of myocardial edema in isolated pig hearts
Lung Transplant. 2001;20(5):534–543. after perfusion with different types of cardioprotective solutions. Thorac
4. Mozaffari K, et al. Diagnostic pitfalls and challenges in interpretation of Cardiovasc Surg. 1998;46(5):285–292.
heart transplantation rejection in endomyocardial biopsies with focus on 27. Huber DJ, et al. The detection of cardiac allograft rejection by alterations
our experience. Res Cardiovasc Med. 2014;3(1):e13986. in proton NMR relaxation times. Invest Radiol. 1985;20(8):796–802.
5. Lund LH, et al. The Registry of the International Society for Heart and 28. Kurland RJ, et al. Magnetic resonance imaging to detect heart transplant
Lung Transplantation: Thirty-Second Official Adult Heart rejection: sensitivity and specificity. Transplant Proc. 1989;21(1 Pt
Transplantation Report—2015; Focus Theme: Early Graft Failure. J Heart 3):2537–2543.
Lung Transplant. 2015;34(10):1244–1254. 29. Nishimura T, et al. Assessment of severity of cardiac rejection in
6. Stewart S, et al. Revision of the 1990 working formulation for the heterotopic heart transplantation using indium-111 antimyosin and
standardization of nomenclature in the diagnosis of heart rejection. magnetic resonance imaging. Cardiovasc Res. 1988;22(2):108–112.
J Heart Lung Transplant. 2005;24(11):1710–1720. 30. Nishimura T, et al. Identification of cardiac rejection with magnetic
7. Angelini A, et al. A web-based pilot study of inter-pathologist resonance imaging in heterotopic heart transplantation model. Heart
reproducibility using the ISHLT 2004 working formulation for biopsy Vessels. 1987;3(3):135–140.
diagnosis of cardiac allograft rejection: the European experience. J Heart 31. Sasaguri S, et al. Early detection of cardiac allograft rejection with proton
Lung Transplant. 2011;30(11):1214–1220. nuclear magnetic resonance. Circulation. 1985;72(3 Pt 2):II231–II236.
8. Baraldi-Junkins C, et al. Complications of endomyocardial biopsy in 32. Sasaki H, et al. The expanded scope of effectiveness of nuclear magnetic
heart transplant patients. J Heart Lung Transplant. 1993;12(1 Pt 1):63–67. resonance imaging to determine cardiac allograft rejection. Transplant
9. Huddleston CB, et al. Biopsy-induced tricuspid regurgitation after cardiac Proc. 1987;19(1 Pt 2):1062–1064.
transplantation. Ann Thorac Surg. 1994;57(4):832–836, discussion 33. Tscholakoff D, et al. Cardiac transplantations in dogs: evaluation with
836–837. MR. Radiology. 1985;157(3):697–702.
10. Marboe CC, et al. Nodular endocardial infiltrates (Quilty lesions) cause 34. Butler CR, et al. Correlation of cardiovascular magnetic resonance
significant variability in diagnosis of ISHLT Grade 2 and 3A rejection in imaging findings and endomyocardial biopsy results in patients
cardiac allograft recipients. J Heart Lung Transplant. 2005;24(7 undergoing screening for heart transplant rejection. J Heart Lung
suppl):S219–S226. Transplant. 2015;34(5):643–650.
11. Tang Z, et al. The natural history of biopsy-negative rejection after heart 35. Marie PY, et al. Detection and prediction of acute heart transplant
transplantation. J Transplant. 2013;2013:236720. rejection with the myocardial T2 determination provided by a black-
12. Subherwal S, et al. Incidence of acute cellular rejection and non-cellular blood magnetic resonance imaging sequence. J Am Coll Cardiol.
rejection in cardiac transplantation. Transplant Proc. 2001;37(3):825–831.
2004;36(10):3171–3172. 36. Marie PY, et al. Detection and prediction of acute heart transplant
13. Abdel-Aty H, et al. Diagnostic performance of cardiovascular magnetic rejection: preliminary results on the clinical use of a “black blood”
resonance in patients with suspected acute myocarditis: comparison of magnetic resonance imaging sequence. Transplant Proc. 1998;30(5):
different approaches. J Am Coll Cardiol. 2005;45(11):1815–1822. 1933–1935.
14. Friedrich MG, et al. Cardiovascular magnetic resonance in myocarditis: a 37. Usman AA, et al. Cardiac magnetic resonance T2 mapping in the
JACC White Paper. J Am Coll Cardiol. 2009;53(17):1475–1487. monitoring and follow-up of acute cardiac transplant rejection: a pilot
15. Abdel-Aty H, et al. Myocardial edema is a feature of Tako-Tsubo study. Circ Cardiovasc Imaging. 2012;5(6):782–790.
cardiomyopathy and is related to the severity of systolic dysfunction: 38. Doornbos J, et al. MR imaging in assessment of cardiac transplant
insights from T2-weighted cardiovascular magnetic resonance. Int J rejection in humans. J Comput Assist Tomogr. 1990;14(1):77–81.
Cardiol. 2009;132(2):291–293. 39. Mousseaux E, et al. Assessing human cardiac allograft rejection using
16. Vignaux O, et al. Clinical significance of myocardial magnetic resonance MRI with Gd-DOTA. J Comput Assist Tomogr. 1993;17(2):237–244.
abnormalities in patients with sarcoidosis: a 1-year follow-up study. 40. Bonnemains L, et al. Diagnostic and prognostic value of MRI T2
Chest. 2002;122(6):1895–1901. quantification in heart transplant patients. Transpl Int. 2014;27(1):69–76.
17. Aherne T, et al. Magnetic resonance imaging of cardiac transplants: the 41. Miller CA, et al. Multiparametric cardiovascular magnetic resonance
evaluation of rejection of cardiac allografts with and without surveillance of acute cardiac allograft rejection and characterisation of
immunosuppression. Circulation. 1986;74(1):145–156. transplantation-associated myocardial injury: a pilot study. J Cardiovasc
18. Almenar L, et al. Utility of cardiac magnetic resonance imaging for the Magn Reson. 2014;16:52.
diagnosis of heart transplant rejection. Transplant Proc. 2003;35(5): 42. Markl M, et al. Myocardial T2-mapping and velocity mapping: changes
1962–1964. in regional left ventricular structure and function after heart
19. Krieghoff C, et al. Assessment of sub-clinical acute cellular rejection after transplantation. Magn Reson Med. 2013;70(2):517–526.
heart transplantation: comparison of cardiac magnetic resonance imaging 43. Ferreira VM, et al. T(1) mapping for the diagnosis of acute myocarditis
and endomyocardial biopsy. Eur Radiol. 2014;24(10):2360–2371. using CMR: comparison to T2-weighted and late gadolinium enhanced
20. Revel D, et al. Magnetic resonance imaging of human orthotopic heart imaging. JACC Cardiovasc Imaging. 2013;6(10):1048–1058.
transplantation: correlation with endomyocardial biopsy. J Heart 44. Jeuthe S, et al. Myocardial T1 maps reflect histological findings in acute
Transplant. 1989;8(2):139–146. and chronic stages of myocarditis in a rat model. J Cardiovasc Magn
21. Smart FW, et al. Magnetic resonance imaging for assessment of tissue Reson. 2016;18(1):19.
rejection after heterotopic heart transplantation. J Heart Lung Transplant. 45. Reinhardt L, Gutberlet M. Diagnostic performance of T1- and
1993;12(3):403–410. T2-mapping in HTx patients to identify acute cellular rejection (ACR) in
22. Taylor AJ, et al. Diagnostic performance of multisequential cardiac comparison to conventional CMR techniques and endomyocardial biopsy
magnetic resonance imaging in acute cardiac allograft rejection. Eur J (EMB) as the standard reference. J Cardiovasc Magn Reson. 2015;17
Heart Fail. 2010;12(1):45–51. (suppl 1):Q54.
439.e2 SECTION III  Functional Disease

46. Yoshida S, et al. Cardiac function of transplanted rat hearts using a 65. Hofmann NP, et al. Comprehensive bio-imaging using myocardial
working heart model assessed by magnetic resonance imaging. J Heart perfusion reserve index during cardiac magnetic resonance imaging and
Lung Transplant. 1999;18(11):1054–1064. high-sensitive troponin T for the prediction of outcomes in heart
47. Donofrio MT, et al. Regional wall motion and strain of transplanted transplant recipients. Am J Transplant. 2014;14(11):2607–2616.
hearts in pediatric patients using magnetic resonance tagging. Am J 66. van Hoeven KH, Factor SM. A comparison of the pathological spectrum
Physiol. 1999;277(5 Pt 2):R1481–R1487. of hypertensive, diabetic, and hypertensive-diabetic heart disease.
48. Hansen DE, et al. Effect of acute human cardiac allograft rejection on left Circulation. 1990;82(3):848–855.
ventricular systolic torsion and diastolic recoil measured by 67. Bellenger NG, et al. Comparison of left ventricular ejection fraction and
intramyocardial markers. Circulation. 1987;76(5):998–1008. volumes in heart failure by echocardiography, radionuclide
49. Yoshida S, et al. Diastolic dysfunction coincides with early mild ventriculography and cardiovascular magnetic resonance; are they
transplant rejection: in situ measurements in a heterotopic rat heart interchangeable? Eur Heart J. 2000;21(16):1387–1396.
transplant model. J Heart Lung Transplant. 1998;17(11):1049– 68. Mastrobuoni S, et al. Allograft morphology and function in heart
1056. transplant recipients surviving more than 15 years by magnetic resonance
50. Badano LP, et al. European Association of Cardiovascular Imaging/ imaging and dual-source computed tomography. Eur J Cardiothorac Surg.
Cardiovascular Imaging Department of the Brazilian Society of 2011;40(1):e62–e66.
Cardiology recommendations for the use of cardiac imaging to assess and 69. Hertz MI. The Registry of the International Society for Heart and Lung
follow patients after heart transplantation. Eur Heart J Cardiovasc Transplantation—Introduction to the 2012 annual reports: new
Imaging. 2015;16(9):919–948. leadership, same vision. J Heart Lung Transplant. 2012;31(10):1045–1051.
51. Konstam MA, et al. Magnetic resonance imaging with gadolinium-DTPA 70. Butler CR, et al. Cardiovascular MRI predicts 5-year adverse clinical
for detecting cardiac transplant rejection in rats. Circulation. 1988;78(5 Pt outcome in heart transplant recipients. Am J Transplant.
2):III87–III94. 2014;14(9):2055–2061.
52. Kanda T, et al. Gadolinium-based contrast agent accumulates in the brain 71. Butler CR, et al. Late gadolinium enhancement in cardiac transplant
even in subjects without severe renal dysfunction: evaluation of autopsy patients is associated with adverse ventricular functional parameters and
brain specimens with inductively coupled plasma mass spectroscopy. clinical outcomes. Can J Cardiol. 2013;29(9):1076–1083.
Radiology. 2015;276(1):228–232. 72. Patel PC, et al. Concentric left ventricular hypertrophy as assessed by
53. McDonald RJ, et al. Intracranial gadolinium deposition after contrast- cardiac magnetic resonance imaging and risk of death in cardiac
enhanced MR imaging. Radiology. 2015;275(3):772–782. transplant recipients. J Heart Lung Transplant. 2010;29(12):1369–
54. Eitel I, et al. Clinical characteristics and cardiovascular magnetic 1379.
resonance findings in stress (takotsubo) cardiomyopathy. JAMA. 73. Armstrong AT, et al. Quantitative investigation of cardiomyocyte
2011;306(3):277–286. hypertrophy and myocardial fibrosis over 6 years after cardiac
55. Prada-Delgado O, et al. Prevalence and prognostic value of cardiac transplantation. J Am Coll Cardiol. 1998;32(3):704–710.
allograft vasculopathy 1 year after heart transplantation according to the 74. Stetson SJ, et al. Cardiac hypertrophy after transplantation is associated
ISHLT recommended nomenclature. J Heart Lung Transplant. with persistent expression of tumor necrosis factor-alpha. Circulation.
2012;31(3):332–333. 2001;104(6):676–681.
56. Kobashigawa JA, et al. Multicenter intravascular ultrasound validation 75. Steen H, et al. Prevalence of different gadolinium enhancement patterns
study among heart transplant recipients: outcomes after five years. J Am in patients after heart transplantation. J Am Coll Cardiol.
Coll Cardiol. 2005;45(9):1532–1537. 2008;52(14):1160–1167.
57. Tuzcu EM, et al. Intravascular ultrasound evidence of angiographically 76. Butler CR, et al. Cardiac MRI of heart transplant recipients with previous
silent progression in coronary atherosclerosis predicts long-term CMV infection demonstrates ventricular hypertrophy and dysfunction.
morbidity and mortality after cardiac transplantation. J Am Coll Cardiol. J Heart Lung Transplant. 2014;33(4):S136.
2005;45(9):1538–1542. 77. Cho JR, et al. Delayed enhancement magnetic resonance imaging is a
58. Wever-Pinzon O, et al. Coronary computed tomography angiography for significant prognostic factor in patients with non-ischemic
the detection of cardiac allograft vasculopathy: a meta-analysis of cardiomyopathy. Circ J. 2010;74(3):476–483.
prospective trials. J Am Coll Cardiol. 2014;63(19):1992–2004. 78. Kuruvilla S, et al. Late gadolinium enhancement on cardiac magnetic
59. Hussain T, et al. Detection and grading of coronary allograft vasculopathy resonance predicts adverse cardiovascular outcomes in nonischemic
in children with contrast-enhanced magnetic resonance imaging of the cardiomyopathy: a systematic review and meta-analysis. Circ Cardiovasc
coronary vessel wall. Circ Cardiovasc Imaging. 2013;6(1):91–98. Imaging. 2014;7(2):250–258.
60. Desai RR, Jha S. Diagnostic performance of cardiac stress perfusion MRI 79. Greulich S, et al. CMR imaging predicts death and other adverse events
in the detection of coronary artery disease using fractional flow reserve as in suspected cardiac sarcoidosis. JACC Cardiovasc Imaging.
the reference standard: a meta-analysis. AJR Am J Roentgenol. 2013;6(4):501–511.
2013;201(2):W245–W252. 80. Barone-Rochette G, et al. Potentially simple score of late gadolinium
61. Greenwood JP, et al. Prognostic value of cardiovascular magnetic enhancement cardiac MR in acute myocarditis outcome. J Magn Reson
resonance and single-photon emission computed tomography in Imaging. 2014;40(6):1347–1354.
suspected coronary heart disease: long-term follow-up of a prospective, 81. Adabag AS, et al. Occurrence and frequency of arrhythmias in
diagnostic accuracy cohort study. Ann Intern Med. 2016;[Epub ahead hypertrophic cardiomyopathy in relation to delayed enhancement on
of print]. cardiovascular magnetic resonance. J Am Coll Cardiol. 2008;51(14):
62. Greenwood JP, et al. Comparison of cardiovascular magnetic resonance 1369–1374.
and single-photon emission computed tomography in women with 82. Bruder O, et al. Myocardial scar visualized by cardiovascular magnetic
suspected coronary artery disease from the Clinical Evaluation of resonance imaging predicts major adverse events in patients with
Magnetic Resonance Imaging in Coronary Heart Disease (CE-MARC) hypertrophic cardiomyopathy. J Am Coll Cardiol. 2010;56(11):
Trial. Circulation. 2014;129(10):1129–1138. 875–887.
63. Muehling OM, et al. Reduced myocardial perfusion reserve and 83. Ingkanisorn WP, et al. Gadolinium delayed enhancement cardiovascular
transmural perfusion gradient in heart transplant arteriopathy assessed magnetic resonance correlates with clinical measures of myocardial
by magnetic resonance imaging. J Am Coll Cardiol. 2003;42(6):1054– infarction. J Am Coll Cardiol. 2004;43(12):2253–2259.
1060. 84. Kwong RY, et al. Impact of unrecognized myocardial scar detected by
64. Miller CA, et al. Multiparametric cardiovascular magnetic resonance cardiac magnetic resonance imaging on event-free survival in patients
assessment of cardiac allograft vasculopathy. J Am Coll Cardiol. presenting with signs or symptoms of coronary artery disease.
2014;63(8):799–808. Circulation. 2006;113(23):2733–2743.
CHAPTER 37  CMR Imaging in the Evaluation of Cardiac Transplantation 439.e3

85. Ghio S, et al. Independent and additive prognostic value of right 87. Shah PK, et al. Variable spectrum and prognostic implications of left and
ventricular systolic function and pulmonary artery pressure in right ventricular ejection fractions in patients with and without clinical
patients with chronic heart failure. J Am Coll Cardiol. 2001;37(1): heart failure after acute myocardial infarction. Am J Cardiol. 1986;58(6):
183–188. 387–393.
86. Polak JF, et al. Right ventricular ejection fraction: an indicator of 88. Morgan JA, et al. Is severe right ventricular failure in left ventricular assist
increased mortality in patients with congestive heart failure associated device recipients a risk factor for unsuccessful bridging to transplant and
with coronary artery disease. J Am Coll Cardiol. 1983;2(2):217–224. post-transplant mortality. Ann Thorac Surg. 2004;77(3):859–863.
38 
Cardiac and Paracardiac Masses
Herbert Frank, Teresa Sykora, and Francisco Alpendurada

Cardiovascular magnetic resonance (CMR) provides a noninvasive and For evaluation of cardiac function and tumor mobility, gradient
three-dimensional (3D) assessment of masses involving the cardiac recalled echo (GRE) techniques (TE 4–12 ms) are recommended.6,7
chambers, the pericardium, and extracardiac structures. CMR has become GRE images are generally T1 weighted and are characterized by bright
an established method to yield complementary diagnostic information signal intensity (SI) from rapidly flowing blood, which is useful for the
and to guide cardiac surgeons in the design of an appropriate therapeutic differentiation of thrombus or the assessment of turbulent flow in case
strategy. The goals of CMR for assessing cardiac and paracardiac masses of valvular regurgitation or intracardiac shunts. Because of the low soft
are to confirm or to exclude a mass initially suspected by echocardiog- tissue contrast, visualization of cardiac masses is not as adequate as
raphy,1 to assess the location, mobility, and its relationship to surrounding with SE techniques, unless they are intraluminal. Steady-state free pre-
tissues,2 to image the degree of vascularity,3 to distinguish solid from cession (SSFP) is a type of GRE optimized for assessment of cardiac
fluid lesions,4 and to determine tissue characteristics and the specific function, offering the highest SNR per unit of time of all imaging
nature of a mass.5 sequences.8 The resulting weighting relies on a T2/T1 ratio, where both
fat and water return high signal as opposed to low signal from the
myocardium. Because of the intrinsic homogeneous contrast between
TECHNICAL CONSIDERATIONS the blood pool and the heart during the cardiac cycle, along with an
For adequate image quality with reduced motion artifacts, CMR is excellent spatial and temporal resolution, SSFP is the sequence of choice
performed using electrocardiogram (ECG) gating.1 Alternatively, the to assess the dynamic relationship of masses with the heart and vessels.
navigator techniques allow for combined ECG and respiratory trigger- Unlike two-dimensional (2D) echocardiography, CMR has the poten-
ing.2 Spin echo (SE) sequences provide detailed morphologic informa- tial to some extent for tissue characterization by comparing the T1 and
tion of the heart, the great vessels, and the adjacent structures.3 For T2 values of the mass to a reference tissue:9 that is based on the obser-
T1-weighted sequences, the echo time (TE) is usually 20 to 30 ms, and vation that significant differences exist in T1 and T2 relaxation times.
the repetition time (TR) is dependent on the R-R interval. A longer Previous studies have tried to determine specific T1 and T2 relaxation
TR is used for T2-weighted sequences and the TE is typically 50 to times of different tissues10; however, precise etiologic diagnoses are not
90 ms.4 T1-weighted images provide a better signal-to-noise ratio (SNR) possible.11,12 T1 and T2 mapping CMR is a novel parametric approach
and excellent soft tissue contrast between epicardial fat, myocardium, to tissue characterization and has shown promising results for the assess-
and rapidly flowing blood.3 T2-weighted images have increased image ment of cardiac thrombus and may have a potential role in overcoming
contrast for water-laden tissues with inflammation or necrosis or fluid- some of the limitations.13
filled cysts. However, T2-weighted images in general have worse SNR
(Table 38.1).
The fast or turbo spin echo (TSE) technique combines the acquisi-
CONTRAST AGENTS
tion of multiple profiles per excitation with the multislice mode, resulting The most commonly used CMR contrast agent uses the paramagnetic
in a marked reduction in imaging time. The contrast of those images element gadolinium in a complex with chelating molecules (e.g., with
is similar to that of SE images with the same TR and an equivalent TE. diethylenetriaminepentaacetic acid [DTPA]14) to reduce toxicity. A
Fat is usually brighter with TSE than regular SE pulse sequences. TSE number of agents are available. The distribution of nearly all currently
permits acquisition of T1- and T2-weighted scans in a fraction of time available intravenous contrast agents is extracellular and contrast is
compared with the conventional spin echo sequence. Furthermore, this enhanced on T1-weighted images. The normal concentration is 0.1 mmol/
technique has a reduced susceptibility to motion artifacts and is insensi- kg body weight.15 Gadolinium provides a better delineation of the mass
tive to field inhomogeneity. The combination with inversion recovery by different enhancement of myocardium and tumor tissue because of
and breath-hold techniques has led to improved image quality and variation of tissue vascularity.16
tissue characterization.5 When combined with an inversion recovery
(IR) technique, the signal from fat can be suppressed by using a short
inversion time before the SE pulse. Most tissues have a T1 relaxation
BENIGN TUMORS OF THE HEART
time longer than that of fat, resulting in a signal increase on T1- and Primary tumors of the heart are very rare, with an estimated inci-
T2-weighted images. This leads to better contrast in these images. Appli- dence between 0.002% and 0.2% in unselected patients at autopsy.
cations of the IR technique are the distinct recognition of fatty tissues Three-quarters of the tumors are benign and nearly half of them are
and the improved visualization of structures that are surrounded by myxomas and about 10% are lipomas.17–19 Fibroelastomas, rhabdomyo-
fatty tissue, which often makes image analysis complicated. mas, fibromas, hemangiomas, teratomas, and mesotheliomas are found

440
CHAPTER 38  Cardiac and Paracardiac Masses 441

TABLE 38.1  Typical Technical Parameters at 1.5 T for Evaluation of Cardiac or


Paracardiac Masses
Sequence Technical Features Indication Advantages/Disadvantages
T1-weighted spin echo 8–12 slices, thickness: 6–10 mm, NSA = 2, Defining anatomic structures, delineation A: Excellent soft tissue contrast
TE: 20–25 ms, TR: shortest, time 4–6 min, of the mass to the adjacent tissue, D: Respiratory and flow artifacts
axial, sagittal, and coronal orientation visualization of vascular walls
T2-weighted spin echo, 8–12 slices, thickness: 6–10 mm, NSA = 2, Detecting the nature of cardiac masses by A: Better tissue contrast,
double echo TE: 50, 90 ms, TR: every 2nd or 3rd abnormal T2 values. demonstration of fluid components
heartbeat, time: 5–8 min, axial D: Very long examination time, lower
orientation SNR, and increased motion artifacts
T1- and T2-weighted Examination time: 2–4 min, also breath- A: Shorter imaging times, reduced
fast spin echo hold technique motion artifacts
D: Less SNR
Short inversion recovery Inversion pulse, combination with spin echo Suppression of fatty tissue A: Eliminating artifacts from fat signal
or TSE
T1-weighted spin echo Axial orientation, antecubital Gd-DTPA Signal intensity behavior of suspected To assess invasive and infiltrative
with contrast agent administration (dose: 0, 1 mmol/kg) masses, assessment of the degree of components of a tumor
administration vascularity
Cine gradient echo with 1–3 slices, thickness 8–10 mm, TE: 9 ms, Imaging the hemodynamic effects of a A: Functional information
flow compensation TR: shortest, 16 phases NSA = 2, time: mass, i.e., mobility, transvalvular flow, D: Low soft tissue contrast
1–3 min differentiation between blood flow and
thrombus, identify turbulent flow regions
Cine gradient echo EPI Breath-hold technique, 1–3 slices, Equal to conventional gradient echo A: Shorter acquisition time, reduced
thickness, 8–10 mm, TE: shortest, TR: motion artifacts
shortest, time: 10–40 s D: Less SNR

EPI, Echo planar imaging; Gd-DTPA, gadolinium-DTPA; NSA, numbers of signal averaged; SNR, signal-to-noise ratio; TE, echo time; TSE, turbo
spin echo; TR, repetition time.
Modified from Hoffman U, Globits S, Frank H. Cardiac and paracardiac masses: current opinion on diagnostic evaluation by magnetic resonance
imaging. Eur Heart J. 1998;19:553–563.

less frequently. Granular cell tumors, neurofibromas, and lymphangiomas be isointense or slightly hyperintense relative to the myocardium but
are very rare.20 hypointense relative to the blood pool. Therefore the tumor can always
be distinguished from the higher signal of the surrounding slowly flowing
Myxoma blood.31,32 Intratumorous areas of subacute or chronic hemorrhage are
Myxomas comprise 30% to 50% of all primary cardiac tumors, occur typically characterized by high SI on both short and long TE.33 Myxomas
frequently between the third and sixth decade of life, and have a slight show a moderately high and heterogeneous contrast enhancement after
female predominance.21 In about 75%, myxomas originate from the intravenous injection of gadolinium, typically at the core, which appears
left atrium, and in 15% to 20% from the right atrium. They usually to be the result of relatively high vascularity.15,31 However, organizing
develop from the interatrial septum close to the fossa ovalis. Only a thrombus may occur in the tumor, which will be responsible for areas
few myxomas are located in the ventricles.22 Myxomas are generally of low SI on gadolinium imaging.
sporadic, but may occur in multiple and unusual locations, usually as
part of the autosomal dominant syndrome known as Carney complex, Lipoma
which is present in about 7% of cases, and also includes lentiginosis Cardiac lipomas (Fig. 38.2) are relatively common and account for
and endocrine overactivity.23 Rare aneurysmatic formations of part of about 10% of all primary tumors of the heart.34 True lipomas are encap-
the septum secundum can lead to a gallbladder-like cystic structure sulated, contain neoplastic fat cells, and occur in young age.35,36 About
and can mimic an atrial myxoma by CMR.24 The histologic structure 50% arise subendocardially, 25% subepicardially, and 25% from the
shows a typical matrix of myxoma cells, large blood vessels at the base, myocardium.37 Subepicardial lipomas may become quite large and may
and variable areas of hemorrhage, calcification, or even ossification.25 alter cardiac function, resulting in dyspnea or fatigue,38 and involvement
They are generally polypoid, often pedunculated, round or oval with of the coronary arteries has also been reported.39 Endocardial lipomas
a smooth surface, often covered with thrombi, and are between 1 and commonly arise from the interatrial septum and are located in the right
15 cm in diameter.26 Clinical symptoms appear as a consequence of atrium. Arrhythmias due to myocardial infiltration have been reported.40
embolism or intracardiac obstruction and are determined by size, loca- Lipomatous hypertrophy of the atrial septum (Fig. 38.3) is histologically
tion, and mobility of the myxoma.27,28 characterized by infiltration of lipomatous cells between atrial muscle
On CMR, myxomas (Fig. 38.1) are mainly diagnosed by the typically fibers. Unlike true lipomas they are unencapsulated and contain lipo-
pedunculated, jellylike, and prolapsing appearance and certain signal blasts as well as mature fat cells.41 This condition has been described
characteristics.29 Therefore cine display should be obtained to show the in older, overweight patients who frequently have atrial fibrillation.42
mobility of the tumor.30 Due to the endocardial origin, myxomas are On CMR, lipomas are characterized by bright signal on T1-weighted
characterized by an intermediate, but variable signal on SE images, images and a slight decrease in signal on T2-weighted images similar
similar to that of the myocardium. On SSFP images, myxomas tend to to subcutaneous fat.43 Injection of gadolinium is not needed because
442 SECTION III  Functional Disease

A B

FIG. 38.1  Left atrial myxoma. Four-chamber


(A) and atrial short-axis (B) steady-state free-
precession images showing a left atrial mass
attached to the fossa ovalis (arrows). On late
gadolinium imaging (C and D), there is typical C D
heterogeneous enhancement within the mass.

RV
LV

A B

FIG. 38.2  Atrial lipoma. (A) T1-weighted turbo


spin echo imaging in a horizontal long-axis plane
without fat suppression showing a large intra-
cardiac mass crossing the interatrial septum
(arrow). Note the bright appearance of subcu-
taneous fat (arrowhead). (B) Imaging in the
same plane with the addition of fat suppression.
Note the reduced chest wall signal intensity
(arrowhead). (C) Surgical removal of the mass
confirmed the diagnosis of lipoma by histopa-
thology. (D) Hematoxylin and eosin stain. LV, C D
Left ventricle; RV, right ventricle.
CHAPTER 38  Cardiac and Paracardiac Masses 443

A B C
FIG. 38.3  Lipomatous hypertrophy of the interatrial septum. Steady-state free precession image (A) showing
fatty deposition within the interatrial septum. Note the characteristic bilobed or hourglass-shaped appearance,
typically sparing the fossa ovalis region (white arrows). The diagnosis of fatty deposition can be confirmed
as demonstrating an area of high signal in the interatrial septum on T1-weighted turbo spin echo images
(B) with a signal drop-out after fat suppression (C). Note the prominent fat deposition around the heart and
chest wall (black arrows).

the SI will remain unchanged. A drop in SI using the fat presaturation fibromas, which are usually solitary. In about 50% of patients, tuberous
technique verifies the diagnosis. In lipomatous hypertrophy a bilobed sclerosis can be diagnosed.52 Rhabdomyomas have typically a solid and
atrial septum thickening with an SI comparable to subcutaneous fat homogeneous appearance, are often isointense to the myocardium on
on T1- and T2-weighted images can be visualized. In contrast to the T1-weighted images, and slightly hyperintense on T2-weighted images.53
benign lipomatous hypertrophy, the very rare liposarcoma usually shows These masses typically enhance after gadolinium injection (Fig. 38.6).
infiltration, inhomogeneities, and fast tumor growth.44
Hemangioma
Papillary Fibroelastoma There are only a few cases reported of arteriovenous hemangiomas of
Papillary fibroelastomas are the commonest tumors of cardiac valves. the interventricular septum of the heart. The location of the tumor is
These papillary structures consist of elastin and collagen layers covered predominantly at the right or left ventricular free walls, but involve-
by endothelium, and predominantly involve the left side of the heart. ment of the septum and multiple locations have been reported.54 The
The aortic valve is most frequently involved, followed by the mitral valve distinction between a hemangioma and a vascular malformation can
and left ventricular outflow tract.45 Recognition is important, because be difficult.55 On CMR, hemangiomas are characterized as regions
neurologic events caused by embolization are common complications. of increased SI on T1- and T2-weighted images compared with the
Because fibroelastomas are relatively small (usually <10 mm) and mobile myocardium because of slow flowing blood. They characteristically
structures, echocardiography is the preferred imaging modality. On show rapid filling during first-pass perfusion, sometimes with higher
CMR, they usually present as regular and round lesions attached to the SI relative to the adjacent myocardium, owing to the vascular nature
cardiac valves or endothelium. They have low signal on GRE, appear of the tumor. In large lesions, filling can be initially appreciated in the
isointense on T1-weighted images and hyperintense on T2-weighted periphery, with subsequent centripetal filling of core. The enhancement
images.46 Despite being relatively avascular, enhancement is usually seen often persists in the latter stages after intravenous gadolinium injection
with late gadolinium imaging (Fig. 38.4).47 (Fig. 38.7).56

Fibroma Leiomyomatosis With Intracardiac Extension


Fibromas are congenital tumors often discovered at young adulthood.48 Intravenous leiomyomatosis is a rare pathologic entity, and all tumors
They are located intramyocardially and frequently within the ventricu- have been observed in females, most of whom were white and pre-
lar septum. The left ventricle is more often involved than the right menopausal.57 The tumor arises either from a uterine myoma or from
ventricle. On T1-weighted SE images, fibromas tend to be isointense the wall of the vessel.58 The tumor generally appears as a large mobile
compared with skeletal muscle. Because of the short T2 relaxation time mass in the right atrium (Fig. 38.8). Because the preoperative evaluation
of fibrous tissue, fibromas show a decrease in SI relative to the myocar- should include assessment of all cardiac chambers and the region of
dium on T2-weighted SE images.49,50 These masses have low signal on the inferior vena cava, CMR can be considered to be a primary diag-
perfusion images and early after gadolinium injection because of low nostic method.59 As a result of the myomatous tissue, SI characteristics
tumor vascularity. However, late after gadolinium injection, they typically are similar to that of muscle.
show significant enhancement (Fig. 38.5), presumably because of the
increased collagen and elastic tissue surrounding the fibroblastic-type Other Benign Tumors
tumor cells.23 Surgical excision is recommended even in asymptomatic There are a large number of unusual conditions affecting the heart
patients,51 attributed to the potential risk of sudden death caused by causing tumors. These may be degenerative, such as mitral annular
arrhythmias. calcification, infective, such as hydatid cysts (Fig. 38.9), or inflamma-
tory (Behçet syndrome mimicking myxoma), among other rare causes.
Rhabdomyoma Mitral annular calcification (MAC) is a chronic noninflammatory
Rhabdomyomas are also congenital in origin and the commonest heart process characterized by calcium deposition in the annular fibrous ring
tumors diagnosed in fetuses and infants.23 They characteristically arise of the mitral valve.60 It usually occurs at an older age and is linked to
from the ventricular myocardium at multiple locations as opposed to chronic renal disease and abnormalities of calcium phosphate metabolism.
A B C

D E F
FIG. 38.4  Papillary fibroelastoma of the aortic valve. Steady-state free precession image (A) demonstrating a solid,
intraluminal mass (arrow) attached to the commissure of the left coronary and noncoronary aortic valve leaflet.
T1-weighted (B) and T2-weighted (C) spin echo images showing a solid lesion (arrows) with intermediate signal
intensity. The macroscopic specimen (D) appeared as a translucent, gelatinous mass. The histologic specimen
in the hematoxylin and eosin stain shows a benign lesion with multiple papillary fronds (E). The papillary fronds
consist of three layers, comprising a collagenous core with low elastin content, an amorphous intermediate
layer, and a delicate coat of single-layer endothelium (F), as demonstrated with the elastica–van Gieson stain.

A C E

B D F
FIG. 38.5  Fibroma of the lateral wall of the left ventricle. Steady-state free precession four-chamber (A) and
short-axis (B) images showing a slightly hypointense mass in the lateral wall of the left ventricle (arrows). First-
pass perfusion image in the short-axis plane (C) showing hypoperfusion of the mass surrounded by the normally
perfused myocardium, which is suggestive of low tumor vascularity (arrow). Late myocardial enhancement image
(D) demonstrating high signal intensity of the mass (arrow) compared with the nulled normal myocardium. Gross
pathology of the resected left ventricular mass (E). The cut section (F) showed an off-white whorled surface with
foci of calcification. The lesion extended to the inked surgical margin. There was no hemorrhage or necrosis.
CHAPTER 38  Cardiac and Paracardiac Masses 445

LV

RV LV

A B

FIG. 38.6  Rhabdomyoma of the interventricular


septum. Short-axis image from a steady-state free
precession (SSFP) image obtained through the
proximal septum (A) showing a markedly thickened
septum (arrow) that appears to be homogeneous
with the rest of the myocardium. SSFP image of the
left ventricular outflow tract (B, arrow). T1-weighted
turbo spin echo image (C) demonstrating that the
C D signal from the markedly thickened septum is isoin-
tense compared with the rest of the myocardium.
T2-weighted image with fat suppression (D) dem-
onstrating high signal within the thickened septum,
suggesting a different structure from the surrounding
myocardium. Early after gadolinium injection (E),
there is enhancement of the left ventricular and
right ventricular cavities, as well as the myocardium,
including the thickened septum. Late after gado-
linium injection (F), the normal myocardium appears
black. There is marked abnormal late enhancement
throughout the entire segment of the thickened
E F
septum. LV, Left ventricle; RV, right ventricle.

A E

B C D G
FIG. 38.7  Hemangioma of the right ventricle. Four-chamber steady-state free precession image (A) demon-
strates a hypointense, spherical, and pedunculated mass within the mid right ventricle, which is attached to
the free wall of the right ventricle via a short stalk (arrow). Short-axis perfusion imaging demonstrates first-
pass contrast arrival in the right ventricle (B), and peripheral contrast enhancement of the mass (C, arrow),
with subsequent complete filling of the tumor on the second-pass arrival of the contrast in the right ventricle
(D, arrow), which indicate vascularity of the lesion. Short-axis view of a 10-minute late gadolinium enhance-
ment (E) demonstrates peripheral enhancement of the right ventricular (RV) mass (arrow). Presence of this
finding favors the diagnosis of cardiac hemangioma. Gross photograph of the resected tumor specimen (F)
exhibits a polypoid, tan, gelatinous, and focally hemorrhagic lesion with a small stalk (arrow). High-power
photomicrograph (G) demonstrates a marked proliferation of capillary-sized blood vessels (short black arrows)
within a slightly blue acid mucopolysaccharide ground substance. Features are consistent with capillary-type
cardiac hemangioma. LV, Left ventricle.
446 SECTION III  Functional Disease

A B

FIG. 38.8  Intravenous leiomyomatosis. Atrial


short-axis (A), right ventricular two-chamber
(B), and right ventricular outflow tract (C) steady-
state free precession showing transvenous
extension of tumor from the inferior vena cava
to the right atrium and right ventricle (arrows).
The mass enhances late after gadolinium injec- C D
tion (D, arrow).

A B

C D
FIG. 38.9  Hydatid cyst. Two-chamber (A) and short-axis (B) steady-state free precession images showing
a large multilocular cystic mass (arrows) involving the inferior wall and producing severe thinning of adjacent
myocardium. Macroscopic image (C) of the excised hydatid material. Microscopic image of Echinococcus
granulosus (D).
CHAPTER 38  Cardiac and Paracardiac Masses 447

It presents as an immobile mass in the basal posterior mitral valve and are the presence of necrosis, calcification, a high degree of vascularity,
adjacent ventricular myocardium, although in more severe cases it can infiltration of the adjacent tissues, inhomogeneous appearance, and
extend circumferentially to involve the whole annulus. It is character- peritumorous edema.67,68
ized by a low signal on GRE, T1- and T2-weighted sequences. After
intravenous gadolinium administration, there is no enhancement at the Sarcoma
core as opposed to a peripheral rim of circumferential enhancement, Sarcomas of the heart or the great vessels comprise the largest group,
in keeping with a calcified core surrounded by a fibrotic envelope.60 accounting for 75% of all primary malignant cardiac tumors in adults.69
Caseous calcification of the mitral valve is a rare variant of MAC, in There are various histologic types of sarcoma, such as angiosarcomas,
which the mass undergoes liquefaction necrosis. The core is a mixture leiomyosarcomas, fibrosarcomas, synovial sarcomas, rhabdomyosarcomas,
of fatty acids, cholesterol, and amorphous eosinophilic material encased undifferentiated pleomorphic sarcomas, and liposarcomas.20,64,70 Angio-
by a rim of fibrosis, calcification, and inflammatory cell infiltration. As sarcoma (Fig. 38.10) is the most frequent primary malignant cardiac
a result, the fatty proteinaceous material in the core can generate high tumor.20 It is located within the right atrium near the atrioventricular
signal on T1-weighted images (and on T2-weighted images as well), as groove in 80% of cases.71 They appear preferentially between the third
opposed to the low signal in MAC.61 Another differentiating sign will be and fifth decade of life, with a male-to-female ratio of about 2 : 1.72 In
the central enhancement in both early and late phases after gadolinium contrast, other types of sarcoma often occur in the left side of the heart,
injection.62 These entities are believed to be benign because they are where they are often clinically mistaken for myxoma. Morphologically,
usually asymptomatic and discovered as incidental findings. However, angiosarcomas are usually hemorrhagic, often with poorly defined
complications such as mitral stenosis or regurgitation, embolization, borders. They frequently invade contiguous structures such as the venae
and infective endocarditis have been described, all of which may prompt cavae and tricuspid valve, and are characterized by irregular anastomos-
surgical intervention. ing sinusoidal structures with papillary intraluminal tufting.72 On CMR,
angiosarcomas have a polymorphic appearance, with a central region
of hyperintensity consistent with necrosis, and moderate SI in peripheral
MALIGNANT TUMORS OF THE HEART regions in T1- and T2-weighted images.32 Because of the high degree
Nearly 25% of all primary cardiac tumors are malignant tumors. Metas- of vascularity, signal enhancement is seen after intravenous injection
tases are 20 to 40 times more common than primary malignant tumors of gadolinium. The typical appearance of angiosarcoma on CMR is a
and appear in 6% of postmortems in malignant diseases. The most “cauliflower” aspect on black-blood images and an avid enhancement
common primary malignant cardiac tumors are various types of sar- with “sunray” aspect after administration of contrast agent.73
comas and lymphomas.63,64 Primary leiomyosarcomas are rare, highly aggressive, locally invasive
Because of the small number of studied cardiac malignancies, and tumors with a frequency of 0.25%. They arise in 75% from the inferior
the differences in tumor age and vascularity, and a widespread vari- vena cava, but also have been reported with an origin of the superior
ability in water content, a reliable tissue differentiation of cardiac malig- vena cava or in the pulmonary veins.70,74 This neoplasm demonstrates
nancies is often not possible.65,66 Distinct features of malignant tumors a SI on T1-weighted SE images slightly higher than liver parenchyma,

A B C D

E F G H
FIG. 38.10  Comprehensive evaluation of a right atrial mass. Four-chamber (A) and atrial short-axis (B and
C) steady-state free precession images showing a large mass in the right atrial free wall invading the right
atrial cavity (arrows). The tumor has intermediate signal on T1-weighted (D) and high signal on T2-weighted
images (E), which persisted after fat suppression (F). The presence of a large, heterogeneous, and invasive
mass in the right heart associated with pericardial effusion suggests malignancy. There is significant enhance-
ment early after gadolinium injection, suggesting vascularity (G). The enhancement pattern persists in the
late phase (H). A biopsy was performed, and the diagnosis of angiosarcoma was made.
448 SECTION III  Functional Disease

A B

C D E
FIG. 38.11  Left ventricular liposarcoma. Left ventricular outflow tract (A) and short axis at papillary muscle
level (B) steady-state free precession images showing abnormal tissue enlarging the posteromedial papillary
muscle. Some abnormal tissue can also be seen in the mitral valve leaflets (black arrows). On a T1-weighted
turbo spin echo image at the papillary level (C), there is a high signal not only in the posteromedial papil-
lary muscle but also in part of the anterolateral papillary muscle (white arrow). There is signal drop-out on
the same plane after fat suppression (D), suggesting fatty infiltration. Increased signal intensity is seen
in the late phase after gadolinium injection (E). Surgical resection of the tumoral masses was performed,
and the diagnosis of low-grade liposarcoma was made on histology.

but not as bright as the adjacent mediastinal fat. The SI is high on T2.
After injection of gadolinium, a slight contrast enhancement of the Lymphoma
tumor can be detected.70 The advantage of CMR is the ability to assess Primary cardiac lymphomas (Fig. 38.12) are rare, usually non-Hodgkin
intravascular tumor extension into the superior vena cava, the pulmo- in origin, with B-cell lymphoma being the most common type.80 Post-
nary veins, and the heart chambers.75 mortem studies in lymphoma patients have shown cardiac involvement
Liposarcomas (Fig. 38.11) are very rare and not represented in most in up to 25% of diagnoses. As a result, the diagnosis of primary cardiac
surgical series of tumors. Grossly, they are bulky tumors as large as lymphoma is made in the absence of extracardiac involvement. New
10 cm.72 Liposarcomas often have a pericardial origin and CMR is able data suggest that primary cardiac lymphoma is more often diagnosed
to detect this pericardial mass with heterogeneous high signal on premortem, maybe because of the awareness of the higher incidence
T1-weighted images and epicardial infiltration. After injection of gado- of lymphoproliferative disorders related to Epstein-Barr virus (EBV)
linium, liposarcomas may show a slight signal enhancement.76 in patients with AIDS and in patients who have had transplantation.72
Rhabdomyosarcomas are the most common primary cardiac malig- The mean age at presentation for cardiac lymphoma is 38 years, with
nancy in children. They arise from any location in the heart and always a slight predominance in men. Involvement of the right heart is much
involve the myocardium and frequently also the valves.77 Patients often more common than of the left heart. On T1- and T2-weighted SE
present with congestive heart failure. On T1-weighted images, rhabdo- images, lymphomas appear isointense or hypointense to cardiac muscle.
myosarcomas are isointense to the myocardium. After administration After injection of gadolinium, lymphomas appear heterogeneous with
of gadolinium, a homogeneous enhancement will be found, unless the less-enhancing central regions consisting of necrosis.66
tumor has necrotic areas, which will then show little or no enhancement.78
Undifferentiated sarcomas are mostly found in the left atrium (80%)
and have no specific histologic markers.78 CMR shows an irregularly
METASTATIC TUMORS OF THE HEART
shaped mass, with heterogeneous signal on T1-weighted images, high- The incidence of cardiac metastases has been described as between 10%
intermediate signal on T2-weighted images, and heterogeneous enhance- and 14%. Primary tumors that metastasize to the heart can be divided
ment after contrast administration.79 into three categories of incidence: uncommon primary tumors that
CHAPTER 38  Cardiac and Paracardiac Masses 449

On CMR images, fresh thrombi on T1-weighted SE images have


often a higher signal than myocardium, and the contrast is further
accentuated on T2-weighted SE images consistent with a high amount
of hemoglobin.81 However, depending on the age of the thrombus,
other signal intensities are possible. After 1 or 2 weeks, the paramagnetic
effect in the organizing thrombus because of deoxyhemoglobin and
methemoglobin causes T1 and T2 shortening, which may result in
increased SI in T1-weighted images and decreased SI in T2-weighted
images.88 Chronic organized thrombi are of low SI because of loss of
water and protons. A problem concerning differentiation between
thrombus and slow-flowing blood occurs especially in laminated or
immobile thrombi on SE images.89 Compared with thrombus forma-
tion, slow-flowing blood shows increased signal on T2-weighted
images.49,90 On GRE images, thrombi have the lowest SI compared with
other cardiac structures, whereas blood appears brightest.88 If thrombi
contain calcification, they appear more heterogeneous.33 T1 and T2
FIG. 38.12  Involvement of the heart by an aggressive large B-cell lym- mapping sequences have not shown significant diagnostic capability
phoma (arrows). Horizontal long-axis steady-state free precession image compared with postgadolinium sequences, which remain the gold stan-
showing a homogeneous mass involving the lateral aspect of the right dard in thrombus imaging.13 To differentiate thrombus from tumor,
ventricle and extending superiorly to the right atrium. intravenous injection of contrast agents will be helpful. Thrombi usually
do not show signal enhancement after intravenous injection of gado-
linium, unless they are already organized.15 Compared with other diag-
nostic procedures, CMR and computed tomography (CT) offer a high
have a high rate of metastasis to the heart (malignant melanoma, thyroid sensitivity of about 90%, with a slightly better specificity when compared
carcinoma, extracardiac sarcomas, lymphomas, malignant germ cell with 2D echocardiography.89
neoplasm, malignant thymoma, renal cell carcinomas); common tumors
that have an intermediate rate of cardiac metastasis but account for the
greatest number of cardiac metastases (carcinoma of the stomach, liver,
PERICARDIAL LESIONS
ovary, colon, and rectum); and common tumors with rare metastases Magnetic resonance evaluation of pericardial neoplasms in most cases
to the heart.72 The most common epithelial malignancies to metastasize involves identification of abnormal anatomic structures and boundaries
to the heart are carcinomas of the breast and of the lung.71 The tumor rather than characterization of relative tissue intensities. A few excep-
burden in the heart is the highest with melanoma as compared with tions to this are included in the differential diagnosis of mediastinal
any other malignancy. There are four ways noncardiac tumors may masses, such as fibroma, lipoma, and pericardial cysts. The value of
invade the heart: (1) direct mediastinal infiltration of heart tissue in CMR for evaluation of potential neoplasm lies largely in treatment
the case of lung cancer (see Fig. 38.9), breast cancer, or mediastinal planning, particularly preoperative assessment. The loss of normal
lymphomas; (2) hematogenous spread by systemic tumors such as anatomic boundaries is an important sign of neoplasm. Neoplastic
malignant melanoma, lymphoma, leukemia, and sarcoma; (3) transve- involvement of the pericardium results in focal or diffuse obliteration
nous spreading from the inferior vena cava in the case of renal or of the normal pericardial signal. In the case of malignancy adjacent to
hepatic tumors and transvenous spreading from the pulmonary vein cardiac structures, visualization of the pericardial line is an indication
in the case of lung cancer; and (4) lymphatic spread.20,38 Metastatic that pericardial invasion has not occurred.
cardiac involvement can be localized or diffuse. Nodular formations
can arise at discrete locations in the myocardium or diffusely encased Tumors
in the epicardial surface. In most cases there is pericardial involvement, Of the patients with primary cardiac tumors, only 7% to 13% arise
with superficial myocardial infiltration. In metastatic melanoma, the primarily from the pericardium.91 The most common benign tumors
myocardium is involved in virtually every patient, with less frequent of the pericardium are lipomas, teratomas, fibromas, hemangiomas,
infiltration of epicardium and endocardium.71 and neuromas. Lipomas and hemangiomas are often incidental findings
without symptoms. On T1-weighted images, lipomas show high SI,
whereas on T2-weighted images, a decrease in SI similar to subcutane-
CARDIAC THROMBUS ous fat is characteristic. Lipomas do not enhance after administration
Intracardiac thrombus formation is often located in the left atrium in of contrast agent. Hemangiomas are characteristically hyperintense
the case of dilatation and chronic atrial fibrillation, or in the left ven- during and after administration of contrast agent because of their sig-
tricle in the case of contractile dysfunction seen in cardiomyopathy nificant vascular content.92 Fibroma is a typical pediatric tumor, but it
(Fig. 38.13) or after myocardial infarction.81 Tumor thrombus in the might also be found in adults. Low SI on T2-weighted images and
inferior vena cava and the right atrium can be seen in renal carcinomas hypoisointense SI on T1-weighted images are typical findings for fibro-
and can mimic a solid cardiac mass.82,83 The diagnosis of cardiac throm- mas. Late gadolinium enhancement (LGE) imaging often reveals no or
bus is clinically important to identify because patients are at risk of very little enhancement.91,92 The rare primary malignancies of the peri-
systemic or pulmonary embolization.84,85 However, in most cases the cardium include mesotheliomas, bronchial pheochromocytomas, lym-
diagnosis of cardiac thrombi is coincidental and patients are asymp- phomas, thymomas, fibrosarcomas, and angiosarcomas.93,94 Hemorrhagic
tomatic. Despite the fact that 2D echocardiography is the method of effusions result from erosion into the intrapericardiac vessels or myo-
choice for diagnosis, false-positive rates as high as 28% in detection of cardial wall, with development of acute or subacute tamponade.
left ventricular thrombi and 59% in detection of left atrial thrombi Mesothelioma is the most common primary malignant neoplasm
have been reported.86,87 of the pericardium,95 originating from the mesothelial cells of parietal
450 SECTION III  Functional Disease

A B

C D
FIG. 38.13  Four-chamber (A) and two-chamber (B) steady-state free precession images showing two relative
hypointense regular masses adjacent to the apical and anterior lateral walls (arrows). Spoiled gradient echo
images with a long inversion time (440 ms) shortly after the administration of gadolinium (C and D). Note
the intense contrast between the strongly enhanced blood pool (bright), the myocardium, and the above-
described abnormal structures in the left ventricular cavity that return no signal (dark). These findings are
consistent with intraventricular thrombi.

pericardium. On CMR studies, mesothelioma presents isointense to proportion to the amount of tumor present, and tamponade is a frequent
myocardium on T1-weighted images and shows a heterogeneous appear- complication.
ance on T2-weighted images. Mesotheliomas have a very poor prognosis
and so do any subtypes of sarcoma as well as lymphoma. Angiosarcoma Cysts
and lymphoma frequently present with large (hemorrhagic) pericardial Pericardial cysts (Fig. 38.14) are uncommon lesions, with an incidence
effusion, as described above. of 1 in 100,000 and account for approximately 7% of mediastinal masses.
They are commonly located in the right cardiophrenic angle (51%–70%)
Metastasis and left cardiophrenic angle (28%–38%).99–101 Pericardial cysts are mostly
Direct tumor invasion or metastasis of the pericardium is much more congenital, with the cyst wall being lined with a single layer of meso-
common than primary pericardial malignancies.96 The involvement of thelial cells. They are usually filled with clear fluid. Patients are generally
the pericardium by metastatic disease at autopsy is much higher than asymptomatic, and the lesion is often discovered on a routine chest
clinically suspected, ranging from 1.5% to 22% in incidence.97 Although film. The appearance is typically stable over a long time. On CMR,
often not detected during the lifetime of a patient, metastatic pericar- pericardial cysts appear as paracardiac masses with long T1 and T2
dial involvement is associated with poor outcomes. It can occur by values and flow void, indicating fluid-filled structures. They have low
direct spread, hematogenous or lymphatic dissemination. Of malignant SI on T1-weighted images and high SI on T2-weighted images.102,103
pericardial disease, 80% is associated with lung or breast carcinoma, After injection of gadolinium, there is usually no fluid enhancement,
leukemia, and lymphoma.98 Typical CMR findings include pericardial but intracystic septae may be observed. In addition, a line of low SI,
effusion, nodular pericardial thickening, and pericardial masses. Focal representing the pericardial layer, can be often visualized. The significant
or diffuse plaque-like thickening of the pericardium may occur with advantage of CMR is its ability to differentiate these lesions from other
signal enhancement after injection of gadolinium. Metastatic involve- mediastinal masses and avoid explorative surgery to determine the
ment of the pericardium is characterized by large effusions out of diagnosis because surgery is not necessary in most cases.104
CHAPTER 38  Cardiac and Paracardiac Masses 451

A B

C D E
FIG. 38.14  Multimodality imaging in the evaluation of a mediastinal mass. Four-chamber (A) and coronal
(B) steady-state free precession images showing a large homogeneous and well-defined mass in the right
cardiophrenic angle (arrows). High signal structures on steady-state free precession images either repre-
sent fat or fluid. The mass returns relatively low signal on T1 (C) and relatively high signal on T2-weighted
(D) images, which is more evident after fat suppression (E). Therefore the diagnosis of a fluid-filled pericardial
cyst was made.

APPROACH TO CARDIOVASCULAR MAGNETIC extracardiac structures. Tissue tagging may be considered to detect
contractile dysfunction because of tumoral infiltration relative to the
RESONANCE ACQUISITION OF CARDIAC MASSES
unaffected contracting myocardium or to assess pericardial involvement
A systematic approach is encouraged for a comprehensive evaluation in masses affecting the epicardium and pericardial space. TSE T1- and
of cardiac masses. Because cardiac masses are complex, direct physician T2-weighted images are indicated for further tissue characterization.
supervision is desirable although the imaging protocols may need to A fat suppression prepulse is used to assess or exclude a fatty component
be adapted to the type of tumor and patient compliance to the study. of the mass. Suppression of signal from a standard T1-weighted SE
The following protocol is therefore suggested. Free-breathing stacks of sequence is virtually diagnostic of a fatty mass (i.e., lipoma or liposar-
images in the three orthogonal planes should be part of the initial coma). A stack of TSE images will provide complete coverage of a mass
evaluation. Dark-blood SE and bright-blood SSFP sequences are comple- but TSE images of only the long-axis cross-cuts may suffice if there are
mentary and provide a rough idea of the location, size, and extent of time constraints. Flow imaging may be considered if the mass has an
the mass, and may also be useful to detect extracardiac lesions suggesting intravascular component and there is suspicion of significant obstruc-
metastasis. The relatively low spatial resolution and SNR will be offset tion to flow or occlusion of the vessel, which will influence surgical
by the fast free-breathing acquisition, which will be especially suitable management. Perfusion imaging indicates the vascularity of the mass.
for the unstable, uncooperative, or claustrophobic patient. SSFP cine Benign masses usually show little or no enhancement, whereas malignant
sequences render the largest amount of SNR per amount of time, allow- masses tend to show visible enhancement during first-pass perfusion
ing good visualization of the mass throughout the cardiac cycle along as a result of higher vascularity and capillary permeability. Hemangiomas
excellent delineation of the contours. An axial stack of cines covering are the exception because they will show significant first-pass perfusion
the mass provides a full 3D impression of the location and extension owing to the high vascularity. When the mass is intravascular, contrast-
of the mass in relation to the adjacent anatomic structures throughout enhanced magnetic resonance angiography (CE-MRA) may be considered
the cardiac cycle. This can be complemented by one or two volume instead of perfusion imaging because the high spatial resolution images
stacks in coronal and sagittal planes, which may be slightly rotated and can give valuable information about the relationship of the mass with
aligned with the axes of the heart for a more detailed assessment. Para- the surrounding vessels and respective patency. After contrast admin-
coronal or parasagittal stacks of cines aligned with the atrial, ventricular, istration, image acquisition in the early phase (1–3 minutes after injec-
or valve planes are ideal to assess the relationship of the mass with the tion) is an alternative way to assess the vascularity of the mass. LGE
cardiac structures and respective functional impact. If not already per- (>10 minutes after injection) may appear in both benign and malignant
formed, dedicated cross-cut cine images along the long-axes of the mass lesions with variable SI and may reflect a combination of different
will allow for optimal information on the size and shape of the mass factors such as vascularity and increased interstitial space suggestive of
and can determine signs of invasion of the adjacent intracardiac or necrosis, fibrosis, and infiltration.
452 SECTION III  Functional Disease

APPROACH TO CARDIOVASCULAR MAGNETIC right-sided cardiac location, tissue inhomogeneity, ill-defined borders,
RESONANCE INTERPRETATION OF tumor displacement or infiltration of adjacent structures, pleural/
pericardial effusion, positive first-pass perfusion and LGE are all suspi-
CARDIAC MASSES
cious for malignancy, whereas the absence of these signs will suggest a
When interpreting a study for a suspected cardiac mass, the first step is benign mass. The following algorithm is suggested:
to confirm the presence of the mass and exclude potential confounders 1. Exclusion of the most common benign cardiac tumors by their
such as aneurysmal interatrial septum, hiatus hernia, aortic aneurysm, or typical and often pathognomonic appearance. Typical features can be
prominent pericardial fat, all of which can mimic and be misinterpreted observed in myxoma (septal atrial origin, hypointense SI, moderate
as a cardiac tumor. When single or multiple masses are confirmed, the contrast enhancement, no effusion or infiltration), fibromas (left-
next step is to provide basic morphologic details such as location, size, sided, homogenous, hypointense SI, no contrast enhancement, infil-
and extension of the mass. It is also worth describing whether the mass tration in up to 50%), lipomas (hyperintense SI on T1-weighted SE
is homogeneous or heterogeneous, and whether the contours are regular images, hypointense in fat-suppressed images), and thrombi (typically
and well-defined or irregular and ill-defined. A third step is to detail in enlarged left atria in atrial fibrillation or left ventricular aneurysms,
the relationship of the mass with adjacent structures, and describe if hypointense SI on GRE and SSFP, no contrast enhancement).
there are signs of compression, erosion, displacement, or invasion. Using 2. Features specific of malignant tumors such as an intracardiac lesion
cine and flow imaging, mobility and influence on cardiac contractil- with pericardial effusion (especially with high SI on T1-weighted
ity and valvular function can be assessed. For intravascular masses, it images suggesting a high proteinaceous or hemorrhagic component),
should be described whether there is partial or complete obstruction a paracardiac lesion with inhomogeneous tissue composition or a
to flow, collateralization, or diversion of flow to other vascular systems mass that infiltrates adjacent tissue but is not an inflammatory
(such as azygous dilatation in obstruction of the vena cavae). The fifth pseudotumor.
step is tissue characterization, with description of the SI relative to the 3. With the exception of a myxoma, lesions of the right heart are always
myocardium on different GRE and TSE sequences. When gadolinium suspicious for malignancy/metastasis.105
is administrated, presence of first-pass perfusion and degree of gado- In summary, CMR has proven to be a valuable tool in differentiating
linium enhancement in earlier and later stages should be documented. benign from malignant tumors of the heart and neoplasms from pseu-
Morphologic CMR features can predict the type of cardiac and dotumors and should therefore be used for the assessment of cardiac
paracardiac masses (Table 38.2). Large tumoral size (usually >5 cm), masses.105–108

TABLE 38.2  Magnetic Resonance Imaging Features of the Most Common Cardiac Tumors
Enhancement After
Administration of
T1-Weighted SE T2-Weighted SE GRE Gd-DTPA
Myxoma Intermediate varying SI, calcified Low SI, especially in iron- Very low SI compared Hyperintense
areas: hypointense and containing myxomas with the surrounding
hemorrhage: increased SI blood pool
Lipoma/lipomatous Brightest SI similar to subcutaneous Intermediate SI parallel to Nonspecific Nonspecific
hypertrophy fat, using fat presaturation subcutaneous fat
technique: reduced SI
Papillary fibroelastoma Intermediate SI Intermediate to high SI Low SI compared with the Hyperintense
surrounding blood pool
Fibroma Intermediate to slightly hyperintense Decrease in SI compared with TI Nonspecific Slight and heterogeneous
SI compared with myocardium,
when calcification (hypointense)
and hemorrhage (hyperintense)
are present: heterogeneous
Rhabdomyoma Homogeneous, slightly lower SI Strong increased SI Very low compared with Nonspecific
than myocardium myocardium
Hemangioma Intermediate SI Increased SI (because of slow- Nonspecific Significant increasing SI,
flowing blood in the tumor heterogeneous
vessels), higher than myocardium
Intravenous Similar to myocardium Similar to myocardium Nonspecific Nonspecific
leiomyomatosis
Pericardial cysts Lowest SI, flow void Highest SI Nonspecific Signal enhancement,
(simple fluid) Low SI, but higher than in normal High SI visualization of
(proteinaceous fluid) fluid, no flow void intracystic septae
Angiosarcoma Central hyperintense spot (blood No change Nonspecific Strong
vessels, hemorrhage, or necrosis),
surrounded by intermediate SI
regions
CHAPTER 38  Cardiac and Paracardiac Masses 453

TABLE 38.2  Magnetic Resonance Imaging Features of the Most Common Cardiac
Tumors—Cont’d
Enhancement After
Administration of
T1-Weighted SE T2-Weighted SE GRE Gd-DTPA
Lymphoma Isointense to hypointense to cardiac Isointense to myocardium Nonspecific Heterogeneous with less-
muscle enhancing central regions
Liposarcoma Bright SI equal to subcutaneous fat, Not published Not published Not published
but heterogeneous: decrease in
SI, when fat presaturation is used
Leiomyosarcoma High SI, slightly higher than liver Not published Not published Not published
parenchyma, but not as high as
subcutaneous fat homogeneous,
commonly connected with
proteinaceous pericardial effusion
Thrombus Intermediate, often slightly higher SI Surrounding slow-flowing blood Thrombus has the No signal enhancement,
than myocardium, slightly higher becomes higher SI than lowest SI unless the thrombus is
than blood thrombus, contrast between organized
thrombus and myocardium is
further accentuated
Fresh High SI (oxyhemoglobin) Decreased SI
Chronic (older than Higher SI (deoxyhemoglobin)
2 weeks)

Gd, Gadolinium; GRE, gradient recalled echo; SE, spin echo; SI, signal intensity.
Modified from Hoffman U, Globits S, Frank H. Cardiac and paracardiac masses: current opinion on diagnostic evaluation by magnetic resonance
imaging. Eur Heart J. 1998;19:553–563.

by assessing extracardiac components of a mass, such as mediastinal


PROGNOSIS OF CARDIAC TUMORS
involvement and extension into large pulmonary vessels. CMR allows
Prognosis after surgery is usually excellent in the case of benign tumors. the exclusion of conditions that can mimic cardiac tumors. The findings
Local recurrence of myxoma is uncommon but could be related to will also be helpful in characterizing paracardiac masses and in guiding
inadequate resection, multicentricity, origin in a chamber other than therapeutic strategies. Comprehensive CMR evaluation of cardiac mass
the left atrium or familial tumors. Nearly all malignant cardiac tumors morphology, tissue composition, and perfusion in conjunction with
are rapidly fatal. Sarcomas of the heart have a better prognosis if they information on tumor spread and operability allows diagnosis of patients
arise in the left atrium, have no necrotic regions, and have not metas- with cardiac mass.107,111 As compared with other image modalities, mul-
tasized at the time of diagnosis.72 Achievement of remission is unusual tiparametric CMR provides a better global cardiac assessment, including
and can be achieved with chemotherapy in lymphomas.109 tumor tissue characterization, which has been proven to be likely pre-
dictive of the pathologic diagnosis.106,112 Nevertheless, histologic con-
firmation is required in most cases, especially when there is a suspicion
CONCLUSION of malignancy.
CMR techniques have contributed significantly to the ability to detect
cardiac and paracardiac masses and play an important role in the diag-
nostic evaluation, which is complementary to echocardiography.110
REFERENCES
Because of its larger field of view, CMR adds diagnostic information A full reference list is available online at ExpertConsult.com
CHAPTER 38  Cardiac and Paracardiac Masses 453.e1

REFERENCES 26. Hall RJ, Cooley DA, Mac Allister HR Jr, Frazier OH. Neoplastic heart
disease. In: Hurst JW, ed. The Heart, Arteries and Veins. 7th ed. New
1. Lanzer P, Barta C, Botvinick EH, Wiesendanger HU, Modin G, Higgins York: McGraw-Hill; 1990:1382–1403.
CB. ECG-synchronized cardiac MR imaging. Method and evaluation. 27. St John Sutton MG, Mercier L-A, Giuliani ER, Lie JT. Atrial myxomas. A
Radiology. 1985;155:681–686. review of clinical experience in 40 cases. Mayo Clin Proc.
2. Wang Y, Rossman PJ, Grimm RC, Riederer SJ, Ehman RL. Navigator- 1980;55:371–376.
echo-based real time respiratory gating and triggering for reduction of 28. Peters MN, Hall RJ, Cooley DA, Leachmann RD, Garcia E. The clinical
respiration effects in three-dimensional coronary MR angiography. syndrome of atrial myxoma. JAMA. 1974;230:695–701.
Radiology. 1996;198(1):55–60. 29. Lund JT, Ehman RL, Julsrud PR, Sinak LJ, Tajik AJ. Cardiac masses.
3. Crooks LE, Barker B, Chang H, et al. Magnetic resonance imaging Assessment by MR. AJR Am J Roentgenol. 1989;152:469–473.
strategies for heart studies. Radiology. 1984;153:459–465. 30. Go RT, O’Donnell JK, Underwood DA, et al. Comparison of gated
4. Edelstein WA, Bottomley PA, Hart HR, Smith LS. Signal, noise, and cardiac MR and 2D echo of intracardiac neoplasms. AJR Am J
contrast in nuclear magnetic resonance imaging. J Comput Assist Roentgenol. 1985;145:21–25.
Tomogr. 1983;7:391–401. 31. Semelka RC, Shoenut JP, Wilson ME, Pellech AE, Patton JN. Cardiac
5. Seelos KC, von Smekal A, Vahlensieck M, Gieseke J, Reiser M. Cardiac masses. Signal intensity features on spin-echo, gradient-echo,
abnormalities: assessment with T2-weighted turbo spin-echo MR gadolinium-enhanced-spin-echo, and TurboFLASH images. J Magn
imaging with electrocardiogram gating at 0.5T. Radiology. Reson Imaging. 1992;2:415–420.
1993;189:517–522. 32. Gomes AS, Lois JF, Child JS, Brown K, Batra P. Cardiac tumors and
6. Pettigrew RI. Cardiovascular imaging techniques. In: Stark DD, Bradley thrombus. Evaluation with MR imaging. AJR Am J Roentgenol.
WB, eds. Magnetic Resonance Imaging. St. Louis: Mosby-Year Book; 1987;149:895–899.
1992:1605–1651. 33. Roos A, Weijers E, Duinen S, Wall EE. Calcified right atrial myxoma
7. Sechtem U, Pflugfelder PW, White RD, et al. Cine MR imaging. Potential demonstrated by MR. Chest. 1989;95:478–479.
for the evaluation of cardiovascular function. AJR Am J Roentgenol. 34. Elbardissi AW, Dearani JA, Daly RC, et al. Survival after resection of
1987;148:239–246. primary cardiac tumors: a 48-year experience. Circulation.
8. Scheffler K, Lehnhardt S. Principles and applications of balanced SSFP 2008;118:S7–S15.
techniques. Eur Radiol. 2003;13:2409–2418. 35. Reyes CV, Jablokow VR. Lipomatous hypertrophy of the cardiac
9. Amparo EG, Higgins CB, Farmer D, Gamsu G, McNamara M. Gated interatrial septum. A report of 38 cases and review of the literature. Am
MR of cardiac and paracardiac masses: initial experience. Am Heart J. J Clin Pathol. 1979;5:785–788.
1984;143:1151–1156. 36. Crocker DW. Lipomatous infiltrates of the heart. Arch Pathol Lab Med.
10. Schulthess GK, McMurdo K, Tscholakoff D, de Geer G, Gamsu G, Higgins 1978;102:69–72.
CB. Mediastinal masses. MR imaging. Radiology. 1986;158:289–296. 37. Fine G. Neoplasms of the pericardium and heart. In: Gould SE, ed.
11. Tscholakoff D. MRT in the diagnosis of cardiovascular system and lung. Pathology of the Heart and Blood Vessels. 3rd ed. Springfield, IL: Charles
Acta Med Austriaca. 1986;3:61–65. C. Thomas; 1968:865.
12. Schmidt HC, Tscholakoff D, Hricak H, Higgins CB. MR image contrast 38. Moulton AL, Jaretzki AIII, Bowman OF, Silverstein EF, Bregman D.
and relaxation times of solid tumors in the chest, abdomen, and pelvis. Massive lipoma of the heart. N Y State J Med. 1976;76:1820–
J Comput Assist Tomogr. 1985;9:738–748. 1825.
13. Caspar T, El Ghannudi S, Ohana M, et al. Magnetic resonance 39. Hananouchi GI, Goff WB. Cardiac lipoma: six-year follow-up with MR
evaluation of cardiac thrombi and masses by T1 and T2 mapping: an characteristics, and a review of the literature. Magn Reson Imaging.
observational study. Int J Cardiovasc Imaging. 2016;10:1034–1036. 1990;8:825–828.
14. Carr DH, Brown J, Bydder GM, et al. Gadolinium-DTPA as a contrast 40. Conces DJ, Vix VA, Tarver RD. Diagnosis of a myocardial lipoma by
agent in MR. Initial experience in 20 patients. AJR Am J Roentgenol. using CT. AJR Am J Roentgenol. 1989;153:725–726.
1984;143:215–224. 41. Hutter Am Jr, Page DL. Atrial arrhythmias and lipomatous hypertrophy
15. Weinmann HJ, Lanaido M, Mutzel W. Pharmakokinetics of Gd-DTPA/ of the cardiac interatrial septum. Am Heart J. 1971;82:16–21.
dimeglumine after IV injection. Physiol Chem Phys Med NMR. 42. Kluge WF. Lipomatous hypertrophy of the interatrial septum. Northwest
1984;16:167–172. Med. 1969;68:25–30.
16. Funari M, Fujita N, Peck WW, Higgins CB. Cardiac tumors. Assessment 43. Levine RA, Weyman AE, Dinsmore RE, et al. Noninvasive tissue
with Gd-DTPA enhanced MR imaging. J Comput Assist Tomogr. characterisation. Diagnosis of lipomatous hypertrophy of the atrial
1991;15:953–958. septum by nuclear MR. J Am Coll Cardiol. 1986;7:688–692.
17. Heath D. Pathology of cardiac tumors. Am J Cardiol. 1968;21:315–327. 44. Applegate PM, Tajik AJ, Ehman RL, Julsrud PR, Miller FA. Two-
18. Straus R, Merliiss R. Primary tumor of the heart. Arch Pathol. dimensional echocardiographic and MR observations in massive
1945;39:74–78. lipomatous hypertrophy of the atrial septum. Am J Cardiol.
19. Wold LE, Lie JT. Cardiac myxomas: a clinicopathologic profile. Am J 1987;59:489–491.
Pathol. 1980;101:219–240. 45. Ngaage DL, Mullany CJ, Daly RC, et al. Surgical treatment of cardiac
20. Mc Allister HA Jr, Fenoglio JJ Jr. Tumors of the Cardiovascular System. papillary fibroelastoma: a single center experience with eighty-eight
Atlas of Tumor Pathology. 2nd series. Fascicle 15. Washington, DC: patients. Ann Thorac Surg. 2007;80:1712–1718.
Armed Forces Institute of Pathology; 1978:1–20. 46. de Arenaza DP, Pietrani M, Moon JC, et al. Cardiac fibroelastoma:
21. Reynen K. Cardiac myxomas. Review article. N Engl J Med. 1995; cardiovascular magnetic resonance characteristics. J Cardiovasc Magn
333(24):1610–1617. Reson. 2007;9:621.
22. Bicer A, Turhan H, Cagrici G, Yasar AS, Sasmaz H. Asymptomatic left 47. Jahnke C, Hamdan A, Fleck E, Paetsch I. Tissue characterization of a
ventricular myxoma diagnosed incidentally by transthoracic suspected aortic valve fibroelastoma with cardiac magnetic resonance
echocardiography. Echocardiography. 2005;22:855–856. imaging. Circ Cardiovasc Imaging. 2008;1:87–88.
23. Burke A, Tavora F. The 2015 WHO classification of tumors of the heart 48. Burke AP, Rosado CM, Templeton PA, Virmani R. Cardiac fibroma:
and pericardium. J Thorac Oncol. 2016;11:441–452. clinicopathologic correlates and surgical treatment. J Thorac Cardiovasc
24. Seebacher G, Binder Th, Frank H, Wolner E, Mohl W. Cystic formation Surg. 1994;108:862–870.
of the foramen ovale mimicking a right atrial myxoma. Ann Thorac 49. Winkler M, Higgins CB. Suspected intracardiac masses: evaluation with
Surg. 2006;82:2296–2298. MR imaging. Radiology. 1987;165:117–122.
25. Pritchard RW. Tumors of the heart. Review of the subject and report of 50. Gamsu G, Stark DD, Webb WR, Moore EH, Sheldon PE. MR of benign
one hundred and fifty cases. Arch Pathol. 1951;51:98–128. mediastinal masses. Radiology. 1984;151:709–713.
453.e2 SECTION III  Functional Disease

51. Oliva PB, Breckinridge JC, Johnson ML, Brantigan CO, O’Meara OP. 77. Kassop D, Donovan MS, Cheezum MK, et al. Cardiac masses on cardiac
Left ventricular outflow obstruction produced by a pedunculated CT: a review. Curr Cardiovasc Imaging Rep. 2014;7:9281.
fibroma in a newborn. Clinical, angiographic, echocardiographic and 78. Sparrow PJ, Kurian JB, Jones TR, Sivananthan M. MR imaging of
surgical observations. Chest. 1978;74:590–593. cardiac tumors. Radiographics. 2005;25:5.
52. Abushaban L, Denham B, Duff D. 10 year review of cardiac tumors in 79. Cheong B, Huber S, Reul R, Frazier OH, Nasser MM, Flamm SD.
childhood. Br Heart J. 1993;70:166–169. Cardiovascular magnetic resonance imaging of sarcoma invading the left
53. Hoffmann U, Globits S, Frank H. Cardiac and paracardiac masses. Eur atrium and pericardium. Tex Heart Inst J. 2005;32:608–609.
Heart J. 1998;19:553–563. 80. Petrich A, Cho SI, Billett H. Primary cardiac lymphoma: an analysis of
54. Brizard C, Latremouille C, Jebara VA, et al. Cardiac hemangiomas. Ann presentation, treatment, and outcome patterns. Cancer. 2011;117:
Thorac Surg. 1993;56:390–394. 581–589.
55. Newell IIJD, Eckel C, Davis M, Tadros NB. MR appearance of an 81. Dooms GC, Higgins CB. MR imaging of cardiac thrombi. J Comput
arteriovenous hemangioma of the interventricular septum. Cardiovasc Assist Tomogr. 1986;10:415–420.
Intervent Radiol. 1988;11:319–321. 82. Asensio JMN, Fernandez MRG, Bravo MA, Herrera AM. Kidney tumour
56. Soberman MS, Plauth WH, Winn KJ, et al. Hemangioma of the right mimicking cardiac mass. Int J Cardiol. 2006;106:401–403.
ventricle causing outflow obstruction. J Thorac Cardiovasc Surg. 83. Gomes AS, Lois JF, Child JS, Brown K, Batra P. Cardiac tumors and
1988;96:307–309. thrombus: evaluation with MR imaging. AJR Am J Roentgenol.
57. Spellacy WN, Maire WJ, Buhi WC. Plasma growth hormone and estradiol. 1987;149:895–899.
Levels in woman with uterine myomas. Obstet Gynecol. 1972;40:829–834. 84. Visser CA, Kan G, Meltzer RS, et al. Embolic potential of left ventricular
58. Bassish MS. Mesenchymal tumors of the uterus. Clin Obstet Gynecol. thrombus after myocardial infarction: a two-dimensional
1974;17:51–88. echocardiographic study of 119 patients. J Am Coll Cardiol.
59. Rosenberg JM, Marvasti MA, Obeid A, Johnson LW, Bonaventura M. 1985;5:1276–1280.
Intravenous leiomyomatosis: a rare cause of right sided cardiac 85. Hamby RI, Wisoff BG, Davison ET, et al. Coronary artery disease and
obstruction. Eur J Cardiothorac Surg. 1988;2:58–60. left ventricular mural thrombi: clinical, hemodynamic, and
60. Shah BN, Babu-Narayan S, Li W, Rubens M, Wong T. Severe mitral angiocardiographic aspects. Chest. 1974;66:488–494.
annular calcification: insights from multimodality imaging. Tex Heart 86. Barakos JA, Brown JJ, Higgins CB. MR imaging of secondary cardiac
Inst J. 2014;41:245–247. and paracardiac lesions. AJR Am J Roentgenol. 1989;153:47–50.
61. Motwani M, Fairbairn TA, Jogiya R, Greenwood JP, Plein S. Caseous 87. Feigenbaum H. Coronary artery disease. In: Feigenbaum H, ed.
calcification of the mitral valve complicated by embolization, mitral Echocardiography. 4th ed. Philadelphia: Lea and Ferbiger; 1986:489–494.
regurgitation, and pericardial constriction. Eur Heart J Cardiovasc 88. Jungehülsing M, Sechtem U, Theissen P, Hilger HH, Schicha H. Left
Imaging. 2012;13(9):792. ventricular thrombi. Evaluation with spin-echo and gradient-echo MR
62. Di Bella G, Masci PG, Ganame J, Dymarkowski S, Bogaert J. imaging. Radiology. 1992;182:225–229.
Liquefaction necrosis of mitral annulus calcification. Circulation. 89. Sechtem U, Theissen P, Heindel W, et al. Diagnosis of left ventricular
2008;117:e292–e294. thrombi by MR and comparison with angiocardiography, computed
63. Roberts WC, Glancy DL, DeVita VT. Heart in malignant lymphoma. A tomography and echocardiography. Am J Cardiol. 1989;64:1195–1199.
study of 196 autopsy cases. Am J Cardiol. 1968;22:85–107. 90. Menegus MA, Greenberg MA, Spindola-Franco H, Fayemi A. MR of
64. Silverman J, Olwin JS, Graettinger JS. Cardiac myxomas with systemic suspected atrial tumors. Am Heart J. 1992;123:1260–1268.
embolization: review of the literature and report of a case. Circulation. 91. Restrepo CS, Vargas D, Ocazionez D, Martínez-Jiménez S, Betancourt
1962;26:99–103. Cuellar SL, Gutierrez FR. Primary pericardial tumors. Radiographics.
65. Zeitler E, Kaiser W, Schuierer G, et al. Magnetic resonance imaging of 2013;33:1613–1630.
aneurysms and thrombi. Cardiovasc Intervent Radiol. 1986;8:321–328. 92. Motwani M, Kidambi A, Herzog BA, Uddin A, Greenwood JP, Plein S.
66. Dorsay TA, Ho VB, Rovira MJ, Armstrong MA, Brisette MD. Primary MR imaging of cardiac tumors and masses: a review of methods and
cardiac lymphoma. CT and MR findings. J Comput Assist Tomogr. clinical applications. Radiology. 2013;268:26–43.
1993;17(6):978–981. 93. Stark DD, Higgins CB, Lanzer P, et al. Magnetic resonance imaging of
67. Hendrick RE, Raff U. Image contrast and noise. In: Stark DD, Bradley the pericardium: normal and pathologic findings. Radiology.
WB, eds. Magnetic Resonance Imaging. St. Louis: Mosby-Year Book; 1984;150:469–474.
1992:109–144. 94. Hort W, Braun H. Untersuchungen ueber Groesse, Wandstärke und
68. Tazelaar HD, Locke TJ, McGregor CGA. Pathology of surgically excised mikroskopischen Aufbau des Herzbeutels unter normalen und
primary cardiac tumors. Mayo Clin Proc. 1992;67:957–965. pathologischen Bedingungen. Arch Kreislaufforsch. 1962;38:1–22.
69. Hudzik B, Miszalski-Jamka K, Glowacki J, et al. Malignant tumors of the 95. Bogaert J, Francone M. Cardiovascular magnetic resonance in
heart. Cancer Epidemiol. 2015;39:665–672. pericardial diseases. J Cardiovasc Magn Reson. 2009;11:14.
70. Hoffstetter P, Djavidani B, Feuerbach S, Hofstädter F, Seitz J. Myxoid 96. Grebenc ML, Rosado de Christenson ML, Burke AP, Green CE, Galvin
fibrosarcoma of a pulmonary vein with extension into the left atrium. JR. Primary cardiac and pericardial neoplasms: radiologic-pathologic
AJR Am J Roentgenol. 2006;186:365–367. correlation. Radiographics. 2000;20:1073–1103.
71. Travis WD, Brabilla R, Burke AP, Marx A, Nicholson AG, eds. 97. Mocanda R, Kotler MN, Churchill RJ, et al. Multimodality approach to
Classification of Tumours of the Lung, Pleura, Thymus and Heart. 4th ed. pericardial imaging. Cardiovasc Clin. 1986;17:409.
Lyon, France: International Agency for Research on Cancer; 2015. 98. Theoligides A. Neoplastic cardiac tamponade. Semin Oncol. 1978;5:181.
72. Butany J, Nair V, Naseemuddin A, Nair GM, Catton C, Yau T. Cardiac 99. Stoller JK, Shaw C, Matthay RA. Enlarging, atypically located pericardial
tumours: diagnosis and management. Lancet Oncol. 2005;6:219–228. cyst. Recent experience and literature review. Chest. 1986;89:402–406.
73. Esposito A, De Cobelli F, Ironi G, et al. CMR in the assessment of 100. Fraser RG, Pare JAP. Diagnosis of Disease of the Chest. 2nd ed.
cardiac masses. JACC Cardiovasc Imaging. 2014;7(10):1–5. Philadelphia: WB Saunders; 1977:656.
74. Lupetin AR, Dash N, Beckman I. Leiomyosarcoma of the superior vena 101. Le Roux BT. Pericardial coelomic cysts. Thorax. 1977;14:27–35.
cava. Diagnosis by cardiac gated MR. Cardiovasc Intervent Radiol. 102. Sechtem U, Tscholakoff D, Higgins CB. MR of the abnormal
1986;9:103–105. pericardium. AJR Am J Roentgenol. 1986;147:245–252.
75. Somers K, Lotte F. Primary lymphosarcoma of the heart. Review of the 103. Verhaert D, Gabriel RS, Johnston D, Lytle BW, Desai MY, Klein AL. The
literature and report of 3 cases. Cancer. 1960;3:449–457. role of multimodality imaging in the management of pericardial disease.
76. Garrigue S, Robert F, Roudaut R, Bonnet J. Assessment of non-invasive Circ Cardiovasc Imaging. 2010;3:333–343.
imaging techniques in the diagnosis of heart liposarcoma. Eur Heart J. 104. Roberts WC, Spray TL. Pericardial heart disease: a study of its causes,
1995;16:139–141. consequences and morphologic features. Cardiovasc Clin. 1976;7:11–65.
CHAPTER 38  Cardiac and Paracardiac Masses 453.e3

105. Hoffmann U, Globits S, Schima W, et al. Usefulness of magnetic 109. Morillas P, Quiles J, Nunez D, et al. Complete regression of cardiac
resonance imaging of cardiac and paracardiac masses. Am J Cardiol. non-Hodgkin lymphoma. Int J Cardiol. 2006;106:426–427.
2003;92:890–895. 110. Patel R, Lim RP, Saric M, et al. Diagnostic performance of cardiac
106. Zhu D, Yin S, Cheng W, et al. Cardiac MRI-based multi-modality magnetic resonance imaging and echocardiography in evaluation of
imaging in clinical decision making: preliminary assessment of a cardiac and paracardiac masses. Am J Cardiol. 2016;117:135–140.
management algorithm for patients with suspected cardiac mass. Int J 111. Beroukhim RS, Prakash A, Buechel ER, et al. Characterization of cardiac
Cardiol. 2016;203:474–481. tumors in children by cardiovascular magnetic resonance imaging: a
107. Fussen S, De Boeck BW, Zellweger MJ, Bremerich J, Goetschalk K, Buser multicenter experience. J Am Coll Cardiol. 2011;58:1044–1054.
PT. Cardiovascular magnetic resonance imaging for diagnosis and 112. Buckley O, Madan R, Kwong R, et al. Cardiac masses, part 2: key
clinical management of suspected cardiac masses and tumours. Eur imaging features for diagnosis and surgical planning. AJR Am J
Heart J. 2011;32:1551–1560. Roentgenol. 2011;197:W842–W851.
108. Pazos-López P, Pozo E, Siqueira ME, et al. Value of CMR for the
differential diagnosis of cardiac masses. JACC Cardiovasc Imaging.
2014;7:896–905.
SECTION IV  Right Ventricular and Congenital Heart Disease

39 
Cardiovascular Magnetic Resonance
Assessment of Right Ventricular Anatomy
and Function
Alicia M. Maceira and Dudley J. Pennell

Accurate noninvasive assessment of right ventricular (RV) mass and thinner walls than the left ventricle. RV wall thickness is usually 3 to
systolic function is important in several pathologies, such as grown- 5 mm, the proportion between right ventricle and left ventricle being
up congenital heart disease,1,2 pulmonary hypertension,3,4 interstitial as 1 to 316; it is thickest at the base and gradually becomes thinner
lung disease,5 valvular heart disease,6 and arrhythmogenic RV car- toward the apex. The whole inner surface except the conus arteriosus is
diomyopathy.7 Right ventricular function is also a prognostic factor covered by more or less prominent muscular columns called trabeculae
in coronary artery disease8,9 and heart failure,10,11 even after cardiac carneae and from some of them (papillary muscles), the chordae ten-
resynchronization therapy.12 This chapter aims to summarize the fea- dineae connect the myocardium to the tricuspid valve, with its septal
tures of the normal right ventricle, briefly describe cardiovascular leaflet more apically placed than the septal leaflet of the mitral valve.17
magnetic resonance (CMR) techniques for assessing RV dimensions The anterior tricuspid valve leaflet is usually the largest and extends
and function, and give reference values for the assessment of the right from the infundibular region anteriorly to the inferolateral wall pos-
ventricle. teriorly; the septal leaflet extends from the interventricular septum to
the posterior ventricular border; the posterior leaflet attaches along the
posterior margin of the annulus from the septum to the inferolateral
NORMAL RIGHT VENTRICULAR ANATOMY wall.18 Finally, a muscular band frequently extends from the base of
The right ventricle is a thin, highly trabeculated structure that is triangular the anterior papillary muscle to the ventricular septum. This band is
in form and, on gross inspection, appears to be wrapped around the considered to prevent overdistension of the ventricle and is called the
left ventricle. It has three well-differentiated components with specific moderator band.19
structure and function. The inlet portion extends from the tricuspid The depictions of the moderator band, the infundibulum,
valve to the insertions of the papillary muscles on the ventricular wall, and the different levels of insertion of the tricuspid and mitral
the trabecular portion involves the RV body and apex and is the main septal leaflets are important diagnostic features for identification
pumping component, and the outlet portion or infundibulum extends of the right ventricle, which can be difficult in some congenital
up to the pulmonary valve and has a thin, nontrabeculated wall (Fig. cardiomyopathies.
39.1). The anterosuperior wall of the right ventricle is rounded and
convex, its inferior surface is flattened and forms a small part of the Importance of Assessing Right Ventricular Dimensions
diaphragmatic surface of the heart, and its posterior wall is formed by and Function
the ventricular septum, which bulges into the right ventricle, owing to The measurement of RV dimensions, morphology, and function is
the much greater left ventricular (LV) systolic pressure,13 so a trans- important in several situations, such as congenital heart disease, LV
verse section of the cavity presents a semilunar outline. The right ven- heart failure, pulmonary hypertension, pulmonary embolism, valvular
tricle has a continuum of muscle bands that rotate by approximately heart disease, lung disease, and arrhythmogenic RV cardiomyopathy.
160 degrees from the epicardium to the endocardium.14 The principal RV failure may result from conditions that lead to impaired RV
axis of these fibers is oblique to the long axis of the right ventricle. contractility, such as RV infarction, right-sided cardiomyopathies, val-
RV contraction is then more dependent on longitudinal shortening vular heart disease or severe sepsis; to RV pressure overload, including
than that of the LV. In the normal adult, the total RV free wall mass pulmonic stenosis, pulmonary primary hypertension, and pulmonary
is 21 ± 13 g/m2.15 hypertension secondary to left heart disease, lung disease, or throm-
The right ventricle has several distinctive features. In its upper left boembolic disease; and to RV volume overload, for instance, tricuspid
portion, there is a conical pouch called the conus arteriosus or infun- or pulmonary regurgitation. Many disorders, such as corrected and
dibulum, from which the pulmonary artery arises. A tendinous band uncorrected adult congenital heart disease and intracardiac shunts, may
connects the posterior surface of the conus arteriosus to the aorta. Also, result in right ventricle failure through a combination of pathophysi-
as a subpulmonary ventricle, the RV has a lower ejection fraction and ologic mechanisms. Also, decompensated right ventricle (both acute

454
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 455

A B C

D E F
FIG. 39.1  Steady-state free precession cines in different views that allow the evaluation of the right ventricle.
Top row depicts a four-chamber view (A), two-chamber view (B), and right ventricular outflow tract view (C).
Bottom row shows three short-axis orientations at basal (D), mid (E), and apical (F) levels. The inlet portion
extends from the tricuspid valve to the insertions of the papillary muscles on the ventricular wall and can
be assessed in A and B, the trabecular portion involves the RV body and apex and is the main pumping
component (A, D, E, F), and the outlet portion or infundibulum extends up to the pulmonary valve and has
a thin, nontrabeculated wall (C). Note the rounded shape of the anterosuperior wall. In the short-axis sections
the right ventricle presents a semilunar outline.

and acute-on-chronic) is increasing as the prevalence of predisposing assessment of RV dimensions and function. More recently, “nongeo-
conditions grows. metric” techniques such as three-dimensional (3D) echocardiography,
The prognostic value of RV function has been shown in several CMR, and multidetector-row computed tomography (CT) permit
conditions such as LV heart failure,20,21 valvular heart disease,22 pulmo- accurate assessment of RV volumes, function, and mass.
nary hypertension,23 congenital heart disease,24 or myocardial infarction
(MI).25 Thus the early detection of RV dysfunction can have an impact Echocardiography
on therapeutic decision making and on prognosis. Finally, improved Echocardiography is the most frequently used technique for assessing
understanding of the RV response to pressure and volume overload the right ventricle; it is cheap, widely available, and can be used bedside
might lead to more optimal surgical and medical treatments. in very ill patients. It provides information about RV morphology,
dimensions, septum convexity, function, tricuspid regurgitation, and
Techniques for Assessing Right Ventricular Dimensions estimates of pulmonary arterial pressure and RV pressure. But the
and Function assessment of the right ventricle with echocardiography has several
Global RV function has been traditionally difficult to assess, given its limitations. First, the location of the right ventricle behind the sternum
irregular shape, which cannot be assumed to any geometrical model. restricts the window that can be accessed by the ultrasound beam.
Several imaging techniques have been used in the past to assess RV Second, the complex shape and thin walls of the right ventricle make
dimensions and function. The chest radiograph is a simple method to it necessary to image the right ventricle from several projections, although
assess RV size with the cardiothoracic ratio, but this may be misleading the short-axis view is usually the most helpful. Third, the thick tra-
because an enlarged RV may compress the LV, resulting in a normal beculations in the chamber may be confused with a thrombus, a tumor,
cardiothoracic ratio. Angiography used to be the gold standard for or hypertrophic cardiomyopathy. Finally, there is a lack of accurate
assessment of RV volumes and regional and global function. But this mathematical models to quantify RV mass and volumes with M-mode
technique is invasive, involves ionizing radiation and the use of poten- or two-dimensional echocardiography (2DE) because quantitative mea-
tially nephrotoxic contrast, and is not as accurate as CMR.26 Echocar- surements are based on geometric assumptions that do not apply to
diography and radionuclide ventriculography have been used for the the right ventricle. A qualitative approach for RV volume assessment
456 SECTION IV  Right Ventricular and Congenital Heart Disease

is usually applied, with the RV size being described as either normal but it may have problems in the case of thin wall or poor image quality
or mildly, moderately, or severely enlarged. If the RV is the same size in the RV lateral wall, and there is variability across vendors. Doppler
as the LV, it is usually characterized as moderately enlarged, and if larger transesophageal echocardiography is another echocardiographic method
than the LV, it is severely enlarged. Also, qualitative evaluation of RV of RV assessment, but it is semiinvasive, is not well suited for evaluation
function is usually applied with RV characterized as normal or mildly, of anteriorly positioned right ventricles, and requires special skills.
moderately, or severely dysfunctional. An approximation to quantifica- Three-dimensional echocardiography (3DE) has emerged as a more
tion has been introduced with M-mode measurement of the tricuspid accurate and reproducible approach to ventricular quantitation, mainly
annular plane systolic excursion (TAPSE).27 This parameter provides a by avoiding the use of geometric assumptions of the ventricular shape.
rough estimate of RV function but it is not angle independent: it only Real-time 3DE is an online acquisition of a 3D dataset of the heart
takes into account the longitudinal function of the RV, which is the without the need for electrocardiographic and respiratory gating.32 It
predominant, but not unique, component of RV systolic function, and, calculates right ventricular ejection fraction (RVEF) using the volumetric
because it is measured at the basal segment, the presence of regional semiautomated border detection method, which needs to be manually
wall motion abnormalities will affect its accuracy. The fractional area adjusted, and after acquisition and display of end-diastolic and end-
change in the four-chamber view has also been used but, again, this systolic frame, long-axis, planes, and volumetric data of the right ventricle
takes into account only the lateral free wall and the presence of regional are analyzed offline. Eventually, curves of regional and global RVEF are
wall motion abnormalities or RV dilatation might affect its accuracy produced and analyzed. However, there are practical problems, such as
(Fig. 39.2). Other indicators of RV function are flow Doppler-derived full cardiac visualization, good-quality endocardial border recognition
indices such as the myocardial performance index,28 tissue Doppler for manual endocardial tracing, and time consumption. Also, these
measurements of myocardial tricuspid annular myocardial velocities methods need a stable cardiac rhythm and constant cardiac function
and time intervals, and strain and strain rate measurements of con- during image acquisition, although new single-beat 3DE techniques
tractility whether with tissue Doppler or speckle tracking imaging (STI). allow a fast acquisition in one short breath-hold. 3DE has been com-
Tissue Doppler is an angle-dependent technique, and incorrect align- pared with CMR for the evaluation of RV function, and improved
ment with the ultrasound beam, poor signal-to-noise ratio (SNR) and results in comparison with 2D echocardiography have been obtained,
variability because of placement of the region of interest may affect the both with the usual technique33,34 and with the new single-beat 3DE,35
results.29 Speckle tracking imaging is a promising technique that has with consistent underestimation of RV volumes and ejection fraction
been applied to the right ventricle30 to measure longitudinal strain in with 3DE. Although 3DE has been used mainly for the left ventricle,
the six segments in which the right ventricle is classically divided.31 assessment of the right ventricle with 3DE is feasible during routine
This technique is useful for the diagnosis of various right heart diseases, standard echocardiography.

A B

TAPSE

C D
FIG. 39.2  M-mode and two dimensional echocardiographic methods for right ventricular (RV) function assess-
ment. (A and B) RV planimetry in diastole (A) and systole (B) from which the fractional area change can be
derived. (C and D) M-mode measurement of the tricuspid annular plane systolic excursion (TAPSE). Both
parameters provide a rough estimate of RV function with significant accuracy and reproducibility problems.
TAPSE only takes into account the longitudinal function of the right ventricle, which is the predominant, but
not unique, component of RV systolic function, and does not account for regional wall motion abnormalities.
The fractional area change is measured in only one plane, so the presence of regional wall motion abnormali-
ties or RV dilatation greatly affect its accuracy.
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 457

A B

C D
FIG. 39.3  Example of ventricular function analysis with computed tomography. (A and B) Analogous to
cardiovascular magnetic resonance, they show myocardial analysis for left ventricular function quantification
both with ejection fraction and with wall thickening. (C and D) The ventricular threshold method for measure-
ment of both LV (C) and right ventricular (D) volumes.

Radionuclide Angiography times requires manual adjustment (Fig. 39.3). In a study that compared
This technique provides a reliable quantitative measurement of ven- accuracy and reproducibility of MSCT with CMR in the quantification
tricular function not based on geometric assumptions with good agree- of RV function, spiral MSCT with reconstruction at 5% intervals had
ment with CMR.36 Although echocardiography and CMR are the two good agreement with CMR for RVEF and volumes, with good accuracy
most commonly used imaging techniques for noninvasive assessment and reproducibility,42 but at the expense of an average radiation dose of
of the right ventricle, nuclear imaging provides new opportunities for 12 to 18 mSv. Another study that compared volumetric quantification
comprehensive evaluation of the right ventricle from a single study, of RV-shaped phantoms with CMR, MSCT, and real-time 3DE showed
because it can assess RV perfusion and metabolism as well as morphol- that CMR images yielded the most accurate measurements, whereas
ogy and ejection fraction. Some years ago this technique did not work MSCT measurements showed slight but consistent overestimation; and
well for the right ventricle, owing to problems such as the limited count 3DE showed small underestimation.43
numbers in this chamber and the overlap of other cardiac chambers.37,38
Currently, gated positron emission tomography (PET) has shown Cardiovascular Magnetic Resonance
moderate-to-high correlation with CMR and CT in the assessments of CMR has some important advantages over other imaging techniques,
RV volume and ejection fraction simultaneously with quantification which have led to the growing enthusiasm for its use. CMR offers accu-
of myocardial glucose metabolism in conditions such as pulmonary rate and reproducible tomographic, static, or cine images of high spatial
hypertension.39 Gated blood-pool single-photon emission computed and temporal resolution in any desired plane without exposure to con-
tomography (SPECT) appears a promising technique because it does trast agents or ionizing radiation. It allows the acquisition of true RV
not require geometric assumptions and provides both global and regional short-axis images encompassing the entire right ventricle with high
RV function quantification with good diagnostic accuracy compared spatial and temporal resolution, thereby providing highly accurate and
with CMR.40 Still, these techniques have disadvantages compared with reproducible quantitative RV mass and functional data regardless of
other imaging modalities, such as poor resolution, the use of ionizing its position in the thorax.44,45 Nowadays, this technique is considered
radiation, the need for an adequate bolus injection for first-pass studies the gold standard for quantitative assessment of RV volume, mass, and
and a regular rhythm, and the lack of clinical experience. Therefore function.
they have been of limited use for the study of the right ventricle so far.
Imaging Strategies for Cardiovascular Magnetic
Multislice Computed Tomography Resonance of the Right Ventricle
Multislice CT (MSCT), although not a first-line technique for RV function Before the study begins, it is essential to obtain an accurate ECG gating
quantification, is emerging as an alternative tool, especially for patients with minimal ectopy and to instruct the patient in breath-holding.
with implantable devices (a contraindication for CMR). However, MSCT Sometimes oxygen may be applied to improve breath-hold length. Ven-
uses ionizing radiation and potentially nephrotoxic contrast, and requires tricular ectopy can be a problem, mainly in patients with congenital
a low and stable heart rate during acquisition.41 The quantification of heart disease or with suspicion of arrhythmogenic RV cardiomyopathy
ventricular volumes throughout the cardiac cycle requires retrospective (ARVC). If this condition is present, pretreatment with an antiarrhythmic
electrocardiogram (ECG)-gated helical acquisition, which increases the agent should be considered or, if this is unavailable or unsuccessful,
radiation significantly. Postprocessing is with either a volume threshold prospective triggering with acquisition window limited to systole can
approach or, similar to CMR, myocardial analysis is automated but most diminish artifacts.
458 SECTION IV  Right Ventricular and Congenital Heart Disease

FIG. 39.4  Short-axis and transaxial spin echo images of the heart in a healthy subject. The right ventricular
free wall is perfectly depicted between the black-blood pool and the black pericardium. Note that there is
epicardial fat deposition around the right coronary artery (anterior atrioventricular groove), left anterior descend-
ing artery (anterior interventricular groove), and left circumflex (posterior atrioventricular groove). It is important
to know the normal patterns of fat distribution to prevent false-positive reading of scans in assessing patients
for possible fat infiltration in arrhythmogenic right ventricular cardiomyopathy.

Spin echo (black-blood) sequences (including turbo spin echo, half- walls. The question has been raised as to whether the RV volumes
Fourier acquisition single-shot turbo spin echo, or spin echo-echo planar should always be measured in the axial orientation. Yet the interstudy
imaging) are used mainly for anatomic assessment (Fig. 39.4), because reproducibility of RV measurements in the short-axis orientation is
the detection of fatty replacement/infiltration in the RV free wall is good,54 with differences not clinically significant and this orientation
subject to high interobserver variability and, at the moment, is not allows the LV and RV dimensions to be measured simultaneously, thus
considered a diagnostic criteria for ARVC to improve their specificity.46 saving time (Fig. 39.8). A recent study55 has shown comparable repro-
Functional evaluation of the RV is performed by using steady-state ducibility for the two approaches when measuring volumes of the sys-
free precession (SSFP) cines with segmented fast imaging, allowing the temic right ventricle in 22 patients with congenital heart disease. Although
whole stack of images to be acquired in 5 to 8 minutes,47 thereby reduc- ejection fraction and stroke volume were measured more consistently
ing breathing and movement artifact (Fig. 39.5). Moreover, real-time in the axial orientation, ventricular mass was measured more consistently
SSFP imaging can acquire the whole stack in just one breath-hold with in the short-axis orientation. Nowadays, nearly all CMR units have
acceptable accuracy and image quality.48 In patients who are unable to adopted the short-axis stack approach, leaving the axial orientation for
hold their breath consistently, solutions using the same sequence with specific congenital heart disease patients.
more signal averages or combined with navigator echo are successful As commented, Simpson’s rule is used for measuring volumes and
with free breathing,49 with a slight increase in the scanning time. function. A standardized method of combined ventricular functional
RV function can be estimated by CMR, in a way analogous to echo- analysis is described in Chapter 16. When using this method, it is
cardiography, with the fractional area change method (Fig. 39.6). This critical to ensure that the most basal part of the free wall of the right
is a fast method but lacks accuracy, owing to the need for geometric ventricle in diastole is included in the most basal cine slice because
assumptions. A better method of measuring volumes, and thereby func- it can be easily truncated. Manual or semiautomatic planimetry of
tion, is by using Simpson’s rule. This method requires a stack of con- the endocardial borders of each ventricle at both end diastole and
tiguous tomographic slices to be acquired that encompass both ventricles end systole and of the epicardial borders at end diastole is done. Care
entirely. must be taken to exclude the right and left atria as they come into the
The right ventricle is well shown in the transaxial plane from the basal imaging planes during systole. The ventricular volume is equal
tricuspid valve to the apex. Consequently, a stack of transverse images to the sum of the endocardial areas multiplied by the distance between
was primarily used for quantification, with good agreement with pul- the centers of each slice (Fig. 39.9).56 The stroke volume is equal to
monary flow and LV stroke volume50,51 (Fig. 39.7), as well as better the difference between the end-diastolic volume (EDV) and the end-
interobserver and intraobserver reproducibility than the short-axis systolic volume (ESV). The ejection fraction (EF) is calculated as the
plane.52,53 However, it can be problematic because it is subject to con- stroke volume divided by the EDV. The mass is calculated as the volume
siderable partial volume effects, especially in the anterior and inferior of tissue occupied by the free wall multiplied by an assumed density
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 459

RV LV

FIG. 39.5  Steady-state free precession cine images in eight short-axis planes from cines acquired from base
to apex. The series of short-axis slices begins in the atrioventricular groove in diastole and goes through the
ventricles down to the apex, thus covering the whole left ventricle (LV) and right ventricle (RV). In the most
basal slice (upper left), the pulmonary artery will appear in systole because of atrioventricular ring descent.
The high contrast between blood and myocardium allows the drawing of the endocardial and epicardial
borders for volume and mass determinations. For the images to be accurate and reproducible, it is important
to always use the same slice thickness and the same gap between slices. Also, retrospective gating should
be used to acquire the whole cardiac cycle.

FIG. 39.6  Measurement of uniplane fractional area change (red shaded area) of the right ventricle through
planimetry of the right ventricle in diastole (left) and systole (right) in a four-chamber view. This is a fast
approximation to right ventricular function but, as for two-dimensional echocardiography, the use of a single
plane makes it insensitive to regional wall motion abnormalities or geometric distortion, affecting its accuracy
and reproducibility.

of 1.05 g/mL. The volumes obtained by this method are independent ventricle in contiguous short-axis slices and tracking of the tricuspid
of geometric assumptions and dimensionally accurate.57 Mass measure- valve throughout the cardiac cycle. A volume–time curve can then be
ments agree well with autopsy studies.58 RV parameters obtained with produced from which a velocity–time curve can be derived for subse-
this technique are reproducible, with an interobserver variability of quent calculation of diastolic function parameters.
6.3% for EDV, 8.6% for ESV, 7% for stroke volume, 4.4% for EF, and If necessary, other CMR techniques could be used in the assess-
7.8% for RV mass and an intraobserver variability of 3.6% for EDV, ment of the right ventricle. Flow velocity maps allow for accurate
6.5% for ESV, 5.9% for stroke volume, 4% for EF, and 5.7% for RV assessment of pulmonary valve regurgitation (regurgitant fraction)60
mass.59 Current analysis software allows for automated quantification and systemic-to-pulmonary flow ratio. Magnetic resonance angiogra-
of the RV, with automated endocardial border detection of the right phy (MRA) must be done in case assessment of great vessel anatomy is
460 SECTION IV  Right Ventricular and Congenital Heart Disease

PA
PV

RV

PA
Ao

RV
LV
RA
LA

B
FIG. 39.7  (A) To plan the transversal slices, a cine sequence through the right ventricular (RV) outflow tract
aligned in an oblique sagittal plane should be acquired. This view is useful for examining the function of the
right ventricular free wall from the apex to the pulmonary valve (PV) and visualizing pulmonary regurgitation.
(B) Multiple transaxial steady-state free precession cine sequences in contiguous planes should be acquired
from the pulmonary valve level down to the inferior right ventricular wall, using the right ventricular outflow
tract view to pilot them. These planes are invaluable for examining regional right ventricular wall motion.
Again, the slice thickness and gap between slices should be consistent among studies. Two features of the
normal right ventricle are seen in this transversal stack: the muscular outflow tract and the more apical
insertion of the tricuspid valve in the septum compared with the septal leaflet of the mitral valve. Ao, Aorta;
LA, left atrium; LV, left ventricle; PA, pulmonary artery; RA, right atrium.

advisable, such as in a number of congenital cardiac conditions.61 New of patients with ischemic heart disease,63 and RV kinetic energy, a 4D
free-breathing time resolved MRA techniques enable accurate diagno- flow-derived parameter, has been suggested as a more robust measure
sis and vessel measures compared with conventional breath-hold MRA of RV efficiency than RV function.64 Diffusion tensor imaging has
and can be performed in children and adults who find breath-holding also been applied to the assessment of RV fibers’ geometry. Although
difficult.62 Recently, four-dimensional (4D) flow has been shown to promising, these techniques are still in development and more work
be able to detect mild impairment of RV function in a small group is needed to fully understand all the information that can be derived
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 461

FIG. 39.8  Volumetric methods for assessment of right ventricular (RV) function (Simpson’s rule). (A) Simul-
taneous quantification of left ventricular and RV volumes and systolic function in the short-axis orientation.
There is difficulty in delineating the most basal slice of the right ventricle, where the right atrium and the
pulmonary trunk may be incorrectly included in the RV blood pool. (B) Quantification of only the right ventricle
in the axial orientation; although the tricuspid valve and right atrium are easily depicted here, problems with
partial volume effects in the inferior slices are significant.

from them, along with their potential clinical use (Fig. 39.10). Myo- absolute and normalized RV mass, EDV, and stroke volume (Figs. 39.11
cardial tissue tagging, feature tracking, and myocardial phase con- and 39.12). It was also an independent predictor of absolute and nor-
trast are briefly summarized later. Finally, CMR with LGE can detect malized active tricuspid peak filling rate (PFRA, PFRA/BSA). There was
myocardial fibrosis in a number of both ischemic and nonischemic a significant decrease with age of normalized RV mass and of absolute
cardiomyopathies.65–67 and normalized EDV and ESV in both males and females. There was
a significant increase with age in absolute RVEF in both males and
NORMAL RIGHT VENTRICULAR VOLUMES AND females and a significant increase in normalized EF in males. For dia-
stolic function, absolute and normalized PFRE decreased significantly
SYSTOLIC FUNCTION with age in males and females, whereas absolute and normalized PFRA
There have been a number of human series describing the normal increased in males. Accordingly, PFRE/PFRA decreased significantly. In
characteristics of RV size and function using autopsy,68 echocardiog- a multivariable analysis, age was an independent predictor of absolute
raphy,69,70 x-ray angiography,71 radionuclide angiography,72 CMR,73–76 and normalized ventricular mass and volumes (EDV, ESV, stroke volume,
and ultrafast CT.77 We have reported on RV reference parameters for EDV/BSA, ESV/BSA, stroke volume/BSA) and of EF. It was also an
mass, volumes, and systolic and diastolic function using cine SSFP CMR independent predictor of diastolic variables (PFRE, PFRA, PFRE/PFRA,
techniques and analysis from 120 healthy adult subjects.78 These gender- PFRE/EDV, PFRA/EDV, PFRE/BSA, PFRA/BSA). Thus the interpretation
specific data are summarized in Tables 39.1–39.5. We observed that of RV parameters in borderline clinical cases, especially in arrhythmo-
many clinical parameters of RV volumes and systolic and diastolic genic RV cardiomyopathy, cardiovascular shunting, and adult congenital
function are significantly dependent on gender, age, and body surface heart disease, should be referred to age-, gender-, and BSA-normalized
area (BSA). On multivariable analysis, BSA was found to significantly values to determine normality or severity of abnormality. In 2016 new
influence RV mass, EDV, ESV, stroke volume, and early tricuspid peak sex-specific normative values for RV measurements with CMR obtained
filling rate (PFRE). Gender had a significant independent influence on from a much larger series of 1336 adults (64 ± 9 years, 576 men) free
462 SECTION IV  Right Ventricular and Congenital Heart Disease

RV LV

B
FIG. 39.9  Summation of discs is used to quantify ventricular volumes and mass. The ventricular volume is
equal to the sum of the endocardial areas multiplied by the distance between the centers of each slice, both
for the end-diastolic volume (A) and the end-systolic volume (B). The ventricular mass is calculated as the
sum of the epicardial minus the endocardial areas multiplied by the distance between the centers of each
slice, as shown in A. LV, Left ventricle; RV, right ventricle.

A B
FIG. 39.10  Diffusion tensor imaging of the left ventricle/right ventricle (A) and four-dimensional flow assess-
ment of the pulmonary artery and branches (B) of the right ventricle. Both techniques are promising for
assessment of fiber geometry and flow dynamics, respectively.
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 463

TABLE 39.1  SSFP Right Ventricular Volumes, Systolic Function, and Mass (Absolute and
Normalized to BSA) by Age Decile (Mean, 95% CI): Males
20–29 Years 30–39 Years 40–49 Years 50–59 Years 60–69 Years 70–79 Years
Absolute Values
EDV (mL) SD 25.4 177 (127–227) 171 (121–221) 166 (116–216) 160 (111–210) 155 (105–205) 150 (100–200)
ESV (mL) SD 15.2 68 (38–98) 64 (34–94) 59 (29–89) 55 (25–85) 50 (20–80) 46 (16–76)
SV (mL) SD 17.4 108 (74–143) 108 (74–142) 107 (73–141) 106 (72–140) 105 (71–139) 104 (70–138)
EF (%) SD 6.5 61 (48–74) 63 (50–76) 65 (52–77) 66 (53–79) 68 (55–81) 70 (57–83)
Mass (g) SD 14.4 70 (42–99) 69 (40–97) 67 (39–95) 65 (37–94) 63 (35–92) 62 (33–90)

Normalized to BSA
EDV/BSA (mL/m2) SD 11.7 91 (68–114) 88 (65–111) 85 (62–108) 82 (59–105) 79 (56–101) 75 (52–98)
ESV/BSA (mL/m2) SD 7.4 35 (21–50) 33 (18–47) 30 (16–45) 28 (13–42) 25 (11–40) 23 (8–37)
SV/BSA (mL/m2) SD 8.2 56 (40–72) 55 (39–71) 55 (39–71) 54 (38–70) 53 (37–69) 52 (36–69)
EF/BSA (%/m2) SD 4 32 (24–40) 32 (25–40) 33 (25–41) 34 (26–42) 35 (27–42) 35 (27–43)
Mass/BSA (g/m2) SD 6.8 36 (23–50) 35 (22–49) 34 (21–48) 33 (20–46) 32 (19–45) 31 (18–44)

BSA, Body surface area; CI, confidence interval; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic volume; SD, standard
deviation; SSFP, steady-state free precession; SV, stroke volume.

TABLE 39.2  SSFP Right Ventricular Volumes, Systolic Function and Mass (Absolute and
Normalized to BSA) by Age Decile (Mean, 95% CI): Females
20–29 Years 30–39 Years 40–49 Years 50–59 Years 60–69 Years 70–79 Years
Absolute Values
EDV (mL) SD 21.6 142 (100–184) 136 (94–178) 130 (87–172) 124 (81–166) 117 (75–160) 111 (69–153)
ESV (mL) SD 13.3 55 (29–82) 51 (25–77) 46 (20–72) 42 (15–68) 37 (11–63) 32 (6–58)
SV (mL) SD 13.1 87 (61–112) 85 (59–111) 84 (58–109) 82 (56–108) 80 (55–106) 79 (53–105)
EF (%) SD 6 61 (49–73) 63 (51–75) 65 (53–77) 67 (55–79) 69 (57–81) 71 (59–83)
Mass (g) SD 10.6 54 (33–74) 51 (31–72) 49 (28–70) 47 (26–68) 45 (24–66) 43 (22–63)

Normalized to BSA
EDV/BSA (mL/m2) SD 9.4 84 (65–102) 80 (61–98) 76 (57–94) 72 (53–90) 68 (49–86) 64 (45–82)
ESV/BSA (mL/m2) SD 6.6 32 (20–45) 30 (17–43) 27 (14–40) 24 (11–37) 21 (8–34) 19 (6–32)
SV/BSA (mL/m2) SD 6.1 51 (39–63) 50 (38–62) 49 (37–61) 48 (36–60) 46 (34–58) 45 (33–57)
EF/BSA (%/m2) SD 5.2 37 (27–47) 38 (27–48) 38 (28–49) 39 (29–49) 40 (30–50) 41 (31–51)
Mass/BSA (g/m2) SD 5.2 32 (22–42) 30 (20–40) 29 (19–39) 27 (17–37) 26 (16–36) 24 (14–35)

BSA, Body surface area; CI, confidence interval; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic volume; SD, standard
deviation; SSFP, steady-state free precession; SV, stroke volume.

TABLE 39.3  SSFP Right Ventricular Diastolic Function and Atrioventricular Plane Descent
(Absolute and Normalized Values) by Age Decile (Mean, 95% CI): Males
20–29 Years 30–39 Years 40–49 Years 50–59 Years 60–69 Years 70–79 Years
Absolute Values
PFRE (mL/s) SD 137 545 (277–814) 491 (223–760) 438 (169–706) 384 (116–652) 330 (62–599) 276 (8–545)
PFRA (mL/s) SD 175 366 (23–709) 413 (70–756) 461 (118–804) 508 (165–852) 556 (213–899) 604 (260–947)
PFRE/PFRA SD* 0.49 1.6 (0.6–2.5) 1.2 (0.3–2.2) 1.0 (0.0–1.9) 0.7 (−0.2–1.7) 0.6 (−0.4–1.5) 0.5 (−0.5–1.4)
Septal AVPD (mm) SD 4.1 16 (8–24) 15 (7–24) 15 (7–23) 14 (6–22) 14 (6–22) 13 (5–21)
Lateral AVPD (mm) SD 4.4 23 (14–32) 23 (14–31) 22 (14–31) 22 (13–30) 21 (13–30) 21 (12–29)

Normalized Values
PFRE/BSA (mL/s/m2) SD 71 280 (142–419) 252 (114–390) 224 (85–362) 195 (57–334) 167 (29–306) 139 (1–277)
PFRE/EDV (s−1) SD 0.75 3.1 (1.6–4.6) 2.8 (1.4–4.3) 2.6 (1.1–4.1) 2.3 (0.9–3.8) 2.1 (0.6–3.6) 1.9 (0.4–3.3)
PFRA/BSA (mL/s/m2) SD 94 190 (6–374) 213 (29–397) 236 (52–420) 259 (75–443) 283 (98–467) 306 (122–490)
PFRA/EDV (s−1) SD 1.07 2.1 (0.0–4.2) 2.5 (0.4–4.6) 2.9 (0.8–4.9) 3.2 (1.1–5.3) 3.6 (1.5–5.7) 4.0 (1.9–6.1)
Septal AVPD/long length (%) SD 4.5 18 (9–27) 18 (9–27) 17 (9–26) 17 (8–26) 17 (8–26) 16 (8–25)
Lateral AVPD/long length (%) SD 4.1 23 (15–31) 23 (15–31) 23 (15–31) 23 (15–31) 23 (15–31) 23 (15–31)

A, Active; AVPD, atrioventricular plane descent; BSA, body surface area; CI, confidence interval; E, early; PFR, peak filling rate; SD, standard
deviation; SD*, standard deviation of log transformed data; SSFP, steady-state free precession.
464 SECTION IV  Right Ventricular and Congenital Heart Disease

TABLE 39.4  SSFP Right Ventricular Diastolic Function and Atrioventricular Plane Descent
(Absolute and Normalized Values) by Age Decile (Mean, 95% CI): Females
20–29 Years 30–39 Years 40–49 Years 50–59 Years 60–69 Years 70–79 Years
Absolute Values
PFRE (mL/s) SD 117 471 (241–701) 419 (189–649) 368 (137–598) 316 (86–546) 264 (34–494) 213 (−17–443)
PFRA (mL/s) SD 153 355 (54–656) 360 (59–660) 365 (64–665) 370 (69–670) 374 (74–675) 379 (79–680)
PFRE/PFRA SD* 0.46 1.6 (0.7–2.5) 1.3 (0.4–2.2) 1.0 (0.1–1.9) 0.8 (−0.1–1.7) 0.7 (−0.2–1.6) 0.5 (−0.4–1.4)
Septal AVPD (mm) SD 3.0 16 (10–22) 15 (9–20) 13 (7–19) 12 (6–18) 11 (5–17) 10 (4–16)
Lateral AVPD (mm) SD 3.5 22 (15–29) 21 (14–28) 21 (14–28) 20 (13–27) 20 (13–27) 19 (12–26)

Normalized Values
PFRE/BSA (mL/s/m2) SD 68 278 (145–411) 247 (114–380) 216 (83–349) 185 (52–318) 153 (20–286) 122 (−11–255)
PFRE/EDV (s−1) SD 0.85 3.4 (1.8–5.1) 3.1 (1.5–4.8) 2.8 (1.2–4.5) 2.5 (0.9–4.2) 2.2 (0.6–3.9) 1.9 (0.3–3.6)
PFRA/BSA (mL/s/m2) SD 89 211 (36–386) 212 (37–388) 214 (39–389) 215 (40–390) 217 (42–392) 218 (43–393)
PFRA/EDV (s−1) SD 1.03 2.4 (0.4–4.4) 2.6 (0.6–4.6) 2.8 (0.8–4.8) 3.0 (1.0–5.0) 3.2 (1.2–5.2) 3.4 (1.4–5.4)
Septal AVPD/long length (%) SD 3.9 19 (11–27) 18 (11–26) 17 (10–25) 17 (9–24) 16 (8–23) 15 (7–22)
Lateral AVPD/long length (%) SD 4.0 24 (16–32) 24 (16–32) 24 (16–32) 24 (16–32) 24 (16–32) 24 (16–31)

A, active; AVPD, atrioventricular plane descent; BSA, body surface area; CI, confidence interval; E, early; PFR, peak filling rate; SD, standard
deviation; SD*, standard deviation of log transformed data; SSFP, steady-state free precession.

TABLE 39.5  SSFP Right Ventricular Summary Data for All Ages (Mean ± Standard Deviation,
95% CI)
All Males Females
EDV (mL) 144 ± 23 (98–190) 163 ± 25 (113–213) 126 ± 21 (84–168)
EDV/BSA (mL/m2) 78 ± 11 (57–99) 83 ± 12 (60–106) 73 ± 9 (55–92)
ESV (mL) 50 ± 14 (22–78) 57 ± 15 (27–86) 43 ± 13 (17–69)
ESV/BSA (mL/m2) 27 ± 7 (13–41) 29 ± 7 (14–43) 25 ± 7 (12–38)
SV (mL) 94 ± 15 (64–124) 106 ± 17 (72–140) 83 ± 13 (57–108)
SV/BSA (mL/m2) 51 ± 7 (37–65) 54 ± 8 (38–70) 48 ± 6 (36–60)
EF (%) 66 ± 6 (54–78) 66 ± 6 (53–78) 66 ± 6 (54–78)
EF/BSA (%/m2) 36 ± 5 (27–45) 34 ± 4 (26–41) 39 ± 5 (29–49)
Mass (g) 48 ± 13 (23–73) 66 ± 14 (38–94) 48 ± 11 (27–69)
Mass/BSA (g/m2) 31 ± 6 (19–43) 34 ± 7 (20–47) 28 ± 5 (18–38)
PFRE (mL/s) 371 ± 125 (126–615) 405 ± 137 (137–674) 337 ± 117 (107–567)
PFRE/BSA (mL/m2) 202 ± 69 (67–337) 207 ± 70 (68–345) 197 ± 68 (64–330)
PFRE/EDV (s−1) 2.6 ± 0.8 (1.0–4.1) 2.4 ± 0.75 (1.0–3.9) 2.7 ± 0.85 (1.0–4.3)
PFRA (mL/s) 429 ± 168 (99–759) 489 ± 175 (146–833) 368 ± 153 (67–668)
PFRA/BSA (mL/m2) 233 ± 93 (50–415) 250 ± 94 (66–434) 215 ± 89 (40–390)
PFRA/EDV (s−1) 3.0 ± 1.0 (1.0–5.1) 3.1 ± 1.0 (1.0–5.2) 2.9 ± 1.0 (0.9–5.0)
PFRE/PFRA 0.9 ± 0.47 (−0.1–1.8) 0.8 ± 0.49 (−0.1–1.8) 0.9 ± 0.46 (0.0–1.8)
Septal AVPD (mm) 14 ± 3.6 (6–21) 15 ± 4.1 (6–23) 13 ± 3.0 (7–19)
Septal AVPD/long length (%) 17 ± 4.2 (9–25) 17 ± 4.5 (8–26) 17 ± 3.9 (9–25)
Lateral AVPD (mm) 21 ± 3.9 (13–29) 22 ± 4.4 (13–30) 21 ± 3.5 (14–27)
Lateral AVPD/long length (%) 23 ± 4.0 (15–31) 23 ± 4.1 (15–31) 24 ± 4.0 (16–32)

A, Active; AVPD, atrioventricular plane descent; BSA, body surface area; CI, confidence interval; E, early; EDV, end-diastolic volume; EF, ejection
fraction; ESV, end-systolic volume; PFR, peak filling rate; SSFP, steady-state free precession; SV, stroke volume.

of prevalent cardiovascular and pulmonary disease have been published,79 was used to show the trajectory of the RV free wall in systole. PSS
with slight differences in methodology. increased through time to an average of 12% across all segments (infe-
Another CMR approach that may be particularly valuable for quan- rior, mid, and superior wall) at the base, 14% at the mid ventricle, and
tifying regional RV free wall systolic function is myocardial tagging,80,81 16% at the apex, with a wave of motion toward the septum and outflow
a technique that enables the clinician to assess the complex mechanism tract. Naito and colleagues83 determined PSS only at the mid ventricle
of myocardial contraction and to quantify myocardial strain. Klein and and found a PSS of 6.7% in the superior wall segment and 20% for
colleagues82 analyzed the RV free wall motion and contraction in humans the midwall segment. Fayad and colleagues84 reported similar PSS values:
with CMR tagging. In this study, percent segmental shortening (PSS) 24.7% in the midwall segment of the midventricular slice and 28.7%
was obtained to measure the amount of contraction, and a vector analysis in the midwall segment of the apical slice. A 3D reconstruction of RV
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 465

FEMALES
120 60

volume/BSA−−Females (mL/m2)
RV end-diastolic volume/ +95% CI 50
BSA−−Females (mL/m2) 100
+95% CI

RV end-systolic
40
Mean Mean
80 30

−95% CI −95% CI
20
60
10

40 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

90 60

RV mass/BSA−−Females
50
fraction−−Females (%)

80
+95% CI +95% CI
40
RV ejection

70

(mL/m2)
Mean
Mean 30
60 −95% CI
20
−95% CI
50 10

40 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

500 600
rate/BSA−−Females (mL/m2)

rate/BSA−−Females (mL/m2)

+95% CI
400 500
RV active peak filling
RV early peak filling

400 +95% CI
300 Mean
300
200 Mean
−95% CI 200
100
100
−95% CI
0 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

3
+95% CI
RV early/active peak filling
rate ratio−−Females

2
Mean

1
−95% CI

0
20 30 40 50 60 70 80
Age (years)
FIG. 39.11  Normal values for right ventricular (RV) end-diastolic volume, end-systolic volume, mass, and
parameters of diastolic function for females normalized to body surface area (BSA). CI, Confidence
interval.
466 SECTION IV  Right Ventricular and Congenital Heart Disease

MALES

120 60

volume/BSA−−Males (mL/m2)
RV end-diastolic volume/
+95% CI 50 +95% CI
BSA−−Males (mL/m2)
100

RV end-systolic
Mean 40
Mean
80 30
−95% CI −95% CI
20
60
10

40 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

90 60

50 +95% CI

RV mass/BSA−−Males
80
fraction−−Males (%)

+95% CI
40
RV ejection

70

(g/m2)
Mean
Mean 30
60
20 −95% CI
−95% CI
50
10

40 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

500 600
+95% CI
rate/BSA−−Males (mL/m2)

rate/BSA−−Males (mL/m2)

400 500
RV active peak filling
RV early peak filling

+95% CI
400
300 Mean
300
200 Mean
−95% CI 200
100
100
−95% CI
0 0
20 30 40 50 60 70 80 20 30 40 50 60 70 80
Age (years) Age (years)

3
+95% CI
RV early/active peak filling
rate ratio−−Males

2
Mean

1
−95% CI

0
20 30 40 50 60 70 80
Age (years)
FIG. 39.12  Normal values for right ventricular (RV) end-diastolic volume, end-systolic volume, mass, and
parameters of diastolic function for males normalized to body surface area (BSA). CI, Confidence interval.
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 467

contraction with CMR tagging85 shows a primary contraction of the


RV tangential to its own surface plane and a circumferential contraction Right Ventricular Assessment in Ischemic
as the RV moves apically, with a twisting motion similar to that described Heart Disease
by Klein and colleagues.86 Still, although CMR tagging seems a promis- Isolated RV infarction is relatively rare, but concurrent RV infarction
ing method for the assessment of regional function, especially with in the setting of an inferior infarction because of proximal right coronary
stress techniques in ischemic heart disease, it remains to be seen whether occlusion97 occurs in up to half of LV infarctions.98 RV infarction can
it provides clinically relevant information beyond that provided by be detected and evaluated in extent by using late gadolinium enhance-
standard cine CMR. Further studies are needed to better define the ment (LGE) CMR. RV necrosis causes a loss of contractile mass, and
clinical role of CMR tagging. Myocardial phase contrast has also been if the inferior interventricular septum is involved, there is also a loss
used in small studies with promising results, but at the time of this of septal augmentation of RV function. The existence of RV dysfunction
writing, this technique is at a very initial stage and far from clinical in patients with inferior MI is associated with high rates of morbidity
use.87 Equally, CMR feature tracking (CMR-FT) assessment of RV strain and mortality.99,100 Cardiogenic shock is more frequent if the RV is
shows acceptable levels of agreement with 2DE and has been used to involved in inferior infarctions,101 whereas reperfusion of acute RV
objectively quantify global and regional RV dysfunction and dyssyn- infarcts by primary angioplasty has been shown to greatly improve RV
chrony in patients with ARVC and other conditions, with incremental function.102 It has been shown that RVEF quantified late after MI is an
value over conventional CMR imaging.88 CMR-FT is promising, although important predictor of prognosis adjusted for patient age, LV infarct
further work is needed to standardize the acquisition and analysis size, and LVEF.103 Accordingly, evaluation of RVEF with CMR is impor-
processes, to define normal values, and to assess its use in disease popu- tant in the setting of acute infarction, not only for RV mass and function
lations with RV dysfunction. quantification but also for detection and quantification of necrosis with
late gadolinium techniques that can improve risk stratification and
CARDIOVASCULAR MAGNETIC RESONANCE potentially refine patient management after MI.
ASSESSMENT OF RIGHT VENTRICULAR ANATOMY
AND FUNCTION IN DISEASE Right Ventricular Assessment in Arrhythmogenic Right
Ventricular Cardiomyopathy
Right Ventricular Assessment in Heart Failure In ARVC, normal myocardium is replaced by fibrofatty tissue, and
In left heart failure, atrial pressure rises, forcing open a number of pul- electrical instability develops. This disorder usually involves the right
monary capillaries. When all reserve capillaries are open, the increase ventricle, but the left ventricle and septum may also be affected.104 After
in pulmonary pressure leads to an increased load on the RV. Therefore hypertrophic heart disease, ARVC is the number one cause of sudden
the function of the RV during exercise in heart failure is important.89 cardiac death in young people, especially athletes. Evident forms of the
The prognostic value of RV function in advanced heart failure of disease are straightforward to diagnose on the basis of a series of diag-
various causes is being increasingly recognized90,91; therefore the esti- nostic criteria proposed by the International Task Force for Cardiomy-
mation of RV function is now warranted in the standard evaluation of opathy7; however, the diagnosis of early and mild forms of the disease
patients with heart failure, because it is helpful in the clinical assess- is often difficult. CMR is regarded as the best imaging technique for
ment and prognostic stratification of such patients. RV dysfunction is detecting RV structural and functional abnormalities.106 The main
also present in one-third of patients with heart failure with preserved advantage of CMR is the possibility of planning any desired view so
ejection fraction, usually associated with an advanced stage and poor that regions such as the outflow tract, which are hard to visualize with
outcomes.92 other techniques, can be assessed with great precision.
Di Salvo and colleagues93 studied 67 patients with heart failure who The most common findings in ARVC are RV wall motion abnor-
had been referred for cardiac transplantation with ischemic (46%) or malities, mainly in the subtricuspid region, systolic dysfunction, and
dilated (54%) cardiomyopathy. An RVEF of 35% or more at rest and dilatation,107 and these features are the only CMR-derived parameters
with exercise predicted overall survival. Maximal oxygen consumption included in the current diagnostic criteria, which try to be very specific
was also predictive of survival, with a modest correlation between RVEF in the general population and very sensitive in individuals who have a
and maximal oxygen consumption. Also, in patients with moderate high likelihood of inherited/genetic disease. CMR can also detect fatty
heart failure, de Groote and colleagues94 showed that three variables, infiltration; however, this alone does not allow a definitive diagnosis
NYHA classification, percent of maximal predicted VO2, and RVEF, of ARVC because fatty infiltration occurs in a high proportion of healthy
were independent predictors of both survival and event-free cardiac people, particularly elderly subjects. It is normally present in areas of
survival. Left ventricular EF and peak VO2 normalized to body weight the heart such as the atrioventricular groove, around the coronaries,
had no predictive value. The event-free survival rates from cardio- and in the epicardium, and there is a high interobserver variability in
vascular mortality and urgent transplantation at 1 year were 80%, its detection.108 LGE may show myocardial fibrosis in the right ventricle
90%, and 95% in patients with an RVEF less than 25%, with an RVEF or in the left ventricle of these patients, but LGE is not incorporated
of 25% or more and less than 35% and with an RVEF of 35% or in the current diagnostic criteria because of several limitations such as
more, respectively. At 2 years, survival rates were 59%, 77%, and 93%, the difficulty of detecting LGE in the thin RV wall and of distinguishing
respectively, in the same subgroups. Throughout the years, a number fibrosis from fat, low specificity of LV LGE, and high variability of
of papers have shown similar results. More recently, it has been shown detection between centers. Importantly, diagnosis of ARVC should always
that increased (>2 SD beyond the mean) RV volumes at a CMR study be made according to the criteria proposed by the Task Force and never
predict mortality in heart failure patients.95 Another study including 250 according to findings from a single test such as CMR.
patients with dilated cardiomyopathy studied with CMR showed that
a RV ejection fraction lower than 45% was an independent predictor Congenital Heart Disease
of death or cardiac transplantation during a median follow-up period A comprehensive summary of the role of CMR in congenital heart
of 6.8 years.96 Therefore CMR quantitative RV volumetric assessment disease is detailed in Chapters 40 and 41. Assessment of RV size, loca-
should be measured on all studies that are performed on patients with tion, and connections as well as of function and pulmonary flow are
heart failure. important.109 In congenital heart disease, the right ventricle may support
468 SECTION IV  Right Ventricular and Congenital Heart Disease

the pulmonary (subpulmonary right ventricle) or the systemic circula- CMR-derived RVEF is an independent noninvasive imaging predictor
tion (systemic right ventricle). In many of these patients, RV dysfunction of adverse outcomes in this patient population.120 CMR has been used
develops and leads to considerable morbidity and mortality. Therefore to determine the time course of changes in ventricular mass and func-
RV function in certain conditions needs close surveillance and timely tion after lung transplantation.121,122 Frist and colleagues123 observed
and appropriate intervention to optimize outcomes. Many of these that RVEF normalized in the early post-lung transplantation period.
patients have come into the adult age, and this has created a patient Other early changes included a decrease in RV end-diastolic volume to
population in which the right ventricle is often the center of attention. below normal levels, with persistence at this level even in the late studies.
Despite major progress being made, assessing the right ventricle in RV mass also regressed early, but remained increased in comparison
either the subpulmonary or the systemic circulation remains challeng- to healthy control subjects. The authors concluded that RV anatomic
ing, often requiring a multi-imaging approach. CMR is of use not only normalization occurred later than functional normalization, and RV
in the assessment of the anatomy and physiology of congenital heart mass remained increased. Fayad and colleagues124 also studied RV tagging
disease but also, in some cases, in risk stratification.110,111 in patients with chronic pulmonary hypertension. Regional short-axis
shortening was reduced in patients in comparison with healthy controls,
Pulmonary Hypertension and Lung Transplantation and the greatest reductions in shortening were found in the outflow
The fact that CMR is a radiation-free, highly accurate, and reproducible tract and basal septal region.
technique for quantitative assessment of RV mass and volume makes
it the most appropriate imaging modality for serial studies on the same
patient. Cine CMR has been used to study pulmonary hypertension,112
CONCLUSION
its response to therapy,113,114 and the progression of RV failure in this The role of the right ventricle in acquired and congenital heart disease
condition.115 CMR can accurately estimate mean pulmonary artery is being increasingly recognized. CMR is a highly accurate, reproducible,
pressure in patients with suspected pulmonary hypertension and cal- and versatile technique that is considered the ideal imaging modality
culate pulmonary vascular resistance (PVR) by estimating all major for the comprehensive evaluation of RV dimensions and global and
pulmonary hemodynamic metrics measured at right heart catheteriza- regional function. Accurate quantitation of RV volumes and systolic
tion.116 CMR has been used to confirm the diagnosis of cor pulmonale function provides important diagnostic and prognostic information in
through increased RV mass measurements above 60 g.117 Pulmonary a wide range of conditions. However, further clinical studies are needed
flow patterns are known to be abnormal in pulmonary hypertension,118 to establish standards for the best use of CMR for predicting patient
and this may affect RV afterload, whereas diastolic function has also outcome and the role of serial evaluation in the case of known RV
been found to be abnormal in pulmonary fibrosis by using tricuspid dysfunction.
flow patterns.119 More recently, it has been shown in a study including
58 patients with pulmonary hypertension that the presence of LGE in
the RV insertion point in patients with pulmonary hypertension is a
REFERENCES
marker for more advanced disease and poor prognosis. In addition, A full reference list is available online at ExpertConsult.com
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 468.e1

REFERENCES 19. Lewis WH. Gray’s Anatomy of the Human Body. 20th ed. New York:
Bartleby.com; 2000.
1. Tobler D, Motwani M, Wald RM, et al. Evaluation of a comprehensive 20. Dandel M, Potapov E, Krabatsch T, et al. Load dependency of right
cardiovascular magnetic resonance protocol in young adults late after ventricular performance is a major factor to be considered in decision
the arterial switch operation for d-transposition of the great arteries. making before ventricular assist device implantation. Circulation.
J Cardiovasc Magn Reson. 2014;16:98. 2013;128:S14–S23.
2. Haddad F, Doyle R, Murphy DJ, Hunt SA. Right ventricular function in 21. Ghio S, Gavazzi A, Campana C, et al. Independent and additive
cardiovascular disease, part II: pathophysiology, clinical importance, and prognostic value of right ventricular systolic function and pulmonary
management of right ventricular failure. Circulation. artery pressure in patients with chronic heart failure. J Am Coll Cardiol.
2008;117:1717–1731. 2001;37:183–188.
3. van de Veerdonk MC, Kind T, Marcus JT, et al. Progressive right 22. Cavalcante JL, Rijal S, Althouse AD, et al. Right ventricular function and
ventricular dysfunction in patients with pulmonary arterial prognosis in patients with low-flow, low-gradient severe aortic stenosis.
hypertension responding to therapy. J Am Coll Cardiol. 2011;58: J Am Soc Echocardiogr. 2016;29:325–333.
2511–2519. 23. Gavazzi A, Ghio S, Scelsi L, et al. Response of the right ventricle to acute
4. Galiè N, Humbert M, Vachiery JL, et al. 2015 ESC/ERS Guidelines for pulmonary vasodilation predicts the outcome in patients with advanced
the diagnosis and treatment of pulmonary hypertension: The Joint Task heart failure and pulmonary hypertension. Am Heart J. 2003;145:310–316.
Force for the Diagnosis and Treatment of Pulmonary Hypertension of 24. Dave HH, Buechel ERV, Dodge-Khatami A, et al. Early insertion of a
the European Society of Cardiology (ESC) and the European pulmonary valve for chronic regurgitation helps restoration of
Respiratory Society (ERS): endorsed by: Association for European ventricular dimensions. Ann Thorac Surg. 2005;80:1615–1620.
Paediatric and Congenital Cardiology (AEPC), International Society for 25. Zornoff LA, Skali H, Pfeffer MA, et al; SAVE Investigators. Right
Heart and Lung Transplantation (ISHLT). Eur Heart J. 2016;37:67–119. ventricular dysfunction and risk of heart failure and mortality after
5. Kato S, Sekine A, Kusakawa Y, et al. Prognostic value of cardiovascular myocardial infarction. J Am Coll Cardiol. 2002;39:1450–1455.
magnetic resonance derived right ventricular function in patients with 26. Rumberger JA, Behrenbeck T, Bell MR, et al. Determination of
interstitial lung disease. J Cardiovasc Magn Reson. 2015;17:10. ventricular ejection fraction: a comparison of available imaging
6. Desai RR, Vargas Abello LM, Klein AL, et al. Tricuspid regurgitation and methods: the cardiovascular imaging working group. Mayo Clin Proc.
right ventricular function after mitral valve surgery with or without 1997;72:860–870.
concomitant tricuspid valve procedure. J Thorac Cardiovasc Surg. 27. Schmid E, Hilberath JN, Blumenstock G, et al. Tricuspid annular plane
2013;146:1126–1132. systolic excursion (TAPSE) predicts poor outcome in patients
7. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic undergoing acute pulmonary embolectomy. Heart Lung Vessel.
right ventricular cardiomyopathy/dysplasia: proposed modification of 2015;7:151–158.
the Task Force criteria. Circulation. 2010;121:1533–1541. 28. Kaul S, Tei C, Hopkins JM, Shah PM. Assessment of right ventricular
8. Di Bella G, Siciliano V, Aquaro GD, et al. Right ventricular dysfunction: function using two-dimensional echocardiography. Am Heart J.
an independent and incremental predictor of cardiac deaths late 1984;107:526–531.
after acute myocardial infarction. Int J Cardiovasc Imaging. 2015;31: 29. Rudski LG, Lai WW, Afilalo J, et al. Guidelines for the echocardiographic
379–387. assessment of the right heart in adults: a report from the American
9. Antoni ML, Scherptong RWC, Atary JZ, et al. Prognostic value of right Society of Echocardiography endorsed by the European Association of
ventricular function in patients after acute myocardial infarction treated Echocardiography, a registered branch of the European Society of
with primary percutaneous coronary intervention. Circ Cardiovasc Cardiology, and the Canadian Society of Echocardiography. J Am Soc
Imaging. 2010;3:264–271. Echocardiogr. 2010;23:685–713.
10. Gulati A, Ismail TF, Jabbour A, et al. The prevalence and prognostic 30. Lemarié J, Huttin O, Girerd N, et al. Usefulness of speckle-tracking
significance of right ventricular systolic dysfunction in nonischemic imaging for right ventricular assessment after acute myocardial
dilated cardiomyopathy. Circulation. 2013;128:1623–1633. infarction: a magnetic resonance imaging/echocardiographic
11. Frea S, Pidello S, Bovolo V, et al. Prognostic incremental role of right comparison within the relation between aldosterone and cardiac
ventricular function in acute decompensation of advanced chronic heart remodeling after myocardial infarction study. J Am Soc Echocardiogr.
failure. Eur J Heart Fail. 2016;18:564–572. 2015;28:818–827.
12. Leong DP, Höke U, Delgado V, et al. Right ventricular function and 31. Geyer H, Caracciolo G, Abe H. Assessment of myocardial mechanics
survival following cardiac resynchronisation therapy. Heart. using speckle tracking echocardiography: fundamentals and clinical
2013;99:722–728. applications. J Am Soc Echocardiogr. 2010;23:351–369.
13. Guyton AC. The pulmonary circulation. In: Textbook of Medical 32. Grapsa J, Dawson D, Nihoyannopoulos P. Assessment of right
Physiology. 7th ed. Philadelphia: WB Saunders; 1986:287–294. ventricular structure and function in pulmonary hypertension.
14. Armour JA, Randall WC. Structural basis for cardiac function. Am J J Cardiovasc Ultrasound. 2011;19:115–125.
Physiol. 1970;218:1517–1523. 33. Shimada YJ, Shiota M, Siegel RRJ, Shiota T. Accuracy of right ventricular
15. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular volumes and function determined by three-dimensional
systolic and diastolic function normalized to age, gender and body echocardiography in comparison with magnetic resonance imaging: a
surface area from steady-state free precession cardiovascular magnetic meta-analysis study. J Am Soc Echocardiogr. 2010;23:943–953.
resonance. Eur Heart J. 2006;27:2879–2888. 34. van der Zwaan HB, Geleijnse ML, McGhie JS, et al. Right ventricular
16. Lorenz CH, Walker ES, Morgan VL, Klein SS, Graham TP. Normal quantification in clinical practice: two-dimensional vs. three-
human right and left ventricular mass, systolic function and gender dimensional echocardiography compared with cardiac magnetic
differences by cine magnetic resonance imaging. J Cardiovasc Magn resonance imaging. Eur J Echocardiogr. 2011;12:656–664.
Reson. 1999;1:7–22. 35. Park JB, Lee SP, Lee JH, et al. Quantification of right ventricular volume
17. Rogers JH, Bolling SF. The tricuspid valve: current perspective and and function using single-beat three-dimensional echocardiography: a
evolving management of tricuspid regurgitation. Circulation. validation study with cardiac magnetic resonance. J Am Soc
2009;119:2718–2725. Echocardiogr. 2016;29:392–401.
18. Lancellotti P, Moura L, Pierard LA, et al; European Association of 36. Hornung TS, Anagnostopoulos C, Bhardwaj P, et al. Comparison of
Echocardiography. European Association of Echocardiography equilibrium radionuclide ventriculography with cardiovascular magnetic
recommendations for the assessment of valvular regurgitation. Part 2: resonance for assessing the systemic right ventricle after Mustard or
mitral and tricuspid regurgitation (native valve disease). Eur Heart J. Senning procedures for complete transposition of the great arteries. Am
2010;11:307–332. J Cardiol. 2003;92:640–643.
468.e2 SECTION IV  Right Ventricular and Congenital Heart Disease

37. Pietras RJ, Kondos GT, Kaplan D, Lam W. Comparative angiographic board of trustees task force on standardized post processing.
right and left ventricular volumes. Am Heart J. 1985;109:321–326. J Cardiovasc Magn Reson. 2013;15:35.
38. Dehmer GJ, Firth DG, Hillis LD, Nicod P, Willerson JT, Lewis SE. Non 57. Longmore DB, Klipstein RH, Underwood SR, et al. Dimensional
geometric determination of right ventricular volumes from equilibrium accuracy of magnetic resonance in studies of the heart. Lancet.
blood pool scans. Am J Cardiol. 1982;49:79–84. 1985;1:1360–1362.
39. Wang L, Zhang Y, Yan C, et al. Evaluation of right ventricular volume 58. Farber NJ, Reddy ST, Doyle M, et al. Ex vivo cardiovascular magnetic
and ejection fraction by gated 18F-FDG PET in patients with resonance measurements of right and left ventricular mass compared
pulmonary hypertension: comparison with cardiac MRI and CT. J Nucl with direct mass measurement in excised hearts after transplantation: a
Cardiol. 2013;20:242–252. first human SSFP comparison. J Cardiovasc Magn Reson. 2014;16:74.
40. Dercle L, Ouali M, Pascal P, et al. Gated blood pool SPECT: the 59. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular
estimation of right ventricular volume and function is algorithm systolic and diastolic function normalized to age, gender and body
dependent in a clinical setting. J Nuclear Cardiology. 2015;22:483–492. surface area from steady state free precession cardiovascular magnetic
41. Schmermund A, Rensing BJ, Sheedy PF, Rumberger JA. Reproducibility resonance. Eur Heart J. 2006;27:2879–2888.
of right and left ventricular volume measurements by electron-beam CT 60. Mercer-Rosa L, Yang W, Kutty S, Rychik J, Fogel M, Goldmuntz E.
in patients with congestive heart failure. Int J Card Imaging. Quantifying pulmonary regurgitation and right ventricular function in
1998;14:201–209. surgically repaired tetralogy of Fallot: a comparative analysis of
42. Maffei E, Messalli G, Martini C, et al. Left and right ventricle assessment echocardiography and magnetic resonance imaging. Circ Cardiovasc
with cardiac CT: validation study vs. cardiac MR. Eur Radiol. Imaging. 2012;5:637–643.
2012;22:1041–1049. 61. Prasad SK, Soukias N, Hornung T, et al. Role of magnetic resonance
43. Sugeng L, Mor-Avi V, Weinert L, et al. Multimodality comparison of angiography in the diagnosis of major aortopulmonary collateral
quantitative volumetric analysis of the right ventricle. JACC Cardiovasc arteries and partial anomalous pulmonary venous drainage. Circulation.
Imaging. 2010;3:10–18. 2004;109:207–214.
44. Pignatelli RH, McMahon CJ, Chung T, Vick GW 3rd. Role of 62. Steeden JA, Pandya B, Tann O, Muthurangu V. Free breathing contrast-
echocardiography versus MRI for the diagnosis of congenital heart enhanced time-resolved magnetic resonance angiography in pediatric
disease. Curr Opin Cardiol. 2003;18:357–365. and adult congenital heart disease. J Cardiovasc Magn Reson. 2015;
45. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ. 17:38.
Interstudy reproducibility of right ventricular volumes, function, and 63. Fredriksson AG, Svalbring E, Eriksson J, et al. 4D flow MRI can detect
mass with cardiovascular magnetic resonance. Am Heart J. subtle right ventricular dysfunction in primary left ventricular disease.
2004;147:218–223. J Magn Reson Imaging. 2016;43:558–565.
46. Tandri H, Castillo E, Ferrari VA, et al. Magnetic resonance imaging of 64. Fogel MA, Sundareswaran KS, de Zelicourt D, et al. Power loss and right
arrhythmogenic right ventricular dysplasia: sensitivity, specificity, and ventricular efficiency in patients after tetralogy of Fallot repair with
observer variability of fat detection versus functional analysis of the pulmonary insufficiency: clinical implications. J Thorac Cardiovasc Surg.
right ventricle. J Am Coll Cardiol. 2006;48(11):2277–2284. 2012;143:1279–1285.
47. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular 65. Sen-Chowdhry S, Prasad SK, McKenna WJ. Arrhythmogenic right
systolic and diastolic function normalized to age, gender, and body ventricular cardiomyopathy with fibrofatty atrophy, myocardial oedema,
surface area from steady-state free precession cardiovascular magnetic and aneurysmal dilation. Heart. 2005;91:784.
resonance. Eur Heart J. 2006;27:2879–2888. 66. Broberg CS, Prasad SK, Carr C, Babu-Narayan SV, Dimopoulos K,
48. Voit D, Zhang S, Unterberg-Buchwald C, Sohns JM, Lotz J, Frahm J. Gatzoulis MA. Myocardial fibrosis in Eisenmenger syndrome: a
Real-time cardiovascular magnetic resonance at 1.5 T using balanced descriptive cohort study exploring associations of late gadolinium
SSFP and 40 ms resolution. J Cardiovasc Magn Reson. 2013;15:79. enhancement with clinical status and survival. J Cardiovasc Magn Reson.
49. Bellenger NG, Gatehouse PD, Rajappan K, Keegan J, Firmin DN, Pennell 2014;16:32.
DJ. Left ventricular quantification in heart failure by CMR using 67. Stirrat J, Rajchl M, Bergin L, Patton DJ, Peters T, White JA. High-
prospective respiratory navigator gating: comparison with breath-hold resolution 3-dimensional late gadolinium enhancement scar imaging in
acquisition. J Magn Reson Imaging. 2000;11:411–417. surgically corrected tetralogy of Fallot: clinical feasibility of volumetric
50. Helbing WA, Rebergen SA, Maliepaard C, et al. Quantification of right quantification and visualization. J Cardiovasc Magn Reson. 2014;16:76.
ventricular function with magnetic resonance imaging in children with 68. Hangartner JRW, Marley NJ, Whitehead A, Thomas AC, Davies MJ. The
normal hearts and with congenital heart disease. Am Heart J. assessment of cardiac hypertrophy at autopsy. Histopathology.
1995;130:828–837. 1985;9:1295–1306.
51. Jauhiainen T, Jarvinen VM, Hekali PE, Poutanen VP, Penttila A, Kupari 69. Schvartzman PR, Fuchs FD, Mello AG, Coli M, Schvartzman M, Moreira
M. MR gradient echo volumetric analysis of human cardiac casts: focus LB. Normal values of echocardiographic measurements: a population-
on the right ventricle. J Comput Assist Tomogr. 1998;22:899–903. based study. Arq Bras Cardiol. 2000;75:107–114.
52. Clarke CJ, Gurka MJ, Norton PT, Kramer CM, Hoyer AW. Assessment of 70. Tamborini G, Marsan NA, Gripari P, et al. Reference values for right
the accuracy and reproducibility of RV volume measurements by CMR ventricular volumes and ejection fraction with real-time three-
in congenital heart disease. JACC Cardiovasc Imaging. 2012;5:28–37. dimensional echocardiography: evaluation in a large series of normal
53. Alfakih K, Plein S, Bloomer T, Jones T, Ridgway J, Sivananthan M. subjects. J Am Soc Echocardiogr. 2010;23:109–115.
Comparison of right ventricular volume measurements between axial 71. Pietras RJ, Kondos GT, Kaplan D, Lam W. Comparative angiographic
and short axis orientation using steady-state free precession magnetic right and left ventricular volumes. Am Heart J. 1985;109:321–326.
resonance imaging. J Magn Reson Imaging. 2003;18:25–32. 72. Kjaer A, Lebech AM, Hesse B, Petersen CL. Right-sided cardiac function
54. Grothues F, Moon JC, Bellenger NG, Smith GS, Klein HU, Pennell DJ. in healthy volunteers measured by first-pass radionuclide
Interstudy reproducibility of right ventricular volumes, function and ventriculography and gated blood-pool SPECT: comparison with cine
mass with cardiovascular magnetic resonance. Am Heart J. MRI. Clin Physiol Funct Imaging. 2005;25:344–349.
2004;147:218–223. 73. Lorenz CH, Walker ES, Morgan VL, Klein SS, Graham TP Jr. Normal
55. van der Bom T, Romeih S, Groenink M, et al. Evaluating the systemic human right and left ventricular mass, systolic function, and gender
right ventricle by cardiovascular magnetic resonance: short axis or axial differences by cine magnetic resonance imaging. J Cardiovasc Magn
slices? Congenit Heart Dis. 2015;10:69–77. Reson. 1999;1:7–21.
56. Schulz-Menger J, Bluemke DA, Bremerich J, et al. Standardized image 74. Rominger MB, Bachmann GF, Pabst W, Rau WS. Right ventricular
interpretation and post processing in cardiovascular magnetic volumes and ejection fraction with fast cine MR imaging in breath-hold
resonance: Society for Cardiovascular Magnetic Resonance (SCMR) technique: applicability, normal values from 52 volunteers and
CHAPTER 39  CMR Assessment of Right Ventricular Anatomy and Function 468.e3

evaluation of 352 adult cardiac patients. J Magn Reson Imaging. 94. de Groote P, Millaire A, Foucher-Hossein C, et al. Right ventricular
1999;10:908–918. ejection fraction is an independent predictor of survival in patients with
75. Alfakih K, Plein S, Thiele H, Jones T, Ridgway JP, Sivananthan MU. moderate heart failure. J Am Coll Cardiol. 1998;32:948–954.
Normal human left and right ventricular dimensions for MRI as 95. Bourantas CV, Loh HP, Bragadeesh T, et al. Relationship between right
assessed by turbo gradient echo and steady-state free precession imaging ventricular volumes measured by cardiac magnetic resonance imaging
sequences. J Magn Reson Imaging. 2003;17:323–329. and prognosis in patients with chronic heart failure. Eur J Heart Fail.
76. Hudsmith LE, Petersen SE, Francis JM, Robson MD, Neubauer S. 2011;13:52–60.
Normal human left and right ventricular and left atrial dimensions 96. Gulati A, Ismail TF, Jabbour A, et al. The prevalence and prognostic
using steady state free precession magnetic resonance imaging. significance of right ventricular systolic dysfunction in nonischemic
J Cardiovasc Magn Reson. 2005;7:775–782. dilated cardiomyopathy. Circulation. 2013;128:1623–1633.
77. Maffei E, Messalli G, Martini C, et al. Left and right ventricle assessment 97. Zehender M, Kasper W, Kauder E, et al. Right ventricular infarction as
with cardiac CT: validation study vs. cardiac MR. Eur Radiol. an independent predictor of prognosis after acute inferior myocardial
2012;22:1041–1049. infarction. N Engl J Med. 1993;328:981–988.
78. Maceira AM, Prasad SK, Khan M, Pennell DJ. Reference right ventricular 98. Kinch JW, Ryan TJ. Right ventricular infarction. N Engl J Med.
systolic and diastolic function normalized to age, gender and body 1994;330:1211–1217.
surface area from steady state free precession cardiovascular magnetic 99. Bueno H, Lopez-Palop R, Bermejo J, Lopez-Sendon JL, Delcan JL.
resonance. Eur Heart J. 2006;27:2879–2888. Inhospital outcome of elderly patients with acute inferior myocardial
79. Foppa M, Arora G, Gona P, et al. Right ventricular volumes and systolic infarction and right ventricular involvement. Circulation.
function by cardiac magnetic resonance and the impact of sex, age, and 1997;96:436–441.
obesity in a longitudinally followed cohort free of pulmonary and 100. Moazami N, Hill L. Right ventricular dysfunction in patients with acute
cardiovascular disease: the Framingham Heart Study. Circ Cardiovasc inferior MI: role of RV mechanical support. Thorac Cardiovasc Surg.
Imaging. 2016;9:e003810. 2003;51:290–292.
80. Young AA, Cowan BR, Occleshaw CJ, Oxenham HC, Gentles TL. 101. Kinch JW, Ryan TJ. Right ventricular infarction. N Engl J Med.
Temporal evolution of left ventricular strain late after repair of 1994;330:1211–1217.
coarctation of the aorta using 3D MR tissue tagging. J Cardiovasc Magn 102. Bowers TR, O’Neill WW, Grines C, Pica MC, Safian RD, Goldstein JA.
Reson. 2002;4:233–243. Effect of reperfusion on biventricular function and survival after right
81. Menteer J, Weinberg PM, Fogel MA. Quantifying regional right ventricular infarction. N Engl J Med. 1998;338:933–940.
ventricular function in tetralogy of Fallot. J Cardiovasc Magn Reson. 103. Larose E, Ganz P, Reynolds HG, et al. Right ventricular dysfunction
2005;7:753–761. assessed by cardiovascular magnetic resonance imaging predicts poor
82. Klein SS, Graham TP, Lorenz CH. Noninvasive delineation of normal prognosis late after myocardial infarction. J Am Coll Cardiol.
right ventricular contractile motion with MRI myocardial tagging. Ann 2007;49:855–862.
Biomed Eng. 1998;26:756–763. 104. Mast TP, Teske AJ, vd Heijden JF, et al. Left ventricular involvement in
83. Naito H, Arisawa J, Yamagami H, Kozuka T, Tamura S. Assessment of arrhythmogenic right ventricular dysplasia/cardiomyopathy assessed by
right ventricular regional contraction and comparison with left ventricle echocardiography predicts adverse clinical outcome. J Am Soc
in normal humans: a cine magnetic resonance study with presaturation Echocardiogr. 2015;28:1103–1113.e9.
myocardial tagging. Br Heart J. 1995;74:186–191. 105. Deleted in review.
84. Fayad ZA, Kraitchman DL, Ferrari VA, Axel L. Right ventricular regional 106. ACCF/ACR/AHA/NASCI/SCMR 2010 Expert Consensus Document on
function in normal subjects using magnetic resonance tissue tagging. In: Cardiovascular Magnetic Resonance. A report of the American College
Book of Abstracts. Berkeley, CA: Society of Magnetic Resonance; of Cardiology Foundation Task Force on Expert Consensus Documents.
1994:1504. J Am Coll Cardiol. 2010;55:2614–2662.
85. Young AA, Cowan BR, Occleshaw CJ, Oxenham HC, Gentles TL. 107. Tandri H, Calkins H, Nasir K, et al. Magnetic resonance imaging
Temporal evolution of left ventricular strain late after repair of findings in patients meeting Task Force criteria for arrhythmogenic right
coarctation of the aorta using 3D MR tissue tagging. J Cardiovasc Magn ventricular dysplasia. J Cardiovasc Electrophysiol. 2003;14:476–482.
Reson. 2002;4:233–243. 108. Bluemke DA, Krupinski EA, Ovitt T, et al. MR imaging of
86. Klein SS, Graham TP, Lorenz CH. Noninvasive delineation of normal arrhythmogenic right ventricular cardiomyopathy: morphologic findings
right ventricular contractile motion with MRI myocardial tagging. Ann and interobserver reliability. Cardiology. 2003;99:153–162.
Biomed Eng. 1998;26:756–763. 109. Rebergen SA, Ottenkamp J, Doornbos J, van der Wall EE, Chin JG, de
87. Auger DA, Zhong X, Epstein FH, Spottiswoode BS. Mapping right Roos A. Postoperative pulmonary flow dynamics after Fontan surgery:
ventricular myocardial mechanics using 3D cine DENSE cardiovascular assessment with nuclear magnetic resonance velocity mapping. J Am
magnetic resonance. J Cardiovasc Magn Reson. 2012;14:4. Coll Cardiol. 1993;21:123–131.
88. Prati G, Vitrella G, Allocca G, et al. Right ventricular strain and 110. Babu-Narayan SV, Goktekin O, Moon JC, et al. Late gadolinium
dyssynchrony assessment in arrhythmogenic right ventricular enhancement cardiovascular magnetic resonance of the systemic
cardiomyopathy. Cardiac magnetic resonance feature-tracking study. right ventricle in adults with previous atrial redirection surgery for
Circ Cardiovasc Imaging. 2015;8:e003647. transposition of the great arteries. Circulation. 2005;111:2091–
89. Brieke A, DeNofrio D. Right ventricular dysfunction in chronic dilated 2098.
cardiomyopathy and heart failure. Coron Artery Dis. 2005;16:5–11. 111. Babu-Narayan SV, Kilner PJ, Li W, et al. Ventricular fibrosis suggested by
90. Meluzin J, Spinarova L, Hude P, et al. Prognostic importance of various cardiovascular magnetic resonance in adults with repaired tetralogy of
echocardiographic right ventricular functional parameters in patients Fallot and its relationship to adverse markers of clinical outcome.
with symptomatic heart failure. J Am Soc Echocardiogr. 2005;18:435–444. Circulation. 2006;113:405–413.
91. de Groote P, Millaire A, Foucher-Hossein C, et al. Right ventricular 112. Boxt LM, Katz J, Kolb T, Czegledy FP, Barst RJ. Direct quantitation of
ejection fraction is an independent predictor of survival in patients with right and left ventricular volumes with nuclear magnetic resonance
moderate heart failure. J Am Coll Cardiol. 1998;32:948–954. imaging in patients with primary pulmonary hypertension. J Am Coll
92. Mohammed SF, Hussain I, AbouEzzeddine OF, et al. Right ventricular Cardiol. 1992;19:1508–1515.
function in heart failure with preserved ejection fraction: a community- 113. Wilkins MR, Paul GA, Strange JW, et al. Sildenafil versus Endothelin
based study. Circulation. 2014;130:2310–2320. Receptor Antagonist for Pulmonary Hypertension (SERAPH) study. Am
93. Di Salvo TG, Mathier M, Semigran MJ, Dec GW. Preserved right J Respir Crit Care Med. 2005;171:1292–1297.
ventricular ejection fraction predicts exercise capacity and survival in 114. Peacock AJ, Crawley S, McLure L, et al. Pulmonary arterial hypertension.
advanced heart failure. J Am Coll Cardiol. 1995;25:1143–1153. Changes in right ventricular function measured by cardiac magnetic
468.e4 SECTION IV  Right Ventricular and Congenital Heart Disease

resonance imaging in patients receiving pulmonary arterial 120. Freed BH, Gomberg-Maitland M, Chandra S, et al. Late gadolinium
hypertension–targeted therapy. The EURO-MR study. Circ Cardiovasc enhancement cardiovascular magnetic resonance predicts clinical
Imaging. 2014;7:107–114. worsening in patients with pulmonary hypertension. J Cardiovasc Magn
115. van de Veerdonk MC, Kind T, Marcus JT, et al. Progressive right Reson. 2012;14:11.
ventricular dysfunction in patients with pulmonary arterial 121. Moulton JM, Creswell LL, Ungacta FF, Downing SW, Szabo BA, Pasque
hypertension responding to therapy. J Am Coll Cardiol. 2011;58: MK. Magnetic resonance imaging provides evidence for remodeling of
2511–2519. the right ventricle after single-lung transplantation for pulmonary
116. Swift AJ, Rajaram S, Hurdman J, et al. Noninvasive estimation of PA hypertension. Circulation. 1996;94(suppl II):312–319.
pressure, flow, and resistance with CMR imaging: derivation and 122. Lorenz CH, Loyd JE, Klein SS, et al. Characterization of different time
prospective validation study from the ASPIRE registry. JACC Cardiovasc courses of left and right ventricular recovery after lung transplantation.
Imaging. 2013;6:1036–1047. J Am Coll Cardiol. 1997;29(suppl A):23A.
117. Pattynama PMT, Willems LNA, Smit AH, van der Wall EE, de Roos A. 123. Frist WH, Lorenz CH, Walker ES, et al. MRI complements standard
Early diagnosis of cor pulmonale with MR imaging of the right assessment of right ventricular function after lung transplantation. Ann
ventricle. Radiology. 1992;182:375–379. Thorac Surg. 1995;60:268–271.
118. Bogren HG, Klipstein RH, Mohiaddin RH, et al. Pulmonary artery 124. Fayad ZA, Ferrari VA, Kraitchman DL, et al. Right ventricular regional
distensibility and blood flow patterns: a magnetic resonance study of function using MR tagging: normals versus chronic pulmonary
normal subjects and of patients with pulmonary arterial hypertension. hypertension. Magn Reson Med. 1998;39:116–123.
Am Heart J. 1989;118:990–999.
119. Kroft LJ, Simons P, van Laar JM, de Roos A. Patients with pulmonary
fibrosis: cardiac function assessed with MR imaging. Radiology.
2000;216:464–471.
40 
Simple and Complex Congenital Heart
Disease: Infants and Children
Andrew J. Powell and Tal Geva

The role of cardiovascular magnetic resonance (CMR) in the evaluation most parameters, particularly in infants and children but also in unusu-
of infants and children with congenital heart disease is now widely ally large or small adults.1 Several investigators have demonstrated that
accepted as a result of ongoing technologic advances and the growing linear dimensions (e.g., ascending aorta diameter) are best adjusted to
realization of its clinical value. Improvements in CMR hardware and the square root of BSA, area measurements to BSA, and volumetric
the development of new, highly efficient/faster imaging methods, have measurements to BSA raised to the 1.3 to 1.4 power.2,3 Thus for example,
allowed for sufficient spatial and temporal resolution to comprehensively one cannot simply apply to children a normative range for ventricular
evaluate the complex cardiac anatomy and function in pediatric patients volume indexed to BSA that was derived in adults. CMR-based norma-
despite their small body size and rapid heart rate. Although transthoracic tive data from healthy children are available, although they are often
echocardiography (TTE) provides much of the necessary noninvasive limited by a smaller than ideal sample size, particularly in the youngest
diagnostic information for most patients in this age group, CMR offers age range.4–12 Furthermore, it is essential to apply the same measure-
an important alternative with added value in several circumstances: (1) ment technique (e.g., whether to include papillary muscles or myocardial
when TTE fails to provide the required diagnostic information; (2) trabeculations in the blood pool) as that which was used to derive the
when clinical assessment and diagnostic tests are inconsistent; (3) as normative data.
an alternative to diagnostic invasive x-ray cardiac catheterization with
its associated higher risk and cost; and (4) to obtain diagnostic infor- ATRIAL SEPTAL DEFECTS AND OTHER
mation for which CMR offers unique advantages (e.g., flow measure-
ments, fibrosis, parametric mapping).
INTERATRIAL COMMUNICATIONS
This chapter reviews the anatomy, clinical management, and CMR Anatomically, five different defects can result in an interatrial shunt
evaluation of common congenital heart disease lesions involving infants (Fig. 40.1):
and children. Patient preparation, sedation, and monitoring strategies 1. A patent foramen ovale is bordered on the left by septum primum
for children undergoing CMR are discussed in Chapter 12. and by the superior limbic band of the fossa ovalis (septum secun-
dum) on the right. A patent foramen ovale is seen in almost all
PRINCIPLES OF CARDIOVASCULAR MAGNETIC newborns and decreases in frequency with age.
RESONANCE EVALUATION IN CONGENITAL 2. A secundum atrial septal defect (ASD) (Fig. 40.2) is the most common
cause of an atrial level shunt after patent foramen ovale. Usually
HEART DISEASE the defect is caused by deficiency of septum primum, but rarely, it
Preexamination planning is crucial given the wide array of CMR imaging results from a deficiency of septum secundum.
sequences available and the often-complex nature of the clinical, ana- 3. A primum ASD is a variant of incomplete common atrioventricular
tomic, and functional issues in patients with congenital heart disease. canal and is the third most common interatrial communication.
It is essential to perform a careful review of the patient’s medical history, This defect involves the septum of the atrioventricular canal and is
including details of all prior cardiovascular surgical procedures, inter- almost always associated with a cleft anterior mitral valve leaflet.
ventional catheterizations, diagnostic test results, and current clinical Any associated defect within the fossa ovalis (e.g., secundum ASD)
status. As with TTE and invasive cardiac catheterization, CMR exami- is regarded as a separate abnormality.
nation of congenital heart disease is a dynamic diagnostic procedure 4. A sinus venosus septal defect results from deficiency of the sinus
that is optimally performed with continuous review and interpretation venosus septum, which separates the pulmonary veins from the
of the data by the supervising physician. Unexpected findings or sub- systemic veins and the sinus venosus component of the right atrium
optimal image quality often require adjustment of the examination (RA) (Fig. 40.3). Most commonly, a sinus venosus defect is between
protocol, imaging planes, techniques, and sequence parameters. Reliance the right upper pulmonary vein and the cardiac end of the superior
on standardized protocols and postexamination review alone in these vena cava. Rarely, the defect involves the right lower and/or middle
patients may result in incomplete or even erroneous interpretation, or pulmonary veins and the inferior aspect of the RA near its junc-
the need to repeat the CMR examination. tion with the inferior vena cava. From an anatomic standpoint,
One of the strengths of CMR is its ability to accurately and precisely a sinus venosus defect is not an ASD because it does not allow
measure ventricular and vascular structures. Care must be taken, however, direct communication between the left and right atria. Instead, the
when determining whether these measurements fall within the normal interatrial flow travels between the left atrium (LA), one or more
range because adjustment for the variation in body size remains chal- of the pulmonary veins, the sinus venosus septal defect, the supe-
lenging. Simply dividing by body surface area (BSA) is inadequate for rior (or inferior) vena cava, and the RA. The defect usually allows

469
470 SECTION IV  Right Ventricular and Congenital Heart Disease

pulmonary vein flow to drain to the RA through the defect as well. unroofed, the left superior vena cava will drain to the LA. The
Patients with sinus venosus defects may have additional accessory coexistence of a coronary sinus septal defect and persistent left
right upper pulmonary veins connecting to the superior vena cava or superior vena cava is termed Raghib syndrome, and may result in
azygous vein. cyanosis.13
5. A coronary sinus septal defect is a rare type of interatrial commu- Regardless of the specific anatomic type, the amount of shunting
nication in which the septum between the coronary sinus and the through an interatrial communication is determined by the defect size
LA is either partially or completely unroofed, allowing the RA and and relative compliance of the right ventricle (RV) and left ventricle
the LA to communicate through the defect and the coronary sinus (LV). Over the first few months of life, RV compliance typically rises,
orifice. Sometimes there is also a persistent left superior vena cava leading to an increasing left-to-right shunt. During adulthood, LV com-
draining to the coronary sinus. If the coronary sinus is completely pliance normally decreases further, augmenting the left-to-right flow.
Shunt flow through the right heart and lungs leads to dilation of the
RA, RV, pulmonary arteries, and pulmonary veins. Up to 5% to 10%
of patients with a significant left-to-right shunt may develop pulmonary
vascular disease by adulthood, leading to pulmonary artery hyperten-
sion. Adults with unrepaired atrial level defects are also at risk for
exercise intolerance, atrial arrhythmias, and paradoxical emboli.
In general, current practice is to refer patients for interatrial defect
closure if the patient is symptomatic or the defect results in a significant
left-to-right shunt. Evidence for the latter includes a defect diameter
>5 mm, RV cavity dilation, flattening of the interventricular septum
ASD 1° in diastole (caused by elevated RV diastolic pressure from the volume
Sinus load), and a pulmonary-to-systemic flow (Qp/Qs) ratio >1.5 to 2.
venosus
defect Although a patent foramen ovale typically produces only a small shunt,
closure may be indicated when there is a history consistent with a
paradoxical embolus.14,15 All of these defect types can be closed surgi-
cally with a very low mortality and morbidity in centers with expertise.
Over the past two decades, transcatheter treatment for secundum ASDs
and patent foramen ovale by occluding them with various devices has
become available at specialized centers.
With regard to surgical technique, secundum ASDs are either closed
primarily or with a patch. For primum ASDs, the defect is closed with
a patch and, in the majority of cases, the associated mitral valve cleft
ASD 2° is partially sutured. Sinus venosus ASDs can often be closed by placing
FIG. 40.1  Anatomic types of atrial communications (see text for details). a patch to reconstruct the missing portion of the sinus venosus septum,
Atrial septal defect: ASD 10 denotes primum atrial septal defect; ASD thereby eliminating interatrial and right pulmonary vein to right atrial
20 denotes secundum atrial septal defect. flow (“single patch repair”). A second patch may be placed anteriorly

SVC RPA

LA

RA

IVC
A B
FIG. 40.2  Secundum atrial septal defect. (A) Oblique balanced steady-state free precession (bSSFP) cine
showing a centrally located defect (arrow). (B) Color-coded velocity-encoded cine showing flow from the left
atrium (LA) to the right atrium (RA) through the defect. IVC, Inferior vena cava; RPA, right pulmonary artery;
SVC, superior vena cava. (From Geva T, Powell AJ. Magnetic resonance imaging. In: Allen HD, Shaddy RE,
Penny DJ, Feltes FT, Cetta F, eds. Moss and Adams’ Heart Disease in Infants, Children, and Adolescents.
9th ed. Philadelphia: Wolters Kluwer; 2016:373–412.)
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 471

SVC
RPA

RUPV SVC Ao

LA RUPV

RA LA

A B C
FIG. 40.3  Sinus venous atrial septal defect. (A) Volume rendered contrast-enhanced three-dimensional magnetic
resonance angiography showing several pulmonary veins from the right upper lobe draining into the superior
vena cava. (B) Cine balance steady-state free precession (bSSFP) in the sagittal plane showing the defect
(*) between the right upper pulmonary vein (RUPV) and the superior vena cava (SVC). (C) Cine bSSFP in the
axial plane showing the defect (*) between the RUPV and the SVC. The arrow points to the left atrial orifice
of the RUPV. A left-to-right shunt results from drainage of the RUPV to the SVC and from left atrial blood
entering the right atrium through the orifice of the RUPV (arrow) and the unroofed wall between the RUPV
and the SVC (*). Ao, Ascending aorta; LA, left atrium. (From Wald RM, Powell AJ. Congenital heart disease.
In: Kwong R, ed. Cardiovascular Magnetic Resonance Imaging. Totowa, NJ: Humana Press; 2008:537–568.)

on the superior vena cava to enlarge it (“two patch repair”). In some and left (Qs) ventricular stroke volumes measured from the short-axis
cases, especially when a right upper pulmonary vein drains relatively cine ventricular stack. In clinical practice, it is recommended to measure
high to the superior vena cava, the superior vena cava is transected the Qp:Qs ratio by both of these methods and check the data for con-
superior to the anomalous veins and the distal caval end anastomosed sistency. In addition, review of the ventricular short-axis cine images
to the right atrial appendage (Warden procedure). The sinus venosus also allows one to make a qualitative estimate of RV systolic pressure
septal defect is then closed in such a way that the proximal superior based on the configuration of the ventricular septum. The septal geom-
vena cava and anomalous veins drain to the LA. Short-term outcomes etry is concave toward the RV when the RV-to-LV pressure ratio is low
for all three techniques are excellent, but caval or pulmonary venous and assumes a flat configuration, or even a concave shape toward the
obstruction may develop over time. For a coronary sinus septal defect, LV, as the RV-to-LV pressure ratio increases. Interpretation of the septal
the os of the coronary sinus is usually patched closed. If a left superior configuration may be confounded by factors such as dysynchronous
vena cava is present, it is redirected to the right atrial side either through contraction, conduction delay (e.g., right or left bundle branch block
ligation when an adequate left innominate vein is present, or via a baffle and preexcitation), and a high LV pressure.
to the RA. The goals of the CMR examination include (1) delineating ASD
number, location, size, and rim length; (2) evaluating pulmonary venous
Cardiovascular Magnetic Resonance return; (3) estimating the RV systolic pressure based on septal configu-
Before Closure ration; (4) measuring the Qp:Qs ratio; and (5) quantifying biventricular
In children, TTE is the primary imaging technique for the evaluation volumes, mass, and function. These goals can be achieved by the fol-
of ASDs and is usually sufficient for clinical decision making. However, lowing protocol26:
CMR can be helpful in older patients, usually adolescents and adults, • Cine bSSFP: stack of contiguous thin slices in an axial (sinus venosus
with a known or suspected ASD and inconclusive clinical or TTE find- defect) or four-chamber plane (secundum ASD) to span the inter-
ings. CMR is also a useful noninvasive alternative to transesophageal atrial septum and cardiac ends of the vena cavae.
echocardiography for determining which patients are candidates for • Cine bSSFP: stack of contiguous thin slices in an oblique sagittal
percutaneous device closure rather than surgery.16 plane perpendicular to the interatrial septum to span the septum
Studies have shown that cine balanced steady-state free precession and entry of the vena cavae (Fig. 40.4).
(bSSFP) imaging and velocity-encoded cine imaging are able to accu- • Velocity-encoded cine: 1 to 3 contiguous slices positioned parallel
rately identify ASD size, shape, proximity to adjacent structures, and to the interatrial septum with through-plane velocity encoding to
associated findings (e.g., anomalous pulmonary venous return).16–20 obtain an en face view of the defect (see Fig. 40.4).
Similarly, CMR is useful for diagnosis and preoperative planning in • Velocity-encoded cine: stack of contiguous thin slices in a four-
patients with sinus venosus ASDs.21,22 Moreover, multiple reports have chamber and/or in an oblique sagittal plane to span the interatrial
demonstrated that calculation of the Qp:Qs ratio by measuring flow septum and with velocity encoding in the direction of ASD flow to
in the main pulmonary artery (Qp) and ascending aorta (Qs) agrees visualize the ASD (see Fig. 40.2B).
closely with invasive catheterization-based oximetry measurements of • Cine bSSFP: ventricular long-axis planes and a ventricular short-axis
Qp:Qs.23–25 In the absence of significant valve regurgitation or an addi- stack from base-to-apex for quantitative assessment of ventricular
tional shunt, the Qp:Qs ratio can also be derived from the right (Qp) volumes and ejection fraction (EF).
472 SECTION IV  Right Ventricular and Congenital Heart Disease

A Axial B Axial Oblique sagittal

C Axial Oblique sagittal ASD en face PC


FIG. 40.4  Atrial septal defect (ASD) imaging protocol. (A) Axial cine balanced steady-state free precession
(bSSFP) image showing a large secundum ASD (white arrow). (B) The axial bSSFP image is used to plan a
stack of oblique sagittal cine bSSFP images to visualize the ASD and the superior and inferior defect margins.
(C) The axial and oblique sagittal images are used together to plan a stack of velocity-encoded phase contrast
(PC) cine images to visualize the ASD flow en face. This provides insight into the oval shape of the defect
and may demonstrate additional ASDs. (From Fratz S, Chung T, Greil GF, et al. Guidelines and protocols for
cardiovascular magnetic resonance in children and adults with congenital heart disease: SCMR expert con-
sensus group on congenital heart disease. J Cardiovasc Magn Reson. 2013;15:51.)

• Contrast-enhanced three-dimensional (3D) magnetic resonance


angiography (MRA) and/or electrocardiogram (ECG) and respira-
tory navigator-gated 3D bSSFP imaging of the thorax.
• Blood flow measurements: ascending aorta (Qs) and main pulmonary
artery (Qp); optional, superior vena cava and descending aorta at
the diaphragm (Qs), and left and right pulmonary arteries (Qp).

After Closure Subpulmonary


The CMR examination goals after ASD closure are similar to the pre-
procedure ones. Additional aims in patients who have undergone trans- Membranous
catheter device closure of an ASD include excluding device malposition,
device interference with the atrioventricular valves and venous blood
flow, and thrombus formation. Patients who have undergone a repair
of a sinus venosus defect are at risk for superior vena cava and right
pulmonary vein obstruction. Mitral regurgitation from a residual mitral AV canal
valve cleft is common after primum ASD repair, and should be assessed type
quantitatively. All of these aims can be accomplished using the imaging
protocol above before closure.

VENTRICULAR SEPTAL DEFECTS


Muscular
A ventricular septal defect (VSD) is a communication between the RV FIG. 40.5  Anatomic types of ventricular septal defects (see text for
and LV through an opening in the interventricular septum. Several details). AV, Atrioventricular.
VSD anatomic classification systems are in use. Fig. 40.5 shows one
such system modified from Van Praagh et al.,27 which includes the
following: (1) defects at the junction between the conal septum and The natural history of a VSD relates to the size and location of the
the muscular septum bordering the membranous septum (referred defect. Defects in the membranous or muscular septum often become
to as membranous defects) or associated with malalignment of the smaller over time and may spontaneously close. In contrast, malalign-
conal septum (conoventricular defects) (Fig. 40.6); (2) defects in the ment conoventricular defects, outlet septum defects, and atrioventricular
muscular septum (called muscular or trabecular defects) (Fig. 40.7); canal-type defects are usually large and rarely spontaneously close.
(3) defects in the inlet septum (known as atrioventricular canal-type Consequently, these patients often undergo surgical closure in infancy.
or inlet defects); and (4) defects in the outlet septum (variably called Venturi effects associated with VSDs in the membranous or outlet septum
outlet, doubly committed subarterial, subpulmonary, conal septal, or may cause aortic valve leaflets (usually the right coronary leaflet) to
supracristal defects). prolapse through the defect. Although the leaflet prolapse typically
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 473

∗ ∗

A B
FIG. 40.6  Malalignment conoventricular septal defect (*) imaged with cine balanced steady-state free pre-
cession in ventricular long-axis (A) and short-axis (B). (From Wald RM, Powell AJ. Congenital heart disease.
In: Kwong R, ed. Cardiovascular Magnetic Resonance Imaging. Totowa, NJ: Humana Press; 2008:537–568.)


A B
FIG. 40.7  Muscular ventricular septal defect (*) imaged with cine balanced steady-state free precession in
ventricular long-axis (A) and short-axis (B) views. Note the turbulent flow jet seen in the right ventricle.

reduces the effective orifice size of the defect, it may also lead to aortic such as those with apical defects, a limited right ventriculotomy may
regurgitation. VSDs, particularly those associated with turbulent flow be required for effective closure. Experience with transcatheter delivery
jets, also predispose patients to the development of endocarditis. of occlusion devices is growing, and this approach may be appropriate
Symptoms are predominantly determined by the size of the shunt in selected circumstances.
through the VSD, which, in turn, is related to the defect size and the
relative resistances of the pulmonary and the systemic vascular beds. Cardiovascular Magnetic Resonance
In most situations, pulmonary resistance is lower than systemic resis- As mentioned, TTE is the primary clinical diagnostic imaging modality
tance, and there is a left-to-right shunt. The resulting increased blood in patients with suspected or known VSD, and is usually adequate.
flow to the lungs and left heart may lead to dilation of the pulmonary Occasionally, in larger patients, acoustic windows may be insufficient
arteries, pulmonary veins, LA, and LV. Because most of the shunt flow and CMR is indicated to define the defect size and location as well as
passes through the VSD into the RV during systole, RV dilation is identify associated conditions such as aortic valve prolapse. CMR may
usually not present. If the shunt is small and the patient is asymptomatic, also be of use when the hemodynamic burden of a defect is uncertain
an intervention to close the defect is not warranted. When the shunt by providing reliable quantitative data on the Qp:Qs ratio, and on
is large, symptoms from pulmonary overcirculation (e.g., tachypnea, ventricular volumes and function. Moreover, CMR has been shown to
diaphoresis, poor feeding, and slow weight gain) may develop in the be an important, noninvasive test in the diagnosis and management of
first few months of life. If the defect is unlikely to become small over defects in the outlet septum when TTE assessment of this region is
time or if these symptoms cannot be managed medically, surgical closure inadequate.28
in infancy is recommended. Untreated, patients with VSDs and large VSD location and size can be demonstrated by cine gradient recalled
left-to-right shunts may develop irreversible pulmonary artery hyper- echo (GRE) or spin echo sequences.29,30 Very small defects may be dif-
tension from elevated pulmonary vascular resistance. In such cases, the ficult to resolve; however, the associated turbulent flow can be made
flow through the defect will become increasingly right-to-left, resulting conspicuous on GRE sequences provided the echo time (TE) is long
in cyanosis (Eisenmenger syndrome). enough to allow for sufficient spin dephasing (see Fig. 40.7). It is useful
A surgical approach is by far the most common technique used to to assess the ventricular septum using stacks of images oriented in at
close VSDs and carries a low mortality risk even when performed in least two planes. The four-chamber plane provides base-to-apex local-
the first few months of life. Typically, the surgeon works through the ization, whereas the short-axis plane shows the location in the superior-
tricuspid valve and applies a patch to cover the defect. In selected patients, to-inferior axis (see Figs. 40.6 and 40.7). Also, as described in the section
474 SECTION IV  Right Ventricular and Congenital Heart Disease

on ASDs, ventricular septal configuration can be used to estimate RV Normally, the ductus arteriosus closes shortly after birth. A persistent
systolic pressure. Finally, quantification of the VSD shunt should be patent ductus arteriosus (PDA) is common in premature infants and is
performed by calculating the Qp:Qs ratio. This can be accomplished associated with increased morbidity. In full-term infants and children,
by measuring the net blood flow in the main pulmonary artery (Qp) the clinical course and sequelae of an isolated PDA are usually related
and the ascending aorta (Qs) using velocity-encoded cine CMR.24,31 to the ductus size and the direction of blood flow. In the absence of
Alternatively, in the absence of significant valve regurgitation or other elevated pulmonary vascular resistance, isolated PDAs have left-to-right
shunts, the ventricular volumetric data can be used. The Qp:Qs ratio flow, leading to increased pulmonary artery flow, and a volume load on
is equal to the RV stroke volume divided by the LV stroke volume. In the left heart. Larger PDAs cause a significant left-to-right shunt and,
practice, both approaches are recommended and the two results should if untreated, lead to pulmonary overcirculation, respiratory distress,
be compared for consistency. growth failure, and, eventually, pulmonary vascular disease. Smaller
The goals of the CMR examination include (1) delineating VSD PDAs pose little hemodynamic burden but place the patient at risk for
number, location, and size; (2) measuring the Qp:Qs ratio; (3) estimat- infective endarteritis; the benefit of closure in older children or adults
ing the RV pressure by evaluating the interventricular septal configura- is debated.32 When indicated, PDA closure can be accomplished either
tion; (4) calculating ventricular volumes, mass, and EF; and (5) surgically or in the catheterization laboratory using occluding devices.
quantifying aortic valve regurgitation if significant. These goals can be The latter is far more common in situations of an isolated PDA.
achieved by the following protocol:
• Cine bSSFP: stack of contiguous thin slices in a four-chamber plane Cardiovascular Magnetic Resonance
to span the interventricular septum for VSD visualization. CMR is seldom requested primarily for assessment of an isolated PDA
• Cine bSSFP: ventricular short-axis stack from base-to-apex for VSD because this usually presents in childhood and is a straightforward TTE
visualization and quantitative assessment of ventricular volumes diagnosis. In several types of complex congenital heart disease, evaluation
and EF. of the ductus arteriosus is an important element of the examination.
• Velocity-encoded cine: 1 to 3 contiguous slices positioned perpen- For example, in patients with tetralogy of Fallot (TOF) and pulmonary
dicular to the ventricular septum with velocity encoding in the atresia, the ductus arteriosus may persist and be a significant source of
direction of VSD flow to visualize the defect. pulmonary blood supply. PDAs can be visualized using a gadolinium-
• Blood flow measurements: ascending aorta (Qs) and main pulmonary enhanced 3D MRA or ECG and respiratory navigator-gated 3D bSSFP
artery (Qp); optional, superior vena cava and descending aorta at imaging of the thorax.33 Cine CMR is also useful in detecting PDAs,
the diaphragm (Qs), and left and right pulmonary arteries (Qp). particularly those that are small with turbulent flow. As with VSD jets,
a longer TE will allow more time for spin dephasing and make the tur-
bulent flow more conspicuous. Blood flow measurements are useful to
PATENT DUCTUS ARTERIOSUS quantify the Qp/Qs ratio. Note that with a PDA and no other shunting
The ductus arteriosus is a vascular channel that usually connects the lesions, systemic flow (Qs) is equal to the main pulmonary artery flow
aortic isthmus with the origin of the left pulmonary artery (Fig. 40.8). and pulmonary flow (Qp) is equal to ascending aorta flow. It is also
During fetal life, the ductus arteriosus allows the majority of the RV helpful to measure ventricular volumes and function, and assess RV
output to bypass the lungs by carrying blood flow to the descending aorta. systolic pressure by evaluating ventricular septal position in systole.

Ao
PDA flow
PDA

MPA

A B
FIG. 40.8  Patent ductus arteriosus (PDA). (A) Cine balanced steady-state free precession image in an oblique
sagittal plane showing a small patent ductus arteriosus. (B) Color-coded velocity-encoded cine showing flow
through the PDA. Ao, Aorta; MPA, main pulmonary artery. (From Geva T, Powell AJ. Magnetic resonance
imaging. In: Allen HD, Shaddy RE, Penny DJ, Feltes FT, Cetta F, eds. Moss and Adams’ Heart Disease in
Infants, Children, and Adolescents. 9th ed. Philadelphia: Wolters Kluwer; 2016:373–412.)
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 475

The goals of the CMR examination include (1) delineating the PDA scimitar syndrome include hypoplasia of the right lung and pulmonary
size, (2) measuring the Qp:Qs ratio, (3) assessing the direction of PDA artery, secondary dextrocardia, and anomalous systemic arterial col-
flow, (4) estimating the RV pressure by evaluating the interventricular lateral vessels, usually from the descending aorta to the right lung.
septal configuration, and (5) calculating biventricular volumes, mass, PAPVC results in a left-to-right shunt because blood draining
and EF. These goals can be achieved by the following protocol: from the lungs returns to the lungs via the systemic veins and right
• Cine bSSFP: stack of contiguous thin slices in an oblique sagittal heart without passing through the systemic arterial circulation. This
plane positioned between the transverse aortic arch and the left physiology leads to increased pulmonary blood flow and resembles
pulmonary artery for PDA visualization (see Fig. 40.8). that of an ASD. The magnitude of the shunt is determined by the
• Velocity-encoded cine: in the same location as above, with velocity number and size of the involved veins, the site of their connections,
encoding in the direction of PDA flow to visualize the defect and the pulmonary vascular resistance, the size of their vascular bed, and
direction of flow (see Fig. 40.8). the presence of associated defects. Dilation is commonly seen in the
• Contrast-enhanced 3D MRA and/or ECG and respiratory navigator- systemic veins downstream of the anomalous pulmonary vein insertion
gated 3D bSSFP imaging of the thorax. site, the RA and RV, and the pulmonary arteries. Young patients are
• Cine bSSFP: ventricular short-axis stack from base-to-apex for quan- usually asymptomatic, but dyspnea on exertion becomes increasingly
titative assessment of biventricular volumes and EF. common in the third and fourth decades of life. Development of pul-
• Blood flow measurements: ascending aorta (Qp) and main pulmo- monary hypertension is very rare. Patients may come to attention after
nary artery (Qs); optional, superior vena cava and descending aorta auscultation of a pulmonary flow murmur or when diagnostic imaging
at the diaphragm (Qs), and left and right pulmonary arteries distal is performed for another indication. Evidence of RV volume overload
to the PDA (Qp). with no apparent intracardiac shunt should prompt a search for PAPVC.
The presentation of scimitar syndrome varies widely depending on
PARTIALLY ANOMALOUS PULMONARY the severity of the associated abnormalities. Infants may be critically
ill with respiratory compromise, whereas adults may have minimal
VENOUS CONNECTION symptoms.
In partially anomalous pulmonary venous connection (PAPVC), one For the most part, PAPVC can be surgically corrected, but the likeli-
or more but not all of the pulmonary veins connect to a systemic vein. hood of success and probable benefits must be weighed carefully. A
PAPVC may be seen in isolation or as a component in complex congenital single small anomalous pulmonary vein is associated with a modest
heart lesions, particularly heterotaxy syndrome with polysplenia. left-to-right shunt and does not require intervention.34 In those with
Common anomalous pulmonary vein connection sites are the left much or all of the left pulmonary veins returning to the left innominate
innominate vein, the right superior vena cava, the azygous vein, and vein, the connecting vertical vein is usually large and long enough to
the inferior vena cava. Anomalous connection of some or all of the detach from the innominate vein and anastomose to the LA. For veins
right pulmonary veins to the inferior vena cava is termed “scimitar connecting to the superior vena cava, a baffle within the superior vena
syndrome” (Fig. 40.9). The name is derived from the curvilinear shadow cava and across the atrial septum can be constructed to channel the
in the right lung on chest radiography caused by the anomalous vein pulmonary venous return to the LA similar to sinus venosus defect
as it descends toward the right hemidiaphragm, which resembles a repair. Alternatively, the superior vena cava can be transected superior
scimitar, or Turkish sword. Other abnormalities commonly seen in to the anomalous veins and the caval end anastomosed to the right
atrial appendage (Warden procedure). A baffle across the atrial septum
is then created to direct the pulmonary venous flow in the cardiac end
of the superior vena cava to the LA. Pulmonary veins entering the
inferior vena cava can either be baffled through the RA to the LA or
directly anastomosed to the LA. The most common postoperative com-
plications seen in patients with PAPVC correction are obstruction and
residual defects in the surgically created pathways.

Cardiovascular Magnetic Resonance


Preoperative
The acoustic properties of lung tissue may make it difficult by TTE to
trace anomalous veins back into the lungs to confirm that they are
pulmonary rather than systemic veins. If a clinical concern for PAPVC
cannot be resolved by TTE with confidence, CMR is usually the most
appropriate diagnostic imaging test.35 Several studies have demonstrated
that CMR angiography techniques for PAPVC have a high level of agree-
ment with surgical inspection and invasive x-ray angiography, and are
more accurate than TTE and transesophageal echocardiography.36–40 In
addition to defining anatomy, the size of the shunt can be calculated
directly by measuring blood flow in the main pulmonary artery (Qp)
and the ascending aorta (Qs). In some cases, it may also be possible to
measure the flow in the anomalously draining vein itself. In the absence
of significant valvular insufficiency, the ventricular stoke volume dif-
FIG. 40.9  Partially anomalous pulmonary venous connection. Contrast- ferential should be equal to the shunt size and thus serves as a useful
enhanced three-dimensional magnetic resonance angiography showing check. For cases in which there is a hypoplastic pulmonary artery or
drainage of the right upper pulmonary vein (arrow) to the inferior vena pulmonary venous pathway obstruction, it is also important to measure
cava–right atrial junction in a patient with scimitar syndrome. blood flow in the branch pulmonary arteries to assess differential flow.
476 SECTION IV  Right Ventricular and Congenital Heart Disease

The goals of the CMR examination include (1) delineating pulmo- have isolated coarctation and may have minimal symptoms. Even in
nary venous drainage, (2) measuring the Qp:Qs ratio and pulmonary asymptomatic patients, relief of the aortic obstruction is indicated for
artery flow distribution, (3) estimating the RV systolic pressure by hemodynamically significant lesions because of the high rate of late
evaluating the interventricular septal configuration, and (4) calculating complications, including heart failure, systemic hypertension, premature
biventricular volumes, mass, and EF. These goals can be achieved by coronary artery disease, ruptured aortic or cerebral aneurysms, stroke,
the following protocol: aortic dissection, infective endarteritis, and premature death.41
• Cine bSSFP: axial stack from the mid liver to the top of the aortic Therapeutic options for coarctation include surgical repair and per-
arch for pulmonary venous and arterial anatomy cutaneous balloon angioplasty and stent placement. Currently, resection
• Cine bSSFP: ventricular long-axis planes and a short-axis stack from of the coarctation with an end-to-end anastomosis and augmentation of
base-to-apex for quantitative assessment of biventricular volumes the transverse arch, if needed, is the most widely practiced surgical repair
and EF and has the lowest incidence of recurrent obstruction. Other approaches
• Contrast-enhanced 3D MRA and/or ECG and respiratory navigator- have included subclavian flap aortoplasty, patch augmentation, and
gated 3D bSSFP imaging of the thorax conduit interposition. Coarctation in infants is treated surgically in the
• Blood flow measurements: ascending aorta (Qs) and main pul- majority of centers because of the lower risk of residual obstruction,
monary artery (Qp); left and right pulmonary arteries (Qp) and recurrence, and technique-related complications compared with percu-
anomalous vein taneous interventions.42 In the event of recurrent coarctation following
surgical repair, balloon angioplasty with or without stent placement is
Postoperative often the preferred approach. Coarctation in older children or adults
The goals of a postoperative assessment include those of the preopera- is increasingly being treated primarily by percutaneous interventions.
tive assessment along with the need to carefully assess for obstruction Regardless of the initial treatment, subsequent surveillance for restenosis,
and defects along the surgically created pulmonary venous drainage aneurysm formation, and dissection is warranted.43
pathway. All of these aims can be accomplished using the preoperative
imaging protocol above and tailored cine bSSFP imaging planes to Cardiovascular Magnetic Resonance
visualize the pulmonary venous connection to the LA. Preintervention and Postintervention
TTE is usually the only diagnostic imaging needed for evaluation of
young children with suspected coarctation or following intervention
COARCTATION OF THE AORTA for coarctation. With increasing age, acoustic windows typically dete-
Coarctation of the aorta is a discrete narrowing most commonly located riorate, leading to an incomplete assessment by TTE. In these circum-
just distal to the left subclavian artery, at the ductus arteriosus insertion stances, CMR is able to provide high-quality anatomic imaging of the
site (Fig. 40.10). Hypoplasia and elongation of the distal transverse arch aortic arch in its entirety (Fig. 40.10), an assessment of the hemodynamic
is a frequent association. Coarctation may be present alone or in com- severity of the obstruction, and an evaluation of LV mass, volumes,
bination with other heart lesions, including bicuspid aortic valve, sub- and function. In a retrospective study of 84 adult patients following
valvar and valvar aortic stenosis, mitral valve abnormalities, ASD, VSD, intervention for coarctation of the aorta, Therrien et al.44 showed that
persistent PDA, and conotruncal anomalies. the combination of clinical assessment and CMR on every patient was
Infants tend to present with symptoms of heart failure and sys- more “cost-effective” for detecting complications than combinations
temic hypoperfusion as the ductus arteriosus closes; if untreated, they that relied on TTE or chest radiography as imaging modalities. Other
may progress to shock or death. Older children and adults typically studies have shown the utility of CMR in infants and children with

A B
FIG. 40.10  Aortic coarctation. (A) Electrocardiogram and respiratory navigator-gated three-dimensional (3D)
balanced steady-state free precession oblique sagittal reformat showing severe discrete narrowing at the
aortic isthmus with an associated turbulent flow jet. (B) Volume rendered contrast-enhanced 3D magnetic
resonance angiography revealing several tortuous collateral vessels and dilated internal mammary arteries.
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 477

coarctation and other anomalies of the aortic arch.45–47 Computed lead to LV hypertrophy and dysfunction. Upper body hypertension
tomography (CT) can also provide excellent anatomic imaging of the may be caused by aortic arch obstruction but systemic hypertension is
aorta but has the disadvantage of ionizing radiation exposure and the also prevalent following coarctation repair even without residual coarc-
need for iodinated contrast make it a less attractive modality for serial tation, particularly in patients who had relief of obstruction at an older
follow-up. age. It is good practice to measure both upper arms and lower extremity
With regard to aortic anatomy, careful attention should be given to cuff blood pressures at the time of the CMR examination to help identify
the transverse aortic arch and isthmus, brachiocephalic vessels, collateral patients with systemic hypertension and estimate the pressure gradient
vessels that may bypass the obstruction, and possible aneurysms or across any aortic obstruction. Note that there may be little upper-to-
dissections at the repair sites. Regions of vessel narrowing should ideally lower extremity blood pressure differential even with important aortic
be measured in cross section as elliptical lesions are common. If a obstruction when there is a significant collateral circulation bypassing
coarctation is present, its diameter, length, and distance to neighboring the obstruction.
vessels should be reported, as this may influence decisions regarding The goals of the CMR examination include (1) assessing the aortic
percutaneous intervention. Given the association of a bicuspid aortic arch anatomy and degree of obstruction; (2) calculating LV volumes,
valve with coarctation, the aortic valve morphology should be noted mass, and EF; and (3) evaluating aortic valve morphology and function.
as well as the dimensions of the aortic root and ascending aorta, as These goals can be achieved by the following protocol26:
they may be dilated. • Cine bSSFP: stack of contiguous thin slices in an oblique sagittal
Blood velocity and flow measurements with CMR have been used plane oriented in long axis (parallel) to the aortic arch for evaluation
to gain insight into the functional significance of a coarctation. One of arch anatomy.
approach has been to assess the flow pattern in the descending aorta • Black-blood spin echo (optional): stack of contiguous thin slices in
distal to the coarctation, preferably at the level of the diaphragm (see an oblique sagittal plane oriented in long axis (parallel) to the aortic
Fig. 40.11). Characteristics suggestive of a hemodynamically significant arch for evaluation of arch anatomy. This is particularly valuable
coarctation include decreased peak flow, decreased time-averaged flow, following endovascular stent placement because there is less metallic
delayed onset of descending aorta flow compared with the onset of susceptibility artifact than with cine sequences.
flow in the ascending aorta, decreased acceleration rate, and prolonged • Cine bSSFP: ventricular long-axis planes and a short-axis stack from
deceleration with increased antegrade diastolic flow.48–51 Another approach base-to-apex for quantitative assessment of LV cavity volume, mass,
to assessing coarctation severity is to measure the peak coarctation jet and EF.
velocity and estimate a pressure gradient using the modified Bernoulli • Contrast-enhanced 3D MRA and/or ECG and respiratory navigator-
equation.52–55 Note, however, that such measurements may be technically gated 3D bSSFP imaging of the thorax (see Fig. 40.10).
difficult in a long, tortuous coarctation segment and that the pressure • Blood flow measurements: ascending aorta, main pulmonary artery,
estimates may not be indicative of anatomic severity because of col- descending aorta at the level of the diaphragm for assessment of
lateral flow. Finally, CMR has been used to quantify collateral flow the descending aorta flow profile (see Fig. 40.11) and aortic valve
entering the descending aorta distal to the obstruction via retrograde function.
flow from the intercostal arteries or vessels arising off the aortic arch • Blood flow measurements (optional): just distal to the coarctation
and arch branches.49,56–58 With this technique, proximal flow is measured in long axis and cross section for measurement of the peak velocity
just proximal or distal to the site of obstruction, and distal flow in the and/or for quantitation of collateral flow.
descending aorta at the level of the diaphragm. A greater distal-to-
proximal flow ratio is seen with increased collateral flow and suggests
more severe obstruction. If little collateral flow is found, the surgeon
TETRALOGY OF FALLOT
may elect to perform a left heart bypass to the descending aorta during TOF is the most common type of cyanotic congenital heart
the repair to reduce the risk of spinal cord ischemic injury when cor- disease.59 Although TOF involves several anatomic components, the
rection requires interruption of aortic flow. anomaly is thought to result from a single developmental anomaly—
CMR evaluation of coarctation should also include calculation of underdevelopment of the subpulmonary infundibulum (conus).60 The
LV volumes, EF, mass, and mass-to-volume ratio. These data are clini- resulting pathology is notable for anterior, superior, and leftward devia-
cally relevant because systemic hypertension is often present and may tion of the infundibular (conal) septum, causing subpulmonary stenosis;
Flow (mL/s)

Flow (mL/s)

A B
FIG. 40.11  Evaluation of coarctation severity based on the flow pattern in the descending aorta at the level
of the diaphragm. (A) Descending aorta flow pattern in a patient with repaired coarctation and no residual
obstruction. Note the sharp upstroke and short deceleration phase. (B) Descending aorta flow pattern in a
patient with severe coarctation. Note the blunted upstroke and prolonged deceleration.
478 SECTION IV  Right Ventricular and Congenital Heart Disease

DAo

A B
FIG. 40.12  Multiple aortopulmonary collateral (APC) vessels in a newborn with tetralogy of Fallot and pulmonary
atresia evaluated by contrast-enhanced three-dimensional magnetic resonance angiography. (A) Subvolume
maximum intensity projection (MIP) image in the coronal plane showing multiple APCs arising from the
descending aorta. (B) Subvolume MIP image in the transverse plane showing APCs arising from the descend-
ing aorta (DAo) and splitting into two large branches, one to the left lung and one to the right lung (arrows).

pulmonary valve stenosis from annular hypoplasia, thickened leaflets, Cardiovascular Magnetic Resonance
and abnormal commissures; a large conoventricular septal defect; and Preoperative
overriding of the aortic valve above the interventricular septum. The For most infants, TTE provides all of the necessary diagnostic informa-
degree of RV outflow tract (RVOT) obstruction varies from mild to tion for surgical repair; however, when the pulmonary arteries are
complete obstruction (i.e., TOF with pulmonary atresia). The mediastinal hypoplastic and aortopulmonary collateral vessels are suspected, other
pulmonary arteries are often small, and may even be discontinuous or modalities are often needed to adequately delineate these structures.
absent. In patients with pulmonary atresia or absent branch pulmonary Contrast-enhanced MRA is well suited for this task (Fig. 40.12). Com-
arteries, pulmonary blood flow comes from a PDA and/or collateral pared with conventional x-ray angiography, MRA has been shown to
vessels arising from the aorta and its branches. Approximately 5% to be highly accurate in depicting all sources of pulmonary blood supply
6% of patients with TOF have a major coronary artery crossing the in patients with complex pulmonary stenosis or atresia, including infants
anterior aspect of the RVOT.61 Most commonly, it is the left anterior with multiple small aortopulmonary collaterals.33,38
descending coronary artery which arises from the right coronary artery
and traverses the infundibular free wall to reach the anterior interven- Postoperative
tricular groove. Preoperative identification of a major coronary artery CMR provides a comprehensive imaging assessment of postoperative
crossing the RVOT is important to avoid inadvertent damage to the TOF patients and its use is recommended by a number of expert guide-
coronary artery during surgical relief of RVOT obstruction. Finally, line statements.35,68–70 Information derived from CMR on pulmonary
approximately 25% of patients have a right aortic arch. regurgitation, biventricular size and function, RVOT aneurysm (Fig.
Although the clinical presentation and course of patients with TOF 40.13), and pulmonary artery stenosis often dictates clinical care. In
varies, most patients develop cyanosis during the first year of life. Surgi- particular, CMR parameters are a key component to determine whether
cal repair is usually performed before the age of 6 months. A typical patients should undergo pulmonary valve replacement.71 Moreover, in
repair includes patch closure of the VSD and RVOT obstruction relief an effort to improve risk stratification, a growing number of studies
using a combination of resection of obstructive muscle bundles and have linked CMR data to adverse outcomes. A report from the Inter-
an overlay patch. When the pulmonary valve annulus is moderately or national Multicenter TOF Registry (INDICATOR) showed that RV
severely hypoplastic, the RVOT patch is extended across the pulmonary hypertrophy and decreased CMR LV and RV EF are associated with
valve annulus into the main pulmonary artery, destroying the valve death or sustained ventricular tachycardia.72 Focal fibrosis, as identified
mechanism and leading to pulmonary regurgitation. In patients with by late gadolinium enhancement (LGE), is common after TOF repair,
TOF and pulmonary atresia, or when a major coronary artery crosses and a greater extent of RV involvement is associated with older age,
the RVOT, a conduit—either a homograft or a prosthetic tube—is placed worse symptoms, exercise intolerance, RV dysfunction, and clinical
between the RVOT and the pulmonary arteries. arrhythmia.73 The utility of diffuse fibrosis assessment using T1 mea-
Patients undergoing surgical repair of TOF as young children have surement techniques is now being explored.74,75
excellent short-term survival62; however, they experience significant The goals of the CMR examination include (1) quantitative assess-
morbidity and mortality related to biventricular dysfunction and arrhyth- ment of biventricular volume, mass, stroke volume, and EF; (2) imaging
mia in their adult years.63–65 These sequelae are believed to be at least the RVOT, pulmonary arteries, aorta, and aortopulmonary collaterals;
in part related to chronic pulmonary regurgitation caused by efforts (3) quantification of pulmonary regurgitation, tricuspid regurgitation,
to relieve pulmonary valve stenosis with the initial TOF repair. Thus cardiac output, and Qp:Qs ratio; and (4) assessment of myocardial
pulmonary valve replacement is often performed subsequently to improve fibrosis. These objectives can be achieved with the following protocol26:
the long-term outcome.66,67 Other common concerns in repaired TOF • Cine bSSFP: ventricular long-axis planes and a short-axis stack from
patients include RVOT tract or pulmonary arterial obstruction, tricuspid base-to-apex for quantitative assessment of biventricular volumes,
regurgitation, and residual ASDs and VSDs. mass, and EF.
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 479

MPA

RV
RV

A B
FIG. 40.13  Right ventricular (RV) outflow tract aneurysm following repair of tetralogy of Fallot. (A) Cine
balanced steady-state free precession (bSSFP) image showing a thin, dyskinetic anterior wall (arrow) of
the right ventricle, at least in part related to placement of an RV outflow tract patch. (B) Cine bSSFP image
demonstrating a massive aneurysm of the RV outflow tract and main pulmonary artery (MPA). Note the
mural thrombus in the MPA (arrow).

• Cine bSSFP: stack parallel to the RVOT. and in the Senning operation native atrial tissue is used. The most
• Cine bSSFP: axial plane to image the branch pulmonary arteries. common sequelae following atrial switch operations are RV (systemic
• Contrast-enhanced 3D MRA and/or ECG and respiratory navigator- ventricle) dysfunction, tricuspid regurgitation, sinus node dysfunction,
gated 3D bSSFP imaging of the thorax. atrial arrhythmias, obstruction of the atrial baffle pathways, and atrial
• Blood flow measurements: main and branch pulmonary arteries, baffle leaks. During the 1980s, the arterial switch operation largely
ascending aorta, and atrioventricular valves. replaced the atrial switch procedures. In this operation, the ascending
• LGE: ventricular long-axis and short-axis planes. aorta and main pulmonary artery are transected and then connected
to the concordant semilunar valve root, and the coronary arteries are
transferred to the new aortic valve. The advantages of the arterial
TRANSPOSITION OF THE GREAT ARTERIES switch operation over the atrial switch procedures include the estab-
Transposition of the great arteries (TGA) is defined as discordant align- lishment of the LV (rather than the RV) as the systemic ventricle, and
ments between the ventricles and the great arteries; the aorta arises the avoidance of extensive suture lines in the atria, which contribute to
from the RV and the pulmonary artery arises from the LV. The most arrhythmia. Recent data on long-term outcomes of the arterial switch
common type of TGA and the focus of the subsequent discussion is operation continue to show excellent overall survival with low morbid-
with visceroatrial situs solitus (S), ventricular D-loop (D), and dextro ity.77,78 The most common postoperative complications include stenosis
malposition of the aortic valve relative to the pulmonary valve (D). of the main and branch pulmonary arteries (Fig. 40.15), dilatation
This anatomical arrangement can be summarized as {S,D,D} TGA. of the neoaortic root, aortic valve regurgitation, and coronary artery
Associated cardiac anomalies with their approximate prevalence stenosis.79 The Rastelli operation is another surgical option for TGA
include VSDs in 45% of patients, coarctation or interrupted aortic patients with an associated subvalvar and valvar pulmonary stenosis and
arch in 12%, pulmonary stenosis in 5%, RV hypoplasia in 4%, and a VSD. It consists of patch closure of the VSD so that the LV outflow is
juxtaposition of the atrial appendages in 2%.76 The principal physi- directed to the aortic valve, and placement of a conduit from the RV to
ologic abnormality is that desaturated systemic venous blood flows to the pulmonary arteries. More recently, this anatomic variant has also
the RA, RV, and the aorta, and oxygenated pulmonary venous blood been treated with complete removal of the native pulmonary root and
flows to the LA and LV, and then back to the lungs. These parallel valve, translocation of the entire aortic root and coronary arteries to
systemic and pulmonary circulations result in profound hypoxemia. the LV outflow tract, and placement of a homograft between the RVOT
Consequently, survival depends on the presence of an ASD, VSD, and/or and the pulmonary arteries.
PDA that allows mixing of blood between the systemic and pulmonary
circulations. Cardiovascular Magnetic Resonance
Surgical management of D-loop TGA in the 1960s and 1970s con- Preoperative
sisted mostly of an atrial switch procedure—the Senning or Mustard CMR is seldom requested for preoperative assessment of infants with
operations. In both procedures, the systemic and pulmonary venous D-loop TGA because TTE usually provides all of the necessary diagnostic
blood is redirected within the atria so that the pulmonary venous blood information.76
reaches the tricuspid valve, RV, and aorta, whereas the systemic venous
blood reaches the mitral valve, LV, and pulmonary arteries (Fig. 40.14). Postoperative Atrial Switch Operation
The main technical difference between these two procedures is that in CMR has a central role in the noninvasive imaging surveillance of
the Mustard operation, pericardium is used to redirect the blood flow patients who have undergone an atrial switch operation.69,80 Assessment
480 SECTION IV  Right Ventricular and Congenital Heart Disease

SVC
RV MPA
Ao

LV
∗ LV

RV LV

IVC
PV SVC

A B C
FIG. 40.14  Mustard palliation for transposition of the great arteries. Electrocardiogram and respiratory nav-
igator-gated three-dimensional balanced steady-state free precession data was reformatted offline in multiple
user-defined planes. (A) Transverse plane showing the pulmonary venous (PV) pathway to the right ventricle
(RV) and superior vena cava (SVC) pathway to the left ventricle (LV). (B) Oblique sagittal plane depicting the
SVC and inferior vena cava (IVC) pathways to the LV, and the PV pathway (*) in cross-section. (C) Oblique
sagittal plane showing the LV outflow tract connecting to the main pulmonary artery (MPA) and the RV
outflow tract connecting to the aorta (Ao).

of the systemic RV, a key concern in this patient group, can be difficult • Cine bSSFP: ventricular short-axis stack from base-to-apex for quan-
by TTE because of its substernal position and complex shape. CMR, titative assessment of biventricular volumes, mass, and EF.
however, routinely provides complete tomographic imaging of the RV, • Contrast-enhanced 3D MRA and/or ECG and respiratory navigator-
which allows for accurate and reproducible measurements of volume, gated 3D bSSFP imaging of the chest (see Fig. 40.14).
mass, and EF.81,82 CMR is also useful for evaluating the systemic and • Blood flow measurements: main pulmonary artery, ascending aorta,
pulmonary venous baffle pathways for obstruction and leaks.83,84 The tricuspid valve, and mitral valve.
physiologic impact of baffle leaks can be gauged by measuring blood • LGE: ventricular long- and short-axis planes.
flow in the main pulmonary artery and ascending aorta to calculate
the Qp:Qs ratio. Note, however, that a Qp:Qs ratio close to 1 can be Postoperative Arterial Switch Operation
seen even in large baffle leaks when there is bidirectional flow; thus, Studies have shown that detection of pulmonary artery stenosis in arte-
anatomic imaging information and the systemic oxygen saturation must rial switch operation patients by CMR is accurate and superior to the
also be considered. information obtained by TTE (see Fig. 40.15).89–91 CMR can also provide
Studies in patients who have undergone an atrial switch operation high-resolution imaging of the proximal coronary arteries and define
have found RV LGE indicative of focal myocardial fibrosis, although their relationship to the surrounding structures such as the aorta and
the reported prevalence of this finding varies.85–88 In cross-sectional main pulmonary artery.92–94 The largest report consisted of 84 CMR
studies, the presence and extent of LGE have been associated with greater examinations and yielded diagnostic image quality of the proximal
age, RV dysfunction, reduced peak oxygen uptake, electrophysiologic coronary arteries in 95% and showed stenosis in 11%.94 There are two
parameters, arrhythmia, and adverse clinical events. The best evidence principal CMR techniques to diagnose inducible myocardial ischemia:
establishing the prognostic value of LGE comes from a longitudinal (1) evaluation for perfusion defects using vasodilator stress agents (e.g.,
study of 55 atrial switch operation patients which showed that RV LGE adenosine or dipyridamole); and (2) evaluation of wall motion, most
was independently associated with a composite endpoint primarily commonly using dobutamine stress. In addition, the LGE technique is
composed of atrial tachyarrhythmia.87 highly sensitive for detecting myocardial infarction and focal fibrosis.
The goals of the CMR evaluation include (1) quantitative assessment In two reports describing a total of 55 asymptomatic arterial switch
of the systemic RV volume, mass, and EF; (2) imaging of the systemic operation patients who underwent stress perfusion CMR, no defects at
and pulmonary venous pathways for obstruction and baffle leaks; (3) stress were detected.93,95 Moreover, the LGE technique has identified prior
quantitation of tricuspid valve regurgitation; (4) evaluation of the LV myocardial infarction in only a small proportion of patients after arterial
and RV outflow tracts for obstruction; and (5) assessment of myocardial switch operation.93,95,96 Thus based on these studies, both CMR stress
fibrosis. These goals can be achieved with the following protocol26: imaging and LGE appear to have a low positive yield in asymptomatic
• Cine bSSFP: axial stack from the mid-liver through the top of the arterial switch operation patients. Larger studies that include symp-
aortic arch for dynamic imaging of the venous pathways, atrioven- tomatic patients and have longer-term follow-up are needed to better
tricular valve regurgitation, and the great arteries. define the indications and prognostic value of these CMR techniques.
• Cine bSSFP: oblique planes to image the superior and inferior limbs The goals of the CMR evaluation include (1) quantitative assessment
of the systemic venous pathway in long axis. of biventricular volumes and function; (2) imaging the LV and RV
• Cine bSSFP: ventricular long-axis planes to assess biventricular func- outflow tracts for obstruction; (3) imaging the aorta, main pulmonary
tion and the outflow tracts. artery, and branch pulmonary arteries for obstruction; (4) measurement
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 481

MPA
MPA RPA

RPA

Ao

LPA

A B

MPA LPA

Ao

MPA

C D
FIG. 40.15  Arterial switch operation with Lecompte maneuver for transposition of the great arteries. Contrast-
enhanced three-dimensional magnetic resonance angiography with subvolume maximum intensity projection
(A–C) and volume rendering (D) showing the main pulmonary artery (MPA) bifurcation positioned anterior to
the ascending aorta (Ao), right pulmonary artery (RPA) stenosis, and an unobstructed left pulmonary artery
(LPA).

of aortic root size; (5) quantitation of aortic and pulmonary valve • If there is a concern for inducible myocardial ischemia, a vasodilator
regurgitation; (6) identification of residual atrial and ventricular septal or dobutamine stress protocol and myocardial LGE imaging should
defects, and calculation of the Qp:Qs ratio; and (7) description of the be considered.
proximal coronary origins, course, and degree of obstruction. These
goals can be achieved with the following protocol26:
SINGLE VENTRICLE
• Cine bSSFP: ventricular long-axis planes and a short-axis stack from
base-to-apex for quantitative assessment of biventricular dimensions The normal human heart is composed of three chambers at the ven-
and EF. tricular level (between the atrioventricular valves and the semilunar
• Cine bSSFP: axial stack to assess for branch pulmonary artery valves): the LV sinus, the RV sinus, and the infundibulum. From an
obstruction. anatomic standpoint, a single ventricle heart is present when one of
• Cine bSSFP: stack in short axis to the aortic root to assess the degree the two ventricular sinuses is absent. The infundibulum is always present
of dilation and visualize any regurgitation jet. and has a variety of names such as infundibular outlet chamber, rudi-
• Cine bSSFP: ascending aorta long-axis plane to assess for obstruction. mentary or hypoplastic RV, and rudimentary chamber. There are two
• Contrast-enhanced 3D MRA (see Fig. 40.15). types of single ventricle:
• ECG and respiratory navigator-gated 3D bSSFP imaging of the 1. Single LV (Fig. 40.16) in which there is an absence of the RV sinus
coronary arteries. and the presence of an LV and an infundibulum. The communica-
• Blood flow measurements: main and branch pulmonary arteries tion between the LV and the infundibulum is termed the bulboven-
and ascending aorta. tricular foramen.
482 SECTION IV  Right Ventricular and Congenital Heart Disease

Inf

RA TV
*
MV
MV
TV
LA

A B
FIG. 40.16  Double-inlet single left ventricle. Cine balanced steady-state free precession images in horizontal
long-axis (A) and short-axis (B) planes. Note that both mitral valve (MV) and the tricuspid valve (TV) enter the
left ventricle, which is characterized by a smooth septal surface (arrow). The bulboventricular foramen (*)
provides an exit from the left ventricle to the infundibulum (Inf). LA, Left atrium; RA, right atrium.

FIG. 40.17  Double-inlet single right ventricle. Stack of cine steady-state free precession images in a short-
axis plane. Note the absence of a ventricular septum.

2. Single RV (Fig. 40.17) in which the ventricular mass consists of the or “functional univentricular hearts,” and are managed similarly to single
RV sinus and the infundibulum, together forming a common chamber. ventricle hearts. Examples include tricuspid atresia, mitral atresia, severely
The septal band is present, indicating the location of a ventricular unbalanced common atrioventricular canal defect, and hypoplastic left
septum, but there is no macroscopically recognizable LV sinus on heart syndrome.
the other side of the septum. Single ventricle physiology is characterized by complete mixing of
There are other congenital cardiac anomalies that do not have a the systemic and pulmonary venous return flows. The proportion of the
single ventricle from a strict anatomic standpoint but whose anatomy ventricular output distributed to the pulmonary versus systemic circula-
precludes the establishment of a normal two-ventricle circulation. These tions is determined by the relative resistance to flow in the two circuits
conditions are often grouped under the term “functional single ventricle” and heavily influences the systemic oxygen saturation. Management is
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 483

RV
Neo-Ao

SVC * MPA

RPA
RV
LPA

DAo
A B

RIA
LIV

AA
Neo-Ao

Az.

RV

C D
FIG. 40.18  Hypoplastic left heart syndrome. Contrast-enhanced three-dimensional (3D) magnetic resonance
angiography in infants before bidirectional Glenn shunt. (A) Subvolume maximum intensity projection (MIP) in
an oblique transverse plane showing a conduit (*) from the right ventricle (RV) to the pulmonary arteries. (B)
3D volume rendering in the same patient showing the conduit (arrow) as well as the anastomosis between
the main pulmonary artery and the reconstructed neo-aorta (Neo-Ao). (C) Oblique coronal subvolume MIP
in a patient with a modified right Blalock-Taussig shunt. The shunt (arrow) extends from the right innomi-
nate artery (RIA) to the right pulmonary artery, and there is pulmonary artery stenosis at the insertion site.
(D) Oblique sagittal subvolume MIP showing a reconstructed Neo-Ao. AA, Ascending aorta; Az., azygos vein;
DAo, descending aorta; LIV, left innominate vein; LPA, left pulmonary artery; MPA, main pulmonary artery;
RPA, right pulmonary artery; SVC, superior vena cava.

typically directed toward a series of surgeries culminating in the Fontan and superior vena cava flow passively draining to the lungs, and pul-
operation. The first surgery is often performed in the first week or two monary venous return actively pumped by the single ventricle out to
of life. Its goals are to establish an appropriate amount of pulmonary the body. This largely eliminates hypoxemia. Since it was first described
blood flow to avoid overcirculation to the lungs, yet adequate systemic in 1971,97 the Fontan operation has undergone multiple modifications
saturation, and to provide unobstructed flow to the systemic circulation. (Fig. 40.19), including direct anastomosis of the right atrial append-
A variety of surgical procedures may be performed, depending on the age to the main pulmonary artery (called an atriopulmonary anasto-
specific patient details and institutional preference. Examples include mosis), RA-to-RV conduit, lateral tunnel between the inferior vena
the stage I procedure, Blalock-Taussig shunt, and pulmonary artery cava and the undersurface of the ipsilateral branch pulmonary artery
band placement (Fig. 40.18). The next surgery is typically a superior (Fig. 40.20), fenestration of the lateral tunnel baffle, and an extracar-
cavopulmonary anastomosis (bidirectional Glenn) operation, performed diac conduit between the inferior vena cava and the ipsilateral branch
at 4 to 6 months of age. As a result of this procedure, the superior vena pulmonary artery.
cava supplies pulmonary flow, and the inferior vena cava blood skips With refinement of patient selection criteria, medical therapy, and sur-
the lungs and is directed to the ventricle and then out to the systemic gical techniques, the results of this stage surgical management approach
circulation. The third and often final surgery is the Fontan operation, to single ventricle heart disease have improved. Nevertheless, this patient
performed between 18 months and 3 years of age. With this procedure, population continues to be at risk for complications such as ventricu-
inferior vena cava blood is also directed to the pulmonary arteries. Thus lar systolic and diastolic dysfunction; valve regurgitation; aortic arch,
the systemic and pulmonary circulations are separated with inferior Fontan pathway, and pulmonary artery obstruction; pulmonary vein
484 SECTION IV  Right Ventricular and Congenital Heart Disease

Atriopulmonary
Fontan

Classic Fontan Lateral tunnel


fenestrated Fontan

Extracardiac
Fontan

FIG. 40.19  Diagrams of four variations of the Fontan operation.

LT
RPA
∗ LT
LPA
RV
LT
RV

A B C

RCA
PR

FIG. 40.20  Lateral tunnel Fontan operation in a patient with hypoplastic left
AoR
heart syndrome. Reformatted electrocardiogram and respiratory navigator-gated
LCA three-dimensional balanced steady-state free precession angiogram. (A) Coronal
plane demonstrating the lateral tunnel (LT) and aorto-pulmonary anastomosis (*).
(B) Axial plane to show the LT in cross section. (C) Axial plane to show the con-
nection of the LT to the right pulmonary artery (RPA) and left pulmonary artery
(LPA). (D) Oblique axial plan to highlight the hypoplastic native aortic root (AoR),
right coronary artery (RCA), left coronary artery (LCA), and native pulmonary root
D
(PR). RV, Right ventricle.
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 485

compression; thromboembolism; protein-losing enteropathy; plastic patients, with a growing body of research to support its use.35,69 One
bronchitis; hepatic and renal dysfunction; arrhythmias; and premature study showed that a larger CMR-derived end-diastolic volume (EDV)
death. Such adverse developments are usually poorly tolerated; thus was independently associated with an increased risk of death and trans-
prompt detection with diagnostic imaging is an important element in plant, and the addition of EDV to clinical variables led to improved
caring for these patients. risk stratification for transplant-free survival.107 In another report, LGE
was seen in 28% of Fontan patients, and a greater extent of LGE was
Cardiovascular Magnetic Resonance associated with lower EF, increased EDV, increased mass, and a higher
Before the First Operation frequency of nonsustained ventricular tachycardia.108 Studies using CMR
CMR is seldom requested before the first palliative operation because blood flow measurements have also revealed new information about
TTE usually provides all of the necessary diagnostic information. the importance of aortopulmonary collateral vessels,100,101,109–113 energy
losses in the Fontan connection,114–116 and the value of computational
Before the Superior Cavopulmonary Anastomosis Operation fluid dynamics in surgical planning.117
Traditionally, the superior cavopulmonary anastomosis operation is The goals of the CMR examination include (1) assessing the systemic
preceded by invasive cardiac catheterization for assessment and plan- veins, Fontan baffle, and pulmonary arteries for obstruction and throm-
ning. However, in a prospective randomized clinical trial, CMR was bus; (2) detecting Fontan baffle fenestrations or leaks; (3) evaluating
shown to be a safe, effective, and less costly alternative to routine cath- the pulmonary veins for compression; (4) quantifying ventricular
eterization in selected patients.98,99 volumes, mass, and function; (5) detecting ventricular outflow tract
The goals of the CMR examination include (1) measuring ventricular obstruction; (6) quantifying significant valve regurgitation; (7) imaging
mass, volume, and function; (2) quantifying significant valve regurgita- the aorta for obstruction or aneurysm; and (8) assessing aortopulmonary
tion; (3) evaluating the pulmonary arteries, pulmonary veins, atrial and venovenous collateral vessels. These goals can be achieved by the
septum, and aortic arch for obstruction; and (4) assessing aortopul- following protocol26:
monary and venovenous collateral vessels.100,101 These goals can be • Cine bSSFP: axial stack from the mid-liver through the top of the
achieved by the following protocol: aortic arch for dynamic imaging of the heart and vasculature.
• Cine bSSFP: axial stack from the mid-liver through the top of the • Cine bSSFP: ventricular long-axis planes and a ventricular short-axis
aortic arch for dynamic imaging of the heart and vasculature. stack from base-to-apex for quantitative assessment of ventricular
• Cine bSSFP: ventricular long-axis planes and a ventricular short-axis volumes and EF.
stack from base-to-apex for quantitative assessment of ventricular • Cine bSSFP: aortic arch long-axis plane.
volumes and EF. • Contrast-enhanced 3D MRA.
• Cine bSSFP: aortic arch long-axis plane. • ECG and respiratory navigator-gated 3D bSSFP imaging of the thorax
• Fast (turbo) spin echo with blood suppression (optional): axial plane (see Fig. 40.20).
to image the branch pulmonary arteries and aortic arch in long axis. • Blood flow measurements: superior vena cava, Fontan baffle above
• Contrast-enhanced 3D MRA and/or ECG and respiratory navigator- and below any fenestration or baffle leak, branch pulmonary arteries,
gated 3D bSSFP imaging of the chest (see Fig. 40.18). pulmonary veins, ascending aorta, descending aorta at the diaphragm,
• Blood flow measurements: main and branch pulmonary arteries, atrioventricular valves.
ascending aorta, descending aorta at the diaphragm; superior vena • LGE: ventricular long-axis and short-axis planes.
cava, inferior vena cava, pulmonary veins, and atrioventricular valves.
• LGE: ventricular long- and short-axis planes.
CONCLUSION
Before the Fontan Operation As outlined in this chapter, CMR provides unique information in the
Several reports have critically examined the necessity of invasive infant and child with simple and complex congenital heart disease
cardiac catheterization before the Fontan operation.102–104 One ret- population—both for guiding management and for monitoring patients
rospective study showed that single ventricle patients who did not after mechanical intervention. As these children often require many
require a catheterization-based intervention and had a pre-Fontan decades of monitoring, the non-ionizing radiation attribute of CMR
CMR and TTE but not a catheterization had similar short-term out- is a particular advantage.
comes as those who underwent an invasive catheterization.105 The
CMR examination goals and imaging protocol are the same as those
before the superior cavopulmonary anastomosis operation noted
ACKNOWLEDGMENT
earlier. We thank the physicians, technologists, nurses, and support personnel
in the CMR Unit at Boston Children’s Hospital for their help. Parts of
After the Fontan Operation the text and figures of this chapter have been included in other chapters
The utility of TTE in patients after a Fontan operation is often limited recently written by the authors on the CMR evaluation of congenital
by increasing body size, acoustic interference from scar tissue, and heart disease.
incomplete visualization of the Fontan pathway and aorta. CMR is not
subject to these drawbacks (see Fig. 40.20) and provides a more reliable
quantitative assessment of ventricular size and function106 and blood
REFERENCES
flow. It has thus become an important clinical tool in these fragile A full reference list is available online at ExpertConsult.com
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 485.e1

REFERENCES transoesophageal echocardiography. J Cardiovasc Magn Reson.


2010;12:44.
1. Dewey FE, Rosenthal D, Murphy DJ Jr, Froelicher VF, Ashley EA. Does 20. Thomson LE, Crowley AL, Heitner JF, et al. Direct en face imaging of
size matter? Clinical applications of scaling cardiac size and function for secundum atrial septal defects by velocity-encoded cardiovascular
body size. Circulation. 2008;117(17):2279–2287. magnetic resonance in patients evaluated for possible transcatheter
2. Sluysmans T, Colan SD. Theoretical and empirical derivation of closure. Circ Cardiovasc Imaging. 2008;1(1):31–40.
cardiovascular allometric relationships in children. J Appl Physiol. 21. Valente AM, Sena L, Powell AJ, Del Nido PJ, Geva T. Cardiac magnetic
2005;99(2):445–457. resonance imaging evaluation of sinus venosus defects: comparison to
3. Cantinotti M, Scalese M, Molinaro S, Murzi B, Passino C. Limitations of surgical findings. Pediatr Cardiol. 2007;28(1):51–56.
current echocardiographic nomograms for left ventricular, valvular, and 22. Prompona M, Muehling O, Naebauer M, Schoenberg SO, Reiser M,
arterial dimensions in children: a critical review. J Am Soc Echocardiogr. Huber A. MRI for detection of anomalous pulmonary venous drainage
2012;25(2):142–152. in patients with sinus venosus atrial septal defects. Int J Cardiovasc
4. Voges I, Jerosch-Herold M, Hedderich J, et al. Normal values of aortic Imaging. 2011;27(3):403–412.
dimensions, distensibility, and pulse wave velocity in children and young 23. Powell AJ, Tsai-Goodman B, Prakash A, Greil GF, Geva T. Comparison
adults: a cross-sectional study. J Cardiovasc Magn Reson. 2012;14:77. between phase-velocity cine magnetic resonance imaging and invasive
5. Buechel EV, Kaiser T, Jackson C, Schmitz A, Kellenberger CJ. Normal oximetry for quantification of atrial shunts. Am J Cardiol.
right- and left ventricular volumes and myocardial mass in children 2003;91(12):1523–1525, A1529.
measured by steady state free precession cardiovascular magnetic 24. Beerbaum P, Korperich H, Barth P, Esdorn H, Gieseke J, Meyer H.
resonance. J Cardiovasc Magn Reson. 2009;11:19. Noninvasive quantification of left-to-right shunt in pediatric patients:
6. Robbers-Visser D, Boersma E, Helbing WA. Normal biventricular phase-contrast cine magnetic resonance imaging compared with invasive
function, volumes, and mass in children aged 8 to 17 years. J Magn oximetry. Circulation. 2001;103(20):2476–2482.
Reson Imaging. 2009;29(3):552–559. 25. Hundley WG, Li HF, Lange RA, et al. Assessment of left-to-right
7. Sarikouch S, Peters B, Gutberlet M, et al. Sex-specific pediatric intracardiac shunting by velocity-encoded, phase-difference magnetic
percentiles for ventricular size and mass as reference values for cardiac resonance imaging. A comparison with oximetric and indicator dilution
MRI: assessment by steady-state free-precession and phase-contrast MRI techniques. Circulation. 1995;91(12):2955–2960.
flow. Circ Cardiovasc Imaging. 2010;3(1):65–76. 26. Fratz S, Chung T, Greil GF, et al. Guidelines and protocols for
8. Alfakih K, Plein S, Thiele H, Jones T, Ridgway JP, Sivananthan MU. cardiovascular magnetic resonance in children and adults with
Normal human left and right ventricular dimensions for MRI as congenital heart disease: SCMR expert consensus group on congenital
assessed by turbo gradient echo and steady-state free precession imaging heart disease. J Cardiovasc Magn Reson. 2013;15:51.
sequences. J Magn Reson Imaging. 2003;17(3):323–329. 27. Van Praagh R, Geva T, Kreutzer J. Ventricular septal defects: how shall
9. Hudsmith LE, Petersen SE, Francis JM, Robson MD, Neubauer S. we describe, name and classify them? J Am Coll Cardiol. 1989;14(5):
Normal human left and right ventricular and left atrial dimensions 1298–1299.
using steady state free precession magnetic resonance imaging. J 28. Bremerich J, Reddy GP, Higgins CB. MRI of supracristal ventricular
Cardiovasc Magn Reson. 2005;7(5):775–782. septal defects. J Comput Assist Tomogr. 1999;23(1):13–15.
10. Kaiser T, Kellenberger CJ, Albisetti M, Bergstrasser E, Valsangiacomo 29. Lowell DG, Turner DA, Smith SM, et al. The detection of atrial and
Buechel ER. Normal values for aortic diameters in children and ventricular septal defects with electrocardiographically synchronized
adolescents—assessment in vivo by contrast-enhanced CMR- magnetic resonance imaging. Circulation. 1986;73(1):89–94.
angiography. J Cardiovasc Magn Reson. 2008;10:56. 30. Baker EJ, Ayton V, Smith MA, et al. Magnetic resonance imaging at a
11. Kawel-Boehm N, Maceira A, Valsangiacomo-Buechel ER, et al. Normal high field strength of ventricular septal defects in infants. Br Heart J.
values for cardiovascular magnetic resonance in adults and children. 1989;62(4):305–310.
J Cardiovasc Magn Reson. 2015;17:29. 31. Mohiaddin RH, Underwood R, Romeira L, et al. Comparison between
12. Sarikouch S, Koerperich H, Boethig D, et al. Reference values for atrial cine magnetic resonance velocity mapping and first-pass radionuclide
size and function in children and young adults by cardiac MR: a study angiocardiography for quantitating intracardiac shunts. Am J Cardiol.
of the German competence network congenital heart defects. J Magn 1995;75(7):529–532.
Reson Imaging. 2011;33(5):1028–1039. 32. Fortescue EB, Lock JE, Galvin T, McElhinney DB. To close or not to
13. Raghib G, Ruttenberg HD, Anderson RC, Amplatz K, Adams P Jr, close: the very small patent ductus arteriosus. Congenit Heart Dis.
Edwards JE. Termination of left superior vena cava in left atrium, atrial 2010;5(4):354–365.
septal defect, and absence of coronary sinus; a developmental complex. 33. Geva T, Greil GF, Marshall AC, Landzberg M, Powell AJ. Gadolinium-
Circulation. 1965;31:906–918. enhanced 3-dimensional magnetic resonance angiography of pulmonary
14. Furlan AJ, Reisman M, Massaro J, et al. Closure or medical therapy for blood supply in patients with complex pulmonary stenosis or atresia:
cryptogenic stroke with patent foramen ovale. N Engl J Med. comparison with x-ray angiography. Circulation. 2002;106(4):
2012;366(11):991–999. 473–478.
15. Udell JA, Opotowsky AR, Khairy P, et al. Patent foramen ovale closure vs 34. Dyme JL, Prakash A, Printz BF, Kaur A, Parness IA, Nielsen JC.
medical therapy for stroke prevention: meta-analysis of randomized Physiology of isolated anomalous pulmonary venous connection of a
trials and review of heterogeneity in meta-analyses. Can J Cardiol. 2014. single pulmonary vein as determined by cardiac magnetic resonance
16. Durongpisitkul K, Tang NL, Soongswang J, Laohaprasitiporn D, Nanal imaging. Am J Cardiol. 2006;98(1):107–110.
A. Predictors of successful transcatheter closure of atrial septal defect by 35. Valsangiacomo Buechel ER, Grosse-Wortmann L, Fratz S, et al.
cardiac magnetic resonance imaging. Pediatr Cardiol. 2004;25(2): Indications for cardiovascular magnetic resonance in children with
124–130. congenital and acquired heart disease: an expert consensus paper of the
17. Beerbaum P, Korperich H, Esdorn H, et al. Atrial septal defects in Imaging Working Group of the AEPC and the Cardiovascular Magnetic
pediatric patients: noninvasive sizing with cardiovascular MR imaging. Resonance Section of the EACVI. Eur Heart J Cardiovasc Imaging.
Radiology. 2003;228(2):361–369. 2015;16(3):281–297.
18. Piaw CS, Kiam OT, Rapaee A, et al. Use of non-invasive phase contrast 36. Riesenkampff EM, Schmitt B, Schnackenburg B, et al. Partial anomalous
magnetic resonance imaging for estimation of atrial septal defect size pulmonary venous drainage in young pediatric patients: the role of
and morphology: a comparison with transesophageal echo. Cardiovasc magnetic resonance imaging. Pediatr Cardiol. 2009;30(4):458–464.
Intervent Radiol. 2006;29(2):230–234. 37. Greil GF, Powell AJ, Gildein HP, Geva T. Gadolinium-enhanced
19. Teo KS, Disney PJ, Dundon BK, et al. Assessment of atrial septal defects three-dimensional magnetic resonance angiography of pulmonary and
in adults comparing cardiovascular magnetic resonance with systemic venous anomalies. J Am Coll Cardiol. 2002;39(2):335–341.
485.e2 SECTION IV  Right Ventricular and Congenital Heart Disease

38. Prasad SK, Soukias N, Hornung T, et al. Role of magnetic resonance significance than arm-leg blood pressure gradient after repair of
angiography in the diagnosis of major aortopulmonary collateral coarctation of the aorta. J Magn Reson Imaging. 2003;17(2):177–183.
arteries and partial anomalous pulmonary venous drainage. Circulation. 58. Julsrud PR, Breen JF, Felmlee JP, Warnes CA, Connolly HM, Schaff HV.
2004;109(2):207–214. Coarctation of the aorta: collateral flow assessment with phase-contrast
39. Ferrari VA, Scott CH, Holland GA, Axel L, Sutton MS. Ultrafast MR angiography. AJR Am J Roentgenol. 1997;169(6):1735–1742.
three-dimensional contrast-enhanced magnetic resonance angiography 59. Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am
and imaging in the diagnosis of partial anomalous pulmonary venous Coll Cardiol. 2002;39(12):1890–1900.
drainage. J Am Coll Cardiol. 2001;37(4):1120–1128. 60. Van Praagh R, Van Praagh S, Nebesar RA, Muster AJ, Sinha SN, Paul
40. Vyas HV, Greenberg SB, Krishnamurthy R. MR imaging and CT MH. Tetralogy of Fallot: underdevelopment of the pulmonary
evaluation of congenital pulmonary vein abnormalities in neonates and infundibulum and its sequelae. Am J Cardiol. 1970;26(1):25–33.
infants. Radiographics. 2012;32(1):87–98. 61. Need LR, Powell AJ, del Nido P, Geva T. Coronary echocardiography in
41. Campbell M. Natural history of coarctation of the aorta. Br Heart J. tetralogy of Fallot: diagnostic accuracy, resource utilization and surgical
1970;32(5):633–640. implications over 13 years. J Am Coll Cardiol. 2000;36(4):1371–1377.
42. Rao PS, Jureidini SB, Balfour IC, Singh GK, Chen SC. Severe aortic 62. Egbe AC, Mittnacht AJ, Nguyen K, Joashi U. Risk factors for morbidity
coarctation in infants less than 3 months: successful palliation by in infants undergoing tetralogy of Fallot repair. Ann Pediatr Cardiol.
balloon angioplasty. J Invasive Cardiol. 2003;15(4):202–208. 2014;7(1):13–18.
43. Chen SS, Dimopoulos K, Alonso-Gonzalez R, et al. Prevalence and 63. Bacha EA, Scheule AM, Zurakowski D, et al. Long-term results after
prognostic implication of restenosis or dilatation at the aortic early primary repair of tetralogy of Fallot. J Thorac Cardiovasc Surg.
coarctation repair site assessed by cardiovascular MRI in adult patients 2001;122(1):154–161.
late after coarctation repair. Int J Cardiol. 2014;173(2):209–215. 64. Nollert G, Fischlein T, Bouterwek S, Bohmer C, Klinner W, Reichart B.
44. Therrien J, Thorne SA, Wright A, Kilner PJ, Somerville J. Repaired Long-term survival in patients with repair of tetralogy of Fallot: 36-year
coarctation: a “cost-effective” approach to identify complications in follow-up of 490 survivors of the first year after surgical repair. J Am
adults. J Am Coll Cardiol. 2000;35(4):997–1002. Coll Cardiol. 1997;30(5):1374–1383.
45. Simpson IA, Chung KJ, Glass RF, Sahn DJ, Sherman FS, Hesselink J. 65. Cuypers JA, Menting ME, Konings EE, et al. Unnatural history of
Cine magnetic resonance imaging for evaluation of anatomy and flow tetralogy of Fallot: prospective follow-up of 40 years after surgical
relations in infants and children with coarctation of the aorta. correction. Circulation. 2014;130(22):1944–1953.
Circulation. 1988;78(1):142–148. 66. Ferraz Cavalcanti PE, Sa MP, Santos CA, et al. Pulmonary valve
46. Mendelsohn AM, Banerjee A, Donnelly LF, Schwartz DC. Is replacement after operative repair of tetralogy of Fallot: meta-analysis
echocardiography or magnetic resonance imaging superior for and meta-regression of 3,118 patients from 48 studies. J Am Coll
precoarctation angioplasty evaluation? Cathet Cardiovasc Diagn. Cardiol. 2013;62(23):2227–2243.
1997;42(1):26–30. 67. O’Byrne ML, Glatz AC, Mercer-Rosa L, et al. Trends in pulmonary valve
47. Didier D, Saint-Martin C, Lapierre C, et al. Coarctation of the aorta: pre replacement in children and adults with tetralogy of Fallot. Am J
and postoperative evaluation with MRI and MR angiography; Cardiol. 2015;115(1):118–124.
correlation with echocardiography and surgery. Int J Cardiovasc Imaging. 68. Valente AM, Cook S, Festa P, et al. Multimodality imaging guidelines for
2006;22(3–4):457–475. patients with repaired tetralogy of Fallot: a report from the American
48. Muzzarelli S, Ordovas KG, Hope MD, Meadows JJ, Higgins CB, Society of Echocardiography: developed in collaboration with the
Meadows AK. Diagnostic value of the flow profile in the distal Society for Cardiovascular Magnetic Resonance and the Society for
descending aorta by phase-contrast magnetic resonance for predicting Pediatric Radiology. J Am Soc Echocardiogr. 2014;27(2):111–141.
severe coarctation of the aorta. J Magn Reson Imaging. 2011;33(6): 69. Kilner PJ, Geva T, Kaemmerer H, Trindade PT, Schwitter J, Webb GD.
1440–1446. Recommendations for cardiovascular magnetic resonance in adults with
49. Steffens JC, Bourne MW, Sakuma H, O’Sullivan M, Higgins CB. congenital heart disease from the respective working groups of the
Quantification of collateral blood flow in coarctation of the aorta by European Society of Cardiology. Eur Heart J. 2010;31(7):794–805.
velocity encoded cine magnetic resonance imaging. Circulation. 70. Silversides CK, Kiess M, Beauchesne L, et al. Canadian Cardiovascular
1994;90(2):937–943. Society 2009 Consensus Conference on the management of adults with
50. Nielsen JC, Powell AJ, Gauvreau K, Marcus EN, Prakash A, Geva T. congenital heart disease: outflow tract obstruction, coarctation of the
Magnetic resonance imaging predictors of coarctation severity. aorta, tetralogy of Fallot, Ebstein anomaly and Marfan’s syndrome. Can
Circulation. 2005;111(5):622–628. J Cardiol. 2010;26(3):e80–e97.
51. Muzzarelli S, Meadows AK, Ordovas KG, et al. Prediction of 71. Geva T. Repaired tetralogy of Fallot: the roles of cardiovascular magnetic
hemodynamic severity of coarctation by magnetic resonance imaging. resonance in evaluating pathophysiology and for pulmonary valve
Am J Cardiol. 2011;108(9):1335–1340. replacement decision support. J Cardiovasc Magn Reson. 2011;139.
52. Mohiaddin RH, Kilner PJ, Rees S, Longmore DB. Magnetic resonance 72. Valente AM, Gauvreau K, Assenza GE, et al. Contemporary predictors of
volume flow and jet velocity mapping in aortic coarctation. J Am Coll death and sustained ventricular tachycardia in patients with repaired
Cardiol. 1993;22(5):1515–1521. tetralogy of Fallot enrolled in the INDICATOR cohort. Heart.
53. Oshinski JN, Parks WJ, Markou CP, et al. Improved measurement of 2014;100(3):247–253.
pressure gradients in aortic coarctation by magnetic resonance imaging. 73. Babu-Narayan SV, Kilner PJ, Li W, et al. Ventricular fibrosis suggested by
J Am Coll Cardiol. 1996;28(7):1818–1826. cardiovascular magnetic resonance in adults with repaired tetralogy of
54. Rengier F, Delles M, Eichhorn J, et al. Noninvasive pressure difference Fallot and its relationship to adverse markers of clinical outcome.
mapping derived from 4D flow MRI in patients with unrepaired and Circulation. 2006;113(3):405–413.
repaired aortic coarctation. Cardiovasc Diagn Ther. 2014;4(2):97–103. 74. Chen CA, Dusenbery SM, Valente AM, Powell AJ, Geva T. Myocardial
55. Riesenkampff E, Fernandes JF, Meier S, et al. Pressure fields by ECV fraction assessed by CMR is associated with type of hemodynamic
flow-sensitive, 4D, velocity-encoded CMR in patients with aortic load and arrhythmia in repaired tetralogy of Fallot. JACC Cardiovasc
coarctation. JACC Cardiovasc Imaging. 2014;7(9):920–926. Imaging. 2016;9(1):1–10.
56. Holmqvist C, Stahlberg F, Hanseus K, et al. Collateral flow in coarctation 75. Broberg CS, Huang J, Hogberg I, et al. Diffuse LV myocardial fibrosis
of the aorta with magnetic resonance velocity mapping: correlation to and its clinical associations in adults with repaired tetralogy of Fallot.
morphological imaging of collateral vessels. J Magn Reson Imaging. JACC Cardiovasc Imaging. 2016;9(1):86–87.
2002;15(1):39–46. 76. Blume ED, Altmann K, Mayer JE, Colan SD, Gauvreau K, Geva T.
57. Araoz PA, Reddy GP, Tarnoff H, Roge CL, Higgins CB. MR findings of Evolution of risk factors influencing early mortality of the arterial
collateral circulation are more accurate measures of hemodynamic switch operation. J Am Coll Cardiol. 1999;33(6):1702–1709.
CHAPTER 40  Simple and Complex Congenital Heart Disease: Infants and Children 485.e3

77. Khairy P, Clair M, Fernandes SM, et al. Cardiovascular outcomes after 95. Manso B, Castellote A, Dos L, Casaldaliga J. Myocardial perfusion
the arterial switch operation for D-transposition of the great arteries. magnetic resonance imaging for detecting coronary function anomalies
Circulation. 2013;127(3):331–339. in asymptomatic paediatric patients with a previous arterial switch
78. Fricke TA, d’Udekem Y, Richardson M, et al. Outcomes of the arterial operation for the transposition of great arteries. Cardiol Young.
switch operation for transposition of the great arteries: 25 years of 2010;20(4):410–417.
experience. Ann Thorac Surg. 2012;94(1):139–145. 96. Taylor AM, Dymarkowski S, Hamaekers P, et al. MR coronary
79. Villafane J, Lantin-Hermoso MR, Bhatt AB, et al; American College of angiography and late-enhancement myocardial MR in children who
Cardiology’s Adult Congenital and Pediatric Cardiology Council. underwent arterial switch surgery for transposition of great arteries.
D-transposition of the great arteries: the current era of the arterial Radiology. 2005;234(2):542–547.
switch operation. J Am Coll Cardiol. 2014;64(5):498–511. 97. Fontan F, Baudet E. Surgical repair of tricuspid atresia. Thorax.
80. Silversides CK, Salehian O, Oechslin E, et al. Canadian Cardiovascular 1971;26(3):240–248.
Society 2009 Consensus Conference on the management of adults with 98. Brown DW, Gauvreau K, Powell AJ, et al. Cardiac magnetic resonance
congenital heart disease: complex congenital cardiac lesions. Can J versus routine cardiac catheterization before bidirectional Glenn
Cardiol. 2010;26(3):e98–e117. anastomosis in infants with functional single ventricle: a prospective
81. Lorenz CH, Walker ES, Graham TP Jr, Powers TA. Right ventricular randomized trial. Circulation. 2007;116(23):2718–2725.
performance and mass by use of cine MRI late after atrial repair of 99. Brown DW, Gauvreau K, Powell AJ, et al. Cardiac magnetic resonance
transposition of the great arteries. Circulation. 1995;92(9 suppl): versus routine cardiac catheterization before bidirectional Glenn
II233–II239. anastomosis: long-term follow-up of a prospective randomized trial.
82. Mooij CF, de Wit CJ, Graham DA, Powell AJ, Geva T. Reproducibility of J Thorac Cardiovasc Surg. 2013;146(5):1172–1178.
MRI measurements of right ventricular size and function in patients 100. Grosse-Wortmann L, Al-Otay A, Yoo SJ. Aortopulmonary collaterals
with normal and dilated ventricles. J Magn Reson Imaging. after bidirectional cavopulmonary connection or Fontan completion:
2008;28(1):67–73. quantification with MRI. Circ Cardiovasc Imaging. 2009;2(3):
83. Groenink M, Mulder BJ, van der Wall EE. Value of magnetic resonance 219–225.
imaging in functional assessment of baffle obstruction after the Mustard 101. Whitehead KK, Gillespie MJ, Harris MA, Fogel MA, Rome JJ.
procedure. J Cardiovasc Magn Reson. 1999;1(1):49–51. Noninvasive quantification of systemic-to-pulmonary collateral flow: a
84. Johansson B, Babu-Narayan SV, Kilner PJ, Cannell TM, Mohiaddin RH. major source of inefficiency in patients with superior cavopulmonary
3-Dimensional time-resolved contrast-enhanced magnetic resonance connections. Circ Cardiovasc Imaging. 2009;2(5):405–411.
angiography for evaluation late after the mustard operation for 102. Prakash A, Khan MA, Hardy R, Torres AJ, Chen JM, Gersony WM. A
transposition. Cardiol Young. 2010;20(1):1–7. new diagnostic algorithm for assessment of patients with single ventricle
85. Babu-Narayan SV, Goktekin O, Moon JC, et al. Late gadolinium before a Fontan operation. J Thorac Cardiovasc Surg. 2009;138(4):
enhancement cardiovascular magnetic resonance of the systemic 917–923.
right ventricle in adults with previous atrial redirection surgery for 103. Ro PS, Rychik J, Cohen MS, Mahle WT, Rome JJ. Diagnostic assessment
transposition of the great arteries. Circulation. 2005;111(16): before Fontan operation in patients with bidirectional cavopulmonary
2091–2098. anastomosis: are noninvasive methods sufficient? J Am Coll Cardiol.
86. Fratz S, Hauser M, Bengel FM, et al. Myocardial scars determined by 2004;44(1):184–187.
delayed-enhancement magnetic resonance imaging and positron 104. Banka P, McElhinney DB, Bacha EA, et al. What is the clinical utility of
emission tomography are not common in right ventricles with systemic routine cardiac catheterization before a Fontan operation? Pediatr
function in long-term follow up. Heart. 2006;92(11):1673–1677. Cardiol. 2010;31(7):977–985.
87. Rydman R, Gatzoulis MA, Ho SY, et al. Systemic right ventricular 105. Fogel MA, Pawlowski TW, Whitehead KK, et al. Cardiac magnetic
fibrosis detected by cardiovascular magnetic resonance is associated with resonance and the need for routine cardiac catheterization in single
clinical outcome, mainly new-onset atrial arrhythmia, in patients after ventricle patients prior to Fontan: a comparison of 3 groups: pre-Fontan
atrial redirection surgery for transposition of the great arteries. Circ CMR versus cath evaluation. J Am Coll Cardiol. 2012;60(12):1094–1102.
Cardiovasc Imaging. 2015;8(5). 106. Margossian R, Schwartz ML, Prakash A, et al; Pediatric Heart Network
88. Preim U, Hoffmann J, Lehmkuhl L, et al. Systemic right ventricles rarely Investigators. Comparison of echocardiographic and cardiac magnetic
show myocardial scars in cardiac magnetic resonance delayed- resonance imaging measurements of functional single ventricular
enhancement imaging. Clin Res Cardiol. 2013;102(5):337–344. volumes, mass, and ejection fraction (from the Pediatric Heart Network
89. Beek FJ, Beekman RP, Dillon EH, et al. MRI of the pulmonary artery Fontan Cross-Sectional Study). Am J Cardiol. 2009;104(3):419–428.
after arterial switch operation for transposition of the great arteries. 107. Rathod RH, Prakash A, Kim YY, et al. Cardiac magnetic resonance
Pediatr Radiol. 1993;23(5):335–340. parameters predict transplantation-free survival in patients with Fontan
90. Weiss F, Habermann CR, Lilje C, et al. [MRI of pulmonary arteries in circulation. Circ Cardiovasc Imaging. 2014;7(3):502–509.
follow-up after arterial-switch-operation (ASO) for transposition of 108. Rathod RH, Prakash A, Powell AJ, Geva T. Myocardial fibrosis identified
great arteries (d-TGA)]. Rofo. 2005;177(6):849–855. by cardiac magnetic resonance late gadolinium enhancement is
91. Gutberlet M, Boeckel T, Hosten N, et al. Arterial switch procedure for associated with adverse ventricular mechanics and ventricular
D-transposition of the great arteries: quantitative midterm evaluation of tachycardia late after Fontan operation. J Am Coll Cardiol.
hemodynamic changes with cine MR imaging and phase-shift velocity 2010;55(16):1721–1728.
mapping-initial experience. Radiology. 2000;214(2):467–475. 109. Grosse-Wortmann L, Drolet C, Dragulescu A, et al. Aortopulmonary
92. Taylor AM, Thorne SA, Rubens MB, et al. Coronary artery imaging in collateral flow volume affects early postoperative outcome after Fontan
grown up congenital heart disease: complementary role of magnetic completion: a multimodality study. J Thorac Cardiovasc Surg.
resonance and x-ray coronary angiography. Circulation. 2012;144(6):1329–1336.
2000;101(14):1670–1678. 110. Glatz AC, Harrison N, Small AJ, et al. Factors associated with systemic
93. Tobler D, Motwani M, Wald RM, et al. Evaluation of a comprehensive to pulmonary arterial collateral flow in single ventricle patients with
cardiovascular magnetic resonance protocol in young adults late after superior cavopulmonary connections. Heart. 2015;101(22):1813–1818.
the arterial switch operation for d-transposition of the great arteries. 111. Whitehead KK, Harris MA, Glatz AC, et al. Status of systemic to
J Cardiovasc Magn Reson. 2014;16:98. pulmonary arterial collateral flow after the fontan procedure. Am J
94. Marin Rodriguez C, Lancharro Zapata A, Rodriguez Ogando A, et al. Cardiol. 2015;115(12):1739–1745.
[Quality of 3D magnetic resonance imaging of coronary arteries in 112. Odenwald T, Quail MA, Giardini A, et al. Systemic to pulmonary
patients with D-transposition of the great arteries after the Jatene switch collateral blood flow influences early outcomes following the total
procedure]. Radiologia. 2015;57(4):326–332. cavopulmonary connection. Heart. 2012;98(12):934–940.
485.e4 SECTION IV  Right Ventricular and Congenital Heart Disease

113. Prakash A, Rathod RH, Powell AJ, McElhinney DB, Banka P, Geva T. real-time magnetic resonance flow study. Circulation. 2003;108(10):
Relation of systemic-to-pulmonary artery collateral flow in single 1227–1231.
ventricle physiology to palliative stage and clinical status. Am J Cardiol. 116. Be’eri E, Maier SE, Landzberg MJ, Chung T, Geva T. In vivo evaluation
2012;109(7):1038–1045. of Fontan pathway flow dynamics by multidimensional phase-velocity
114. Khiabani RH, Whitehead KK, Han D, et al. Exercise capacity in magnetic resonance imaging. Circulation. 1998;98(25):2873–2882.
single-ventricle patients after Fontan correlates with haemodynamic 117. Haggerty CM, Kanter KR, Restrepo M, et al. Simulating hemodynamics
energy loss in TCPC. Heart. 2015;101(2):139–143. of the Fontan Y-graft based on patient-specific in vivo connections.
115. Hjortdal VE, Emmertsen K, Stenbog E, et al. Effects of exercise and J Thorac Cardiovasc Surg. 2013;145(3):663–670.
respiration on blood flow in total cavopulmonary connection: a
41 
Simple and Complex Congenital Heart
Disease: Adults
Eric V. Krieger, Sara L. Partington, and Anne Marie Valente

The number of adults with congenital heart disease living in the United Surgical Repairs
States is estimated to be at least 1.4 million, and at least 300,000 of these When possible, the surgical intervention for Ebstein anomaly involves
people have complex forms of congenital heart disease.1 The major- tricuspid valve repair. In the current era, the repair generally involves
ity of these patients have undergone surgical repairs in childhood, a right atrial (RA) reduction, plication of the atrialized right ventricle
and lifelong follow-up is recommended. Many adults with congenital and a tricuspid valve repair involving the creation of a monocusp tri-
heart disease do not recognize subtle changes in exercise capacity and cuspid valve using the redundant anterior tricuspid valve leaflet with
serial imaging of adults with congenital heart disease is important a posterior annuloplasty (cone procedure).2 In patients who are unsuit-
to monitor for hemodynamic and anatomic sequelae. Cardiovascular able for tricuspid valve repair, a bioprosthetic tricuspid valve replacement
magnetic resonance (CMR) imaging has become an attractive imaging is generally performed. Evaluation of the patient after Ebstein repair
modality for surveillance of the long-term cardiac complications in involves assessing the degree of residual tricuspid regurgitation or ste-
this population because of its excellent tissue border delineation, tissue nosis, assessing RA size and the RV size and function. The right coronary
characterization, and quantification of biventricular volumes and val- artery lies in close proximity to the repaired tricuspid valve annulus
vular regurgitation. CMR allows for serial comparisons without the site and it is important to assess for evidence of late gadolinium enhance-
need for ionizing radiation or iodinated contrast. The objective of this ment (LGE) following surgery for Ebstein anomaly.
chapter is to review select congenital heart disease diagnoses that are
referred for CMR imaging. For each congenital heart disease condition, Cardiovascular Magnetic Resonance Evaluation of
we will present a suggested CMR protocol with the acknowledge- Ebstein Anomaly
ment that a there is considerable anatomic variability and individu- Tricuspid Valve Anatomy and Tricuspid
alization of protocols is often required. Knowledge of the patient’s Regurgitation Fraction
anatomy, surgical interventions, and prior imaging is critical to focus An accurate assessment of the tricuspid valve anatomy, including assess-
the protocol so that the essential information is obtained within a ing the leaflet sizes, the subvalvar apparatus, the presence of tethering
reasonable amount of time, as many of these adults will undergo serial attachments of the anterior leaflet to the RV free wall, and the location
examinations. of the tricuspid valve functional annulus, is important for planning
repair. Understanding the amount and structure of the tricuspid leaflet
EBSTEIN ANOMALY tissue provides the surgeon information on whether enough leaflet
tissue is accessible to repair, rather than replace, the tricuspid valve.
Anatomy and Natural History Balanced steady-state free precession (bSSFP) cine imaging of the
Ebstein anomaly is a malformation of the tricuspid valve and right tricuspid valve can be performed in the four-chamber view and the RV
ventricle, which encompasses a large spectrum of disease severity. There two-chamber view. It is important to determine the orientation of the
is failure of delamination of the septal and posterior tricuspid valve tricuspid valve inflow because it may be oriented toward the right ven-
leaflets from the myocardium, resulting in apical displacement of the tricular outflow tract (RVOT) or directed at the RV apex; therefore
tricuspid valve (≥0.8 cm/m2 relative to the anterior mitral leaflet inser- off-axis views are often required. Determining the severity of tricuspid
tion site). The portion of the right ventricle proximal to the functional regurgitation may be challenging in patients with Ebstein anomaly but
tricuspid valve becomes “atrialized.” The anterior leaflet of the tricuspid may be quantified with phase contrast CMR (PCMR).3
valve is large and redundant, and often has fenestrations and tethering
attachments to the right ventricular (RV) free wall. The posterior leaflet Right Atrial and Right Ventricular Size and Function
is often dysplastic and atrially displaced. Progressive tricuspid regurgitation and abnormal RV myocardium lead
The etiology of Ebstein anomaly is not known; however, in rare to right heart dilation. Measurements of RA and RV volumes are typi-
cases, genetic factors such as mutations in the transcription factor cally performed in axial or short-axis orientation with endocardial
NKX2.5,10p13–p14 deletion, or 1p34.3–p36.11 deletion have been contours drawn in end systole and end diastole.4 When contouring the
described. The most severe cases of Ebstein anomaly result in fetal right ventricle, it can be useful to assess the functional RV size that
hydrops and fetal demise. Patients with Ebstein anomaly often develop excludes the atrialized right ventricle and only includes the RV volume
progressive right heart failure and exercise intolerance. However, some distal to the functional tricuspid valve annulus and coaptation point.
individuals with mild forms of this disease can be asymptomatic into Many centers will report both the functional RV size and function and
adulthood. the anatomic RV size and function (using the anatomic tricuspid annulus

486
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 487

as a landmark). The ability to accurately and reproducibly measure RV noncompaction. CMR can be used to assess the morphology of the
volume is important because RV size has been associated with clinical interatrial septum and to calculate the Qp:Qs ratio by performing PCMR
outcomes in patients with Ebstein anomaly.5 Additionally, mismeasure- across the aorta and pulmonary artery.7
ment of RV stroke volume can result in errors when calculating tricuspid A suggested CMR protocol for adults with Ebstein anomaly is pre-
regurgitation severity. sented in Box 41.1.
Progressive RV dilation or dysfunction in the setting of severe tri-
cuspid regurgitation is imaging criteria for tricuspid valve surgical COARCTATION OF THE AORTA
intervention. The index of right-sided to left-sided heart volumes derived
from CMR has been shown to correlate well with other established Anatomy and Natural History
markers of heart failure.6 Contouring the end-diastolic volume mea- Coarctation of the aorta is typically a fibrous ridge in the aortic isthmus,
surements of the right ventricle, RA, left ventricle, and left atrium (LA) just distal to the insertion of the left subclavian artery. Although often
can provide a total right/left-volume index (Fig. 41.1): discrete, aortic coarctation can also be long segment or associated with
a diffusely hypoplastic transverse aorta. Aortic coarctation is associated
Total right left-volume index with a diffuse vascular abnormality and is associated with aortic aneu-
= (right atrium + atrialized right ventricle rysm, cerebral aneurysms, or endothelial dysfunction.8 Awareness of
+ functional right ventricle) (LA + left ventricle) Eq. 41.1 these abnormalities is important because the entire aorta and cerebral
vessels should be imaged in patients with aortic coarctation.
Indexes such as these may eventually be used to help guide timing Aortic coarctation sometimes presents in adulthood as difficult-to-
of tricuspid valve intervention. manage systemic hypertension and the diagnosis may be suspected
from a murmur, diminished lower extremity pulses, or arm–arm or
Associated Cardiac Abnormalities arm–leg blood pressure discrepancy. Those who had an aortic coarcta-
Patients with Ebstein anomaly commonly have atrial septal defects tion repair early in life have up to a 5% risk of re-coarctation in adult-
(ASDs), and may occasionally have RVOT obstruction, or myocardial hood and present similarly.9

BOX 41.1  Cardiovascular Magnetic


Resonance Protocol for Ebstein Anomaly
RV Localizers
Electrocardiogram-gated cine balanced steady-state free precession
• Axial stack through the heart and branch pulmonary arteries
• Three-chamber right ventricular stack, two-chamber left, four-chamber
RA views
• Ventricular short-axis stack from the base to the apex
• Right ventricular outflow tract view parallel to the right ventricular outflow
tract
• Right ventricular two-chamber view
A • Oblique planes to image tricuspid valve inflow orientation
• Oblique sagittal plane to image the atrial septum
Contrast-enhanced three-dimensional magnetic resonance angiograma
Electrocardiogram-gated phase contrast through-plane flow in the main pul-
monary artery, aorta, tricuspid and mitral valvesb
fRV Late gadolinium enhancement to assess for myocardial fibrosisc
These protocols were adapted from Boston Children’s Hospital
aRV Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-
RA dimensional magnetic resonance angiogram with the first
cardiovascular magnetic resonance (CMR) examination, and then
optional for subsequent examinations.
b
In cases where Qp:Qs ratio is of interest, flow across the branch
pulmonary arteries, superior vena cava, and inferior vena cava/
B descending thoracic aorta may be assessed to improve level of
confidence.
FIG. 41.1  Balanced steady-state free precession four-chamber view of c
In general, our practice is to obtain late gadolinium enhancement
a patient with Ebstein anomaly (A) demonstrating the severely dilated (LGE) imaging to assess for myocardial fibrosis with the first CMR
right atrium (RA) and the relatively smaller right ventricle. End-diastolic examination, and the frequency of repeating this technique in
contours (B) outlining the RA, the atrialized right ventricle (aRV, the subsequent examinations is dependent on the initial findings and
portion of the right ventricle distal to the anatomic tricuspid valve annulus specific condition. For example, some CMR laboratories repeat LGE
but proximal to the coaptation point of the tricuspid valve) and the imaging every 3 years in patients with conotruncal anomalies
functional right ventricle (fRV; the portion of the right ventricle distal to (tetralogy of Fallot, transposition of the great arteries, double outlet
the coaptation of the tricuspid valve). right ventricle).
488 SECTION IV  Right Ventricular and Congenital Heart Disease

BOX 41.2  Cardiovascular Magnetic


Resonance Protocol for Aortic Coarctation
Localizers
Electrocardiogram-gated cine balanced steady-state free precession
• Four-chamber view, left ventricular two-chamber and, left ventricular
long-axis view to the aorta
• Ventricular short-axis stack from the base to the apex
• Oblique sagittal view of the aortic arch (“candy-cane” view)
Consider double-inversion recovery black-blood images in the oblique sagittal
plane if artifact on balanced steady-state free precession imaging
Contrast-enhanced three-dimensional magnetic resonance angiogram in sagittal
orientation with first acquisition timed to the aortaa
Electrocardiogram-gated phase contrast through-plane flow in the main pul-
monary artery, aorta and consider proximal descending aorta proximal to
the coarctation (distal to left subclavian artery) and descending aorta at
the level of the diaphragm
Late gadolinium enhancement to assess for myocardial fibrosisb

These protocols were adapted from Boston Children’s Hospital


Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-
FIG. 41.2  Three-dimensional volume-rendered magnetic resonance dimensional magnetic resonance angiogram with the first
angiogram in a patient with an aortic coarctation (arrow). Note the large cardiovascular magnetic resonance (CMR) examination, and then
left internal thoracic artery that functions as a bridging collateral and is optional for subsequent examinations.
a sign of hemodynamically significant coarctation. b
In general, our practice is to obtain late gadolinium enhancement
(LGE) imaging to assess for myocardial fibrosis with the first CMR
examination, and the frequency of repeating this technique in
subsequent examinations is dependent on the initial findings and
Surgical Repairs specific condition. For example, some CMR laboratories repeat LGE
imaging every 3 years in patients with conotruncal anomalies
There are several types of surgical repairs for native aortic coarctation,
(tetralogy of Fallot, transposition of the great arteries, double outlet
and they are dictated by the anatomy. For patients with a discrete coarc-
right ventricle).
tation, an end-to-end anastomosis is performed. If there is associated
transverse aortic arch hypoplasia, an extended end-to-end procedure
may be required.
Some patients with complex aortic coarctation have undergone
interposition graft repairs, and those with Dacron material are particu- Patients with aortic coarctation are prone to aneurysms in the ascend-
larly susceptible to aneurysm formation. The subclavian flap procedure ing aorta, cerebral vasculature, and at the site of surgical repair. Aneu-
was performed in the early surgical experience, and involved transecting rysms at the site of previous Dacron patch repair are particularly prone
the left subclavian artery and placing it over the narrowed portion of to rupture and should be managed aggressively.17–19 Screening for cerebral
the descending aorta. Patients who have undergone this procedure will aneurysms is recommended, although neither optimal management
no longer have the left subclavian artery attached to the transverse arch. nor frequency of surveillance is well defined.20

Cardiovascular Magnetic Resonance Evaluation of Left Ventricular Cavity Size and Function
Aortic Coarctation Patients with native or re-coarctation are predisposed to systemic hyper-
Aortic Arch tension, particularly exercise-induced hypertension. Therefore standard
CMR imaging of native and re-coarctation will be discussed together. measurements of left ventricular (LV) mass and function should be
The anatomic features of aortic coarctation can be seen with bright- performed.21
blood cine or double-inversion black-blood spin echo images in the
oblique sagittal plane. Gadolinium-enhanced magnetic resonance angi- Associated Cardiac Lesions
ography (MRA) also demonstrates the coarctation (Fig. 41.2) and double Over 50% of those with aortic coarctation have a bicommissural aortic
oblique reformats can be used to measure the minimal luminal diameter valve and these patients are at greater risk for ascending aortic aneu-
and the reference vessel size.10,11 If collateral vessels are seen on contrast- rysms.17 Patients may also have subaortic stenosis, mitral valve abnor-
enhanced (CE) MRA or PCMR demonstrates flow augmentation in malities (e.g., parachute mitral valve and/or mitral stenosis) or ventricular
the descending aorta, the obstruction is probably hemodynamically septal defect (VSD).
important.12,13 A prolonged deceleration time of the systolic flow in the A suggested CMR protocol for adults with aortic coarctation is pre-
distal descending thoracic aorta also indicates hemodynamically sig- sented in Box 41.2.
nificant aortic coarctation.11,14
Four-dimensional velocity mapping is not widely available but is TETRALOGY OF FALLOT
an emerging technique to show turbulence at the coarctation site, and
characterize altered aortic velocity profiles, providing insight into which Anatomy and Natural History
patients may be at higher risk for aortic wall complications, such as Evaluation of patients with repaired tetralogy of Fallot (TOF) is one
dissection.15,16 of the most common adult congenital heart disease diagnoses referred
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 489

for CMR. TOF represents the most common form of cyanotic congenital
heart disease affecting up to 0.5 per 1000 live births.22 Survival following Cardiovascular Magnetic Resonance Evaluation of
TOF repair is excellent, but there is a 3-fold increase in mortality begin- Tetralogy of Fallot Repair
ning in the third postoperative decade of life,23 and 14% of patients Right Ventricular Outflow Tract Obstruction and
develop markedly impaired functional status late after surgical repair, Pulmonary Arteries
highlighting the importance of regular surveillance of these patients.24 A goal of TOF surgery is to relieve the RVOT obstruction, yet many
This congenital anomaly results from the anterior deviation of the conal patients are left with varying degrees of obstruction that may be located
septum, resulting in varying degrees of RVOT obstruction, VSD, an in the subpulmonary area, at the level of the pulmonary valve, or more
overriding aorta, and RV hypertrophy. Importantly, the degree of RVOT distally in the main or branch pulmonary arteries (PA). Transannular
obstruction can range from only mild subpulmonary stenosis to the patch repairs can result in RVOT regional wall motion abnormalities
most severe form involving complete absence of the main pulmonary and aneurysms (Fig. 41.3). RVOT aneurysms are not only associated
artery (TOF/pulmonary atresia), which occurs in approximately 15% with decreased RV ejection fraction (EF) but are also associated with
of patients with TOF. unfavorable ventricular interactions, resulting in a reduced LVEF.25
Visualization of the entire RVOT is important using specific RVOT
Surgical Repairs long-axis (Fig. 41.4A) and two-chamber RV cine views (Fig. 41.4B).
In the current era, the majority of patients undergo surgical repair The RV two-chamber view allows visualization of the right atrium,
in infancy or childhood, although older adults may have first under- tricuspid valve, RV, and main pulmonary artery all in one plane. Assess-
gone a palliative shunt (Blalock-Taussig, Waterston, or Potts shunt) as ing for downstream stenosis in the branch pulmonary arteries can be
a young child and then undergone a complete repair at an older age. performed with either a bSSFP stack in an axial plane or an MRA.
Strategies to repair TOF have evolved over time. The early experience Newer catheter-based pulmonary valves are promising developments
involved placing a transannular patch to eliminate the RVOT obstruc- for patients with congenital heart disease affecting the right heart. At
tion; however, current strategies have been modified to alleviate most the current time, most percutaneous valves are placed inside existing
of the RVOT obstruction while trying to preserve some of the integ- RV-PA conduits or dysfunctional bioprosthetic valves. CMR is useful
rity of the pulmonary valve. Patients with TOF/pulmonary atresia and to determine the geometry of the RVOT and identify potential can-
those with anomalous left coronary artery from the right sinus may didates for percutaneous pulmonary valve placement. Delineation of
undergo an RV-to-pulmonary artery (RV-PA) conduit. Knowledge of the the coronary artery course is essential before any RVOT intervention
patient’s surgical history before CMR is ideal because it will determine because 5% to 7% of patients with TOF have an anomalous left coro-
the specific CMR protocol to employ, focusing on the potential residual nary artery that may course across the RVOT which can complicate
lesions. both surgical or transcatheter pulmonic valve implantation. Cardiac
Many patients with repaired TOF undergo a pulmonary valve replace- and respiratory-gated MRA images have sufficient spatial resolution
ment in adulthood. CMR is increasingly being used to aid in the decision to assess the origins and proximal coronary artery courses.
making of timing of these interventions. Box 41.3 lists suggested imaging
criteria for consideration of pulmonary valve replacement in asymp- Pulmonary Regurgitation
tomatic patients with severe pulmonary regurgitation (PR). Pulmonary regurgitation is the most common sequelae of TOF repair
and is often associated with RV dilation (Fig. 41.5), predisposing to RV
dysfunction, arrhythmias, and death. PCMR sequences assess antegrade

BOX 41.3  Imaging Criteria for Pulmonary


Valve Replacement in Asymptomatic
Patients With Repaired Tetralogy of Fallot
With Severe Pulmonary Regurgitation
• Right ventricular end-diastolic volume index >150 mL/m2 or Z-score >4. In
patients whose body surface area falls outside published normal data: right
ventricle/left ventricle end-diastolic volume ratio >2
• Right ventricular end-systolic volume index >80 mL/m2
• Right ventricular ejection fraction <47%
• Left ventricle ejection fraction <55%
*
• Large right ventricular outflow tract aneurysm
• Other significant hemodynamic abnormalities: RA
• Right ventricular outflow tract obstruction with right ventricular systolic
pressure >2/3 systemic
• Severe branch pulmonary artery stenosis (<30% flow to affected lung)
RV
• More than moderate tricuspid regurgitation
• Left-to-right shunt from residual ventricular or atrial septal defect with
pulmonary to aortic flow ratio >1.5
• Severe aortic regurgitation
• Severe aortic root dilation (>5 cm)
Data from Geva T. Repaired tetralogy of Fallot: the roles of FIG. 41.3  Three-dimensional volume-rendered magnetic resonance
cardiovascular magnetic resonance in evaluating pathophysiology and angiogram in a patient with a transannular patch repair for tetralogy
for pulmonary valve replacement decision support. J Cardiovasc of Fallot with a right ventricular outflow tract aneurysm (*). RA, Right
Magn Reson. 2011;13:9. atrium; RV, right ventricle.
490 SECTION IV  Right Ventricular and Congenital Heart Disease

MPA
MPA

LV RA
RV RV

A B
FIG. 41.4  Cine balanced steady-state free precession with right ventricular views for evaluating patients
with tetralogy of Fallot. (A) Right ventricular outflow tract view demonstrates the right ventricle (RV) and
pulmonary valve and delineates right ventricular outflow tract. (B) Right ventricular two-chamber view dem-
onstrates the RV, tricuspid and pulmonary valve in a single plane. Arrows denote right ventricular outflow
tract aneurysm. LV, Left ventricle; MPA, main pulmonary artery; RA, right atrium.

in dramatic decreases in RV volumes and improvement in functional


status,30 but studies have demonstrated that RV systolic function gener-
ally remains unchanged.31,32
CMR is the gold standard for quantifying RV size and function in
patients with repaired TOF and results in significantly better interob-
server variability than transthoracic echocardiography (TTE).33,34 LV
RV dysfunction is present in >20% of adults with TOF repair, particularly
those who were repaired later in life, had prior palliative shunts, or
concomitant RV dysfunction. LV dysfunction has been associated with
RA sudden cardiac death in this patient population.28,35

LV Tricuspid Regurgitation
There are several mechanisms that lead to tricuspid regurgitation in
repaired TOF patients, including annular dilation, structural valve
abnormalities, or as a consequence of valve disruption during prior
TOF repair.36 CMR allows for quantification of tricuspid regurgitation,
and significant tricuspid regurgitation should be considered in surgical
plans at the time of pulmonic valve replacement.
FIG. 41.5  Balanced steady-state free precession imaging of the four-
chamber view in a patient with tetralogy of Fallot demonstrating right Ascending Aorta
ventricular dilation secondary to long-standing pulmonary regurgitation. Ascending aortic dilation is common in adults with repaired TOF.
When planning the short-axis stack, it is important to include the basal However, despite large aortic dimensions, aortic dissection is exceed-
lateral portion of the right ventricle that often extends well beyond the ingly rare. In fact, up to 25% of adults with repaired TOF have an aortic
mitral annular plane. LV, Left ventricle; RA, right atrium; RV, right ventricle. root diameter >4 cm; however, only 2.3% have an aortic diameter >5 cm.37
Some patients develop progressive dilation of the aortic root that can
and retrograde flow across the main pulmonary artery (Fig. 41.6), and lead to significant aortic regurgitation.38
studies demonstrate this to be a highly reproducible technique for
quantifying pulmonary regurgitation volume.26 Pulmonary regurgita- Myocardial Fibrosis
tion fraction is calculated as retrograde flow volume divided by antegrade LGE occurs commonly in locations of prior surgery (RVOT, VSD patch).
flow volume and, in our laboratory, a pulmonary regurgitation fraction Repaired TOF patients with greater degrees of LGE are at a higher risk
greater than 40% is considered severe. of sustained symptomatic arrhythmia; however, it is unclear if LGE is
associated with increased mortality in this patient population. LGE in
Right Ventricular Size and Function the area of the VSD patch, RVOT patch, or septal insertion sites are
Accurate quantification of RV size, function, and mass is particularly not associated with worse outcomes.39 Novel techniques such as T1
important in repaired TOF patients because RV dilatation, dysfunc- mapping (see Chapter 2), which acts as a marker for diffuse fibrosis,
tion, and hypertrophy are associated with adverse outcomes in this may also prove to act as prognostic indicators in patients with TOF;
group.27,28 Pulmonary valve replacement usually eliminates significant however, T1 mapping in the RV can be difficult, and definitive studies
pulmonary regurgitation; however, optimal timing of pulmonic valve are lacking.40,41
replacement to prevent the adverse sequelae of RV dilation and dys- A suggested CMR protocol for adults after TOF repair is presented
function remains unclear.29 Pulmonic valve replacement usually results in Box 41.4.
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 491

MPA MPA

A B
600

400
Antegrade
Flow (mL/s)

200 PA flow

0
Retrograde
PA flow
–200

–400
0 100 200 300 400 500 600 700 800
C Time (ms)
FIG. 41.6  Phase contrast cardiovascular magnetic resonance of the pulmonary valve to assess pulmonary
regurgitation volume and regurgitation fraction. (A) Magnitude imaging of the main pulmonary artery (MPA).
(B) Phase contrast imaging of the MPA. (C) Flow profile demonstrating the antegrade flow, representing the
area under the curve above the horizontal axis, and the retrograde flow, representing area under the curve
below the horizontal axis. There is severe pulmonary regurgitation with a regurgitation fraction of 47%. PA,
Pulmonary artery.

BOX 41.4  Cardiovascular Magnetic Resonance Protocol for Repaired Tetralogy of Fallot
Localizers Three-dimensional contrast-enhanced magnetic resonance angiograma
Electrocardiogram-gated cine balanced steady-state free precession Electrocardiogram-gated phase contrast through-plane flow in the main pulmonary
• Four-chamber view and two-chamber left ventricular, right ventricular views artery, aorta, atrioventricular valvesb
• Ventricular short-axis stack from the base to the apex Late gadolinium enhancement to assess for myocardial fibrosisc
• Right ventricular outflow tract outflow view parallel to the right ventricular Consider coronary artery imaging with electrocardiogram and respiratory navigator
outflow tract, then a second view orthogonal to the first right ventricular gated three-dimensional balanced steady-state free precession
outflow tract view, right ventricular outflow tract short-axis view Consider T1 mapping to assess for diffuse fibrosis
• Axial stack to assess for branch pulmonary arteries and outflow tracts

These protocols were adapted from Boston Children’s Hospital Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-dimensional magnetic resonance angiogram with the first cardiovascular
magnetic resonance (CMR) examination, and then optional for subsequent examinations.
b
In cases where Qp:Qs ratio is of interest, flow across the branch pulmonary arteries, superior vena cava, and inferior vena cava/descending
thoracic aorta may be assessed to improve level of confidence.
c
In general, our practice is to obtain late gadolinium enhancement (LGE) imaging to assess for myocardial fibrosis with the first CMR
examination, and the frequency of repeating this technique in subsequent examinations is dependent on the initial findings and specific
condition. For example, some CMR laboratories repeat LGE imaging every 3 years in patients with conotruncal anomalies (tetralogy of Fallot,
transposition of the great arteries, double outlet right ventricle).
492 SECTION IV  Right Ventricular and Congenital Heart Disease

RV

* LV

A B
FIG. 41.7  (A) The atrial switch procedure (Mustard or Senning) for D-loop transposition of the great arteries.
The atrial switch procedure uses intra-atrial baffles to redirect systemic venous return to the subpulmonic
left ventricle and pulmonary venous return to the systemic right ventricle. (B) Balanced steady-state free
precession image in a four-chamber orientation. Note the thin-walled subpulmonic left ventricle (LV) and the
hypertrophied systemic right ventricle (RV). Asterisk denotes the pulmonary venous pathway. (A, From Otto
C, ed. The Practice of Clinical Echocardiography. 5th ed. Philadelphia: Elsevier; 2017:951.)

D-LOOP TRANSPOSITION OF THE GREAT ARTERIES


Anatomy and Natural History
D-loop transposition of the great arteries (TGA) is characterized by
atrioventricular concordance and ventriculoarterial discordance. This
results in parallel circulations, which sends oxygenated blood from the
LA to the left ventricle to the pulmonary artery, while deoxygenated
blood flows from the RA to the right ventricle to the aorta. It is a
common form of cyanotic congenital heart disease and occurs in 3 out
of 10,000 live births, accounting for 5% to 7% of all congenital heart
disease defects. Without intervention, the mortality rate is >90% in the
first year of life.42 Approximately 50% those with D-loop TGA have an
associated cardiac anomaly. Common lesions include VSD (40%), LV
outflow tract (LVOT) obstruction (25%), or aortic coarctation (5%).
The coronary artery pattern is also variable, with a high frequency of
anomalous and interarterial coronary courses.43–45

Surgical Repairs
From the late 1950s until the early 1990s the atrial switch procedure
was used to treat D-loop TGA and is often described by the eponyms
the Mustard or Senning procedures (Fig. 41.7).46,47 Even though the
atrial switch has been largely abandoned (see later), there is a large
population of adults previously treated with the atrial switch who are
referred for CMR. During the atrial switch, systemic venous (deoxygen-
ated) blood is rerouted leftward through the atrium to the mitral valve
and left ventricle by intraatrial baffles. Pulmonary venous blood is
FIG. 41.8  The arterial switch procedure. This provides for a systemic
redirected rightwards to the tricuspid valve and right ventricle. Follow-
left ventricle and a subpulmonic right ventricle but requires a neonatal
ing the atrial switch, the right ventricle is the systemic ventricle and coronary transfer. (From Otto C, ed. The Practice of Clinical Echocar-
the left ventricle is the subpulmonic ventricle. Common complications diography. 5th ed. Philadelphia: Elsevier; 2017:951.)
after the atrial switch include venous pathway stenosis, baffle leak, sys-
temic right ventricular systolic dysfunction, tricuspid regurgitation, and
LVOT obstruction.48–50
In the 1990s the arterial switch operation succeeded the atrial they straddle the aorta to come into alignment with the RV. A coro-
switch as the preferred treatment for D-loop TGA (Fig. 41.8). With nary artery translocation is performed. Late complications after the
the arterial switch operation, the great arteries are transected above arterial switch operation include neoaortic dilation and regurgitation,
the sinuses, the aorta is brought posteriorly to align with the left supravalvar pulmonic stenosis, branch pulmonary artery stenosis, and
ventricle while the pulmonary arteries are brought anteriorly where coronary artery obstruction.51–53
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 493

The Rastelli operation is performed for those with a VSD and obstruction so computed tomography (CT) angiography should be
pulmonic stenosis. In the Rastelli operation, a VSD patch is fash- considered.
ioned to baffle oxygenated blood across the VSD to the anterior Many patients with hemodynamically important superior vena cava
aorta. The pulmonic artery is oversewn and an RV to pulmonary obstruction will decompress down the azygous vein. Axial through-
artery conduit is inserted. Late complications include dysfunction plane PCMR should normally confirm azygous flow directed cranially.
of the RV to pulmonary artery conduit, residual VSD, or LVOT If flow is reversed (the descending aorta can be used for reference),
obstruction.54 superior vena caval obstruction is likely.
Baffle leaks allows communication between the systemic and pul-
Cardiovascular Magnetic Resonance Evaluation of the monary venous pathways. These are often small but can be seen using
Atrial Switch Operation thin-slice no-gap bSSFP orthogonal stacks through the atria. Dephasing
Baffle Complications jets can sometimes be seen. If comparison of flow by PCMR through
Baffle complications are the most frequent indication for re-intervention the aorta and pulmonary suggests a shunt, a baffle leak should be
late after the atrial switch operation.43 Narrowing typically occurs in suspected.
the superior vena cava pathway and is particularly common in those
with transvenous pacing or defibrillator leads. Few patients are symp- Systemic Right Ventricle and Tricuspid Valve
tomatic and most are managed conservatively. Inferior vena caval and RV systolic dysfunction is nearly universal in the adult with an atrial
pulmonary venous obstruction are less common. CMR evaluation of switch operation and is associated with poor outcomes.43,57 Detailed
stenosis within the venous pathways requires careful planning to align discussion of CMR evaluation of the systemic RV is discussed in detail
imaging planes. Sagittal planes can identify the systemic and pulmonary in the section on L-loop TGA. Either a short-axis or axial stack should
venous pathways. Imaging planes are obtained through the superior be contoured in end diastole and end systole for measures of RV volumes
vena cava, inferior vena cava, and pulmonary venous pathways (Fig. and function. Reference values for normal RV volumes are not estab-
41.9). The use of orthogonal planes can overcome artifact caused by lished for this population.55,58,59 Myocardial fibrosis, detected by LGE
partial volume averaging.55 Black-blood imaging can be performed in in the systemic right ventricle, has been associated with adverse outcomes,
areas of suspected pathway obstruction for additional anatomic defini- mostly atrial arrhythmias.60
tion. CE-MRA is sensitive in detecting hemodynamically important Tricuspid (systemic atrioventricular) valve regurgitation is common
venous pathway obstruction but may miss milder narrowings.56 Although and often functional, secondary to annular dilation rather than primary
CMR is generally safe in patients with pacemakers (see Chapter 11), leaflet disease (in contrast to L-loop TGA). Tricuspid regurgitation
local susceptibility artifact from pacemaker leads (and especially defi- volume can be calculated by comparing RV stroke volume to flow mea-
brillator leads) reduces the diagnostic accuracy of detecting pathway sured across the aortic valve.

SVC
PV

IVC

FIG. 41.9  An approach to initial plane selection for cardiovascular magnetic resonance of a Mustard/Senning
patient. In sagittal projections the superior vena cava (SVC) and inferior vena cava (IVC) are identified at the
point where the pulmonary venous (PV) pathway transects them. Using these landmarks provides horizontal
and vertical long-axis views (smaller panels at right) that can be used for additional imaging, starting with a
short-axis scout stack. Red denotes the location of the baffle. (From Broberg CS. Cardiac magnetic imaging
of the patient with an atrial switch palliation for transposition of the great arteries. Prog Pediat Cardiol.
2014;38:49–55.)
494 SECTION IV  Right Ventricular and Congenital Heart Disease

Lecompte maneuver. Obstruction in the main pulmonary artery is less


Left Ventricular Outflow Tract Obstruction common but can occur if large coronary artery buttons had been har-
After the atrial switch, LVOT obstruction acts as subpulmonic stenosis. vested from the neopulmonic root. Cine bSSFP images can show the
Obstruction can be caused by a discrete subpulmonic ridge, posterior area of obstruction.
malalignment VSD, or systolic anterior motion of the mitral valve. Cine Branch pulmonary artery stenosis is best shown with CE-MRA
bSSFP images aligned to the LVOT will elucidate the mechanism of (Fig. 41.10). Narrowing is often eccentric leading to an oval lumen.
obstruction.55 Subpulmonic obstruction is not always undesirable because Double-oblique measurements should be performed. PCMR in the
pressure loading the subpulmonic left ventricle may produce a more main, right and left pulmonary arteries allow for calculation of dif-
favorable position of the interventricular septum.48 ferential pulmonary blood flow.
With these potential complications in mind, a suggested CMR pro-
tocol for adults after an atrial switch is presented in Box 41.5. Neoaortic Dilation and Regurgitation
The neoaortic root is native pulmonary artery tissue and often dilates
Cardiovascular Magnetic Resonance Evaluation of the after the arterial switch operation. Dilation is typically slowly progres-
Arterial Switch Operation sive (<0.5 mm/year) and dissection is rare.52,62 Electrocardiogram-gated
Outcomes after the arterial switch operation are better than the out- respiratory navigated three-dimensional (3D) isotropic sequences are
comes after an atrial switch operation, with less arrhythmia, less heart ideal for measuring the neoaortic sinuses because they minimize blur-
failure, and improved functional status.61 ring from cardiac motion. Neoaortic valve regurgitation is often sec-
ondary to annular dilation and can be severe. PCMR in the ascending
Supravalvar Pulmonic Stenosis and Branch Pulmonary aorta can measure regurgitation volume. Slice location affects the degree
Artery Stenosis of regurgitation detected, so consistency is important.63
Supravalvar pulmonic stenosis can occur at the anastomotic site or
where the pulmonary arteries are draped over the aorta as part of the Coronary Arteries
Coronary artery occlusion or ostial stenosis occurs after the coronary
translocation in ~5% after the arterial switch operation.64,65 Wall motion
BOX 41.5  Cardiovascular Magnetic abnormalities or LGE should raise suspicion for coronary injury. Stress
Resonance Protocol for Transposition of the perfusion CMR and coronary imaging using free-breathing navigated
3D bSSFP sequences triggered in diastole can be used to evaluate for
Great Arteries With Atrial Switch Operation
proximal coronary occlusions, but invasive coronary angiography is
Localizers often needed for confirmation.66,67
Electrocardiogram-gated cine balanced steady-state free precession A suggested CMR protocol for imaging adults after the arterial switch
• Superior and inferior vena cava baffles operation is presented in Box 41.6.
• Pulmonary venous baffle
• Four-chamber view L-LOOP TRANSPOSITION OF THE GREAT ARTERIES
• Ventricular short-axis stack from the base to the apex
• Right ventricular long-axis view to the aorta, two orthogonal views Anatomy and Natural History
• Left ventricular long-axis view to the pulmonary artery, two orthogonal L-loop transposition of the great arteries (also known as “physiologi-
views cally corrected” or “congenitally corrected” TGA) is characterized by
• Branch pulmonary artery axial stack to assess for pulmonary artery atrioventricular discordance and ventriculoarterial discordance (Fig.
stenosis 41.11). Most patients have atrial situs solitus with ventricular inversion
Three-dimensional contrast-enhanced magnetic resonance angiogram or elec-
trocardiogram and respiratory navigator gated three-dimensional balanced
steady-state free precessiona
Electrocardiogram-gated phase contrast through-plane flow in the main pul-
monary artery, aorta, tricuspid and mitral valvesb
Late gadolinium enhancement to assess for myocardial fibrosisc
These protocols were adapted from Boston Children’s Hospital
Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-dimen-
sional magnetic resonance angiogram with the first cardiovascular Ao
RPA
magnetic resonance (CMR) examination, and then optional for sub- LPA
sequent examinations.
b
In cases where Qp:Qs ratio is of interest, flow across the branch
pulmonary arteries, superior vena cava, and inferior vena cava/
descending thoracic aorta may be assessed to improve level of
confidence.
c
In general, our practice is to obtain late gadolinium enhancement
(LGE) imaging to assess for myocardial fibrosis with the first CMR FIG. 41.10  Axial contrast-enhanced magnetic resonance angiogram
examination, and the frequency of repeating this technique in demonstrating the relationship of the pulmonary arteries and ascending
subsequent examinations is dependent on the initial findings and aorta after an arterial switch operation. Branch pulmonary artery stenosis
specific condition. For example, some CMR laboratories repeat LGE often occurs where the pulmonary arteries are draped over the ascend-
imaging every 3 years in patients with conotruncal anomalies ing aorta. Quantifying differential pulmonary blood flow gives clues to
(tetralogy of Fallot, transposition of the great arteries, double outlet the hemodynamic significance of narrowing. Ao, Aorta; LPA, left pul-
right ventricle). monary artery; RPA, right pulmonary artery.
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 495

BOX 41.6  Cardiovascular Magnetic


Resonance Protocol for Transposition of the
Great Arteries With the Arterial Switch
Operation Ao

Localizers
Electrocardiogram-gated cine balanced steady-state free precession
• Four-chamber view, left ventricular two-chamber, and left ventricular
MPA
long-axis view to the neoaortic valve
• Ventricular short-axis stack from the base to the apex
LA
Electrocardiogram and respiratory navigator gated three-dimensional balanced
steady-state free precession for coronary artery imaging
Three-dimensional contrast-enhanced magnetic resonance angiogram timed
to the pulmonary arteriesa
Electrocardiogram-gated phase contrast through-plane flow in the main pul- RA
monary artery and aortab
Late gadolinium enhancement to assess for myocardial fibrosisc

These protocols were adapted from Boston Children’s Hospital


Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-
dimensional magnetic resonance angiogram with the first RV
cardiovascular magnetic resonance (CMR) examination, and then
optional for subsequent examinations.
b
In cases where Qp:Qs ratio is of interest, flow across the branch
pulmonary arteries, superior vena cava, and inferior vena cava/ LV
descending thoracic aorta may be assessed to improve level of
confidence.
c
In general, our practice is to obtain late gadolinium enhancement
(LGE) imaging to assess for myocardial fibrosis with the first CMR
examination, and the frequency of repeating this technique in
subsequent examinations is dependent on the initial findings and FIG. 41.11  L-loop transposition of the great arteries. In this rare acya-
specific condition. For example, some CMR laboratories repeat LGE notic congenital heart defect there is ventricular inversion and transposed
imaging every 3 years in patients with conotruncal anomalies great arteries. The right ventricle (RV) pumps oxygenated blood to the
(tetralogy of Fallot, transposition of the great arteries, double outlet aorta (Ao), while the left ventricle (LV) pumps deoxygenated blood to
right ventricle). the pulmonary artery. LA, Left atrium; MPA, main pulmonary artery; RA,
right atrium. (From Otto C, ed. The Practice of Clinical Echocardiography.
5th ed. Philadelphia: Elsevier; 2017.)

and an aorta which is anterior and leftward. In this configuration the


right ventricle will lie posterior and leftward. Approximately 10% of
patients have mirror-image dextrocardia. The right ventricle can be
identified by the moderator band and trabeculated septal surface (Fig.
41.12). L-loop TGA is rare, accounting for <1% of congenital heart LV
disease. In isolation, patients with isolated L-loop TGA are acyanotic,
although the majority of patients have associated defects (VSD 70%,
abnormal tricuspid valve >50%, and pulmonary outflow obstruction
in 40%). Patients with isolated L-loop TGA, without associated defects
and normal tricuspid (systemic atrioventricular) valves, have a good
RA * RV

prognosis. However, this is a minority of patients and those with addi-


tional anatomic abnormalities have a worse prognosis. Progressive
tricuspid regurgitation is common and is associated with heart failure LA
and death.68,69

Surgical Repairs
Patients with L-loop TGA and no associated defects often do not require
surgery. For those who do require intervention, options include a physi-
ologic repair in which the associated anatomic defects are corrected
but the right ventricle remains the systemic ventricle. Alternatively, an
anatomic repair (the double-switch) can be performed in young patients, FIG. 41.12  Four-chamber balanced steady-state free precession cine
which requires an atrial switch procedure and either an arterial switch image showing the anatomy of L-loop transposition of the great arteries.
or Rastelli. Evaluation of patients after the double-switch procedure Asterisk denotes the septal leaflet of the tricuspid valve. LA, Left atrium;
requires evaluation of both the atrial switch and the arterial switch LV, left ventricle; RA, right atrium; RV, right ventricle.
496 SECTION IV  Right Ventricular and Congenital Heart Disease

portions. Refer to the text on evaluation of the patient with D-loop DOUBLE OUTLET RIGHT VENTRICLE
TGA for details.50 The atrioventricular node and His bundles are abnor-
mally located and complete heart block occurs at a rate of approximately Anatomy and Natural History
2% per year in adults. Double outlet right ventricle (DORV) is a conotruncal anomaly in
which both great arteries are completely or nearly completely aligned
Cardiovascular Magnetic Resonance Evaluation of with the right ventricle. Rather than a single congenital abnormality,
L-Loop Transposition of the Great Arteries Following DORV encompasses a variety of configurations with very different pre-
Physiologic Repair sentations and types of surgical repair. In DORV, the VSD is the only
Systemic Right Ventricle source of egress from the left ventricle. The VSD size, relationship of the
The right ventricle is not optimally suited to support the systemic circula- VSD to the great arteries, conal morphology, presence of outflow tract
tion and up to 50% of patients with L-loop TGA will have heart failure obstruction, and associated cardiovascular defects primarily determine
by age 50 years.50,70 TTE evaluation of systemic RV function is difficult, the physiology as well as the treatment strategy.
with poor reproducibility, and CMR is the clinical gold standard for
evaluation of systemic RV volumes and systolic function.71 Nonetheless, Surgical Repairs
there is wide variability in how different CMR laboratories process and There are several physiologic variations of DORV that dictate the clini-
contour RV images. The right ventricle can be contoured from either cal presentation and approach to surgical repair:
short-axis stacks perpendicular to the interventricular septum or axial 1. TOF physiology: DORV with subaortic VSD and pulmonic stenosis.
stacks. Axial stacks allow easier tracking of the tricuspid valve plane In the most common form of DORV (subaortic VSD), the surgical
but partial volume averaging can make the diaphragmatic surface dif- goal is to establish left ventricle-to-aortic continuity by patching
ficult to contour and volumes and EF are different from those acquired the VSD to the aorta. If the VSD is aligned beneath the aorta, the
in short axis.59,72 The systemic right ventricle also has prominent tra- pulmonary stenosis is present and the physiology is very similar to
beculations and there is variability as to whether trabeculations are TOF. The left ventricle will eject blood across the VSD to stream
included in the blood pool or in the myocardial mass. When included out the aorta and the degree of right-to-left shunt depends on the
in the blood pool, postprocessing is faster, ventricular volumes are amount of pulmonic stenosis. This configuration is repaired similarly
larger, and EF is smaller. Centers should develop internal consistency to TOF. The subpulmonic obstruction is resected and a VSD patch
to allow comparison of individual patient’s ventricular contours with is angled to baffle oxygenated blood from the left ventricle to the
prior studies.55,73 aorta. However, in DORV the pathway from the left ventricle to
the aorta can be longer and more complex than is typical in TOF.
Tricuspid Valve The aortic valve is not just anatomically displaced from the left
In L-loop TGA the tricuspid valve (systemic atrioventricular valve) is ventricle but also structurally separated from it with loss of normal
usually morphologically abnormal, demonstrating apical displacement, mitral-aortic continuity. Subaortic stenosis is therefore a common
a dysplastic septal leaflet, and is often described as Ebstein-like. Regur- cause of re-intervention.76
gitation volume can be calculated by comparing RV stroke volume and 2. VSD physiology: DORV with large subaortic VSD and no pulmonic
anterograde aortic flow using PCMR or by comparing RV and LV stoke stenosis. With a subaortic VSD and no pulmonic stenosis, the physi-
volumes, assuming no shunt. Tricuspid regurgitation places a volume ology and repair will mirror a VSD. Unlike a simple VSD patch,
load on an already pressure-loaded right ventricle and is associated however, the aorta is remote from the mitral valve so the pathway
with heart failure. Tricuspid valve repair is typically ineffective and from the left ventricle to the aorta may become obstructed, as above.
early valve replacement improves outcomes.70,74,75 3. TGA physiology: DORV with subpulmonary VSD ± aortic obstruc-
A suggested CMR protocol for imaging adults with L-loop TGA is tion. DORV with subpulmonary VSD, bilateral conus, and side-by-
presented in Box 41.7. side semilunar valves is known as the “Taussig-Bing” anomaly. With

BOX 41.7  Cardiovascular Magnetic Resonance Protocol for L-Loop Transposition of the Great
Arteries
Localizers • Right ventricular long-axis view to the aortic valve
Electrocardiogram-gated cine balanced steady-state free precession • Left ventricular long-axis view to the pulmonary valve
• Four-chamber view and left ventricular two-chamber view Three-dimensional contrast-enhanced magnetic resonance angiograma
• Ventricular short-axis stack from the base to the apex Electrocardiogram-gated phase contrast through-plane flow in the main pulmonary
• Right ventricular two-chamber view to assess right ventricle and tricuspid artery, aorta, tricuspid and mitral valvesb
valve Late gadolinium enhancement to assess for myocardial fibrosisc

These protocols were adapted from Boston Children’s Hospital Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-dimensional magnetic resonance angiogram with the first cardiovascular
magnetic resonance (CMR) examination, and then optional for subsequent examinations.
b
In cases where Qp:Qs ratio is of interest, flow across the branch pulmonary arteries, superior vena cava, and inferior vena cava/descending
thoracic aorta may be assessed to improve level of confidence.
c
In general, our practice is to obtain late gadolinium enhancement (LGE) imaging to assess for myocardial fibrosis with the first CMR
examination, and the frequency of repeating this technique in subsequent examinations is dependent on the initial findings and specific
condition. For example, some CMR laboratories repeat LGE imaging every 3 years in patients with conotruncal anomalies (tetralogy of Fallot,
transposition of the great arteries, double outlet right ventricle).
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 497

BOX 41.8  Cardiovascular Magnetic


Resonance Protocol for Double Outlet Right
Ventricle
Ao Localizers
Electrocardiogram-gated cine balanced steady-state free precession
* • Four-chamber view and left ventricular two-chamber view
LV
• Left ventricular long-axis view to the aortic valve with careful orientation
to align the left ventricle, ventricular septal defect, and the anteriorly
displaced aorta
LA • Ventricular short-axis stack from the base to the apex
• Right ventricular outflow tract outflow view parallel to the right ventricular
outflow tract
Three-dimensional contrast-enhanced magnetic resonance angiogram timed
to the pulmonary arteriesa
FIG. 41.13  Balanced steady-state free precession cine image oriented Electrocardiogram-gated phase contrast through-plane flow in the main pul-
to the left ventricular outflow tract in a patient with double outlet right
monary artery, aorta, tricuspid and mitral valvesb
ventricle. Note that the aortic valve is remote from the mitral valve and
Late gadolinium enhancement to assess for myocardial fibrosisc
the long complex pathway from the left ventricle (LV) to the anteriorly
malposed aorta (Ao), which is frequently narrowed. Asterisk denotes These protocols were adapted from Boston Children’s Hospital
ventricular septal defect through which LV flow has been redirected Cardiac Magnetic Resonance Imaging Program.
and a common site of obstruction. LA, Left atrium. a
In general, our practice is to obtain a gadolinium-enhanced three-
dimensional magnetic resonance angiogram with the first
cardiovascular magnetic resonance (CMR) examination, and then
optional for subsequent examinations.
a subpulmonic VSD, LV flow will stream out the pulmonary artery b
In cases where Qp:Qs ratio is of interest, flow across the branch
and the physiology and repair strategies will be similar to D-loop pulmonary arteries, superior vena cava, and inferior vena cava/
TGA and is typically repaired with the arterial switch operation (see descending thoracic aorta may be assessed to improve level of
earlier). confidence.
c
In general, our practice is to obtain late gadolinium enhancement
Cardiovascular Magnetic Resonance Evaluation of (LGE) imaging to assess for myocardial fibrosis with the first CMR
examination, and the frequency of repeating this technique in
Double Outlet Right Ventricle
subsequent examinations is dependent on the initial findings and
CMR imaging planes must be planned to align the LVOT with the aorta,
specific condition. For example, some CMR laboratories repeat LGE
which is more remote than in TOF (Fig. 41.13). Associated cardiac imaging every 3 years in patients with conotruncal anomalies
defects such as anomalous pulmonary or systemic veins are also common (tetralogy of Fallot, transposition of the great arteries, double outlet
in DORV and can be seen on MRA.77 right ventricle).
CMR imaging of patients with DORV and subpulmonary VSD can
be equally as challenging. Subaortic stenosis is common and LVOT
planes must take into account the long pathway from the left ventricle
to the neoaorta. Aortic coarctation is common in this configuration,
so arch imaging with MRA and oblique sagittal “candy-cane” bSSFP Surgical Repairs
images can assess arch patency. The coronary transfer can be more The Fontan palliation generally involves a series of cardiac surgeries
challenging in DORV than D-loop TGA, so careful assessment for myo- with the goal to separate the pulmonary and systemic circulations to
cardial ischemia with LGE imaging and evaluation for segmental wall decrease mixing of oxygenated and deoxygenated blood. This series of
motion abnormalities is warranted.64,78 surgeries generally culminates in connecting the superior and inferior
A suggested CMR protocol for imaging adults with repaired DORV vena cava to the pulmonary arteries without a ventricular pumping
is presented in Box 41.8. chamber ejecting to the pulmonary arteries. In the current era, there
is generally a direct anastomosis made between the superior vena cava
SINGLE VENTRICLE (FONTAN PROCEDURE) and a branch pulmonary artery, creating a superior caval-pulmonary
anastomosis (Glenn procedure). A baffle is created either within the
Anatomy and Natural History right atrium (lateral tunnel Fontan) or outside the heart involving a
Patients with Fontan repairs represent a broad spectrum of rare and tube graft (extracardiac Fontan) to connect the inferior vena cava to
complex cardiac abnormalities in which usually only one well-formed the pulmonary artery (Fig. 41.14). The one functional ventricular
ventricle exists and cardiac repair resulting in two functional ventricles pumping chamber ejects oxygenated blood through the aorta to the
is not feasible. This may be due to a severely hypoplastic ventricle, systemic circulation.
hypoplastic atrioventricular valve, or in some cases, a common atrio-
ventricular valve in the setting of a large VSD. The unoperated survival
of patients with single ventricle physiology depends of the cardiac Cardiovascular Magnetic Resonance Evaluation of the
anatomy, the presence of adequate systemic outflow, and on the pres- Patient After Fontan Repair
ence of adequate, but not excessive, pulmonary blood flow. Patients Fontan Baffle, Branch Pulmonary Arteries, and
with some types of single ventricle cardiac anatomy, such as hypoplastic Pulmonary Veins
left heart syndrome, do not generally survive beyond the first few weeks The baffles or conduits that are used to create the Fontan may become
of life without intervention. obstructed, particularly at the caval-pulmonary anastomosis site. The
498 SECTION IV  Right Ventricular and Congenital Heart Disease

RV

Fontan RA LV

FIG. 41.15  Balanced steady-state free precession cine image of the


four-chamber view of a patient with hypoplastic left heart with a fenes-
trated extracardiac Fontan demonstrating the Fontan baffle with a dephas-
ing jet passing across a patent fenestration (arrow). LV, Hypoplastic left
ventricle; RA, right atrium; RV, right ventricle.

Ventricular Size and Function


FIG. 41.14  The completed extracardiac Fontan circulation. The superior Heart failure remains the leading cause of death for patients with only
vena cava has been directly anastomosed to the right pulmonary artery one well-formed ventricle following Fontan surgery.79 Ventricular size
and the inferior vena cava has been brought to the undersurface of the and systolic function should be quantified and the values are generally
right pulmonary artery using an interposition graft. There is a small reported as combined ventricular volumes and EF. It is important to
fenestration between the inferior limb of the Fontan and the pulmonary remember that in cases where there is a VSD and two ventricles both
venous atrium. (From Kutty S, Rathod RH, Danford DA, et al. Role of ejecting to the aorta, ventricular contours of both the right ventricle
imaging in the evaluation of single ventricle with the Fontan palliation. and left ventricle should be performed in the same end-systolic and
Heart. 2016;102:174–183.) end-diastolic phases.

Ventricular Outflow Tract Obstruction


Patients with a hypoplastic left ventricle frequently have hypoplastic
branch pulmonary arteries may be of small caliber and should be evalu- aortas and usually undergo reconstruction of the ascending aorta with
ated for any discrete stenosis. Small gradients within these connec- pulmonary artery tissue (Damus-Kaye-Stansel or Norwood procedure)
tions can have significant impact in the hemodynamics because there as well as relief of any aortic arch narrowing or coarctation. Cine bSSFP
is no ventricular pump to the pulmonary circulation and pulmonary of the LVOT and oblique sagittal views of the aortic arch compliment
flow depends solely on the systemic venous pressure. Fontan baffles MRA images to assess these features.
can develop leaks, which result in shunting from the Fontan pathway
to the systemic circulation, resulting in the right-to-left shunt and Systemic Venous to Pulmonary Venous Collaterals
cyanosis. Many patients also have a fenestration within their Fontan Patients with Fontan physiology are prone to develop systemic venous
baffle created at the time of Fontan surgery to act as a “pop-off ” to collateral vessels from the cavae to the pulmonary veins to offload the
allow ventricular filling in the setting of elevated Fontan pressures elevated caval pressures. These collaterals act as right-to-left shunts
(Fig. 41.15), at the expense of mild arterial desaturation. Blood flow is and result in arterial desaturation. Occluding the collaterals can be
often sluggish in the Fontan pathway and evaluation for thrombus is performed percutaneously in the catheterization laboratory and results
warranted. in increased saturations but may worsen hemodynamics. The pres-
An axial bSSFP stack from the mid liver to aortic arch is often used ence of coils is not a safety concern, but stainless-steel coils lead to
to assess cardiac and vascular anatomy. Narrowing within the Fontan significant image degradation.80 Collaterals can also occur between the
baffle, branch pulmonary arteries, and Fontan baffle leaks or fenestra- aorta and the pulmonary arteries in the setting of cyanosis and these
tions can be identified on cine images. Pulmonary venous obstruction collaterals result in volume loading of the ventricle and may require
can also occur, particularly if there is atrial dilation resulting in pul- occluding if the ventricular volume load is significant. Collateral flow
monary venous compression. MRA complements cine images in defining can be calculated with PCMR measuring flow in multiple vessels
this anatomy. PCMR across the pulmonary arteries assesses for physi- (see Chapter 5).
ologic consequences of branch pulmonary artery stenosis or preferential Fontan patients, particularly those who underwent a classic Glenn
flow to one lung. Fontan thrombus may be seen on cine images or be procedure, are at risk for pulmonary arterial venous malformations
identified as a filling defect on MRA. If there is concern for thrombus, which are frequently too small to be seen by MRA but may be appreci-
LGE with a long TI time (~ 600 ms) may be useful to delineate thrombus ated by rapid transit time from pulmonary artery flow to the pulmonary
from native cardiac tissue. veins by time-resolved MRA.
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 499

BOX 41.9  Cardiovascular Magnetic Resonance Protocol for Fontan


Localizers Three-dimensional contrast-enhanced magnetic resonance angiograma
Electrocardiogram-gated cine balanced steady-state free precession Electrocardiogram-gated phase contrast images: superior vena cava, inferior vena
• Axial stack mid-liver to aortic arch, including visualization of the branch cava, branch pulmonary arteries, ascending and descending aorta, pulmonary
pulmonary arteries veins, Fontan baffle just below caval pulmonary anastomosis, Fontan baffle
• Ventricular two- and four-chamber views just above hepatic vein insertion to assess fenestration of Fontan baffle
• Ventricular short-axis stack from the base to the apex leak flow
• Ventricular outflow tract view Late gadolinium-enhanced to assess for myocardial fibrosisb
Consider double-inversion recovery black-blood images if artifact on balanced Consider postcontrast late gadolinium-enhanced with long inversion time if concern
steady-state free precession imaging for thrombus within the Fontan pathway

These protocols were adapted from Boston Children’s Hospital Cardiac Magnetic Resonance Imaging Program.
a
In general, our practice is to obtain a gadolinium-enhanced three-dimensional magnetic resonance angiogram with the first cardiovascular
magnetic resonance (CMR) examination, and then optional for subsequent examinations.
b
In general, our practice is to obtain late gadolinium-enhanced (LGE) imaging to assess for myocardial fibrosis with the first CMR examination,
and the frequency of repeating this technique in subsequent examinations is dependent on the initial findings and specific condition. For
example, some CMR laboratories repeat LGE imaging every 3 years in patients with conotruncal anomalies (tetralogy of Fallot, transposition of
the great arteries, double outlet right ventricle).

A suggested CMR protocol for adults after Fontan palliation is pre- protocol for each individual patient is crucial to perform a successful
sented in Box 41.9. CMR examination. The information provided by the CMR may identify
prognostic information regarding future risk for adverse outcomes in
this unique set of patients.
CONCLUSION
In conclusion, an increasing number of adults with congenital heart
disease are undergoing CMR imaging. Knowledge of the congenital
REFERENCES
heart anatomy, prior surgical interventions, and incorporating an imaging A full reference list is available online at ExpertConsult.com
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 499.e1

REFERENCES 20. Warnes CA, Williams RG, Bashore TM, et al. ACC/AHA 2008 guidelines
for the management of adults with congenital heart disease: a report of
1. Gilboa SM, Devine OJ, Kucik JE, et al. Congenital heart defects in the the American College of Cardiology/American Heart Association Task
United States: estimating the magnitude of the affected population in Force on Practice Guidelines (Writing Committee to Develop Guidelines
2010. Circulation. 2016;134:101–109. on the Management of Adults with Congenital Heart Disease). Developed
2. da Silva JP, Baumgratz JF, da Fonseca L, et al. The cone reconstruction of in collaboration with the American Society of Echocardiography, Heart
the tricuspid valve in Ebstein’s anomaly. The operation: early and Rhythm Society, International Society for Adult Congenital Heart
midterm results. J Thorac Cardiovasc Surg. 2007;133:215–223. Disease, Society for Cardiovascular Angiography and Interventions, and
3. Nayak KS, Nielsen JF, Bernstein MA, et al. Cardiovascular magnetic Society of Thoracic Surgeons. J Am Coll Cardiol. 2008;52:e143–e263.
resonance phase contrast imaging. J Cardiovasc Magn Reson. 2015; 21. Krieger EV, Clair M, Opotowsky AR, et al. Correlation of exercise
17:71. response in repaired coarctation of the aorta to left ventricular mass and
4. Yalonetsky S, Tobler D, Greutmann M, et al. Cardiac magnetic resonance geometry. Am J Cardiol. 2013;111:406–411.
imaging and the assessment of Ebstein anomaly in adults. Am J Cardiol. 22. Murphy JG, Gersh BJ, Mair DD, et al. Long-term outcome in patients
2011;107:767–773. undergoing surgical repair of tetralogy of Fallot. N Engl J Med.
5. Tobler D, Yalonetsky S, Crean AM, et al. Right heart characteristics and 1993;329:593–599.
exercise parameters in adults with Ebstein anomaly: new perspectives 23. Nollert G, Fischlein T, Bouterwek S, Bohmer C, Klinner W, Reichart B.
from cardiac magnetic resonance imaging studies. Int J Cardiol. Long-term survival in patients with repair of tetralogy of Fallot: 36-year
2013;165:146–150. follow-up of 490 survivors of the first year after surgical repair. J Am Coll
6. Hosch O, Sohns JM, Nguyen TT, et al. The total right/left-volume index: Cardiol. 1997;30:1374–1383.
a new and simplified cardiac magnetic resonance measure to evaluate the 24. Geva T, Sandweiss BM, Gauvreau K, Lock JE, Powell AJ. Factors
severity of Ebstein anomaly of the tricuspid valve: a comparison with associated with impaired clinical status in long-term survivors of
heart failure markers from various modalities. Circ Cardiovasc Imaging. tetralogy of Fallot repair evaluated by magnetic resonance imaging. J Am
2014;7:601–609. Coll Cardiol. 2004;43:1068–1074.
7. Powell AJ, Tsai-Goodman B, Prakash A, Greil GF, Geva T. Comparison 25. Davlouros PA, Kilner PJ, Hornung TS, et al. Right ventricular function in
between phase-velocity cine magnetic resonance imaging and invasive adults with repaired tetralogy of Fallot assessed with cardiovascular
oximetry for quantification of atrial shunts. Am J Cardiol. 2003;91:1523– magnetic resonance imaging: detrimental role of right ventricular
1525, A1529. outflow aneurysms or akinesia and adverse right-to-left ventricular
8. Krieger EV, Stout K. The adult with repaired coarctation of the aorta. interaction. J Am Coll Cardiol. 2002;40:2044–2052.
Heart. 2010;96:1676–1681. 26. Wald RM, Redington AN, Pereira A, et al. Refining the assessment of
9. Younoszai AK, Reddy VM, Hanley FL, Brook MM. Intermediate term pulmonary regurgitation in adults after tetralogy of Fallot repair: should
follow-up of the end-to-side aortic anastomosis for coarctation of the we be measuring regurgitant fraction or regurgitant volume? Eur Heart J.
aorta. Ann Thorac Surg. 2002;74:1631–1634. 2009;30:356–361.
10. Muzzarelli S, Meadows AK, Ordovas KG, Higgins CB, Meadows JJ. 27. Knauth AL, Gauvreau K, Powell AJ, et al. Ventricular size and function
Usefulness of cardiovascular magnetic resonance imaging to predict the assessed by cardiac MRI predict major adverse clinical outcomes late after
need for intervention in patients with coarctation of the aorta. Am J tetralogy of Fallot repair. Heart. 2008;94:211–216.
Cardiol. 2012;109:861–865. 28. Valente AM, Gauvreau K, Assenza GE, et al. Contemporary predictors of
11. Nielsen JC, Powell AJ, Gauvreau K, Marcus EN, Prakash A, Geva T. death and sustained ventricular tachycardia in patients with repaired
Magnetic resonance imaging predictors of coarctation severity. tetralogy of Fallot enrolled in the indicator cohort. Heart.
Circulation. 2005;111:622–628. 2014;100:247–253.
12. Araoz PA, Reddy GP, Tarnoff H, Roge CL, Higgins CB. MR findings of 29. Harrild DM, Berul CI, Cecchin F, et al. Pulmonary valve replacement in
collateral circulation are more accurate measures of hemodynamic tetralogy of Fallot: impact on survival and ventricular tachycardia.
significance than arm-leg blood pressure gradient after repair of Circulation. 2009;119:445–451.
coarctation of the aorta. J Magn Reson Imaging. 2003;17:177–183. 30. Vliegen HW, van Straten A, de Roos A, et al. Magnetic resonance imaging
13. Chen SS, Dimopoulos K, Alonso-Gonzalez R, et al. Prevalence and to assess the hemodynamic effects of pulmonary valve replacement in
prognostic implication of restenosis or dilatation at the aortic coarctation adults late after repair of tetralogy of Fallot. Circulation.
repair site assessed by cardiovascular MRI in adult patients late after 2002;106:1703–1707.
coarctation repair. Int J Cardiol. 2014;173:209–215. 31. Geva T. Repaired tetralogy of Fallot: the roles of cardiovascular magnetic
14. Muzzarelli S, Ordovas KG, Hope MD, Meadows JJ, Higgins CB, Meadows resonance in evaluating pathophysiology and for pulmonary valve
AK. Diagnostic value of the flow profile in the distal descending aorta by replacement decision support. J Cardiovasc Magn Reson. 2011;13:9.
phase-contrast magnetic resonance for predicting severe coarctation of 32. Geva T, Gauvreau K, Powell AJ, et al. Randomized trial of pulmonary
the aorta. J Magn Reson Imaging. 2011;33:1440–1446. valve replacement with and without right ventricular remodeling surgery.
15. Hope MD, Meadows AK, Hope TA, et al. Clinical evaluation of aortic Circulation. 2010;122:S201–S208.
coarctation with 4D flow MR imaging. J Magn Reson Imaging. 33. Puchalski MD, Askovich B, Sower CT, Williams RV, Minich LL, Tani LY.
2010;31:711–718. Pulmonary regurgitation: determining severity by echocardiography and
16. Rengier F, Delles M, Eichhorn J, et al. Noninvasive 4D pressure difference magnetic resonance imaging. Congenit Heart Dis. 2008;3:168–175.
mapping derived from 4D flow MRI in patients with repaired aortic 34. Kilner PJ, Geva T, Kaemmerer H, Trindade PT, Schwitter J, Webb GD.
coarctation: comparison with young healthy volunteers. Int J Cardiovasc Recommendations for cardiovascular magnetic resonance in adults with
Imaging. 2015;31:823–830. congenital heart disease from the respective working groups of the
17. von Kodolitsch Y, Aydin MA, Koschyk DH, et al. Predictors of aneurysmal European Society of Cardiology. Eur Heart J. 2010;31:794–805.
formation after surgical correction of aortic coarctation. J Am Coll 35. Ghai A, Silversides C, Harris L, Webb GD, Siu SC, Therrien J. Left
Cardiol. 2002;39:617–624. ventricular dysfunction is a risk factor for sudden cardiac death in adults
18. Bromberg BI, Beekman RH, Rocchini AP, et al. Aortic aneurysm after late after repair of tetralogy of Fallot. J Am Coll Cardiol.
patch aortoplasty repair of coarctation: a prospective analysis of 2002;40:1675–1680.
prevalence, screening tests and risks. J Am Coll Cardiol. 1989;14: 36. Mahle WT, Parks WJ, Fyfe DA, Sallee D. Tricuspid regurgitation in
734–741. patients with repaired tetralogy of Fallot and its relation to right
19. Cook SC, Hickey J, Maul TM, et al. Assessment of the cerebral circulation ventricular dilatation. Am J Cardiol. 2003;92:643–645.
in adults with coarctation of the aorta. Congenit Heart Dis. 37. Mongeon FP, Gurvitz MZ, Broberg CS, et al; Alliance for Adult Research
2013;8:289–295. in Congenital Cardiology. Aortic root dilatation in adults with surgically
499.e2 SECTION IV  Right Ventricular and Congenital Heart Disease

repaired tetralogy of Fallot: a multicenter cross-sectional study. complete transposition of great arteries and previous atrial switch
Circulation. 2013;127:172–179. operation. Am J Cardiol. 2013;111:908–913.
38. Niwa K. Aortic Root dilatation in tetralogy of Fallot long-term after 59. van der Bom T, Romeih S, Groenink M, et al. Evaluating the systemic
repair—histology of the aorta in tetralogy of Fallot: evidence of intrinsic right ventricle by cardiovascular magnetic resonance: short axis or axial
aortopathy. Int J Cardiol. 2005;103:117–119. slices? Congenit Heart Dis. 2015;10:69–77.
39. Babu-Narayan SV, Kilner PJ, Li W, et al. Ventricular fibrosis suggested by 60. Rydman R, Gatzoulis MA, Ho SY, et al. Systemic right ventricular fibrosis
cardiovascular magnetic resonance in adults with repaired tetralogy of detected by cardiovascular magnetic resonance is associated with clinical
Fallot and its relationship to adverse markers of clinical outcome. outcome, mainly new-onset atrial arrhythmia, in patients after atrial
Circulation. 2006;113:405–413. redirection surgery for transposition of the great arteries. Circ Cardiovasc
40. Broberg CS, Huang J, Hogberg I, et al. Diffuse LV myocardial fibrosis and Imaging. 2015;8.
its clinical associations in adults with repaired tetralogy of Fallot. JACC 61. Junge C, Westhoff-Bleck M, Schoof S, et al. Comparison of late results of
Cardiovasc Imaging. 2016;9:86–87. arterial switch versus atrial switch (Mustard procedure) operation for
41. Chen CA, Dusenbery SM, Valente AM, Powell AJ, Geva T. Myocardial transposition of the great arteries. Am J Cardiol. 2013;111:1505–1509.
ECV fraction assessed by CMR is associated with type of hemodynamic 62. Co-Vu JG, Ginde S, Bartz PJ, Frommelt PC, Tweddell JS, Earing MG.
load and arrhythmia in repaired tetralogy of Fallot. JACC Cardiovasc Long-term outcomes of the neoaorta after arterial switch operation for
Imaging. 2016;9:1–10. transposition of the great arteries. Ann Thorac Surg. 2013;95:1654–
42. Liebman J, Cullum L, Belloc NB. Natural history of transpositon of the 1659.
great arteries. Anatomy and birth and death characteristics. Circulation. 63. Chaturvedi A, Hamilton-Craig C, Cawley PJ, Mitsumori LM, Otto CM,
1969;40:237–262. Maki JH. Quantitating aortic regurgitation by cardiovascular magnetic
43. Cuypers JA, Eindhoven JA, Slager MA, et al. The natural and unnatural resonance: significant variations due to slice location and breath holding.
history of the Mustard procedure: long-term outcome up to 40 years. Eur Eur Radiol. 2016;26:3180–3189.
Heart J. 2014;35:1666–1674. 64. Khairy P, Clair M, Fernandes SM, et al. Cardiovascular outcomes after the
44. Pasquali SK, Hasselblad V, Li JS, Kong DF, Sanders SP. Coronary artery arterial switch operation for D-transposition of the great arteries.
pattern and outcome of arterial switch operation for transposition of the Circulation. 2013;127:331–339.
great arteries: a meta-analysis. Circulation. 2002;106:2575–2580. 65. Legendre A, Losay J, Touchot-Kone A, et al. Coronary events after arterial
45. Lange R, Horer J, Kostolny M, et al. Presence of a ventricular septal defect switch operation for transposition of the great arteries. Circulation.
and the Mustard operation are risk factors for late mortality after the 2003;108(suppl 1):II186–II190.
atrial switch operation: thirty years of follow-up in 417 patients at a 66. Tobler D, Motwani M, Wald RM, et al. Evaluation of a comprehensive
single center. Circulation. 2006;114:1905–1913. cardiovascular magnetic resonance protocol in young adults late after the
46. Senning A. Surgical correction of transposition of the great vessels. arterial switch operation for D-transposition of the great arteries. J
Surgery. 1959;45:966–980. Cardiovasc Magn Reson. 2014;16:98.
47. Mustard WT. Successful two-stage correction of transposition of the great 67. Manso B, Castellote A, Dos L, Casaldaliga J. Myocardial perfusion
vessels. Surgery. 1964;55:469–472. magnetic resonance imaging for detecting coronary function anomalies
48. Roubertie F, Thambo JB, Bretonneau A, et al. Late outcome of 132 in asymptomatic paediatric patients with a previous arterial switch
Senning procedures after 20 years of follow-up. Ann Thorac Surg. operation for the transposition of great arteries. Cardiol Young.
2011;92:2206–2213, discussion 2213–2204. 2010;20:410–417.
49. Moons P, Gewillig M, Sluysmans T, et al. Long term outcome up to 30 68. Prieto LR, Hordof AJ, Secic M, Rosenbaum MS, Gersony WM.
years after the Mustard or Senning operation: a nationwide multicentre Progressive tricuspid valve disease in patients with congenitally corrected
study in Belgium. Heart. 2004;90:307–313. transposition of the great arteries. Circulation. 1998;98:997–1005.
50. Krieger EV, Deen JF. Transposition of the great arteries. In: Otto CM, ed. 69. Connelly MS, Liu PP, Williams WG, Webb GD, Robertson P, McLaughlin
Practice of Clinical Echocardiography. Philadelphia: Elsevier Saunders; PR. Congenitally corrected transposition of the great arteries in the adult:
2017. functional status and complications. J Am Coll Cardiol.
51. Villafane J, Lantin-Hermoso MR, Bhatt AB, et al. D-transposition of the 1996;27:1238–1243.
great arteries: the current era of the arterial switch operation. J Am Coll 70. Graham TP Jr, Bernard YD, Mellen BG, et al. Long-term outcome in
Cardiol. 2014;64:498–511. congenitally corrected transposition of the great arteries: a multi-
52. van der Bom T, van der Palen RL, Bouma BJ, et al. Persistent neo-aortic institutional study. J Am Coll Cardiol. 2000;36:255–261.
growth during adulthood in patients after an arterial switch operation. 71. Eindhoven JA, Menting ME, van den Bosch AE, et al. Quantitative
Heart. 2014;100:1360–1365. assessment of systolic right ventricular function using myocardial
53. Fricke TA, d’Udekem Y, Richardson M, et al. Outcomes of the arterial deformation in patients with a systemic right ventricle. Eur Heart J
switch operation for transposition of the great arteries: 25 years of Cardiovasc Imaging. 2015;16:380–388.
experience. Ann Thorac Surg. 2012;94:139–145. 72. Jimenez-Juan L, Joshi SB, Wintersperger BJ, et al. Assessment of right
54. Kreutzer C, De Vive J, Oppido G, et al. Twenty-five-year experience with ventricular volumes and function using cardiovascular magnetic
Rastelli repair for transposition of the great arteries. J Thorac Cardiovasc resonance cine imaging after atrial redirection surgery for complete
Surg. 2000;120:211–223. transposition of the great arteries. Int J Cardiovasc Imaging.
55. Broberg CS. Cardiac magnetic imaging of the patient with an atrial 2013;29:335–342.
switch palliation for transposition of the great arteries. Prog Pediatr 73. Winter MM, Bernink FJ, Groenink M, et al. Evaluating the systemic right
Cardiol. 2014;38:49–55. ventricle by CMR: the importance of consistent and reproducible
56. Johansson B, Babu-Narayan SV, Kilner PJ, Cannell TM, Mohiaddin RH. delineation of the cavity. J Cardiovasc Magn Reson. 2008;10:40.
3-Dimensional time-resolved contrast-enhanced magnetic resonance 74. Mongeon FP, Connolly HM, Dearani JA, Li Z, Warnes CA. Congenitally
angiography for evaluation late after the Mustard operation for corrected transposition of the great arteries ventricular function at the
transposition. Cardiol Young. 2010;20:1–7. time of systemic atrioventricular valve replacement predicts long-term
57. van der Bom T, Winter MM, Groenink M, et al. Right ventricular ventricular function. J Am Coll Cardiol. 2011;57:2008–2017.
end-diastolic volume combined with peak systolic blood pressure during 75. Scherptong RW, Vliegen HW, Winter MM, et al. Tricuspid valve surgery
exercise identifies patients at risk for complications in adults with a in adults with a dysfunctional systemic right ventricle: repair or replace?
systemic right ventricle. J Am Coll Cardiol. 2013;62:926–936. Circulation. 2009;119:1467–1472.
58. Khattab K, Schmidheiny P, Wustmann K, Wahl A, Seiler C, Schwerzmann 76. Belli E, Serraf A, Lacour-Gayet F, et al. Surgical treatment of subaortic
M. Echocardiogram versus cardiac magnetic resonance imaging for stenosis after biventricular repair of double-outlet right ventricle. J
assessing systolic function of subaortic right ventricle in adults with Thorac Cardiovasc Surg. 1996;112:1570–1578, discussion 1578–1580.
CHAPTER 41  Simple and Complex Congenital Heart Disease: Adults 499.e3

77. Artrip JH, Sauer H, Campbell DN, et al. Biventricular repair in double 79. Kiaffas MG, Van Praagh R, Hanioti C, Green DW. The modified Fontan
outlet right ventricle: surgical results based on the STS-EACTS procedure: morphometry and surgical implications. Ann Thorac Surg.
International Nomenclature classification. Eur J Cardiothorac Surg. 1999;67:1746–1753.
2006;29:545–550. 80. Garg R, Powell AJ, Sena L, Marshall AC, Geva T. Effects of metallic
78. Schwarz F, Blaschczok HC, Sinzobahamvya N, et al. The Taussig-Bing implants on magnetic resonance imaging evaluation of Fontan palliation.
anomaly: long-term results. Eur J Cardiothorac Surg. 2013;44:821–827. Am J Cardiol. 2005;95:688–691.
SECTION V Vascular/Pericardium

42 
Pulmonary Vein and Left Atrial Imaging
Thomas H. Hauser and Dana C. Peters

The development of radiofrequency ablation for the treatment of atrial lung signal is very low. Often a second time frame is acquired imme-
fibrillation has led to an increased interest in the accurate determination diately after the first-pass image to ensure acquisition during peak
of pulmonary vein anatomy and left atrial fibrosis assessment to help contrast. Timing of the acquisition to the first pass of contrast through
plan the procedure and to monitor for postablation pulmonary vein the pulmonary veins is critical, and is achieved using either a bolus
stenosis. Contrast-enhanced magnetic resonance angiography (CE-MRA) timing scan15 or with fluoroscopic triggering.16 For either method,
readily demonstrates the pulmonary veins and is the method of choice imaging is timed to begin with the appearance of contrast in the left
for these imaging studies. In this chapter, we review the methods used atrium (LA). Time-resolved imaging using view-sharing methods17,18
to image the pulmonary veins and normal and anomalous pulmonary is also valuable, providing multiple 3D volumes during the passage
venous anatomy. We also describe how late gadolinium enhancement of contrast.
(LGE) imaging is used to identify atrial fibrosis and scar. The utility of More intravascular contrast agents (gadobenate dimeglumine,
cardiovascular magnetic resonance (CMR) before and after atrial fibril- gadofosveset trisodium, or ferumoxytol)19–21 are available to extend the
lation ablation is described. duration of shortened blood T1, and improve quality, but they may
reduce the contrast between blood and scar/fibrosis, if LGE CMR is
acquired.
IMAGING METHODS: PULMONARY VEINS
The pulmonary veins can be identified by using standard anatomic Image Display
and functional CMR imaging sequences. Although these methods are Once the 3D MRA dataset is obtained, the images can be transferred
sufficient for identifying the anatomic relationship of the pulmonary to a workstation for further manipulation and analysis (Fig. 42.1).
veins to the heart and the other major vascular structures, CE-MRA The simplest and often most informative is to dynamically view two-
is usually used for a volumetric three-dimensional (3D) understand- dimensional (2D) slices within the 3D dataset in the axial, coronal,
ing of the intricate pulmonary venous anatomy. A 3D spoiled gradi- and sagittal planes. The axial images usually provide a good over-
ent echo sequence is acquired during the first pass of gadolinium view of the pulmonary veins and their relationship to the LA, but
(Gd) contrast.1 Clinical protocols vary but have mainly common the coronal and sagittal images are frequently required to determine
elements.2–10 The technique uses short repetition times (TR; 2–5 ms), specific anatomic findings, such as a left common or anomalous
a high flip angle (25–60 degrees), and fractional echoes, all of which pulmonary vein.
provide T1 weighting and minimal flow artifacts. The spatial resolu- Although 2D slices are very useful for viewing the individual pul-
tion varies from 1–2 × 1–2 mm in-plane with 2 to 4 mm slices, before monary veins, it is difficult to produce a single summary image of
interpolation. A single 3D volume requires a 10 to 20 second breath- the anatomy. Maximal intensity projection (MIP) and 3D reconstruc-
hold to suppress ventilatory motion, but scan time can be shortened tions displayed as shaded surface or volume-rendered images take full
using smaller fields of view, shorter TRs, partial Fourier, lower spatial advantage of the 3D dataset and provide very good summary images.
resolution, parallel imaging, or compressed sensing.11 Electrocardio- By convention, the LA and pulmonary veins are viewed in the posterior-
gram (ECG) triggering is not employed, although it is recognized that anterior orientation. These 3D views are most useful when the displayed
the position and shape of the pulmonary veins changes throughout volume is limited to the LA and pulmonary veins. Because the aorta is
the cardiac cycle.7,12 Images obtained with this method reflect the pul- directly posterior to the left-sided pulmonary vein, it frequently obscures
monary veins at their maximal size.13 Axial or coronal slabs are usually them from view in the MIP images. Three-dimensional reconstructed
acquired, using either sequential or centric k-space filling. For the images are frequently preferred because the aorta can be excluded from
pulmonary vasculature, the arterial-venous transit time is very short the displayed volume. Three-dimensional rendered images can also be
(4–7 seconds)14 and therefore artery-vein separation is highly challeng- rotated, to better appreciate the anatomy. Software is also available for
ing and generally not targeted. Contrast is injected with a gadolinium generating an endovascular reconstruction, simulating the view of the
dose of 0.1 to 0.2 mmol/kg at a rate of 1 to 2 mL/s, followed by a saline pulmonary vein ostia from “inside” the LA. Direct anatomic measure-
flush. A precontrast mask can be acquired, although mask subtraction ments should not be obtained from these postprocessed images but
is not essential for pulmonary venography because the background from the 2D slices instead.

500
CHAPTER 42  Pulmonary Vein and Left Atrial Imaging 501

RI
LS

LA
LA LI LA
RS
Ao RS Ao
A B C

LS
RS RS

LA LS
LA
LI
RI Ao

RI
D E F LI

LS
RS RS

LI

LA

RI
G RI H
FIG. 42.1  Normal anatomy and quantification of pulmonary vein size. These images show the normal comple-
ment of four pulmonary veins, along with left atrium (LA) and descending aorta (Ao). The right inferior (RI),
right superior (RS), and left inferior (LI) pulmonary veins are shown in the axial plane (A and B). The left
superior (LS) and right superior pulmonary veins are shown in the coronal plane from the posterior-anterior
orientation (C). The left-sided (D) and right-sided (E) pulmonary veins are shown in the sagittal plane (anterior
to the left). All of the pulmonary veins are shown in the axial maximal intensity projection (F) and posterior-
anterior volume-rendered (G) images. The ostia of the right-sided pulmonary veins can be identified in the
endovascular reconstruction (H).

pulmonary veins is asymmetrical, with the two right-sided pulmonary


PULMONARY VEIN EMBRYOLOGY
veins developing first whereas the left-sided pulmonary venous drainage
A clear understanding of pulmonary vein embryology is important for enters the LA through a single trunk that eventually bifurcates to form
understanding both normal pulmonary vein anatomy, nonpathologic two veins.24
variations from the normal anatomy, and congenital anomalies. The
pulmonary veins and associated apical LA are derived from the primi- Normal and Variant Pulmonary Venous Anatomy
tive common pulmonary vein. The primitive pulmonary venous system Most commonly, there are four pulmonary veins that enter the LA:
initially has no connection with the heart and drains into the cardinal right superior, right inferior, left superior, and left inferior (see Fig.
veins and the umbilico vitelline system. At approximately the fourth 42.1). Each of the veins is directed laterally, with the inferior veins
week of gestation, the pulmonary venous drainage coalesces into a directed posteriorly and the superior veins directed anteriorly. The left
single vessel.22 At the same time, an outgrowth of the primitive LA superior pulmonary vein frequently has a cranial angulation and may
extends toward the pulmonary venous system to meet this vessel to appear to arise from the superior portion of the LA.
form the primitive common pulmonary vein and the venous connec- Variant, nonpathologic pulmonary vein anatomy is very common,
tions to the cardinal veins and the umbilic vitelline system degenerate. present in approximately 40% of patients.2,25 Although numerous varia-
The common pulmonary vein then expands to form the smooth-walled tions have been described, the most common variations in the usual
body of the LA, whereas the primitive LA forms the trabeculated left anatomy are a single left common pulmonary vein or an additional
atrial appendage.23 The branches of the primitive common pulmonary right middle pulmonary vein (Fig. 42.2).3 These variations occur because
vein form the adult pulmonary veins. The development of the LA and of more or less incorporation of the primitive common pulmonary
502 SECTION V Vascular/Pericardium

LC

LA
LA

RM Ao
RI
A B

LC
PA
RS

Ao RM

LA RI

C D

RS LC
RS

RM

RM LA
LA
LC

Ao
RI RI
E F

RS

RM

LA

RI
G
FIG. 42.2  Variant pulmonary venous anatomy. These images were obtained from a patient with right middle
and left common pulmonary veins. The right middle (RM) pulmonary vein is shown in the axial plane (A).
The left common (LC) pulmonary vein is shown in the coronal plane from the posterior-anterior orientation
(B). The single left common (C) and all three right pulmonary veins (D) are shown in the sagittal plane (anterior
to the left) along with the pulmonary artery (PA) immediately adjacent to the right superior pulmonary vein.
All of the pulmonary veins are shown in the axial maximal intensity projection (E) and posterior-anterior volume-
rendered (F) images. The aorta has been removed from the volume-rendered image. The right middle pul-
monary vein is obscured by the right inferior pulmonary vein and is best seen with cranial angulation (G). It
is frequently necessary to manipulate the point of view to see all of the pulmonary veins. Ao, Aorta; LA, left
atrium; RI, right inferior pulmonary vein; RS, right superior pulmonary vein.
CHAPTER 42  Pulmonary Vein and Left Atrial Imaging 503

vein into the LA. Less incorporation leads to apparent fusion of pul- Anomalous pulmonary venous connections are the most common
monary veins before entering the LA, whereas more incorporation results congenital anomaly.27 In total anomalous pulmonary venous connec-
in additional pulmonary veins (Fig. 42.3).26 Because the right-sided tion, there is no connection of the pulmonary veins to the LA such that
pulmonary veins form first and have more developmental time to be all of the pulmonary venous drainage enters the right atrium directly
incorporated into the LA, it is more common to have additional veins or via a systemic vein. This anomaly is necessarily associated with an
on the right. Conversely, the left-sided pulmonary veins form later and atrial right-to-left shunt. Pulmonary venous hypertension is common
are more likely to have a common trunk. These variations in pulmonary because of twists in the artery or compression from adjacent vascular
venous anatomy have not yet been identified as a cause of pathology. structures,29 whereas small atrial septal defects (ASDs) restrict systemic
blood flow.30 These conditions results in cyanosis and heart failure.
Although the mortality rate for symptomatic infants is 80% at 1 year,27
CONGENITAL PULMONARY VENOUS ANOMALIES surgical repair is usually feasible and reduces the mortality rate to less
Congenital pulmonary venous anomalies account for up to 3% of than 25%.31 In partial anomalous pulmonary venous return, one or
all congenital heart disease and approximately 2% of all deaths from more pulmonary veins but not all enter the right atrium or a systemic
congenital heart disease in the first year of life.27 The congenital anomalies vein. There is usually an associated ASD, frequently of the sinus venosus
that affect pulmonary veins are atresia, stenosis, and anomalous con- type with the right superior or right middle pulmonary veins draining
nections, which can be total or partial. These conditions occur when into the superior vena cava.27 The physiology of this anomaly is similar
the normal connections of the primitive pulmonary venous system to that of an ASD and depends on the magnitude of the left-to-right
form abnormally or if embryologic connections to the cardinal vein shunt and the presence of increased pulmonary vascular resistance.28
or umbilic vitelline systems persist, and are frequently associated with Patients are often asymptomatic if the shunt is relatively small and the
other major congenital cardiac anomalies.28 pulmonary vascular resistance is normal. As a result, the diagnosis may
not be made until adulthood. The scimitar syndrome, named after the
characteristic chest radiograph finding, is a specific form of partial
anomalous pulmonary venous connection in which all of the venous
drainage from the right lung enters the inferior vena cava (Fig. 42.4).
This rare syndrome is also associated with anomalous arterial supply
of the right lower lobe from the aorta, dextroposition of the heart, and
hypoplasia of the right lung.32
Congenital pulmonary vein atresia is defined as the absence of any
connection of the pulmonary veins to either the LA or any other vas-
cular structure. This is a very rare condition that is not compatible with

FIG. 42.3  Incorporation of the primitive common pulmonary vein into


the left atrium. The incorporation of the primitive common pulmonary
vein is variable and results in nonpathologic variations in the normal
anatomy. This figure shows the results of variable incorporation of the FIG. 42.4  Coronal thoracic cardiovascular magnetic resonance image
left-sided pulmonary veins. The most common pattern is two left-sided in a patient with scimitar syndrome. This coronal maximal intensity
pulmonary veins (plane B). With less incorporation of the common pul- projection image in the anterior-posterior orientation shows the typical
monary vein into the left atrium, there is only a single left common findings of the scimitar syndrome. The single right pulmonary vein enters
pulmonary vein (plane C). With more incorporation, there are additional the inferior vena cava. The right atrium and descending aorta are also
pulmonary veins (plane A). (From Ghaye B, Szapiro D, Dacher JN, et al. shown. (From Greil GF, Powell AJ, Gildein HP, et al. Gadolinium-enhanced
Percutaneous ablation for atrial fibrillation: the role of cross-sectional three-dimensional magnetic resonance angiography of pulmonary and
imaging. Radiographics. 2003;23 Spec No:S19–33; discussion S48–S50.) systemic venous anomalies. J Am Coll Cardiol. 2002;39:335–341.)
504 SECTION V Vascular/Pericardium

life, although infants may survive for a short period of time because paroxysmal atrial fibrillation, with a reduction in morbidity and improved
of small connections between the pulmonary veins and esophageal or quality of life.53
brachial veins.28
Congenital pulmonary vein stenosis can involve a focal segment of IMAGING BEFORE AND AFTER ATRIAL
one or more pulmonary veins or more diffusely involve an entire pul-
monary vein and is usually associated with other congenital cardiac
FIBRILLATION ABLATION
malformations. Severe stenosis frequently results in cyanosis, heart Imaging is usually performed before atrial fibrillation ablation to deter-
failure, and death, although surgical repair is possible if only focal mine the pulmonary vein anatomy (size and number/orientation of
stenosis is present.28 the pulmonary veins). In addition, CE-MRA may be performed after
Imaging patients with congenital anomalies using CE-MRA is a the ablation if the patient has signs/symptoms suggestive of pulmonary
valuable method for determining the pulmonary venous anatomy. It vein stenosis (see later). The accurate determination of pulmonary vein
is generally able to identify all pulmonary venous anomalies, providing anatomy is critical for the planning and execution of atrial fibrillation
new information in 75% and identifying previously unsuspected anoma- ablation. To achieve success, the operator must place a series of radio-
lies in 30%.33 frequency lesions that encircle the pulmonary veins and electrically
isolate them from the LA.54 This necessarily requires that the pulmonary
PULMONARY VEINS AND THE PATHOPHYSIOLOGY vein anatomy be determined before the procedure. In the initial devel-
opment of the procedure the pulmonary veins were identified using
OF ATRIAL FIBRILLATION invasive contrast venography.55 Although this can be done successfully,
Atrial fibrillation is the most common sustained cardiac arrhythmia, it greatly increases the procedure time and only provides projection
affecting more than 5 million people in the United States,34,35 and is a images of the pulmonary veins. Most centers now use CE-MRA, non-
major cause of morbidity and mortality. It accounts for more than contrast MRA, or computed tomography (CT) angiography to determine
400,000 hospitalizations each year36 and increasing the risk of death by the pulmonary vein anatomy before the procedure. Either technique
50% for men and 90% for women.37 Atrial fibrillation quintuples the provides high-resolution 3D tomographic images of the pulmonary
risk of stroke38 and is the attributed cause for 15% of all strokes.36 The veins and other mediastinal structures. These images can also be imported
costs associated with the treatment of atrial fibrillation are estimated into the 3D electrophysiologic mapping systems that are an integral
at US$6 billion.39 Although several antiarrhythmic drugs are available part of the procedure to combine anatomic and functional information
for the treatment of atrial fibrillation, maintenance of sinus rhythm is during the procedure to form an integrated image.56 Routine utilization
frequently suboptimal40–42 and all these drugs are associated with sig- of image integration has been associated with shorter procedure times
nificant side effects or adverse events.43 and improved outcome after ablation for atrial fibrillation.57,58
Increasingly, evidence has shown that the pulmonary veins play a It is also important to identify the relationship of the pulmonary
critical role in the pathophysiology of atrial fibrillation. As noted, the veins to other mediastinal structures to avoid complications during the
pulmonary veins and LA are both derived from the primitive common procedure. The formation of an atrial-esophageal fistula is a rare but
pulmonary vein23 and therefore have many anatomic similarities. Both catastrophic complication caused by excessive heating of the posterior
are smooth-walled structures that have electrically active myocardium. LA wall and the adjacent esophagus.59–63 The esophagus and its relation-
Approximately 90% of pulmonary veins contain atrial myocardium.44 ship to the LA can be readily identified on standard anatomic CMR
Although the myocardium in the LA is uniform, myocardium in the sequences and on LGE (Fig. 42.5). The esophagus almost always directly
pulmonary veins is frequently discontinuous and fibrotic. Patients with abuts the posterior LA and is usually closer to the left-sided pulmonary
a history of atrial fibrillation uniformly have myocardium in the pul- veins, but the location is highly variable.64–66 The esophagus is frequently
monary veins and an increased rate of structural abnormalities. These within 5 mm of the pulmonary veins at a location that probably increases
structural abnormalities result in abnormal electrical activation, with the risk for the formation of an atrial-esophageal fistula.67,68 Indeed,
slow and anisotropic conduction. Proarrhythmic re-entrant beats and the esophagus often appears enhanced on postablation LGE.69 The risk
sustained focal activity can be easily induced.45 of causing an atrial-esophageal fistula may be reduced by avoiding
A landmark study demonstrated that the proarrhythmic electrical ablation in the region of the LA closest to the esophagus, but this may
activity in pulmonary veins is directly responsible for the generation be difficult because the esophagus is mobile and may move during the
of atrial fibrillation in many patients.46 Among those with paroxysmal course of the procedure.70
atrial fibrillation, 94% were found to have ectopic foci in the pulmonary The pulmonary veins are sometimes imaged after the procedure
veins that were responsible for the induction of atrial fibrillation. Radio- if there is a suspicion for pulmonary vein stenosis. Pulmonary vein
frequency ablation of these foci resulted in complete suppression of stenosis is an uncommon but severe complication of atrial fibrillation
atrial fibrillation in a majority of patients. ablation (Fig. 42.6).2,25,55,71–77 The application of radiofrequency energy
Although these studies are suggestive that re-entry may be impor- to the pulmonary veins causes intimal proliferation and myocardial
tant in the initiation of atrial fibrillation, the mechanisms that sustain necrosis that can result in stenosis or occlusion.78 Severe stenosis occurs
atrial fibrillation are still not known. Proposed mechanisms include in up to 3% of patients after the procedure and results in pulmonary
triggers and an abnormal LA substrate. Triggers potentially include hypertension and decreased perfusion of the affected lung segments.79–81
regions with ectopic electrical activity or atrial rotors. The presence Patients frequently present with cough or dyspnea, but a significant
of a dilated LA, by itself and with associated electrical, metabolic, and proportion are asymptomatic.73 Stenosis is most likely to occur in smaller
structural remodeling,47,48 is the abnormal substrate that may sustain pulmonary veins in which the ablation lesions were placed further into
atrial fibrillation. the pulmonary vein trunk and with greater extent of ablation.76,77 If
Based on these findings, several related procedures were developed stenosis does occur, pulmonary vein angioplasty is usually success-
for the treatment of atrial fibrillation.46,49–52 Each of these procedures ful in restoring normal flow and alleviating symptoms.82 Techniques
uses radiofrequency ablation to electrically isolate the pulmonary veins that have emphasized placing ablation lesions within the body of the
from the LA, with or without additional ablation in the body of the LA under intracardiac echocardiographic guidance have dramatically
LA. Short-term success rates range from 65% to 85% in patients with reduced the rate of pulmonary vein stenosis72 such that screening for
CHAPTER 42  Pulmonary Vein and Left Atrial Imaging 505

LA

Rl

Eso
A B Eso

FIG. 42.5  Anatomic relationship of the esophagus to the pulmonary veins and the left atrium (LA). (A) This
axial T1-weighted fast spin echo image shows the esophagus immediately posterior to the LA and adjacent
to the right inferior (RI) pulmonary vein. The esophagus is compressed between the enlarged left atrium and
the spinal column. (B) In a different subject, postablation late gadolinium enhancement image also shows
the esophagus (Eso), adjacent to a region of ablation.

vein diameter measurements performed using CT, intracardiac


RS echocardiography, transesophageal echocardiography, and x-ray
venography in the same patients.84 Each of these methods identified
different numbers and positions of pulmonary veins, with a poor cor-
relation between diameter measurements obtained with each imaging
LA
modality.
LI Tomographic imaging of the pulmonary veins using CMR has
several advantages. All of the anatomic information is obtained in
a single 3D dataset that can be manipulated in numerous ways. This
allows for anatomic measurements in any desired plane, including
determination of the perimeter and cross-sectional area that may be
more meaningful measures of pulmonary vein size. A simple method
for determining pulmonary vein size in the sagittal plane is highly
FIG. 42.6  Contrast-enhanced magnetic resonance angiography in a
patient with pulmonary vein stenosis 6 months following pulmonary reproducible and provides these additional measures.3 The maximal
vein isolation. There is severe stenosis of the left inferior (LI) pulmonary diameter, perimeter, and cross-sectional area are measured at the loca-
vein and moderate stenosis of the right superior (RS) pulmonary vein, tion in the sagittal plane at which the pulmonary veins separate from
indicated by the dashed arrows. There is prestenotic dilation of the LI the LA and from each other (see Fig. 42.1). This is easily determined
pulmonary vein. LA, Left atrium. by scrolling through a reconstruction of the 3D dataset in the sagittal
plane. Because the measurements are made in a standard plane and
location, reproducibility is greatly improved compared with standard
pulmonary vein stenosis after the procedure is no longer routinely diameter measurements.3 This allows for more accurate determination
performed. of interstudy differences in pulmonary vein size and increased statisti-
cal power in research studies. Even in the absence of severe stenosis,
Quantification of Pulmonary Vein Size Before and this method can identify small changes in pulmonary vein size after
After Ablation atrial fibrillation ablation that may be due to hemodynamic changes
The accurate measurement of pulmonary vein size is essential for related to the restoration of sinus rhythm.4 The determination of the
serial assessment of pulmonary vein stenosis and to further investi- perimeter and cross-sectional area is also advantageous. Patients with
gate the role of pulmonary veins in the initiation and maintenance larger summed total pulmonary vein cross-sectional area are more
of atrial fibrillation. Most investigators have measured pulmonary likely to have recurrent atrial fibrillation after ablation independent of
vein diameters in a specified plane, usually at the ostia.2,55,77 These the type of atrial fibrillation or LA size.85 Diameter measurements do
measurements tend to have poor reproducibility for several reasons not have predictive value.53
(Fig. 42.7). Identification of the true ostia is very difficult because the
pulmonary veins and LA are embryologically related with no clear Left Atrial and Left Atrial Appendage Morphology
anatomic border between them. The pulmonary vein ostia are not Increased LA “sphericity” (how closely the LA shape resembles a sphere)
round, such that measurements taken at the same location vary sig- has been demonstrated to be a marker of poor prognosis,86 related to
nificantly with the plane of measurement.2,3 A further complication is atrial fibrillation recurrence after ablation, and more predictive than
that most measurements are derived from nongated images, although LA volume. Additionally, the LA appendage morphology may help risk
the pulmonary vein size varies significantly over the cardiac cycle.7,83 stratify patients for thromboembolic event,87 with “chicken wing” mor-
These difficulties were highlighted in a study comparing pulmonary phologies predicting fewer events.
506 SECTION V Vascular/Pericardium

LI
RS

LA
LA

A B
FIG. 42.7  Difficulty in measurement of pulmonary vein diameters. These images show a right superior (RS)
pulmonary vein in the axial plane (A) and a left inferior (LI) pulmonary vein in the sagittal plane (B). There is
no clear anatomic border between the RS pulmonary vein and the left atrium (LA) (A) such that several
potential diameter measurements are possible (solid lines). The left inferior pulmonary vein is oval (B). The
diameters measured in the axial plane (horizontal line) and coronal plane (vertical line) differ from each other
and from the true maximal diameter (dashed line).

RS RS RS
RM RM RM

LA LA LA
LI LI LI

Ao Ao Ao
A RI B RI C RI
FIG. 42.8  Late gadolinium enhancement (LGE) imaging. These images show the pulmonary vein anatomy
(A) in a patient who underwent LGE imaging before (B) and 6 weeks after (C) atrial fibrillation ablation. There
is variant anatomy with an additional right middle (RM) pulmonary vein (A). Before ablation (B), there is no
enhancement of the pulmonary veins. There is increased signal in the right superior (RS) and RM pulmonary
veins due to artifact from the ventilatory compensation technique. After ablation (C) there is evidence of
LGE/pulmonary vein scar (dashed arrows). The inset shows a reformatted image of the left inferior (LI)
pulmonary vein that shows circumferential scar. Ao, Aorta; LA, left atrium; RI, right inferior pulmonary vein.

It also demonstrates the effects of timing of the 3D volume after contrast


Late Gadolinium Enhancement of the Left Atrium and injection.
Pulmonary Veins As with left ventricular thrombus imaging,91a long inversion time
Scar and fibrosis imaging using LGE has the potential to noninvasively LGE has been shown to be superior for the identification of left atrial
assess both the extent of LA remodeling (fibrosis) associated with atrial appendage thrombus.91b This offers the opportunity to combine pul-
fibrillation and the completeness of ablation lines after pulmonary vein monary vein CE-MRA with long inversion time LGE to both assess
isolation. LGE may have applications in both risk stratifying patients pulmonary vein anatomy and exclude LA appendage thrombus.91c
for ablation and in periprocedural or real-time guidance of an ablation,
to ensure minimal gaps. The LGE technique used for atrial imaging88 Scar and Fibrosis Measurement
is a T1-weighted inversion recovery gradient echo (GRE) sequence, Similar to LGE in the left ventricle, scar can be segmented by carefully
with imaging acquired >15 to 20 minutes after the injection of gado- drawn regions of interest and thresholding (Fig. 42.10). First, a blood-
linium contrast.89,89a Gadolinium contrast remains concentrated in the pool region of interest and an LA wall region of interest are constructed,
regions of scar/fibrosis, compared with muscle or blood, because of from which scar is segmented using a threshold. The threshold for atrial
reduced clearance and the large contrast distribution volume in fibrotic fibrosis and scar depends on the image contrast, but generally signal
regions.90 To detect LA wall scar, the standard LGE method is modified >3 SDs (measured in the blood) above blood pool signal is identified
to include fat suppression and to achieve higher spatial resolution (1.3 as scar.92,93 Fibrosis extent is measured by segmenting the enhanced
× 1.3 × 3–5 mm3) by acquiring a 3D volume during free breathing with atrial myocardium (volume), and normalizing it by the total atrial
ventilatory motion compensated imaging (Fig. 42.8).91 Fig. 42.9 describes myocardial volume, which is approximately equal to the atrial wall
potential pitfalls in atrial LGE imaging, including the importance of surface area × 2 mm (mean atrial wall thickness).94 Such segmentations
fat suppression, TI choice, phase-encoding direction choice, and the can be visualized in 3D, and the percentage of the wall which is enhanced
appearance of artifacts because of poor fat suppression and arrhythmia. can be quantified. However, this type of analysis is time-intensive and
CHAPTER 42  Pulmonary Vein and Left Atrial Imaging 507

A B C

D E F
FIG. 42.9  Pitfalls in late gadolinium enhancement (LGE) imaging of atrial fibrosis and scar. (A–E) Preablation
LGE. (A) Poor fat suppression (arrows), with poor TI choice which nulls blood. The fat which surrounds the
left atrium is visible. (B) Phase-encoding direction should right–left. Arrows show respiratory motion artifact.
(C) The same subject and slice as in B, except phase-encoding direction is right–left; the TI was too short.
(D) Patient with arrhythmia, showing ghosted, poor-quality images. (E) Good-quality image, with inflow artifact
in the right pulmonary vein (arrow). This can be reduced using a navigator pulse at the end of the data
acquisition. (F). Excellent quality postablation image, with evident scar (arrows).

A B C D
FIG. 42.10  (A–C) Axial late gadolinium enhancement images in a patient following pulmonary vein isolation
demonstrating left atrial scar/hyperenhancement. (A) Segmentation of the left atrial cavity in one slice.
(B) The left atrial wall can be considered to extend about 2 mm outward from the left atrial cavity. (C) The
enhanced signal, based on thresholding, within the left atrial wall is identified as fibrosis or scar. (D) After
segmenting the left atrial wall and scar/fibrosis in each slice, the percentage of the atrial wall that is enhanced
can be measured and displayed. Images were obtained from a preablation patient with atrial fibrillation.
508 SECTION V Vascular/Pericardium

has moderate reproducibility.93 Automated and semiquantitative methods III, 20% to 30%; and IV, >30%. Some 260 patients were enrolled and
are in development. monitored over a mean of 213 days of follow-up. Few (15%) of stage
I patients and many (51%) of stage IV patients had atrial fibrillation
Postablation Assessments of Atrial Scar recurrence, although Utah IV patients represent a small minority of
Radiofrequency ablation for the treatment of atrial fibrillation results the total population. In this study no other variable except for mitral
in scarring of the pulmonary vein and LA.78 Several studies have found valve disease (hazard ratio 3.45) predicted recurrence. LGE evidence
increased ablation scar or an increased number of fully ablated veins of complete pulmonary vein encirclement was rarely achieved, though
in subjects without recurrence.95–97 Other groups have worked toward the lack of complete encirclement at 90 days did not predict atrial
the goal of identifying gaps in the ablation lines around individual arrhythmia recurrence.108
pulmonary veins using LGE, with mixed success. In studies of patients Most of these studies are from a single group, and it is critical to
undergoing repeat ablations, some studies found no correlation between note that atrial fibrosis imaging by LGE has not yet been correlated
sites of electrical reconnection and minimal LGE,98,99 whereas others with pathology because of a lack of animal models of atrial fibrosis.
found a relationship.100 Probably, improved LGE quality and interpreta- However, effort has been made to correlate LA voltage mapping with
tion is required before LGE is able to guide reablation. CMR to guide fibrosis, with the goal of establishing one-to-one correspondence between
ablations in real time has employed both T2-weighted images, low voltage and LA enhancement, as a surrogate validation. These studies
T1-weighted images, and LGE.96,101–104 find decreased bipolar voltage in regions of LGE enhancement,92,98,110
with optimal cutoffs for low voltage corresponding to contrast-to-noise
Atrial Fibrosis by Late Gadolinium Enhancement to ratio >3 or enhancement ratio (LA wall to blood) >1.6. These studies
Predict Atrial Fibrillation Recurrence Postablation range from strong to moderate to weak voltage to LGE correlations,
A series of studies48,105–108 have developed evidence for the hypothesis that and were mainly in mixed pre-PVI and post-PVI populations. Finally,
patients with more significant atrial fibrosis before ablation are more patients with other cardiovascular diseases (i.e., heart failure without
prone to atrial fibrillation recurrence after ablation. The largest and atrial fibrillation) also exhibit atrial fibrosis.111,112
most recent multicenter study109 used frequent monitoring of recurrence
post-pulmonary vein isolation (PVI) procedure. Fibrosis extent indicates
the percent of atrial wall tissue that is enhanced/fibrotic. Fibrosis extent
REFERENCES
(Utah stages) corresponded to: I, 0% to 10% fibrosis; II, 10% to 20%; A full reference list is available online at ExpertConsult.com
CHAPTER 42  Pulmonary Vein and Left Atrial Imaging 508.e1

REFERENCES 19. Camren GP, Wilson GJ, Bamra VR, Nguyen KQ, Hippe DS, Maki JH. A
comparison between gadofosveset trisodium and gadobenate
1. Prince MR, Narasimham DL, Stanley JC, et al. Breath-hold gadolinium- dimeglumine for steady state MRA of the thoracic vasculature. Biomed
enhanced MR angiography of the abdominal aorta and its major Res Int. 2014;2014:625614.
branches. Radiology. 1995;197:785–792. 20. Han F, Rapacchi S, Khan S, et al. Four-dimensional, multiphase,
2. Wittkampf FH, Vonken EJ, Derksen R, et al. Pulmonary vein ostium steady-state imaging with contrast enhancement (MUSIC) in the heart:
geometry: analysis by magnetic resonance angiography. Circulation. a feasibility study in children. Magn Reson Med. 2015;74:1042–1049.
2003;107:21–23. 21. Rustogi R, Galizia M, Thakrar D, et al. Steady-state MRA techniques
3. Hauser TH, Yeon SB, McClennen S, et al. A method for the with a blood pool contrast agent improve visualization of pulmonary
determination of proximal pulmonary vein size using contrast-enhanced venous anatomy and left atrial patency compared with time-resolved
magnetic resonance angiography. J Cardiovasc Magn Reson. MRA pre- and postcatheter ablation in atrial fibrillation. J Magn Reson
2004;6:927–936. Imaging. 2015;42:1305–1313.
4. Hauser TH, Yeon SB, McClennen S, et al. Subclinical pulmonary vein 22. Blom NA, Gittenberger-de Groot AC, Jongeneel TH, DeRuiter MC,
narrowing after ablation for atrial fibrillation. Heart. 2005;91:672–673. Poelmann RE, Ottenkamp J. Normal development of the pulmonary
5. Mansour M, Holmvang G, Sosnovik D, et al. Assessment of pulmonary veins in human embryos and formulation of a morphogenetic concept
vein anatomic variability by magnetic resonance imaging: implications for sinus venosus defects. Am J Cardiol. 2001;87:305–309.
for catheter ablation techniques for atrial fibrillation. J Cardiovasc 23. Moore KL. The Developing Human. Philadelphia: WB Saunders; 1988.
Electrophysiol. 2004;15:387–393. 24. Webb S, Kanani M, Anderson RH, Richardson MK, Brown NA.
6. Mlcochova H, Tintera J, Porod V, Peichl P, Cihak R, Kautzner J. Development of the human pulmonary vein and its incorporation in
Magnetic resonance angiography of pulmonary veins: implications for the morphologically left atrium. Cardiol Young. 2001;11:632–642.
catheter ablation of atrial fibrillation. Pacing Clin Electrophysiol. 25. Kato R, Lickfett L, Meininger G, et al. Pulmonary vein anatomy in
2005;28:1073–1080. patients undergoing catheter ablation of atrial fibrillation: lessons
7. Syed MA, Peters DC, Rashid H, Arai AE. Pulmonary vein imaging: learned by use of magnetic resonance imaging. Circulation.
comparison of 3D magnetic resonance angiography with 2D cine MRI 2003;107:2004–2010.
for characterizing anatomy and size. J Cardiovasc Magn Reson. 26. Ghaye B, Szapiro D, Dacher JN, et al. Percutaneous ablation for atrial
2005;7:355–360. fibrillation: the role of cross-sectional imaging. Radiographics. 2003;23
8. Tamborero D, Mont L, Nava S, et al. Incidence of pulmonary vein Spec No:S19–S33, discussion S48–S50.
stenosis in patients submitted to atrial fibrillation ablation: a 27. Krabill KA, Lucas RV. Abnormal pulmonary venous connections. In:
comparison of the selective segmental ostial ablation vs the Emmanouilides GC, Riemenschneider TA, Allen HD, et al, eds. Moss and
circumferential pulmonary veins ablation. J Interv Card Electrophysiol. Adams’ Heart Disease in Infants, Children and Adolescents. 5th ed.
2005;14:21–25. Baltimore: Williams and Wilkins; 1995:838.
9. Tsao HM, Wu MH, Huang BH, et al. Morphologic remodeling of 28. Friedman WF, Silverman N. Congenital heart disease in infancy and
pulmonary veins and left atrium after catheter ablation of atrial childhood. In: Braunwald E, Zipes DP, Libby P, eds. Heart Disease. 6th
fibrillation: insight from long-term follow-up of three-dimensional ed. Philadelphia: WB Saunders; 2001:1552.
magnetic resonance imaging. J Cardiovasc Electrophysiol. 2005;16:7–12. 29. Wang JK, Lue HC, Wu MH, Young ML, Wu FF, Wu JM. Obstructed total
10. Vonken EP, Velthuis BK, Wittkampf FH, Rensing BJ, Derksen R, Cramer anomalous pulmonary venous connection. Pediatr Cardiol.
MJ. Contrast-enhanced MRA and 3D visualization of pulmonary venous 1993;14:28–32.
anatomy to assist radiofrequency catheter ablation. J Cardiovasc Magn 30. Ward KE, Mullins CE, Huhta JC, Nihill MR, McNamara DG, Cooley DA.
Reson. 2003;5:545–551. Restrictive interatrial communication in total anomalous pulmonary
11. Roujol S, Foppa M, Basha TA, et al. Accelerated free breathing ECG venous connection. Am J Cardiol. 1986;57:1131–1136.
triggered contrast enhanced pulmonary vein magnetic resonance 31. Lamb RK, Qureshi SA, Wilkinson JL, Arnold R, West CR, Hamilton DI.
angiography using compressed sensing. J Cardiovasc Magn Reson. Total anomalous pulmonary venous drainage. Seventeen-year surgical
2014;16:91. experience. J Thorac Cardiovasc Surg. 1988;96:368–375.
12. Lickfett L, Dickfeld T, Kato R, et al. Changes of pulmonary vein orifice 32. Gao YA, Burrows PE, Benson LN, Rabinovitch M, Freedom RM.
size and location throughout the cardiac cycle: dynamic analysis using Scimitar syndrome in infancy. J Am Coll Cardiol. 1993;22:873–882.
magnetic resonance cine imaging. J Cardiovasc Electrophysiol. 33. Greil G, Powell A, Gildein H, Geva T. Gadolinium-enhanced three-
2005;16:582–588. dimensional magnetic resonance angiography of pulmonary and
13. Hauser TH, Yeon SB, McClennen S, et al. Variability in Pulmonary Vein systemic venous anomalies. J Am Coll Cardiol. 2002;39:335–341.
Anatomy During the Cardiac Cycle. Mount Royal, NJ: Society for 34. Colilla S, Crow A, Petkun W, Singer DE, Simon T, Liu X. Estimates of
Cardiovascular Magnetic Resonance; 2005. current and future incidence and prevalence of atrial fibrillation in the
14. Schoenberg SO, Bock M, Floemer F, et al. High-resolution pulmonary U.S. adult population. Am J Cardiol. 2013;112:1142–1147.
arterio- and venography using multiple-bolus multiphase 3D-Gd-mRA. 35. Feinberg WM, Blackshear JL, Laupacis A, Kronmal R, Hart RG.
J Magn Reson Imaging. 1999;10:339–346. Prevalence, age distribution, and gender of patients with atrial
15. Earls JP, Rofsky NM, DeCorato DR, Krinsky GA, Weinreb JC. Breath- fibrillation. Analysis and implications. Arch Intern Med.
hold single-dose gadolinium-enhanced three-dimensional MR 1995;155:469–473.
aortography: usefulness of a timing examination and MR power 36. Mozaffarian D, Benjamin EJ, Go AS, et al. Executive summary: heart
injector. Radiology. 1996;201:705–710. disease and stroke statistics—2016 update: a report from the American
16. Wilman AH, Riederer SJ, King BF, Debbins JP, Rossman PJ, Ehman RL. Heart Association. Circulation. 2016;133:447–454.
Fluoroscopically triggered contrast-enhanced three-dimensional MR 37. Benjamin EJ, Wolf PA, D’Agostino RB, Silbershatz H, Kannel WB, Levy
angiography with elliptical centric view order: application to the renal D. Impact of atrial fibrillation on the risk of death: the Framingham
arteries. Radiology. 1997;205:137–146. Heart Study. Circulation. 1998;98:946–952.
17. Korosec FR, Frayne R, Grist TM, Mistretta CA. Time-resolved 38. Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent
contrast-enhanced 3D MR angiography. Magn Reson Med. risk factor for stroke: the Framingham Study. Stroke. 1991;22:983–988.
1996;36:345–351. 39. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke
18. Schonberger M, Usman A, Galizia M, Popescu A, Collins J, Carr JC. statistics—2015 update: a report from the American Heart Association.
Time-resolved MR venography of the pulmonary veins precatheter- Circulation. 2015;131:e29–e322.
based ablation for atrial fibrillation. J Magn Reson Imaging. 40. Hauser TH, Pinto DS, Josephson ME, Zimetbaum P. Early recurrence of
2013;37:127–137. arrhythmia in patients taking amiodarone or class 1C agents for
508.e2 SECTION V Vascular/Pericardium

treatment of atrial fibrillation or atrial flutter. Am J Cardiol. radiofrequency ablation within the left atrium. Circulation.
2004;93:1173–1176. 2005;112:459–464.
41. Singh BN, Singh SN, Reda DJ, et al. Amiodarone versus sotalol for atrial 62. Redfearn DP, Trim GM, Skanes AC, et al. Esophageal temperature
fibrillation. N Engl J Med. 2005;352:1861–1872. monitoring during radiofrequency ablation of atrial fibrillation.
42. Roy D, Talajic M, Dorian P, et al. Amiodarone to prevent recurrence of J Cardiovasc Electrophysiol. 2005;16:589–593.
atrial fibrillation. Canadian Trial of Atrial Fibrillation Investigators. 63. Cappato R, Calkins H, Chen SA, et al. Updated worldwide survey on the
N Engl J Med. 2000;342:913–920. methods, efficacy, and safety of catheter ablation for human atrial
43. Hauser TH, Pinto DS, Josephson ME, Zimetbaum P. Safety and fibrillation. Circ Arrhythm Electrophysiol. 2010;3:32–38.
feasibility of a clinical pathway for the outpatient initiation of 64. Lemola K, Sneider M, Desjardins B, et al. Computed tomographic
antiarrhythmic medications in patients with atrial fibrillation or atrial analysis of the anatomy of the left atrium and the esophagus:
flutter. Am J Cardiol. 2003;91:1437–1441. implications for left atrial catheter ablation. Circulation.
44. Hassink RJ, Aretz HT, Ruskin J, Keane D. Morphology of atrial 2004;110:3655–3660.
myocardium in human pulmonary veins: a postmortem analysis in 65. Tsao HM, Wu MH, Higa S, et al. Anatomic relationship of the
patients with and without atrial fibrillation. J Am Coll Cardiol. esophagus and left atrium: implication for catheter ablation of atrial
2003;42:1108–1114. fibrillation. Chest. 2005;128:2581–2587.
45. Arora R, Verheule S, Scott L, et al. Arrhythmogenic substrate of the 66. Cury RC, Abbara S, Schmidt S, et al. Relationship of the esophagus and
pulmonary veins assessed by high-resolution optical mapping. aorta to the left atrium and pulmonary veins: implications for catheter
Circulation. 2003;107:1816–1821. ablation of atrial fibrillation. Heart Rhythm. 2005;2:1317–1323.
46. Haissaguerre M, Jais P, Shah DC, et al. Spontaneous initiation of atrial 67. Sanchez-Quintana D, Cabrera JA, Climent V, Farre J, Mendonca MC,
fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Ho SY. Anatomic relations between the esophagus and left atrium and
Med. 1998;339:659–666. relevance for ablation of atrial fibrillation. Circulation.
47. De Jong AM, Maass AH, Oberdorf-Maass SU, Van Veldhuisen DJ, Van 2005;112:1400–1405.
Gilst WH, Van Gelder IC. Mechanisms of atrial structural changes 68. Monnig G, Wessling J, Juergens KU, et al. Further evidence of a close
caused by stretch occurring before and during early atrial fibrillation. anatomical relation between the oesophagus and pulmonary veins.
Cardiovasc Res. 2011;89:754–765. Europace. 2005;7:540–545.
48. Oakes RS, Badger TJ, Kholmovski EG, et al. Detection and 69. Meng J, Peters DC, Hsing JM, et al. Late gadolinium enhancement of the
quantification of left atrial structural remodeling with delayed- esophagus is common on cardiac MR several months after pulmonary
enhancement magnetic resonance imaging in patients with atrial vein isolation: preliminary observations. Pacing Clin Electrophysiol.
fibrillation. Circulation. 2009;119:1758–1767. 2010;33:661–666.
49. Pappone C, Rosanio S, Oreto G, et al. Circumferential radiofrequency 70. Good E, Oral H, Lemola K, et al. Movement of the esophagus during
ablation of pulmonary vein ostia. A new anatomic approach for curing left atrial catheter ablation for atrial fibrillation. J Am Coll Cardiol.
atrial fibrillation. Circulation. 2000;102:2619–2628. 2005;46:2107–2110.
50. Arentz T, von Rosenthal J, Blum T, et al. Feasibility and safety of 71. Moak J, Moore H, Lee S, et al. Case report: pulmonary vein stenosis
pulmonary vein isolation using a new mapping and navigation system following RF ablation of paroxysmal atrial fibrillation: successful
in patients with refractory atrial fibrillation. Circulation. treatment with balloon dilation. J Interv Card Electrophysiol.
2003;108:2484–2490. 2000;4:621–631.
51. Haissaguerre M, Jais P, Shah DC, et al. Electrophysiological end point 72. Saad EB, Rossillo A, Saad CP, et al. Pulmonary vein stenosis after
for catheter ablation of atrial fibrillation initiated from multiple radiofrequency ablation of atrial fibrillation: functional characterization,
pulmonary venous foci. Circulation. 2000;101:1409–1417. evolution, and influence of the ablation strategy. Circulation.
52. Oral H, Knight BP, Ozaydin M, et al. Segmental ostial ablation to isolate 2003;108:3102–3107.
the pulmonary veins during atrial fibrillation: feasibility and mechanistic 73. Saad EB, Marrouche NF, Saad CP, et al. Pulmonary vein stenosis after
insights. Circulation. 2002;106:1256–1262. catheter ablation of atrial fibrillation: emergence of a new clinical
53. Pappone C, Rosanio S, Augello G, et al. Mortality, morbidity, and quality syndrome. Ann Intern Med. 2003;138:634–638.
of life after circumferential pulmonary vein ablation for atrial 74. Scanvacca M, Kajita L, Vieira M, Sosa E. Pulmonary vein stenosis
fibrillation: outcomes from a controlled nonrandomized long-term complicating catheter ablation of focal atrial fibrillation. J Cardiovasc
study. J Am Coll Cardiol. 2003;42:185–197. Electrophysiol. 2000;1:677–681.
54. Marine JE, Dong J, Calkins H. Catheter ablation therapy for atrial 75. Yang M, Akbari H, Reddy GP, Higgins CB. Identification of pulmonary
fibrillation. Prog Cardiovasc Dis. 2005;48:178–192. vein stenosis after radiofrequency ablation for atrial fibrillation using
55. Lin WS, Prakash VS, Tai CT, et al. Pulmonary vein morphology in MRI. J Comput Assist Tomagr. 2001;25:34–35.
patients with paroxysmal atrial fibrillation initiated by ectopic beats 76. Arentz T, Jander N, von Rosenthal J, et al. Incidence of pulmonary vein
originating from the pulmonary veins: implications for catheter stenosis 2 years after radiofrequency catheter ablation of refractory atrial
ablation. Circulation. 2000;101:1274–1281. fibrillation. Eur Heart J. 2003;24:963–969.
56. Calkins H. Three dimensional mapping of atrial fibrillation: techniques 77. Dill T, Neumann T, Ekinci O, et al. Pulmonary vein diameter reduction
and necessity. J Interv Card Electrophysiol. 2005;13(suppl 1):53–59. after radiofrequency catheter ablation for paroxysmal atrial fibrillation
57. Bertaglia E, Bella PD, Tondo C, et al. Image integration increases efficacy evaluated by contrast-enhanced three-dimensional magnetic resonance
of paroxysmal atrial fibrillation catheter ablation: results from the imaging. Circulation. 2003;107:845–850.
CartoMerge Italian Registry. Europace. 2009;11:1004–1010. 78. Taylor GW, Kay GN, Zheng X, Bishop S, Ideker RE. Pathological effects
58. Della Bella P, Fassini G, Cireddu M, et al. Image integration-guided of extensive radiofrequency energy applications in the pulmonary veins
catheter ablation of atrial fibrillation: a prospective randomized study. in dogs. Circulation. 2000;101:1736–1742.
J Cardiovasc Electrophysiol. 2009;20:258–265. 79. Holmes DR Jr, Monahan KH, Packer D. Pulmonary vein stenosis
59. Scanavacca MI, D’Avila A, Parga J, Sosa E. Left atrial-esophageal fistula complicating ablation for atrial fibrillation: clinical spectrum and
following radiofrequency catheter ablation of atrial fibrillation. interventional considerations. JACC Cardiovasc Interv. 2009;2:267–276.
J Cardiovasc Electrophysiol. 2004;15:960–962. 80. Kluge A, Dill T, Ekinci O, et al. Decreased pulmonary perfusion in
60. Pappone C, Oral H, Santinelli V, et al. Atrio-esophageal fistula as a pulmonary vein stenosis after radiofrequency ablation: assessment with
complication of percutaneous transcatheter ablation of atrial fibrillation. dynamic magnetic resonance perfusion imaging. Chest. 2004;126:
Circulation. 2004;109:2724–2726. 428–437.
61. Cummings JE, Schweikert RA, Saliba WI, et al. Assessment of 81. Arentz T, Weber R, Jander N, et al. Pulmonary haemodynamics at rest
temperature, proximity, and course of the esophagus during and during exercise in patients with significant pulmonary vein stenosis
CHAPTER 42  Pulmonary Vein and Left Atrial Imaging 508.e3

after radiofrequency catheter ablation for drug resistant atrial 96. McGann C, Kholmovski E, Blauer J, et al. Dark regions of no-reflow on
fibrillation. Eur Heart J. 2005;26:1410–1414. late gadolinium enhancement magnetic resonance imaging result in scar
82. Qureshi AM, Prieto LR, Latson LA, et al. Transcatheter angioplasty for formation after atrial fibrillation ablation. J Am Coll Cardiol.
acquired pulmonary vein stenosis after radiofrequency ablation. 2011;58:177–185.
Circulation. 2003;108:1336–1342. 97. Badger TJ, Daccarett M, Akoum NW, et al. Evaluation of left atrial
83. Bowman AW, Kovacs SJ. Prediction and assessment of the time-varying lesions after initial and repeat atrial fibrillation ablation: lessons learned
effective pulmonary vein area via cardiac MRI and Doppler from delayed-enhancement MRI in repeat ablation procedures. Circ
echocardiography. Am J Physiol Heart Circ Physiol. Arrhythm Electrophysiol. 2010;3:249–259.
2005;288:H280–H286. 98. Harrison JL, Sohns C, Linton NW, et al. Repeat left atrial catheter
84. Wood MA, Wittkamp M, Henry D, et al. A comparison of pulmonary ablation: cardiac magnetic resonance prediction of endocardial voltage
vein ostial anatomy by computerized tomography, echocardiography, and gaps in ablation lesion sets. Circ Arrhythm Electrophysiol.
and venography in patients with atrial fibrillation having radiofrequency 2015;8:270–278.
catheter ablation. Am J Cardiol. 2004;93:49–53. 99. Spragg DD, Khurram I, Zimmerman SL, et al. Initial experience with
85. Hauser TH, Essebag V, Baldessin F, et al. Larger pulmonary vein magnetic resonance imaging of atrial scar and co-registration with
cross-sectional area is associated with recurrent atrial fibrillation after electroanatomic voltage mapping during atrial fibrillation: success and
pulmonary vein isolation. Circulation. 2005;112(S):II–555. limitations. Heart Rhythm. 2012;9:2003–2009.
86. Bisbal F, Guiu E, Calvo N, et al. Left atrial sphericity: a new method to 100. Bisbal F, Guiu E, Cabanas-Grandio P, et al. CMR-guided approach to
assess atrial remodeling. Impact on the outcome of atrial fibrillation localize and ablate gaps in repeat AF ablation procedure. JACC
ablation. J Cardiovasc Electrophysiol. 2013;24:752–759. Cardiovasc Imaging. 2014;7:653–663.
87. Lupercio F, Ruiz JC, Briceno DF, et al. Left atrial appendage morphology 101. Badger TJ, Oakes RS, Daccarett M, et al. Temporal left atrial lesion
assessment for risk stratification of embolic stroke in patients with atrial formation after ablation of atrial fibrillation. Heart Rhythm.
fibrillation: a meta-analysis. Heart Rhythm. 2016;13:1402–1409. 2009;6:161–168.
88. Kim RJ, Wu E, Rafael A, et al. The use of contrast-enhanced magnetic 102. Hsing J, Peters DC, Knowles BR, Manning WJ, Josephson ME.
resonance imaging to identify reversible myocardial dysfunction. N Engl Cardiovascular magnetic resonance imaging of scar development
J Med. 2000;343:1445–1453. following pulmonary vein isolation: a prospective study. PLoS One.
89. Simonetti OP, Kim RJ, Fieno DS, et al. An improved MR imaging 2014;9:e104844.
technique for the visualization of myocardial infarction. Radiology. 103. Knowles BR, Caulfield D, Cooklin M, et al. Three-dimensional
2001;218:215–223. visualization of acute RF ablation lesions using MRI for the
89a.  Chubb H, Aziz S, Karim R, et al. Optimization of late gadolinium simultaneous determination of the patterns of necrosis and edema. IEEE
enhancement cardiovascular magnetic resonance imaging of post- Trans Biomed Eng. 2010;57:1467–1475.
ablation atrial scar: a cross-over study. J Cardiovasc Magn Reson. 2018. 104. Celik H, Ramanan V, Barry J, et al. Intrinsic contrast for characterization
In press. of acute radiofrequency ablation lesions. Circ Arrhythm Electrophysiol.
90. Judd RM, Lugo-Olivieri CH, Arai M, et al. Physiological basis of 2014;7:718–727.
myocardial contrast enhancement in fast magnetic resonance images of 105. McGann C, Akoum N, Patel A, et al. Atrial fibrillation ablation outcome
2-day-old reperfused canine infarcts. Circulation. 1995;92:1902–1910. is predicted by left atrial remodeling on MRI. Circ Arrhythm
91. Peters DC, Wylie JV, Kissinger KV, et al. Detection of Pulmonary Vein Electrophysiol. 2014;7:23–30.
Ablation with High Resolution MRI. Mount Royal, NJ: Society for 106. Daccarett M, Badger TJ, Akoum N, et al. Association of left atrial fibrosis
Cardiovascular Magnetic Resonance; 2006. detected by delayed-enhancement magnetic resonance imaging and the
91a. Weinsaft JW, Kim HW, Shah DJ, et al. Detection of left ventricular risk of stroke in patients with atrial fibrillation. J Am Coll Cardiol.
thrombus by delayed-enhancement cardiovascular magnetic resonance. 2011;57:831–838.
Prevalence and markers in patients with systolic dysfunction. J Am Coll 107. Akoum N, Daccarett M, McGann C, et al. Atrial fibrosis helps select
Cardiol. 2008;52:148–157. the appropriate patient and strategy in catheter ablation of atrial
91b. Kitkungvan D, Nabi F, Ghosn MG, et al. Detection of LA and LAA fibrillation: a DE-MRI guided approach. J Cardiovasc Electrophysiol.
thrombus by CMR in patients referred for pulmonary vein isolation. 2011;22:16–22.
JACC Cardiovasc Imaging. 2016;9:809–821. 108. Akoum N, Wilber D, Hindricks G, et al. MRI assessment of ablation-
91c. Manning WJ, Spahillari A. Combined pulmonary vein and LA/LAA induced scarring in atrial fibrillation: analysis from the DECAAF study. J
thrombus assessment. Can CMR kill two birds with one stone? J Am Cardiovasc Electrophysiol. 2015;26:473–480.
Coll Cardiol. 2016;9:10. 109. Marrouche NF, Wilber D, Hindricks G, et al. Association of atrial tissue
92. Malcolme-Lawes LC, Juli C, Karim R, et al. Automated analysis of atrial fibrosis identified by delayed enhancement MRI and atrial fibrillation
late gadolinium enhancement imaging that correlates with endocardial catheter ablation: the DECAAF study. JAMA. 2014;311:498–506.
voltage and clinical outcomes: a 2-center study. Heart Rhythm. 110. Khurram IM, Beinart R, Zipunnikov V, et al. Magnetic resonance image
2013;10:1184–1191. intensity ratio, a normalized measure to enable interpatient
93. Karim R, Housden RJ, Balasubramaniam M, et al. Evaluation of current comparability of left atrial fibrosis. Heart Rhythm. 2014;11:85–92.
algorithms for segmentation of scar tissue from late gadolinium 111. Grunseich K, Mekonnen G, Simprini LA, Mojibian H, Marieb M. Left
enhancement cardiovascular magnetic resonance of the left atrium: an atrial volume, congestive heart failure, and obesity are associated with
open-access grand challenge. J Cardiovasc Magn Reson. 2013;15:105. extent of left atrial fibrosis by late gadolinium enhancement.
94. Whitaker J, Rajani R, Chubb H, et al. The role of myocardial wall J Cardiovasc Magn Reson. 2015;17(suppl 1):P368.
thickness in atrial arrhythmogenesis. Europace. 2016;18:1758–1772. 112. Cochet H, Mouries A, Nivet H, et al. Age, atrial fibrillation, and
95. Peters DC, Wylie JV, Hauser TH, et al. Recurrence of atrial fibrillation structural heart disease are the main determinants of left atrial fibrosis
correlates with the extent of post-procedural late gadolinium detected by delayed-enhanced magnetic resonance imaging in a general
enhancement: a pilot study. JACC Cardiovasc Imaging. 2009;2:308–316. cardiology population. J Cardiovasc Electrophysiol. 2015;26:484–492.
43 
Thoracic Aortic Disease
Christoph A. Nienaber

The anatomic and functional characteristics of the aorta, which may whereas repetition time (TR) is determined from the RR interval of the
at first glance appear relatively straightforward, are now recognized to ECG. A shorter acquisition time can be achieved with fast spin echo
be complex. Recent insights from both modern imaging technology pulse sequences whereby a long train of echoes is acquired by using
and better understanding of the hydraulic principles associated with a series of 180-degree radiofrequency (RF) pulses; washout effects are
the variety of aortic diseases have helped the medical community to even more substantial than in conventional spin echo techniques. A
realize the multiple facets of in vivo aortic pathology, as well as its superior black-blood effect is achieved by using preparatory pulses12
varied clinical presentation. (such as presaturation, dephasing gradients, and preinversion) with one
Diagnostic modalities such as transesophageal echocardiography or more additional RF pulses outside the plane to suppress the signal
(TEE), cardiovascular magnetic resonance (CMR), and spiral computed intensity of in-flowing blood and nullify the blood signal (Fig. 43.1).
tomography (CT) have all been shown to be useful to interrogate the Therefore “black-blood” fast T1-weighted and T2-weighted spin echo
aorta, both in chronic disease and in acute aortic syndromes. X-ray sequences have improved image quality, and constitute the method of
contrast angiography, the former gold standard in acute and chronic choice for morphologic assessment of the thoracic aorta. Images are
aortic syndromes, has been relegated to a secondary role after the emer- acquired in axial and additional planes, depending on the anatomy
gence of the noninvasive techniques, most importantly CMR, with their and diagnostic problems, to define the extent of the disease in three-
high sensitivity, specificity, and practical advantages.1–5 However, none dimensional (3D) space.
of the diagnostic modalities listed above is ideal for all patients, and
for a given individual, knowledge of both accuracy and limitations in Gradient Echo Cardiovascular Magnetic Resonance and
the presenting clinical scenario are required.4–7 Although the informa- Flow Mapping
tion content of CMR may greatly overlap with established methods Gradient echo techniques provide dynamic and functional informa-
such as echocardiography, CT, or angiography, the technique is more tion, although with fewer details of the vessel wall. The bright signal
comprehensive and offers more options including four-dimensional of the blood pool on gradient echo images results from flow-related
(4D) functional imaging. enhancement obtained by applying RF pulses to saturate a volume of
Besides images with high soft tissue contrast without any radiation, tissue. With a short TR (4 to 8 ms) and low flip angle (20 to 30 degrees),
CMR can demonstrate and quantify functional parameters beyond maximal signal is emitted by blood flowing in the voxel and ECG-gated
anatomic depiction. Combining anatomical and functional information acquisition provides a high degree of temporal resolution throughout
in a single acquisition means that CMR can potentially provide a more the cardiac cycle (up to 20 to 25 frames) to be displayed in cine format.
comprehensive evaluation of thoracic aortic disease, including aortic Flow-related enhancement is produced by inflow of unsaturated blood
valve morphology and function. CMR is an ideal imaging modality for exposed to only one RF pulse. As result, the laminar moving blood
surveillance in a relatively young patient population requiring long-term displays a bright signal in contrast to stationary tissues. The signal can
or even lifelong follow-up care.8–10 Although the cost-effectiveness of be reduced if the flow is low, as in aortic aneurysms. Mural thrombi
CMR has not been proven in all areas,11 CMR is the preferred modality can be identified by persistent low-signal intensity in different phases
in both aortic disease, including aneurysm and dissection, and its pre- of the cardiac cycle. Turbulent flow produces rapid spin dephasing
cursors, and in congenital and inherited heart diseases. This chapter and results in a signal void, providing additional information in many
focuses on emerging advantages of CMR with respect to a spectrum pathologic conditions such as coarctation, aortic valve insufficiency,
of aortic pathologies. aortic aneurysm, and dissection.13 Particularly in aortic dissection, the
detection of entry and reentry sites is a special capability of functional
PRINCIPLES OF CARDIOVASCULAR MAGNETIC CMR that can be helpful in planning both surgical and endovascular
RESONANCE IN AORTIC IMAGING therapy. Accurate quantitative information on blood flow is obtained
from modified gradient echo sequences with parameter reconstruction
Spin Echo Cardiovascular Magnetic Resonance from the phase rather than the amplitude of the magnetic resonance
Spin echo T1-weighted imaging provides the anatomic detail of the (MR) signal; this is also known as flow mapping or phase contrast
aortic wall and pathologic conditions such as atheromatous plaques, or velocity-encoded cine CMR14 (Fig. 43.2). In each pixel of velocity
intimal flaps, or intramural hemorrhage and is still the basis of any images, the phase of the signal is related to the velocity component
aortic study, whereas T2-weighted images (repetition time: 2/3 RR; in the direction of a bipolar velocity phase-encoding gradient. In the
echo time: 80 to 100 ms) can be used in tissue characterization of the phase image, the velocity of blood flow can be determined for any site
aortic wall or blood components. Electrocardiogram (ECG) triggering of the vascular system. Flow velocity is calculated by using a formula in
is essential in minimizing motion and pulsatility artifacts. Slice thick- which velocity is proportional to change in the phase angle of protons
ness of 3 to 8 mm and an echo time (TE) of 20 to 30 ms are standard, in motion. MR maps of flow velocity are obtained two-dimensionally,

509
510 SECTION V Vascular/Pericardium

which is particularly important in profiles of nonuniform flow, such as slow flow or turbulence-induced signal voids are avoided. During the
in the great vessels. Quantitative data on flow velocity and flow volume short intravascular phase, the paramagnetic contrast agent provides a
are obtained from the velocity maps through a region of interest. The precise signal in the arterial or venous system, enhancing the vessel-
mean blood flow is estimated by multiplying the spatial mean velocity to-background contrast-to-noise ratio, irrespective of flow patterns and
and the cross-sectional area of the vessel. Vector mapping has been used velocity and not only delineates the dissection lamella, but also filling
to describe flow patterns in different aortic diseases (e.g., hypertension, of critical side branches. Intravenous bolus timing is necessary to ensure
aneurysms, dissection, Marfan syndrome, coarctation).15,16 peak enhancement during the middle of CMR acquisition and not
exceeding the acquisition time.19 Improved gradient systems allow a
Magnetic Resonance Angiography considerable reduction of the minimum TRs and TEs and the acquisi-
A variety of magnetic resonance angiography (MRA) techniques, includ- tion of complex 3D datasets within a breath-hold interval of under 30
ing various pulse sequences, methods of data acquisition, and postpro- seconds. With the support of maximum intensity projection (MIP)
cessing, have been developed, but first-pass 3D contrast-enhanced MRA images and the 3D multiplanar reformation, this technique delineates
constitutes the method of choice for the evaluation of the aorta.8,17,18 all the morphologic details of the aorta and its side branches in any
The technique relies on the contrast-induced T1-shortening effects of plane in a 3D format (Fig. 43.3). Some caution has to be exercised in
the gadolinium (Gd) contrast agent, whereby saturation problems with patients with poor renal function because this agent has been associated
with the development of nephrogenic systemic fibrosis. From a technical
point of view, blood pool imaging can be acquired without the use of
a contrast agent using an ECG-gated and respiratory-navigated balanced
steady-state free precession (SSFP) acquisition. After an intervention,
first-pass contrast-enhanced angiography is instrumental in assessing
the success of a procedure and in quantifying false lumen thrombosis,
a well-established prognostic indicator. The image acquisition is timed
AAo
according to the arrival of the contrast bolus in the proximal unaffected
aorta and thrombosis is assumed to be present when there is no contrast
agent in the false lumen. However, recent studies have shown that the
flow rates in the false lumen are highly variable and often very slow.20–22
The use of a new CMR technique with gadofosveset trisodium blood
RPA pool agent in equilibrium state is encouraging.20,21
Finally, phase contrast sequences may be used to assess blood flow
and velocity in both lumens of a dissection. This imaging technique
does not play a major role in the context of diagnostic imaging; however,
it appears to be an interesting research tool and potentially provides
DAo parameters for a prognostic assessment of a given patient. These sequences
can also be used to acquire 3D velocity information (Vx, Vy, Vz) for
each voxel, within a 3D volume, over time (frequently called 4D phase
contrast-CMR) (Fig. 43.4). This acquisition offers the potential to study
aortic hemodynamics, flow patterns, and derived vessel wall parameters,
such as wall shear stress. This technique can help to identify entry tears
FIG. 43.1  Black-blood vascular imaging of the aorta obtained with cardiac between the true and false lumens and to stratify patients according to
gating and breath holding. Axial image shows the ascending (AAo) and their risk of aneurysm formation.21,23
descending (DAo) aorta at the level of the right pulmonary artery (RPA). Four-dimensional time-resolved angiography with keyhole (TRAK)
Note the excellent suppression of the luminal blood signal and demon- is a contrast-enhanced MR angiography technique that can acquire
stration of the vessel wall. multiple time-resolved 3D volumes over time using image acceleration

AAo
MPA

DAo

A B
FIG. 43.2  Phase-contrast imaging of the aorta. Magnitude (A) and phase (B) axial images show the ascend-
ing (AAo) and descending (DAo) aorta at the level of the main pulmonary artery (MPA). Flow encoding was
superior to inferior. On the phase image (B), the ascending aorta and main pulmonary artery appear black,
and the descending aorta appears white, owing to the opposite directions of flow in these arteries.
CHAPTER 43  Thoracic Aortic Disease 511

techniques.24 It can be used to acquire several phases of contrast dis- originates in the ascending aorta, and the dissecting hematoma extends
tribution, including, but not limited to, the arterial and venous phases. past the origin of the left subclavian artery, whereas type II dissections
This technique can be used clinically to characterize flow-related phe- are confined to the ascending aorta. Type III dissections begin after the
nomena, such as false lumen thrombus distribution and endoleak.21,24 origin of the left subclavian artery and extend distally. The Stanford
classification is conceptually founded on prognostic grounds, where
type A involves the ascending aorta (Fig. 43.6), regardless of the site
DISSECTION OF THE THORACIC AORTA of the entry tear, and type B dissections spare the ascending aorta and
Acute aortic dissection is a life-threatening medical emergency requir- often imply a better prognosis.25 In general, acute dissections of the
ing prompt diagnosis and treatment. The 14-day period after onset ascending aorta require emergency surgery, whereas descending aortic
has been designated as an acute phase because the rates of morbidity dissections may be managed with medical therapy and stent-grafting;
and mortality are highest during this period.26,27 The two most fre- in any case rapid diagnosis of the dissecting process and a delineation
quently used classifications (DeBakey and Stanford) are based on the of its anatomic details are critical for successful management.28,29 The
anatomic location and extension of intimal flap (Fig. 43.5). DeBakey’s primary imaging goal is not only to establish the diagnosis of dissection,
nomenclature is based on the anatomic site of the intimal tear and the but also detection of entry and reentry sites, presence and degree of
extent of the resulting dissection. In a type I dissection, the intimal tear aortic insufficiency, and the flow pattern in the true and false lumen as
well as in critical aortic branches.5 The latter is crucial for patient selec-
tion for transcatheter endovascular repair of type B acute and chronic
aortic dissection as an alternative to open surgery.30,31 For diagnostic
purposes the true lumen can be differentiated from the false lumen by
the anatomic and functional (flow-related) features. In addition, the
visualization of remnants of the dissected media as cobwebs adjacent
to the outer wall of the lumen may help to identify the false lumen.
The leakage of blood from the descending aorta into the periaortic
space, which can appear with high signal intensity and can result in a
left-sided pleural effusion, is usually better visualized on axial images.
A high signal intensity of a pericardial effusion indicates a bloody com-
ponent and is considered to be a sign of impending rupture of the
ascending aorta into the pericardial space. A detailed anatomic map of
aortic dissection must indicate the type and extension of dissection and
distinguish the origin and perfusion of branch vessels from the true or
false channels. In stable patients, adjunctive gradient echo sequences or
phase contrast images can be instrumental in identifying aortic insuf-
ficiency and entry or reentry sites, as well as in differentiating slow flow
from thrombus in the false lumen.20,32 The third step in the diagnosis
of aortic dissection and definition of its anatomic detail relies on the
use of Gd-enhanced 3D MRA. Because 3D MRA is rapidly acquired
without any need of ECG triggering and Gd has minimal toxicity in
patients with good renal function, this technique may even be used
with severely ill patients.33 With spin echo sequences, artifacts caused
by imperfect ECG gating, respiratory motion, or slow blood pool can
FIG. 43.3  Gadolinium-enhanced three-dimensional magnetic resonance result in intraluminal signal, simulating or obscuring an intimal flap.
angiography of the thoracic aorta (surface-shaded display algorithm). In Gd-enhanced 3D MRA, the intimal flap is easily detected, and the

A B
FIG. 43.4  Transverse images of type B aortic dissection (A and B) acquired using four-dimensional phase
contrast magnetic resonance imaging. These images demonstrate the velocity of blood flow in the heart and
aorta. High-velocity blood flow is shown in turquoise and low velocity in blue.
512 SECTION V Vascular/Pericardium

Stanford Type A Stanford Type B

Type II Type IIIa

De Bakey Type I

Type IIIb
FIG. 43.5  Commonly used classification systems for aortic dissection. Although the DeBakey classification
(I, II, IIIa, IIIb) focuses on the anatomic extent, the Stanford classification (A, B) highlights the involvement
of the ascending aorta and the prognostic aspects of dissection.

definition.34 At present, CMR is one of the most accurate tools in the


detection of aortic dissection. A high degree of spatial resolution and
contrast, and the capability for multiplanar acquisition provide excel-
lent sensitivity and specificity rating at approximately 100% in the
published series.34–35
In selecting the diagnostic modality of choice, begin by considering
what diagnostic information needs to be obtained. First, any chosen study
must confirm or refute the diagnosis of dissection. Second, the study
must determine whether the dissection involves the ascending aorta (type
A) or is confined to the descending aorta (type B). Third, a number of
anatomic features of the dissection should be identified, including its
extent, the sites of entry and reentry, the presence of thrombus in the
false lumen, the extent of branch vessel involvement, the presence and
severity of aortic insufficiency, the presence of a pericardial effusion,
and the presence of coronary artery involvement. It is also important to
A consider the accuracy of the diagnostic information obtained because a
false-negative diagnosis may result in avoidable death, whereas a false-
positive diagnosis might lead to unnecessary surgery.
According to the present information, CMR and TEE are the most
sensitive modalities, with both performing better than aortography.
The sensitivities of aortography, CT, and CMR are all quite high, whereas
the specificity of TEE may be comparable only when a strict definition
of a positive study is applied. Finally, availability, speed, safety, and cost
B should be taken into consideration in comparing various modalities.
FIG. 43.6  Stanford type A aortic dissection. (A) Axial T1-weighted black- Aortography is rarely immediately available, requires transport of the
blood image shows nearly circumferential compression of the true aortic patient, is a lengthy study, has the associated risks of both an invasive
lumen by a false lumen (arrowhead). High signal intensity in the false study and intravenous contrast, and is the most expensive. Yet it may
lumen makes it difficult to differentiate thrombosis from flowing blood. be necessary in selected patients who are being considered for combined
(B) Axial reformatted image from contrast-enhanced magnetic resonance surgical treatment, especially those with a high likelihood of coronary
angiography shows an intimal flap (black arrowhead) with flow in the artery disease or evidence of involvement of major arterial trunks arising
false lumen (white arrowhead).
from the aorta. CT scanning has the advantage that it is more easily
obtained in less time and is noninvasive, but it is overall less accurate
than the other techniques. CMR is usually less available in most hos-
relationship with aortic vessels is clearly depicted. Entry and reentry sites pitals, requires transportation of the patient, and is considered undesir-
appear as a segmental interruption of the linear intimal flap on axial or able for unstable patients or those requiring very close monitoring.
sagittal images. The analysis of MRA images should not be limited to Meanwhile, TEE is readily obtained, quick to complete at the bedside
viewing MIP images or surface-shaded display; it should also include a and thus ideal for unstable patients, and is the least costly of the four
complete evaluation of reformatted images in all three planes to confirm imaging techniques. These options render angiography obsolete in the
or improve spin echo information and exclude artifacts. Combining diagnostic workup of aortic dissection. Thus detecting dissection of
the spin echo with MRA images completes the diagnosis and anatomic the thoracic aorta should be a noninvasive strategy using CT or CMR
CHAPTER 43  Thoracic Aortic Disease 513

in hemodynamically stable patients and TEE in patients who are too


AORTIC INTRAMURAL HEMATOMA
unstable for transportation. Comprehensive and detailed evaluation
can thus be reduced to a single noninvasive imaging modality in the An important differential diagnosis of aortic dissection is intramural
evaluation of suspected aortic dissection. hematoma (IMH), which usually presents with the same clinical picture
Although CMR may be less practical than CT and TEE to evaluate and risk profile as overt aortic dissection.37 Tomographic modalities
patients presenting with suspected aortic dissection, it is well suited for such as CMR may be used to identify IMH with no luminal component,
patients with stable chronic dissections. The extraordinary accuracy of which is considered an imminent precursor of aortic dissection.38,39
CMR and the optional functional images may render CMR the gold With high-resolution tomographic imaging, the imaging diagnosis of
standard for defining aortic anatomy and risk of rupture in dissection IMH is now feasible, suggesting IMH as a precursor of dissection, with
patients; for surveillance of dissection, CMR has already been widely a 30% progression to overt dissection.39,40 Because of these findings in
accepted regardless of medical treatment, surgical repair, or endovascular vivo before death, IMH appears more likely to be a variant of dissection
management. than a separate entity.41,42 Typical epiphenomena of dissection such as
CMR can be used to custom design an individual stent graft for aortic insufficiency, pericardial or pleural effusion may also occur in
lesions such as aneurysms, aortic dissections, and localized aortic ulcers. IMH. Spontaneous rupture of aortic vasa vasorum, especially of nutri-
With customized aortic endoprosthesis, such entities are becoming more ent vessels to the media layer, has been suggested to initiate the process
frequently an ideal target for interventional treatment rather than surgi- of aortic disintegration without an intimal tear. With a pathogenesis
cal approaches that have high mortality and morbidity36 (Fig. 43.7). that explains the high rate of progression to overt aortic dissection and
a prognosis and survival that are similar to those in aortic dissection,
urgent diagnosis of IMH is very important.
The diagnosis of IMH relies on the visualization of intramural
blood and/or evidence of localized increased wall thickness.43 CMR
techniques, however, not only visualize the blood in the wall, but also
allow an assessment of the age of the hematoma based on signal changes
caused by the formation of methemoglobin (Fig. 43.8). Acute IMH
(early stage) is well imaged on T2-weighted spin echo images because
of high initial signal intensity of blood, whereas blood of 1 to 5 days
of age has lower signal intensity on T2 images. High signal intensity
within the aortic wall on T1 spin echo images suggests subacute IMH,
whereas acute IMH may be determined on T1 images from the isodense
appearance of blood and aortic wall.41,43 Although TEE has an excellent
sensitivity to detect aortic dissection, the definite distinction between
IMH and normal findings may require a second tomographic modality
such as CT or CMR, because a false-negative result (or false exclusion
A B of IMH) is more likely to be avoided with independent morphologic
information. In conclusion, as a precursor of dissection, IMH requires
FIG. 43.7  Contrast-enhanced magnetic resonance angiography (MRA)
of chronic type B dissection originating from the aortic arch region.
diagnostic attention by use of high-resolution tomographic imaging;
(A) Follow-up MRA at 7 days after stent-graft placement shows a com- owing to its physical properties, CMR may play a prominent role not
pletely sealed proximal entry to the thrombosed false lumen. (B) The only to diagnose IMH, but also to assess its age and to differentiate
diameter of the true lumen is normalized, and the descending aorta is IMH from mural thrombosis; angiography certainly is not diagnostic
reconstructed. in the setting of IMH.

A B C
FIG. 43.8  T1-weighted spin echo axial image of intramural hematoma of the ascending and descending
aorta. The abnormal wall thickening (arrowheads) present intermediate signal intensity in panel A (oxyhemo-
globin, acute phase) and high signal intensity in panel B (methemoglobin, subacute phase). (C) T2-weighted
spin echo image signal intensity is high in the acute phase (recent hemorrhage).
514 SECTION V Vascular/Pericardium

PENETRATING AORTIC ULCER THORACIC AORTIC ANEURYSM


Ulcers occur in the presence of aortic atherosclerosis and may mimic Limited dilatation of a blood vessel can be either a true aneurysm or a
subacute dissection or may develop without major symptoms. In contrast false aneurysm. True aneurysms involve all layers of the aortic wall and
to IMH, aortic ulcers are characterized on angiography by focal contrast result from the degeneration of the elastin fibers within the media. A
enhancement beyond the confines of the aortic lumen but communi- false aneurysm, or pseudoaneurysm, is not a true aneurysm but rather a
cating with the lumen (Fig. 43.9).44 At present, there is emerging con- contained perforation of the vessel wall with penetration of the intima
sensus on the prognosis and outcome of aortic ulcers. Persistent pain, and media. Pseudoaneurysms form after focal penetration of the intima
hemodynamic instability, or expansion more than 20 mm width or and media by trauma (20% to 25% of thoracic aneurysms) or by infec-
depth should trigger surgical or endovascular treatment. An incidence tion (about 5% of thoracic aneurysms). Atherosclerosis may result in
of transmural rupture ranging from 8% to 42% has been reported in the formation of pseudoaneurysms if a true aneurysm ruptures but is
the medical literature.45,46 Both IMH and ulcers are unrelated to intimal contained by the adventitia and periaortic tissues. Pseudoaneurysms,
lacerations as in acute aortic dissection. Lacerations and IMH usually therefore, have a narrow “neck” leading to the “aneurysm.” Thoracic
occur at points of greatest hydraulic stress (right lateral ascending aorta aortic aneurysms are common, with a recently reported incidence of
or adjacent to the ligamentum arteriosum), whereas penetrating ulcers 10.9 cases per 100,000 persons per year.49 They may involve all thoracic
are typically found in the descending or abdominal aorta. Conversely, aortic segments, requiring different therapeutic strategies with a surgi-
discrete penetrating atheromatous ulcers (“giant ulcers”) have been cal, interventional, or hybrid approach (Fig. 43.10). In modern series,
suspected as one cause of intramural bleeding. Both MRI and TEE have aneurysms of the ascending aorta occur most commonly (60%), fol-
helped to elucidate the pathogenic background and the complex ana- lowed by aneurysms of the descending aorta (40%), while isolated arch
tomic peculiarities of these important features, but accurate diagnosis aneurysms or thoracoabdominal aneurysms are seen less often and are
of penetrating ulcers is sometimes difficult, and no test is ideal.47 CT considered integral parts of those two groups.50 Atherosclerotic aneurysms
may demonstrate the surrounding hematoma and displaced calcifica- are usually fusiform, involving long segments of the aorta. Conversely,
tions in most cases; in addition to this, CMR can differentiate subacute ascending aortic aneurysm or annuloaortic ectasia are often caused by
intramural hematoma from chronic intraluminal thrombus.48 No large- Marfan syndrome, syphilis, or poststenotic dilatation resulting from
scale studies of CMR and aortic ulcers are currently available, but owing aortic stenosis; the sinotubular junction is preserved in aortic stenosis
to its versatility and imaging features, CMR appears best suited to but markedly dilated in the other entities. If isolated to the descending
characterize this form of aortic pathology as a constellation of diffuse aorta, saccular aneurysms may be traumatic or infective, but atheroscle-
aortic atherosclerosis, an ulceration with focal wall thickening, and rosis remains the leading cause. Saccular and sinus Valsalva aneurysms
missing evidence of aortic dissection. in the aortic root very often have associated aortic insufficiency. The
clear delineation of the location, extent, and shape of an aneurysm; its
relation to branch vessels and adjacent structures; and its associated
complicating factors, such as rupture, periaortic hematoma or infection,
hemopericardium, or aortic valve insufficiency, are all of importance.
CMR is effective in identification and characterization of thoracic
aortic aneurysms, as well as in evaluation of their pathophysiologic
consequences.51–53 Standard spin echo sequences are helpful in evalu-
ation of alterations of the aortic wall and periaortic space. Periaortic
hematoma and areas of high signal intensity within the thrombus may
indicate instability of the aneurysm and are well depicted on spin echo
images. Atherosclerotic lesions are visualized as areas of increased thick-
ness with high signal intensity and irregular profiles. In contrast with
transaxial imaging, oblique planes allow precise determination of lumen
diameter. With fat suppression techniques, the outer wall of the aneu-
rysm can easily be distinguished from periadventitial fat tissue. Contrast-
enhanced 3D MRA can provide precise topographic information about
the extent of an aneurysm and its relationship to the aortic branches
(Fig. 43.11). The capability of contrast MRA to visualize the Adamkie-
wicz artery represents an important advance in avoiding postoperative
neurologic deficit secondary to spinal cord ischemia.54,55
At present, no trials exist to compare the value of CMR and offer
modalities for the diagnosis of thoracic aneurysms; there is, however,
consensus that CMR is accurate and versatile but most likely not as
cost-effective in serial studies. Although CMR promises to emerge as
the standard in the diagnosis and management of thoracic aortic aneu-
rysms, cost considerations have to be evaluated before recommendation
of widespread use of MRI scanning to follow stable patients with sub-
critical aneurysms.
FIG. 43.9  Penetrating atherosclerotic ulcer in a 61-year-old man with
high blood pressure. Sagittal oblique contrast-enhanced magnetic reso-
nance angiography shows a diffusely aneurysmal descending aorta with TRAUMA TO THE AORTA
a large outpouching in the medial anterior region (arrowhead), an appear-
ance consistent with penetrating atherosclerotic ulcer. Prior studies Aortic trauma is usually generated by deceleration of the body in serious
showed development of a focal aneurysm 2 years earlier. motor vehicle crashes, pedestrian injuries, and falls from height.56 The
CHAPTER 43  Thoracic Aortic Disease 515

A B
FIG. 43.10  (A) Magnetic resonance angiography images of an aortic arch aneurysm in parasagittal-oblique
and axial orientation. (B) Follow-up study after a hybrid procedure with initial head-vessel-debranching and
staged endovascular stent-graft repair demonstrates perfect reconstruction of the proximal aorta.

A B
FIG. 43.12  Contrast-enhanced magnetic resonance angiography of
chronic traumatic aortic lesion. (A) Partial laceration of the aortic wall
results in a diverticular aneurysm (shaded surface rendering). (B) Dis-
charge imaging after endovascular stent-graft repair reveals complete
restitution of thoracic aortic integrity.

FIG. 43.11  Three-dimensional shaded-surface-rendered contrast-enhanced been described in pathologic series but was not recognized in the clini-
magnetic resonance angiography depicts a large true aneurysm of the cal setting before the advent of high-resolution tomographic imaging
thoracic descending aorta in a 38-year-old man. modalities. Recent reports indicate that intimal hemorrhage with and
without partial intimal laceration tends to heal spontaneously. When
the lesion involves intimal and medial layers, false aneurysm formation
aortic segment subjected to the greatest strain by rapid deceleration occurs. The aneurysm is fusiform in the case of a circumferential lesion,
forces is located just beyond the area of the isthmus aortae. In clinical whereas in a partial laceration, it appears as localized diverticulum (Fig.
series, aortic rupture occurs at this location in 90% of cases.57,58 The 43.12). Periaortic hemorrhage may occur irrespective of the type of
traumatic lesion is a transverse tear variably extending from the intimal lesion. Complete rupture of the aorta, including the adventitial layer
to adventitial layers. Intimal hemorrhage without any laceration has and periadventitial connective tissue, leads to immediate exsanguination.
516 SECTION V Vascular/Pericardium

However, false aneurysm or occlusion of the side of rupture may permit


temporary survival. A long examination time and difficult access to the
polytraumatized patient have been considered to be the main limitations
of CMR in acute aortic pathology. The development of fast CMR tech-
niques has shortened the examination time to a few minutes; therefore
MRI can be used even in critically ill patients. The value of CMR in
detecting traumatic aortic rupture in comparison with angiography and
conventional CT was reported in a series of 24 consecutive patients.59
The potential for CMR to detect the hemorrhagic component of a lesion
by its high signal intensity is beneficial in traumatized patients. On spin
echo images in the sagittal plane, a longitudinal visualization of the tho-
racic aorta makes it possible to distinguish a partial lesion from a lesion
encompassing the entire aortic circumference. This discrimination is of
prognostic significance because a circumferential lesion may be more
likely to rupture.59 The presence of periadventitial hematoma and/or A B
pleural and mediastinal hemorrhagic effusion may also be considered
a sign of instability. In the same sequence used to evaluate the aortic FIG. 43.13  (A) Magnetic resonance angiography of aortic coarcta-
lesion, without the need of any additional time, the wide field of view tion depicts focal stenotic segment (arrowhead) at the isthmic zone
of CMR provides a comprehensive evaluation of chest trauma such as and multiple intercostal collateral arteries joining the descending aorta.
(B) Medium-sized Dacron patch aneurysm (arrowhead) diagnosed 30
lung contusion and edema and pleural effusion and rib fractures. MR
years after coarctation surgery.
angiography does not add any diagnostic value to spin echo CMR, and
it cannot supply information on parietal lesions and hemorrhagic fluids
outside the aortic vessel. Recently, the development of endovascular the coarctation severity with good sensitivity and specificity (95% and
technique has provided additional opportunities in the treatment of 82%, respectively) compared with catheter angiography. Flow mapping
acute onset and chronic traumatic aortic disease (see Fig. 43.12).60–62 is also able to quantify the flow pattern and volume of collateral flow
in the descending aorta, which are other important parameters of the
Aortic Coarctation severity of coarctation, and this information may be crucial in the
Coarctation is a common congenital anomaly, with an incidence of 20 choice of surgical strategy. Collateral flow is present if distal aortic flow
to 60 per 100,000 live births, and represents 5% to 8% of all congenital is greater than proximal flow and it is possible to quantify collateral
cardiovascular disorders. The obstructive lesion results from an abnor- circulation by subtracting flow volume in the proximal descending aorta
mality in the aortic media and refers to an enfolding of the posterolateral from that in the distal portion. The presence of collateral flow indicates
aortic wall in the region of the ligamentum or ductus arteriosus. This a hemodynamically significant coarctation. Furthermore, a pressure
is usually a discrete phenomenon occurring just distal to the ductus gradient calculated with flow mapping CMR by use of the modified
and is also labeled postductal coarctation. Because it is usually asymp- Bernoulli equation (P = 4V2)66 can be used to determine the need for
tomatic in the neonatal period, it is also referred to as adult coarctation surgical/endovascular repair (a pressure gradient greater than 15 mm Hg
versus preductal or infantile coarctation, which is less common than is considered an indication for intervention). However, this threshold
adult coarctation and usually associated with hypoplasia of the arch. is arbitrary and therefore it may be more appropriate to use the mea-
There is usually a dilation of the descending aorta distal to the coarcta- surement of collateral flow.
tion. As a result of the obstruction caused by the coarctation, collateral Several therapeutic strategies are available for the treatment of aortic
vessels develop to increase flow into the descending aorta. Increased coarctation, depending on the morphology of the affected aorta as
flow through intercostal arteries results in their dilation, and notching well as the age and clinical condition of the patient.67,68 Surgery for
along the inferior aspect of the ribs, which usually takes 8 to 10 years aortic coarctation is recommended at an early age because long-term
to become significant enough to be observed on a chest radiograph. results seem to be better. Recently, interventional procedures and balloon
With its multiplanar image acquisition, large field of view, and angioplasty have come into wide use and provide good results, espe-
dynamic quantitative flow imaging capacity, CMR appears the modality cially in mild or moderate cases.69,70 An accurate selection of favorable
of choice for evaluation of coarctation (Fig. 43.13). The left oblique anatomy by high-resolution imaging modalities is particularly impor-
sagittal view centered on the middle of the ascending and descending tant in interventional procedure to ensure a low rate of complications
aortas is an ideal orientation that may also demonstrate associated and restenosis.71 An increased risk for aneurysm formation at the site
aortic stenosis, left ventricular hypertrophy, and ventricular septal defects. of repair has been reported after both synthetic patch aortoplasty and
This is important because there is a high association of bicuspid aortic subclavian-flap arterioplasty (see Fig. 43.13).72–74 Moreover, restenosis,
valve and ventricular septal defects with coarctation. The severity of aortic dissection, and pseudoaneurysms have been reported after surgery
the stenosis can be expressed as the ratio of the diameters or cross- or balloon angioplasty in up to 42% of patients.75 Therefore routine
sectional areas measured at the coarcted segment and above the dia- follow-up is recommended for patients who underwent repair of an
phragm.63 However, although the anatomic narrowing of the aorta aortic coarctation, independently of surgical technique used and timing
establishes the diagnosis of coarctation, an assessment of its clinical of the repair. New interventional techniques, such as endovascular stent-
significance depends on determining its hemodynamic effects. Cine grafting, have currently been applied to the treatment of postsurgical
CMR has been applied to evaluate flow turbulence across the coarcta- patch aneurysms with excellent results, avoiding the need for further
tion; the severity of coarctation is quantified on the basis of the length surgical intervention.76,77
of flow void.64 Further functional information can be provided by CMR
flow mapping, which can define the severity of the stenosis by measur- Aortitis
ing velocity jets at the level of coarctation and mean flow deceleration Although there are many causes of aortitis, Takayasu arteritis is the
in the descending aorta.65 With this technique, it is possible to predict type most often studied with CMR because of the diffuse stenotic nature
CHAPTER 43  Thoracic Aortic Disease 517

A B C
FIG. 43.14  Takayasu arteritis. (A) Coronal maximum intensity projection three-dimensional magnetic reso-
nance angiography shows significant stenosis of right common carotid artery at its origin (small arrow). The
left subclavian artery has two stenotic segments (large arrows) with a small area of poststenotic dilatation
in between. There are also some luminal irregularities of the left common carotid artery. Axial unenhanced
(B) and gadolinium (Gd)-enhanced (C) T1-weighted cardiovascular magnetic resonance shows wall thickening
of the ascending aorta (arrow), which is enhanced on the Gd-enhanced image.

of the disease, which often makes vascular access impossible by catheter- instrument for CMR visualization requires no hardware or instrument
ization.78 In Takayasu disease, the aortic arch vessels are primarily affected, modifications and appears to be promising in terms of potential clinical
but thoracic and abdominal aorta may also be involved (Fig. 43.14). applications. However, CMR-compatible instruments with satisfactory
Active inflammatory disease demonstrates diffuse thickening of the susceptibility artifacts are required, as well as CMR-compatible guide
aortic wall, typically enhanced after Gd administration in T1-weighted wires with adequate mechanical support. To date, the limitations of
images. The chronic stage, however, is characterized by extensive peri- interventional CMR are long procedure times, lack of true real-time
vascular fibrosis without postcontrast enhancement.79 MRA is the pre- monitoring, and stent artifacts, which necessitate further modifications
ferred imaging modality for the study of branch vessels stenosis and before they can be recommended for clinical use.86,87 However, CMR-
has replaced invasive angiography, which carries a risk of pseudoaneurysm guided interventions are feasible and may become an important advance
formation at the site of arterial puncture. Recently, the capability of in the near future.
F-18 fluorodeoxyglucose hybrid camera positron emission tomography
(PET) combined with CMR to detect early stages of Takayasu arteritis
has been demonstrated.80
CONCLUSION
With recent advances in the understanding of aortic diseases, both power
INTERVENTIONAL CARDIOVASCULAR and versatility have put magnetic resonance imaging in the focus of
diagnostic workup and surveillance of all aortic pathology. Technical
MAGNETIC RESONANCE refinements, from classic anatomic imaging, 3D Gd-enhanced MRA
CMR guidance of vascular interventional procedures offers several and tissue characterization to 4D functional imaging, have rendered
potential advantages over fluoroscopy-guided techniques, including CMR ideal for assessment of acquired disease, such as aortic dissec-
image acquisition in any desired orientation, superior 3D soft tissue tion, intramural hematoma, and aneurysm, along with postoperative
contrast with simultaneous visualization of the interventional device follow-up evaluation, with better diagnostic reliability and prognostic
and 4D functional imaging, and absence of ionizing radiation. The information than other imaging modalities. The guidance of vascular
feasibility of real-time CMR-guided interventions has been demonstrated interventional procedures by CMR offers potential advantages over
for a wide range of vascular interventional procedures in animals and fluoroscopy-guided techniques and will continue to advance in the
patients.81–84 In terms of endovascular aortic stent-graft placement, CMR near future.
appears to be particularly useful because it can provide preinterventional
evaluation of aortic pathology, real-time interventional image guidance,
and immediate evaluation of treatment success or procedure-related
complications, as well as follow-up examinations.85 Passive device track-
REFERENCES
ing using material-induced susceptibility artifacts of the interventional A full reference list is available online at ExpertConsult.com
CHAPTER 43  Thoracic Aortic Disease 517.e1

REFERENCES 23. Francois CJ, Markl M, Schiebler ML, et al. Four-dimensional, flow-
sensitive magnetic resonance imaging of blood flow patterns in
1. Hayashi K, Meaney TF, Zelch JV, et al. Aortographic analysis of aortic thoracic aortic dissections. J Thorac Cardiovasc Surg. 2013;145(5):
dissection. Am J Roentgenol Radium Ther Nucl Med. 1974;122(4):769–782. 1359–1366.
2. Singh H, Fitzgerald E, Ruttley MS. Computed tomography: the 24. Willinek WA, Hadizadeh DR, Von Falkenhausen M, et al. 4D time-
investigation of choice for aortic dissection? Br Heart J. 1986;56(2): resolved MR angiography with keyhole (4D-TRAK): more than 60 times
171–175. accelerated MRA using a combination of CENTRA, keyhole, and SENSE
3. Wilbers CR, Carrol CL, Hnilica MA. Optimal diagnostic imaging of at 3.0T. Magn Reson Imaging. 2008;27(6):1455–1460.
aortic dissection. Tex Heart Inst J. 1990;17(4):271–278. 25. Daily PO, Trueblood HW, Stinson EB, et al. Management of acute aortic
4. Nienaber CA, von Kodolitsch Y, Nicolas V, et al. The diagnosis of thoracic dissection. Ann Thorac Surg. 1970;10(3):237–247.
aortic dissection by noninvasive imaging procedures. N Engl J Med. 26. Mid-term outcomes and aortic remodelling after thoracic endovascular
1993;328(1):1–9. repair for acute, subacute, and chronic aortic dissection: the VIRTUE
5. Cigarroa JE, Isselbacher EM, DeSanctis RW, et al. Diagnostic imaging in Registry. Eur J Vasc Endovasc Surg. 2014;48(4):363–371.
the evaluation of suspected aortic dissection: old standards and new 27. Acosta S, Blomstrand D, Gottsater A. Epidemiology and long-term
directions. N Engl J Med. 1993;328(1):35–43. prognostic factors in acute type B aortic dissection. Ann Vasc Surg.
6. Ballal RS, Nanda NC, Gatewood R, et al. Usefulness of transesophageal 2007;21(4):415–422.
echocardiography in assessment of aortic dissection. Circulation. 28. Nienaber CA, Eagle KA. Aortic dissection: new frontiers in diagnosis and
1991;84(5):1903–1914. management: part II: therapeutic management and follow-up.
7. Galvin IF, Black IW, Lee CL, et al. Transesophageal echocardiography in Circulation. 2003;108(6):772–778.
acute aortic transection. Ann Thorac Surg. 1991;51(2):310–311. 29. Nienaber CA, Eagle KA. Aortic dissection: new frontiers in diagnosis and
8. Russo V, Renzulli M, Buttazzi K, Fattori R. Acquired diseases of the management: part I: from etiology to diagnostic strategies. Circulation.
thoracic aorta: role of MRI and MRA. Eur Radiol. 2006;16(4):852–865. 2003;108(5):628–635.
9. Nienaber CA, Kische S, Skriabina V, Ince H. Noninvasive imaging 30. Nienaber CA, Fattori R, Lund G, et al. Nonsurgical reconstruction of
approaches to evaluate the patient with known or suspected aortic thoracic aortic dissection by stent-graft placement. N Engl J Med.
disease. Circ Cardiovasc Imaging. 2009;2(6):499–506. 1999;340(20):1539–1545.
10. Trimarchi S, Nienaber CA, Rampoldi V, et al. Role and results of surgery 31. Dake MD, Kato N, Mitchell RS, et al. Endovascular stent-graft placement
in acute type B aortic dissection: insights from the International Registry for the treatment of acute aortic dissection. N Engl J Med. 1999;340(20):
of Acute Aortic Dissection (IRAD). Circulation. 2006;114(suppl 1): 1546–1552.
I357–I364. 32. Chang JM, Friese K, Caputo GR, et al. MR measurement of blood flow in
11. Abernethy LJ. Cost effectiveness of magnetic resonance imaging. BMJ. the true and false channel in chronic aortic dissection. J Comput Assist
1992;304(6820):183. Tomogr. 1991;15(3):418–423.
12. Stemerman DH, Krinsky GA, Lee VS, et al. Thoracic aorta: rapid 33. Fernandez GC, Tardaguila FM, Duran D, et al. Dynamic 3-dimensional
black-blood MR imaging with half-Fourier rapid acquisition with contrast-enhanced magnetic resonance angiography in acute aortic
relaxation enhancement with or without electrocardiographic triggering. dissection. Curr Probl Diagn Radiol. 2002;31(4):134–145.
Radiology. 1999;213(1):185–191. 34. Bogaert J, Meyns B, Rademakers FE, et al. Follow-up of aortic dissection:
13. Powell AJ, Maier SE, Chung T, et al. Phase-velocity cine magnetic contribution of MR angiography for evaluation of the abdominal aorta
resonance imaging measurement of pulsatile blood flow in children and and its branches. Eur Radiol. 1997;7(5):695–702.
young adults: in vitro and in vivo validation. Pediatr Cardiol. 35. Sommer T, Fehske W, Holzknecht N, et al. Aortic dissection: a
2000;21(2):104–110. comparative study of diagnosis with spiral CT, multiplanar
14. Bogren HG, Buonocore MH. 4D magnetic resonance velocity mapping transesophageal echocardiography, and MR imaging. Radiology.
of blood flow patterns in the aorta in young vs. elderly normal subjects. 1996;199(2):347–352.
J Magn Reson Imaging. 1999;10(5):861–869. 36. Nienaber CA, Zannetti S, Barbieri B, et al. Investigation of stent grafts in
15. Hopkins KD, Lehmann ED, Gosling RG. Aortic compliance patients with type B aortic dissection: design of the INSTEAD trial—a
measurements: a non-invasive indicator of atherosclerosis? Lancet. prospective, multicenter, European randomized trial. Am Heart J.
1994;343(8911):1447. 2005;149(4):592–599.
16. Groenink M, de Roos A, Mulder BJ, et al. Biophysical properties of the 37. Nienaber CA, Sievers HH. Intramural hematoma in acute aortic
normal-sized aorta in patients with Marfan syndrome: evaluation with syndrome: more than one variant of dissection? Circulation.
MR flow mapping. Radiology. 2001;219(2):535–540. 2002;106(3):284–285.
17. Lee VS, Martin DJ, Krinsky GA, et al. Gadolinium-enhanced MR 38. Nienaber CA, von Kodolitsch Y, Petersen B, et al. Intramural hemorrhage
angiography: artifacts and pitfalls. AJR Am J Roentgenol. 2000;175(1): of the thoracic aorta: diagnostic and therapeutic implications. Circulation.
197–205. 1995;92(6):1465–1472.
18. Sodickson DK, McKenzie CA, Li W, et al. Contrast-enhanced 3D MR 39. Sueyoshi E, Imada T, Sakamoto I, et al. Analysis of predictive factors for
angiography with simultaneous acquisition of spatial harmonics: a pilot progression of type B aortic intramural hematoma with computed
study. Radiology. 2000;217(1):284–289. tomography. J Vasc Surg. 2002;35(6):1179–1183.
19. Riederer SJ, Bernstein MA, Breen JF, et al. Three-dimensional contrast- 40. Evangelista A, Dominguez R, Sebastia C, et al. Long-term follow-up of
enhanced MR angiography with real-time fluoroscopic triggering: design aortic intramural hematoma: predictors of outcome. Circulation.
specifications and technical reliability in 330 patient studies. Radiology. 2003;108(5):583–589.
2000;215(2):584–593. 41. Murray JG, Manisali M, Flamm SD, et al. Intramural hematoma of the
20. Clough RE, Hussain T, Uribe S, et al. A new method for quantification of thoracic aorta: MR image findings and their prognostic implications.
false lumen thrombosis in aortic dissection using magnetic resonance Radiology. 1997;204(2):349–355.
imaging and a blood pool contrast agent. J Vasc Surg. 2011;1251–1258. 42. Sawhney NS, DeMaria AN, Blanchard DG. Aortic intramural hematoma:
21. Clough RE, Waltham M, Giese D, Taylor PR, Schaeffter T. A new imaging an increasingly recognized and potentially fatal entity. Chest. 2001;120(4):
method for assessment of aortic dissection using four-dimensional phase 1340–1346.
contrast magnetic resonance imaging. J Vasc Surg. 2012;55(4):914–923. 43. Wolff KA, Herold CJ, Tempany CM, et al. Aortic dissection: atypical
22. Karmonik C, Bismuth J, Redel T, et al. Impact of tear location on patterns seen at MR imaging. Radiology. 1991;181(2):489–495.
hemodynamics in a type B aortic dissection investigated with 44. Kazerooni EA, Bree RL, Williams DM. Penetrating atherosclerotic ulcers
computational fluid dynamics. Conf Proc IEEE Eng Med Biol Soc. of the descending thoracic aorta: evaluation with CT and distinction
2010;2010:3138–3141. from aortic dissection. Radiology. 1992;183(3):759–765.
517.e2 SECTION V Vascular/Pericardium

45. Coady MA, Rizzo JA, Elefteriades JA. Pathologic variants of thoracic 66. Mohiaddin RH, Kilner PJ, Rees S, et al. Magnetic resonance volume flow
aortic dissections: penetrating atherosclerotic ulcers and intramural and jet velocity mapping in aortic coarctation. J Am Coll Cardiol.
hematomas. Cardiol Clin. 1999;17(4):637–657. 1993;22(5):1515–1521.
46. Movsowitz HD, Lampert C, Jacobs LE, et al. Penetrating atherosclerotic 67. Carr JA. The results of catheter-based therapy compared with surgical
aortic ulcers. Am Heart J. 1994;128(6 Pt 1):1210–1217. repair of adult aortic coarctation. J Am Coll Cardiol. 2006;47(6):
47. Hayashi H, Matsuoka Y, Sakamoto I, et al. Penetrating atherosclerotic 1101–1107.
ulcer of the aorta: imaging features and disease concept. Radiographics. 68. Gibbs JL. Treatment options for coarctation of the aorta. Heart.
2000;20(4):995–1005. 2000;84(1):11–13.
48. Yucel EK, Steinberg FL, Egglin TK, et al. Penetrating aortic ulcers: 69. Mullen MJ. Coarctation of the aorta in adults: do we need surgeons?
diagnosis with MR imaging. Radiology. 1990;177(3):779–781. Heart. 2003;89(1):3–5.
49. Clouse WD, Hallett JW Jr, Schaff HV, et al. Improved prognosis of 70. Ovaert C, McCrindle BW, Nykanen D, et al. Balloon angioplasty of native
thoracic aortic aneurysms: a population-based study. JAMA. coarctation: clinical outcomes and predictors of success. J Am Coll
1998;280(22):1926–1929. Cardiol. 2000;35(4):988–996.
50. Isselbacher EM. Thoracic and abdominal aortic aneurysms. Circulation. 71. Paddon AJ, Nicholson AA, Ettles DF, et al. Long-term follow-up of
2005;111(6):816–828. percutaneous balloon angioplasty in adult aortic coarctation. Cardiovasc
51. Russo V, Renzulli M, Buttazzi K, et al. Acquired diseases of the thoracic Intervent Radiol. 2000;23(5):364–367.
aorta: role of MRI and MRA. Eur Radiol. 2006;16(4):852–865. 72. Ala-Kulju K, Jarvinen A, Maamies T, et al. Late aneurysms after patch
52. Goldfarb JW, Holland AE, Heijstraten FM, et al. Cardiacsynchronized aortoplasty for coarctation of the aorta in adults. Thorac Cardiovasc Surg.
gadolinium-enhanced MR angiography: preliminary experience for the 1983;31(5):301–306.
evaluation of the thoracic aorta. Magn Reson Imaging. 73. Hehrlein FW, Mulch J, Rautenburg HW, et al. Incidence and pathogenesis
2006;24(3):241–248. of late aneurysms after patch graft aortoplasty for coarctation. J Thorac
53. Tatli S, Lipton MJ, Davison BD, et al. From the RSNA refresher courses: Cardiovasc Surg. 1986;92(2):226–230.
MR imaging of aortic and peripheral vascular disease. Radiographics. 74. von Kodolitsch Y, Aydin MA, Koschyk DH, et al. Predictors of aneurysmal
2003;23(Spec No):S59–S78. formation after surgical correction of aortic coarctation. J Am Coll
54. Yoshioka K, Niinuma H, Kawakami T, et al. Three-dimensional Cardiol. 2002;39(4):617–624.
demonstration of the artery of Adamkiewicz with contrast-enhanced 75. Varma C, Benson LN, Butany J, et al. Aortic dissection after stent
magnetic resonance angiography. Ann Thorac Surg. 2005;79(5):1785. dilatation for coarctation of the aorta: a case report and literature review.
55. Yamada N, Takamiya M, Kuribayashi S, et al. MRA of the Adamkiewicz Catheter Cardiovasc Interv. 2003;59(4):528–535.
artery: a preoperative study for thoracic aortic aneurysm. J Comput Assist 76. Bell RE, Taylor PR, Aukett M, et al. Endoluminal repair of aneurysms
Tomogr. 2000;24(3):362–368. associated with coarctation. Ann Thorac Surg. 2003;75(2):530–533.
56. Richens D, Kotidis K, Neale M, et al. Rupture of the aorta following road 77. Ince H, Petzsch M, Rehders T, et al. Percutaneous endovascular repair of
traffic accidents in the United Kingdom 1992–1999: the results of the aneurysm after previous coarctation surgery. Circulation.
co-operative crash injury study. Eur J Cardiothorac Surg. 2003;23(2): 2003;108(24):2967–2970.
143–148. 78. Nastri MV, Baptista LP, Baroni RH, et al. Gadolinium-enhanced
57. Hunt JP, Baker CC, Lentz CW, et al. Thoracic aorta injuries: management three-dimensional MR angiography of Takayasu arteritis. Radiographics.
and outcome of 144 patients. J Trauma. 1996;40(4):547–555, discussion 2004;24(3):773–786.
555–556. 79. Choe YH, Kim DK, Koh EM, et al. Takayasu arteritis: diagnosis with MR
58. Kodali S, Jamieson WR, Leia-Stephens M, et al. Traumatic rupture of the imaging and MR angiography in acute and chronic active stages. J Magn
thoracic aorta: a 20-year review: 1969–1989. Circulation. 1991;84(suppl Reson Imaging. 1999;10(5):751–757.
5):III40–III46. 80. Meller J, Grabbe E, Becker W, et al. Value of F-18 FDG hybrid camera
59. Fattori R, Celletti F, Descovich B, et al. Evolution of post-traumatic aortic PET and MRI in early Takayasu aortitis. Eur Radiol. 2003;13(2):
aneurysm in the subacute phase: magnetic resonance imaging follow-up 400–405.
as a support of the surgical timing. Eur J Cardiothorac Surg. 81. Buecker A, Spuentrup E, Grabitz R, et al. Real-time-MR guidance for
1998;13(5):582–586, discussion 586–587. placement of a self-made fully MR-compatible atrial septal occluder: in
60. Rousseau H, Soula P, Perreault P, et al. Delayed treatment of traumatic vitro test. Rofo. 2002;174(3):283–285.
rupture of the thoracic aorta with endoluminal covered stent. Circulation. 82. Buecker A, Adam GB, Neuerburg JM, et al. Simultaneous real-time
1999;99(4):498–504. visualization of the catheter tip and vascular anatomy for MR-guided
61. Scheinert D, Krankenberg H, Schmidt A, et al. Endoluminal stent-graft PTA of iliac arteries in an animal model. J Magn Reson Imaging.
placement for acute rupture of the descending thoracic aorta. Eur Heart 2002;16(2):201–208.
J. 2004;25(8):694–700. 83. Raval AN, Karmarkar PV, Guttman MA, et al. Real-time magnetic
62. Demers P, Miller C, Scott Mitchell R, et al. Chronic traumatic aneurysms resonance imaging-guided endovascular recanalization of chronic total
of the descending thoracic aorta: mid-term results of endovascular repair arterial occlusion in a swine model. Circulation. 2006;113(8):1101–
using first and second-generation stent-grafts. Eur J Cardiothorac Surg. 1107.
2004;25(3):394–400. 84. Raman VK, Karmarkar PV, Guttman MA, et al. Real-time magnetic
63. Godart F, Labrot G, Devos P, et al. Coarctation of the aorta: comparison resonance-guided endovascular repair of experimental abdominal aortic
of aortic dimensions between conventional MR imaging, 3D MR aneurysm in swine. J Am Coll Cardiol. 2005;45(12):2069–2077.
angiography, and conventional angiography. Eur Radiol. 85. Eggebrecht H, Kuhl H, Kaiser GM, et al. Feasibility of real-time magnetic
2002;12(8):2034–2039. resonance-guided stent-graft placement in a swine model of descending
64. Simpson IA, Chung KJ, Glass RF, et al. Cine magnetic resonance imaging aortic dissection. Eur Heart J. 2006;27(5):613–620.
for evaluation of anatomy and flow relations in infants and children with 86. Eggebrecht H, Zenge M, Ladd ME, et al. In vitro evaluation of current
coarctation of the aorta. Circulation. 1988;78(1):142–148. thoracic aortic stent-grafts for real-time MR-guided placement.
65. Weber OM, Higgins CB. MR evaluation of cardiovascular physiology in J Endovasc Ther. 2006;13(1):62–71.
congenital heart disease: flow and function. J Cardiovasc Magn Reson. 87. Ladd ME, Quick HH, Debatin JF. Interventional MRA and intravascular
2006;8(4):607–617. imaging. J Magn Reson Imaging. 2000;12(4):534–546.
44 
Cardiovascular Magnetic Resonance
Angiography: Carotids, Aorta, and
Peripheral Vessels
Harrie van den Bosch, Jos J.M. Westenberg, and Albert de Roos

Magnetic resonance angiography (MRA) is an important imaging chelates is their chemical stability, which depends on the chemical
modality for the diagnosis, clinical workup, and treatment planning structure and ionicity of the complex.2 A stable chelate has less ten-
in patients suspected of a wide range of vascular pathology. The aim dency to release free Gd ions in the human body. Gd chelates can be
of MRA is to visualize the arterial and/or venous system by creating distinguished in two structural categories: macrocyclic and linear Gd
high contrast between the blood flow and its surrounding stationary chelates. In macrocyclic structures, the Gd chelate is more stable and
tissue. To fulfill this aim, several techniques may be used in clinical therefore the chance of free Gd ions in the body is reduced. Breaking of
practice. Depending on its demands and the conditions required to the bond between Gd and the chelate (transmetallation) is more likely
optimally visualize specific vessels of interest, imaging techniques are to occur with linear agents than with macrocyclic agents. However,
chosen. In the first part of this chapter, techniques will be addressed even though the macrocyclic agents are more resistant to transmetalla-
that are currently available and applied for MRA. Contrast-enhanced tion, development of NSF is described in patients with advanced renal
MRA (CE-MRA) is probably the most widely used MRA technique. failure (effective glomerular infiltration rate <20 mL/min/1.74 m2,
CE-MRA is applied either by fast imaging of the first pass of an intra- acute deterioration of renal function or on dialysis) before Gd con-
venously injected bolus of a gadolinium (Gd) chelate agent or by a trast administrations.3 Most reported cases of NSF fibrosis are linked
prolonged imaging approach with a blood-pool contrast agent. The with linear Gd-chelates. Awareness of this association has given rise
latter approach is applied when imaging requires a time window that to effective screening of patient with possible renal failure using the
surpasses the first arterial pass. Because of the reported adverse events, Choyke questionnaire,4 use of more stable Gd-chelates, and inter-
such as nephrogenic systemic fibrosis (NSF) (see Chapter 3), accumula- national recommendations for the use of Gd-based contrast agents in
tion of Gd in the brain, kidneys, and bone tissue associated with Gd- daily practice.5–7 Somewhat concerning are anecdotal reports of NSF-
based contrast agents, an increasing interest in non–CE-MRA techniques like symptoms following Gd administration but in the absence of renal
is currently observed. Black-blood and bright-blood non–CE-MRA dysfunction.7a
techniques will be discussed. Finally, flow imaging by time-resolved Gd-chelates create intravascular signal by shortening the T1 relax-
three-dimensional (3D) phase contrast (i.e., four-dimensional [4D] flow ation time of blood in proportion to the concentration of contrast
magnetic resonance imaging [MRI]), with the aid of newly developed material. At 1.5 T, T1 of blood is 1200 ms. The T1 relaxation time is
visualization tools, is a relatively new technique that may be used to shortened by Gd according to Eq. 44.18:
visualize complex flow structures and add quantitative hemodynamic
information. 1 T1 = 1 1200 ms + R1[Gd] Eq. 44.1
In the second part of this chapter, we will address the anatomical
regions imaged by MRA and discuss the state of the art. Special focus where R1 = T1 relaxivity of gadolinium, and [Gd] = gadolinium con-
will be on the carotid arteries, thoracic and abdominal aorta, renal centration in blood.
arteries, mesenteric artery, and the peripheral arteries. Gd CE-MRA is insensitive to blood flow, in contrast to non-CE-
MRA techniques as time-of-flight (TOF) and phase-contrast (PCA)
CONTRAST-ENHANCED MAGNETIC RESONANCE MRA. Consequently, in CE-MRA, image quality is not degraded by
flow disturbances.
ANGIOGRAPHY: TECHNICAL APPROACH Gd contrast agents can be classified depending on their distribution
In CE-MRA, vessels in the field of view of interest are imaged during in tissue after intravenous administration, that is, extracellular or intra-
the arterial first pass of intravenously injected paramagnetic contrast vascular. Most of the Gd chelates used in clinical routine are extracellular
material.1 These contrast agents have a short T1 relaxation time and agents. After intravascular administration they diffuse rapidly through
will produce high signal on T1-weighted images. In clinical practice, the capillary walls into the extravascular space. In CE-MRA, arteries
Gd-bound chelates, for example diethylenetriamine pentaacetic acid are preferably imaged during the initial passage of contrast. Timing of
(DTPA), are used as the paramagnetic contrast agents. The Gd ion is a starting the image acquisition is essential to reduce signal from Gd
rare-earth element, and toxic to humans when unbound. Therefore in diffused to the extracellular space and to overcome venous enhance-
MRA, Gd contrast is based on chelates to control the distribution of ment, which can lead to artifacts and image degradation.
Gd within the body and to overcome toxicity while maintaining their The application of CE-MRA was improved by the introduction of
contrast enhancement capacity. A very important property of these Gd-chelates that have the capability to bind to larger molecules in the

518
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 519

blood such as albumin and thus remain relatively intravascular. These filling can be used in the available 3D pulse sequences. It is important
contrast agents (e.g., gadobenate dimeglumine) provide a prolonged to adjust bolus timing to the order of k-space filling used.
imaging time window because of a decreased decay time of contrast In linear ordering, lines of k-space can be acquired in any order
in blood. The use of such agents enables imaging beyond the initial (high k-space lines first, low k-space lines first, or at random). Linear
passage of contrast bolus, which may be a benefit when acquisition ordering can be used in a CE-MRA protocol when precise timing of
needs to be gated to cardiac or respiratory motion, or when high reso- the contrast bolus is difficult or arrival time may be prolonged.
lution is required in small vessels such as the coronary arteries.9 When For most CE-MRA examinations, elliptical-centric ordering is the
compared with an extravascular Gd-chelate, blood pool agents show preferred k-space sampling method used. Typically, the center of k-space
an improved conspicuity of small vessels.10 Further development of is acquired first and the periphery later (e.g., elliptic-centric Contrast-
macromolecular blood pool agents in the near future may play an ENhanced Timing Robust Angiography [CENTRA],15 Differential Rate
important role in CE-MRA. K-space Sampling [DRKS], or PEak Arterial K-Space filling [PEAKS]).
To determine the delay between the start of the venous contrast
Three-Dimensional Contrast-Enhanced Magnetic injection and the start of the acquisition, either a small test bolus or
Resonance Angiography Pulse Sequences fluoroscopic real-time imaging can be used. The timing method by
The CMR sequence used in CE-MRA needs to fulfill the following: using a test bolus (1 to 2 mL) is robust and easy to perform. However,
T1-weighting is required, as well as the obtaining of a large 3D volume it will lengthen the procedure by a few minutes and requires an addi-
with sufficient spatial resolution, preferably fast within the first pass of tional administration of contrast. In fluoroscopic real-time imaging,
contrast and within a breath-hold to suppress respiratory motion. the inflow of contrast is imaged in the field of view with the vessels of
Therefore fast T1-weighted 3D spoiled gradient-echo (GRE) sequences interest. At the moment of contrast arrival, the acquisition is started
are used for image acquisition. In clinical routine, 3D CE-MRA pref- automatically; however, the short delay of a few seconds, which is needed
erentially is performed on a 1.5 T or 3 T system with strong gradient to switch from the fluoroscopic real-time imaging to the actual start
systems to reduce scan time, such that 3D datasets can be acquired of CE-MRA acquisition, can be a disadvantage.
during breath-holding. Typically, a short repetition time (TR) and echo
time (TE) is used, 3 to 8 ms and 1 to 3 ms, respectively. Flip angle is CONTRAST-ENHANCED VERSUS
20 to 40 degrees to achieve optimized T1 weighting. NON–CONTRAST-ENHANCED MAGNETIC
Furthermore, subsampling imaging techniques such as parallel
imaging are implemented to reduce total scan time.11 Advanced k-space
RESONANCE ANGIOGRAPHY
sampling techniques (spiral, keyhole, echo planar imaging [EPI]) may The association between CE-MRA and NSF16,17 in patients with severe
also be used to speed up acquisition. renal dysfunction and linear Gd-chelates5 has been appreciated for over
At postprocessing, the 3D nature of the dataset allows viewing of a decade.
the data from any desired angle, that is by creating multiplanar reformats Moreover, Gd accumulation in tissue in patients without renal
(MPR) or rotational maximum intensity projection (MIP). This results impairment has been reported in several studies. Kanda et al. reported
in multiple images in various anatomic orientations. Thin (i.e., <3 mm residual Gd concentrations in the brain, particularly in the dentate
thick) slices are required to provide a useful evaluation of these images. nucleus and globus pallidus, of patients without severe renal dysfunc-
In some situations, it may be useful to use thicker slices (e.g., 10 mm) tion18 and foci of hyperintensity on unenhanced T1-weighted magnetic
to reduce the number of slices and allow faster scanning. Although the resonance (MR) images associated with previous administration of
ability to rotate the MIP is lost with such thick slices, scan times can linear Gd-chelates, and which may be associated with the total number
be as short as 1 second. of previous Gd-based contrast material administrations.19 Deposition
of Gd has also been demonstrated in bone, skin, and liver.20,21 Currently,
Bolus Timing previous administration of macrocyclic Gd-chelates shows no associa-
Precise bolus timing is essential for first-pass CE-MRA. Arterial imaging tion with Gd accumulation in tissue22,23; however, these macrocyclic
is performed in the time window between arterial and venous enhance- contrast chelates have been in use in clinical practice for a shorter time
ment and this time window depends on the rate of contrast agent than the linear chelates. Nevertheless, the application of CE-MRA and
injection, and will be patient specific.12 The transit time of contrast the amount of administered Gd contrast is of clinical importance, espe-
from the injection site (usually in the veins in the arm) to the field of cially in patients with impaired renal function, and issues of long-term
view with the vessels of interest depends on several factors, for example, retention may be of particular concern in children/young adults. Aware-
injection rate, injection site, heart rate, and stroke volume. ness of NSF-related incidents associated with CE-MRA and reports on
The Gd contrast concentration in the arterial blood is proportional accumulation of Gd in various tissues have also led to a renewed interest
to the injection rate and inversely proportional to the cardiac output: in non–CE-MRA. Improvements in CMR hardware and software, includ-
ing the widespread availability of parallel imaging have helped to reduce
[Gd]arterial = injection rate (mol s) cardiac output (L s) Eq. 44.2 acquisition times and have made some non–CE-MRA methods clinically
practical. Non–CE-MRA methods may be classified into three categories:
T1 for blood is related to the injection rate and cardiac output by black-blood, bright-blood, and 4D flow visualization.
combining Eq. 44.1 and Eq. 44.2.13
Maximum arterial signal intensity is achieved when the start of the Black-Blood Imaging
acquisition and the contrast infusion are synchronized such that peak Black-blood imaging of blood vessels uses double-inversion recovery
arterial Gd concentration coincides with the acquisition of the central to null the signal of flowing blood.24 This technique is flow-sensitive.
portion of k-space.14 The center of k-space contributes most to overall Two 180 degree inversion recovery prepulses are applied to null the
image contrast, whereas the periphery of k-space provides image infor- signal of flowing blood: the first prepulse is nonslice selective and inverts
mation of details and contributes to spatial resolution. Therefore for the longitudinal magnetization vector in the entire body, whereas the
improved arterial/venous differentiation, central k-lines have to be second inversion prepulse is slice selective and inverts the magnetization
sampled before venous return. In CE-MRA, different types of k-space in the imaging slice back to its original orientation. Signal of blood
520 SECTION V Vascular/Pericardium

entering the imaging slice after the second prepulse has undergone flowing blood) will accumulate phase in the transverse magnetiza-
reinversion and will appear dark in the images. Electrocardiogram tion. This phase shift is proportional to its velocity and the gradi-
(ECG)-gated partial Fourier fast spin echo (FSE) is the sequence of ent strength.30 In addition to such a velocity-sensitive acquisition, a
choice,25,26 using a slice thickness of 6 to 8 mm stacked in the transverse velocity-compensation acquisition is defined. Such acquisition uses a
plane of the chest or in a double-oblique sagittal view of the aorta (i.e., bipolar gradient with equal positive and negative effect to achieve no
the candy-cane view). Single-shot FSE (SSFSE) sequences are much difference in net phase shift between spins moving with uniform veloc-
faster than basic FSE sequences and allow for acquisition of the entire ity. PCA uses the subtraction of both velocity-sensitive and velocity-
imaging stack in a single breath-hold. Fat saturation is recommended compensation acquisitions to visualize moving spins. The velocity
to increase the conspicuity of aortic wall hematoma.27 Despite being can be directly quantified from the resultant phase images. A priori
an established technique, black-blood imaging is prone to artifacts to the acquisition the velocity sensitivity needs to be determined by
because inadequate nulling of blood signal may occur by slow flow, or choosing the Venc (velocity-encoding range), that is, the outer limit of
when in-plane blood flow is present. the velocity range that is encoded by the phase shift of +180 degrees
and −180 degrees.
Bright-Blood Imaging: Time of Flight TOF and PCA, two conventional bright-blood non–CE-MRA tech-
TOF methods depend on the inflow enhancement of flowing blood niques, were highly time consuming and moreover, suffered from artifacts
and the relative saturation of longitudinal magnetization of stationary caused by turbulence and in-plane saturation. In a metaanalysis,31 the
tissue to produce high contrast. The longitudinal magnetization from sensitivity and specificity for the detection of significant stenosis (>50%
the background tissue is suppressed by multiple radiofrequency (RF) luminal reduction) for 2D TOF ranged from 64% to 100% and 68%
excitations, such that the magnetic spins associated with background to 96%, compared with 92% to 100% and 91% to 99%, respectively,
tissue do not have sufficient time to fully regain their longitudinal for CE-MRA. Such limitations resulted in declining interest for TOF
magnetization. Saturation of background signal will reach a steady state and PCA in favor of CE-MRA. However, in association with the reported
determined by the flip angle, repetition time, and the T1 of the back- adverse events from Gd usage, an increase in interest in new bright-
ground tissue. This saturation will make stationary tissue appear dark blood non–CE-MRA techniques is occurring.
in TOF images. The bright signal intensity of flowing blood is the
consequence of the continuous inflow of fresh, unsaturated blood into Bright-Blood Imaging: Steady-State Free Precession
the imaging slice, which produces more signal than the surrounding Steady-state free precession (SSFP) produces high signal intensity for
stationary tissue because the latter was repeatedly exposed to RF pulses.28 blood, because of the inherently T2/T1-weighted image contrast, which
This effect is known as flow-related enhancement29 and can be maxi- is high for all fluids, independent from inflow effects. ECG-gated cine
mized by using sequences with a short TR. Ideally, the imaging slice acquisitions with balanced SSFP (bSSFP) are used to visualize the anatomy
should be thin and oriented perpendicular to the direction of flow. of thoracic aorta with breath-hold sequences. Aneurysms, coarctations,
ECG gating can be used to optimize synchronization of acquisition to stenoses, and dissection flaps can be visualized using this approach.
the blood flow to the cardiac cycle and consequently to the signal present The aortic root is best evaluated in multiple planes, including long-axis
in the artery of interest. planes of the left ventricular outflow tract (LVOT). The remainder of
Two-dimensional (2D) and 3D TOF techniques are in use. Two- the thoracic aorta is best evaluated with images in the axial plane of
dimensional TOF with consecutive slices stacked together can produce the chest and in the double-oblique sagittal “candy-cane” view. bSSFP
excellent contrast in the vessels when the imaging slices are planned sequences are susceptible to magnetic field inhomogeneity, which are
perpendicular to the vessels of interest. When in-plane flow occurs, the particularly present at high field strength. Off-resonance artifacts may
longitudinal magnetization of blood will become saturated and signal result in nondiagnostic images. Dedicated shimming algorithms may
is progressively lost. Additionally, venous signal may be suppressed with aid in minimizing these off-resonance artifacts.
presaturation slabs, positioned parallel and next to the imaging slices, to
saturate longitudinal magnetization of blood flowing into the imaging Bright-Blood Imaging: Dixon
slice from the opposite side of arterial blood flow. Resolution of 2D Another relatively new bright-blood approach relies on an old CMR
TOF is often limited by the slice thickness used. However, thin slice technique based on the chemical shift between both water and fat.
acquisitions are possible, but acquisition time will increase with the Dixon’s water–fat separation32 uses the phase shift from the water–fat
number of slices required to cover the vasculature and signal-to-noise resonance frequency and, by using carefully chosen multiple echo times,
ratio (SNR) will decrease with decreasing slice thickness. excellent separation between water and fat is achieved in four distinct
Three-dimensional TOF may be performed with isotropic voxel images: separate water and fat images as well as images with signal from
sizes of 1 mm3. High resolution is essential to visualize small branch both tissues in and out of phase. The Dixon technique may be applied
vessels, stenosis, or to provide a clear delineation of tortuous vessels. for noncontrast MRA purposes and its application may be promising
Three-dimensional TOF has to deal with saturation effects of arterial for imaging of the coronaries as well as the thoracic aorta.33
blood flow, especially slow flow. On the other hand, the SNR will be
increased by using 3D imaging. Decreasing the flip angle may reduce Four-Dimensional Flow Visualization
excessive saturation effects. Phase-contrast imaging enables quantitation of blood flow velocity in
the direction of a magnetic field gradient. In addition to conventional
Bright-Blood Imaging: Phase-Contrast Angiography through-plane one-directional velocity-encoding, advanced 3D time-
In PCA, the contrast between flowing blood and the surrounding resolved imaging is possible with velocity-encoding in all three direc-
stationary tissue is generated by the flow-induced phase shift in the tions, providing the temporal distribution of a velocity vector field,
transverse magnetization of the moving blood. As the magnetic field representing the flow of blood inside a 3D volume (i.e., so-called 4D
strength and the precession frequency of spins are linearly related, flow data).34 Visualization tools such as streamline and pathline visual-
the precession frequency will increase when spins move along a posi- ization have been developed to display this data and to segment blood
tive gradient in the magnetic field. Compared with spins that remain vessels of interest from their surrounding tissue. The usability of this
fixed in position (i.e., in stationary tissue), moving spins (i.e., in technique has been shown in the aorta, pulmonary arteries, carotid
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 521

artery, and abdominal circulation as well as the circulation in the brain. first-stroke patients and 185,000 patients with recurrent stroke. Stroke
Although this technique is still a research application, a lot of effort can lead to significant disability in nonfatal cases and accounts for
has been made to translate this technique toward clinical use.35 The approximately 1 of every 20 deaths in the United States.
additional hemodynamic information (e.g., wall shear stress, stenotic Carotid atherosclerosis is a significant factor for risk stratification
pressure drop, pulse wave velocity, viscous energy dissipation) which and is associated with more than a doubling in the stroke risk after a
becomes available with 4D flow CMR in combination with the excel- transient ischemic attack (TIA).40 Therefore accurate evaluation of the
lent 3D visualization possibilities of complex vascular structures and extracranial carotid artery is essential.
pathologies, may eventually push this modality to be a clinically viable For evaluating the severity of carotid stenosis, x-ray digital subtrac-
application. tion angiography (DSA) remains the standard of reference. This imaging
approach has certain limitations, including invasiveness and the use of
ARTIFACTS IN CONTRAST-ENHANCED MAGNETIC ionizing radiation and iodinated contrast. Moreover, DSA involves a
small risk of stroke, estimated at 0.5% to 4%.41
RESONANCE ANGIOGRAPHY Therefore, today, noninvasive imaging techniques play an important
In CE-MRA, images with maximum arterial signal intensity are obtained role in the routine assessment of patients with suspected carotid artery.
when the acquisition of the central portion of k-space coincides with Noninvasive techniques such as duplex ultrasound (DUS), CE-MRA and
peak arterial Gd concentration. Timing of bolus arrival and imaging computed tomography angiography (CTA) are widely available and have
within the time window of arterial enhancement is critical to minimize largely replaced invasive x-ray arteriography in daily clinical routine.
artifacts. Starting the acquisition too early, when the contrast bolus is still When compared with DSA, noninvasive imaging techniques gener-
arriving in the vessels of interest, can cause ringing or so-called Maki ally are less accurate for low-degree carotid artery stenoses. Metaanalysis
artifacts.14 Starting the acquisition too late will lead to imaging outside showed sensitivity and specificity for DUS of 36% and 91%, respectively,
the time window of arterial enhancement, causing venous enhance- 50% to 69% for stenosis degree of the internal carotid artery, 77% and
ment hampering the evaluation of arterial vasculature. Such artifacts 97%, respectively, for CE-MRA, and 67% and 79%, respectively, for
may result in degradation of image quality and misclassified diagnosis. CTA.42 Noninvasive techniques revealed higher sensitivity and specificity
Artifacts are most evident when the center of k-space is acquired for high grade stenosis (70%–99%), DUS 89% and 84%, CE-MRA 94%
before the peak of intravascular Gd arrival.14 The Maki artifact, for and 93%, and CTA 77% and 95%, respectively.
example, results in a central dark line in the vessel, from which a so- DUS is widely available, easy accessible, cost effective and often used
called pseudodissection can appear, when the contrast bolus arrives in as a first-line investigation in standardized protocols assessing carotid
the vessel of interest after sampling of the central k-lines. Overestimation artery disease.43 In experienced hands it is an accurate test; however, it
of the length of a stenosis can occur due to signal loss because of has higher interobserver variability and lower sensitivity and specificity
turbulence and in-plane saturation. Generally, this latter artifact is not when compared with CE-MRA and CTA. Guidelines of the American
present in CE-MRA; however, signal loss can occur in the stenosis caused Heart Association (AHA) and the American College of Cardiology
by high velocity rates, especially in combination with low contrast mate- Foundation (ACCF) recommend evaluation of the carotid artery by
rial concentration.36 MRA or CTA, when DUS provides equivocal or nondiagnostic results.44
Erroneously, an occlusion can be suggested in tortuous arteries when When DUS detects a significant stenosis (>70%) it is useful to evaluate
the vessel of interest is not completely covered inside the field of view. stenosis severity by performing MRA or CTA. For assessing carotid
Parallel to vessels with very high signal intensity, ghosts (i.e., phase artery stenosis, CE-MRA shows a comparable accuracy.
artifact) can be seen.37 However, CTA has important limitations. It employs potentially
Movement of the patient between the acquisition of the mask and harmful ionizing radiation and the administration of larger volume of
the CE-MRA may lead to subtraction artifacts, leading to image deg- iodinated contrast with possible toxicity and adverse reaction. Moreover,
radation. Signal loss caused by susceptibility can be seen when clips, plaque calcification may hamper stenosis gradation. Therefore it is
metallic stents or prostheses are present. becoming common practice to combine DUS as first-line screening
In non–CE-MRA, artifacts may also result in image degradation or modality followed by CE-MRA for gradation of carotid stenosis.
even lead to misdiagnosis. Poststenotic signal loss due to turbulence or CE-MRA is the MRA technique of choice to evaluate carotid artery
in-plane saturation in phase-contrast imaging may lead to an overes- disease (Fig. 44.1). Typically, 3D CE-MRA data are acquired in the
timation of a stenosis. When the a priori set velocity encoding (Venc) coronal plane. Bolus timing is essential, because venous return in the
sensitivity is set too low, aliasing will then occur in the velocity images
and when uncorrected will result in a decrease in signal in the phase-
contrast magnitude images.
On the other hand, slow flow in time-of-flight CMR imaging may
lead to similar misdiagnosis when vascular signal decreases. In black-
blood imaging, in-plane flow may lead to vascular enhancement and
inadequate signal suppression when the imaging slice is too thick.
bSSFP is very susceptible to off-resonance artifacts, especially at
higher field strength and nonoptimal shimming, which may degrade
image quality to the point of nonevaluable for clinical diagnosis.

MAGNETIC RESONANCE ANGIOGRAPHY OF


EXTRACRANIAL CAROTID ARTERIES
Atherosclerosis of the extracranial carotid arteries is an important cause FIG. 44.1  Neck contrast-enhanced magnetic resonance angiography
of stroke.38 Worldwide stroke is a major health concern, with nearly demonstrating a severe bilateral stenoses of the proximal internal carotid
800,000 strokes in the United States every year,39 including ~610,000 arteries (arrows).
522 SECTION V Vascular/Pericardium

veins of the neck is quite fast. Spatial resolution of 0.71 mm3 at 1.5 T first-line imaging modality, but MRA and CTA are widely used. The
and 0.54 mm3 at 3 T are recommended.45 Generally, single dose abdominal aorta is most typically imaged by ultrasound, MRA, and
(0.1 mmol/kg) of a Gd-based contrast agent is sufficient to achieve CTA,57 whereas TTE is the most frequently used imaging modality for
excellent image quality and resolution at 1.5 T. Several studies showed evaluating the aortic root and proximal ascending aorta.
that dose reduction to half dose (0.05 mmol/kg) is feasible at 3 T without In a standardized TTE examination the thoracic aorta is routinely
compromising SNR and spatial resolution.46,47 assessed.58 Despite the fact that TTE is limited for assessing the complete
In several studies, time of flight MRA showed good sensitivity and thoracic aorta, it still permits adequate assessment of several aortic
specificity in detecting stenosis of the carotid bifurcation.48,49 However, segments, especially when monitoring aortic root diameters.59 Moreover,
CE-MRA offers high-resolution imaging of larger field of view in shorter TTE can be used as a screening tool for proximal ascending aorta dila-
acquisition times50,51 and therefore, has largely replaced time of flight tation, but is not sensitive for detection of aortic dissection, particularly
MRA in clinical practice. beyond the proximal ascending aorta; however TTE is quite useful to
Development of new non–CE-MRA sequences show promising assess for associated complications of type A dissections (e.g., aortic
results and could have the potential to become an alternative imaging valve disruption/regurgitation, hemopericardium).
approach in evaluating carotid artery disease. Zhoa et al.52 showed that Because of the proximity of the esophagus to the thoracic aorta,
3D black-blood CMR imaging provided high agreement for stenosis TEE is often used in thoracic aorta assessment and overcomes several
measurements as compared with DSA. Three-dimensional black-blood of TTE limitations. TEE reveals high resolution images and, unlike
CMR imaging depicted longer lesion length than DSA, because of its TTE, allows gradual assessment of the thoracic aorta from the aortic
ability to define vessel wall morphology and distribution of plaque. root to the descending aorta.60,61 However, if TTE and/or TEE reveal
When compared with CE-MRA, 3D black-blood MRA shows accu- inconclusive information or abnormalities are present, another imaging
rate measurement of carotid stenosis53 and sensitivity and specificity modality is required for confirmation/further evaluation.
comparable with CTA.52 CTA is an important imaging technique in vascular diseases of the
Other vascular disease of the carotid artery can also be diagnosed thoracic and abdominal aorta. Especially in acute aortic syndrome
by CE-MRA. Fibromuscular dysplasia is a nonatherosclerotic, nonin- (i.e., aorta dissection, intramural hematoma, penetrating atheroscle-
flammatory arteriopathy with segmental involvement of small-sized rotic ulcer, and contained aortic rupture) CTA plays a key role and is
and medium-sized arteries. The etiology is unknown and the extracranial the preferred primary imaging technique because of its speed, wide
carotid, vertebral, and renal arteries are most frequently affected. The availability, and high spatial resolution. As a result, CTA has essentially
so-called string-of-beads sign of the involved arteries is a classic pattern replaced invasive DSA. Diagnostic accuracy of CTA for the detection of
seen in fibromuscular dysplasia, but also other imaging findings are dissection and traumatic injury is excellent. Sensitivity and specificity
described as aneurysm, fusiform arterial ectasia, and vascular loops. for the detection of dissection are 100%.62 Steenburg et al.63 reported
Fibromuscular dysplasia can accurately be diagnosed using CE-MRA; a sensitivity of 96%, a specificity of 99.8%, and accuracy of 99.8% for
however, a negative result on CE-MRA does not exclude fibromuscular multislice CTA as compared with DSA in patients with acute thoracic
dysplasia. Therefore DSA is still recommended in patients with high aortic injury. False positives have been described with CTA if ECG
clinical suspicion of fibromuscular dysplasia.54 gating is not performed.64
Carotid artery dissection is one of the most common causes of The role of MRA in the evaluation of acute aortic syndrome is
stroke in young patients. Dissection may occur after trauma or may limited. Monitoring acute, unstable patients in the CMR scanner is
arise spontaneously. In patients suspected for carotid artery dissection, more complicated and acquisition times are longer. However, MRA has
CE-MRA is the imaging modality of choice.55,56 Mural hematoma can several advantages over CTA, such as the previously mentioned radiation-
be visualized by performing additional axial T1 weighted images with free nature of the imaging modality in which the need for iodinated
fat suppression. Although DSA is the reference standard for evaluation contrast medium is avoided. Therefore MRA is highly suitable for evalu-
of the lumen of the carotid artery, it does not provide details of the ation of aortic disease in young patients and for follow-up studies.65
arterial wall. In suspected cases it is important to reformat acquired CE-MRA has emerged as the preferred MRA technique for evalu-
high-resolution isovolumetric voxels in axial thin slices to eventually ation of thoracic and abdominal vascular pathology independent of
depict an intimal flap. blood flow. High-resolution 3D image acquisition permits multiplanar
and MIP reconstructions in any desired plane for detailed image evalu-
ation (Fig. 44.2). Three-dimensional CE-MRA acquisition does not
AORTIC MAGNETIC RESONANCE ANGIOGRAPHY
Diseases of the thoracic and abdominal aorta can be examined by several
imaging modalities. Over the last decade, noninvasive imaging techniques
have improved significantly and therefore the need for DSA as an imaging
tool in clinical practice has decreased. With noninvasive techniques,
the risks of arterial catheterization and the use of iodinated contrast
medium can be avoided.
A wide spectrum of vascular diseases affecting the thoracic and
abdominal aorta can be evaluated by angiographic imaging, including
aortic aneurysm, suspected aortic dissection, penetrating atherosclerotic
ulcer, atherosclerotic arterial occlusive disease, genetic diseases (e.g.,
Marfan syndrome), and congenital abnormalities such as coarctation
of the aorta. For imaging the thoracic aortic lumen, current established
noninvasive techniques such as transthoracic echocardiography (TTE),
transesophageal echocardiography (TEE), MRA, and CTA are able to FIG. 44.2  Abdominal–pelvic contrast-enhanced magnetic resonance
provide additional information about the aortic wall and the relation- angiography shows a short distal aortic occlusion (arrow) and a significant
ship of vascular disease to surrounding tissue. TTE is the most common stenosis of the proximal left common iliac artery (arrowhead).
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 523

require ECG triggering and can be performed during a single breath- Additionally, ECG-triggered T1-weighted images with fat suppres-
hold of 15 to 20 seconds. sion can be acquired to analyze the aortic wall. Phase-contrast sequences
For optimizing SNR, surface phased array coils are used for imaging can be performed to quantify flow.
the thoracic and abdominal aorta, although sufficient image quality Non–CE-MRA of the thoracic aorta can also provide additional
can often be achieved with the body coil. Positioning of the surface information about the aortic wall and surrounding tissue, and there-
phased array coil in thoracic aorta imaging is important because the fore this acquisition is an important sequence within the standard
volume stack should include the proximal brachiocephalic arteries. MRA imaging protocol. Non–CE-MRA by bSSFP may serve as a
When imaging the abdominal aorta, positioning of the 3D volume is useful alternative for evaluation and follow-up of aortic disease when
essential so that the acquired volume contains the proximal course of compared with CE-MRA in patients with contraindications to Gd
major arterial braches, such as the cephalic trunk, superior mesenteric use or with poor intravenous access. bSSFP showed high diagnostic
artery, renal arteries and iliac arteries. accuracy for aortic dimensions and vascular pathology, and similar
For both the thoracic and abdominal aortic CE-MRA, patients are reader confidence, when compared with CE-MRA.68,69 For the detection
routinely imaged in supine position. of aortic disease, free breathing 3D SSFP MRA with navigator gating
The imaging protocol of the thoracic aorta consists of a survey or showed a 100% diagnostic accuracy with CE-MRA serving as refer-
localizer sequence, bSSFP cine imaging, contrast bolus timing sequence ence standard.70 Furthermore, bSSFP showed significantly higher image
(test bolus or fluoroscopic real-time imaging), 3D CE-MRA sequence quality of the aortic root, whereas non–ECG-gated CE-MRA images
precontrast and postcontrast and, depending on vascular disease, may suffer motion artifacts in this region hindering diagnostic image
T1-weighted postcontrast sequence to rule out, for example, aortitis. evaluation.
Single-shot fast spin echo or bSSFP acquired during breath-holding For CE-MRA, postprocessing of the acquired 3D dataset is essential.
can serve as a localizer sequence in three directions. For optimal plan- On MPR and MIP reconstructions aortic vascular pathology can be
ning and positioning of the 3D CE-MRA imaging volume, it is essential assessed in relation to the origin of branching vessels. For serial follow-
that the survey scan is performed in the same breath-holding position up examinations in patients with aortic dilatation, consistency and
as used for the 3D CE-MRA acquisition. Preferably, this breath-holding accuracy of aortic diameter measurements is critical, because increase
is end-expiration because it is more reproducible when compared with in diameter may have important impact on risk stratification and on
end-inspiration.66 bSSFP images are acquired with ECG triggering and planning of surgical intervention.71,72 The maximal diameter should be
during breath-holding. Preferably, two directions (axial and parasagittal measured perpendicular to the axis of flow, double oblique or so-called
planes) are acquired. bSSFP images provide additional information centerline reconstruction.73 Double oblique measurements of aortic
about the vessel wall and surrounding tissue. diameter provided better agreement with the reference standard of
The precontrast 3D scan is acquired to verify proper positioning of planimetry than axial measurements and, therefore, may have potentially
the 3D volume and to exclude possible aliasing artifacts. For 3D CE-MRA, important impact on surgical decision-making.74
0.1 to 0.2 mmol/kg body weight Gd contrast is administered. The 3D An aneurysm of the thoracic aorta is often an incidental finding
dataset can be obtained in coronal, sagittal, or parasagittal plane. Cover- during TTE for another indication or on chest x-ray in asymptomatic
ing the thoracic aorta in a coronal plane requires a larger volume and patients. Aortic aneurysm may lead to aortic rupture or aortic dissec-
consequently, longer breath-holding maintaining high spatial resolution. tion. These acute events may be rapidly fatal in a large proportion of
Therefore in clinical practice, sagittal or parasagittal plane is favored. patients75; therefore serial follow-up examinations in this patient popu-
Typically, the field of view ranges from 30 to 40 cm. lation are vital. CE-MRA is a robust, accurate, and reproducible imaging
Usually, the abdominal aorta is imaged in the coronal plane and a modality for evaluation of the entire aorta and is therefore favorable
field of view of 35 and 40 cm. For both thoracic and abdominal CE-MRA, for long-term monitoring examinations because it does not employ
spatial resolution of at least 1 to 1.5 mm is recommended.67 ionizing radiation or iodinated contrast (Fig. 44.3).
In CE-MRA of the thoracic and abdominal aorta, optimally two Reports showed a high accuracy for the detection of thoracic and
postcontrast datasets are obtained. Large aneurysms or false lumen in abdominal aneurysms with a sensitivity of 100% and specificity of
aortic dissection may have slow flow and therefore require a longer 100%, respectively, when compared with surgery or DSA.76,77
time interval between contrast injection and 3D image acquisition to Atherosclerosis more often causes aneurysm formation in the descend-
become fully enhanced. As a consequence, they may be only fully estab- ing and abdominal aorta, whereas dilatation of the ascending aorta more
lished during the second 3D volume acquisition. Moreover, distinction frequently occurs in aortic valvular disease, as in aortic valvular regurgita-
between slow flow and therefore potential nonfilling with contrast of tion and in patients with bicuspid valves. Other causes of aortic aneurysm
the lumen can be better distinguished from thrombus. are connective tissue disease (e.g., Marfan syndrome, Ehlers–Danlos,

A B
FIG. 44.3.  (A) Axial, bright-blood balanced steady-state free-precession axial image shows a fusiform aneu-
rysm of the ascending aorta (*). (B) Contrast-enhanced magnetic resonance angiography (sagittal maximum-
intensity-projection) of the same patient.
524 SECTION V Vascular/Pericardium

and Loeys–Dietz syndrome), Turner syndrome, inflammatory disease, T1-weighted imaging can determine blood in the aortic wall and
trauma, chronic dissection or aortic surgery. Hypertension, smoking, additional T2-weighted imaging can further help to analyze the blood
hypercholesterolemia, and low high-density lipoprotein cholesterol levels compounds, characteristics, and age of intramural hemorrhage. CE-MRA
are also significantly associated with aortic dilatation.78,79 clearly reveals the extent of aortic dissection and the possible extension
Aneurysms can be classified in true and false aneurysms. A true into arterial side braches. Moreover, branching arteries originating from
aneurysm retains all three layers of the aortic wall—the intima, media, the true or false lumen can be evaluated. Both bSSFP images and CE-MRA
and adventitia. In a so-called false aneurysm, a disruption in the arterial can clearly visualize the intimal flap. However, MIP images may obscure
wall causes blood leakage and an aneurysm contained by only the the intimal flap and dissection may be missed. Therefore it remains
adventitia or surrounding tissue. The disruption may be caused by important to review both source images of the CE-MRA sequence,
trauma, iatrogenic origin (e.g., percutaneous or surgical procedures), together with bSSFP images and MPR in patients with suspicion of or
or inflammation. Risk of aneurysm rupture is much higher in a false known aortic dissection. It is not advisable to make a diagnosis based
aneurysm when compared with true aneurysms.80 solely on the projection images.
Morphologically, two types of aneurysms can be recognized: fusiform
(see Fig. 44.3) and saccular. A fusiform aneurysm is concentric and RENAL ARTERY MAGNETIC
involves dilatation of the full circumference of the aortic wall. A saccular
aneurysm is eccentric and consists of an asymmetric outpouching of
RESONANCE ANGIOGRAPHY
a part of the aortic wall. Renal artery stenosis is regarded one of the most common manifestations
Bacterial infections of the aortic wall can cause the formation of of systemic atherosclerosis. It is a progressive disease and is more seen in
mycotic aneurysms. The most common infecting organisms are Staphy- the elderly. Increased prevalence in patients with coronary artery disease
lococcus aureus and Salmonella. Mycotic aneurysms are usually saccular and peripheral artery disease are reported.86,87 Typically, stenosis of the
and are more prone to rupture. For optimal treatment early diagnosis renal artery either involves the origin of the renal artery, the proximal
is essential. Normally, patients present with signs of infection, including third, or it is caused by an aortal atherosclerotic plaque impinging on
fever or sometimes sepsis. Often bacteremia is present; however, nega- the ostium of the renal artery. Patients with renal artery stenosis often
tive blood cultures do not exclude a mycotic aneurysm.81 Therefore have bilateral renal artery stenoses.88 Luminal diameter narrowing of
imaging is crucial for diagnosis. Besides the saccular aneurysm, CE-MRA more than 50% to 70% is considered hemodynamically significant.
can demonstrate additional perivascular inflammatory changes as soft In young patients, renal arterial stenosis can be caused by fibromus-
tissue inflammation, edema, gas, and irregularity of the aortic wall cular dysplasia. Fibromuscular dysplasia is an idiopathic, nonathero-
leading to the diagnosis of mycotic aneurysm. sclerotic, segmental disease of the arterial wall leading to stenosis,
In aortic dissection, intraluminal blood penetrates into the media aneurysm, dissection and/or occlusion of small-sized to medium-sized
layer of the aortic wall through a tear in the intima, thereby dissecting arteries, especially renal and carotid arteries, and predominantly occurs
the aortic wall. The proximal ascending aorta and the descending aorta in young and female patients. When the renal arteries are involved, it
distal to the origin of the left subclavian artery are the two most common may cause secondary hypertension.89
initiating sites for the intimal tear. The bleeding between the dissected The standard of reference to determine the diagnosis of renal artery
layers of the aortic wall forms the false lumen and may propagate proxi- stenosis anatomically is catheter renal angiography, that is, DSA. Direct
mally or distally. Distention of the false lumen may narrow and compress measurement of hemodynamic characteristics is possible by involving
the true lumen. Bleeding within the medial layer of the aortic wall can transstenotic pressure measurements and if necessary, revascularization
also cause the formation of a localized intramural hematoma and facili- can be performed immediately. However, DSA has known limitations,
tate the development of an intimal tear. such as the use of ionizing radiation and iodinated contrast. Moreover,
Currently, two classification systems—DeBakey and Stanford DSA is an invasive imaging tool with possible risk of complications.
classification—are most frequently used for aortic dissection.82 The Nowadays, noninvasive imaging modalities have replaced DSA in
DeBakey classification subdivides aortic dissection in type I, II, and III. the diagnosis and workup in patients with suspected renal artery stenosis
A DeBakey type I aortic dissection involves the ascending and descending in clinical practice. Doppler ultrasonography is a frequently used imaging
thoracic aorta; in type II DeBakey, only the ascending aorta and aortic modality to evaluate renal vasculature. DUS is a widely available, non-
arch are involved; and in type III, only the descending aorta is involved. invasive and radiation-free technique that allows anatomic assessment
The Stanford classification differentiates only type A and type B aortic of renal artery stenosis and measurement of renal artery flow patterns.
dissection. Stanford type A dissection involves the ascending aorta and To characterize significant renal artery stenosis, several DUS measures
may also involve the arch and descending thoracic aorta. Stanford type B have been proposed, for example, renal-to-aortic peak systolic ratio,90,91
aortic dissections start out in the descending aorta and continue distally, and renal peak systolic velocity.92,93 Zeisbrich et al.94 showed that real-
sparing the ascending aorta and arch. If not surgically intervened upon time contrast-enhanced ultrasonography was superior to conventional
emergently, patients with acute type A aortic dissection have a very Doppler ultrasonography in microvascular perfusion changes in allografts.
high mortality of 50% within the first 48 hours83; therefore surgery is Contrast-enhanced ultrasonography can play a role as a complementary
the treatment of choice. Hemodynamically stable patients with type B imaging tool to assess renal artery stenosis and detailed qualitative and
dissection are often treated medically with close surveillance to identify quantitative information on renal microvascular perfusion. Doppler
possible progression of the dissection (an indication for mechanical ultrasonography is, however, time consuming, highly operator depen-
intervention). For patients with suspected aortic dissection, imaging of dent, and can be technically challenging in obese patients.
the thoracic and, if necessary, abdominal aorta is essential. CTA is the Modern multislice CT permits rapid high-resolution imaging with
imaging technique of choice in most patients with suspected aortic dissec- accurate first-pass contrast timing in patients with renal arterial disease.
tion in acute setting because of the availability of computed tomography High accuracy of CTA for diagnosing significant atherosclerotic renal
(CT) in the emergency department. CE-MRA is an excellent technique stenosis has been reported, with a sensitivity and specificity of 94% and
for multiple follow-up examinations, without ionizing radiation or the 93%, respectively.95 Also high sensitivity and specificity of 100% and
use of iodinated contrast. For CMR, high sensitivity and specificity, 99% respectively was described for detecting in-stent recurrent stenosis
98% for both, in the diagnosis of aortic dissection were reported.84,85 in patients following percutaneous revascularization.96 Limitations of
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 525

A B
FIG. 44.4.  (A) Abdominal three-dimensional (3D) contrast-enhanced magnetic resonance angiography (CE-
MRA) (maximum-intensity-projection) shows a short stenosis of the right proximal renal artery (arrow).
(B) Coronal 3D phase-contrast MRA shows a signal void at the origin of the hemodynamic significant renal
artery stenosis (arrow).

CTA are the use of ionizing radiation and iodinated contrast. Another a reduction of 2 mm Hg in systolic blood pressure, in patients with
disadvantage of CTA is its inability to perform physiologic assessment revascularization in combination with medical therapy when compared
of renal arterial stenosis and, moreover, severe renal artery calcification with medical therapy alone.102 Moreover, renal artery revascularization
may obscure arterial lumen narrowing and limit evaluation of stenosis. did not show significant clinical benefit concerning prevention of clini-
CE-MRA is a reliable and fast noninvasive imaging technique provid- cal events.101,102
ing accurate visualization of the renal arteries and accessory renal arter- However, metaanalysis also showed that patients with atherosclerotic
ies.97 Metaanalysis showed a sensitivity and specificity of 88% to 100%, renal artery stenosis provided better diastolic blood pressure control
respectively.98 Three-dimensional CE-MRA provides anatomic evaluation and a reduction in the mean number of antihypertensive drugs after
of renal arteries. Additionally, PCA images can be acquired, adding endovascular treatment.103,104
functional information about renal arterial flow (Fig. 44.4). In clinical Selected subgroups of patients, for example, patients with acceler-
practice this can help, for example, in the analysis of the hemodynamic ated or malignant hypertension, difficult to control blood pressure,
significance of an intermediate-grade renal artery stenosis (40%–70%), young patients with fibromuscular dysplasia, or at risk of progressive
which cannot be reliably derived from vessel diameter measurements nephropathy and end-organ failure may benefit from renal arterial
alone. In these cases, PCA can be used as an adjunct to CE-MRA for revascularization.105,106
assessing and grading the hemodynamic relevance of intermediate-
grade renal stenosis for decision on proper treatment strategies.99,100 MAGNETIC RESONANCE ANGIOGRAPHY OF THE
Typically, a CMR imaging protocol for renal artery evaluation consists
of a survey or localizer sequence, a contrast bolus timing sequence (test
MESENTERIC ARTERIES
bolus or fluoroscopic real-time imaging), a 3D CE-MRA sequence pre- Atherosclerotic vascular disease of the mesenteric arteries is the leading
contrast and postcontrast, and a PCA sequence. For optimizing signal cause of chronic mesenteric ischemia. In a population-based prevalence
and image quality a body phased-array coil should be applied. A fast study, significant mesenteric arterial disease (i.e., a stenosis of more
2D balanced-gradient echo sequence or fast spin echo sequence can be than 70% or an occlusion) has been reported in 18% of asymptomatic
used as survey sequence. The survey scan is performed in the same elderly patients.107 However, significant mesenteric arterial stenosis does
breath-holding position as used for the 3D CE-MRA acquisition. Prefer- not necessarily indicate the presence of mesenteric ischemia clinically.108
ably, this breath-holding is end expiration.66 A precontrast 3D scan is The reason for this is that the mesenteric arterial circulation contains
acquired to control proper positioning of the 3D volume and rule out an extensive collateral circulation between the celiac artery, superior
aliasing artifacts. After intravenous administration of Gd contrast mesenteric artery, and inferior mesenteric artery. Therefore involvement
(0.1–0.2 mmol/kg body weight), the 3D CE-MRA sequence is performed. of atherosclerosis in the mesenteric arteries in patients with generalized
Typically, the 3D CE-MRA images have an acquired in-plane resolution atherosclerosis infrequently leads to symptoms of chronic mesenteric
of approximately 1 mm × 1 mm and a slice thickness of 2 to 2.5 mm. ischemia. The collateral pathways of the mesenteric arterial circulation
The 3D volume is positioned in coronal plane and acquired during include the pancreatico–duodenal arcade, arc of Riolan, arc of Buhler,
breath-holding (less than 20 seconds). Following the 3D CE-MRA the arc of Barkow, and the marginal artery of Drummond. In general, in
PCA sequence is acquired with a standardized typical velocity of 35 cm/s patients with symptoms of chronic mesenteric ischemia, two of the
in the right–left direction, 35 cm/s in the anterior–posterior, and 100 cm/s three mesenteric arteries show significant stenosis or occlusion.109,110
in the feet–head direction, respectively. Postprocessing of the acquired Mostly, patients with stenosis of one of the three mesenteric arteries
3D CE-MRA and PCA data, using MPR and MIP, is essential for renal do not present chronic mesenteric ischemia symptoms.111 However,
artery evaluation and permits detailed analysis in all desired planes. In chronic mesenteric ischemia has been reported in isolated mesenteric
all patients, but especially in those with suspected renal artery stenosis, arterial stenosis.112
referred for CE-MRA, the estimated glomerular filtration rate should In patients with chronic mesenteric ischemia, mesenteric arterial
be assessed before the CE-MRA examination, to minimize the risk of stenosis or occlusion compromise the blood flow to the intestine, leading
NFS-related complications. to postprandial abdominal pain, so-called angine abdominale. Typically,
Significant renal artery stenosis may activate the renin–angiotensin– this pain starts within 30 minutes of eating and can last for several
aldosterone system and is associated with arterial hypertension, ischemic hours. This may lead to avoidance of food and significant weight loss.
nephropathy, and chronic kidney disease. The main options for treat- Patients may also present with atypical symptoms as nausea, vomiting,
ment of renal arterial stenosis include percutaneous or surgical bypass diarrhea, constipation, or colitis. In the majority of patients, diagnosis
revascularization, or medical therapy. Large, randomized trials showed of chronic mesenteric ischemia is established in an advanced stage of
no significant difference in systolic blood pressure,101 or only revealed the disease.
526 SECTION V Vascular/Pericardium

The incidence of chronic mesenteric ischemia increases with age, is be evaluated. For stenosis grading of mesenteric arteries, high sensitivity
more prevalent in women and in patients with risk factors for athero- and specificity of 100% and 95%, respectively, with DSA as the refer-
sclerotic vascular disease such as history of smoking, hypertension, and ence standard were reported.119 However, the diagnostic value of CTA
hyperlipidemia.113 Less commonly, chronic mesenteric ischemia occurs may be limited with the presence of extensive calcifications. Other known
in young patients with fibromuscular dysplasia, Takayasu vasculitis, drawbacks include the use of radiation and iodinated contrast; therefore
polyarteritis nodosa, arterial dissection, abdominal aortic aneurysm, CE-MRA is an accurate and reliable alternative noninvasive imaging
or postradiation therapy. tool for diagnosing chronic mesenteric ischemia. Three-dimensional
Clinical diagnosis of chronic mesenteric ischemia is established on CE-MRA has a high sensitivity and specificity, ranging from 92% to
clinical symptoms and consistent imaging characteristics. Identification 100% and 95% to 100%, respectively.119–121 CE-MRA has, however, a
of stenotic arterial mesenteric disease without associated clinical symp- limited role in the evaluation of distal mesenteric arteries and depicting
toms is not diagnostic of chronic mesenteric ischemia. stenosis in the inferior mesenteric artery.122,123
For diagnosing chronic mesenteric ischemia, conventional DSA Routinely, 3D CE-MRA is acquired with the patient in supine posi-
remains the reference standard. Advantageously, an endovascular thera- tion using a surface phased-array coil for optimizing SNR. As with
peutic procedure can be performed at the time of diagnosis of mesenteric other previously described 3D CE-MRA protocols, the mesenteric arteries
arterial stenosis. However, DSA is an invasive procedure with the already are evaluated with a protocol which comprises a survey or localizer
mentioned disadvantages such as the exposure to radiation, nephrotoxic sequence, a contrast bolus timing sequence (test bolus or fluoroscopic
iodinated contrast, and the invasive procedure is associated with com- real-time imaging), and a 3D CE-MRA sequence precontrast and post-
plications.114 In common clinical practice, MRA and CTA have replaced contrast. Spatial resolution and slice thickness should be tailored for
DSA as an imaging tool in the workup of patients suspected for chronic the individual patient such that the 3D volume can be imaged in one
mesenteric ischemia. single breath-hold. The same breath-holding position is used for the
In patients suspected of chronic mesenteric ischemia, Doppler ultra- survey scan and the 3D CE-MRA acquisition. The precontrast 3D scan
sound is useful for screening and is the preferred initial imaging modality. can be used to check correct positioning of the 3D volume and rule
Visualization of high-grade stenoses in the celiac artery or superior out aliasing artifacts, as was discussed previously. Three-dimensional
mesenteric artery associated with chronic mesenteric ischemia may be CE-MRA sequence is performed after intravenous administration of
technically challenging, especially because of overlying bowel gas or Gd contrast (0.1–0.2 mmol/kg body weight). The 3D volume slab is
patient’s habitus. When compared with angiography, the celiac and positioned in sagittal plane, if the primary clinical indication is to deter-
superior mesenteric artery are accurately visualized in more than 80% mine arterial disease of the mesenteric arteries, which occurs mostly
and 90%, respectively.115 The inferior mesenteric artery can rarely be proximal in the course of the mesenteric arteries. Also the coronal plane
evaluated by Doppler ultrasound because of its anatomical location.116 can be used to include the aorta and portal vein. In addition to the
Peak systolic velocity has been widely used for evaluating celiac and arterial phase, the sequence can be repeated to obtain the portal and
superior mesenteric arterial disease with a sensitivity and specificity systemic venous phase. Typically, images are required with an in-plane
for diagnosing hemodynamically significant stenosis of 92% and 96%, resolution of approximately 1 to 2 mm and a slice thickness of 1 to
and 87% and 80%, respectively.117 2 mm, resulting in a breath-hold of 10 to 20 seconds.124
Van Petersen et al.118 showed that stenosis in the celiac or superior CE-MRA should be performed in both inspiration and expiration
mesenteric artery results in an increase of flow velocities in the other when median arcuate ligament syndrome is suspected (Fig. 44.5). In
mesenteric arteries. The increase in flow was correlated with the pres- median arcuate ligament syndrome, the celiac artery is compressed by
ence of collateral vessels. This may lead to overgrading or mimicking the central tendon of the diaphragm crura. The classical finding is a
stenosis in the other mesenteric artery. Therefore Doppler ultrasound smooth narrowing of the proximal celiac artery in expiration, diminish-
should be used only as an initial screening imaging tool and in case of ing or disappearing in inspiration. With inspiration, the celiac artery
demonstrated arterial stenosis further evaluation by performing MRA moves inferiorly and more vertically alleviating the compression of the
or CTA is required. ligamentous band. By performing 3D CE-MRA in both inspiration and
CTA is considered the first-line alternative to diagnostic DSA for expiration the diagnosis can be obtained and differentiated from ath-
evaluation of chronic mesenteric ischemia.116 Especially in the emergency erosclerotic arterial disease.
situation of the suspicion of an acute mesenteric ischemia, CTA is the Non–CE-MRA such as time of flight and PCA are flow-dependent
most appropriate imaging modality. It is a fast, noninvasive imaging CMR techniques and may suffer from decreased sensitivity and specific-
technique and is widely available. Acquired 3D data sets can be post- ity because of slow flow and artifacts from turbulence overestimating
processed in varying planes and other causes for abdominal pain can stenosis. bSSFP techniques demonstrated a superior visualization of

A B C
FIG. 44.5.  A patient diagnosed with the arcuate ligament syndrome shows significant narrowing of the
celiac artery (arrow) on contrast-enhanced magnetic resonance angiography (CE-MRA) (maximum intensity
projection) (A) during expiration, (B) disappearing with inspiration (arrow), and (C) CE-MRA during inspiration
after surgical release of the median arcuate ligament (arrow).
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 527

the inferior mesenteric artery when compared with CE-MRA125 and index are widely available. These inexpensive techniques are easily repeat-
may play a role in patients with contraindications for intravenous able and commonly used for the initial screening, to analyze the presence
administration of Gd-based or iodinated contrast media. and location of atherosclerotic arterial disease of the run-off arteries.141,142
However, Doppler ultrasonography is highly operator dependent143 and
MAGNETIC RESONANCE ANGIOGRAPHY OF THE has several other drawbacks, such as hampered evaluation of peripheral
arteries in obese patients and difficulty obtaining consistent high image
PERIPHERAL ARTERIES quality in the run-off vessels distal to the popliteal artery. The ankle–
Peripheral arterial disease is a common clinical manifestation of ath- brachial index is commonly used as a first-line diagnostic imaging tool
erosclerosis and is—together with coronary artery disease and stroke— in clinical practice. A normal ankle–brachial index ranges from 1 to
one of the leading causes of atherosclerotic cardiovascular morbidity.126 1.4, and an abnormal value is defined as ≤0.9.144 Ankle–brachial index
It has been estimated that around 8.5 million people in the United values of 0.91 to 0.99 are regarded “borderline” and values >1.4 are an
States are affected by peripheral arterial disease over the age of 40 indication for noncompressible arteries. Moreover, several studies
years.127 Because of raised life expectancy, the prevalence of peripheral reported ankle–brachial index to predict all-cause mortality and car-
arterial disease increased over the last few decades by more than 20% diovascular events in cardiovascular disease.145–147 A low ankle–brachial
and has become a global problem affecting more than 200 million index is a strong indicator of the presence of peripheral arterial disease;
people worldwide,126 causing a global increase in morbidity and mortal- however, because of false-negative rates, a normal ankle–brachial index
ity associated with peripheral arterial disease.128 Prevalence rates increase does not exclude peripheral arterial disease148 and ankle–brachial index
with age, and the prevalence in men and women is similar.129 However, may underestimate the prevalence of atherosclerotic arterial disease of
in the last decades women experienced a more dramatic increase in the lower extremities when compared with CE-MRA.149
morbidity and mortality from peripheral arterial disease when compared DSA is still considered as a standard reference for the evaluation of
with men.128 Risk factors significantly associated with the incidence of stenosis in peripheral arteries. However, because of the invasive character
peripheral arterial disease include smoking, diabetes mellitus, hyperten- and the risk of possible complications, DSA is generally not used as a
sion, hypercholesterolemia, and chronic kidney disease.130 Markers of diagnostic imaging tool. Moreover, because of technical developments
inflammation and thrombosis are also associated with peripheral arterial of noninvasive imaging techniques such as CTA and MRA, DSA is
disease.131 Intermittent claudication is the classic symptom in peripheral consequently indicated only in patients with symptomatic peripheral
arterial disease, however described in only about 10% of patients with arterial disease for whom percutaneous revascularization is planned
peripheral arterial disease,132 whereby men are more likely to have classic or in patients with contraindications for performing a CTA or MRA
symptoms of claudication. The large majority of patients show a wide examination.
range of leg symptoms or are even asymptomatic. For further workup and planning of revascularization in patients
Peripheral arterial disease is associated with increased risk for cardiac with symptomatic peripheral arterial disease, CTA and MRA are widely
and cerebral events133,134 and, moreover, several reports showed an up available and used in clinical practice. CTA is an accurate technique
to 6 times higher risk for mortality from cardiovascular causes and a for evaluating stenosis severity in patients with peripheral arterial disease.
2.5 times higher overall mortality rate in patients with symptomatic Modern multislice CT scanners allow rapid acquisition of high-resolution,
peripheral arterial disease.129,135 Therefore assessment of risk factors for thin CT images. Accurate timing of the data acquisition during maximal
risk stratification in patients with peripheral arterial disease is very iodinated arterial contrast enhancement provides detailed imaging of
relevant.136 Assessment of risk factors such as hypertension, diabetes the run-off arteries. Metaanalysis estimated a sensitivity and specificity
mellitus, body mass index, levels of triglyceride and high-density lipo- for CTA of 96% and 95%, respectively. However, CTA employs poten-
protein in blood plasma samples, and ankle–brachial index is essential tially harmful radiation and requires the use of iodinated contrast
when risk factor reduction is pursued. Moreover, the usefulness of material as stated before. Moreover, arterial wall calcification may com-
noninvasive imaging for risk assessment in cardiovascular disease is promise diagnostic image quality and hamper stenosis evaluation espe-
recognized137; for example, the assessment of CMR imaging biomarkers cially in patients with diabetes mellitus and renal failure.150
such as mean MRA stenosis and pulse wave velocity showed prognostic For the detection of peripheral arterial disease, CE-MRA is superior
value for all-cause mortality and cardiac events in patients with symp- to the conventional non–CE-MRA techniques, such as 2D time of flight
tomatic peripheral arterial disease.138 or PCA. These conventional and older non–CE-MRA techniques were
Clinically, symptoms in patients with peripheral arterial disease can quite lengthy. Moreover, they suffered from artifacts caused by turbu-
be staged according the Fontaine or Rutherford classification. The Fon- lence and in-plane saturation. In a metaanalysis,151 the sensitivity and
taine classification139,140 is divided into four different stages: stage I, specificity for the detection of significant stenosis (>50% luminal reduc-
asymptomatic; stage IIa, intermittent claudication after more than 200 m tion) for 2D time of flight ranged from 64% to 100% and 68% to 96%,
of pain free walking; stage IIb, intermittent claudication after less than and for CE-MRA 92% to 100% and 91% to 99%, respectively.
200 m of walking; stage III, rest pain or nocturnal pain; and stage IV, In CE-MRA the run-off vessels are imaged during the arterial first
ischemic ulcers or gangrene. pass of Gd-based contrast material after intravenous injection (Fig.
The Rutherford classification139,140 is divided into three grades: (1) 44.6). As with CTA, precise bolus timing is essential for high quality
grade I compromises category 0: asymptomatic, category 1: mild clau- CE-MRA. Starting the acquisition too early, when the contrast bolus
dication, category 2: moderate claudication, and category 3: severe is still arriving in the vessels of interest, can cause ringing or Maki
claudication; (2) grade II compromises category 4: ischemic rest pain, artifacts.14 Starting the acquisition too late can cause venous enhance-
and category 5: minor tissue loss, nonhealing ulcer or focal gangrene; ment, hampering image quality, especially in the lower legs. Arterial
and (3) grade III compromises category 6: major, severe ischemic ulcers imaging is thus performed in the time window between arterial and
or gangrene. Both the Fontaine and the Rutherford classifications may venous enhancement. This time window depends on the rate of contrast
be used routinely in clinical practice or in research settings. agent injection, and will be patient specific.12 Arterial enhancement
For the evaluation of patients with symptomatic peripheral arterial must coincide with data readout of the central k-lines and, for improved
disease, several noninvasive tests such as segmental blood pressure, arterial/venous differentiation, the central k-lines have to be sampled
pulse volume recording, Doppler ultrasonography, and ankle–brachial before venous return.
528 SECTION V Vascular/Pericardium

A B C
FIG. 44.6.  (A) Contrast-enhanced magnetic resonance angiography of the distal aorta and common iliac arteries.
(B) Peripheral arteries show an occlusion of the right superficial femoral artery (arrow). (C) Run-off arteries.

To cover the whole region of the distal abdominal aorta and the multiple risk factors, adequate arterial enhancement with minimal
run-off vessels, two methods are clinically used: the single-injection venous enhancement can be obtained with this technique.155 The first
3-station moving-table (bolus-chase) technique and the multistation/ station covering the distal abdominal aorta and pelvic region is acquired
multiinjection method. In both methods, imaging is performed in during breath-holding and the acquired resolution is of the first two
a coronal orientation, usually with three stations with overlapping stations is 1.3 mm × 1.3 mm with a slice thickness of 3 mm, typically.
fields of view, covering the complete anatomic region. Typically, the The third station covering the lower leg scan be acquired with a submil-
first station covers the distal abdominal aorta and pelvic region, the limeter voxel size, allowing accurate diagnostic evaluation.156
second station the upper legs, and the third station lower legs. The Acquired high-resolution image data can be postprocessed using
quadrature body coil or phased-array surface coils are used for signal multiplanar reconstructions or MIPs; however, for obtaining an adequate
transmission and reception. In both techniques, 3D volumes are planned diagnosis, evaluation of 2D source images is essential. Furthermore,
on acquired 2D time of flight survey images. Thereafter, precontrast the role of CE-MRA in the evaluation of possible restenosis in (metal-
mask images are acquired in all stations. Subtraction of the precon- lic) stents is limited because of dephasing of CMR signal, causing signal
trast mask images from the contrast-enhanced images will suppress void. New developments in CMR imaging technology provide large
surrounding tissue.152 homogeneous field of view at 3T, revealing robust CE-MRA of the
In the bolus-chase technique, a biphasic contrast material injection complete vascular tree of the run-off vessels and allowing accurate
protocol is typically used. The first half of the contrast bolus is admin- clinical evaluation of patient on high field 3T scanners.153 Moreover,
istered at a flow rate of 1.0 to 1.2 mL/s and the remaining half at 0.5 two-point Dixon fat suppression allows subtractionless CE-MRA, avoid-
to 0.6 mL/s. Contrast injection is followed by a saline flush at 0.5 to ing possible misregistration artifacts between precontrast mask dataset
0.6 mL/s. Modern commercially available 1.5 T and 3 T CMR scanners and contrast-enhanced images.157 Furthermore, new techniques, such
allow acquisition of CE-MRA of the run-off vessels with a bolus-chase as three-station bolus-chase MRA with real-time fluoroscopic tracking
technique with excellent diagnostic performance.153 and precise triggering of table motion can provide high-resolution
In the multistation/multiinjection approach, the three regions of imaging of the run-off vessels. These new developments potentially can
the legs stations are acquired sequentially with two or three separate reduce contrast material dose and examination time.158
bolus injections of contrast medium.154 Gd-contrast is injected at a rate
of 1 to 1.5 mL/s, resulting in high signal intensity in the arteries. A
REFERENCES
disadvantage of this approach is that it is more time consuming, when
compared with the bolus-chase technique. However, in patients with A full reference list is available online at ExpertConsult.com
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 528.e1

REFERENCES T1-weighted MR images: relationship with increasing cumulative dose


of a gadolinium-based contrast material. Radiology. 2014;270(3):
1. Prince M, Yucel E, Kaufman J, Harrison D, Geller S. Dynamic 834–841.
gadolinium-enhanced three-dimensional abdominal MR arteriography. J 20. Murata N, Gonzalez-Cuyar L, Murata K, et al. Macrocyclic and other
Magn Reson Imaging. 1993;3(6):677–881. non-group 1 gadolinium contrast agents deposit low levels of
2. Port M, Idée J, Medina C, Robic C, Sabatou M, Corot C. Efficiency, gadolinium in brain and bone tissue: preliminary results from 9 patients
thermodynamic and kinetic stability of marketed gadolinium chelates with normal renal function. Invest Radiol. 2016;51(7):447–453.
and their possible clinical consequences: a critical review. Biometals. 21. Wang Y-X, Schroeder J, Siegmund H, et al. Total gadolinium tissue
2008;21(4):469–490. deposition and skin structural findings following the administration of
3. Reilly R. Risk for nephrogenic systemic fibrosis with Gadoteridol structurally different gadolinium chelates in healthy and ovariectomized
(ProHance) in patients who are on long-term hemodialysis. Clin J Am female rats. Quant Imaging Med Surg. 2015;5(4):534–545.
Soc Nephrol. 2008;3(3):747–751. 22. Kanda T, Osawa M, Oba H, et al. High signal intensity in dentate
4. Too CW, Ng WY, Tan CC, Mahmood MI, Tay KH. Screening for nucleus on unenhanced T1-weighted MR images: association with linear
impaired renal function in outpatients before iodinated contrast versus macrocyclic gadolinium chelate administration. Radiology.
injection: comparing the Choyke questionnaire with a rapid point-of- 2015;275(3):803–809.
care-test. Eur J Radiol. 2015;84:1227–1231. 23. Radbruch A, Weberling L, Kieslich P, et al. Gadolinium retention in the
5. Thomson H, Morcos S, Almen T, et al. Nephrogenic systemic fibrosis dentate nucleus and globus pallidus is dependent on the class of
and gadolinium-based contrast media: updated ESUR contrast medium contrast agent. Radiology. 2015;275(3):783–791.
safety committee guidelines. Eur Radiol. 2013;23(2):307–318. 24. Simonetti O, Finn J, White R, Laub G, Henry D. “Black blood”
6. Huckle J, Altun E, Jay M, Semelka R. Gadolinium deposition in humans: T2-weighted inversion-recovery MR imaging of the heart. Radiology.
when did we learn that gadolinium was deposited in vivo? Invest Radiol. 1996;199(1):49–57.
2016;51(4):236–240. 25. Miyazaki M, Akahane M. Non-contrast enhanced MR angiography:
7. Edwards B, Laumann A, Nardone B, et al. Advancing pharmacovigilance established techniques. J Magn Reson Imaging. 2012;35(1):1–19.
through academic-legal collaboration: the case of gadolinium-based 26. Miyazaki M, Lee V. Nonenhanced MRangiography. Radiology.
contrast agents and nephrogenic systemic fibrosis—a Research on 2008;248(1):20–43.
Adverse Drug Events and Reports (RADAR) report. Br J Radiol. 27. Holloway B, Rosewarne D, Jones R. Imaging of thoracic aortic disease.
2014;87(1042):20140307. Br J Radiol. 2011;84(Spec Iss 3):S338–S354.
7a.  Casey B. Norris lawsuit puts personal face on gadolinium debate. 28. Axel L. Blood flow effects in magnetic resonance imaging. AJR Am J
https://www.auntminnie.com/index.aspx?sec=ser&sub=def&pag=dis Roentgenol. 1984;143(6):1157–1166.
&ItemID=118804. Accessed March 26, 2018. 29. Bradley W, Waluch V. Blood flow: magnetic resonance imaging.
8. Prince M. Gadolinium-enhanced MR aortography. Radiology. Radiology. 1985;154(2):443–450.
1994;191(1):155–164. 30. Lotz J, Meier C, Leppert A, Galanski M. Cardiovascular flow
9. Prompona M, Cyran C, Nikolaou K, Bauner K, Reiser M, Huber A. measurement with phase-contrast MR imaging: basic facts and
Contrast-enhanced whole-heart MR coronary angiography at 3.0 T implementation. Radiographics. 2002;22(3):651–671.
using the intravascular contrast agent gadofosveset. Invest Radiol. 31. Nelemans P, Leiner T, de Vet H, van Engelshoven J. Peripheral arterial
2009;44(7):369–374. disease: meta-analysis of the diagnostic performance of MR
10. Jaspers K, Versluis B, Leiner T, et al. MR angiography of collateral angiography. Radiology. 2000;217(1):105–114.
arteries in a hind limb ischemia model: comparison between blood pool 32. Dixon W. Simple proton spectroscopic imaging. Radiology. 1984;153(1):
agent Gadomer and small contrast agent Gd-DTPA. PLoS One. 189–194.
2011;6(1):e16159. 33. Nezafat M, Henningsson M, Ripley D, et al. Coronary MR angiography
11. Wilson G, Hoogeveen R, Willinek W, Muthupillai R, Maki J. Parallel at 3T: fat suppression versus water-fat separation. J Cardiovasc Magn
imaging in MR angiography. Top Magn Reson Imaging. 2004;15(3): Reson. 2014;16(suppl 1):175.
169–185. 34. Calkoen E, Roest A, van der Geest R, de Roos A, Westenberg J.
12. Prince M, Chabra S, Watts R, et al. Contrast material travel times in Cardiovascular function and flow by 4-dimensional magnetic resonance
patients undergoing peripheral MR angiography. Radiology. 2002; imaging techniques: new applications. J Thorac Imaging.
224(1):55–61. 2014;29(3):185–196.
13. Maki J, Prince M, Chenevert T. Optimizing three-dimensional 35. Dyverfeldt P, Bissell M, Barker A, et al. 4D flow cardiovascular magnetic
gadolinium-enhanced magnetic resonance angiography. Invest Radiol. resonance consensus statement. J Cardiovasc Magn Reson. 2015;
1998;33(9):528–537. 17(1):72.
14. Maki J, Prince M, Londy F, Chenevert T. The effects of time-varying 36. Mitsuzaki K, Yamashita Y, Onomichi M, et al. Delineation of simulated
intravascular signal intensity and k-space acquisition order on vascular stenosis with Gd-DTPA-enhanced 3D gradient echo MR
three-dimensional MR angiography image quality. J Magn Reson angiography: an experimental study. J Comput Assist Tomogr.
Imaging. 1996;6(4):642–651. 2000;24(1):77–82.
15. Willinek W, Gieseke J, Conrad R, et al. Randomly segmented central 37. Korosec F, Mistretta C. MR angiography: basic principles and theory.
k-space ordering in high-spatial-resolution contrast-enhanced MR Magn Reson Imaging Clin N Am. 1998;6(2):223–256.
angiography of the supra-aortic arteries: initial experience. Radiology. 38. Inzitari D, Eliasziw M, Gates P, et al. The causes and risk of stroke in
2002;225(2):583–588. patients with asymptomatic internal-carotid-artery stenosis. N Engl J
16. Grobner T. Gadolinium, a specific trigger for the development of Med. 2000;342(23):1693–1701.
nephrogenic fibrosing dermopathy and nephrogenic systemic fibrosis? 39. Mozaffarian D, Benjamin E, Go AS, et al; on behalf of the American
Nephrol Dial Transplant. 2006;21(4):1104–1108. Heart Association Statistics Committee and Stroke Statistics
17. Prince M, Zhang H, Roditi G, Leiner T, Kucharczyk W. Risk factors for Subcommittee. Heart disease and stroke statistics—2016 update. A
NSF: a literature review. J Magn Reson Imaging. 2009;30(6):1298–1308. report from the American Heart Association. Circulation. 2016;133(4):
18. Kanda T, Fukusato T, Matsuda M, et al. Gadolinium-based contrast 38–361.
agent accumulates in the brain even in subjects without severe renal 40. Amarenco P, Lavallée P, Labreuche J, et al. One-year risk of stroke after
dysfunction: evaluation of autopsy brain specimens with inductively transient ischemic attack or minor stroke. N Engl J Med.
coupled plasma mass spectroscopy. Radiology. 2015;276(1):228–232. 2016;374(16):1533–1542.
19. Kanda T, Ishii K, Kawaguchi H, Kitajima K, Takenaka D. High signal 41. Hankey G, Warlow C, Sellar R. Cerebral angiographic risk in mild
intensity in the dentate nucleus and globus pallidus on unenhanced cerebrovascular disease. Stroke. 1990;21(2):209–222.
528.e2 SECTION V Vascular/Pericardium

42. Wardlaw J, Chapel F, Best J, Wartolowska K, Berry E. Noninvasive 63. Steenburg S, Ravenel J. Acute traumatic thoracic aortic injuries:
imaging compared with intra-arterial angiography in the diagnosis of experience with 64-MDCT. AJR Am J Roentgenol.
symptomatic carotid stenosis: a meta-analysis. Lancet. 2006;367(9521): 2008;191(5):1564–1569.
1503–1512. 64. Freeman LA, Young PM, Foley TA, et al. CT and MRI assessment of the
43. Arous E, Simons J, Flahive J, et al. National variation in preoperative aortic root and ascending aorta. AJR Am J Roentgenol. 2013;200:W581.
imaging, carotid duplex ultrasound criteria, and threshold for surgery 65. Erbel R, Aboyans V, Boileau C, et al; ESC Committee for Practice
for asymptomatic carotid artery stenosis. J Vasc Surg. 2015;62(4): Guidelines. 2014 ESC guidelines on the diagnosis and treatment of
937–944. aortic diseases. Eur Heart J. 2014;35(41):2873–2926.
44. Brot T, Halperin J, Abbara S, et al. 2011 guideline on management of 66. Plathow C, Ley S, Zaporozhan J, et al. Assessment of reproducibility and
patients with extracranial carotid and vertebral artery disease: executive stability of different breath-hold maneuvers by dynamic MRI:
summary. J Am Coll Cardiol. 2011;57(8):1002–1044. comparison between healthy adults and patients with pulmonary
45. DeMarco J, Willinek W, Finn J, Huston IIIJ. Current state-of-the-art hypertension. Eur Radiol. 2006;16(1):173–179.
1.5T and 3T extracranial carotid contrast-enhanced magnetic resonance 67. Kramer C, Barkhausen J, Flamm S, Kim R, Nagel E. Standardized
angiography. Neuroimag Clin N Am. 2012;22(2):235–257. cardiovascular magnetic resonance imaging (CMR) protocols,
46. Dehkharghani S, Qiu D, Albin L, Saindane A. Dose reduction in Society for Cardiovascular Magnetic Resonance: Board of Trustees
contrast-enhanced cervical MR angiography: field strength dependency Task Force on Standardized Protocols. J Cardiovasc Magn Reson.
of vascular signal intensity, contrast administration, and arteriographic 2008;10:35.
quality. AJR Am J Roentgenol. 2015;204(6):W701–W706. 68. Lim R, Winchester P, Bruno M, et al. Highly accelerated single
47. Tomasian A, Salamon N, Lohan DG, Jalili M, Villablanca J, Finn J. breath-hold noncontrast thoracic MRA: evaluation in a clinical
Supraaortic arteries: contrast material dose reduction at 3.0-T population. Invest Radiol. 2013;48(3):145–151.
high-spatial-resolution MR angiography-feasibility study. Radiology. 69. Krishnam MS, Tomasian A, Malik S, Desphande V, Laub G, Ruehm S.
2008;249:980–990. Image quality and diagnostic accuracy of unenhanced SSFP MR
48. Townsend T, Saloner D, Pan X, Rapp J. Contrast material-enhanced angiography compared with conventional contrast-enhanced MR
MRA overestimates severity of carotid stenosis, compared with 3D angiography for the assessment of thoracic aortic diseases. Eur Radiol.
time-of-flight MRA. J Vasc Surg. 2003;38(1):36–40. 2010;20(6):1311–1320.
49. Patel M, Kuntz K, Klufas R, et al. Preoperative assessment of the carotid 70. Srichai M, Kim S, Axel L, Babb J, Hecht E. Non-gadolinium-enhanced
bifurcation. Can magnetic resonance angiography and duplex 3-dimensional magnetic resonance angiography for the evaluation of
ultrasonography replace contrast arteriography? Stroke. thoracic aortic disease: a preliminary experience. Tex Heart Inst J.
1995;26(10):1753–1758. 2010;37(1):58–65.
50. DeMarco J, Schonfeld S, Keller I, Bernstein M. Contrast-enhanced 71. Hiratzka L, Bakris G, Beckman J, et al; American College of Cardiology
carotid MR angiography with commercially available triggering Foundation/American Heart Association Task Force on Practice
mechanisms and elliptic centric phase encoding. AJR Am J Roentgenol. Guidelines; American Association for Thoracic Surgery; American
2001;12(3):175–181. College of Radiology; American Stroke Association; Society of
51. Wright V, Olan W, Dick B, et al. Assessment of CE-MRA for the rapid Cardiovascular Anesthesiologists; Society for Cardiovascular
detection of supra-aortic vascular disease. Neurology. 2005;65(1): Angiography and Interventions; Society of Interventional Radiology;
27–32. Society of Thoracic Surgeons; Society for Vascular Medicine. 2010
52. Zhao H, Wang J, Liu X, et al. Assessment of carotid artery atherosclerotic ACCF/AHA/AATS/ACR/ASA/SCA/SCAI/SIR/STS/SVM Guidelines for
disease by using three-dimensional fast black-blood MR imaging: the diagnosis and management of patients with thoracic aortic disease.
comparison with DSA. Radiology. 2005;274(2):508–516. A report of the American College of Cardiology Foundation/American
53. Mihai G, Winner M, Raman S, Rajagopalan S, Simonetti O, Chung Y. Heart Association Task Force on Practice Guidelines, American
Assessment of carotid stenosis using three-dimensional T2-weighted Association for Thoracic Surgery, American College of Radiology,
dark blood imaging: initial experience. J Magn Reson Imaging. American Stroke Association, Society of Cardiovascular
2012;35(2):449–455. Anesthesiologists, Society for Cardiovascular Angiography and
54. Varennes L, Tahon F, Kastler A, et al. Fibromuscular dysplasia: what the Interventions, Society of Interventional Radiology, Society of Thoracic
radiologist should know: a pictorial review. Insights Imaging. Surgeons, and Society for Vascular Medicine. J Am Coll Cardiol.
2015;6(3):295–307. 2010;55(14):e27–e129.
55. Debette S, Leys D. Cervical-artery dissections: predisposing factors, 72. Cozijnsen L, Braam R, Waalewijn R, et al. What is new in dilatation of
diagnosis, and outcome. Lancet Neurol. 2009;8(7):668–678. the ascending aorta? Review of current literature and practical advice for
56. Ben Hassena W, Machet A, Edjlali-Goujon M, et al. Imaging of cervical the cardiologist. Circulation. 2011;123(8):924–928.
artery dissection. Diagn Interv Imaging. 2014;95(12):1151–1161. 73. Svensson L, Adams D, Bonow R, et al. Aortic valve and ascending aorta
57. Desjardins B, Dill KE, Flamm SD, et al. ACR Appropriateness Criteria® guidelines for management and quality measures. Ann Thorac Surg.
pulsatile abdominal mass, suspected abdominal aortic aneurysm. Int J 2013;95(suppl 6):S1–S66.
Cardiovasc Imaging. 2013;29(1):177–183. 74. Mendoza D, Kochar M, Devereux R, et al; GenTAC (National Registry of
58. Douglas P, Garcia M, Haines D, et al. Appropriate use of Genetically Triggered Thoracic Aortic Aneurysms and Cardiovascular
echocardiography. J Am Soc Echocardiogr. 2011;24(3):229–267. Conditions) Study Investigators. Impact of image analysis methodology
59. Evangelista A, Flachskampf F, Erbel R, et al. Echocardiography in aortic on diagnostic and surgical classification of patients with thoracic aortic
diseases: EAE recommendations for clinical practice. Eur J Echocardiogr. aneurysms. Ann Thorac Surg. 2011;92(3):904–912.
2010;11(3):645–658. 75. Landenhed M, Engstrom G, Gottsater A, et al. Risk profiles for aortic
60. Flachskampf F, Decoodt P, Fraser A, Daniel W, Roelandt J. dissection and ruptured or surgically treated aneurysms: a prospective
Recommendations for performing transesophageal echocardiography. cohort study. J Am Heart Assoc. 2015;4(1):e001513.
Eur J Echocardiogr. 2001;2(1):8–21. 76. Prince M. Gadolinium-enhanced MR aortography. Radiology.
61. Flachskampf F, Badano L, Daniel W, et al. Recommendations for 1994;191(1):155–164.
transoesophageal echocardiography-update 2010. Eur Heart J Cardiovasc 77. Arlart I, Gerlach A, Kolb M, Erpenbach S, Würstlin S. [MR angiography
Imaging. 2010;11(7):557–576. using Gd-DTPA in staging of abdominal aortic aneurysm: a correlation
62. Sommer T, Fehske W, Holzknecht N, et al. Aortic dissection: a with DSA and CT]. Rofo. 1997;167(9):257–263. [Article in German]
comparative study of diagnosis with spiral CT, multiplanar 78. Singh K, Bønaa K, Jacobsen B, Bjørk L, Solberg S. Prevalence of and risk
transesophageal echocardiography, and MR imaging. Radiology. factors for abdominal aortic aneurysms in a population-based study: the
1996;199(2):347–352. Tromsø Study. Am J Epidemiol. 2001;154(3):236–244.
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 528.e3

79. Forsdahl S, Singh K, Solberg S, Jacobsen B. Risk factors for abdominal 100. Leiner T, Nelemans P, de Haan M, de Leeuw P, van Engelshoven J,
aortic aneurysms: a 7-year prospective study: the Tromsø Study, Vasbinder G. Addition of phase-contrast data to anatomical evaluation
1994-2001. Circulation. 2009;119(16):2202–2208. substantially improves the diagnostic accuracy of dynamic contrast-
80. Tessier D, Stone W, Fow R, et al. Clinical features and management of enhanced renal MR angiography for the detection of renal artery
splenic artery pseudoaneurysm: case series and cumulative review of stenosis. In: RSNA Annual Meeting Abstract Book. 2006:540.
literature. J Vasc Surg. 2003;38(5):969–974. 101. ASTRAL Investigators, Wheatley K, Ives N, Gray R, et al.
81. Deipolyi A, Rho J, Khademhosseini A, Oklu R. Diagnosis and Revascularization versus medical therapy for renal-artery stenosis. N
management of mycotic aneurysms. Clin Imaging. 2016;40(2): Engl J Med. 2009;361(20):1953–1962.
256–262. 102. Cooper C, Murphy T, Cutlip D, et al; CORAL Investigators. Stenting and
82. Nienaber CA. The role of imaging in acute aortic syndromes. Eur Heart medical therapy for atherosclerotic renal-artery stenosis. N Engl J Med.
J Cardiovasc Imaging. 2013;14(1):15–23. 2014;370(1):13–22.
83. Chiappini B, Schepens M, Tan E, et al. Early and late outcomes of acute 103. Caielli P, Frigo A, Pengo M, et al. Treatment of atherosclerotic
type A aortic dissection: analysis of risk factors in 487 consecutive renovascular hypertension: review of observational studies and a
patients. Eur Heart J. 2005;26(2):180–186. meta-analysis of randomized clinical trials. Nephrol Dial Transplant.
84. Nienaber C, von Kodolitsch Y, Nicolas V, et al. The diagnosis of thoracic 2015;30(4):541–553.
aortic dissection by noninvasive imaging procedures. N Engl J Med. 104. Kumbhani D, Bavry A, Harvey J, et al. Clinical outcomes after
1993;328(1):1–9. percutaneous revascularization versus medical management in patients
85. Shiga T, Wajima Z, Apfel C, Inoue T, Ohe Y. Diagnostic accuracy of with significant renal artery stenosis: a meta-analysis of randomized
transesophageal echocardiography, helical computed tomography, and controlled trials. Am Heart J. 2011;161(3):622–630.
magnetic resonance imaging for suspected thoracic aortic dissection 105. Bavishi C, de Leeuw P, Messerli F. Atherosclerotic renal artery stenosis
systematic review and meta-analysis. Arch Intern Med. 2006;166(13): and hypertension: pragmatism, pitfalls, and perspectives. Am J Med.
1350–1356. 2016;129(6):635.e5–635.e14.
86. Harding M, Smith L, Himmelstein S, et al. Renal artery stenosis: 106. Mohan I, Bourke V. The management of renal artery stenosis: an
prevalence and associated risk factors in patients undergoing routine alternative interpretation of ASTRAL and CORAL. Eur J Vasc Endovasc
cardiac catheterization. J Am Soc Nephrol. 1992;2(11):1608–1616. Surg. 2015;49(4):465–473.
87. Olin J, Melia M, Young J, Graor R, Risius B. Prevalence of atherosclerotic 107. Hansen K, Wilson D, Craven T, et al. Mesenteric artery disease in the
renal artery stenosis in patients with atherosclerosis elsewhere. Am J elderly. J Vasc Surg. 2004;40(1):45–52.
Med. 1990;88(1N):46N–51N. 108. Roobottom C, Dubbins P. Significant disease of the celiac and superior
88. Rimmer J, Gennari F. Atherosclerotic renovascular disease and mesenteric arteries in asymptomatic patients: predictive value of
progressive renal failure. Ann Intern Med. 1993;118(9):712–719. Doppler sonography. AJR Am J Roentgenol. 1993;161(5):985–988.
89. Sanidas E, Seferou M, Papadopoulos D, et al. Renal fibromuscular 109. Parameshwarappa S, Savlania A, Viswanathan S, Gadhinglajkar S,
dysplasia: a not so common entity of secondary hypertension. J Clin Raman K, Unnikrishnan M. Chronic mesenteric ischemia and
Hypertens. 2016;18(3):240–246. therapeutic paradigm of mesenteric revascularization. Indian J
90. Kohler T, Zierler R, Martin R, et al. Noninvasive diagnosis of renal Gastroenterol. 2014;33(2):169–174.
artery stenosis by ultrasonic duplex scanning. J Vasc Surg. 1986;4(5): 110. Matsumoto A, Tegtmeyer C, Fitzcharles E, et al. Percutaneous
450–456. transluminal angioplasty of visceral arterial stenoses: results and
91. Taylor D, Kettler M, Moneta G, et al. Duplex ultrasound scanning in the long-term clinical follow-up. J Vasc Interv Radiol. 1995;6(2):165–174.
diagnosis of renal artery stenosis: a prospective evaluation. J Vasc Surg. 111. Brandt L, Boley S. AGA technical review on intestinal ischemia.
1988;7(2):363–369. Gastroenterology. 2000;118(5):954–968.
92. Olin J, Piedmonte M, Young J, De Anna S, Grubb M, Childs M. The 112. Carrick R, Borge M, Labropolous N, Rodriguez H. Chronic mesenteric
utility of duplex ultrasound scanning of the renal arteries for diagnosing ischemia resulting from isolated lesions of the superior mesenteric
significant renal artery stenosis. Ann Intern Med. 1995;122(1):833–838. artery a case report. Angiology. 2005;56(6):785–788.
93. Hoffmann U, Edwards JM, Carter S, et al. Role of duplex scanning for 113. Cangemi J, Picco M. Intestinal ischemia in the elderly. Gastroenterol Clin
the detection of atherosclerotic renal artery disease. Kidney Int. North Am. 2009;38(3):527–540.
1991;39(6):1232–1239. 114. Balduf L, Langsfeld M, Marek J, Tullis M, Kasirajan K, Matteson B.
94. Zeisbrich M, Kihm L, Drüschler F, Zeier M, Schwenger V. When is Complication rates of diagnostic angiography performed by vascular
contrast-enhanced sonography preferable over conventional ultrasound surgeons. Vasc Endovascular Surg. 2002;36(6):439–445.
combined with Doppler imaging in renal transplantation? Clin Kidney J. 115. Moneta G, Lee R, Yeager R, Taylor L, Porter J. Mesenteric duplex
2015;8(5):606–614. scanning: a blinded prospective study. J Vasc Surg. 1993;17(1):79–84.
95. Rountas C, Vlychou M, Vassiou K, et al. Imaging modalities for renal 116. Oliva I, Davarpanah A, Rybicki F, et al. ACR appropriateness criteria:
artery stenosis in suspected renovascular hypertension: prospective imaging of mesenteric ischemia. Abdom Imaging. 2013;38(4):
intraindividual comparison of color Doppler US, CT angiography, 714–719.
GD-enhanced MR angiography, and digital subtraction angiography. 117. Moneta GL. Screening for mesenteric vascular insufficiency and
Ren Fail. 2007;29(13):295–302. follow-up of mesenteric artery bypass procedures. Semin Vasc Surg.
96. Steinwender C, Schutzenberger W, Fellner F, et al. 64-Detector CT 2001;14(3):186–192.
angiography in renal artery stent evaluation: prospective comparison 118. Van Petersen A, Kolkman J, Meerwaldt R, et al. Mesenteric stenosis,
with selective catheter angiography. Radiology. 2009;252(1):299–305. collaterals, and compensatory blood flow. J Vasc Surg. 2014;60(1):
97. Bakker J, Beek F, Beutler J, et al. Renal artery stenosis and accessory 111–119.
renal arteries: accuracy of detection and visualization with gadolinium- 119. Schaefer P, Pfarr J, Trentmann J, et al. Comparison of noninvasive
enhanced breath-hold MR angiography. Radiology. 1998;207(2): imaging modalities for stenosis grading in mesenteric arteries. Rofo.
497–504. 2013;185(7):628–634.
98. Vasbinder G, Nelemans P, Kessels A, Kroon A, de Leeuw P, van 120. Meaney J, Prince M, Nostrant T, Stanley J. Gadolinium-enhanced MR
Engelshoven J. Diagnostic tests for renal artery stenosis in patients angiography of visceral arteries in patients with suspected chronic
suspected of having renovascular hypertension: a meta-analysis. Ann mesenteric ischemia. J Magn Reson Imaging. 1997;7(1):171–176.
Intern Med. 2001;135(6):401–411. 121. Holland G, Dougherty L, Carpenter J, et al. Breath-hold ultrafast
99. Prince M, Schoenberg S, Ward J, Londy F, Wakefield T, Stanley J. three-dimensional gadolinium-enhanced MR angiography of the aorta
Hemodynamically significant atherosclerotic renal artery stenosis: MR and the renal and other visceral abdominal arteries. AJR Am J
angiographic features. Radiology. 1997;205(1):128–136. Roentgenol. 1996;166(4):971–981.
528.e4 SECTION V Vascular/Pericardium

122. Jaster A, Choudhery S, Ahn R, et al. Anatomic and radiologic review of ACC/AHA Task Force on Practice Guidelines Writing Committee to
chronic mesenteric ischemia and its treatment. Clin Imaging. Develop Guidelines for the Management of Patients With Peripheral
2016;40(5):961–969. Arterial Disease; American Association of Cardiovascular and
123. Laissy J, Trillaud H, Douek P. MR angiography: noninvasive vascular Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute;
imaging of the abdomen. Abdom Imaging. 2002;27(5):488–506. Society for Vascular Nursing; TransAtlantic Inter-Society Consensus;
124. Hagspiel K, Flors L, Hanley M, Norton P. Computed tomography Vascular Disease Foundation. ACC/AHA 2005 Practice guidelines for the
angiography and magnetic resonance angiography imaging of the management of patients with peripheral arterial disease (lower
mesenteric vasculature. Tech Vasc Interv Radiol. 2015;18(1):2–13. extremity, renal, mesenteric, and abdominal aortic): a collaborative
125. Iozzelli A, D’Orta G, Aliprandi A, Secchi F, Di Leo G, Sardanelli F. The report from the American Association for Vascular Surgery/Society for
value of true-FISP sequence added to conventional gadolinium- Vascular Surgery, Society for Cardiovascular Angiography and
enhanced MRA of abdominal aorta and its major branches. Eur J Radiol. Interventions, Society for Vascular Medicine and Biology, Society of
2009;72(3):489–493. Interventional Radiology, and the ACC/AHA Task Force on Practice
126. Fowkes FG, Rudan D, Rudan I, et al. Comparison of global estimates of Guidelines (Writing Committee to Develop Guidelines for the
prevalence and risk factors for peripheral artery disease in 2000 and Management of Patients With Peripheral Arterial Disease): endorsed by
2010: a systematic review and analysis. Lancet. the American Association of Cardiovascular and Pulmonary
2013;382(9901):1329–1340. Rehabilitation; National Heart, Lung, and Blood Institute; Society for
127. Allison MA, Ho E, Denenberg JO, et al. Ethnic-specific prevalence of Vascular Nursing; TransAtlantic Inter-Society Consensus; and Vascular
peripheral arterial disease in the United States. Am J Prev Med. Disease Foundation. Circulation. 2006;113(11):e463–e654.
2007;32(4):328–333. 142. Rooke TW, Hirsch AT, Misra S, et al; Society for Cardiovascular
128. Sampson UK, Fowkes FG, McDermott MM, et al. Global and regional Angiography and Interventions; Society of Interventional Radiology;
burden of death and disability from peripheral artery disease: 21 world Society for Vascular Medicine; Society for Vascular Surgery. 2011 ACCF/
regions, 1990 to 2010. Glob Heart. 2014;9(1):145–158. AHA focused update of the guideline for the management of patients
129. Norgren L, Hiatt WR, Dormandy JA, et al. Inter-society consensus for with peripheral artery disease (updating the 2005 guideline): a report of
the management of peripheral arterial disease (TASC II). J Vasc Surg. the American College of Cardiology Foundation/American Heart
2007;45(suppl S):S5–S67. Association Task Force on Practice Guidelines. J Am Coll Cardiol.
130. Eraso LH, Fukaya E, Mohler ER 3rd, Xie D, Sha D, Berger JS. Peripheral 2011;58(19):2020–2045.
arterial disease, prevalence and cumulative risk factor profile analysis. 143. O’Keeffe ST, Persson AV. Use of noninvasive vascular laboratory in
Eur J Prev Cardiol. 2014;21(6):704–711. diagnosis of venous and arterial disease. Cardiol Clin.
131. Khawaja FJ, Kullo IJ. Novel markers of peripheral arterial disease. Vasc 1991;9(3):429–442.
Med. 2009;4(4):381–392. 144. Ankle Brachial Index Collaboration, Fowkes FG, Murray GD, et al. Ankle
132. Hirsch AT, Criqui MH, Treat-Jacobson D, et al. Peripheral arterial brachial index combined with Framingham Risk Score to predict
disease detection, awareness, and treatment in primary care. JAMA. cardiovascular events and mortality: a meta-analysis. JAMA.
2001;286(11):1317–1324. 2008;300(2):197–208.
133. Criqui MH, Ninomiya JK, Wingard DL, Ji M, Fronek A. Progression of 145. Newman AB, Shemanski L, Manolio TA, et al. Ankle-arm index as a
peripheral arterial disease predicts cardiovascular disease morbidity and predictor of cardiovascular disease and mortality in the Cardiovascular
mortality. J Am Coll Cardiol. 2008;52(21):1736–1742. Health Study: the Cardiovascular Health Study Group. Arterioscler
134. Araki Y, Kumakura H, Kanai H, et al. Prevalence and risk factors for Thromb Vasc Biol. 1999;19(3):538–545.
cerebral infarction and carotid artery stenosis in peripheral arterial 146. McDermott MM, Feinglass J, Slavensky R, Pearce WH. The ankle-
disease. Atherosclerosis. 2012;223(2):473–477. brachial index as a predictor of survival in patients with peripheral
135. Sigvant B, Lundin F, Wahlberg E. The risk of disease progression in vascular disease. J Gen Intern Med. 1994;9(8):445–449.
peripheral arterial disease is higher than expected: a meta-analysis of 147. McKenna M, Wolfson S, Kuller L. The ratio of ankle and arm arterial
mortality and disease progression in peripheral arterial disease. Eur J pressure as an independent predictor of mortality. Atherosclerosis.
Vasc Endovasc Surg. 2016;51(3):395–403. 1991;87(2–3):119–128.
136. Goff DC Jr, Lloyd-Jones DM, Bennett G, et al; American College of 148. Doobay AV, Anand SS. Sensitivity and specificity of the ankle-brachial
Cardiology/American Heart Association Task Force on Practice index to predict future cardiovascular outcomes: a systematic review.
Guidelines. 2013 ACC/AHA guideline on the assessment of Arterioscler Thromb Vasc Biol. 2005;25(7):1463–1469.
cardiovascular risk: a report of the American College of Cardiology/ 149. Wikström J, Hansen T, Johansson L, Lind L, Ahlström H. Ankle brachial
American Heart Association Task Force on Practice Guidelines. J Am index <0.9 underestimates the prevalence of peripheral artery occlusive
Coll Cardiol. 2014;63(25):2935–2959. disease assessed with whole-body magnetic resonance angiography in
137. Greenland P, Alpert JS, Beller GA, et al; American College of Cardiology the elderly. Acta Radiol. 2008;49(2):143–149.
Foundation; American Heart Association. ACCF/AHA guideline for 150. Meyer BC, Werncke T, Foert E, et al. Do the cardiovascular risk profile
assessment of cardiovascular risk in asymptomatic adults: a report of and the degree of arterial wall calcification influence the performance of
the American College of Cardiology Foundation/American Heart MDCT angiography of lower extremity arteries? Eur Radiol.
Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2010;20(2):497–505.
2010;56(25):e50–e103. 151. Nelemans PJ, Leiner T, de Vet HC, van Engelshoven JM. Peripheral
138. van den Bosch H, Westenberg J, Setz-Pels W, et al. Prognostic value of arterial disease: meta-analysis of the diagnostic performance of MR
cardiovascular MR imaging biomarkers on outcome in peripheral angiography. Radiology. 2000;217(1):105–114.
arterial disease: a 6-year follow-up pilot study. Int J Cardiovasc Imaging. 152. Ho KY, de Haan MW, Kessels AG, Kitslaar PJ, van Engelshoven JM.
2016;32(8):1281–1288. Peripheral vascular tree stenosis: detection with subtracted and
139. Dormandy JA, Rutherford RB. Management of peripheral arterial nonsubtracted MR angiography. Radiology. 1998;206(3):673–681.
disease (PAD). TASC Working Group. TransAtlantic Inter-Society 153. van den Bosch HC, Westenberg JJ, Caris R, et al. Peripheral arterial
Consensus (TASC). J Vasc Surg. 2000;31(1):S1–S296. occlusive disease: 3.0-T versus 1.5-T MR angiography compared with
140. Norgren L, Hiatt WR, Dormandy JA, et al. Inter-Society Consensus for digital subtraction angiography. Radiology. 2013;266(1):337–346.
the Management of Peripheral Arterial Disease (TASC II). J Vasc Surg. 154. Watanabe Y, Dohke M, Okumura A, et al. Dynamic subtraction
2007;45(suppl S):S5–S67. contrast-enhanced MR angiography: technique, clinical applications,
141. Hirsch AT, Haskal ZJ, Hertzer NR, et al; Society for Vascular Surgery; and pitfalls. Radiographics. 2000;20(1):135–152.
Society for Cardiovascular Angiography and Interventions; Society for 155. Dinter DJ, Neff KW, Visciani G, et al. Peripheral bolus-chase MR
Vascular Medicine and Biology; Society of Interventional Radiology; angiography: analysis of risk factors for nondiagnostic image quality of
CHAPTER 44  Cardiovascular MRA: Carotids, Aorta, and Peripheral Vessels 528.e5

the calf vessels—a combined retrospective and prospective study. AJR 157. Leiner T, Habets J, Versluis B, et al. Subtractionless first-pass single
Am J Roentgenol. 2009;193(1):234–240. contrast medium dose peripheral MR angiography using two-point
156. Bezooijen R, van den Bosch HC, Tielbeek AV, et al. Peripheral arterial Dixon fat suppression. Eur Radiol. 2013;23(8):2228–2235.
disease: sensitivity-encoded multiposition MR angiography compared 158. Johnson CP, Weavers PT, Borisch EA, et al. Three-station three-
with intraarterial angiography and conventional multiposition MR dimensional bolus-chase MR angiography with real-time fluoroscopic
angiography. Radiology. 2004;231(1):263–271. tracking. Radiology. 2014;272(1):241–251.
45 
Pulmonary Artery
Csilla Celeng, R. Nils Planken, David A. Bluemke, and Tim Leiner

Pathologies involving the pulmonary arterial system include pulmonary deep venous system, and occasionally the reference standard, catheter-
embolus (PE), pulmonary arterial hypertension (PAH), congenital based pulmonary angiography. From the resulting array of diagnostic
anomalies, and pulmonary artery tumors. These diseases present a information, the clinician must then assess the patient’s likelihood of
unique clinical diagnostic challenge often requiring multiple diagnostic having PE and initiate treatment. The most extensively used clinical
examinations spanning the entire radiologic armamentarium. Initially, score in acute PE is the pulmonary embolism severity index (PESI)
cardiovascular magnetic resonance (CMR) played only a minor role in or its simplified version (sPESI), which is helpful in the prognostic
evaluation of the pulmonary arterial system. However, with the advent assessment of 30-day mortality. For hemodynamically stable patients,
of improved, high-performance gradients and bolus gadolinium, contrast- pretest probability relies on the revised Geneva score and/or Wells rule,
enhanced cardiovascular magnetic resonance (CE-CMR) is now well which both aim to stratify patients into three-level (low, intermedi-
suited for this challenge. It not only provides precise, three-dimensional ate, and high risk) or two-level (PE unlikely and PE likely) schemes
(3D) anatomic information, but also provides functional data critical based on information that is easy to obtain.4 Elevated plasma levels of
to a complete evaluation of the pulmonary arterial system. Beyond the D-dimer as well as markers of myocardial injury (cardiac troponin I
basic flow parameters, four-dimensional (4D)-CMR is able to assess or T) and/or ventricular dysfunction (brain natriuretic peptide-BNP
information about more advanced parameters (e.g., wall shear stress); and N-terminal-pro BNP) support the diagnosis of PE. However, the
however this technique requires standardization before routine clinical relatively low positive predictive values of laboratory biomarkers do
application. Further developments are expected in the field of molecular not allow reliable exclusion of PE. The suspicion of PE is therefore a
imaging by using target-specific contrast agents in humans, allowing common indication for imaging.
the interaction and detection of pathological mechanisms. CMR is CTA is the most used noninvasive imaging method for detection of
noninvasive and requires neither iodinated contrast nor ionizing radia- PE. A negative CTA in patients with low/intermediate risk allows for
tion, and consistently demonstrates anatomic accuracy comparable with safely ruling out PE, although in patients with negative CTA, but high
that of computed tomography (CT) while providing reproducible func- probability of PE, further investigations are required.4 The high spatial
tional data previously only available by catheter based pulmonary resolution of current CT scanners and the administration of contrast
angiography. media allows for the identification of PE at least to the segmental level.
However, potential pitfalls can occur, especially at distal levels of the
pulmonary arteries. A retrospective review of PE on CTA showed dis-
PULMONARY EMBOLISM cordance between subspecialists in chest radiology and general radiolo-
Venous thromboembolism (VTE) incorporates deep venous thrombosis gists in 26% of the cases. The disagreement was most frequent if the
(DVT) and PE. VTE is one of the major contributors to global disease PE was adjudicated as solitary (46% of solitary PEs were later adjudicated
burden: the incidence ranges from 100 to 200 per 100,000 persons per as negative) or segmental/subsegmental (27% of segmental and 59%
year with an increased incidence from 200 to 700 per 100,000 in those of subsegmental PE were later considered as negative).6 Additionally,
≥70 years.1 The risk for the disease doubles in each decade after the CTA exposes the patient to ionizing radiation and requires iodinated
age of 40 years.2 PE represents the most serious and potentially life- intravenous contrast, which is potentially nephrotoxic and can provoke
threatening condition related to VTE. It often occurs as a consequence allergic reactions. Radiation exposure is of particular concern in young
of DVT when a blood clot breaks off, migrates, and eventually obstructs adults and those who are referred for multiple CTA scans because of
the pulmonary arteries. An estimation model based on data from six a history of prior PE.
countries of the European Union showed that 317,000 deaths could V̇ /Q̇ scintigraphy is a combined noninvasive procedure that uses
be attributable to VTE.3 Approximately one-third of patients suffered nuclear radiotracers for diagnosing PE. The reported radiation exposure
from sudden death caused by PE, 59% died as a result of existing but of the examination is significantly lower compared with CTA (1.1 mSv
undiagnosed PE, and only 7% of patients who died were appropriately vs. 4 mSv) and might be a preferred alternative over CTA in younger
identified with PE. Because the presenting signs and symptoms are patients, especially women, in whom radiation exposure of CTA is
nonspecific and overlap in a wide variety of common ailments, diag- associated with higher risk of breast cancer.4,7,8 However, V̇ /Q̇ scans are
nosing PE is challenging and relies on a multidimensional prognostic frequently nondiagnostic, which raised concerns because of the need
model. Risk stratification of patients into high-risk, intermediate-risk, for further diagnostic tests. Diagnostic accuracy can be improved by
and low-risk categories is based on the additive value of clinical scores, incorporation of a single photon emission tomography (SPECT) to V̇ /Q̇
imaging tests, and laboratory markers.4 Therefore the current workup scintigraphy.9
typically includes two or more of the following tests: blood assays, Catheter-based pulmonary angiography remains the reference
computed tomography angiography (CTA) or ventilation/perfusion standard technique for the exclusion of PE. The use of this method,
(V̇ /Q̇ ) scintigraphy, compression venous ultrasonography (CUS) of the however, is infrequent due to the good diagnostic accuracy provided by

529
530 SECTION V Vascular/Pericardium

noninvasive CTA. Furthermore, the invasive nature of the test carries ≥25 mm Hg measured by right heart catheterization.21 Poor reporting
risk for procedure-related complications, especially in patients with of PAH hampers the estimation of its incidence on a global level. In
hemodynamic instability and/or respiratory failure.10 the United Kingdom, a prevalence of 97 cases per million has been
CUS is useful for detection of DVT, which is thought to be a pre- reported; in the United States the age-standardized death rate ranges
disposing condition for development of PE. However, a negative test between 4.5 and 12.3 per 100,000 persons.21 The clinical classification
does not exclude the presence of PE: in one study, CUS found DVT in of the disease has recently been updated attributed to newly recognized
only 30% of patients with proven PE.11 A proximal DVT detected by clinical entities (e.g., new gene mutations) and other conditions that
CUS in patients with isolated subsegmental PE is an adequate finding might require the establishment of new subcategories. The new clas-
to start anticoagulation therapy.4 However, an ongoing trial raised con- sification includes PAH (previously primary PAH), PAH as a result of
cerns about treating small blood clots attributed to the elevated risk of left heart disease, PAH caused by lung disease and/or hypoxia, chronic
bleeding and investigates withholding anticoagulants in patients with thromboembolic PAH and other pulmonary artery obstructions, and
subsegmental PE without cancer.11a PAH with unclear or multifactorial mechanism. The specific cause of
The prognostic value of CMR for diagnosing PE has been investi- the disease initiates several pathophysiologic mechanisms (vasoconstric-
gated in the prospective, multicenter Prospective Investigation of Pul- tion, cell proliferation, remodeling of the pulmonary arterial wall),
monary Embolism Diagnosis (PIOPED) III trial. However, the results which increase pulmonary vascular resistance and eventually lead to
were disappointing: CMR images were technically unsatisfactory in the development of right ventricular (RV) dysfunction. As pulmonary
25% of patients.12 Another study found that 30% of CMR scans were arterial pressure rises, patients experience an insidious onset of non-
inconclusive.13 Furthermore, the latter investigation reported high sen- specific symptoms, such as progressive dyspnea and exercise intolerance,
sitivity for proximal PE (97.7%–100% assessed by two readers) but which are often mistaken for signs of coronary artery disease, thereby
significantly lower sensitivity for segmental (68%–91.7%) and sub- delaying diagnosis and treatment. Although comprehensive registries
segmental PE (21.4%–33.3%). Another issue is that access to CMR is demonstrated improved prognosis,22–24 early diagnosis is of utmost
relatively limited, particularly outside of office hours. Current guidelines importance because patients experience poor quality of life.
therefore do not include CMR in the primary imaging work-up of PE. Establishing PAH currently requires numerous tests interpreted in
Nevertheless, because of its radiation-free nature CMR remains a useful concert. CMR has been suggested as a “one-stop shop” imaging tech-
imaging modality for detection of PE, especially in young women and nique for the evaluation of PAH and its impact on the right ventricle.25
children.7,14 The assessment of the pulmonary vascular and circulatory changes
There are two fundamental and complementary approaches to CMR encompasses the following CMR techniques: 2D and 3D CE-CMR,
in the diagnosis of PE. The earliest approach builds on the model of V̇ /Q̇ CMR lung perfusion, and velocity-encoded phase-contrast (PC) imaging.
scintigraphy and uses CE-CMR to evaluate pulmonary perfusion, to Radiological signs on CE-CMR indicative of PAH include the dilation
identify segmental, wedge-shaped perfusion defects in the pulmonary of the main pulmonary artery, abnormal proximal-to-distal tapering
parenchyma. In this regard, several investigations demonstrated good (“pruning”) of pulmonary vasculature, and reduction and tortuosity
agreement between CMR lung perfusion and V̇ /Q̇ scintigraphy.15–17 of the distal vessels (Fig. 45.1). CE-CMR also assists in establishing the
Another method of diagnosing acute PE on CMR is the visualization etiology of the hypertension. Findings associated with chronic throm-
of vascular alterations, which include intravascular filling defects, abrupt boembolic PAH such as recanalization and organization of thrombus
cutoff of intravascular signal and the absence of contrast material distal to reflects the underlying pathology of the disease. CE-CMR demonstrates
the obstructed artery. Furthermore, CMR allows for accurate evaluation thrombotic wall thickening and irregularity, as well as intraluminal
of flow changes in the bronchial arteries in patients with chronic PE.18 webs. Because CTA has essentially supplanted catheter angiography for
Molecular CMR with the use of specific contrast agents might also the clinical diagnosis of PE, it has quickly become the study of choice
allow for direct visualization of thrombus formation.19 Fluorine (F) for evaluation of PAH. However, CE-CMR shows similar accuracy when
CMR and α2-antiplasmin (α2AP) labeled contrast agents have been
shown to be able to detect thrombi with a diameter <0.8 mm in murine
inferior vena cava. Further development of this method in humans
might help in the identification of evolving thrombi following invasive
procedures with increased risk for thrombosis.19
Ferumoxytol is an intravenous iron preparation intended for treat-
ment of the anemia associated with chronic kidney disease. It is a
carbohydrate-coated, superparamagnetic iron oxide nanoparticle. Feru-
moxytol has been used off-label for CMR as a contrast agent for vascular
imaging. It does not contain gadolinium and therefore might be beneficial
in patients with renal impairment. The prolonged steady state of feru-
moxytol allows T1-weighted CMR to be performed under steady-state
conditions (rather than only first-pass imaging with conventional gado-
linium contrast agents). Thus ferumoxytol has been proposed to be A B
useful in patients who have difficulty with breath-holding.20
FIG. 45.1  Chronic thromboembolic pulmonary arterial hypertension.
(A) Coronal thick maximum-intensity projection contrast-enhanced car-
PULMONARY ARTERY HYPERTENSION diovascular magnetic resonance reconstruction shows cardiomegaly with
dilation of the main pulmonary artery as well as the proximal main
PAH is an enigmatic progressive condition characterized by abnormally pulmonary arteries with a reduction in caliber (pruning) in the diseased
elevated blood pressure in the pulmonary artery circulation. Normally, segmental pulmonary arteries, most prominent in the right lung (middle
the pulmonary arterial system is a low-resistance vascular bed with a lobe and lower lobe). (B) Corresponding catheter-based pulmonary angi-
mean arterial pressure of 14 ± 3 mm Hg and with an upper limit of ography confirms the contrast-enhanced magnetic resonance angiography
21 mm Hg. PAH is defined as a mean pulmonary arterial pressure of findings.
CHAPTER 45  Pulmonary Artery 531

compared with CTA in identifying the presence of chronic thrombo- gas exchange. The total area of bronchial arteries is significantly larger
embolic disease in patients with PAH.26 Furthermore, CE-CMR is the in patients with chronic thromboembolic PAH than in those with non-
desirable imaging modality in clinical scenarios such as chronic PE in thromboembolic PAH.36 Larger total area of the bronchial arteries is
pregnant women, young patients, or in case of multiple follow-up not only indicative of the presence of chronic thromboembolic PAH,
examinations. CMR lung perfusion imaging can also be used to diagnose but also correlates with improved outcomes after surgery for thrombotic
and characterize PAH. Patients with PAH typically demonstrate perfu- hypertension.36,37 Previously, the degree of bronchial arterial shunting
sion defects that are characteristically segmental and/or focal (in chronic could be measured only by an invasive contrast-medium dilution tech-
thromboembolic PAH) (Fig. 45.2A) and patchy and/or diffuse (in non- nique. However, by using velocity-encoded PC CMR of the pulmonary
thromboembolic PAH). The diagnostic accuracy of CE-CMR–generated arteries, increased bronchial arterial shunting can be identified nonin-
perfusion for the detection of chronic thromboembolic-related perfusion vasively in patients with chronic thromboembolic PAH.18 Finally, late
defects is comparable with CTA26 or conventional radionuclide scin- gadolinium enhancement at the RV insertion points of interventricular
tigraphy (Fig. 45.2B).27 Furthermore, the combined use of CMR lung septum has been shown to be associated with more advanced disease
perfusion and CE-CMR is able to distinguish chronic thromboembolic and poor prognosis.38–40
PAH from nonthromboembolic PAH with an accuracy of 90%.28 A CE-CMR and CMR perfusion techniques used for detecting acute
recent investigation demonstrated the potential of CMR lung perfusion PE are equally applicable to diagnose the anatomic changes of PAH,
for the quantification of perfusion changes in patients before and after including thrombotic wall thickening, increased pulmonary arterial
pulmonary artery endarterectomy.29 diameter, abnormal vessel tapering, and segmental or patchy perfusion
CMR assessment of the pulmonary arteries can not only accurately defects. Additionally, CMR allows for the acquisition of structural and
diagnose PAH and differentiate its cause, but also can provide functional functional changes of the heart, and provides information for systemic-
assessments, which can help guide treatment and predict response. CMR pulmonary arterial shunting. Further refinement in CMR including
can provide structural and quantitative functional analysis of the RV, vascular imaging and the incorporation of 4D-CMR in the clinical
including RV ejection fraction and mass, which are not readily obtained setting can considerably contribute to the differential diagnosis and
by echocardiography. A metaanalysis of eight studies investigated 21 monitoring of PAH without ionizing radiation exposure or iodinated
CMR parameters and found RV ejection fraction to be the strongest contrast material.41
predictor of mortality in PAH.30 CMR also allows assessment of the
interventricular septum, which bows into the left ventricle when RV
pressure exceeds left ventricular pressure.31 The amount of septal cur-
CONGENITAL VASCULAR DISORDERS
vature is linearly related to right heart pressure as described by Laplace’s A wide spectrum of congenital heart diseases (CHD) and congenital
law.32 PAH has also been associated with a vortex of blood flow in the vascular anomalies affect the pulmonary arteries. Precise anatomic
main pulmonary artery on velocity encoded PC CMR: the relative period delineation of the abnormality is essential to plan and perform opera-
of existence of the vortex showed high correlation with pulmonary tive therapy. Echocardiography is the initial imaging modality for the
artery pressure in patients with manifest PAH.33 The assessment of wall diagnosis of pulmonary vascular anomalies. However, during the past
shear stress (WSS) profile is also feasible by both velocity encoded PC years the use of other imaging modalities, such as CTA and CMR, has
CMR and 4D-CMR. In patients with PAH, significantly reduced WSS considerably increased to complement echocardiography. Numerous
was observed, which might affect the endothelial function and may studies demonstrated the efficacy of CMR for detecting complex CHD
lead to remodeling of the proximal pulmonary artery.34,35 affecting the pulmonary arterial and venous circulation and preopera-
Dilatation of the bronchial arterial system has been noted in patients tive planning for a wide range of disorders, including the spectrum of
with PAH as a collateral pathway for providing blood to the lungs for tetralogy of Fallot,42 hypoplastic pulmonary arteries,43 partial anomalous
pulmonary venous connection (Fig. 45.3),44 pulmonary artery stenosis
(Fig. 45.4),45 pulmonary artery sling (Fig. 45.5),46 and patent ductus
arteriosus (Fig. 45.6).47
Perfusion anterior Postoperative complications of surgical repair affecting the aorta and
pulmonary artery are also readily evaluated with near 100% accuracy
when compared with catheter-based pulmonary angiography and have
R even delineated vascular anomalies that were not seen at catheterization
I but were later proven at surgery.43 Over the past decade use of CMR
G
H
T

A B
FIG. 45.2  Assessment of pulmonary parenchymal perfusion in chronic
thromboembolic pulmonary arterial hypertension. (A) Coronal thin max-
imum-intensity projection contrast-enhanced cardiovascular magnetic
resonance reconstruction of the proximal right and left pulmonary arter-
ies, as well as the lung parenchyma. Note the decrease in vessel caliber
from proximal to distal and also the mosaic perfusion with evident large A B
perfusion defects (low signal-intensity areas). Large perfusion defects
in the right lower lobe and right middle lobe and smaller perfusion FIG. 45.3  Partial anomalous pulmonary venous connection (PAPVC).
defects in the right upper lobe, left upper lobe, and left lower lobe are (A) Axial noncontrast enhanced steady-state free precession image shows
shown. (B) Lung perfusion scintigraphy images of the same patient that a right upper pulmonary vein (arrows) draining to the superior vena cava.
also confirms the contrast-enhanced magnetic resonance angiography (B) Corresponding computed tomography angiography image of the
results. same patient showing the PAPVC (arrows).
532 SECTION V Vascular/Pericardium

in the workup of CHD has greatly expanded, which has required the
standardization of the used sequences. The purpose of using different
sequences encompasses the visualization of morphological variations
Ao
and the quantification of functional changes (e.g., blood flow, ven-
tricular volumes). In case of pulmonary arteries, CE-CMR allows for
exact evaluation of RV outflow tract, bifurcation of the pulmonary
RA trunk, side branches, and their relation to other structures, such as
airways. In patients with contraindications to contrast agent injection,
electrocardiogram-synchronized 3D steady-state free precession (SSFP)
acquisitions are preferred, during which blood pool appears bright.
Furthermore, CMR is the only imaging modality enabling flow measure-
ments in the right and left pulmonary artery by using velocity-encoded
A B PC cine sequence.48 Regarding the pulmonary veins, CMR has become a
FIG. 45.4  Pulmonary artery stenosis and ventricular septal defect.
reference standard imaging technique for the visualization of anomalous
(A) Axial noncontrast enhanced steady-state free precession image clearly of pulmonary vein connections and stenoses.49 Signs of pulmonary vein
depicts the narrowed pulmonary trunk (white arrows) between the aorta stenosis include narrowing of the lumen and the presence of veno-venous
(Ao) and the enlarged right atrium (RA). (B) Ventricular septal defect collaterals. Furthermore, distal to the site of narrowing, velocity-encoded
(black arrows) in the same patient. PC CMR can measure a higher flow. Velocity-encoded PC CMR has
been also proven to successfully quantify pulmonary valve insufficiency
by measuring regurgitation volumes after the surgical or percutaneous
relief of pulmonary stenosis in tetralogy of Fallot.50 Because the size
of the RV is crucial in patients who are planned to undergo pulmo-
nary valve replacement, careful evaluation of the RV volumes and RV
outflow tract (RVOT) geometry is required. By using velocity-encoded
Ao PC flow, 2D SSFP, 3D SSFP, or 3D CE-CMR these data can be obtained
Asc Ao PT in a single examination to provide a comprehensive assessment of the
PS
unique anatomy and physiology of tetralogy of Fallot.
RPA CE-CMR has also been proven useful in the evaluation of pulmonary
PS T LPA vascular anomalies such as arteriovenous malformations (AVM). Pul-
Ao
LA monary AVMs cause pulmonary-to-systemic shunting and a resultant
DAo decrease in oxygen saturation, as well as a potential conduit for para-
doxical embolism and systemic embolism/stroke. Embolization of these
A B AVMs is the current mainstay of treatment, requiring precise prepro-
cedural anatomic delineation of the anomaly or anomalies and accurate
FIG. 45.5  Pulmonary artery sling. (A) Axial contrast-enhanced cardio- assessment of the subsequent flow disturbances. The combined use of
vascular magnetic resonance reconstruction demonstrating an aberrant 3D CMR and velocity-encoded PC cine sequence allows for the detec-
left pulmonary artery (LPA) also known as pulmonary artery sling (PS),
tion of shunt location and assessment of the direction of flow, and thus
coursing between the ascending aorta (Asc Ao) and descending aorta
(DAo), posterior to the trachea (T). (B) Three-dimensional volume rendered
the magnitude of the shunt.
image of the same patients showing the PS arising from the right pul- CMR is also ideally suited for the evaluation of the univentricular
monary artery (RPA) coursing posterior to the aorta (Ao). LA, Left atrium. heart because it is able to provide detailed information on complex
anatomical configurations, particularly the large vessels and enables
flow quantification in Fontan circulations.

PULMONARY ARTERY TUMORS


Primary pulmonary artery tumors are rare malignancies with an esti-
mated incidence of 0.001% to 0.03%.51 The exact incidence of pulmonary
artery tumors is, however, unknown because not all patients who die
as a result of PE or pulmonary hypertension undergo autopsy. Pulmo-
nary artery sarcomas (PASs) are the most common primary malignancies
of pulmonary arteries. The vast majority of PASs originate from the
intimal layer of the vessel wall (intimal sarcomas) and consist of poorly
differentiated pluripotent cells with fibroblastic or myofibroblastic dif-
ferentiation.52 In some instances, cells can arise from the media of the
vessel wall (mural sarcomas) leading to the formation of leiomyosarcoma,
with a better prognosis compared with undifferentiated sarcomas.53
A B PASs predominantly show an intraluminal growth pattern and most
FIG. 45.6  Patent ductus arteriosus. (A) Sagittal noncontrast enhanced frequently involve the pulmonary trunk (85%). Other common loca-
balanced steady-state free precession image showing the patent ductus tions are the right (71%) and left (65%) pulmonary arteries, and they
Botalli (arrow) between the aorta and main pulmonary artery. (B) Coronal occasionally affect the pulmonary valve (32%) and the RVOT (10%).54
image of the same patient demonstrating the patent ductus arteriosus The diagnosis of PAS is challenging because the clinical symptoms such
(arrow). as dyspnea, chest pain, and cough can be attributable to different
CHAPTER 45  Pulmonary Artery 533

underlying medical conditions. Moreover, tumors appear as large filling


defects and can masquerade as PE on CTA and CMR images.54–56 However,
the presence of few specific imaging markers might favor the differen-
tiation of PAS. In contrast to abrupt narrowing typical to PE, sarcomas
generally occupy the entire lumen of the proximal pulmonary artery
with the potential of extravascular expansion. On CTA images PASs
can appear as low-attenuation filling defect but may also show hetero-
geneous density owing to areas of calcification, hemorrhage, or necrosis.57
Novel refinements in CTA technology using quantitative analytic meth-
odology might further promote the differentiation of PAS from PE.58 A B
The diagnostic potential of CE-CMR regarding PAS assessment was
described in several case reports with variable findings, covering the
axis from minimal to avid enhancement of the intraluminal masses.
These results were further confirmed by an investigation using multi-
parametric CMR data: hyperintense filling defects on fat-suppressed
T2-weighted and diffusion-weighted images, as well as contrast enhance-
ment were the major findings in PAS.59
Secondary pulmonary vasculature tumors can be both metastatic
and embolic, and frequently originate from breast, stomach, and lung
cancer.60 The estimated incidence rates of pulmonary tumor embolism
vary between 3% and 26% in patients with known carcinoma.60 Intra- C D
luminal expansion of the tumors might lead to total or partial blockage FIG. 45.7  Hydatid cyst. (A) Anteroposterior view of chest x-ray showing
of the pulmonary arteries with subsequent pulmonary infarction. CTA a well-defined, homogenous, spherical radio-opacity (arrows) in the right
is the reference-standard imaging method for the differentiation of supradiaphragmatic region, as well as widened right pulmonary hilum
parenchymal lung alterations related to tumors (e.g., atelectasis, pneu- (arrowheads). (B) Coronal computed tomography angiography image
monitis, and infarction), however the associated radiation exposure showing hydatid cyst in the right lung along the right pulmonary artery
remains a matter of concern. CMR lung imaging is fraught with dif- (arrows). (C) Coronal T2-weighted images, showing mass with hetero-
ficulty, both because the lungs have little proton density, and therefore geneous intermediate-to-high signal intensity. (D) Dynamic contrast-
generate little signal, and because the air within the lungs creates an enhanced cardiovascular magnetic resonance showing perfusion defect
in the lower lobe of the right lung.
extensive susceptibility artifact. However, implementation of advanced
imaging strategies over the last decade allows for more frequent use of
this technique in the clinical routine. Based on the contrast intensity
on T1-weighted and T2-weighted images, CMR is able to provide com- Echinococcus granulosus. Most commonly cysts form in the liver but in
prehensive tissue characterization, which can aid in the differentiation 20% to 30% of cases they form in the lungs.69 They can affect the
of pulmonary consolidations in a good correlation with CTA.61,62 pulmonary arteries and might lead to the development of PE.70 Hydatid
Primary cardiac tumors rarely can affect the pulmonary valve. Papil- cysts on CE-CMR appear as single or multiple, well-defined, homog-
lary fibroelastomas (PFEs) are the third most common primary cardiac enous, intermediate-to-high signal-intensity lesions and as a perfusion
tumors after myxomas and lipomas. PFEs are benign endocardial papil- defect in the lung parenchyma on CE-CMR (Fig. 45.7).
lomas mostly originating from the aortic (44.5%) or mitral valve (36.4%),
whereas the involvement of pulmonary valve is rare (8%).63 Although
they have a low tendency toward growth, the friable consistency of the
CONCLUSION
tumor aids the formation of emboli, which can cause obstruction in CMR provides precise anatomic and functional information regarding
cerebral, retinal, and pulmonary arteries.64 In most of the cases PFE the pulmonary arterial system in a single examination without iodin-
can be diagnosed by echocardiography, however if inconclusive, differ- ated contrast or exposure to ionizing radiation. Technological advances
ent CMR sequences are able to aid the diagnosis. On CMR images PFEs have enabled fast scan times while obtaining high spatial-resolution
appear as a homogenous mass, showing intermediate signal intensity images. More advanced analysis of blood flow might provide insights
on T1-weighted images and increased signal intensity on T2-weighted into the association between pulmonary artery hemodynamics and
images. Fat-saturation sequences also allow for the differentiation of vascular remodeling. Furthermore, the application of molecular CMR
PFEs from lipomas: PFEs do not show signal dropout.65,66 In cases of with specific contrast agents has the potential to diagnose a wide variety
left-sided heart valve-derived PFEs, surgical removal is promoted. of pathologies in a single noninvasive examination. Therefore in the
However, treatment management of pulmonary valve-derived PFEs is future, CMR might emerge as the examination of first choice for the
uncertain, because these tumors are less frequent and usually asymp- evaluation of the pulmonary arterial system.
tomatic. The potential for clot formation and obstruction of pulmonary
arteries might favor surgical excision of the tumor. Other benign tumors
of the pulmonary valve, such as myxomas and pulmonary valve cysts,
REFERENCES
were also reported.67,68 Hydatid cyst is a parasitic disease caused by A full reference list is available online at ExpertConsult.com
CHAPTER 45  Pulmonary Artery 533.e1

REFERENCES 21. Galie N, Humbert M, Vachiery JL, et al. 2015 ESC/ERS guidelines for the
diagnosis and treatment of pulmonary hypertension: The Joint Task
1. Raskob GE, Angchaisuksiri P, Blanco AN, et al. Thrombosis: a major Force for the Diagnosis and Treatment of Pulmonary Hypertension of
contributor to global disease burden. Arterioscler Thromb Vasc Biol. the European Society of Cardiology (ESC) and the European Respiratory
2014;34:2363–2371. Society (ERS): endorsed by: Association for European Paediatric and
2. Anderson FA Jr, Spencer FA. Risk factors for venous thromboembolism. Congenital Cardiology (AEPC), International Society for Heart and Lung
Circulation. 2003;107:I9–I16. Transplantation (ISHLT). Eur Heart J. 2016;37:67–119.
3. Cohen AT, Agnelli G, Anderson FA, et al. Venous thromboembolism 22. Thenappan T, Shah SJ, Rich S, Gomberg-Maitland M. A USA-based
(VTE) in Europe. The number of VTE events and associated morbidity registry for pulmonary arterial hypertension: 1982-2006. Eur Respir J.
and mortality. Thromb Haemost. 2007;98:756–764. 2007;30:1103–1110.
4. Konstantinides SV, Torbicki A, Agnelli G, et al. 2014 ESC guidelines on 23. Benza RL, Miller DP, Gomberg-Maitland M, et al. Predicting survival in
the diagnosis and management of acute pulmonary embolism. Eur Heart pulmonary arterial hypertension: insights from the Registry to Evaluate
J. 2014;35:3033–3069, 3069a–3069k. Early and Long-Term Pulmonary Arterial Hypertension Disease
5. Deleted in review. Management (REVEAL). Circulation. 2010;122:164–172.
6. Hutchinson BD, Navin P, Marom EM, Truong MT, Bruzzi JF. 24. Humbert M, Sitbon O, Chaouat A, et al. Survival in patients with
Overdiagnosis of pulmonary embolism by pulmonary CT angiography. idiopathic, familial, and anorexigen-associated pulmonary arterial
AJR Am J Roentgenol. 2015;205:271–277. hypertension in the modern management era. Circulation.
7. Einstein AJ, Henzlova MJ, Rajagopalan S. Estimating risk of cancer 2010;122:156–163.
associated with radiation exposure from 64-slice computed tomography 25. Nagendran J, Michelakis E. MRI: one-stop shop for the comprehensive
coronary angiography. JAMA. 2007;298:317–323. assessment of pulmonary arterial hypertension? Chest. 2007;132:2–5.
8. Sabel BO, Buric K, Karara N, et al. High-pitch CT pulmonary 26. Rajaram S, Swift AJ, Capener D, et al. Diagnostic accuracy of contrast-
angiography in third generation dual-source CT: image quality in an enhanced MR angiography and unenhanced proton MR imaging
unselected patient population. PLoS One. 2016;11:e0146949. compared with CT pulmonary angiography in chronic thromboembolic
9. Phillips JJ, Straiton J, Staff RT. Planar and SPECT ventilation/perfusion pulmonary hypertension. Eur Radiol. 2012;22:310–317.
imaging and computed tomography for the diagnosis of pulmonary 27. Rajaram S, Swift AJ, Telfer A, et al. 3D contrast-enhanced lung perfusion
embolism: a systematic review and meta-analysis of the literature, and MRI is an effective screening tool for chronic thromboembolic
cost and dose comparison. Eur J Radiol. 2015;84:1392–1400. pulmonary hypertension: results from the ASPIRE registry. Thorax.
10. Stein PD, Athanasoulis C, Alavi A, et al. Complications and validity of 2013;68:677–678.
pulmonary angiography in acute pulmonary embolism. Circulation. 28. Nikolaou K, Schoenberg SO, Attenberger U, et al. Pulmonary arterial
1992;85:462–468. hypertension: diagnosis with fast perfusion MR imaging and high-
11. Righini M, Le Gal G, Aujesky D, et al. Diagnosis of pulmonary embolism spatial-resolution MR angiography–preliminary experience. Radiology.
by multidetector CT alone or combined with venous ultrasonography 2005;236:694–703.
of the leg: a randomised non-inferiority trial. Lancet. 2008;371: 29. Schoenfeld C, Cebotari S, Hinrichs J, et al. MR Imaging-derived regional
1343–1352. pulmonary parenchymal perfusion and cardiac function for monitoring
11a.  Ottawa Hospital Research Institute. A study to evaluate the safety of patients with chronic thromboembolic pulmonary hypertension before
withholding anticoagulation in patients with PE who have a negative and after pulmonary endarterectomy. Radiology. 2016;279:925–934.
serial bilateral lower extremity ultrasound (SSPE); 2011. https:// 30. Baggen VJ, Leiner T, Post MC, et al. Cardiac magnetic resonance findings
clinicaltrials.gov/ct2/show/NCT01455818. Accessed January 26, 2018. predicting mortality in patients with pulmonary arterial hypertension: a
12. Stein PD, Chenevert TL, Fowler SE, et al. Gadolinium-enhanced magnetic systematic review and meta-analysis. Eur Radiol. 2016;26:3771–3780.
resonance angiography for pulmonary embolism: a multicenter 31. Haddad F, Guihaire J, Skhiri M, et al. Septal curvature is marker of
prospective study (PIOPED III). Ann Intern Med. 2010;152:434–443, hemodynamic, anatomical, and electromechanical ventricular
W142–W143. interdependence in patients with pulmonary arterial hypertension.
13. Revel MP, Sanchez O, Couchon S, et al. Diagnostic accuracy of magnetic Echocardiography. 2014;31:699–707.
resonance imaging for an acute pulmonary embolism: results of the 32. Roeleveld RJ, Marcus JT, Faes TJ, et al. Interventricular septal
‘IRM-EP’ study. J Thromb Haemost. 2012;10:743–750. configuration at MR imaging and pulmonary arterial pressure in
14. Li X, Samei E, Segars WP, Sturgeon GM, Colsher JG, Frush DP. pulmonary hypertension. Radiology. 2005;234:710–717.
Patient-specific radiation dose and cancer risk for pediatric chest CT. 33. Reiter G, Reiter U, Kovacs G, et al. Magnetic resonance-derived
Radiology. 2011;259:862–874. 3-dimensional blood flow patterns in the main pulmonary artery as a
15. Amundsen T, Torheim G, Kvistad KA, et al. Perfusion abnormalities in marker of pulmonary hypertension and a measure of elevated mean
pulmonary embolism studied with perfusion MRI and ventilation- pulmonary arterial pressure. Circ Cardiovasc Imaging. 2008;1:23–30.
perfusion scintigraphy: an intra-modality and inter-modality agreement 34. Truong U, Fonseca B, Dunning J, et al. Wall shear stress measured by
study. J Magn Reson Imaging. 2002;15:386–394. phase contrast cardiovascular magnetic resonance in children and
16. Fink C, Bock M, Puderbach M, Schmahl A, Delorme S. Partially parallel adolescents with pulmonary arterial hypertension. J Cardiovasc Magn
three-dimensional magnetic resonance imaging for the assessment of Reson. 2013;15:81.
lung perfusion–initial results. Invest Radiol. 2003;38:482–488. 35. Schafer M, Kheyfets VO, Schroeder JD, et al. Main pulmonary arterial
17. Yilmaz E, Akkoclu A, Degirmenci B, et al. Accuracy and feasibility of wall shear stress correlates with invasive hemodynamics and stiffness in
dynamic contrast-enhanced 3D MR imaging in the assessment of lung pulmonary hypertension. Pulm Circ. 2016;6:37–45.
perfusion: comparison with Tc-99 MAA perfusion scintigraphy. Clin 36. Shimizu H, Tanabe N, Terada J, et al. Dilatation of bronchial arteries
Radiol. 2005;60:905–913. correlates with extent of central disease in patients with chronic
18. Ley S, Kreitner KF, Morgenstern I, Thelen M, Kauczor HU. thromboembolic pulmonary hypertension. Circ J. 2008;72:1136–1141.
Bronchopulmonary shunts in patients with chronic thromboembolic 37. Kauczor HU, Schwickert HC, Mayer E, Schweden F, Schild HH, Thelen
pulmonary hypertension: evaluation with helical CT and MR imaging. M. Spiral CT of bronchial arteries in chronic thromboembolism. J
AJR Am J Roentgenol. 2002;179:1209–1215. Comput Assist Tomogr. 1994;18:855–861.
19. Temme S, Grapentin C, Quast C, et al. Noninvasive imaging of early 38. Swift AJ, Rajaram S, Condliffe R, et al. Diagnostic accuracy of
venous thrombosis by 19F magnetic resonance imaging with targeted cardiovascular magnetic resonance imaging of right ventricular
perfluorocarbon nanoemulsions. Circulation. 2015;131:1405–1414. morphology and function in the assessment of suspected pulmonary
20. Hope MD, Hope TA, Zhu C, et al. Vascular imaging with ferumoxytol as hypertension results from the ASPIRE registry. J Cardiovasc Magn Reson.
a contrast agent. AJR Am J Roentgenol. 2015;205:W366–W373. 2012;14:40.
533.e2 SECTION V Vascular/Pericardium

39. Blyth KG, Groenning BA, Martin TN, et al. Contrast enhanced- 54. Restrepo CS, Betancourt SL, Martinez-Jimenez S, Gutierrez FR. Tumors
cardiovascular magnetic resonance imaging in patients with pulmonary of the pulmonary artery and veins. Semin Ultrasound CT MR.
hypertension. Eur Heart J. 2005;26:1993–1999. 2012;33:580–590.
40. Freed BH, Gomberg-Maitland M, Chandra S, et al. Late gadolinium 55. Wong HH, Gounaris I, McCormack A, et al. Presentation and
enhancement cardiovascular magnetic resonance predicts clinical management of pulmonary artery sarcoma. Clin Sarcoma Res. 2015;5:3.
worsening in patients with pulmonary hypertension. J Cardiovasc Magn 56. El-Sayed Ahmed MM, Aftab M, Al-Najjar RM, de la Cruz KI, Benjamin
Reson. 2012;14:11. RS, Hallman CH. Pulmonary artery sarcoma mimicking pulmonary
41. Dyverfeldt P, Bissell M, Barker AJ, et al. 4D flow cardiovascular magnetic embolism. Tex Heart Inst J. 2014;41:515–517.
resonance consensus statement. J Cardiovasc Magn Reson. 2015;17:72. 57. Yi CA, Lee KS, Choe YH, Han D, Kwon OJ, Kim S. Computed
42. Kutty S, Kuehne T, Gribben P, et al. Ascending aortic and main tomography in pulmonary artery sarcoma: distinguishing features from
pulmonary artery areas derived from cardiovascular magnetic resonance pulmonary embolic disease. J Comput Assist Tomogr. 2004;28:34–39.
as reference values for normal subjects and repaired tetralogy of Fallot. 58. Chang S, Hur J, Im DJ, et al. Dual-energy CT-based iodine quantification
Circ Cardiovasc Imaging. 2012;5:644–651. for differentiating pulmonary artery sarcoma from pulmonary
43. Geva T, Greil GF, Marshall AC, Landzberg M, Powell AJ. Gadolinium- thromboembolism: a pilot study. Eur Radiol. 2016;26:3162–3170.
enhanced 3-dimensional magnetic resonance angiography of pulmonary 59. Liu M, Luo C, Wang Y, et al. Multiparametric MRI in differentiating
blood supply in patients with complex pulmonary stenosis or atresia: pulmonary artery sarcoma and pulmonary thromboembolism: a
comparison with x-ray angiography. Circulation. 2002;106:473–478. preliminary experience. Diagn Interv Radiol. 2017;23:15–21.
44. Valente AM, Sena L, Powell AJ, Del Nido PJ, Geva T. Cardiac magnetic 60. Roberts KE, Hamele-Bena D, Saqi A, Stein CA, Cole RP. Pulmonary
resonance imaging evaluation of sinus venosus defects: comparison to tumor embolism: a review of the literature. Am J Med. 2003;115:228–232.
surgical findings. Pediatr Cardiol. 2007;28:51–56. 61. Gaeta M, Minutoli F, Ascenti G, et al. MR white lung sign: incidence and
45. Myerson SG. Heart valve disease: investigation by cardiovascular significance in pulmonary consolidations. J Comput Assist Tomogr.
magnetic resonance. J Cardiovasc Magn Reson. 2012;14:7. 2001;25:890–896.
46. Leonardi B, Secinaro A, Cutrera R, et al. Imaging modalities in children 62. Barreto MM, Rafful PP, Rodrigues RS, et al. Correlation between
with vascular ring and pulmonary artery sling. Pediatr Pulmonol. computed tomographic and magnetic resonance imaging findings of
2015;50:781–788. parenchymal lung diseases. Eur J Radiol. 2013;82:e492–e501.
47. Broadhouse KM, Finnemore AE, Price AN, et al. Cardiovascular magnetic 63. Generali T, Tessitore G, Mushtaq S, Alamanni F. Pulmonary valve
resonance of cardiac function and myocardial mass in preterm infants: a papillary fibroelastoma: management of an unusual, tricky pathology.
preliminary study of the impact of patent ductus arteriosus. J Cardiovasc Interact Cardiovasc Thorac Surg. 2013;16:88–90.
Magn Reson. 2014;16:54. 64. Gowda RM, Khan IA, Nair CK, Mehta NJ, Vasavada BC, Sacchi TJ.
48. Caputo GR, Kondo C, Masui T, et al. Right and left lung perfusion: in Cardiac papillary fibroelastoma: a comprehensive analysis of 725 cases.
vitro and in vivo validation with oblique-angle, velocity-encoded cine Am Heart J. 2003;146:404–410.
MR imaging. Radiology. 1991;180:693–698. 65. Jahnke C, Hamdan A, Fleck E, Paetsch I. Tissue characterization of a
49. Valsangiacomo Buechel ER, Grosse-Wortmann L, Fratz S, et al. suspected aortic valve fibroelastoma with cardiac magnetic resonance
Indications for cardiovascular magnetic resonance in children with imaging. Circ Cardiovasc Imaging. 2008;1:87–88.
congenital and acquired heart disease: an expert consensus paper of the 66. O’Donnell DH, Abbara S, Chaithiraphan V, et al. Cardiac tumors: optimal
Imaging Working Group of the AEPC and the Cardiovascular Magnetic cardiac MR sequences and spectrum of imaging appearances. AJR Am J
Resonance Section of the EACVI. Eur Heart J Cardiovasc Imaging. Roentgenol. 2009;193:377–387.
2015;16:281–297. 67. Pemberton J, Raudkivi P. Right ventricular myxoma causing pulmonary
50. Wald RM, Redington AN, Pereira A, et al. Refining the assessment of outflow tract obstruction. Interact Cardiovasc Thorac Surg.
pulmonary regurgitation in adults after tetralogy of Fallot repair: should 2012;14:362–363.
we be measuring regurgitant fraction or regurgitant volume? Eur Heart J. 68. Ghawi H, Polimenakos AC, Gonzalez I, Abdulla R, Bokowski J. Giant
2009;30:356–361. pulmonary valve cyst in a 7-year-old child. Pediatr Cardiol.
51. Bleisch VR, Kraus FT. Polypoid sarcoma of the pulmonary trunk: analysis 2012;33:656–658.
of the literature and report of a case with leptomeric organelles and 69. Kunst H, Mack D, Kon OM, Banerjee AK, Chiodini P, Grant A. Parasitic
ultrastructural features of rhabdomyosarcoma. Cancer. 1980;46: infections of the lung: a guide for the respiratory physician. Thorax.
314–324. 2011;66:528–536.
52. Burke AP, Virmani R. Sarcomas of the great vessels. A clinicopathologic 70. Mnari W, Zrig A, Maatouk M, Golli M. Acute pulmonary embolism due
study. Cancer. 1993;71:1761–1773. to hydatid cysts. Int J Infect Dis. 2014;25:142–144.
53. Huo L, Moran CA, Fuller GN, Gladish G, Suster S. Pulmonary artery
sarcoma: a clinicopathologic and immunohistochemical study of 12 cases.
Am J Clin Pathol. 2006;125:419–424.
46 
The Pericardium: Anatomy and
Spectrum of Disease
Jay S. Leb and Susie N. Hong

The pericardium is an important structure in the evaluation of patients moderately invasive, and the field of view remains limited to the available
with cardiovascular disease. Understanding pericardial anatomy and acoustic windows.7
the complex hemodynamics associated with pericardial pathology is
critical in assessing the pericardium and its impact on cardiovascular Computed Tomography
function. Unfortunately, clinical evaluation through history and physical Cardiac CT and CMR provide a larger field of view than echocardiog-
examination, clinical laboratory tests, and electrocardiograms (ECG) raphy, enabling visualization of the entire pericardium and the sur-
may be nonspecific and overlap with other clinical syndromes in patients rounding structures, providing excellent anatomic definition without
with pericardial disease. Advanced noninvasive imaging, including limitation to specific windows.2,8,9 Although optimal imaging of the
echocardiography, cardiac chest computed tomography (CT), and car- pericardium with CT and CMR requires ECG gating to minimize motion
diovascular magnetic resonance (CMR), is vital in the evaluation and artifact, standard chest CT and nongated magnetic resonance imaging
diagnosis of pericardial disease. When combined with a high degree of (MRI) performed for other indications often reveal most pericardial
clinical suspicion, these modalities help advance our understanding abnormalities, particularly pericardial effusions or calcifications.3 Cardiac
pericardial pathophysiology and, ultimately, guide clinicians to optimal CT and CMR may be useful when TTE findings are nondiagnostic,
treatment and therapies. particularly for assessment of pericardial thickness and evaluation of
pericardial constriction, loculated pericardial effusions, hematomas,
and/or pericardial masses.5,9,10
IMAGING MODALITIES A specific advantage of CT (as compared with CMR) is its ability
Several noninvasive imaging modalities are available to assess the peri- to identify pericardial calcification, a frequent, but not obligatory, finding
cardium, with each modality having its strengths and limitations. As a in constrictive pericarditis.8 Cardiac CT with multidetector scanners
result of the complex nature of the pericardium and its impact on and ECG synchronized data acquisition is now widely available, offering
cardiovascular hemodynamics, multimodality imaging is often required the advantage of very short scan times with limited cardiac and respira-
to completely evaluate the pericardium. tory motion artifact.11,12 Additionally, CT provides excellent spatial
resolution, as a result of the use of multidetector, high-resolution volu-
Chest X-Ray metric acquisition. Retrospective ECG-gated CT may also provide rea-
Chest x-radiography is often the first imaging modality that may suggest sonable temporal resolution to assess interventricular septal motion in
pericardial disease and prompt further testing. An enlarged cardiac the evaluation of constrictive physiology, although this is better assessed
silhouette may suggest the presence of a pericardial effusion, although by TTE or real-time CMR.13
this finding has limited sensitivity and poor specificity.1 Additionally, Disadvantages of CT include use of ionizing radiation, the need for
the chest x-ray (CXR) may detect pericardial calcifications and raise intravenous iodinated contrast for adequate blood–tissue contrast, and
the possibility of constrictive pericarditis; however, calcification is not limited hemodynamic assessment. Additionally, CT images may not
necessary for pericardial constriction and, in fact, is frequently absent adequately discriminate pericardial fluid from thickened pericardial
in clinical presentations of constriction.2–4 tissue.8

Echocardiography Cardiovascular Magnetic Resonance Imaging


Direct visualization of the heart through transthoracic echocardiography CMR provides a comprehensive evaluation of the pericardium, both
(TTE) is the most common noninvasive cardiovascular imaging modal- anatomically and hemodynamically, without ionizing radiation. CMR
ity for the evaluation of the heart and pericardium. Two-dimensional has the ability to obtain morphological, functional, and hemodynamic
(2D) TTE with Doppler examination is an excellent, readily available, information in a single examination, which is frequently required
portable tool for the identification and assessment of pericardial disease. when assessing pericardial disease and its impact on cardiovascular
Real-time TTE can detect abnormal cardiac function and hemody- function. Balanced steady-state free precession (bSSFP) cine imaging
namic compromise secondary to underlying pericardial disease, includ- is used to evaluate left ventricular (LV) and right ventricular (RV)
ing pericardial effusions and tamponade physiology. However, patient motion and can assess the impact of pericardial disease on overall
body habitus, chest trauma, and/or poor acoustic windows attributed cardiac function. CMR also provides advantages over CT and echo-
to underlying lung disease (e.g., emphysema) may result in subop- cardiography in its ability to characterize the contents of pericardial
timal views and limit TTE in visualizing the entire pericardium.5,6 effusions and pericardial masses through the use of T1-weighted and
Pericardial thickness is also not reliably assessed by TTE. Although T2-weighted imaging, first-pass perfusion, and late gadolinium enhance-
transesophageal echocardiography (TEE) may be more accurate, it is ment (LGE).6,14 Additionally, real-time imaging by CMR may give vital

534
CHAPTER 46  The Pericardium: Anatomy and Spectrum of Disease 535

hemodynamic assessments regarding cardiac motion and inflow pat- compliance; (3) balancing RV and LV output over several cardiac cycles
terns while the patient is free-breathing, allowing for the identification through diastolic and systolic interactions; (4) buffering of changes in
of constrictive and/or tamponade physiology. Multiplanar and multi- chamber filling and output; (5) assisting atrial filling via more negative
sequence CMR can be used to characterize the extent and composi- pericardial pressure during ventricular ejection; (6) limitation of acute
tion of pericardial abnormalities and real-time CMR sequences allow dilatation; (7) minimizing friction between cardiac chambers and sur-
for functional assessment, including chamber compression caused by rounding structures; and (8) providing an anatomic and immunologic
ventricular interdependence and interventricular septal motion.2,15,16 barrier to inflammation and infection from contiguous structures such
Administration of conventional extracellular gadolinium (Gd)-based as the lungs.14,19 However, the presence of an intact pericardium may
contrast agents may assist in detecting of pericardial inflammation or be detrimental in clinical situations in which fluid rapidly fills the peri-
enhancing masses. cardial space, resulting tamponade physiology.

Imaging Findings of Normal Anatomy


NORMAL PERICARDIAL ANATOMY The normal pericardium is seen as a very thin linear density surround-
The pericardium is an elastic dual-layered fibroserous membrane that ing the heart. Discrimination of the pericardium from the myocardium
surrounds nearly the entire heart and extends superiorly to the origins by noninvasive imaging requires the presence of interposed epicardial
of the great vessels.17 It consists of an outer fibrous component and a fat or the presence of pericardial fluid with associated enlargement of
dual-layered inner serous sac. The tough fibrous outer pericardium is the pericardial space.18 The thickness of the epicardial fat is increased
loosely attached to the sternum and costal cartilage (anteriorly) and in patients with obesity and/or diabetes and has been found to increase
the proximal great vessels (superiorly), and is more firmly attached to the risk of coronary atherosclerosis.26–28 The pericardium is best visual-
the central tendon of the diaphragm (inferiorly).18,19 The serous peri- ized over the right ventricle and, as a result of less epicardial and peri-
cardium consists of an outer parietal layer, which is intimately adherent cardial fat to provide adjacent tissue contrast, may not be well visualized
to the inside of the fibrous pericardium, and commonly referred to as around the left ventricle.8 Technical factors, such as spatial and temporal
the pericardium. The inner visceral serosal layer is adjacent to the heart resolution, may affect pericardial thickness measurements because the
(but not directly attached), covers the epicardial fat and vessels, and normal pericardium is a thin irregularly shaped sac that moves with
typically referred to as the epicardium. A film of clear pericardial fluid cardiac and respiratory motion, affecting the pericardial recesses to
(~15–50 mL) normally separates the two serosal surfaces.3,19 The small varying degrees. The superior aortic and left pulmonic recesses are less
amount of fluid minimizes the friction between the two layers as the impacted by cardiac motion, whereas the inferior aortic recess, left
heart expands and contracts. The serosal layers, consisting of the visceral pulmonary venous recess, and right pulmonic recess border the LA or
and parietal pericardium, merge at two complex lines of reflection: one left ventricle, and are often blurred because of cardiac motion.29
at the base of the aorta and pulmonary trunk, and the other at the The maximum normal pericardial thickness measured through
insertions of the superior and inferior vena cavae and the four pulmo- advanced noninvasive imaging is typically 2 mm, which is slightly
nary veins.17 The entire pericardium generally lies between variable greater than normal cadaveric pericardial thickness, which measures
amounts of epicardial and pericardial adipose tissue.8 up 1 mm.30–32 This difference is primarily caused by the inherent limi-
The pericardial cavity is a complex space consisting of the pericardial tations of spatial and temporal resolution of CT and CMR. A possible
cavity proper and interconnecting cul-de-sacs known as sinuses, which pitfall in determining pericardial thickness by CT is that trace pericardial
are further subdivided and connected to multiple recesses.13,20,21 Because effusions, as well as partial volume effects, may produce the appearance
of the merging of the serosal layers at two distinct points, the reflections of focal thickening.33 These imaging effects, along with normal intrinsic
of the serous pericardium become arranged as two complex tubes; one inhomogeneity in pericardial thickness, produce significant variations
enclosing the base of the aorta and main pulmonary artery and the in pericardial thickness, and should not be mistaken for disease. The
other enclosing the vena cavae and the four pulmonary veins.22 The superior aortic recess of the transverse pericardial sinus, which sur-
transverse sinus is the space between these two pericardial tubes and rounds the ascending aorta, may be mistaken for an aortic dissection
lies superior to the left atrium (LA) and posterior to the ascending or lymphadenopathy.21,24 The oblique pericardial sinus, which is located
aorta and main pulmonary artery. The transverse sinus connects to the behind the LA, may simulate abnormalities in the esophagus, descending
following four recesses: superior aortic, inferior aortic, left pulmonic, thoracic aorta, and subcarinal and bronchopulmonary lymph nodes.23,25
and right pulmonic. The cul-de-sac delineated by the inferior border Recognizing the appearance of these normal structures is important to
of the tube, surrounding the venous structures, is the oblique sinus. avoid mistaking them for mediastinal disease.21,25
The oblique sinus is an inverted U-shaped space which lies posteriorly CMR, with its superior tissue contrast, is an excellent modality to
to the LA and includes the posterior pericardial recess. The pericardial evaluate the heart and pericardium. It is able to differentiate between
cavity proper includes the postcaval, left pulmonary venous, and right small amounts of pericardial fluid and actual thickening. With CMR,
pulmonary venous recesses.13 The transverse sinus is the space between the normal pericardium appears as a linear low-intensity signal between
these two pericardial tubes and lies superior to the LA and posterior the high-intensity mediastinal and sub-epicardial fat. It is best visual-
to the ascending aorta and main pulmonary artery. The transverse sinus ized during systole (Fig. 46.1).32 bSSFP cine imaging is used to evaluate
connects to the following four recesses: superior aortic, inferior aortic, cardiac motion and can be helpful in detecting pericardial effusions.
left pulmonic, and right pulmonic recesses. These recesses are usually Black-blood T1-weighted spin echo sequences, such as double inver-
linear in appearance and become more band-like and rounded as they sion recovery fast spin echo, are commonly performed to evaluate the
accumulate with fluid. Appreciating the normal appearance of these pericardial anatomy thickness and morphology. T2-weighted spin echo
sinuses and recesses is important because these spaces can be misin- sequences, usually with a short tau inversion recovery sequence to null
terpreted as adenopathy, dissection, or an abnormality of an adjacent the signal from fat (referred to as a triple inversion spin echo sequence),
mediastinal structure.13,23–25 is used to depict difference between pericardial thickening and small
Although the pericardium is not required for normal cardiac func- amounts of fluid. Normal pericardium will demonstrate low signal
tion, it has several homeostatic roles. These include (1) limitation of on both T1-weighted and T2-weighted sequences, whereas pericardial
intrathoracic cardiac displacement; (2) maintenance of normal ventricular fluid will be high signal on the T2-weighted imaging.8,34 Pericardial
536 SECTION V Vascular/Pericardium

A B
FIG. 46.1  (A) Axial T1-weighted spin echo image of the normal pericardium (arrows). (B) Steady-state free
precession four-chamber view demonstrating normal anterior pericardium (arrows).

inflammation may also demonstrate increased signal on T2-weighted attached with or without a peduncle to the pericardium.19,37 They are
images, but will commonly exhibit abnormal enhancement on the LGE commonly found at the right anterior cardiophrenic angle but can be
imaging, helping to differentiate it from a small pericardial effusion. located throughout the mediastinum.8,19 Most patients are asymptomatic,
Real-time cine gradient echo imaging in the short axis during dynamic with lesions detected incidentally on CXR or other chest imaging.34
breathing allows for the detection of ventricular coupling in patients However, they may be associated with chest pain, dyspnea, cough, or
with suspected constrictive pericarditis. T1-weighted gradient echo arrhythmias, likely as a result of compression of adjacent structures,
myocardial tagged cine can be useful to detect the presence or absence and may require aspiration or removal.19
of epicardial–pericardial slippage in cases with suspected fibrinous adhe-
sions exist between pericardial layers. Phase contrast sequences using Imaging Findings of Pericardial Cysts
velocity encoded gradients can be used to assess for diastolic dysfunction Pericardial cysts may present as a mass or abnormal contour adjacent
and pulmonary venous flow patterns, which are abnormal in patients to the cardiac silhouette on radiographic imaging. The location and
with restrictive physiology.2,15,35 incidental nature of the lesion will allow the imager to suspect the
presence of this benign finding. Cross-sectional imaging of a suspected
PERICARDIAL DISEASES pericardial cyst will often distinguish it from a mediastinal tumor, hema-
toma, or loculated pericardial effusion.19 The CT and CMR appearance
Pericardial Defects of the pericardial cyst is generally a nonenhancing, well-circumscribed,
Congenital pericardial defects are rare and uncommon. A spectrum of ovoid structure adjacent to the pericardium. The imaging characteris-
abnormalities exists, ranging from small defects in various locations to tics of the contents are usually similar to water and will demonstrate
complete bilateral absence of the pericardium (extremely rare).19 The low attenuation on CT, as well as low signal intensity on T1-weighted
most common defect is an absence of the entire left side of the peri- images, with homogeneous high intensity on T2-weighted images by
cardium.36 Pericardial agenesis may be associated with congenital heart CMR (Fig. 46.2).38 A cyst may occasionally contain highly proteinaceous
disease in a minority of cases, including atrial septal defect, patent ductus fluid that has high signal intensity on T1-weighted images.38 Intracystic
arteriosus, and tetralogy of Fallot.34 Although pericardial defects are septae may be observed, especially after the administration of contrast,
generally found incidentally, smaller defects of the left pericardium may but cyst contents do not enhance with administration of standard Gd-
potentially cause herniation and entrapment of portions of the heart.19 based contrast agents.9,39 Although typically benign, cysts often change
size and shape with respiratory motion or body position.18
Imaging Findings of Pericardial Defects
CXR findings are nonspecific and may reveal evidence of cardiac dis- Pericarditis
placement.19 Frontal CXR will demonstrate cardiac levorotation, with Pericarditis refers to any cause of pericardial inflammation, which may
loss of the right heart border, and prominence of the pulmonary artery be local or systemic, and is the most common disorder involving the
contour with complete absence of the left pericardium. Evaluation for pericardium.40–42 The term pericarditis is a generalized term used for
pericardial defects on CT and CMR may be difficult because the normal various forms of inflammation of the pericardium, with a spectrum
pericardium is frequently not well visualized over portions of the heart, of clinical presentations. These include (1) acute pericarditis without
particularly along the left heart border.8 The diagnosis may be made clinical evidence of a significant effusion; (2) pericardial inflammation
on cross-sectional imaging by assessing for indirect signs of pericardial with an effusion, but without major hemodynamic compromise; (3)
absence, including prominent cardiac displacement, interposition of cardiac tamponade, and (4) constrictive pericarditis.19,43 Categories of
lung tissue between the aorta and the main pulmonary artery, or between disease associated with pericardial disease are broad and include infection
the inferior cardiac border and the diaphragm.10,19 Excessive LV apical (e.g., bacterial or viral), radiation, autoimmune diseases, drug toxic-
motion on CMR cine images may also suggest the diagnosis. ity, inflammatory processes of contiguous structures (e.g., myocarditis
and myocardial infarction), metabolic disorders (e.g., uremia, dialysis,
Pericardial Cysts myxedema), trauma (including iatrogenic causes), and neoplasms, as
A pericardial cyst is a congenital anomalous fluid collection arising well as idiopathic etiologies.19 Most cases of infectious pericarditis are
from the pericardium and develops from persistent defective embryo- of viral causes. However, nearly any infectious disease (bacterial, fungal,
logical pericardial remnants.37 They are usually unilocular, smooth, parasitic, etc.) may involve the pericardium. In the Western hemisphere,
thin-walled structures filled with clear transudative fluid and may be most cases of acute pericarditis are idiopathic and presumed to be
CHAPTER 46  The Pericardium: Anatomy and Spectrum of Disease 537

RA RA

LA LA

A B
FIG. 46.2  (A) Axial T1-weighted image demonstrating a pericardial cyst (arrow) with intracystic septae with
low intensity signal. (B) Axial T2-weighted imaging of pericardial cyst (arrow), demonstrating homogenous
high intensity signal. LA, Left atrium; RA, right atrium.

viral.44,45 Iatrogenic pericarditis from prior cardiac surgery or radiation is Purulent pericarditis is an infection of the pericardial space associ-
a major cause for pericarditis in the developed countries.46,47 Tuberculosis ated with purulent pericardial fluid. Fortunately, purulent pericarditis
related pericarditis is still a common entity in developing countries.45 is less common since the advent of antibiotics.51 It may be caused by
Signs and symptoms of acute pericarditis include chest pain (which is bacterial or fungal pathogens, with Staphylococcus aureus being the
often pleuritic and positional in nature), a pericardial friction rub on most common.52,53 When purulent pericardial infection is suspected,
auscultation, and widespread ST elevation on the electrocardiogram.43 drainage, combined with intravenous antibiotic therapy, is generally
required.54
Imaging Findings of Pericarditis Cardiac tamponade results in life-threatening hemodynamic com-
A pericardial effusion may be present with pericarditis, although its promise via external compression of the heart by accumulating pericardial
absence does not exclude the diagnosis, particularly in cases of acute fluid.41,42,55 The accumulation of pericardial fluid surrounding the heart
pericarditis. CT or CMR may identify pericardial thickening, as well as causes cardiac chamber compression leading to compromise of venous
pericardial effusions. However, differentiation between pericardial thick- systemic return and cardiac output. It can be caused by pericardial
ening and small effusions on echocardiography or CT may require disease of any etiology. Progression to cardiac tamponade is related to
further imaging.3,8,48 High intensity signal on T2-weighted imaging and the following: (1) the rate of fluid accumulation in the pericardial space,
pericardial enhancement after contrast administration by CMR suggest (2) the ability of the pericardium to stretch (pericardial compliance),
pericardial inflammation (Fig. 46.3), which may be secondary to infec- and (3) the ability to increase venous pressure to support right heart
tion or neoplastic involvement.3,10 However, a lack of pericardial enhance- filling.19 Because the pericardium has limited short-term capacity to
ment in the face of a medium or large pericardial effusion suggests a stretch, rapidly accumulating pericardial fluid collections (e.g., hemor-
chronic, longstanding effusion. Metastatic disease or infection of the rhage) can produce tamponade at relatively low volumes (~250 mL).
pericardium may also cause pericardial nodularity and thickening.49 On the other hand, more slowly accumulating effusions may be accom-
CMR enables detection of pericardial thickening, effusion, and peri- modated by pericardial stretching over time, as in the case of hypothy-
cardial contrast enhancement.3,48 roidism. Large, slowly accumulating pericardial effusions (greater than
1–2 L) may be tolerated with little or no hemodynamic compromise.41
Pericardial Effusion The symptoms of tamponade are nonspecific, so rapid diagnosis through
Pericardial effusions are excess fluid accumulations in the pericardial clinical assessment and early imaging, typically by TTE, is required.
sac. Accumulation may be attributed to a variety of causes, with most Prompt treatment is typically required through drainage of the effusion,
accumulations arising from the visceral pericardium.19 Types of effu- most commonly by pericardiocentesis. However, a surgical approach
sions include simple fluid (either related to systemic fluid retention or with a pericardial window/pericardiectomy may be indicated when the
underlying inflammation), hemopericardium, or rarely, chylopericar- effusion is loculated, recurrent, or associated with a malignancy.42
dium.19,42 Hemopericardium may consist of either frank blood (e.g.,
direct trauma, iatrogenic perforation, or aortic dissection) or a bloody Imaging Findings of Pericardial Effusion
effusion (e.g., uremia or tuberculosis). Pericardial effusions related to Pericardial effusions are frequently detected in subjects with suspected
a myocardial infarct may develop within days in response to transmural cardiac or pericardial disease, as well as incidentally when imaging is
infarct, or later (1 week to several months) related to underlying inflam- performed for other indications. On CT, common transudative effu-
mation, secondary to an immune response (Dressler syndrome). Similarly, sions have attenuation similar to water, usually measuring less than 20
pericardial effusions after cardiac surgery can be seen within days or Hounsfield units. Attenuation higher than water suggests more complex,
later (1 week to 2 months, known as postpericardiotomy syndrome). more proteinaceous fluid, as in malignancy, hemopericardium, purulent
Infectious or postinfectious pericardial effusions often resolve sponta- exudates, or myxedema.10 On CMR, transudative effusions will demon-
neously but may continue as chronic relapsing pericarditis or progress strate water characteristics and will demonstrate high signal intensity
to constrictive pericarditis. Myopericarditis occurs when an infectious on T2-weighted imaging and low signal on T1-weighted imaging.3,6,10
or inflammatory process involves both the pericardium and the myo- Transudative pericardial effusions are often even brighter than epicardial
cardium simultaneously. Malignant effusions are more commonly caused fat on gradient echo images.18 More proteinaceous effusions will tend to
by metastatic disease or local invasion rather than primary neoplastic show intermediate signal intensity on both T1-weighted and T2-weighted
disease of the pericardium.50 images and lower signal intensity than blood in the ventricular cavities
538 SECTION V Vascular/Pericardium

A B

C
FIG. 46.3  (A) Axial T2-weighted imaging in a short-axis (SAX) view demonstrating bright circumferential
pericardial enhancement, consistent with fluid/edema. (B) Phase sensitive inversion recovery sequence with
fat-saturated imaging in SAX and (C) four-chamber views demonstrating significant pericardial late gadolinium
enhancement (LGE). Findings of both pericardial LGE and bright pericardial T2-weighted imaging are consistent
with pericardial inflammation and patient’s diagnosis of viral pericarditis.

A B
FIG. 46.4  Large circumferential pericardial effusion in a patient found to have underlying lymphoma
(A) balanced steady-state free precession imaging with high signal intensity of pericardial effusion.
(B) T1-weighted imaging with low-to-intermediate signal intensity as a result of underlying malignant pericardial
effusion.

on bSSFP cine imaging (Fig. 46.4).10,34,37 In hemorrhagic pericardial effu- thrombi may have a dark rim and low signal intensity foci, which may
sions, the appearance of blood will depend on the age of the collection.3 represent areas of fibrosis, calcification, or hemosiderin deposition.58,59
A fresh hematoma will appear homogenously bright on T1-weighted and The size of pericardial effusions can be readily assessed quantitatively
T2-weighted images, whereas older collections show the effects of T1 by CMR using volumetric methods.38
and T2 shortening caused by methemoglobin formation and generally Because cardiac tamponade may occur with pericardial effusions
show heterogeneous signal intensity, with areas of high signal intensity of varying sizes as well as with loculated accumulations, the diagnosis
on both T1-weighted and T2-weighted images.3,10,56,57 Chronic organized of tamponade should be considered even for small effusions if they
CHAPTER 46  The Pericardium: Anatomy and Spectrum of Disease 539

have accumulated rapidly and in cases in which the pericardial effusion effusion.63 Effusive–constrictive disease may be the result of any of the
or hematoma is not readily apparent in some of the standard views. many causes of pericarditis, particularly radiation therapy.66 It is best
TTE with Doppler interrogation is the imaging method most commonly demonstrated via right heart catheterization at the time of pericardio-
used to detect pericardial effusions, assess their hemodynamic signifi- centesis to assess for residual elevations in right atrial and in ventricular
cance, and guide therapeutic pericardiocentesis.60 TTE features of tam- diastolic pressures after normalization of intrapericardial pressure.63
ponade include accentuated respiratory variation in transmitral (>25%) Although this syndrome frequently leads to persistent pericardial con-
and transtricuspid (>50%) Doppler inflow velocities, diastolic collapse striction that may ultimately require pericardiectomy, spontaneous
of the right atrium, LA, and/or RV free wall, and inferior vena cava resolution of idiopathic cases has been reported.66
dilation.42,61,62 However, suboptimal views may limit the usefulness of
TTE in certain individuals, particularly after cardiac surgery, for locu- Imaging Findings of Constrictive Pericarditis
lated pericardial effusions and hematomas.5,6 Although the role of CT The CXR is neither sensitive nor specific for diagnosing constrictive
and CMR is limited in evaluating for tamponade, findings that may be pericarditis. Pericardial calcification is commonly detected in constric-
present to suggest tamponade physiology include deformity of the cardiac tion; is best visualized on the lateral view, although this finding is not
chambers, particularly the right atrium and ventricle, flattening of the diagnostic for clinical constriction4; and is best appreciated by CT.
interventricular septum, compression of the coronary sinus, and disten- Pericardial calcifications can also occur in the absence of physiologic
tion of the superior vena cava and intraabdominal inferior vena cava.42,61,62 constriction, and constriction may be present without pericardial cal-
cifications. Pericardial calcification is often associated with idiopathic
Constrictive Pericarditis disease and is an independent predictor of increased perioperative
Constrictive pericarditis is usually caused by chronic fibrous thickening mortality with pericardiectomy.4
and scarring of the pericardium with consequent loss of its normal Pericardial thickness is not reliably assessed by echocardiography
elasticity and compliance that impedes diastolic cardiac filling.19,42 Several but can be readily determined using CT and CMR. Similar to pericardial
forms exist, including the acute inflammatory, effusive–constrictive, calcification, the presence of pericardial thickening by itself does not
and adhesive–constrictive pericarditis. Adhesive pericarditis is attributed indicate constriction.38 Additionally, in a significant minority of cases
to the development of fibrous adhesions between the two layers of with surgically proven constriction, the pericardial thickness is normal.67
pericardium. The effusive–constrictive and adhesive–constrictive forms When pericardial thickening is present in constriction, it may be local-
are both related to chronic inflammation of the pericardium. The ized, commonly along the right side of the heart. Nonspecific findings
effusive–constrictive pericarditis is a relatively uncommon syndrome suggestive of constriction caused by poor diastolic filling include ascites,
in which individuals with pericardial effusion and tamponade demon- pleural effusions, and, occasionally, pericardial effusion. Often, there is
strate clinical and hemodynamic evidence of pericardial constriction dilatation of the atria, coronary sinus, inferior vena cava, and hepatic
after normalization of intrapericardial pressure by drainage of the effu- veins.8 The cardiac cavities may have a flattened or tubular appearance
sion.63 Pericardial constriction has a wide variety of potential causes on cross-sectional imaging, because of the constriction from the sur-
including cardiac surgery, radiation, trauma, infection (tuberculosis, rounding pericardium
bacterial, or viral), neoplasia, uremia, connective diseases, Dressler syn- Echocardiography may identify intermittent diastolic flattening of
drome, sarcoidosis, and drugs. However, the majority of cases are of LV inferolateral wall with respiratory variation, “septal shutter,” and
uncertain etiology.4,19 Although constriction is typically a chronic process, premature opening of the pulmonic valve.68 Additionally, TTE may
the acute inflammatory form may present from within days to months identify elevated atrial pressures and venous congestion. Conventional
after the initial insult, as has been described following cardiac surgery.19 Doppler imaging may reveal a restrictive LV inflow pattern, although
Constrictive pericarditis remains a challenging diagnosis and requires this pattern may also be seen in restrictive cardiomyopathy and other
a high degree of clinical suspicion. It usually presents with symptoms conditions associated with high atrial pressure. However, respiratory
and signs of right-heart failure including edema, anasarca, and fatigue.4 variation in transvalvular inflow patterns suggests constriction.69
Clinically, it may be difficult to distinguish constrictive pericarditis from CT detection of pericardial thickening demonstrates good correla-
other causes of heart failure, specifically restrictive cardiomyopathy, tion with histopathologic findings of abnormally thickened pericar-
because of their overlapping clinical presentations. Current imaging dium.67,70 CT is the most sensitive noninvasive modality for detection
techniques are critical in the differentiation of these two entities. In of pericardial calcifications because calcium produces high attenuation
both constrictive pericarditis and restrictive cardiomyopathy, ventricular on CT with small amounts of calcification recognizable. In contrast,
filling is restricted, leading to an increase in cardiac diastolic pressures. calcification on CMR manifests as signal deficit, although significant
In restrictive cardiomyopathy, ventricular filling is limited by abnormal deposits may be missed, if otherwise not known.8
myocardial compliance and relaxation, whereas in constriction, there On CMR, normal pericardium has low signal intensity on T1-weighted
is no inherent abnormality with myocardial relaxation. In constrictive spin echo imaging, typically seen as a darker stripe between the bright
pericarditis, the abnormal pericardium forms a noncompliant, rigid layers of epicardial fat and fat around the pericardium.3,34 Thickened
encasement around the heart, externally limiting diastolic filling and pericardium may have moderate-to-high signal intensity on spin echo
restricting cardiac output. Differentiating between restrictive cardio- images.9 The appearance on T2-weighted imaging is variable, but the
myopathy and constrictive pericarditis is critical, because restrictive signal intensity is usually lower than that of transudative fluid. Thickened
cardiomyopathy is typically treated medically, whereas constrictive peri- pericardium demonstrates low signal intensity on both T1-weighted
carditis is best treated with surgical pericardial stripping/pericardiectomy. and T2-weighted spin echo images and gradient echo cine images
Pericardiectomy may not be indicated in patients with very mild disease (including bSSFP).34 Normal pericardial thickness by CMR is ≤2 mm.32
or with severe advanced disease for whom the operative risk is exces- A thickness of >4 mm indicates pericardial thickening and is strongly
sively high.55,64,65 suggestive of constrictive pericarditis, in the appropriate clinical context.
The effusive–constrictive pericarditis is an uncommon syndrome CMR best distinguishes small pericardial effusions from thickening.
in which individuals with pericardial effusion and tamponade demon- Enhancement of the pericardium after contrast administration is an
strate clinical and hemodynamic evidence of pericardial constriction indicator of ongoing pericardial inflammation, which may be seen in
after normalization of intrapericardial pressure by drainage of the either the acute inflammatory or effusive–constrictive subtypes.48 Larger
540 SECTION V Vascular/Pericardium

pericardial calcifications may be visualized as regions of low signal lines passing through the myocardium–pericardium interface maintain
intensity by CMR. continuity during systolic deformation.3,73
A key finding in the diagnosis of pericardial constriction is the Ventricular filling patterns can be assessed by CMR using flow veloc-
presence of early diastolic septal flattening giving the appearance of ity encoding (phase contrast) sequences. In constrictive pericarditis, an
a “septal shutter,” which occurs as a result of the differential timing increased mitral level E-wave (early filling) may be observed as a con-
of the opening and closing of the mitral and tricuspid valve with sequence of increased diastolic pressure, whereas the A-wave (atrial
restricted right and left heart filling pressures. The diseased, noncom- filling) may be of reduced height owing to reduced late diastolic filling.16
pliant pericardium limits cardiac filling, causing rapid, high-velocity
early diastolic filling with minimal mid-to-late diastolic filling, because PERICARDIAL TUMORS
of the imposed external volume limitation. Additionally, the cardiac
volume constraint causes the function of one ventricle to be linked Primary Pericardial Tumors
to the other, resulting in ventricular interdependence, resulting in a Primary pericardial tumors are uncommon, with most pericardial tumors
“shuttering” of the interventricular septum with opening and closing a result of invasion (e.g., lung cancer), hematogenous seeding (e.g.,
of the mitral and tricuspid valves. Additionally, rapid diastolic filling melanoma), or lymphatic spread (e.g., breast cancer) of other primary
and ventricular interdependence leads to septal flattening or abnormal pathologies. Primary pericardial tumors are more commonly malignant
leftward bowing of the interventricular septum, rather than its normal and include mesothelioma, angiosarcoma, lymphoma, or liposarcoma.
rightward convexity. This is most pronounced during inspiration because Benign pericardial tumors include lipomas, teratomas, fibromas, neu-
the decline in intrathoracic pressure results in a decrease in pulmonary romas, and hemangiomas, and are extremely rare.9,19 Thymomas involving
venous pressure and, therefore, less LA filling (the rigid pericardium the pericardium may be either malignant or benign.
limits transmission of intrathoracic pressures to the heart). Right atrial
filling is not as impacted by changes in intrathoracic pressure, result- Imaging Findings of Primary Pericardial Tumors
ing in increased right-sided filling relative to the left. When encased An underlying pericardial neoplasm should be suspected if hemor-
in a rigid pericardium, the septum shifts leftward and flattens during rhagic pericardial effusions are found with diffuse or nodular pericar-
inspiration because of the increase in RV filling, and rightward during dial thickening. When evaluating any suspected pericardial mass, CT
expiration, resulting in pronounced septal shift (Fig. 46.5). Originally and CMR imaging are often necessary to accurately assess the lesion
noted on TTE, both the septal shutter and septal shift with respiratory characteristics and its relationship with the adjacent structures. Benign
variation can be visualized on real-time midventricular short axis or pericardial tumors found in children are often associated with large
four-chamber cine CMR and may be helpful in distinguishing constric- pericardial effusions.19 Primary mesothelioma of the pericardium, the
tion from restrictive cardiomyopathy.34,71 most common primary pericardial malignancy, usually presents with
Constriction is often localized to the right side of the heart and may a hemorrhagic pericardial effusion or pericardial plaques and may lead
be localized to just the region of the right atrioventricular groove.38,72 to pericardial constriction.19,74,75 Lymphoma, angiosarcoma, and liposar-
The use of contrast-enhanced CMR may suggest pericardial inflam- coma typically appear as large irregular masses, often associated with
mation in effusive–constrictive pericarditis.48 CMR tagging methods, pericardial effusions.9
such as spatial modulation of magnetization (SPAMM), may also be Lipomas can be readily recognized by their typical signal character-
useful in diagnosing constriction. Tagging stripes laid down in end istics with CMR or CT.3 On CT, lipomas generally have low attenuation.
diastole are successively imaged during ventricular systole, where normal On CMR, T1-weighted spin-echo images, lipomas have characteristic
slippage between myocardium and pericardium results in the appear- high-signal intensity that is not generally altered by contrast adminis-
ance of discontinuities or breaks in the stripes at the myocardium– tration and may demonstrate an “India ink artifact,” which is an out-
pericardium interface among normal patients. In patients with constrictive of-phase, T1-weighted imaging related to peripheral signal drop out in
pericarditis with adhesion of the parietal pericardium, this slippage is voxels containing both fat and nonfat components.9 Confirmation of
lost in the affected regions. Lines now appear “tethered” and the tag the presence of fat signal is achieved by demonstrating signal loss on

A B
FIG. 46.5  Real-time free-breathing cardiovascular magnetic resonance sequence in a short axis view in a
patient with acute pericarditis and constrictive physiology. (A) Normal filling during expiration with (B) septal
flattening and phasic change in left and right ventricles during inspiration.
CHAPTER 46  The Pericardium: Anatomy and Spectrum of Disease 541

A B
FIG. 46.6  Metastatic invasion of mediastinal liposarcoma with local invasion of the pericardium. (A) Steady-
state free precession imaging of pericardial mass (arrow). (B) Pericardial mass demonstrating late gadolinium
enhancement, suggestive of underlying malignancy (arrow).

fat-suppressed imaging.39 Depiction of regions of calcium or fat in a However, pericardial effusions and pericardial thickening may also occur
pericardial mass on CT or CMR suggests a teratoma.9 Fibromas more in patients with malignancy because radiation, drugs, and infections
commonly arise from the pleura, but may arise from the pericardium may also cause pericardial disease in this population.77,81
as well. They are usually homogenous in appearance and appear isoin- Hemorrhagic pericardial effusions are strongly suggestive of under-
tense to hypointense, as compared with myocardium on T1-weighted lying malignancy/metastatic pericardial disease.8,79 The other major
images, and hypointense on T2-weighted images. Fibromas may or may cause of a hemorrhagic pericardial effusion is aortic dissection propa-
not demonstrate contrast enhancement.39,76,77 Hemangiomas are gener- gating proximally to involve the pericardium, although these patients
ally bright on T1-weighted and T2-weighted images because of their often die before reaching the hospital. Acute hemorrhagic effusions
content of slow moving blood and demonstrate intense heterogeneous may be identified by their high signal intensity on T1-weighted and
enhancement following contrast administration.39 T2-weighted spin echo images. Extension of local tumor to the peri-
cardium may be confirmed by focal loss of the pericardial line with or
Secondary Malignant Pericardial Tumors without associated effusion (Fig. 46.6).8,34 On the other hand, an intact
Secondary malignant pericardial involvement is much more common pericardial line may be observed if an adjacent tumor extends up to
than primary pericardial tumors and reported in approximately 10% to the pericardium but not through it.9 Cine gradient echo CMR may
12% of all patients dying with malignancy.78,79 Among 110 patients with help determine if the tumor is adherent to the pericardium.77
cardiac metastases, autopsy studies revealed the pericardium was involved Most malignant tumors enhance after contrast administration.9,76
in more than 70% of cases, yet a pericardial effusion was found in only On noncontrast imaging, most neoplasms have low signal intensity on
one-third of those with pericardial involvement.79 Malignant pericardial T1-weighted images and high signal intensity on T2-weighted images.
involvement is often clinically silent and may be found incidentally. However, metastatic melanoma may have high signal intensity on
However, it can also present with symptoms of pericarditis, tamponade, T1-weighted and T2-weighted images because of the paramagnetic
or constriction. Patients with large pericardial effusions who present metals bound by melanin.84 Lymphomas appear isointense to hypoin-
with tamponade without clinical signs of pericarditis (e.g., chest pain, tense to myocardium on T1-weighted and T2-weighted images.39 They
rub, fever, and ECG changes), may be more likely to have a malignant may show lesser contrast enhancement in central regions that may be
effusion than other patients with large pericardial effusions.80 Pericardial necrotic.9,85
effusions caused by either malignancy or treatment of malignancy are
more likely than other effusions to require repeat pericardiocentesis or
surgical management.81 Malignant involvement of the pericardium may
CONCLUSION
result from local invasion (lung, breast, esophagus, stomach, lymphoma, The pericardium is a complex and important structure in the evaluation
thymoma, and pleural mesothelioma) or metastatic spread (breast, lung, of the cardiovascular system. Pericardial pathology can result in a spec-
melanoma, renal, and others) (Fig. 46.6).3,19,77,79 Malignancies most likely trum of clinical presentations, ranging from little or no symptoms to
to metastasize to the pericardium are typically lung, breast, and esoph- severe hemodynamic compromise and collapse. Advanced noninvasive
ageal cancer, melanoma, leukemia, and lymphoma.50,78,79,81,82 Patients imaging modalities such as CMR have broadened our understanding
with symptomatic malignant pericardial effusion generally have a poor of pericardial diseases and their impact on the cardiovascular system.
prognosis, although those with breast cancer, leukemia, or lymphoma When combined with a high degree of clinical suspicion, CMR can
may have a better prognosis than others.82,83 serve as an important tool to diagnose and guide clinical management
and therapeutic strategies.
Imaging Findings of Metastatic Pericardial Tumors
Metastatic involvement of the pericardium may be suggested by the
existence of a pericardial effusion with nodular pericardial thickening
REFERENCES
or pericardial mass, which can be visualized by TTE, CT, or CMR.18 A full reference list is available online at ExpertConsult.com
CHAPTER 46  The Pericardium: Anatomy and Spectrum of Disease 541.e1

REFERENCES 25. Truong MT, Erasmus JJ, Gladish GW, et al. Anatomy of pericardial
recesses on multidetector CT: implications for oncologic imaging. AJR
1. Eisenberg MJ, Dunn MM, Kanth N, Gamsu G, Schiller NB. Diagnostic Am J Roentgenol. 2003;181(4):1109–1113.
value of chest radiography for pericardial effusion. J Am Coll Cardiol. 26. Rabkin SW. Epicardial fat: properties, function and relationship to
1993;22(2):588–593. obesity. Obes Rev. 2007;8(3):253–261.
2. Bogaert J, Francone M. Pericardial disease: value of CT and MR imaging. 27. Iacobellis G, Leonetti F. Epicardial adipose tissue and insulin resistance in
Radiology. 2013;267(2):340–356. obese subjects. J Clin Endocrinol Metab. 2005;90(11):6300–6302.
3. Axel L. Assessment of pericardial disease by magnetic resonance and 28. Iacobellis G, Ribaudo MC, Assael F, et al. Echocardiographic epicardial
computed tomography. J Magn Reson Imaging. 2004;19(6):816–826. adipose tissue is related to anthropometric and clinical parameters of
4. Ling LH, Oh JK, Breen JF, et al. Calcific constrictive pericarditis: is it still metabolic syndrome: a new indicator of cardiovascular risk. J Clin
with us? Ann Intern Med. 2000;132(6):444–450. Endocrinol Metab. 2003;88(11):5163–5168.
5. Yousem D, Traill TT, Wheeler PS, Fishman EK. Illustrative cases in 29. Kodama F, Fultz PJ, Wandtke JC. Comparing thin-section and thick-
pericardial effusion misdetection: correlation of echocardiography and section CT of pericardial sinuses and recesses. AJR Am J Roentgenol.
CT. Cardiovasc Intervent Radiol. 1987;10(3):162–167. 2003;181(4):1101–1108.
6. Mulvagh SL, Rokey R, Vick GW 3rd, Johnston DL. Usefulness of nuclear 30. Ferrans VJIT, Roberts WC. Anatomy of the Pericardium. New York: Raven
magnetic resonance imaging for evaluation of pericardial effusions, and Press; 1982.
comparison with two-dimensional echocardiography. Am J Cardiol. 31. Hort W, Braeun H. Studies on the dimensions, wall thickness and
1989;64(16):1002–1009. microscopic structure of the pericardium in normal and pathological
7. Ling LH, Oh JK, Tei C, et al. Pericardial thickness measured with conditions. Arch Kreislaufforsch. 1962;38:1–22.
transesophageal echocardiography: feasibility and potential clinical 32. Sechtem U, Tscholakoff D, Higgins CB. MRI of the normal pericardium.
usefulness. J Am Coll Cardiol. 1997;29(6):1317–1323. AJR Am J Roentgenol. 1986;147(2):239–244.
8. Breen JF. Imaging of the pericardium. J Thorac Imaging. 2001;16(1): 33. Bull RK, Edwards PD, Dixon AK. CT dimensions of the normal
47–54. pericardium. Br J Radiol. 1998;71(849):923–925.
9. Wang K, Asinger RW, Marriott HJ. ST-segment elevation in conditions 34. Glockner JF. Imaging of pericardial disease. Magn Reson Imaging Clin N
other than acute myocardial infarction. N Engl J Med. Am. 2003;11(1):149–162, vii.
2003;349(22):2128–2135. 35. Rajiah P, Kanne JP. Computed tomography of the pericardium and
10. Wang ZJ, Reddy GP, Gotway MB, Yeh BM, Hetts SW, Higgins CB. CT and pericardial disease. J Cardiovasc Comput Tomogr. 2010;4(1):3–18.
MR imaging of pericardial disease. Radiographics. 2003;23(Spec No): 36. Nasser WK. Congenital diseases of the pericardium. Cardiovasc Clin.
S167–S180. 1976;7(3):271–286.
11. O’Leary SM, Williams PL, Williams MP, et al. Imaging the pericardium: 37. Smith WH, Beacock DJ, Goddard AJ, Bloomer TN, Ridgway JP,
appearances on ECG-gated 64-detector row cardiac computed Sivananthan UM. Magnetic resonance evaluation of the pericardium. Br J
tomography. Br J Radiol. 2010;83(987):194–205. Radiol. 2001;74(880):384–392.
12. American College of Cardiology Foundation Task Force on Expert 38. Frank H, Globits S. Magnetic resonance imaging evaluation of
Consensus Documents, Mark DB, Berman DS, et al. ACCF/ACR/AHA/ myocardial and pericardial disease. J Magn Reson Imaging.
NASCI/SAIP/SCAI/SCCT 2010 expert consensus document on coronary 1999;10(5):617–626.
computed tomographic angiography: a report of the American College of 39. Hoffmann U, Globits S, Frank H. Cardiac and paracardiac masses.
Cardiology Foundation Task Force on Expert Consensus Documents. Current opinion on diagnostic evaluation by magnetic resonance
Circulation. 2010;121(22):2509–2543. imaging. Eur Heart J. 1998;19(4):553–563.
13. Maffei E, Messalli G, Martini C, et al. Left and right ventricle assessment 40. Kyto V, Sipila J, Rautava P. Clinical profile and influences on outcomes in
with cardiac CT: validation study vs. cardiac MR. Eur Radiol. patients hospitalized for acute pericarditis. Circulation.
2012;22(5):1041–1049. 2014;130(18):1601–1606.
14. Goldstein JA. Cardiac tamponade, constrictive pericarditis, and restrictive 41. Spodick DH. Acute cardiac tamponade. N Engl J Med. 2003;349(7):
cardiomyopathy. Curr Probl Cardiol. 2004;29(9):503–567. 684–690.
15. Bogaert J, Francone M. Cardiovascular magnetic resonance in pericardial 42. Troughton RW, Asher CR, Klein AL. Pericarditis. Lancet. 2004;363(9410):
diseases. J Cardiovasc Magn Reson. 2009;11:14. 717–727.
16. Francone M, Dymarkowski S, Kalantzi M, Bogaert J. Magnetic resonance 43. Shaebetai RIM. Evaluation and Management of Acute Pericarditis.
imaging in the evaluation of the pericardium. A pictorial essay. Radiol Waltham, MA: UpToDate; 2005.
Med (Torino). 2005;109(1–2):64–74, quiz 75–76. 44. Zayas R, Anguita M, Torres F, et al. Incidence of specific etiology and role
17. Ellis H. Gray’s Anatomy: The Anatomical Basis of Clinical Practice. 39th ed. of methods for specific etiologic diagnosis of primary acute pericarditis.
New York: Elsevier Churchill Livingstone; 2005. Am J Cardiol. 1995;75(5):378–382.
18. Oyama N, Oyama N, Komuro K, Nambu T, Manning WJ, Miyasaka K. 45. Permanyer-Miralda G, Sagrista-Sauleda J, Soler-Soler J. Primary acute
Computed tomography and magnetic resonance imaging of the pericardial disease: a prospective series of 231 consecutive patients. Am J
pericardium: anatomy and pathology. Magn Reson Med Sci. Cardiol. 1985;56(10):623–630.
2004;3(3):145–152. 46. Stewart JR, Fajardo LF, Gillette SM, Constine LS. Radiation injury to the
19. Spodick D. The Pericardium: A Comprehensive Textbook. New York: heart. Int J Radiat Oncol Biol Phys. 1995;31(5):1205–1211.
Marcel Dekker, Inc.; 1997. 47. Stevenson LW, Child JS, Laks H, Kern L. Incidence and significance of
20. Vesely TM, Cahill DR. Cross-sectional anatomy of the pericardial sinuses, early pericardial effusions after cardiac surgery. Am J Cardiol.
recesses, and adjacent structures. Surg Radiol Anat. 1986;8(4):221–227. 1984;54(7):848–851.
21. Groell R, Schaffler GJ, Rienmueller R. Pericardial sinuses and recesses: 48. Watanabe A, Hara Y, Hamada M, et al. A case of effusive-constructive
findings at electrocardiographically triggered electron-beam CT. pericarditis: an efficacy of GD-DTPA enhanced magnetic resonance
Radiology. 1999;212(1):69–73. imaging to detect a pericardial thickening. Magn Reson Imaging.
22. Ellis H. Heart and mediastinum: heart and great vessels. In: Gray’s 1998;16(3):347–350.
Anatomy: The Anatomical Basis of Clinical Practice. 39th ed. New York: 49. Chiles C, Woodard PK, Gutierrez FR, Link KM. Metastatic involvement
Elsevier Churchill Livingstone; 2005. of the heart and pericardium: CT and MR imaging. Radiographics.
23. Levy-Ravetch M, Auh YH, Rubenstein WA, Whalen JP, Kazam E. CT of 2001;21(2):439–449.
the pericardial recesses. AJR Am J Roentgenol. 1985;144(4):707–714. 50. Lam KY, Dickens P, Chan AC. Tumors of the heart. A 20-year experience
24. Batra P, Bigoni B, Manning J, et al. Pitfalls in the diagnosis of thoracic with a review of 12,485 consecutive autopsies. Arch Pathol Lab Med.
aortic dissection at CT angiography. Radiographics. 2000;20(2):309–320. 1993;117(10):1027–1031.
541.e2 SECTION V Vascular/Pericardium

51. Sexton DJCG. Purulent Pericarditis. Watham, MA: UpToDate; 2005. 68. Engel PJ, Fowler NO, Tei CW, et al. M-mode echocardiography in
52. Rubin RH, Moellering RC Jr. Clinical, microbiologic and therapeutic constrictive pericarditis. J Am Coll Cardiol. 1985;6(2):471–474.
aspects of purulent pericarditis. Am J Med. 1975;59(1):68–78. 69. Hatle LK, Appleton CP, Popp RL. Differentiation of constrictive
53. Klacsmann PG, Bulkley BH, Hutchins GM. The changed spectrum of pericarditis and restrictive cardiomyopathy by Doppler echocardiography.
purulent pericarditis: an 86 year autopsy experience in 200 patients. Am J Circulation. 1989;79(2):357–370.
Med. 1977;63(5):666–673. 70. Oren RM, Grover-McKay M, Stanford W, Weiss RM. Accurate
54. Maisch B, Seferovic PM, Ristic AD, et al. Guidelines on the diagnosis and preoperative diagnosis of pericardial constriction using cine computed
management of pericardial diseases executive summary; The Task Force tomography. J Am Coll Cardiol. 1993;22(3):832–838.
on the Diagnosis and Management of Pericardial Diseases of the 71. Giorgi B, Mollet NR, Dymarkowski S, Rademakers FE, Bogaert J.
European Society of Cardiology. Eur Heart J. 2004;25(7):587–610. Clinically suspected constrictive pericarditis: MR imaging assessment of
55. Hoit BD. Management of effusive and constrictive pericardial heart ventricular septal motion and configuration in patients and healthy
disease. Circulation. 2002;105(25):2939–2942. subjects. Radiology. 2003;228(2):417–424.
56. Vilacosta I, Gomez J, Dominguez J, et al. Massive pericardiac hematoma 72. Masui T, Finck S, Higgins CB. Constrictive pericarditis and restrictive
with severe constrictive pathophysiologic complications after insertion of cardiomyopathy: evaluation with MR imaging. Radiology. 1992;182(2):
an epicardial pacemaker. Am Heart J. 1995;130(6):1298–1300. 369–373.
57. Seelos KC, Funari M, Chang JM, Higgins CB. Magnetic resonance 73. Kojima S, Yamada N, Goto Y. Diagnosis of constrictive pericarditis by
imaging in acute and subacute mediastinal bleeding. Am Heart J. tagged cine magnetic resonance imaging. N Engl J Med. 1999;341(5):
1992;123(5):1269–1272. 373–374.
58. Ferguson ER, Blackwell GG, Murrah CP, Holman WL. Evaluation of 74. Grebenc ML, Rosado de Christenson ML, Burke AP, Green CE, Galvin JR.
complex mediastinal masses by magnetic resonance imaging. J Cardiovasc Primary cardiac and pericardial neoplasms: radiologic-pathologic
Surg (Torino). 1998;39(1):117–119. correlation. Radiographics. 2000;20(4):1073–1103, quiz 1110–1111, 1112.
59. Jungehulsing M, Sechtem U, Theissen P, Hilger HH, Schicha H. Left 75. Song H, Choi YW, Jang IS, et al. Pericardium: anatomy and spectrum of
ventricular thrombi: evaluation with spin-echo and gradient-echo MR disease on computed tomography. Curr Probl Diagn Radiol.
imaging. Radiology. 1992;182(1):225–229. 2002;31(5):198–209.
60. Cheitlin MD, Armstrong WF, Aurigemma GP, et al. ACC/AHA/ASE 2003 76. Funari M, Fujita N, Peck WW, Higgins CB. Cardiac tumors: assessment
guideline update for the clinical application of echocardiography: with Gd-DTPA enhanced MR imaging. J Comput Assist Tomogr.
summary article: a report of the American College of Cardiology/ 1991;15(6):953–958.
American Heart Association Task Force on Practice Guidelines (ACC/ 77. Gilkeson RC, Chiles C. MR evaluation of cardiac and pericardial
AHA/ASE Committee to Update the 1997 Guidelines for the Clinical malignancy. Magn Reson Imaging Clin N Am. 2003;11(1):173–186, viii.
Application of Echocardiography). Circulation. 2003;108(9): 78. Abraham KP, Reddy V, Gattuso P. Neoplasms metastatic to the heart:
1146–1162. review of 3314 consecutive autopsies. Am J Cardiovasc Pathol.
61. Leeman DE, Levine MJ, Come PC. Doppler echocardiography in cardiac 1990;3(3):195–198.
tamponade: exaggerated respiratory variation in transvalvular blood flow 79. Klatt EC, Heitz DR. Cardiac metastases. Cancer. 1990;65(6):1456–1459.
velocity integrals. J Am Coll Cardiol. 1988;11(3):572–578. 80. Sagrista-Sauleda J, Merce J, Permanyer-Miralda G, Soler-Soler J. Clinical
62. Levine MJ, Lorell BH, Diver DJ, Come PC. Implications of clues to the causes of large pericardial effusions. Am J Med.
echocardiographically assisted diagnosis of pericardial tamponade in 2000;109(2):95–101.
contemporary medical patients: detection before hemodynamic 81. Gornik HL, Gerhard-Herman M, Beckman JA. Abnormal cytology
embarrassment. J Am Coll Cardiol. 1991;17(1):59–65. predicts poor prognosis in cancer patients with pericardial effusion. J
63. Hancock EW. A clearer view of effusive-constrictive pericarditis. N Engl J Clin Oncol. 2005;23(22):5211–5216.
Med. 2004;350(5):435–437. 82. Wilkes JD, Fidias P, Vaickus L, Perez RP. Malignancy-related pericardial
64. Seifert FC, Miller DC, Oesterle SN, Oyer PE, Stinson EB, Shumway NE. effusion. 127 cases from the Roswell Park Cancer Institute. Cancer.
Surgical treatment of constrictive pericarditis: analysis of outcome and 1995;76(8):1377–1387.
diagnostic error. Circulation. 1985;72(3 Pt 2):II264–II273. 83. Tsang TS, Seward JB, Barnes ME, et al. Outcomes of primary and
65. Ling LH, Oh JK, Schaff HV, et al. Constrictive pericarditis in the modern secondary treatment of pericardial effusion in patients with malignancy.
era: evolving clinical spectrum and impact on outcome after Mayo Clin Proc. 2000;75(3):248–253.
pericardiectomy. Circulation. 1999;100(13):1380–1386. 84. Mousseaux E, Meunier P, Azancott S, Dubayle P, Gaux JC. Cardiac
66. Sagrista-Sauleda J, Angel J, Sanchez A, Permanyer-Miralda G, Soler-Soler metastatic melanoma investigated by magnetic resonance imaging. Magn
J. Effusive-constrictive pericarditis. N Engl J Med. 2004;350(5):469–475. Reson Imaging. 1998;16(1):91–95.
67. Talreja DR, Edwards WD, Danielson GK, et al. Constrictive pericarditis in 85. Dorsay TA, Ho VB, Rovira MJ, Armstrong MA, Brissette MD. Primary
26 patients with histologically normal pericardial thickness. Circulation. cardiac lymphoma: CT and MR findings. J Comput Assist Tomogr.
2003;108(15):1852–1857. 1993;17(6):978–981.
SECTION VI Interventional

47 
Interventional Cardiovascular
Magnetic Resonance
Toby Rogers and Robert Lederman

An ideal imaging guidance system for cardiovascular, catheter-based and sometimes embedded x-ray fluoroscopy systems.4,5 Major limitations
interventional procedures would offer real-time, high-resolution, three- included relatively low signal-to-noise ratio (SNR), inhomogeneous
dimensional (3D) imaging of important anatomic tissues and chambers, magnetic field interfering with rapid imaging, and limited patient access
irrespective of respiratory, cardiac, or patient motion alongside excellent when employing long, closed bores. Newer, short-bore, 1.5 T systems
visualization of catheters, guidewires, and other interventional devices. with high performance gradients provide excellent field homogeneity
Such tools would quickly enable novel, minimally invasive alternatives and higher SNR. Newer scanners also include large arrayed radiofre-
to open surgical procedures. quency (RF) receivers (≥32), intended for parallel imaging techniques
X-ray fluoroscopy guides most contemporary catheter-based pro- to improve imaging speed. These additional receiver channels can also
cedures. However, fluoroscopy has important limitations (Table 47.1). be used to attach “active” catheter devices to improve their visibility. By
Iodinated radiocontrast, which provides the ability to outline chamber contrast, higher field 3 T systems disproportionately increase heating
and vascular lumens, is injected only periodically. Tissue detail is minimal. but do not notably enhance image quality using workhorse (balanced
Additionally, fluoroscopy provides only two-dimensional (2D) “projec- steady state free precession [bSSFP]) pulse sequences, and are therefore
tion” imaging, with limited depth perception. Iodinated radiocontrast difficult to justify for interventional CMR (iCMR) applications.
is nephrotoxic in susceptible individuals. Ionizing radiation increases In addition, commercial vendors now provide interactive scan user
lifetime cancer risk, particularly in children.1,2 Finally, operators and interfaces that increasingly resemble echocardiography. These interfaces
personnel risk disabling orthopedic injuries from the weight of protec- drive sophisticated image reconstruction hardware and software that
tive lead apparel.3 permit images to be acquired with ease and displayed to operators with
Ultrasound is often used in combination with x-ray fluoroscopy minimal delay in multiple scan planes.
procedures. For example, transesophageal echocardiography (TEE) Combined x-ray and CMR catheterization laboratories, so-called
provides adjunctive imaging during atrial septal defect closure or trans- XMR labs, now enable rapid and convenient CMR catheterization
catheter aortic valve replacement, by assisting with device sizing, posi- procedures, especially in pediatrics and heart failure (Fig. 47.1). In
tioning, and postdeployment assessment. However, transthoracic these laboratories, conventional x-ray fluoroscopy guidance is imme-
echocardiography (TTE) offers limited acoustic windows, and suffers diately available to perform interventions using CMR fusion imaging
“shadowing” artifacts caused by devices. or for situations requiring emergency bailout. XMR laboratories are
Cardiovascular magnetic resonance (CMR) more closely approaches now commercially available. For example, the National Heart Lung
the idealized imaging guidance system described earlier. CMR offers and Blood Institute (NHLBI) laboratory at Children’s National
superior tissue imaging, in any arbitrary orientation, without ioniz- Medical Center in Washington, DC, combines a biplane x-ray fluo-
ing radiation or nephrotoxic contrast. Rapid imaging techniques and roscopy system with a 1.5 T cardiac scanner (Fig. 47.1). These systems
modified CMR visible devices now permit real-time CMR guided are separated by RF-shielded doors and can be used independently
catheter applications. The ability to visualize both tissue and inter- or together for combined procedures.6 An automated transport table
ventional devices may enable catheter-based procedures in the future moves patients seamlessly between the two modalities. Alternative
that currently require open surgical exposure. In the meantime, CMR designs include single-room layouts, in which both x-ray and CMR
right heart catheterization provides superior hemodynamic char- systems are located inside the RF shield.7 Wave guides and penetration
acterization combining high fidelity flow, pressure, and function panels are strategically positioned in the room so that communication
measurements. and patient monitoring hardware can be installed without disrupting
the RF shield.
INTERVENTIONAL CARDIOVASCULAR MAGNETIC
Communication, Monitoring and Image Display
RESONANCE LABORATORY CONFIGURATION Rapid CMR requires rapid gradient switching that causes substantial
Early real-time magnetic resonance imaging (MRI) systems employed low acoustic noise. This noise prevents verbal communications between
field strength magnets (0.2–0.5 T), open or closed bore configurations, operators, the patient, and staff. Wireless noise-cancelling headsets are

542
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 543

TABLE 47.1  Advantages and Disadvantages of Different Imaging Modalities to Guide


Interventional Procedures
Imaging Modality Advantages Disadvantages
iCMR • Excellent soft tissue imaging • Clinical devices currently unavailable and require hardware
• Simultaneous display of multiple arbitrary imaging attachments
perspectives • Acoustic noise
• Real-time device tracking in 3D • Claustrophobia
• Image-based physiology assessment • Rapid emergency response more difficult
• No ionizing radiation • ECG monitoring for cardiac procedures more difficult
• No lead aprons • Peripheral nerve stimulation from rapidly switching gradients
• No nephrotoxic contrast • Imaging systems require large capital outlay
• Uninterrupted imaging to detect complications more
efficiently
X-ray fluoroscopy • Excellent temporal resolution • Poor soft tissue imaging
• Widely available in most centers • 2D “projection” imaging offers limited depth perception
• Clinical devices available • Cancer risk from ionizing radiation
• ECG monitoring simple • Nephrotoxic contrast required
• Floating table permits remote imaging (i.e., groin site) • Chronic orthopedic injuries from lead apron apparel
• Greater physician/nurse access to patient • Angiography interrupted during device deployment resulting
in complication detection after contrast injection
Ultrasound • Good soft tissue imaging • Limited acoustic windows
• Excellent temporal resolution • Device related “shadow” artifacts
• Widely available • No tip tracking capability
• Imaging based physiology assessment (i.e., Doppler) • Transesophageal and intracardiac echo are invasive
• New x-ray + ultrasound fusion capability • Limited orientation-dependent imaging
(CartoMerge, HeartNavigator, etc.)

2D, Two-dimensional; 3D, three-dimensional; ECG, electrocardiogram; iCMR, interventional cardiovascular magnetic resonance.

FIG. 47.1  National Heart Lung and Blood Institute/Children’s National Medical Center combined x-ray and
interventional cardiovascular magnetic resonance (iCMR) suite, Washington, DC. The iCMR suite combines
a biplane x-ray system (foreground) and a 1.5 T CMR scanner (background). The x-ray table docks with the
CMR table to enable the patient to be easily transferred between the two modalities.

used at NHLBI (Fig. 47.2). This versatile system allows staff and patients heart rate monitoring; however, ECG waveform morphology is not
to speak simultaneously on open microphones and allows separate and readily interpretable for ST-segment monitoring of myocardial ische-
parallel conversations with patients and among staff. mia or injury.8,9 Multichannel invasive pressure waveform monitoring,
Gradients, RF interference, and magnetohydrodynamic effects temperature monitoring, and oxygen saturation monitoring can use
severely distort the electrocardiogram (ECG). Electronic filters enable commercial systems, without modification from x-ray catheterization
544 SECTION VI Interventional

FIG. 47.2  Working inside the interventional cardiovascular magnetic resonance (iCMR) catheterization labora-
tory. The operator communicates with the patient and with staff inside and outside the iCMR room with
sound-suppression microphones and headsets. Real-time CMR, scanner control interface and patient hemo-
dynamics are displayed inside the room using video projectors.

laboratories. Several manufacturers are developing dedicated hardware in a 3D-rendered image (Fig. 47.3).12 Other laboratories are experi-
that will interface with existing catheterization laboratory high-fidelity menting with voice-control, automatic adjustment of slice location to
hemodynamic recording systems. In the meantime, open source kits correspond to catheter position, and automatic adaptation of imaging
for local assembly of suitable hemodynamics interfaces are available parameters to speed of catheter manipulation.
(http://nhlbi-mr.github.io/PRiME).
Interventional catheterization suites require continuous operator Safety Considerations
monitoring of multiple information streams, including hemodynamics, Patient care staff must undergo formal comprehensive training in CMR
imaging control, and interventional images. This is similarly true during safety. For example, the hospital emergency medical response team is
iCMR procedures, especially using active devices.6,10 MRI-conditional unlikely to be familiar with CMR operations and may unwittingly jeop-
liquid crystal display (LCD) video displays are available from most ardize themselves, their colleagues, or the patient by rushing into the
scanner manufacturers. We prefer two banks of less expensive LCD room with a steel oxygen tank. Rapid patient evacuation from the CMR
video projectors that project onto a fabric screen, which depict hemo- catheterization laboratory needs to be practiced repeatedly in mock
dynamics, scanner interface, and 3D-rendered images separately, one drills. Ferromagnetic objects that cannot be removed from the room
at each end of the CMR system (Fig. 47.2). The projectors are housed must be attached to the wall during CMR guided procedures. It is good
in RF-shielded boxes and video signals are passed into the room via practice for all devices on wheels in the x-ray section of the iCMR suite
fiber optic cables through the waveguide. (e.g., anesthetic machine, intraaortic balloon pump, or defibrillator)
to be tethered to the wall or floor. Oxygen, inhalational anesthetic, and
Interventional Cardiovascular Magnetic Resonance vacuum for suction can be fed through RF-protective wall ports, elimi-
Real-Time Scanner Interface nating in-room cylinders. At NHLBI, we drill emergency evacuation
The iCMR scanner user interface requires several key features not typical from the CMR room on a quarterly basis.13 Swift evacuation relies on
for diagnostic CMR. Commercial or investigational graphic user interface clear delineation of roles for all staff. In an emergency, we can start
based scanning host computer systems are available from most systems chest compressions within 10 seconds and move the patient back into
vendors (Interactive Front End, Siemens11; MR Echo, General Electric; the x-ray room for defibrillation in under 1 minute. In the near future,
eXTernal Control, Philips Healthcare) and as standalone commercial CMR-conditional external defibrillators should be commercially avail-
(RT Hawk, Heart Vista) or open-source solutions (Vurtigo, Sunnybrook able so that arrhythmias can be managed without evacuating the patient
Health Sciences Center, http://www.vurtigo.ca). from the scanner.
One of the most useful features is the colorized display of individual RF energy from CMR transmit coils will concentrate on long con-
receiver channels that are attached to “active” catheter receiver coil devices. ductive devices causing local heating, potentially leading to burns.14
This makes catheter devices readily apparent and has proven useful in Box 47.1 lists factors associated with heating, especially long conductive
vivo. Another is interactive “projection-mode” imaging wherein slice devices and cables. This complex problem is described later under “active”
select is disabled and catheter devices are displayed as the operator is catheter devices. Interventionists must also take care that connections
used to seeing them under conventional x-ray fluoroscopy. For tracking to intravascular coils as well as surface coils do not inadvertently form
balloon catheters filled with dilute gadolinium (Gd) contrast during loops, which can cause patient burns.
CMR catheterization, we find it helpful to interactively adjust slice
thickness and saturation magnetization preparation, to help find the
catheter tip should it move outside of the selected plane, and to inter-
REAL-TIME IMAGING
actively adjust temporal resolution for catheterization steps requiring Rapid CMR is required for invasive procedures, for catheter visualiza-
fine motor control. Operationally, we find it useful to display multiple tion, and for imaging of anatomic structures.15 Efficient image data
slices during interventional procedures, both separately and combined sampling methods,16–25 parallel imaging,26–28 and coherent steady state
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 545

FIG. 47.3  Screenshot of a commercial interventional cardiovascular magnetic resonance (iCMR) user inter-
face. The iCMR interface is designed to accommodate interleaved multislice real-time CMR image acquisition
(three panels on left), a volume rendering of the slices indicating their three-dimensional relationship (center
panel), “postage stamps” to store and recall important graphical slice prescriptions (bottom row), and interac-
tive scanner parameter control (right panels).

techniques29–33 have enabled real-time CMR without significant degra-


BOX 47.1  Factors Influencing Heating-
dation in image quality in the field of interest. Frame rates as high as
Related Injury by Cardiovascular Magnetic 10 to 15 images per second are now possible using multichannel (≥32)
Resonance Interventional Devices coils that use parallel imaging techniques to achieve acceleration factors
Interventional Device Factors of three or more. In addition, interactive, real-time color flow imaging
• Length of conductive elements may supplement anatomic detail with critical physiologic detail, such
• Geometric shape as leaks or gradients, during therapeutic procedures and interventions.34
• Orientation in the magnet Slice orientation can automatically follow the tip of catheter devices as
• Distance from the radiofrequency transmitter they move, so-called adaptive imaging, so that catheter features are kept
• Physical proximity to tissues in view during manipulation. These methods also can be applied auto-
• Insulation matically to alter scanning parameters such as field-of-view and temporal
resolution.35
Patient Factors
• Body mass and surface area INTERVENTIONAL CARDIOVASCULAR MAGNETIC
• Convective cooling of intravascular devices by local blood flow
• General body temperature
RESONANCE CATHETER DEVICES
• Implanted conductive devices Interventional devices must be conspicuous and clearly distinguishable
• Tissue thermosensitivity from tissue to conduct therapeutic procedures. Conventional x-ray devices
are generally unsuitable, because most incorporate steel braids to increase
Magnetic Resonance Imaging Scanner Factors x-ray attenuation for visibility and to enhance catheter performance
• Field strength characteristics such as steerability, pushability, and trackability. The steel
• Pulse sequence causes severe blooming or susceptibility artifacts that destroy much
• Flip angle of the CMR image (Fig. 47.4). Removing these ferrous components
• Scanning duty cycle usually renders the catheter devices virtually invisible under CMR, and
• Position relative to bore isocenter140 (closer is better) usually renders them floppy and mechanically unsuitable as catheters.
iCMR catheter devices are generally classified as passive or active.
Passive devices have elements that cause discrete susceptibility imaging
artifacts that darken images, or T1 shortening elements that brighten
images. Alternatively, active devices have built-in micro-coils and
546 SECTION VI Interventional

B
A B FIG. 47.5  Passive and active device imaging during aortic coarctation
repair. (A) A dilute gadolinium partially filled balloon, delivered over a
FIG. 47.4  Cardiovascular magnetic resonance (CMR) artifact caused nitinol guidewire, is positioned across the coarctation (arrow). The wire
by steel braiding in standard x-ray catheters. (A) Photograph of stainless
and balloon catheter shaft are poorly visualized against the anatomic
steel braided Kumpe catheter. (B) In a water phantom, steel braiding
background. (B) The same balloon, delivered over an active guidewire
causes severe ‘blooming’ signal void artifact rendering it useless for
from which the signal is colored in green, is clearly more conspicuous
CMR guided interventional procedures.
compared with solely passive catheter devices.

electrical circuitry that allow the device to act as a RF receiver and/or


transmitter. A minimum of two coils at the tip of a catheter is required to enable the
computer to calculate the position of the catheter in 3D space and depict
Passive Devices it on an image. This approach is particularly attractive to procedures
Stents36 and guidewires37–40 made from copper, dysprosium, cobalt– that require high spatial location accuracy, for example electrophysiology
chromium alloy, nitinol, titanium, and platinum suffer less severe sus- mapping and ablation. Tracking also permits the user interface to store
ceptibility artifacts compared with iron-alloys. They can be coated to coordinates of key locations, for example ablation points.
improve biocompatibility. Carbon dioxide creates a dark CMR signal Alternatively, devices incorporate conductive wires (or loop antenna)
by excluding proton spins; carbon dioxide gas has been injected in along the length of the device. Signals from these devices are displayed
humans for selective CMR angiography and has been used to fill balloon on top of previously acquired anatomic roadmaps (device localization
catheters for diagnostic CMR-guided catheter tracking in patients.41–43 only) or combined into the array of receivers used to update real-
Unfortunately, volume averaging makes it difficult to distinguish passive time CMR images. The device position or signal receiver can be color
devices from neighboring anatomic features. Guidewires44–46 have been coded, to distinguish them from grey-scale background images of the
made out of nonmetallic polymers, and clinical cardiovascular interven- anatomy. Devices displayed in this way are readily visible inside thick-
tions have been conducted using polymer guidewires.47 Polymer guide- slab projections resembling projection x-ray images showing devices in
wires lack the trackability, torquability, and tactile responsiveness of profile. Distinct signals can be imparted to the tip of devices so that it
metallic guidewires; a new segmented nitinol design overcomes these can be tracked in 3D, ideal for intracardiac applications. Alternatively,
limitations.48 direct current applied to conductive elements along the device induce
Devices that appear bright are less vulnerable to volume averaging magnetic field inhomogeneities, disrupt local signal, and create dark
effects and are more easily visualized against anatomic background. impressions.63
Dilute Gd chelates such as gadopentate dimeglumine (Gd-DTPA) have Unfortunately, the long conductive transmission wires used to connect
been used to fill49 and coat catheters50,51 and balloons,52 offering bright catheter coils to the CMR transmitter or receiver system are susceptible
device imaging. Ultimately, we have found this passive approach to to RF heating, which may damage neighboring tissue. Multiple approaches
be inferior in vivo compared with active catheter device visualization to reduce heating can be combined to make catheter devices safe, such
(Fig. 47.5). Investigational, off-resonance, chemical-selective visualiza- as attaching circuitry to decouple and detune the transmission lines,
tion using F-1953 or C-1354 and other hyperpolarized agents have been intermittent chokes and transformers in the transmission lines, and
proposed for catheter tracking. Off-resonance effects have been exploited insulation.
to track metallic catheter and guidewire components.55,56 Clinical diag- Another hybrid approach is to incorporate closed-loop receiver coils
nostic catheterization for hemodynamic studies can be conducted using into stents or catheter devices, without connecting via transmission
dilute Gd-filled balloon catheters,6 iron-contrast filled,57 or CO2-filled58 lines to the CMR system. These “inductively-coupled” devices resonate
catheters as an alternative. at predetermined geometric shapes and thereby amplify local RF
signal.64,65 Unfortunately, inductively coupled catheter devices cannot
Active Devices easily be displayed in color during real-time CMR, as can other actively
Highly sensitive, ultrasmall receiver coils can be incorporated into visualized catheter devices, and are best visualized at lower flip angles
devices to locate (catheter-tracking59) or to visualize them,60–62 or both. that compromise imaging.
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 547

The NHLBI active guidewire for cardiovascular applications incor- whether remote-controlled catheters really decrease procedure time in
porates a loop antenna for active visualization of the whole shaft and experienced hands.73
a separate active marker to distinguish the guidewire tip.66 The device
also incorporates a fiberoptic temperature probe for continuous moni- APPLICATIONS
toring of RF-induced heating. The device appears in color overlay on
real-time CMR images (Fig. 47.6). Extra-Anatomic Bypass
Finally, CMR pulse sequence techniques can reduce the energy depos- Advances in transcatheter therapy for many congenital cardiovascular
ited per image, and therefore heating, with an acceptable penalty in image conditions have reduced the need for invasive open surgery. As noted
quality.15 Such techniques include radial sampling, spiral sampling, and earlier, children may be particularly sensitive to the harmful effects of
echo planar imaging (EPI), which use fewer excitation pulses and longer x-ray radiation. Children with complex congenital cardiovascular disease
repetition times, variable flip angle sequences67 and parallel imaging. often undergo multiple x-ray procedures and have greater cumulative
Techniques that reduce specific absorption rate at high fields, such as radiation exposure and greater lifetime risk of radiation-induced malig-
adaptive parallel transmission, may also prove helpful at lower fields to nancy. Radiation-sparing procedural guidance with CMR is attractive
reduce device heating during CMR guided interventions.68 and forms the basis for a number of real-time CMR guided research
applications.
Device Solutions for Cardiovascular Applications Full visualization of all anatomic structures and soft tissue may
In our experience, it has been useful to use a combination of both active facilitate complex procedures that involve crossing anatomic boundaries
and passive devices during complex interventional procedures, albeit and connecting remote vascular structures. Using a double-doughnut
in animal models. Real-time, active visualization of the device tip in CMR configuration, Kee et al. conducted preclinical74 and clinical75
3D is desirable during certain procedures such as atrial transseptal transjugular intrahepatic porto-systemic shunt (TIPS) procedures. Even
puncture or myocardial wall injection. Similarly, active approaches prove in this proof-of-concept experiment, MRI reduced the number of tran-
useful while surveying devices for common failure modes such as buck- shepatic needle punctures compared with historical controls. Arepally
ling, looping, or kinking. Passive approaches unencumbered by electrical et al. created an elegant catheter-based mesocaval shunt outside the
hardware attachments are sufficient for simple procedures such as placing liver capsule.76
introducer sheaths that typically do not move once positioned. More Ratnayaka et al. developed the preclinical capability to use catheters
importantly, combining passive devices (such as balloons filled with instead of surgery for one of the three staged surgical repairs to palliate
dilute Gd-DTPA) with active devices (such as an active guidewire) children with single ventricle physiology. They demonstrated real-time
generates wonderful and useful images (Fig. 47.5). iCMR guided cavopulmonary shunt using active needles, Gd-filled
Catheters for iCMR are usually manipulated manually by the opera- balloon-mounted covered stents, and custom-built endografts (Fig.
tor in a similar fashion to a standard x-ray catheterization laboratory. 47.7).77 Although theoretically feasible using x-ray fluoroscopy and
However, by applying current to wire coils wound at the tip of a catheter, contrast angiography, CMR enables visualization of origin and destina-
it is possible to remotely steer a catheter under real-time iCMR guid- tion vascular structures in orthogonal planes as well as all interposed
ance.69,70 Using this approach, Hetts et al. developed an endovascular tissues. Furthermore, CMR enables immediate identification of important
catheter system and demonstrated comparable procedure times for navi- complications, for example, pericardial effusion or vascular dissection.
gation through a vascular phantom with MRI and conventional x-ray The ultimate goal is to develop percutaneous approaches to replace
guidance.71 The same group used these catheters to perform renal artery some of the open-chest surgeries that children with complex congenital
catheterization and embolization in swine.72 However, it is questionable heart disease will require over their lifetime.

A B C
FIG. 47.6  Cardiac catheterization in a pig using the National Heart Lung and Blood Institute active guidewire.
The full length of the guidewire is conspicuous and the tip has a separate and distinct signal (arrow). During
left heart catheterization, the guidewire is navigated to (A) the aortic arch, (B) the aortic valve, and (C) through
the aortic valve into the left ventricle under real-time cardiovascular magnetic resonance guidance.
548 SECTION VI Interventional

A B C
FIG. 47.7  Real-time cardiovascular magnetic resonance (CMR) guided percutaneous cavopulmonary shunt.
Central venous access is obtained via the right internal jugular vein. (A) An active needle is used to puncture
from the superior vena cava in to the right pulmonary artery under real-time CMR guidance (arrow). (B) A
covered endograft is then deployed from the superior vena cava into the right pulmonary artery. (C) The
endograft now connects the superior vena cava and the right pulmonary artery.

Endomyocardial Biopsy Atrial Transseptal Procedures


Endomyocardial biopsy remains the preferred diagnostic test in patients Atrial transseptal puncture is usually conducted as the first step in
with unexplained cardiomyopathy and in heart transplant recipients numerous cardiac procedures, such as pulmonary vein ablation. A needle
with suspected rejection. Yet x-ray guided biopsy is essentially a blind is advanced from a vein, through the right atrium into left atrium (LA)
procedure because neither important structures, such as valves or across the interatrial septum. Currently, this procedure is conducted
chordae, nor the target endocardium can be visualized. In patholo- using subtle x-ray fluoroscopic visual cues and tactile feedback from
gies affecting the heart in a nonuniform distribution, biopsy can often sharp catheter devices, with or without adjunctive TEE or intracardiac
provide inconclusive results because a negative specimen may simply echocardiography. Poor tissue visualization and limited acquisition
reflect that a nonaffected part of the heart was sampled. In contrast, windows, combined with unusual atrial anatomy, can lead to life-
CMR offers a number of tools for advanced tissue characterization threatening perforation and pericardial tamponade in as many as 1%
that may help distinguish normal from abnormal myocardium, for to 6% of procedures, even in experienced hands. Using custom active
example, late Gd enhancement (LGE) and T1 mapping. Real-time iCMR needles, real-time iCMR guided atrial transseptal puncture has been
guided endomyocardial biopsy could potentially enhance diagnostic successfully performed in swine (Fig. 47.10).82,83 Related therapeutic
yield and safety by targeting abnormal myocardium and avoiding vul- procedures, such as closure of atrial septal defects and patent foramen
nerable structures. Both passive-visualization and active-visualization ovale, have been reported in swine using passively visualized nitinol
bioptomes have been developed for in vitro and preclinical testing.78,79 devices delivered with catheters.84,85
We developed an animal model of focal myocardial pathology and
demonstrated significantly higher diagnostic yield with iCMR versus Transthoracic Cardiac Access and Closure
x-ray guided biopsy (Fig. 47.8).79 Using active needles and guidewires, and passive sheaths and catheters,
we have successfully performed real-time iCMR guided percutaneous
Aortic Aneurysm, Dissection and Coarctation Stenting transthoracic access to the left ventricle (LV) and the LA.86–88 After
Percutaneous endograft repair for thoracoabdominal aneurysms and delivery of large-bore sheaths, the access port was closed using off-the-
aortic dissection are performed for patients with suitable anatomy, con- shelf nitinol occluder devices or collagen vascular plugs. The delivery
sidered high risk for surgery. Aorta size, proximal and distal landing system for the nitinol occluders was modified to incorporate a loop
zones for stents and grafts, and vicinity to crucial arterial branches are antenna for active visualization.66 Via closed-chest transthoracic approach,
vital measurements required for these procedures. These procedures are Ratnayaka et al. performed muscular ventricular septal defect (VSD)
typically performed using x-ray fluoroscopy, with adjunctive intravascular closure using nitinol occluders and the same modified delivery system
ultrasound. Bulky stent/graft devices may distort the native anatomy, (Fig. 47.11).89 Alternative applications could include delivery of partial
preventing operator confidence in preacquired fluoroscopic roadmaps. or full mitral or tricuspid prosthetic valves, which are large devices
Ultrasound scatters within stents/endograft, offering limited external currently under investigation, and which are usually delivered via surgi-
visualization. Real-time iCMR guided abdominal aortic aneurysm80 and cal transthoracic access to the heart.
aortic dissection81 endograft repair have been performed successfully in The ability to manage complications in the CMR scanner is impor-
swine models using active and passive nitinol stents (Fig. 47.9). Post- tant. Using off-the-shelf titanium needles, Halabi et al. performed real-
procedure assessment using phase contrast flow within and adjacent to time iCMR guided pericardiocentesis in a swine model of pericardial
the endograft demonstrated the versatility of iCMR-guided endograft effusion.90 CMR-conditional nitinol or Inconel needles are available
therapy. Real-time iCMR guided aortic coarctation stent repair has commercially but, although they are safe to use in the iCMR environ-
also successfully been performed in a swine coarctation model using ment, they are practically invisible. Needles with passive or active markers
commercially available clinical-grade devices38 (see Fig. 47.5). are sorely needed.
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 549

iCMR
bioptome

FIG. 47.8  Real-time interventional cardiovascular magnetic resonance (iCMR) guided endomyocardial biopsy.
The active visualization CMR bioptome appears in color overlay on real-time CMR images. The arrows indicate
the jaws of the bioptome. Orthogonal views ensure the bioptome is accurately directed to the desired
endocardial surface. Inversion-recovery real-time CMR can be used to highlight areas of abnormal tissue
after administration of systemic gadolinium contrast.

A B C
FIG. 47.9  Real-time interventional cardiovascular magnetic resonance (iCMR) guided aortic dissection repair
in a swine model. (A) Baseline CMR showing dissection of the descending aorta. (B) Passively visualized
stent positioned in the descending aorta at the level of the dissection. (C) Final CMR demonstrates success-
fully delivered stent and repair of the dissection. (Courtesy Holger Eggebrecht and Harold H. Quick, PhD,
University Essen, Germany.)
550 SECTION VI Interventional

A B C
FIG. 47.10  Atrial transseptal puncture in a pig using an active needle. (A) Photograph of the active trans-
septal puncture needle. (B) Cardiovascular magnetic resonance of the needle in a water phantom and (C) in
vivo across the interatrial septum.

A B
FIG. 47.11  Real-time interventional cardiovascular magnetic resonance (iCMR) guided direct transthoracic
ventricular septal defect closure. The right ventricle is accessed directly through the chest wall using a CMR
active needle under real-time iCMR guidance. The ventricular septal defect (VSD) is crossed antegradely and
then closed using a nitinol muscular occluder device. The delivery cable for the occluder device is also active.
(A) The left ventricular disk (arrow) is deployed and pulled back against the septum. (B) The right ventricular
disk is deployed and pushed forward against the septum to close the VSD. The right ventricle free wall
puncture is closed using an off-the-shelf vascular closure plug.

Invasive Coronary Artery Imaging Gd-DTPA (bright signal), undiluted Gd-DTPA (dark signal), or carbon
Percutaneous, real-time iCMR guided coronary selective angiography,91 dioxide gas have been used to inflate balloons and provide balloon-
angioplasty,92 and stent insertion93 has been reported in healthy animals. tissue contrast ensuring full inflation. Active receiver coils, either embed-
Although these works are impressive, clinical translation of these coro- ded on the balloon catheter or inserted through the wire port of the
nary artery therapeutic procedures remains hindered by seemingly balloon filled with dilute Gd-DTPA, provide added contrast to adjacent
insurmountable obstacles. Currently x-ray fluoroscopy provides spatial tissue by further enhancing the bright signal within the inflated balloon.
resolution of 100 µm at a usual working temporal resolution of 66 ms, Radio-opaque markers are typically added to balloon catheters to indi-
to manipulate guidewires that are 350 µm wide and stent devices that cate the shoulder points of the balloon and assist with lesion length
are 600 µm across. It seems unlikely—barring an unforeseen technical assessment before and during deployment. These markers act as small
breakthrough—that real-time CMR can provide comparable spatial susceptibility markers to assist with positioning and balloon deployment
and temporal resolution currently required for safe guidewire and cath- under iCMR.
eter manipulation through delicate diseased human coronary arteries. Both balloon-expandable and self-expanding stents are implanted
Similarly, very low profile, distinctly conspicuous CMR-compatible to prevent arterial recoil, and to alleviate flow-limiting dissection. Both
catheter devices would be required for clinical implementation, and stent designs have been successfully deployed under iCMR guidance in
such devices are not currently available. animals101–103 and humans.104 Local susceptibility and shielding effects
result in imaging voids within and adjacent to stents. Inductively coupled
Peripheral Vascular Disease stents (described earlier) may ameliorate this problem.64 Chronic total
Several groups have shown the feasibility of iCMR guided balloon arterial occlusion recanalization is particularly challenging under x-ray
angioplasty in healthy animals,94,95 animal models of arterial stenosis,96–99 fluoroscopy because only the patent inflow, occluded artery, and patent
and humans with obstructive peripheral artery disease.100 Dilute outflow distal artery beyond the obstruction can be visualized with
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 551

conventional angiography. Guidewires and catheter traversal through depending on the patient’s clinical presentation; for example, lung per-
the “invisible” occluded segment may cause perforation and hemor- fusion to screen for chronic thromboembolic pulmonary artery disease.111
rhage. These procedures are often long, and require excessive nephrotoxic With procedural workflow optimization it is possible to repeat mea-
contrast. Successful real-time iCMR guided chronic total occlusion surements at rest and under physiological provocation, such as exercise,
recanalization and subsequent balloon angioplasty was successfully inhaled nitric oxide or intravascular volume challenge, all within a
performed in swine model of peripheral artery occlusion, using modi- reasonable timeframe.112 Investigators from the University of Leuven
fied active chronic total occlusion wires and support catheters.105 in Belgium expertly applied this technology in patients with chronic
thromboembolic pulmonary hypertension and in endurance athletes.113,114
X-Ray Fused With Magnetic Resonance Imaging Subjects performed supine exercise in the CMR scanner with a pulmo-
3D structures can be segmented from CMR datasets and coregistered nary artery catheter in place for assessment of pulmonary vascular
with live x-ray fluoroscopy to provide anatomic context. XFM has been resistance and RV function during physiological stress. Hemodynamic
used successfully to enhance imaging guidance in a wide range of car- parameters only measureable with exercise CMR catheterization, such
diovascular interventions, including pulmonary stenosis angioplasty, as pulmonary vascular and RV reserve, may be valuable prognostic
aortic coarctation stenting (Fig. 47.12), VSD closure, and for cardiac indicators in patients with little or no overt pathology at rest.115
resynchronization therapy to optimally place the LV lead.106 Because
anatomy is clearer and frequent contrast angiography is not required, Tissue Delivery and Ablation
total fluoroscopy time and iodinated contrast volume can be substan- Real-time iCMR guided endomyocardial cell delivery has achieved
tially reduced with XFM.107 To be useful, XFM requires accurate coreg- millimeter-scale precision using modified CMR needle-catheters in
istration of both imaging modalities. A number of groups have developed animal models for many years.116–121 These technically impressive delivery
techniques to improve coregistration and to incorporate respiratory systems await suitable cell preparations or interstitial tissue augmenta-
and cardiac motion.108,109 tion materials for applications to patients.
High intensity focused ultrasound is an alternative to RF energy for
Diagnostic Right Heart Catheterization targeted tissue ablation. Preclinical CMR-conditional, high intensity
Using off-the-shelf nonbraided balloon catheters, it is feasible to perform focused ultrasound catheters have been developed and successfully used
diagnostic right-heart catheterization in patients.41 Dilute Gd-filled to ablate tissue such as renal parenchyma.122 Importantly, histopathol-
balloons are conspicuous using real-time CMR (Fig. 47.13). Ratnayaka ogy demonstrated coagulation necrosis with comparable volumes to
et al. performed both x-ray and iCMR-guided right-heart catheteriza- the lesions seen with contrast-enhanced CMR. However, just like RF
tion in 16 patients, demonstrating that CMR was useful to navigate ablation lesion visualization, high intensity focused ultrasound depends
through the right heart and selectively engage left and right pulmonary on administration of systemic Gd contrast for lesion conspicuity.
arteries.6 iCMR-guided cardiac catheterization is now classified as a Maximum safe dose of Gd contrast therefore precludes repeated abla-
standard medical procedure at the National Institutes of Health for all tion and imaging cycles. Although this may be acceptable for large solid
patients requiring right-heart catheterization, unless there is a clear tumor ablation, it is not helpful for more targeted and complex ablation
contraindication (e.g., a pacemaker or other MRI-unsafe metallic procedures, such as arrhythmia ablation in the heart.
implant). As mentioned before, iCMR-guided catheterization is of par- Minimally invasive procedures involving the myocardium are par-
ticular value in pediatric patients for reducing exposure to ionizing ticularly amenable to CMR guidance because of the high contrast between
radiation. At Children’s National Medical Center, most routine right myocardium and blood, and because of the readily obtained contrast
heart catheterizations for cardiac transplant patients are now performed between normal and pathologic myocardial tissue. Unfortunately, con-
under CMR guidance. Combining a full CMR study with invasive temporary approaches to the use of CMR to treat rhythm disorders
hemodynamics provides accurate measurement of key parameters, such fail to address the key shortcoming of RF ablation, that without surgical
as pulmonary vascular resistance110 and right ventricular (RV) function. exposure, there is no interactive visualization of irreversibly necrotic
Additional scans can be easily added to the scanning protocol, myocardium after ablation. Edema lesions take minutes to appear and

A B
FIG. 47.12  Coarctation stent angioplasty in a 17-year-old patient. (A) X-ray fused with cardiovascular magnetic
resonance (XFM) coregistration confirmation with contrast aortogram. (B) Coarctation stent angioplasty under
XFM guidance allows for accurate stent positioning to avoid left subclavian artery occlusion. (Courtesy Elena
Grant et al., Children’s National Health System, Washington, DC.)
552 SECTION VI Interventional

A B

C D
FIG. 47.13  Real-time interventional cardioavascular magnetic resonance (iCMR) guided right-heart catheter-
ization using passive catheters. Nonbraided balloon wedge end-hole catheters are filled with dilute gadolinium.
The balloon at the tip of the catheter appears as a white ball on real-time CMR (arrow). Catheter in the
inferior (A) and superior (B) vena cavae, right ventricle (C), and right pulmonary artery (D).

exaggerate the zone of irreversible conduction block.123 As an alternative however, these roadmaps are subject to intrinsic registration errors, to
to RF energy, Kholmovsky and colleagues recently demonstrated cryo- nonperiodic cardiac and respiratory motion,128 to alterations in volume
ablation lesions during real-time iCMR.124 Using active needle-tip cath- as loading conditions change, and to catheter-induced geometric distor-
eters and passive visualization deflectable sheathes, originally designed tion. That said, electroanatomic mapping systems have become suffi-
for cell delivery, our laboratory demonstrated feasibility of real-time ciently sophisticated that near-zero fluoroscopy guided electrophysiology
iCMR guided myocardial chemoablation using Gd-doped caustic agents procedures are now widely attainable.129,130
such as ethanol or acetic acid.125 iCMR enabled real-time visualization Catheter treatment of atrial fibrillation is performed by creating
of chemoablation lesions as they were created; needle delivery of caustic lines of ablation to isolate all four pulmonary veins. Even in experi-
agents into the thick ventricular myocardium created fully transmural enced hands, these procedures, guided by x-ray fluoroscopy and elec-
lesions; and acetic acid caused immediate tissue necrosis with very tromagnetic mapping, usually require hours of radiation exposure. Of
homogenous and well-circumscribed lesions. Real-time iCMR guided note, “image-guided” treatment of atrial fibrillation, conducted under
chemoablation could improve the efficacy of ventricular tachycardia direct surgical exposure, can take minutes. It is tantalizing to speculate
ablation by ensuring accurate targeting of pathological myocardial that comparable image-guided treatment of atrial fibrillation might
substrate with fully transmural and irreversible lesions. be afforded by real-time iCMR guidance without surgical exposure.
Real-time iCMR guidance systems using actively tracked catheters and
Cardiac Electrophysiology filtered local electrograms have been developed.131 Preclinical experiments
Therapeutic endomyocardial catheter ablation is widely performed using have demonstrated close correlation between LGE and electroanatomic
endomyocardial mapping systems and x-ray fluoroscopy to abolish atrial maps, and feasibility of performing electrophysiology studies with real-
and ventricular tachyarrhythmia. In these procedures, a mapping catheter time CMR guidance.132–135 Using an early generation iCMR-conditional
is advanced into the cardiac chambers, guided by endocardial electro- ablation system and passively visualized catheters, ablation of simple
gram patterns to localize the arrhythmia. Key targets are subjected to atrial arrhythmias, such as atrial flutter, was successfully performed in
RF or cryoablation to create nonconductive zones to abolish the arrhyth- patients in Europe (Fig. 47.14).136,137 Newer generation systems provide
mia. Because available imaging modalities afford poor visualization of more accurate catheter tracking through active visualization, combin-
tissue and anatomic structures, these procedures can be challenging ing 3D electroanatomic maps, LGE and real-time iCMR, which should
and time consuming. Roadmaps created using prior electromagnetic permit electrophysiologists to target more complex arrhythmias such
maps, CMR, or CT can be used to fuse with updated catheter images126,127; as atrial fibrillation or ventricular tachycardia (Fig. 47.15).138,139 CMR
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 553

AA

MV MV
RA

RV RA RA

A B C
FIG. 47.14  Interventional cardiovascular magnetic resonance (iCMR) guided ablation using passive cath-
eters. The catheter tip, positioned at the level of the cavo–tricuspid isthmus, contains a passive marker for
CMR conspicuity (arrow). The newly created ablation lesions (arrows) are visible using T2-weighted (B) and
late gadolinium enhancement (C) images. AA, Ascending aorta; MV, mitral valve; RA, right atrium; RV, right
ventricle. (Courtesy M. Grothoff, M. Gutberlet, and G. Hindricks, Departments of Radiology and Cardiology
of the Heart Center Leipzig, University Leipzig.)

electrophysiology is a fertile topic and much progress can be expected in the


A coming years.
Tricuspid valve
annulus CONCLUSION
Real-time CMR combined with CMR conspicuous devices may offer
a complete imaging solution for therapeutic cardiovascular interven-
tions. Superior tissue imaging, no ionizing radiation or nephrotoxic
Coronary contrast, imaging based physiology assessment, and 3D perspective are
Late sinus among many advantages over existing guidance modalities. The require-
IVC ment for conspicuous commercial-grade catheter devices remains a
[ms] challenge. Nonetheless, minimally invasive and novel therapeutic inter-
ventions once considered impossible with traditional imaging may now
Early
be possible using this rapidly evolving technology.

B
Tricuspid valve
annulus
REFERENCES
A full reference list is available online at ExpertConsult.com

Coronary
sinus

3.5
Signal
intensity/
3.4 StdDev IVC
from blood
pool mean
3.3

FIG. 47.15  Interventional cardiovascular magnetic resonance (iCMR)-


guided ablation using active catheters. Screenshot of an iCMR-electro-
physiology user interface showing an activation map acquired during
coronary sinus pacing and before ablation. White dots are planned ablation
points and red dots are delivered ablation sites. Catheters are depicted
on the three-dimensional (3D) image of the right atrium with the pacing
catheter in the coronary sinus and the ablation catheter in the inferior vena
cava. Smaller panels on the right display CMR images in multiple planes
that change automatically to follow the ablation catheter. (B) Maximum
intensity projection 3D late gadolinium enhancement map of the right
atrium 3 months postablation. Scar is thresholded approximately three
standard deviations above the blood pool mean. IVC, Inferior vena cava.
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 553.e1

REFERENCES 23. Duerk JL, Lewin JS, Wu DH. Application of keyhole imaging to
interventional MRI: a simulation study to predict sequence
1. American Academy of Pediatrics CoEH. Risk of ionizing radiation requirements. J Magn Reson Imaging. 1996;6:918–924.
exposure to children: a subject review. Pediatrics. 1998;101:717–719. 24. Harvey PR. A keyhole echo planar hybrid imaging technique. MAGMA.
2. Johnson JN, Hornik CP, Li JS, et al. Cumulative radiation exposure and 1999;9:16–20.
cancer risk estimation in children with heart disease. Circulation. 25. Hwang KP, Lim J, Wendt M, Merkle E, Lewin JS, Duerk JL. Improved
2014;130:161–167. device definition in interventional magnetic resonance imaging using a
3. Klein LW, Tra Y, Garratt KN, et al. Occupational health hazards of rotated stripes keyhole acquisition. Magn Reson Med. 1999;42:554–560.
interventional cardiologists in the current decade: results of the 2014 26. Pruessmann KP, Weiger M, Scheidegger MB, Boesiger P. SENSE:
SCAI membership survey. Catheter Cardiovasc Interv. 2015;86:913–924. sensitivity encoding for fast MRI. Magn Reson Med. 1999;42:952–962.
4. Adam G, Neuerburg J, Bucker A, et al. Interventional magnetic 27. Sodickson DK, Manning WJ. Simultaneous acquisition of spatial
resonance. Initial clinical experience with a 1.5-tesla magnetic resonance harmonics (SMASH): fast imaging with radiofrequency coil arrays.
system combined with c-arm fluoroscopy. Invest Radiol. 1997;32: Magn Reson Med. 1997;38:591–603.
191–197. 28. Griswold MA, Jakob PM, Heidemann RM, et al. Generalized
5. Silverman SG, Jolesz FA, Newman RW, et al. Design and implementation autocalibrating partially parallel acquisitions (GRAPPA). Magn Reson
of an interventional MR imaging suite. AJR Am J Roentgenol. Med. 2002;47:1202–1210.
1997;168:1465–1471. 29. Busse RF, Kruger DG, Debbins JP, Fain SB, Riederer SJ. A flexible view
6. Ratnayaka K, Faranesh AZ, Hansen MS, et al. Real-time MRI-guided ordering technique for high-quality real-time 2DFT MR fluoroscopy.
right heart catheterization in adults using passive catheters. Eur Heart J. Magn Reson Med. 1999;42:69–81.
2013;34:380–389. 30. Debbins JP, Riederer SJ, Rossman PJ, et al. Cardiac magnetic resonance
7. White MJ, Thornton JS, Hawkes DJ, et al. Design, operation, and safety fluoroscopy. Magn Reson Med. 1996;36:588–595.
of single-room interventional MRI suites: practical experience from two 31. Farzaneh F, Riederer SJ, Lee JN, Tasciyan T, Wright RC, Spritzer CE. MR
centers. J Magn Reson Imaging. 2015;41:34–43. fluoroscopy: initial clinical studies. Radiology. 1989;171:545–549.
8. Gregory TS, Schmidt EJ, Zhang SH, Ho Tse ZT. 3DQRS: a method to 32. Rasche V, Holz D, Proksa R. MR fluoroscopy using projection
obtain reliable QRS complex detection within high field MRI using reconstruction multi-gradient-echo (prMGE) MRI. Magn Reson Med.
12-lead electrocardiogram traces. Magn Reson Med. 2014;71:1374–1380. 1999;42:324–334.
9. Tse ZT, Dumoulin CL, Clifford GD, et al. A 1.5T MRI-conditional 33. Riederer SJ, Tasciyan T, Farzaneh F, Lee JN, Wright RC, Herfkens RJ. MR
12-lead electrocardiogram for MRI and intra-MR intervention. Magn fluoroscopy: technical feasibility. Magn Reson Med. 1988;8:1–15.
Reson Med. 2014;71:1336–1347. 34. Nayak KS, Pauly JM, Kerr AB, Hu BS, Nishimura DG. Real-time color
10. White MJ, Thornton JS, Hawkes DJ, et al. Design, operation, and safety flow MRI. Magn Reson Med. 2000;43:251–258.
of single-room interventional MRI suites: practical experience from two 35. Elgort DR, Wong EY, Hillenbrand CM, Wacker FK, Lewin JS, Duerk JL.
centers. J Magn Reson Imaging. 2014. Real-time catheter tracking and adaptive imaging. J Magn Reson
11. Meyer BC, Brost A, Kraitchman DL, et al. Percutaneous punctures with Imaging. 2003;18:621–626.
MR imaging guidance: comparison between MR imaging-enhanced 36. Wang Y, Truong TN, Yen C, et al. Quantitative evaluation of
fluoroscopic guidance and real-time MR imaging guidance. Radiology. susceptibility and shielding effects of nitinol, platinum, cobalt-alloy, and
2013;266:912–919. stainless steel stents. Magn Reson Med. 2003;49:972–976.
12. Guttman MA, Ozturk C, Raval AN, et al. Interventional cardiovascular 37. Buecker A, Spuentrup E, Schmitz-Rode T, et al. Use of a nonmetallic
procedures guided by real-time MR imaging: an interactive interface guide wire for magnetic resonance-guided coronary artery
using multiple slices, adaptive projection modes and live 3D renderings. catheterization. Invest Radiol. 2004;39:656–660.
J Magn Reson Imaging. 2007;26:1429–1435. 38. Raval AN, Telep JD, Guttman MA, et al. Real-time magnetic resonance
13. Mazal JR, Rogers T, Schenke WH, et al. Interventional-cardiovascular imaging-guided stenting of aortic coarctation with commercially
MR: role of the interventional MR technologist. Radiol Technol. available catheter devices in Swine. Circulation. 2005;112:699–706.
2016;87:261–270. 39. Wacker FK, Reither K, Branding G, Wendt M, Wolf KJ. Magnetic
14. Atalar E. Radiofrequency safety for interventional MRI procedures. Acad resonance-guided vascular catheterization: feasibility using a passive
Radiol. 2005;12:1149–1157. tracking technique at 0.2 Tesla in a pig model. J Magn Reson Imaging.
15. Campbell-Washburn AE, Faranesh AZ, Lederman RJ, Hansen MS. 1999;10:841–844.
Magnetic resonance sequences and rapid acquisition for MR-guided 40. Kochli VD, McKinnon GC, Hofmann E, von Schulthess GK. Vascular
interventions. Magn Reson Imaging Clin N Am. 2015;23:669–679. interventions guided by ultrafast MR imaging: evaluation of different
16. Guttman MA, Kellman P, Dick AJ, Lederman RJ, McVeigh ER. Real-time materials. Magn Reson Med. 1994;31:309–314.
accelerated interactive MRI with adaptive TSENSE and UNFOLD. Magn 41. Razavi R, Hill DL, Keevil SF, et al. Cardiac catheterisation guided by
Reson Med. 2003;50:315–321. MRI in children and adults with congenital heart disease. Lancet.
17. Madore B, Glover GH, Pelc NJ. Unaliasing by fourier-encoding the 2003;362:1877–1882.
overlaps using the temporal dimension (UNFOLD), applied to cardiac 42. Miquel ME, Hegde S, Muthurangu V, et al. Visualization and tracking of
imaging and fMRI. Magn Reson Med. 1999;42:813–828. an inflatable balloon catheter using SSFP in a flow phantom and in the
18. van Vaals JJ, Brummer ME, Dixon WT, et al. “Keyhole” method for heart and great vessels of patients. Magn Reson Med. 2004;51:988–995.
accelerating imaging of contrast agent uptake. J Magn Reson Imaging. 43. Kuehne T, Yilmaz S, Schulze-Neick I, et al. Magnetic resonance imaging
1993;3:671–675. guided catheterisation for assessment of pulmonary vascular resistance:
19. Peters DC, Lederman RJ, Dick AJ, et al. Undersampled projection in vivo validation and clinical application in patients with pulmonary
reconstruction for active catheter imaging with adaptable temporal hypertension. Heart. 2005;91:1064–1069.
resolution and catheter-only views. Magn Reson Med. 2003;49:216–222. 44. Wolska-Krawczyk M, Rube MA, Immel E, Melzer A, Buecker A. Heating
20. Rasche V, de Boer RW, Holz D, Proksa R. Continuous radial data and safety of a new MR-compatible guidewire prototype versus a
acquisition for dynamic MRI. Magn Reson Med. 1995;34:754–761. standard nitinol guidewire. Radiol Phys Technol. 2014;7:95–101.
21. Boll DT, Merkle EM, Seaman DM, et al. Comparison of ECG-gated 45. Kramer NA, Kruger S, Schmitz S, et al. Preclinical evaluation of a novel
rectilinear vs. real-time radial K-space sampling schemes in cine fiber compound MR guidewire in vivo. Invest Radiol. 2009;44:390–397.
True-FISP cardiac MRI. J Cardiovasc Magn Reson. 2004;6:793–802. 46. Mekle R, Hofmann E, Scheffler K, Bilecen D. A polymer-based
22. Schaeffter T, Rasche V, Bornert P, Mens G. Interactive reduced FOV MR-compatible guidewire: a study to explore new prospects for
imaging for projection reconstruction and spiral acquisition. Magn interventional peripheral magnetic resonance angiography (ipMRA). J
Reson Imaging. 2001;19:677–684. Magn Reson Imaging. 2006;23:145–155.
553.e2 SECTION VI Interventional

47. Tzifa A, Krombach GA, Kramer N, et al. Magnetic resonance-guided 69. Settecase F, Sussman MS, Wilson MW, et al. Magnetically-assisted
cardiac interventions using magnetic resonance-compatible devices: a remote control (MARC) steering of endovascular catheters for
preclinical study and first-in-man congenital interventions. Circ interventional MRI: a model for deflection and design implications. Med
Cardiovasc Interv. 2010;3:585–592. Phys. 2007;34:3135–3142.
48. Basar B, Rogers T, Ratnayaka K, et al. Segmented nitinol guidewires with 70. Wilson MW, Martin AB, Lillaney P, et al. Magnetic catheter
stiffness-matched connectors for cardiovascular magnetic resonance manipulation in the interventional MR imaging environment. J Vasc
catheterization: preserved mechanical performance and freedom from Interv Radiol. 2013;24:885–891.
heating. J Cardiovasc Magn Reson. 2015;17:105. 71. Losey AD, Lillaney P, Martin AJ, et al. Magnetically assisted remote-
49. Omary RA, Unal O, Koscielski DS, et al. Real-time MR imaging-guided controlled endovascular catheter for interventional MR imaging: in vitro
passive catheter tracking with use of gadolinium-filled catheters. J Vasc navigation at 1.5 T versus X-ray fluoroscopy. Radiology.
Interv Radiol. 2000;11:1079–1085. 2014;271:862–869.
50. Unal O, Korosec FR, Frayne R, Strother CM, Mistretta CA. A rapid 2D 72. Lillaney PV, Yang JK, Losey AD, et al. Endovascular MR-guided renal
time-resolved variable-rate k-space sampling MR technique for passive embolization by using a magnetically assisted remote-controlled
catheter tracking during endovascular procedures. Magn Reson Med. catheter system. Radiology. 2016;281(1):219–228. doi:10.1148/
1998;40:356–362. radiol.2016152036.
51. Strother CM, Unal O, Frayne R, et al. Endovascular treatment of 73. Moftakhar P, Lillaney P, Losey AD, et al. New-generation laser-
experimental canine aneurysms: feasibility with MR imaging guidance. lithographed dual-axis magnetically assisted remote-controlled
Radiology. 2000;215:516–519. endovascular catheter for interventional MR imaging: in vitro
52. Bakker CJ, Hoogeveen RM, Weber J, van Vaals JJ, Viergever MA, Mali multiplanar navigation at 1.5 T and 3 T versus x-ray fluoroscopy.
WP. Visualization of dedicated catheters using fast scanning techniques Radiology. 2015;277:842–852.
with potential for MR-guided vascular interventions. Magn Reson Med. 74. Kee ST, Rhee JS, Butts K, et al. 1999 Gary J. Becker Young Investigator
1996;36:816–820. Award. MR-guided transjugular portosystemic shunt placement in a
53. Kozerke S, Hegde S, Schaeffter T, Lamerichs R, Razavi R, Hill DL. swine model. J Vasc Interv Radiol. 1999;10:529–535.
Catheter tracking and visualization using 19F nuclear magnetic 75. Kee ST, Ganguly A, Daniel BL, et al. MR-guided transjugular
resonance. Magn Reson Med. 2004;52:693–697. intrahepatic portosystemic shunt creation with use of a hybrid
54. Mansson S, Johansson E, Magnusson P, et al. 13C imaging-a new radiography/MR system. J Vasc Interv Radiol. 2005;16:227–234.
diagnostic platform. Eur Radiol. 2005. 76. Arepally A, Karmarkar PV, Weiss C, Atalar E. Percutaneous MR
55. Campbell-Washburn A, Rogers T, Ratnayaka K, et al. Spiral imaging imaging-guided transvascular access of mesenteric venous system: study
with off-resonance reconstruction for MRI-guided cardiovascular in swine model. Radiology. 2006;238:113–118.
catheterizations using commercial off-the-shelf nitinol guidewires. J 77. Ratnayaka K, Rogers T, Schenke WH, et al. Magnetic resonance
Cardiovasc Magn Reson. 2015;17(suppl 1):Q15. imaging-guided transcatheter cavopulmonary shunt. JACC Cardiovasc
56. Campbell-Washburn AE, Rogers T, Basar B, et al. Positive contrast spiral Interv. 2016;9:959–970.
imaging for visualization of commercial nitinol guidewires with reduced 78. Lossnitzer D, Seitz SA, Krautz B, et al. Feasibility of real-time magnetic
heating. J Cardiovasc Magn Reson. 2015;17:114. resonance imaging-guided endomyocardial biopsies: an in-vitro study.
57. Krueger JJ, Ewert P, Yilmaz S, et al. Magnetic resonance imaging-guided World J Cardiol. 2015;7:415–422.
balloon angioplasty of coarctation of the aorta: a pilot study. Circulation. 79. Rogers T, Ratnayaka K, Karmakar P, et al. Real-time magnetic resonance
2006;113:1093–1100. imaging guidance improves the diagnostic yield of endomyocardial
58. Pushparajah K, Tzifa A, Bell A, et al. Cardiovascular magnetic resonance biopsy. JACC Basic Transl Sci. 2016;in press.
catheterization derived pulmonary vascular resistance and medium-term 80. Raman VK, Karmarkar PV, Guttman MA, et al. Real-time magnetic
outcomes in congenital heart disease. J Cardiovasc Magn Reson. resonance-guided endovascular repair of experimental abdominal
2015;17:28. aortic aneurysm in swine. J Am Coll Cardiol. 2005;45:2069–
59. Dumoulin CL, Souza SP, Darrow RD. Real-time position monitoring of 2077.
invasive devices using magnetic resonance. Magn Reson Med. 81. Eggebrecht H, Kuhl H, Kaiser GM, et al. Feasibility of real-time
1993;29:411–415. magnetic resonance-guided stent-graft placement in a swine model of
60. McKinnon GC, Debatin JF, Leung DA, Wildermuth S, Holtz DJ, von descending aortic dissection. Eur Heart J. 2006;27:613–620.
Schulthess GK. Towards active guidewire visualization in interventional 82. Arepally A, Karmarkar PV, Weiss C, Rodriguez ER, Lederman RJ, Atalar
magnetic resonance imaging. MAGMA. 1996;4:13–18. E. Magnetic resonance image-guided trans-septal puncture in a swine
61. Ocali O, Atalar E. Intravascular magnetic resonance imaging using a heart. J Magn Reson Imaging. 2005;21:463–467.
loopless catheter antenna. Magn Reson Med. 1997;37:112–118. 83. Raval AN, Karmarkar PV, Guttman MA, et al. Real-time MRI guided
62. Ladd ME, Zimmermann GG, Quick HH, et al. Active MR visualization atrial septal puncture and balloon septostomy in swine. Catheter
of a vascular guidewire in vivo. J Magn Reson Imaging. 1998;8:220–225. Cardiovasc Interv. 2006;67:637–643.
63. Glowinski A, Adam G, Bucker A, Neuerburg J, van Vaals JJ, Gunther RW. 84. Rickers C, Jerosch-Herold M, Hu X, et al. Magnetic resonance
Catheter visualization using locally induced, actively controlled field image-guided transcatheter closure of atrial septal defects. Circulation.
inhomogeneities. Magn Reson Med. 1997;38:253–258. 2003;107:132–138.
64. Quick HH, Kuehl H, Kaiser G, Bosk S, Debatin JF, Ladd ME. Inductively 85. Buecker A, Spuentrup E, Grabitz R, et al. Magnetic resonance-guided
coupled stent antennas in MRI. Magn Reson Med. 2002;48:781–790. placement of atrial septal closure device in animal model of patent
65. Quick HH, Zenge MO, Kuehl H, et al. Interventional magnetic foramen ovale. Circulation. 2002;106:511–515.
resonance angiography with no strings attached: wireless active catheter 86. Halabi M, Ratnayaka K, Faranesh AZ, et al. Transthoracic delivery of
visualization. Magn Reson Med. 2005;53:446–455. large devices into the left ventricle through the right ventricle and
66. Bell JA, Saikus CE, Ratnayaka K, et al. Active delivery cable tuned to interventricular septum: preclinical feasibility. J Cardiovasc Magn Reson.
device deployment state: enhanced visibility of nitinol occluders during 2013;15:10.
preclinical interventional MRI. J Magn Reson Imaging. 2012;36:972–978. 87. Barbash IM, Saikus CE, Faranesh AZ, et al. Direct percutaneous left
67. Srinivasan S, Ennis DB. Variable flip angle balanced steady-state free ventricular access and port closure: pre-clinical feasibility. JACC
precession for lower SAR or higher contrast cardiac cine imaging. Magn Cardiovasc Interv. 2011;4:1318–1325.
Reson Med. 2014;71:1035–1043. 88. Rogers T, Ratnayaka K, Schenke WH, et al. Fully percutaneous
68. Wu X, Akgun C, Vaughan JT, et al. Adapted RF pulse design for SAR transthoracic left atrial entry and closure as a potential access route for
reduction in parallel excitation with experimental verification at 9.4 T. J transcatheter mitral valve interventions. Circ Cardiovasc Interv.
Magn Reson. 2010;205:161–170. 2015;8:e002538.
CHAPTER 47  Interventional Cardiovascular Magnetic Resonance 553.e3

89. Ratnayaka K, Saikus CE, Faranesh AZ, et al. Closed-chest transthoracic invasive pressure monitoring and phase-contrast magnetic resonance
magnetic resonance imaging-guided ventricular septal defect closure in flow. Circulation. 2004;110:826–834.
swine. JACC Cardiovasc Interv. 2011;4:1326–1334. 111. Schoenfeld C, Cebotari S, Hinrichs J, et al. MR imaging-derived regional
90. Halabi M, Faranesh AZ, Schenke WH, et al. Real-time cardiovascular pulmonary parenchymal perfusion and cardiac function for monitoring
magnetic resonance subxiphoid pericardial access and pericardiocentesis patients with chronic thromboembolic pulmonary hypertension before
using off-the-shelf devices in swine. J Cardiovasc Magn Reson. and after pulmonary endarterectomy. Radiology. 2016;279:925–934.
2013;15:61. 112. Rogers T, Ratnayaka K, Lederman RJ. MRI catheterization in
91. Omary RA, Green JD, Schirf BE, Li Y, Finn JP, Li D. Real-time magnetic cardiopulmonary disease. Chest. 2014;145:30–36.
resonance imaging-guided coronary catheterization in swine. 113. Claessen G, La Gerche A, Wielandts JY, et al. Exercise pathophysiology
Circulation. 2003;107:2656–2659. and sildenafil effects in chronic thromboembolic pulmonary
92. Serfaty JM, Yang X, Foo TK, Kumar A, Derbyshire A, Atalar E. hypertension. Heart. 2015;101:637–644.
MRI-guided coronary catheterization and PTCA: a feasibility study on a 114. La Gerche A, Claessen G, Dymarkowski S, et al. Exercise-induced right
dog model. Magn Reson Med. 2003;49:258–263. ventricular dysfunction is associated with ventricular arrhythmias in
93. Spuentrup E, Ruebben A, Schaeffter T, Manning WJ, Gunther RW, endurance athletes. Eur Heart J. 2015;36:1998–2010.
Buecker A. Magnetic resonance–guided coronary artery stent placement 115. Claessen G, La Gerche A, Dymarkowski S, Claus P, Delcroix M,
in a swine model. Circulation. 2002;105:874–879. Heidbuchel H. Pulmonary vascular and right ventricular reserve in
94. Wildermuth S, Dumoulin CL, Pfammatter T, Maier SE, Hofmann E, patients with normalized resting hemodynamics after pulmonary
Debatin JF. MR-guided percutaneous angioplasty: assessment of tracking endarterectomy. J Am Heart Assoc. 2015;4:e001602.
safety, catheter handling and functionality. Cardiovasc Intervent Radiol. 116. Dick AJ, Guttman MA, Raman VK, et al. Magnetic resonance
1998;21:404–410. fluoroscopy allows targeted delivery of mesenchymal stem cells to infarct
95. Feng L, Dumoulin CL, Dashnaw S, et al. Feasibility of stent placement in borders in Swine. Circulation. 2003;108:2899–2904.
carotid arteries with real-time MR imaging guidance in pigs. Radiology. 117. Kraitchman DL, Heldman AW, Atalar E, et al. In vivo magnetic
2005;234:558–562. resonance imaging of mesenchymal stem cells in myocardial infarction.
96. Yang X, Bolster BD Jr, Kraitchman DL, Atalar E. Intravascular MR Circulation. 2003;107:2290–2293.
monitored balloon angioplasty: an in vivo feasibility study. J Vasc Interv 118. Corti R, Badimon J, Mizsei G, et al. Real time magnetic resonance
Radiol. 1998;9:953–959. guided endomyocardial local delivery. Heart. 2005;91:348–353.
97. Buecker A, Adam GB, Neuerburg JM, et al. Simultaneous real-time 119. Krombach GA, Pfeffer JG, Kinzel S, Katoh M, Gunther RW, Buecker A.
visualization of the catheter tip and vascular anatomy for MR-guided MR-guided percutaneous intramyocardial injection with an
PTA of iliac arteries in an animal model. J Magn Reson Imaging. MR-compatible catheter: feasibility and changes in T1 values after
2002;16:201–208. injection of extracellular contrast medium in pigs. Radiology.
98. Godart F, Beregi JP, Nicol L, et al. MR-guided balloon angioplasty of 2005;235:487–494.
stenosed aorta: in vivo evaluation using near-standard instruments and 120. Saeed M, Lee R, Martin A, et al. Transendocardial delivery of
a passive tracking technique. J Magn Reson Imaging. 2000;12:639–644. extracellular myocardial markers by using combination X-ray/MR
99. Omary RA, Frayne R, Unal O, et al. MR-guided angioplasty of renal fluoroscopic guidance: feasibility study in dogs. Radiology.
artery stenosis in a pig model: a feasibility study. J Vasc Interv Radiol. 2004;231:689–696.
2000;11:373–381. 121. Rickers C, Gallegos R, Seethamraju RT, et al. Applications of magnetic
100. Paetzel C, Zorger N, Seitz J, et al. Intraarterial contrast material- resonance imaging for cardiac stem cell therapy. J Interv Cardiol.
enhanced magnetic resonance angiography of the aortoiliac system. J 2004;17:37–46.
Vasc Interv Radiol. 2004;15:981–984. 122. Saeed M, Krug R, Do L, Hetts SW, Wilson MW. Renal ablation using
101. Buecker A, Neuerburg JM, Adam GB, et al. Real-time MR fluoroscopy magnetic resonance-guided high intensity focused ultrasound: magnetic
for MR-guided iliac artery stent placement. J Magn Reson Imaging. resonance imaging and histopathology assessment. World J Radiol.
2000;12:616–622. 2016;8:298–307.
102. Dion YM, Ben El Kadi H, Boudoux C, et al. Endovascular procedures 123. Lardo AC, McVeigh ER, Jumrussirikul P, et al. Visualization and
under near-real-time magnetic resonance imaging guidance: an temporal/spatial characterization of cardiac radiofrequency ablation
experimental feasibility study. J Vasc Surg. 2000;32:1006–1014. lesions using magnetic resonance imaging. Circulation. 2000;102:698–705.
103. Mahnken AH, Gunther RW, Tacke J. Radiofrequency ablation of renal 124. Kholmovski EG, Coulombe N, Silvernagel J, et al. Real-time MRI-guided
tumors. Eur Radiol. 2004;14:1449–1455. cardiac cryo-ablation: a feasibility study. J Cardiovasc Electrophysiol.
104. Manke C, Nitz WR, Djavidani B, et al. MR imaging-guided stent 2016;27:602–608.
placement in iliac arterial stenoses: a feasibility study. Radiology. 125. Rogers T, Mahapatra S, Kim S, et al. Transcatheter myocardial needle
2001;219:527–534. chemoablation during real-time magnetic resonance imaging: a new
105. Raval AN, Karmarkar PV, Guttman MA, et al. Real-time magnetic approach to ablation therapy for rhythm disorders. Circ Arrhythm
resonance imaging-guided endovascular recanalization of chronic total Electrophysiol. 2016;9.
arterial occlusion in a swine model. Circulation. 2006;113:1101–1107. 126. Sermesant M, Rhode K, Sanchez-Ortiz GI, et al. Simulation of cardiac
106. Choi J, Radau P, Xu R, Wright GA. X-ray and magnetic resonance pathologies using an electromechanical biventricular model and XMR
imaging fusion for cardiac resynchronization therapy. Med Image Anal. interventional imaging. Med Image Anal. 2005;9:467–480.
2016;31:98–107. 127. Reddy VY, Malchano ZJ, Holmvang G, et al. Integration of cardiac
107. Abu Hazeem AA, Dori Y, Whitehead KK, et al. X-ray magnetic magnetic resonance imaging with three-dimensional electroanatomic
resonance fusion modality may reduce radiation exposure and contrast mapping to guide left ventricular catheter manipulation: feasibility in a
dose in diagnostic cardiac catheterization of congenital heart disease. porcine model of healed myocardial infarction. J Am Coll Cardiol.
Catheter Cardiovasc Interv. 2014;84:795–800. 2004;44:2202–2213.
108. Xu R, Athavale P, Nachman A, Wright GA. Multiscale registration of 128. Noseworthy PA, Malchano ZJ, Ahmed J, Holmvang G, Ruskin JN, Reddy
real-time and prior MRI data for image-guided cardiac interventions. VY. The impact of respiration on left atrial and pulmonary venous
IEEE Trans Biomed Eng. 2014;61:2621–2632. anatomy: implications for image-guided intervention. Heart Rhythm.
109. Faranesh AZ, Kellman P, Ratnayaka K, Lederman RJ. Integration of 2005;2:1173–1178.
cardiac and respiratory motion into MRI roadmaps fused with x-ray. 129. Cano O, Andres A, Osca J, et al. Safety and feasibility of a minimally
Med Phys. 2013;40:032302. fluoroscopic approach for ventricular tachycardia ablation in patients
110. Muthurangu V, Taylor A, Andriantsimiavona R, et al. Novel method of with structural heart disease: influence of the ventricular tachycardia
quantifying pulmonary vascular resistance by use of simultaneous substrate. Circ Arrhythm Electrophysiol. 2016;9:e003706.
553.e4 SECTION VI Interventional

130. Clark BC, Sumihara K, McCarter R, Berul CI, Moak JP. Getting to zero: resonance guided electrophysiology system. J Cardiovasc Magn Reson.
impact of electroanatomical mapping on fluoroscopy use in pediatric 2015;17:27.
catheter ablation. J Interv Card Electrophysiol. 2016. 136. Piorkowski C, Grothoff M, Gaspar T, et al. Cavotricuspid isthmus
131. Susil RC, Yeung CJ, Halperin HR, Lardo AC, Atalar E. Multifunctional ablation guided by real-time magnetic resonance imaging. Circ
interventional devices for MRI: a combined electrophysiology/MRI Arrhythm Electrophysiol. 2013;6:e7–e10.
catheter. Magn Reson Med. 2002;47:594–600. 137. Grothoff M, Piorkowski C, Eitel C, et al. MR imaging-guided
132. Ashikaga H, Sasano T, Dong J, et al. Magnetic resonance-based electrophysiological ablation studies in humans with passive catheter
anatomical analysis of scar-related ventricular tachycardia: implications tracking: initial results. Radiology. 2014;271:695–702.
for catheter ablation. Circ Res. 2007;101:939–947. 138. Hilbert S, Sommer P, Gutberlet M, et al. Real-time magnetic resonance-
133. Nazarian S, Kolandaivelu A, Zviman MM, et al. Feasibility of real-time guided ablation of typical right atrial flutter using a combination of
magnetic resonance imaging for catheter guidance in electrophysiology active catheter tracking and passive catheter visualization in man: initial
studies. Circulation. 2008;118:223–229. results from a consecutive patient series. Europace. 2016;18:572–577.
134. Dukkipati SR, Mallozzi R, Schmidt EJ, et al. Electroanatomic mapping 139. Krahn PRP, Singh SM, Ramanan V, et al. Cardiovascular magnetic
of the left ventricle in a porcine model of chronic myocardial infarction resonance guided ablation and intra-procedural visualization of evolving
with magnetic resonance-based catheter tracking. Circulation. radiofrequency lesions in the left ventricle. J Cardiovasc Magn Reson.
2008;118:853–862. 2018;20:20.
135. Oduneye SO, Pop M, Shurrab M, et al. Distribution of abnormal 140. Yeung CJ, Karmarkar P, McVeigh ER. Minimizing RF heating of
potentials in chronic myocardial infarction using a real time magnetic conducting wires in MRI. Magn Reson Med. 2007;58:1028–1034.
48 
Pediatric Interventional Cardiovascular
Magnetic Resonance
Kuberan Pushparajah and Reza S. Razavi

The last two and a half decades have seen phenomenal advances made the aid of new parallel imaging techniques, while maintaining suitable
in the field of cardiovascular magnetic resonance (CMR), and these spatial resolution for interventional applications.29–34
advances have supported research into interventional applications Despite the inherent potential and promise of CMR-guided inter-
using CMR.1–4 Conventional x-ray fluoroscopically guided cardiac ventions and operations, there are still obstacles associated with per-
catheterization and interventions are associated with the risk of expo- forming the complete procedure in the CMR scanner, particularly
sure to ionizing radiation for both patients and staff. This is particu- because of the lack of CMR-compatible catheters and devices. There-
larly relevant in younger patients, who are often required to undergo fore the initial work in interventional CMR exploited multimodality
multiple procedures. The need for an imaging modality offering mul- imaging, such as x-ray and CMR (XMR) or XMR and ultrasound. Such
tiplanar imaging, superior structural delineation of complex cardiac hybrid units already in existence allow the use of separate modalities
anatomy, and additional physiologic information, without the risk of or a combination of them when needed. Cross-modality image inte-
ionizing radiation, has brought CMR guidance of cardiac catheter- gration, with spatial and temporal information about the anatomy,
ization procedures to the fore. In the last 10 years, clinical programs pathology, and therapy devices, can be provided to the users of these
using CMR-guided cardiac catheterization have developed and show systems. XMR systems, which combine x-ray and CMR by having both
promise.5–8 modalities in the same room with a tabletop design, allow patients
Since the first magnetic resonance (MR) images showing live human to be moved from one modality to the other in less than 1 minute
anatomy were produced,9–11 this technique evolved to enable a variety (Fig. 48.1).35–38
of clinical applications of MR.12,13 Over the years, improvements in Image fusion modalities are now in existence as a commercial product
signal detection, fast data handling, advanced understanding of spin for clinical use. The use of x-ray fused with CMR (XFM) uses previ-
systems, pulse sequences, and artifact suppression have resulted in much ously acquired CMR images and overlays them onto x-ray fluoroscopic
faster scan times and considerable improvements in image resolution.14–22 images in the cardiac catheterization laboratory. These can be aligned
These ultrafast imaging techniques form the basis of real-time imaging, by means of external fiduciary markers or internal anatomical struc-
used for CMR-guided cardiac catheterization. However, the first impor- tures,39,40 with the ability to correct for cardiac and respiratory motion.41
tant step in making CMR cardiac catheterization a clinical reality is the This has already been shown to reduce radiation exposure, screening
design of a suitable interventional CMR system.23–27 time, and use of iodinated contrast agents in selected cases of cardiac
catheterization in congenital heart disease.42 Although not a form of
INTERVENTIONAL CARDIOVASCULAR MAGNETIC solely CMR-guided catheterization, such technology allows the applica-
tion of CMR technology in the cardiac catheter laboratory even in
RESONANCE SYSTEMS nonhybrid suites.
In the design of an interventional CMR suite, it is important to retain
the full capabilities of a state-of-the-art diagnostic scanner without MERITS OF CARDIOVASCULAR MAGNETIC
encumbering the interventionalist or creating a risk of high radiofre- RESONANCE GUIDANCE
quency (RF) or switched magnetic field exposure. Open-magnet designs
allow easier access to the patient, but are not typically available in field Improved Visualization of Cardiac Anatomy
strengths higher than 1 T. The cylindrical horizontal bore systems offer A problem with x-ray-guided cardiac catheterization is the inherent
higher field strengths and gradient slew rates, allowing higher-resolution poor contrast of soft tissues, such as the heart and great vessels. This
imaging, shorter scan times, higher signal-to-noise ratio (SNR), reduced makes it difficult for the cardiologist to manipulate or position guide-
image distortion, and improved functionality with real-time imaging, wires, catheters, balloons, or interventional devices within the heart
all of which are of paramount importance when endovascular interven- and surrounding vessels. A skilled operator usually relies on recognizing
tions are considered.28 A trade off with the traditional cylindrical magnet anatomic structures from previous experience or on contrast angio-
design is access to the patient. More recently, magnets with shorter graphic images acquired earlier in the procedure. The lack of adequate
bores and flared margins have been introduced and offer better patient visualization increases the risk of perforating the heart or great vessels,
access, especially for cardiovascular interventions, without compromis- especially when performing complex interventional procedures.
ing the advanced CMR features of diagnostic scanners. Rapid improve- Certain interventional cardiac procedures involve selection of an
ments in the processing power of computers, along with the use of appropriate cardiac device and its successful deployment within the
powerful and intuitive software, have allowed researchers to develop heart, which requires accurate measurement of the size of defects and
novel strategies for image data acquisition and reconstruction. It is now nearby anatomic structures. Such measurements are possible under
possible to achieve frame rates of as high as 20 images per second with x-ray fluoroscopy (XRF), but can be difficult.

554
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 555

FIG. 48.1  Schematic room plan of a typical x-ray and magnetic resonance (XMR) suite. XMR room with the
x-ray and magnetic resonance (MR) equipment joined by a movable tabletop. The C-arm of the x-ray unit is
seen in the foreground, ceiling-mounted MR monitor and controls are seen in the distance, and the 5-gauss
area is demarcated by a change in the floor coloring from the MR to the x-ray end of the room.

A successful interventional cardiac procedure therefore relies heavily been validated in numerous phantom experiments, allowing for a novel
on adequate visualization of the heart or vessel. This implies the need method of quantification of PVR in patients with pulmonary hyperten-
for superior imaging methods that provide excellent visualization without sion by using invasive pressure measurements and CMR flow data.65–67
increasing the risk to the patient. CMR fits this role very well because This method has since been used in a large clinical case series with
it provides exceptional structural delineation of both the heart and its good results and forms part of routine clinical practice in institutions
surrounding vasculature and therefore allows safe guidance of inter- where this is available.8,68
ventional procedures. Assessment of global and regional ventricular function can also be
carried out much more accurately with cine steady-state free precession
Reduced Ionizing Radiation (SSFP) CMR than with x-ray angiography. When using CMR for assess-
There is a strong case for pediatric cardiac catheterization procedures ing global ventricular function, there is no need to make assumptions
to be made safer, especially in terms of ionizing radiation. According about cardiac geometry, unlike with XRF or even echocardiography.
to the UK National Radiation Protection Board, the mean risk that a This is particularly important when assessing right ventricular (RV)
solid tumor will develop as a result of a single cardiac catheterization function and regional wall motion in the normal or systemic ventricle
procedure is approximately 1 in 2500 in adults. This risk increases to in patients with functionally single ventricle physiology.
1 in 1000 in children if exposure occurs at 5 years of age.43–46 Also, the Finally, combining invasive pressure measurements with CMR-derived
proportion of the body that is irradiated increases as the size of the blood flow and ventricular volumes also opens up interesting new ways
patient decreases, and some procedures in patients with congenital of looking at pathophysiology. It allows for the study of pulmonary
heart disease often require much longer x-ray exposure. These risks vascular compliance, derived ventricular pressure–volume loops, and
are multiplied in children in particular, because they often undergo assessment of load-independent ventricular function.69–72 Pharmaco-
multiple cardiac catheter procedures.47 In addition to the patients, logical stress studies have also been applied in CMR/XMR catheter
there is also a significant risk from ionizing radiation to the staff in studies to assess hemodynamic responses.72–76
the catheter laboratory during these procedures, despite the use of
protective shields.48–51 MAGNETIC INSTRUMENTATION AND
Physiologic Information VISUALIZATION STRATEGIES
Conventional cardiac catheterization is used not only to provide ana- Crucial to the success of interventional CMR is real-time tracking and
tomic information and perform intervention but also to obtain functional visualization of catheters, guidewires, and devices in the CMR environ-
information. Invasive pressures and blood gases are commonly used to ment. Several groups around the world are putting considerable effort
calculate systemic and pulmonary blood flow and resistance with the into developing CMR-suitable catheters and devices. Device localization
Fick principle. XRF angiography is also used to assess global ventricular under CMR is made possible by a variety of approaches that can be
function as well as regional wall motion abnormalities. The functional broadly classified as either electrically passive or electrically active.77
information obtained at cardiac catheterization is used alongside ana-
tomic information to assess patient suitability for surgery or interven- Passive Catheter Tracking and Visualization
tional cardiac catheterization or the need for long-term vasodilator The passive tracking technique is commonly based on visualization of
therapy in patients with pulmonary vascular disease. susceptibility artifacts or signal voids caused by the interventional device
The Fick principle to quantify flow is dependent on multiple mea- under CMR imaging. This is a well-studied technique and to date it is
surements (hemoglobin, aortic/pulmonary artery oxygen saturation, the most clinically feasible (Figs. 48.2 and 48.3).78–82 Passive visualization
partial pressure, oxygen consumption), which can be a considerable often does not require any special hardware or software and therefore
source of inaccuracy. In addition, in patients with large intracardiac it can be performed on any commercial CMR system.
shunts and high pulmonary blood flow, accuracy is further reduced.52–57 The ideal passive tracking catheter or guidewire must be made of
Therefore there is a need for a method of flow quantification that allows a material that provides adequate physical properties such as torque
accurate and reproducible measurement of pulmonary vascular resis- and steerability, and allows tracking without obscuring the underlying
tance (PVR). Velocity encoded phase contrast CMR enables noninvasive anatomy. Ferromagnetic materials cause large susceptibility artifacts
quantification of blood flow in major vessels. Cardiac output and the and therefore are not generally suitable for CMR-guided procedures.
pulmonary-to-systemic flow ratio (Qp:Qs) measured using this tech- This rules out most metals used for making cardiac devices. However,
nique have been shown to be accurate.58–64 In addition, phase CMR has certain alloys, such as nitinol (nickel and titanium), have magnetic
556 SECTION VI Interventional

A B C
FIG. 48.2  Passive tracking. (A) Inflated balloon angiographic Bermann catheter filled with 0.8 mL CO2.
(B) CO2-filled balloon catheter manipulated in a phantom. (C) Dysprosium catheter: a catheter impregnated
with dysprosium oxide is manipulated in an in vitro set-up mimicking endovascular intervention. The catheter
is clearly visualized along its full length, despite being orientated along B0. (A, Courtesy Arrow International,
Reading, PA.)

susceptibility close to that of tissue. Therefore they are best suited for which were used in preclinical and clinical trials and led to successful
making guidewires and braided catheters that are MR compatible but interventions in congenital heart disease, but these were difficult to
not necessarily CMR safe. steer and proved to be fragile.89,90 There continues to be a drive for the
The polymeric materials used for making catheters typically have development of an optimal guidewire and techniques to measure heating
low magnetic susceptibility and therefore cannot be easily localized on risks in vivo have been employed.91 Consequently, a newer nitinol based
CMR images.83 This implies that, if materials with higher susceptibility guidewire with iron oxide markers along the length to impart visibility
can be incorporated into the wall of the catheters or sheaths or the has been developed with good preclinical results.92
lumen filled with a suitable contrast agent, then improved visualization In the case of balloon angiographic catheters, if the balloon is inflated
can be achieved. with carbon dioxide, as is done conventionally with x-ray, then the
One approach to generating susceptibility artifacts is locally impreg- inflated balloon creates a signal void in the CMR image, thus enabling
nating the catheter wall with gadolinium (Gd)-like compounds, such visualization (see Fig. 48.2A and B). This method has been used suc-
as dysprosium oxide, in the form of rings or along the length of the cessfully to guide catheters in patients under CMR (see Fig. 48.3).5,93
catheter during the extrusion process (see Fig. 48.2C).82 Another approach Although this technique allows easy visualization of the tip, the length is
is to use Gd contrast agents in varying concentrations within catheter impossible to visualize because the signal void from the catheter length
lumens84 or impregnated into catheter walls to create either a positive is masked by volume averaging and dephasing effects of thicker slices.94
or negative signal on CMR imaging.85 A similar approach is to inflate the balloon of the angiographic catheter
Metallic devices and guidewires produce susceptibility artifacts, which with a 1% concentration of Gd contrast agent7 and the balloon appears
aid visualization by way of the artifacts, but different metals behave as a white ball because of the signal from the contrast-filled balloon.
differently under CMR. Titanium alloys produce narrower artifacts The success of passive visualization also relies on dedicated scan
compared with ferromagnetic, or even certain other nonferromagnetic techniques. A dynamic gradient echo sequence, such as SSFP, has been
alloys such as nickel–chromium, which can produce large RF and sus- shown to be ideal for passive catheter tracking, especially when signal
ceptibility artifacts. However, commercial guidewires can heat up during voids or susceptibility artifacts are used for visualization.93,95 Cardiac
CMR because of standing wave formation along the conductive parts catheterization under XRF guidance is usually performed at imaging
longer than a quarter wavelength at the resonant frequency, which cor- speeds of 25 to 30 frames per second. The frame rates available for
responds to approximately 12 cm in humans at 1.5 T,86 which is relevant CMR-guided interventions are not comparable because of the post-
for cardiac catheterization where wires are inserted to at least that length processing of CMR images and their subsequent display, allowing a
and nearly always much further. maximum of 10 to 14 frames routinely. Some of the proposed passive
Guidewires with a fiberglass core and nonmetallic guidewires made catheter tracking techniques require image subtraction or positive con-
of resin microparticle compound covered by polytetrafluoroethylene trast to improve visualization of markers on the catheter, which means
have been used for MR-guided interventions in animals.87,88 More recent that, along with faster scan techniques, faster image processing algorithms
wire developments include the use of fiberglass MR safe guidewires, are required.81,96–99
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 557

FIG. 48.3  Manipulation of carbon dioxide-filled balloon catheter (arrows) from the inferior vena cava to the
right pulmonary artery using solely magnetic resonance guidance. Real-time interactive images: repetition
time 2.9 ms, echo time 1.45 ms, flip angle 45 degrees, matrix 128 × 128, field of view 250 to 350, and
temporal resolution 10 to 14 frames per second. Arrows show the signal void of the catheter tip as it tra-
verses the inferior vena cava, right atrium, tricuspid valve, and right ventricular outflow tract and enters the
pulmonary artery.

Active Catheter Tracking and Visualization coil or an antenna that functions in either receive-only mode or transmit/
The active catheter tracking and visualization method uses an electrical receive mode.
connection to the CMR scanner, and localization or tracking of the Active catheters used as receivers have a coil or an antenna that
device requires the device itself, along with any additional hardware or receives signal from tissue in its immediate vicinity.100 These devices
software that comes with it. Typically, the device is equipped with a do not transmit signal into the patient but rely on the body coil to
558 SECTION VI Interventional

Another approach to device localization is what some authors


refer to as semiactive catheter tracking, implying passive localiza-
tion of an electrically isolated resonant coil.77 These resonant coils
locally enhance B1 and signal reception so that, for very low global
flip angles, the signal from the fiducial is prominent.119–124 The reso-
nant coils can be interrogated by gradient echo sequences, such as
SSFP with low flip angles. Catheters with multiple resonant coils can
be tracked easily compared with passive catheters and have a relatively
better safety profile compared with some of the active catheter designs
(Fig. 48.6).
Catheter visualization and localization using 19F CMR in conjunc-
tion with proton imaging appears to be a promising alternative to existing
methods that either are associated with safety concerns if active markers
are used or have insufficient direction-dependent contrast if passive
visualization is used (Fig. 48.7).125 Other multispectral CMR methods
tried before include catheter tracking and angiography using hyperpo-
FIG. 48.4  Active catheter designed for intramyocardial injection. (Cour- larized gases.126,127 Catheter-tracking techniques using inductively coupled
tesy Dr. Parag Karmarkar, Johns Hopkins University, Baltimore, MD.) RF coils or hyperpolarized 13C and visualization strategies using novel
k-space sampling also hold promise.128–130

SAFETY ISSUES
transmit into the patient. The signal received by these coils can then
be used to pinpoint their position, for imaging of local tissue, or both. Bioeffects of Magnetic Fields
There are two important types of active catheters: those based on small The patient undergoing a CMR scan typically is exposed to three forms
coils positioned, for example, at the end of a catheter, and those based of electromagnetic radiation: static magnetic field, gradient magnetic
on a loopless antenna that can run along a catheter or can be made field, and RF electromagnetic field. These can cause bioeffects at signifi-
into a guidewire (Fig. 48.4).101–105 In addition, active designs in which cantly high exposure levels. A health care worker in such a setting can
signal voids along the catheter are created by electrically controlled also be exposed to electromagnetic fields, although exposure is more
magnetic field inhomogeneities have also been investigated.106 chronic and intermittent. However, numerous studies have shown no
A small resonant coil at the tip of a catheter can be identified by a substantial risks to patients from the electromagnetic fields used in clini-
series of three one-dimensional (1D) projections along each axis.100 cal CMR scanners.131–134 The risks to the health care worker, especially
This can be done quickly (in three repetition times) and so could be in a CMR setting, are fiercely debated, but the consensus is that more
repeated for very fast update of the catheter position, allowing real-time work needs to be carried out before occupational electromagnetic field
tracking of the catheter. The position of the catheter could then be exposure limits can be set.135,136 Furthermore, the bioeffects specifi-
projected over a previously acquired road map. Similar techniques have cally related to the use of interventional CMR have not yet been fully
been combined with fast/real-time sequences, imaging the heart or investigated.
vessels using surface coils, and the combined (interleaved) sequence Many reports in the literature regarding the bioeffects of static
has allowed simultaneous localization of the catheter and imaging of magnetic fields are conflicting. There is no strong evidence to suggest
the surrounding tissue. Further adaptation of these sequences has allowed that there are any significant cardiac or neurologic effects from static
automatic changing of the imaging plane to match the change in the magnetic fields of less than 2 T. In addition, several studies have shown
position of the catheter. Another development of active catheter track- that high static magnetic fields do not significantly alter skin and body
ing by the group at the National Institutes of Health allows the visu- temperature.137–142
alization of two simultaneously acquired planes as well as visualization Gradient magnetic fields can induce electrical fields and current
of the catheter or device positions in real time, thus reducing the major in conductive media, including biologic tissue, according to Faraday
problem of the catheter moving through the plane when only one law of induction. The thermal effects of switched magnetic fields are
imaging plane is visualized.107–110 considered negligible and are not believed to be clinically significant.
The great advantage of these active systems is that location of the Electrical stimulation of the retina is believed to cause magnetophos-
catheter is unambiguous. Active visualization has great potential because phenes, which are completely reversible, with no known residual side
it allows the whole length of the catheter or guidewire to be visualized effects. Some volunteers have also reported experiencing a metallic taste
and the imaging plane to be adapted to the moving catheter automati- and vertigo while undergoing imaging within ultrahigh field magnets.
cally. It may even allow high-resolution imaging of a small area of These bioeffects caused by gradient fields are unusual in fields of less
interest, such as a plaque in the vessel, when the coil or antenna is used than 2 T.143
in its imaging mode.111 However, the main disadvantage is concerned The exposure limits for RF radiation are set in terms of specific
with safety.86,112–116 These devices use intravascular coils as RF antennas, absorption rate in Wkg-1, which is the mass normalized rate at which
and the connection to the external circuits via a long wire in the strong RF power is coupled with biologic tissue. The main bioeffects associated
magnetic field makes induction of an electrical current and heating with exposure to RF radiation relate to the generation of heat in tissues.
possible. There have been developments to overcome this risk, such as Controversially, some researchers have reported that electromagnetic
electrical decoupling of loopless antennas and the use of optical coupling fields cause cancer and developmental abnormalities in animal models.
and long fiber optic connections.117 An innovative active catheter design However, the efficiency and absorption pattern of RF radiation is mainly
that uses miniaturized transformers showed no significant RF heating determined by the physical dimensions of the tissue in relation to the
and holds promise for a safe transmission line for interventional appli- incident wavelength, which implies that laboratory animal experiments
cations (Fig. 48.5).118 cannot be simply scaled or extrapolated to humans.144–146
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 559

6F = 2 mm

Coaxial
Transformer
Transformer
loop
A

Passive
markers

Cross at
tip coil

B
FIG. 48.5  (A) Safe transmission line for active catheter tracking created with integrated miniaturized trans-
formers. (B) To evaluate the transformer concept for active tracking in vivo, a 6 Fr (6F) catheter was built
for catheterization of the arterial and venous system of a swine. The catheter is seen being manipulated in
the heart by the active tracking method. (Courtesy Dr. Steffen Weiss, Philips Research, Hamburg, Germany.)
560 SECTION VI Interventional

A B

C
FIG. 48.6  (A) A 5 Fr balloon angiographic catheter with six prewound fiducial markers mounted onto the
surface was manipulated in a 20 mm polyethylene tube taped to the chest of a volunteer. A real-time spoiled
gradient echo sequence (fast field echo [FFE]: repetition time 2.3 ms, echo time 1.2 ms, flip angle 50 degrees,
slice thickness 20 mm) followed by an interactive FFE sequence with interleaving of scans with flip angles
of 2 degrees and 50 degrees and a frame rate of 4 frames per second was used. All six markers are visual-
ized along the length of the catheter. (B) Distal end (arrowhead) of a 6 Fr catheter with an integrated self-
resonant radiofrequency circuit (arrow). (C) The active wireless catheter is shown being guided with real-time
projection reconstruction steady-state free procession imaging into the celiac trunk. (A, Courtesy Arrow
International, Reading, PA. B and C, Courtesy Dr. Harald H. Quick, University of Essen, Germany.)
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 561

FIG. 48.7  Corresponding 1H (top left) and 19F (top right) images of a 7 Fr catheter containing perfluorooc-
tylbromide. With a simple peak search algorithm in the image space, the catheter tip position was extracted
and two orthogonal 19F projections were used to determine the position of the catheter tip (+), as shown.
(From Kozerke S, Hedge S, Schaeffter T, et al. Catheter tracking and visualization using 19F nuclear magnetic
resonance. Magn Reson Med. 2004;52:693–697.)

Heating and Electrical Safety of conductive media and cause current to flow. The circulating currents
Interventional Equipment cause power loss by heating that is referred to as induction heating. A
The heating of wires, devices, implants, and other instruments is an loop in a monitoring cable would increase the inductance of the circuit;
important safety issue that is holding back the rapid advance of inter- therefore larger currents would be induced, resulting in greater heating
ventional CMR. Heating as a result of RF radiation occurs by three of the cable.86,149,150
mechanisms, according to Maxwell’s theory of electromagnetism.112 If a circuit is in a resonant state, then there is maximum current
When a conductive device or instrument is moved through a mag- induction such that significant electromagnetic induction heating occurs.
netic field, small “rings” of current are induced called eddy currents Lengths of wire, for example, can behave as RF antennas that capture
and create internal magnetic fields opposing the change. The kinetic electromagnetic waves to extract power from them. The electromagnetic
energy that goes into driving the eddy currents inside the metal will waves entering the antennas have electrical charges and corresponding
give off that energy as heat. Therefore intravascular guidewires or device currents associated with them. When the antenna is approximately half
delivery systems with a metal core are unsafe in the CMR environment, a wavelength long, resonance occurs and the electrical energy remains
with documented heating up to 165°F of the tip.112,113,147,148 confined to the immediate vicinity of a given antinode. Hence, the highest
Electromagnetic induction heating has often been blamed for thermal electrical field of the antennas is believed to be at the tip. The electrical
injuries caused by monitoring cables used in CMR. RF electromagnetic properties of the media surrounding the antennas and the operating
fields and time-varying gradient magnetic fields can induce voltage in frequency also determine the wavelength.115,116 Newer designs of wires
562 SECTION VI Interventional

and cables aimed at reducing heating are currently being investigated,


BOX 48.1  X-Ray and Cardiovascular
along with novel RF shielding technologies.151,152
Magnetic Resonance Facility: Safety Features
Magnetic Force and Torque • Compulsory safety training of all CMR interventional staff
In addition to the bioeffects of CMR and heating and electrical safety • Specially designed clothes without pockets
of interventional devices, a significant risk to interventional procedures • Safety officer restricting entry to the main room during x-ray and CMR
is magnetic force and torque exerted by the magnetic field on metallic intervention
devices.153,154 • Clear demarcation of ferromagnetic safe and unsafe areas within the room
Conventional guidewires made of ferromagnetic materials, such as • CMR-compatible anesthetic and monitoring equipment
stainless steel, and catheters with metallic braiding, are inherently unsafe • Noise-proof headphone systems for all staff within the room
for use in the CMR environment. Interventional devices that are fer- • X-ray-shielded and radiofrequency-shielded scrub room
romagnetic will be subject to both deflection force (translational move- • Positive pressure air handling and filtration system
ment) and torque (rotational movement); therefore they cannot be • Tethering of all ferromagnetic equipment to the wall or floor
used for procedures within a CMR scanner. Hence, all CMR imaging • Safety checks whenever a patient is transferred between x-ray and CMR
facilities must have safeguards to ensure that ferromagnetic objects are to ensure that metallic instruments used for catheterization are not taken
not brought into the vicinity of the magnet. across to the CMR end of the room
However, there are certain other metallic alloys, such as nitinol, that • Written log of all safety infringements and regular review of safety
are CMR compatible. They produce minimal susceptibility artifacts procedures
and are not affected by the magnetic field in terms of deflection force
and torque. This is an important consideration in developing suitable CMR, Cardiovascular magnetic resonance.
catheters and guidewires for use in interventional CMR procedures.
It should be remembered however that any conducting wire includ-
ing those made from nonferromagnetic materials such as nitinol can lines are designed with extra length and are secured to the movable
still be susceptible to heating and still be unsuitable for use in CMR tabletop to ensure smooth patient transfer.
procedures. The electrocardiogram (ECG) and invasive pressure data are sent
from the MR-compatible monitoring equipment via an optical network
X-RAY AND CARDIOVASCULAR MAGNETIC to a computer in the control room, where the cardiac technician is
RESONANCE GUIDANCE stationed. The appropriate measurement and recording of the data is
made in the usual way. The technician has access to monitors that show
X-Ray and Cardiovascular Magnetic Resonance the appropriate x-ray or CMR images of the procedure. The person
Facility Design carrying out the procedure in the room can view the CMR images and
The room design of a typical XMR facility is shown in Fig. 48.1. There any monitoring data (e.g., ECG, invasive pressure data) with the addi-
are many design features that make this room different from standard tion of commercially available MR conditional LCD monitors or pro-
CMR facilities. The design and clinical practice framework for our jectors available for use in the magnetic resonance imaging (MRI) room
XMR facility is outlined in a paper by White et al.25 for display.
The XMR suite is designed so that half of the room is outside the Blood samples taken during the procedure are labeled in the room
5-gauss line of the magnet, permitting the use of traditional instruments and passed to the technician in the control room via a wave guide.
and devices as well as echocardiography and RF ablation equipment Reliable and accurate ECG synchronization is essential for CMR and
when required. A movable tabletop allows patients to be moved easily in particular CMR-guided cardiac catheterization. When catheters are
between modalities in less than 60 seconds. The paramount consider- manipulated in the heart, there is the potential to cause arrhythmias
ation in the design, construction, and operation of an XMR facility is (tachyarrhythmia or heart block). It is therefore important to perform
safety, and a comprehensive safety protocol must be drawn up to mini- accurate monitoring of the cardiac rhythm at all times during XMR
mize possible hazards (Box 48.1). catheterization. Obtaining a reliable ECG in the magnet, particularly
Traditionally, CMR scans are planned and conducted from the control during some CMR sequences, can be difficult. The magnetohydrodynamic
room, away from the magnet and the patient. However, during CMR- effect and gradient noise can seriously disturb the ECG signal.156,157
guided cardiac catheterization, there is a need for real-time changes to Vector electrocardiogram (VCG) is a QRS detection algorithm that
the scanning plane and sequence parameters to follow catheter manipu- automatically adjusts to the actual electrical axis of the patient’s heart
lation in the heart and great vessels. Also, the person carrying out the and the specific multidimensional QRS waveform. In our experience,
procedure needs to have a clear view of the CMR images while perform- this greatly improves the reliability of R-wave detection to nearly 100%.
ing the cardiac catheter. Therefore it is useful to have a fully functional A reliable R-wave, with the P-waves and T-waves that are also always
set of ceiling-mounted, movable screens and scanner controls within clearly seen with VCG, allows detection of nearly all arrhythmias. There
the CMR scanner room that can be placed at either end of the bore of are now MR-conditional hemodynamic monitoring systems available
the scanner, in close proximity to the patient. Some units also have the (such as Invivo, GE, Medrad) that are sufficient for basic monitoring,
facility for image overlay with semiautomated whole heart segmentation but there are still not commercial hemodynamic monitoring systems
tools155 and these are now commercially available (examples include available specifically for use during MR guided cardiac catheterization.
Interactive Front End, Siemens; RTHawk, HeartVista; Cleartrace, MRI Another complication of performing cardiac catheterization under
Interventions; iSuite, Philips). CMR guidance is the noise generated during scanning. There is a head-
The XMR suite includes appropriate CMR-compatible anesthetic phone and microphone system in the room reducing the noise, allowing
equipment and monitoring equipment for invasive pressure monitoring staff to communicate with each other in both the scanner and control
via the catheter. A great deal of thought has been given to the safety of rooms. There are techniques using infrared technology to allow full
patients under anesthesia, especially during the transfer between the wireless coverage in the scanning and control rooms to allow use of
x-ray and CMR tables. All of the anesthetic and monitoring tubing and multiple headsets (such as Optoacoustics, Clear-Com, Gaven).
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 563

Some CMR coils have x-ray–visible components and would need


to be removed between CMR imaging and x-ray imaging of patients.
It is therefore necessary to have specifically designed coils sufficiently
radiotranslucent to be left in place during XRF without deterioration
of image quality. We use these types of coils in our procedures so that
patients do not have to be disturbed when moving from one imaging
modality to the other.158
The XMR suite has positive-pressure air handling and filtration
appropriate for a catheterization laboratory. There is a scrub room that
is also RF and x-ray shielded and can be accessed both from the XMR
suite and control room. This room acts as an RF lock, allowing access
to the XMR suite during CMR scanning.

Performing X-Ray and Cardiovascular Magnetic


Resonance Interventions A
In a typical XMR interventional procedure, after the induction of anes-
thesia, the patient is transferred from an MR-compatible trolley to the
CMR end of the XMR facility and positioned on the CMR scanner
tabletop (Fig. 48.8A). The monitoring and anesthetic equipment are
attached. A three-lead ECG, separate from the VCG, is used for cardiac
monitoring during MR scanning. The VCG electrodes are placed on
the subcostal margin, outside the x-ray field of view, and the VCG is
used for triggering CMR scans. An MR-compatible pulse oximeter and
noninvasive blood pressure monitoring equipment are also attached.
The exhaled anesthetic gases are monitored for end-tidal carbon dioxide
as well as the concentration of the volatile anesthetic agents. Flexible
phase array RF coils are used. These coils are relatively x-ray lucent and
thus do not need to be removed between MR and x-ray imaging.
The patient is then placed in the CMR scanner, and a multibreath-
hold three-dimensional (3D) SSFP scan of the heart and great vessels
(echo time 2, repetition time 4, flip angle 50 degrees, 80 to 120 slices
B
reconstructed to 1 mm cubic voxels) is obtained.8,159 Using an interactive
SSFP sequence (8 to 10 frames per second), with real-time manipulation
of scan parameters, the likely imaging planes needed for subsequent
catheter tracking, ventricular function, and flow quantification are stored.
The patient is then transferred to the x-ray end of the room. Draping and
vascular access are carried out as for routine cardiac catheterization; in
addition, a second large drape is placed over the patient (see Fig. 48.8B).
The patient is transferred back to the MR scanner after safety checks
are performed, including an operating theater-style check of all metallic
objects used under x-ray. The second drape is then lifted up and taped
to the top of the magnet, which in effect provides sterile draping of the
bore and sides of the magnet (see Fig. 48.8C). An end-hole or side-hole
balloon angiographic catheter (4 to 7 Fr) is placed in the sheath, and
with the balloon inflated with CO2 (see Fig. 48.3), the catheter tip is
passively visualized using the interactive sequences described earlier.
The previously stored imaging planes are used, along with interactive C
slice selection, to track the catheter. Because only the tip of the catheter
is visualized, care is taken not to push the catheter too fast and thus FIG. 48.8  X-ray and magnetic resonance intervention. (A) Patient is
placed on the magnetic resonance tabletop. (B) Patient is slid across to
beyond the CMR imaging plane. This also ensures that the catheter
the x-ray half of the room for sheath insertion. (C) Passive catheter
does not accidentally form loops and possible knots.
manipulation is performed under magnetic resonance guidance.
A duplicate CMR control console is positioned next to the bore
of the magnet so that the interactive window can be easily visualized
while the catheter is being manipulated. Therefore this procedure requires as for routine cardiac catheterization. In addition, ventricular function
two experienced operators, one to move the catheter and one to alter (short-axis balanced SSFP) and flow (phase contrast) scans can be per-
the CMR imaging planes to ensure that the catheter tip is tracked, formed using the appropriate previously stored imaging planes. If catheter
using the real-time interactive sequence. Alternatively simple maneuvers manipulation into a particular heart chamber or vessel using CMR
such as moving the imaging plane or toggling between different views, guidance alone is difficult, the patient is transferred back to the x-ray
as well as starting or stopping scanning can be done using pedals that end of the room, where catheterization can be continued under XRF
are connected to the scanner host computer. (e.g., to use a guidewire or a braided catheter). The patient can be
Once the catheter is positioned in the desired vessel or chamber, transferred back to the CMR scanner for further CMR measurements
appropriate pressure data and saturation/blood gas samples are obtained, once the catheter is positioned satisfactorily.
564 SECTION VI Interventional

Early Experience in Humans techniques include balloon angioplasty of arterial stenoses,162–167 stenting
In our institution we had the first clinical experience of CMR and of vessels,121,168–171 and atrial septal puncture/septostomy.172,173 Device
combined CMR and x-ray (XMR) guided cardiac catheterizations,5,67,93 closure of atrial septal defects is another application that has been
which allowed for a significant reduction of overall x-ray dose. CMR/ explored.174–177 CMR-guided percutaneous pulmonary and aortic valve
XMR catheterizations were initially employed and validated against stent implantation have also been performed successfully (Fig. 48.11).168,178
standard cardiac catheterization for the assessment of pulmonary vas- In addition, more complex interventions, such as percutaneous coronary
cular resistance (PVR).5,67 We used CMR to assess pulmonary vascular catheterization and intervention, have been demonstrated in healthy
resistance in the patients because it allowed for simultaneous measure- animals using CMR179–182 but limitations in spatial resolution are unlikely
ment of pulmonary arterial flow and invasive pressures. We found to result in coronary interventions being a key area for CMR guided
moderate-to-good agreement between the Fick method and the CMR interventions.
method of deriving PVR at baseline conditions. However, in the pres- Further animal studies employing XFM also offer potential interven-
ence of nitric oxide, which is used to assess pulmonary vasoreactivity, tions in congenital heart disease such as percutaneous VSD closure183
there was less agreement between the two methods. There was not only and successful transcatheter creation of bidirectional Glenn shunts in
worsening in agreement but also a large bias when PVR was measured pigs.184 The application of interventions has now been extended to
in the presence of 100% oxygen and nitric oxide. We believe this is the humans.90,167 This includes balloon dilation of aortic coarctation and
result of errors in the Fick method rather than the XMR method, which pulmonary valvuloplasty in patients under CMR guidance alone. In
has important implications for patient management. This novel MR our experience, the youngest patient where this has been achieved was
technique proved to be a more accurate method to quantify PVR in 3.5 years.8,90
humans; it also offers reduced exposure to ionizing radiation.6,67 Novel catheters and guidewires have made possible targeted intra-
In the past few years the indications have widened to include assess- myocardial injection of progenitor stem cells in myocardial infarction
ment of anatomy and function, cardiac output, and hemodynamic in animal models.39,109,185,186 Using real-time CMR and direct apical access
measurements during pharmacological stress.8,68,70,72,160 We have also in porcine hearts, prosthetic aortic valves were implanted in the beating
described an initial clinical experience of CMR guided structural cardiac heart.187 This breakthrough application may allow CMR guidance of
interventions using a CMR compatible guide wire.90 CMR catheteriza- minimally invasive extraanatomic bypass and beating-heart valve repair.
tion has been employed successfully into routine clinical practice at MR guidance of intramyocardial gene therapy is another exciting field.188
several centers with experience of over 100 cases.7 At our own unit, we The ability of CMR to detect myocardial fibrosis and scar tissue could
have performed over 214 MRI catheterizations in the first 10 years of also open up the utility for targeted myocardial biopsy, with researchers
the program8 in a range of patient weights from 2.3 kg to 108 kg, with working on developing appropriate biotomes.3 This may well improve
a good safety profile. This includes the CMR-guided interventions in the poor diagnostic yield of myocardial biopsy in the evaluation of
humans, as described later. The majority of the assessments were for cardiomyopathy.189
PVR evaluation. We found that PVR assessments in this way were a
safe and accurate tool, which allowed for risk stratification of patients Electrophysiology and Radiofrequency Ablation
with congenital heart disease being considered for intervention.68 We Electrophysiology studies and radiofrequency ablation have long been
also identified discrete PVR thresholds below which good long-term done under XRF and ultrasound guidance because of the lack of suitable
outcomes could be achieved following surgical repair or intervention, CMR compatible hardware. However, CMR offers significant advantages
with a successful biventricular repair at resting PVR values ≤6 WU/m2 over XRF with rapid 3D segmentation of the heart and myocardial tissue
and Fontan completion at ≤4 WU/m2. Additionally, we identified that characterization, which allows for visualization of the arrhythmogenic
a baseline Qp:Qs ≤2.75 in biventricular circulations with left-to-right substrate as a target for radiofrequency ablation of arrhythmias and
shunts predicted a PVR ≥6 WU/m2 with 100% sensitivity and 48% assessment ablation induced cardiac lesions. Initial experience in the
specificity as a possible noninvasive surrogate. application of CMR required part of the procedure to be performed
Pharmacological stress studies with dobutamine were employed to under x-ray fluoroscopy with CMR imaging performed at the beginning
increase the heart rate and simulate physiological heart rate responses of the procedure for planning purposes, used in guiding the procedure
to exercise to assess the circulation at rest and under stress. These with XFM, and performed again at the end of the procedure for evalu-
studies involved measurements of cardiac output and invasive pres- ation. XRF guidance is conventionally used to guide such procedures
sures at baseline and were repeated with dobutamine infused at a rate because it offers excellent temporal resolution and good visualization
of 10 µg/kg per minute for 10 minutes or once a stable heart rate or of catheters. However, as a projection imaging modality, more than one
blood pressure rise had been observed and repeated at 20 µg/kg per view is necessary to gain an appreciation of the 3D location and path
minute. Thus far, these have been employed in the assessment of patients of catheters. This implies moving the x-ray c-arm to obtain different
preliver transplant8,73 and in patients with a functionally single ven- projections. A few centers use a biplane x-ray system for the same
tricle.72,74 Our experience is that titration in this manner has a very good purpose. The anatomic context of the acquired images can be difficult
safety profile. to interpret because soft tissues, such as the heart and blood vessels, are
Isoprenaline stress studies have been used to assess for latent coarc- not visible during x-ray exposure. Therefore we developed a real-time
tation.76,161 This involved measurement of aortic blood flow and pressure XMR guidance system for cardiovascular interventions that allows the
gradients across the site of aortic coarctation at baseline and with iso- use of both CMR and x-ray imaging for guidance, thereby overcoming
prenaline (isoprenaline sulphate) at a dose of 0.02 µg/kg per minute some of the failings of exclusive XRF guidance.190
increasing to a maximum of 0.7 µg/kg per minute. The dose was titrated During intervention, the guidance system can provide a real-time
upwards until the heart rate increased by ≥50% from baseline and MR anatomy overlay onto x-ray images. One monitor is used to display
maintained once a stress steady state was achieved. the control interface and the second monitor shows the image overlay.
During fluoroscopy, the system acquires x-ray images and computes the
Interventional Cardiac Applications registration matrix from the tracking data at 10 frames per second and
Animal models have shown immense potential for interventional CMR. updates the overlay display at 3 frames per second. Using this unique
The interventions shown to be feasible with passive and active catheter XMR technology, we have carried out RF ablation in pulmonary veins,
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 565

atria, and ventricles to treat arrhythmias successfully in 30 patients (Fig. These have led to successful electrophysiological studies and RF ablation
48.9).190 This CMR to x-ray registration method also allows us to relate of right atrial flutter in humans.198,199 Early clinical studies have proved
the position of measured electrophysiology data to cardiac motion data safe, with reliable navigation and mapping for RF ablation. RF ablation
from 3D CMR images. The XMR technology is also being used to perform of the cavo-tricuspid isthmus was performed under active MR guidance,
stent implantation in patients with coarctation of the aorta (Figs. 48.10 with brief cine sequences for catheter position confirmation (Fig. 48.13).
and 48.11). Three-dimensional electromechanical models of the heart This is currently limited to adults and, to date, there are no reports of
have been created that allow simulation of cardiovascular pathologies this approach in children. It is anticipated that further development of
to test therapeutic strategies and plan interventions (Fig. 48.12).191,192 this technique would benefit adults and children with congenital heart
However, there are tools for real-time visualization and active track- disease with complex intracardiac anatomy and potential arrhythmic
ing of cardiac catheters for diagnostic and ablation procedures.193–197 substrate from multiple scar sites.

A B C
FIG. 48.9  (A, B) Biplane x-ray views of the linear ablating catheter in the left atrial roof position.
(C) Posterior three-dimensional view of the left atrium derived from a gadolinium cardiovascular magnetic
resonance angiography scan. The green dots show the mapped locations of the linear ablating catheter in
three positions: (1) left atrial roof position; (2) left upper pulmonary vein to mitral valve annulus position; and
(3) right upper pulmonary vein to mitral valve annulus position.

Prestent Poststent
A B
FIG. 48.10  Cardiovascular magnetic resonance angiography (MRA) image superimposed onto the x-ray
cardiac catheter image during stent implantation. (A) Undilated stent and guidewire across the coarctation
site. (B) The combined images show that the implanted open stent lies in a satisfactory position, distal to
the origin of the left subclavian artery and across the coarctation narrowing. Stent implantation was performed
in the x-ray half of the x-ray and magnetic resonance facility. Magnetic resonance imaging was used before
stent insertion to acquire the three-dimensional cardiovascular MRA images and after the procedure (guide-
wires removed) to confirm satisfactory position of the stent and relief of aortic obstruction.
566 SECTION VI Interventional

A B
FIG. 48.11  Percutaneous aortic valve stent implantation in a swine before (A) and after (B) valve stent
implantation under cardiovascular magnetic resonance. White arrow indicates guidewire, and black arrows
note the position of the aortic valve annulus and subsequent stent. (Courtesy Dr. Titus Kuehne, German
Heart Institute, Berlin, Germany.)

image registration and overlay techniques also allow for immediate use
of 3D datasets for procedural guidance in the CMR scanner. Improve-
ments in the accessibility of these imaging platforms will increase their
application in the clinical arena. Meanwhile, there still needs to be
improved spatial and temporal resolution of CMR, particularly to guide
pediatric interventions.
There is a pressing need for industry participation in the develop-
ment of CMR compatible cardiac catheters and devices specifically
designed for CMR-guided cardiac catheterization. This is particularly
relevant in congenital heart disease, where complex anatomy requires
wires and end-hole catheters with good steerabilty and torque to negoti-
ate the bends of the relevant cardiac and vascular structures. Develop-
ment of such equipment needs to keep pace with the rigorous processes
of regulatory approval involved in bringing devices and sequences from
a prototype stage to clinical applications.
The cost associated with installing expensive XMR suites does also
limit the widespread application of interventional CMR but costs will
eventually come down. Over time, there will need to be some verifica-
tion in terms of the cost effectiveness of these techniques and its role
FIG. 48.12  Patient undergoing x-ray and cardiovascular magnetic reso- in improving patient outcomes. However, CMR-guided catheterization
nance (CMR)-guided biventricular pacing. Composite image showing in children will continue to develop as a consequence of the continued
one slice of a CMR cardiac anatomic scan with a superimposed surface strive for better anatomical and physiological data and avoidance of
model of the left ventricle. Cardiac electrical modeling was used to
radiation.
estimate myocardial conductivity for the left ventricle. The conductivity
is represented by the color coding, with blue showing areas of low
conductivity and yellow showing areas of normal conductivity. The white CONCLUSION
region shows the area of scarring segmented from late enhancement
magnetic resonance imaging. There is good correspondence with pre- CMR-guided catheterization is clearly feasible and safe in pediatric
dicted low conductivity and the region of the scar. and adult practice for diagnostic purposes with promising early data
on interventions, limited by the lack of available hardware. The poten-
tial benefits of 3D anatomic guidance for interventional cardiologists,
radiologists, and surgeons, including the useful additional physiologic
Future Directions information and the ability to assess tissue response to therapy with CMR,
CMR-guided catheterization is clearly feasible and safe in pediatric and makes this remarkable imaging modality unique and one that offers
adult practice for diagnostic purposes, particularly in the accurate assess- great promise for safe guidance of complex cardiovascular interventions.
ment of PVR. The early experience in CMR guided interventions is
also promising, with major advances being made in electrophysiological
studies and RF ablation in adults. The main benefits include reduction
ACKNOWLEDGMENTS
of x-ray dose and better visualization of complex anatomy for both Some of the work described in this chapter was performed by a team
diagnostic and interventional cardiac catheterization. Developments in of academic and clinical staff at the Evelina London Children’s Hospital,
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 567

SVC

Tricuspid
valve
annulus
Body of
right
atrium

Coronary
sinus
Late

[ms]

IVC
Early

B C
Tricuspid valve
Ao Body of right atrium annulus
PA

Coronary
RA sinus
RV

Ao
IVC
3.5

RA Signal
intensity/
3.4
StdDev
RV from blood
3.3 pool mean

FIG. 48.13  (A) Screenshot of the iSuite interface. The left side demonstrates a magnetic resonance gener-
ated mesh of the right atrium (RA). Local activation time is overlaid on the map, in a manner analogous to
electroanatomical mapping systems such as Carto. The green catheter, in the coronary sinus, is used for
pacing. A line of block is seen at the site of the red dots (ablation lesions), with superior to inferior activation
of the lateral wall of the RA, confirming block. The dotted arrow indicates direction of electrical activation.
On the right side are three orthogonal multiplanar reconstructions of the balanced steady-state free preces-
sion whole heart that is used as a map for navigation. The multiplanar reconstruction planes are automatically
recalculated based upon the catheter position. (B) T2-weighted imaging immediately postablation. The white
arrow indicates regions of increased enhancement at the cavotricuspid isthmus, at the site of the projected
ablation points (red dots). (C) Magnetic resonance imaging performed three months post ablation. A three-
dimensional late gadolinium enhanced image has been reconstructed on a mesh of the RA, in the same
patient as A and B. The enhancement is seen at the sites of prior ablation. Ao, Aorta; IVC, inferior vena cava;
PA, pulmonary artery; RV, right ventricle; SVC, superior vena cava. (Courtesy Dr. Henry Chubb, King’s College
London.)

London, United Kingdom and the Division of Imaging Sciences and Sciences, Pediatric and Adult Cardiology. We would also like to acknowl-
Biomedical Engineering at King’s College London. The authors thank edge members of the Anesthetic Department; and staff from the Radiol-
Sanjeet Hegde for his work on the previous edition of this chapter. In ogy Department who have provided considerable support.
addition, we wish to acknowledge Tobias Schaeffter, Kawal Rhode, Aph-
rodite Tzifa, Stephen Keevil, Marc O’Neill, Henry Chubb, Israel Valverde,
James Wong, Sujeev Mathur, Aaron Bell, Jas Gill, Shakeel Qureshi, Eric
REFERENCES
Rosenthal, Edward Baker and Derek Hill in the Departments of Imaging A full reference list is available online at ExpertConsult.com
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 567.e1

REFERENCES 28. Elgort DR, Duerk JL. A review of technical advances in interventional
magnetic resonance imaging. Acad Radiol. 2005;12:1089–1099.
1. Jolesz FA, Blumenfeld SM. Interventional use of magnetic resonance 29. Bücker A, Neuerburg JM, Adam GB, et al. Real-time MR guidance for
imaging. Magn Reson Q. 1994;10:85–96. inferior vena cava filter placement in an animal model. J Vasc Interv
2. Lufkin RB. Interventional MR imaging. Radiology. 1995;197:16–18. Radiol. 2001;12:753–756.
3. Rogers T, Lederman RJ. Interventional CMR: clinical applications and 30. Pruessmann KP, Weiger M, Scheidegger MB, Boesiger P. SENSE:
future directions. Curr Cardiol Rep. 2015;17:31–39. sensitivity encoding for fast MRI. Magn Reson Med. 1999;42:952–962.
4. Rogers T, Ratnayaka K, Lederman RJ. MRI catheterization in 31. Pruessmann KP, Weiger M, Boesiger P. Sensitivity encoded cardiac MRI.
cardiopulmonary disease. Chest. 2014;145:30–36. J Cardiovasc Magn Reson. 2001;3:1–9.
5. Razavi R, Hill DLG, Keevil SF, et al. Cardiac catheterisation guided by 32. McVeigh ER, Guttman MA, Kellman P, Raval AN, Lederman RJ.
MRI in children and adults with congenital heart disease. Lancet. Real-time, Interactive MRI for Cardiovascular Interventions. Acad
2003;362:1877–1882. Radiol. 2005;12:1121–1127.
6. Kuehne T, Yilmaz S, Schulze-Neick I, et al. Magnetic resonance imaging 33. Yutzy SR, Duerk JL. Pulse sequences and system interfaces for
guided catheterisation for assessment of pulmonary vascular resistance: interventional and real-time MRI. J Magn Reson Imaging.
in vivo validation and clinical application in patients with pulmonary 2008;27:267–275.
hypertension. Heart. 2005;91:1064–1069. 34. Uecker M, Zhang S, Voit D, Karaus A, Merboldt K-D, Frahm J. Real-time
7. Ratnayaka K, Faranesh AZ, Hansen MS, et al. Real-time MRI-guided MRI at a resolution of 20 ms. NMR Biomed. 2010;23:986–994.
right heart catheterization in adults using passive catheters. Eur Heart J. 35. Fahrig R, Butts K, Rowlands JA, et al. A truly hybrid interventional
2013;34:380–389. MR/X-ray system: feasibility demonstration. J Magn Reson Imaging.
8. Pushparajah K, Tzifa A, Razavi R. Cardiac MRI catheterization: a 10-year 2001;13:294–300.
single institution experience and review. Interv Cardiol. 2014;6: 36. Fahrig R, Butts K, Wen Z, et al. Truly hybrid interventional MR/X-ray
335–346. system. Acad Radiol. 2001;8:1200–1207.
9. Mansfield P, Maudsley AA. Line scan proton spin imaging in biological 37. Adam G, Neuerburg J, Bücker A, et al. Interventional magnetic
structures by NMR. Phys Med Biol. 1976;21:847–852. resonance. Initial clinical experience with a 1.5-tesla magnetic resonance
10. Mansfield P, Maudsley AA. Medical imaging by NMR. Br J Radiol. system combined with c-arm fluoroscopy. Invest Radiol.
1977;50:188–194. 1997;32:191–197.
11. Damadian R. Tumor detection by nuclear magnetic resonance. Science. 38. Rhode KS, Hill DLG, Edwards PJ, et al. Registration and tracking to
1971;171:1151–1153. integrate X-ray and MR images in an XMR facility. IEEE Trans Med
12. Pykett IL, Newhouse JH, Buonanno FS, et al. Principles of nuclear Imaging. 2003;22:1369–1378.
magnetic resonance imaging. Radiology. 1982;143:157–168. 39. de Silva R, Gutiérrez LF, Raval AN, McVeigh ER, Ozturk C, Lederman
13. Henderson RG. Nuclear magnetic resonance imaging: a review. J R Soc RJ. X-ray fused with magnetic resonance imaging (XFM) to target
Med. 1983;76:206–212. endomyocardial injections: validation in a swine model of myocardial
14. Riederer SJ, Tasciyan T, Farzaneh F, Lee JN, Wright RC, Herfkens RJ. MR infarction. Circulation. 2006;114:2342–2350.
fluoroscopy: technical feasibility. Magn Reson Med. 1988;8:1–15. 40. Dori Y, Sarmiento M, Glatz AC, et al. X-ray magnetic resonance fusion
15. Farzaneh F, Riederer SJ, Lee JN, Tasciyan T, Wright RC, Spritzer CE. MR to internal markers and utility in congenital heart disease
fluoroscopy: initial clinical studies. Radiology. 1989;171:545–549. catheterization. Circ Cardiovasc Imaging. 2011;4:415–424.
16. Wright RC, Riederer SJ, Farzaneh F, Rossman PJ, Liu Y. Real-time MR 41. Faranesh AZ, Kellman P, Ratnayaka K, Lederman RJ. Integration of
fluoroscopic data acquisition and image reconstruction. Magn Reson cardiac and respiratory motion into MRI roadmaps fused with x-ray.
Med. 1989;12:407–415. Med Phys. 2013;40:032302.
17. Holsinger AE, Wright RC, Riederer SJ, Farzaneh F, Grimm RC, Maier JK. 42. Abu Hazeem AA, Dori Y, Whitehead KK, et al. X-ray magnetic
Real-time interactive magnetic resonance imaging. Magn Reson Med. resonance fusion modality may reduce radiation exposure and contrast
1990;14:547–553. dose in diagnostic cardiac catheterization of congenital heart disease.
18. Riederer SJ, Wright RC, Ehman RL, et al. Real-time interactive color Catheter Cardiovasc Interv. 2014;84:795–800.
flow MR imaging. Radiology. 1991;181:33–39. 43. Le Heron J. Guidelines on patient dose to promote the optimisation of
19. Cox RW, Jesmanowicz A, Hyde JS. Real-time functional magnetic protection for diagnostic medical exposures: documents of the NRPB v
resonance imaging. Magn Reson Med. 1995;33:230–236. 10(1), 1999. Radiation protection news and notes. 1999;:17–18.
20. Debbins JP, Riederer SJ, Rossman PJ, et al. Cardiac magnetic resonance 44. Little MP, De Vathaire F, Charles MW, Hawkins MM, Muirhead CR.
fluoroscopy. Magn Reson Med. 1996;36:588–595. Variations with time and age in the risks of solid cancer incidence after
21. Gmitro AF, Ehsani AR, Berchem TA, Snell RJ. A real-time reconstruction radiation exposure in childhood. Stat Med. 1998;17:1341–1355.
system for magnetic resonance imaging. Magn Reson Med. 45. Modan B, Keinan L, Blumstein T, Sadetzki S. Cancer following cardiac
1996;35:734–740. catheterization in childhood. Int J Epidemiol. 2000;29:424–428.
22. Kerr AB, Pauly JM, Hu BS, et al. Real-time interactive MRI on a 46. American Academy of Pediatrics, Committee on Environmental Health.
conventional scanner. Magn Reson Med. 1997;38:355–367. Risk of ionizing radiation exposure to children: a subject review.
23. Schenck JF, Jolesz FA, Roemer PB, et al. Superconducting open- Pediatrics. 1998;101:717–719.
configuration MR imaging system for image-guided therapy. Radiology. 47. Johnson JN, Hornik CP, Li JS, et al. Cumulative radiation exposure and
1995;195:805–814. cancer risk estimation in children with heart disease. Circulation.
24. Silverman SG, Collick BD, Figueira MR, et al. guided biopsy in an 2014;130:161–167.
open-configuration MR imaging system. Radiology. 1995;197:175–181. 48. Bashore TM, Bates ER, Berger PB, et al. Task Force on Clinical Expert
25. White MJ, Thornton JS, Hawkes DJ, et al. Design, operation, and safety Consensus Documents. American College of Cardiology/Society for
of single-room interventional MRI suites: practical experience from two Cardiac Angiography and Interventions Clinical Expert Consensus
centers. J Magn Reson Imaging. 2015;41:34–43. Document on cardiac catheterization laboratory standards. A report of
26. Hoult DI, Saunders JK, Sutherland GR, et al. The engineering of an the American College of Cardiology Task Force on Clinical Expert
interventional MRI with a movable 1.5 Tesla magnet. J Magn Reson Consensus Documents; 2001:2170–2214.
Imaging. 2001;13:78–86. 49. McCullough PA, Wolyn R, Rocher LL, Levin RN, O’Neill WW. Acute
27. Guttman MA, Ozturk C, Raval AN, et al. Interventional cardiovascular renal failure after coronary intervention: incidence, risk factors, and
procedures guided by real-time MR imaging: an interactive interface relationship to mortality. Am J Med. 1997;103:368–375.
using multiple slices, adaptive projection modes and live 3D renderings. 50. Venneri L, Rossi F, Botto N, et al. Cancer risk from professional
J Magn Reson Imaging. 2007;26:1429–1435. exposure in staff working in cardiac catheterization laboratory: insights
567.e2 SECTION VI Interventional

from the National Research Council’s Biological Effects of Ionizing 70. Kuehne T. Magnetic resonance imaging analysis of right ventricular
Radiation VII Report. Am Heart J. 2009;157:118–124. pressure-volume loops: in vivo validation and clinical application in
51. Chambers CE, Fetterly KA, Holzer R, et al. Radiation safety program for patients with pulmonary hypertension. Circulation. 2004;110:
the cardiac catheterization laboratory. Catheter Cardiovasc Interv. 2010–2016.
2011;77:546–556. 71. Schmitt B, Steendijk P, Lunze K, et al. Integrated assessment of diastolic
52. Cigarroa RG, Lange RA, Hillis LD. Oximetric quantitation of and systolic ventricular function using diagnostic cardiac magnetic
intracardiac left-to-right shunting: limitations of the Qp/Qs ratio. Am J resonance catheterization: validation in pigs and application in a clinical
Cardiol. 1989;64:246–247. pilot study. JACC Cardiovasc Imaging. 2009;2:1271–1281.
53. Dhingra VK. Lack of agreement between thermodilution and 72. Schmitt B, Steendijk P, Ovroutski S, et al. Pulmonary vascular resistance,
Fick cardiac output in critically ill patients. Chest. 2002;122: collateral flow, and ventricular function in patients with a Fontan
990–997. circulation at rest and during dobutamine stress. Circ Cardiovasc
54. Hillis LD, Firth BG, Winniford MD. Variability of right-sided cardiac Imaging. 2010;3:623–631.
oxygen saturations in adults with and without left-to-right intracardiac 73. Razavi RS, Baker A, Qureshi SA, et al. Hemodynamic response to
shunting. Am J Cardiol. 1986;58:129–132. continuous infusion of dobutamine in Alagille’s syndrome.
55. van den Berg E, Pacifico A, Lange RA, Wheelan KR, Winniford MD, Transplantation. 2001;72:823.
Hillis LD. Measurement of cardiac output without right heart 74. Pushparajah K, Wong JK, Bellsham-Revell HR, et al. Magnetic resonance
catheterization: reliability, advantages, and limitations of a left-sided imaging catheter stress haemodynamics post-Fontan in hypoplastic left
indicator dilution technique. Cathet Cardiovasc Diagn. 1986;12: heart syndrome. Eur Heart J Cardiovasc Imaging. 2016;17:644–651.
205–208. 75. Wong J, Pushparajah K, de Vecchi A, et al. Pressure–volume loop-derived
56. Hillis LD, Firth BG, Winniford MD. Analysis of factors affecting the cardiac indices during dobutamine stress: a step towards understanding
variability of Fick versus indicator dilution measurements of cardiac limitations in cardiac output in children with hypoplastic left heart
output. Am J Cardiol. 1985;56:764–768. syndrome. Int J Cardiol. 2017;230:439–446.
57. Dehmer GJ, Firth BG, Hillis LD. Oxygen consumption in adult patients 76. Valverde I, Staicu C, Grotenhuis H, et al. Predicting hemodynamics in
during cardiac catheterization. Clin Cardiol. 1982;5:436–440. native and residual coarctation: preliminary results of a Rigid-Wall
58. Beerbaum P, Korperich H, Barth P, Esdorn H, Gieseke J, Meyer H. Computational-Fluid-Dynamics model (RW-CFD) validated against
Noninvasive quantification of left-to-right shunt in pediatric patients: clinically invasive pressure measures at rest and during pharmacological
phase-contrast cine magnetic resonance imaging compared with invasive stress. J Cardiovasc Magn Reson. 2011;13:P49.
oximetry. Circulation. 2001;103:2476–2482. 77. Duerk J. A brief review of hardware for catheter tracking in magnetic
59. Beerbaum P, Korperich H, Gieseke J, Barth P, Peuster M, Meyer H. resonance imaging. MAGMA. 2002;13:199–208.
Rapid left-to-right shunt quantification in children by phase-contrast 78. Rubin DL, Ratner AV, Young SW. Magnetic susceptibility effects and
magnetic resonance imaging combined with sensitivity encoding their application in the development of new ferromagnetic catheters for
(SENSE). Circulation. 2003;108:1355–1361. magnetic resonance imaging. Invest Radiol. 1990;25:1325–1332.
60. Beerbaum P, Korperich H, Gieseke J, Barth P, Peuster M, Meyer H. 79. Bakker CJ, Hoogeveen RM, Weber J, van Vaals JJ, Viergever MA, Mali
Blood flow quantification in adults by phase-contrast MRI combined WP. Visualization of dedicated catheters using fast scanning techniques
with SENSE—a validation study. J Cardiovasc Magn Reson. with potential for MR-guided vascular interventions. Magn Reson Med.
2005;7:361–369. 1996;36:816–820.
61. Firmin DN, Nayler GL, Klipstein RH, Underwood SR, Rees RSO, 80. Bakker CJ, Smits HF, Bos C, et al. MR-guided balloon angioplasty: in
Longmore DB. In vivo validation of MR velocity imaging. J Comput vitro demonstration of the potential of MRI for guiding, monitoring,
Assist Tomogr. 1987;11:751–756. and evaluating endovascular interventions. J Magn Reson Imaging.
62. Kilner PJ, Manzara CC, Mohiaddin RH, et al. Magnetic resonance jet 1998;8:245–250.
velocity mapping in mitral and aortic valve stenosis. Circulation. 81. Bakker CJG, Bos C, Weinmann HJ. Passive tracking of catheters and
1993;87:1239–1248. guidewires by contrast-enhanced MR fluoroscopy. Magn Reson Med.
63. Hundley WG, Li HF, Hillis LD, et al. Quantitation of cardiac output 2001;45:17–23.
with velocity-encoded, phase-difference magnetic resonance imaging. 82. Bakker CJ, Hoogeveen RM, Hurtak WF, Van Vaals JJ, Viergever MA, Mali
Am J Cardiol. 1995;75:1250–1255. WP. MR-guided endovascular interventions: susceptibility-based catheter
64. Hundley WG, Li HF, Lange RA, et al. Assessment of left-to-right and near-real-time imaging technique. Radiology. 1997;202:273–276.
intracardiac shunting by velocity-encoded, phase-difference magnetic 83. Lardo AC. Real-time magnetic resonance imaging: diagnostic and
resonance imaging a comparison with oximetric and indicator dilution interventional applications. Pediatr Cardiol. 2000;21:80–98.
techniques. Circulation. 1995;91:2955–2960. 84. Omary RA, Unal O, Koscielski DS, et al. Real-time MR imaging-guided
65. Mousseaux E, Tasu JP, Jolivet O, Simonneau G, Bittoun J, Gaux J-C. passive catheter tracking with use of gadolinium-filled catheters. J Vasc
Pulmonary arterial resistance: noninvasive measurement with indexes of Interv Radiol. 2000;11:1079–1085.
pulmonary flow estimated at velocity-encoded MR imaging— 85. Unal O, Li J, Cheng W, Yu H, Strother CM. MR-visible coatings for
preliminary experience. Radiology. 1999;212:896–902. endovascular device visualization. J Magn Reson Imaging.
66. Kondo C, Caputo GR, Masui T, et al. Pulmonary hypertension: 2006;23:763–769.
pulmonary flow quantification and flow profile analysis with velocity- 86. Yeung CJ, Susil RC, Atalar E. RF safety of wires in interventional MRI:
encoded cine MR imaging. Radiology. 1992;183:751–758. using a safety index. Magn Reson Med. 2002;47:187–193.
67. Muthurangu V, Taylor A, Andriantsimiavona R, et al. Novel method of 87. Mekle R, Hofmann E, Scheffler K, Bilecen D. A polymer-based
quantifying pulmonary vascular resistance by use of simultaneous MR-compatible guidewire: a study to explore new prospects for
invasive pressure monitoring and phase-contrast magnetic resonance interventional peripheral magnetic resonance angiography (ipMRA).
flow. Circulation. 2004;110:826–834. J Magn Reson Imaging. 2006;23:145–155.
68. Pushparajah K, Tzifa A, Bell A, et al. Cardiovascular magnetic resonance 88. Buecker A, Spuentrup E, Schmitz-Rode T, et al. Use of a nonmetallic
catheterization derived pulmonary vascular resistance and medium-term guide wire for magnetic resonance-guided coronary artery
outcomes in congenital heart disease. J Cardiovasc Magn Reson. catheterization. Invest Radiol. 2004;39:656–660.
2015;17:28. 89. Krueger S, Schmitz S, Weiss S, et al. An MR guidewire based on
69. Muthurangu V, Atkinson D, Sermesant M, et al. Measurement of total micropultruded fiber-reinforced material. Magn Reson Med.
pulmonary arterial compliance using invasive pressure monitoring and 2008;60:1190–1196.
MR flow quantification during MR-guided cardiac catheterization. Am J 90. Tzifa A, Krombach GA, Krämer N, et al. Magnetic resonance-guided
Physiol Heart Circ Physiol. 2005;289:H1301–H1306. cardiac interventions using magnetic resonance-compatible devices: a
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 567.e3

preclinical study and first-in-man congenital interventions. Circ 113. Liu C-Y, Farahani K, Lu DSK, Duckwiler G, Oppelt A. Safety of
Cardiovasc Interv. 2010;3:585–592. MRI-guided endovascular guidewire applications. J Magn Reson
91. Sonmez M, Saikus CE, Bell JA, et al. MRI active guidewire with an Imaging. 2000;12:75–78.
embedded temperature probe and providing a distinct tip signal to 114. Yeung CJ, Atalar E. RF Transmit power limit for the barewire loopless
enhance clinical safety. J Cardiovasc Magn Reson. 2012;14:38. catheter antenna. J Magn Reson Imaging. 2000;12:86–91.
92. Basar B, Rogers T, Ratnayaka K, et al. Segmented nitinol guidewires with 115. Yeung CJ, Atalar E. A Green’s function approach to local RF heating in
stiffness-matched connectors for cardiovascular magnetic resonance interventional MRI. Med Phys. 2001;28:826.
catheterization: preserved mechanical performance and freedom from 116. Yeung CJ, Susil RC, Atalar E. RF heating due to conductive wires during
heating. J Cardiovasc Magn Reson. 2015;17:105. MRI depends on the phase distribution of the transmit field. Magn
93. Miquel ME, Hegde S, Muthurangu V, et al. Visualization and tracking Reson Med. 2002;48:1096–1098.
of an inflatable balloon catheter using SSFP in a flow phantom and in 117. Konings MK, Bartels LW, van Swol CFP, Bakker CJG. Development of an
the heart and great vessels of patients. Magn Reson Med. 2004;51: MR-safe tracking catheter with a laser-driven tip coil. J Magn Reson
988–995. Imaging. 2001;13:131–135.
94. Zimmermann-Paul GG, Ladd ME, Pfammatter T, Hilfiker PR, Quick 118. Weiss S, Vernickel P, Schaeffter T, Schulz V, Gleich B. Transmission line
HH, Debatin JF. MR versus fluoroscopic guidance of a catheter/ for improved RF safety of interventional devices. Magn Reson Med.
guidewire system: in vitro comparison of steerability. J Magn Reson 2005;54:182–189.
Imaging. 1998;8:1177–1181. 119. Hegde S, Miquel ME, Boubertakh R, et al. Interactive MR imaging and
95. Martin AJ, Weber OM, Saeed M, Roberts TPL. Steady-state imaging for tracking of catheters with multiple tuned fiducial markers. J Vasc Interv
visualization of endovascular interventions. Magn Reson Med. Radiol. 2006;17:1175–1179.
2003;50:434–438. 120. Kuehne T, Fahrig R, Butts K. Pair of resonant fiducial markers for
96. Bakker CJG, Seppenwoolde J-H, Bartels LW, van der Weide R. Adaptive localization of endovascular catheters at all catheter orientations. J Magn
subtraction as an aid in MR-guided placement of catheters and Reson Imaging. 2003;17:620–624.
guidewires. J Magn Reson Imaging. 2004;20:470–474. 121. Kuehne T, Weiss S, Brinkert F, et al. Catheter visualization with resonant
97. Smits HFM, Bos C, van der Weide R, Bakker CJG. Interventional MR markers at MR imaging-guided deployment of endovascular stents in
vascular applications. Eur Radiol. 1999;9:1488–1495. swine. Radiology. 2004;233:774–780.
98. van der Weide R, Bakker CJG, Viergever MA. Localization of 122. Quick HH, Zenge MO, Kuehl H, et al. Interventional magnetic
intravascular devices with paramagnetic markers in MR images. IEEE resonance angiography with no strings attached: wireless active catheter
Trans Med Imaging. 2001;20:1061–1071. visualization. Magn Reson Med. 2005;53:446–455.
99. Duerk JL, Butts K, Hwang K-P, Lewin JS. Pulse sequences for 123. Wong EY, Zhang Q, Duerk JL, Lewin JS, Wendt M. An optical system for
interventional magnetic resonance imaging. Top Magn Reson Imaging. wireless detuning of parallel resonant circuits. J Magn Reson Imaging.
2000;11:147–162. 2000;12:632–638.
100. Dumoulin CL, Souza SP, Darrow RD. Real-time position monitoring of 124. Weiss S, Kuehne T, Brinkert F, et al. In vivo safe catheter visualization
invasive devices using magnetic resonance. Magn Reson Med. and slice tracking using an optically detunable resonant marker. Magn
1993;29:411–415. Reson Med. 2004;52:860–868.
101. Ladd ME, Zimmermann GG, McKinnon GC, et al. Visualization of 125. Kozerke S, Hegde S, Schaeffter T, Lamerichs R, Razavi R, Hill DL.
vascular guidewires using MR tracking. J Magn Reson Imaging. Catheter tracking and visualization using 19F nuclear magnetic
1998;8:251–253. resonance. Magn Reson Med. 2004;52:693–697.
102. Ladd ME, Zimmermann GG, Quick HH, et al. Active MR visualization 126. Månsson S, Johansson E, Magnusson P, et al. 13C imaging—a new
of a vascular guidewire in vivo. J Magn Reson Imaging. 1998;8:220–225. diagnostic platform. Eur Radiol. 2006;16:57–67.
103. Ladd ME, Quick HH. Reduction of resonant RF heating in intravascular 127. Svensson J, Månsson S, Johansson E, Petersson JS, Olsson LE.
catheters using coaxial chokes. Magn Reson Med. 2000;43:615–619. Hyperpolarized 13C MR angiography using trueFISP. Magn Reson Med.
104. Zhang Q, Wendt M, Aschoff AJ, Zheng L, Lewin JS, Duerk JL. Active MR 2003;50:256–262.
guidance of interventional devices with target-navigation. Magn Reson 128. Celik H, Ulutürk A, Tali T, Atalar E. A catheter tracking method using
Med. 2000;44:56–65. reverse polarization for MR-guided interventions. Magn Reson Med.
105. Hillenbrand CM, Elgort DR, Wong EY, et al. Active device tracking and 2007;58:1224–1231.
high-resolution intravascular MRI using a novel catheter-based, 129. Magnusson P, Johansson E, Månsson S, et al. Passive catheter tracking
opposed-solenoid phased array coil. Magn Reson Med. 2004;51: during interventional MRI using hyperpolarized 13C. Magn Reson Med.
668–675. 2007;57:1140–1147.
106. Glowinski A, Adam G, Bücker A, Neuerburg J, van Vaals JJ, Günther RW. 130. Peng H, Draper JN, Frayne R. Rapid passive MR catheter visualization
Catheter visualization using locally induced, actively controlled field for endovascular therapy using nonsymmetric truncated k-space
inhomogeneities. Magn Reson Med. 1997;38:253–258. sampling strategies. Magn Reson Imaging. 2008;26:293–303.
107. Guttman MA, Lederman RJ, Sorger JM, McVeigh ER. Real-time volume 131. McRobbie D, Foster MA. Thresholds for biological effects of time-
rendered MRI for interventional guidance. J Cardiovasc Magn Reson. varying magnetic fields. Clin Phys Physiol Meas. 1984;5:67–78.
2002;4:431–442. 132. McRobbie D, Foster MA. Cardiac response to pulsed magnetic fields
108. Lederman RJ. Cardiovascular interventional magnetic resonance with regard to safety in NMR imaging. Phys Med Biol. 1985;30:
imaging. Circulation. 2005;112:3009–3017. 695–702.
109. Lederman RJ, Guttman MA, Peters DC, et al. Catheter-based 133. Budinger TF. MR safety: past, present, and future from a historical
endomyocardial injection with real-time magnetic resonance imaging. perspective. Magn Reson Imaging Clin N Am. 1998;6:701–714.
Circulation. 2002;105:1282–1284. 134. Budinger TF. Emerging nuclear magnetic resonance technologies. Health
110. Bell JA, Saikus CE, Ratnayaka K, et al. A deflectable guiding catheter for and safety. Ann N Y Acad Sci. 1992;649:1–18.
real-time MRI-guided interventions. J Magn Reson Imaging. 135. Hill DLG, McLeish K, Keevil SF. Impact of electromagnetic field
2011;35:908–915. exposure limits in Europe: is the future of interventional MRI safe? Acad
111. Atalar E, Bottomley PA, Ocali O, et al. High resolution intravascular Radiol. 2005;12:1135–1142.
MRI and MRS by using a catheter receiver coil. Magn Reson Med. 136. Keevil SF, Gedroyc W, Gowland P, et al. Electromagnetic field exposure
1996;36:596–605. limitation and the future of MRI. Br J Radiol. 2005;78:973.
112. Konings MK, Bartels LW, Smits HF, Bakker CJ. Heating around 137. Tenforde TS. Biological interactions and potential health effects of
intravascular guidewires by resonating RF waves. J Magn Reson Imaging. extremely-low-frequency magnetic fields from power lines and other
2000;12:79–85. common sources. Annu Rev Public Health. 1992;13:173–196.
567.e4 SECTION VI Interventional

138. Schenck JF, Dumoulin CL, Redington RW, Kressel HY, Elliott RT, validation against invasive pressure catheterization at rest and
McDougall IL. Human exposure to 4.0-Tesla magnetic fields in a pharmacological stress. J Cardiovasc Magn Reson. 2015;17:Q78.
whole-body scanner. Med Phys. 1992;19:1089–1098. 162. Wildermuth S, Dumoulin CL, Pfammatter T, Maier SE, Hofmann E,
139. Schenck JF. MR safety at high magnetic fields. Magn Reson Imaging Clin Debatin JF. MR-guided percutaneous angioplasty: assessment of tracking
N Am. 1998;6:715–730. safety, catheter handling and functionality. Cardiovasc Intervent Radiol.
140. Kelley DA, Schenck JF. Very-high-field magnetic resonance imaging: 1998;21:404–410.
instrumentation and safety issues. Top Magn Reson Imaging. 163. Yang X, Atalar E. Intravascular MR imaging-guided balloon angioplasty
1999;10:79–89. with an MR imaging guide wire: feasibility study in rabbits. Radiology.
141. Schenck JF. Safety of strong, static magnetic fields. J Magn Reson 2000;217:501–506.
Imaging. 2000;12:2–19. 164. Yang X, Bolster BD, Kraitchman DL, Atalar E. Intravascular
142. Schenck JF. Physical interactions of static magnetic fields with living MR-monitored balloon angioplasty: an in vivo feasibility study. J Vasc
tissues. Prog Biophys Mol Biol. 2005;87:185–204. Interv Radiol. 1998;9:953–959.
143. Budinger TF, Fischer H, Hentschel D, Reinfelder HE, Schmitt F. 165. Godart F, Beregi JP, Nicol L, et al. MR-guided balloon angioplasty of
Physiological effects of fast oscillating magnetic field gradients. stenosed aorta: in vivo evaluation using near-standard instruments and
J Comput Assist Tomogr. 1991;15:909–914. a passive tracking technique. J Magn Reson Imaging. 2000;12:639–644.
144. Guidelines on limits of exposure to static magnetic fields. International 166. Omary RA, Frayne R, Unal O, et al. MR-guided angioplasty of renal
Commission on Non-Ionizing Radiation Protection. Health Phys. artery stenosis in a pig model: a feasibility study. J Vasc Interv Radiol.
1994;66:100–106. 2000;11:373–381.
145. Kanal E, Borgstede JP, Barkovich AJ, et al. American College of Radiology 167. Krueger JJ, Ewert P, Yilmaz S, et al. Magnetic resonance imaging-guided
white paper on MR safety. AJR Am J Roentgenol. 2002;178:1335–1347. balloon angioplasty of coarctation of the aorta: a pilot study. Circulation.
146. International Commission on Non-Ionizing Radiation Protection. 2006;113:1093–1100.
Medical magnetic resonance (MR) procedures: protection of patients. 168. Kuehne T, Saeed M, Higgins CB, et al. Endovascular stents in pulmonary
Health Phys. 2004;87:197–216. valve and artery in swine: feasibility study of MR imaging-guided
147. Shellock FG, Shellock VJ. Cardiovascular catheters and accessories: ex deployment and postinterventional assessment. Radiology. 2003;226:
vivo testing of ferromagnetism, heating, and artifacts associated with 475–481.
MRI. J Magn Reson Imaging. 1998;8:1338–1342. 169. Raval AN, Telep JD, Guttman MA, et al. Real-time magnetic resonance
148. Nitz WR, Oppelt A, Renz W, Manke C, Lenhart M, Link J. On the imaging-guided stenting of aortic coarctation with commercially
heating of linear conductive structures as guide wires and catheters in available catheter devices in Swine. Circulation. 2005;112:699–706.
interventional MRI. J Magn Reson Imaging. 2001;13:105–114. 170. Buecker A, Neuerburg JM, Adam GB, et al. Real-time MR fluoroscopy
149. Armenean C, Perrin E, Armenean M, Beuf O, Pilleul F, Saint-Jalmes H. for MR-guided iliac artery stent placement. J Magn Reson Imaging.
RF-induced temperature elevation along metallic wires in clinical 2000;12:616–622.
magnetic resonance imaging: influence of diameter and length. Magn 171. Quick HH, Kuehl H, Kaiser G, Bosk S, Debatin JF, Ladd ME. Inductively
Reson Med. 2004;52:1200–1206. coupled stent antennas in MRI. Magn Reson Med. 2002;48:781–790.
150. Dempsey MF, Condon B, Hadley DM. Investigation of the factors 172. Arepally A, Karmarkar PV, Weiss C, Rodriguez ER, Lederman RJ, Atalar
responsible for burns during MRI. J Magn Reson Imaging. E. Magnetic resonance image-guided trans-septal puncture in a swine
2001;13:627–631. heart. J Magn Reson Imaging. 2005;21:463–467.
151. Gray RW, Bibens WT, Shellock FG. Simple design changes to wires to 173. Raval AN, Karmarkar PV, Guttman MA, et al. Real-time MRI guided
substantially reduce MRI-induced heating at 1.5 T: implications for atrial septal puncture and balloon septostomy in swine. Catheter
implanted leads. Magn Reson Imaging. 2005;23:887–891. Cardiovasc Interv. 2006;67:637–643.
152. Helfer JL, Gray RW, MacDonald SG, Bibens TW. Can pacemakers, 174. Buecker A, Spuentrup E, Grabitz R, et al. Magnetic resonance-guided
neurostimulators, leads, or guide wires be MRI safe? Technological placement of atrial septal closure device in animal model of patent
concerns and possible resolutions. Minim Invasive Ther Allied Technol. foramen ovale. Circulation. 2002;106:511–515.
2006;15:114–120. 175. Rickers C, Jerosch-Herold M, Hu X, et al. Magnetic resonance
153. Luechinger R, Duru F, Scheidegger MB, Boesiger P, Candinas R. Force image-guided transcatheter closure of atrial septal defects. Circulation.
and torque effects of a 1.5-Tesla MRI scanner on cardiac pacemakers 2003;107:132–138.
and ICDs. Pacing Clin Electrophysiol. 2001;24:199–205. 176. Schalla S, Saeed M, Higgins CB, Martin A, Weber O, Moore P. Magnetic
154. Buecker A, Spuentrup E, Grabitz R, et al. Real-time-MR guidance for resonance–guided cardiac catheterization in a swine model of atrial
placement of a self-made fully MR-compatible atrial septal occluder: in septal defect. Circulation. 2003;108:1865–1870.
vitro test. Rofo. 2002;174:283–285. 177. Schalla S, Saeed M, Higgins CB, Weber O, Martin A, Moore P. Balloon
155. Peters J, Ecabert O, Meyer C, et al. Automatic whole heart segmentation sizing and transcatheter closure of acute atrial septal defects guided by
in static magnetic resonance image volumes. Med Image Comput magnetic resonance fluoroscopy: assessment and validation in a large
Comput Assist Interv. 2007;10:402–410. animal model. J Magn Reson Imaging. 2005;21:204–211.
156. Dimick RN, Hedlund LW, Herfkens RJ, Fram EK, Utz J. Optimizing 178. Kuehne T, Yilmaz S, Meinus C, et al. Magnetic resonance imaging-
electrocardiograph electrode placement for cardiac-gated magnetic guided transcatheter implantation of a prosthetic valve in aortic valve
resonance imaging. Invest Radiol. 1987;22:17–22. position: feasibility study in swine. J Am Coll Cardiol. 2004;44:
157. Tenforde TS. Magnetically induced electric fields and currents in the 2247–2249.
circulatory system. Prog Biophys Mol Biol. 2005;87:279–288. 179. Spuentrup E, Ruebben A, Schaeffter T, Manning WJ, Günther RW,
158. Rieke V, Ganguly A, Daniel BL, et al. X-ray compatible radiofrequency Buecker A. Magnetic resonance–guided coronary artery stent placement
coil for magnetic resonance imaging. Magn Reson Med. 2005;53: in a swine model. Circulation. 2002;105:874–879.
1409–1414. 180. Zhang S, Rafie S, Chen Y, et al. In vivo cardiovascular catheterization
159. Razavi RS, Hill DLG, Muthurangu V, et al. Three-dimensional magnetic under real-time MRI guidance. J Magn Reson Imaging. 2006;24:
resonance imaging of congenital cardiac anomalies. Cardiol Young. 914–917.
2003;13:461–465. 181. Serfaty JM, Yang X, Aksit P, et al. Guided coronary catheterization:
160. Tzifa A, Schaeffter T, Razavi R. MR imaging-guided cardiovascular visualization of guiding catheters, guidewires, and anatomy in real time.
interventions in young children. Magn Reson Imaging Clin N Am. J Magn Reson Imaging. 2000;12:590–594.
2012;20:117–128. 182. Serfaty J-M, Yang X, Foo TK, Kumar A, Derbyshire A, Atalar E.
161. Sotelo JA, Valverde I, Beerbaum PB, et al. Pressure gradient prediction in MRI-guided coronary catheterization and PTCA: a feasibility study on a
aortic coarctation using a computational-fluid-dynamics model: dog model. Magn Reson Med. 2003;49:258–263.
CHAPTER 48  Pediatric Interventional Cardiovascular Magnetic Resonance 567.e5

183. Ratnayaka K, Raman VK, Faranesh AZ, et al. Antegrade percutaneous 192. Sermesant M, Rhode K, Sanchez-Ortiz GI, et al. Simulation of cardiac
closure of membranous ventricular septal defect using X-ray fused with pathologies using an electromechanical biventricular model and XMR
magnetic resonance imaging. JACC Cardiovasc Interv. 2009;2:224–230. interventional imaging. Med Image Anal. 2005;9:467–480.
184. Ratnayaka K, Rogers T, Schenke WH, et al. Magnetic resonance 193. Sommer P, Grothoff M, Eitel C, et al. Feasibility of real-time magnetic
imaging–guided transcatheter cavopulmonary shunt. JACC Cardiovasc resonance imaging-guided electrophysiology studies in humans.
Interv. 2016;9:959–970. Europace. 2012;15:101–108.
185. Dick AJ, Guttman MA, Raman VK, et al. Magnetic resonance 194. Grothoff M, Piorkowski C, Gaspar T, et al. Comprehensive MRI-guided
fluoroscopy allows targeted delivery of mesenchymal stem cells to infarct intracardiac electrophysiological interventions in swine using a
borders in swine. Circulation. 2003;108:2899–2904. combination of active tracking and passive real-time catheter imaging. J
186. Dick AJ, Lederman RJ. MRI-guided myocardial cell therapy. Int J Cardiovasc Magn Reson. 2014;16:1.
Cardiovasc Intervent. 2005;7:165–170. 195. Piorkowski C, Grothoff M, Gaspar T, et al. Cavotricuspid isthmus
187. McVeigh ER, Guttman MA, Lederman RJ, et al. Real-time interactive ablation guided by real-time magnetic resonance imaging. Circ
MRI-guided cardiac surgery: aortic valve replacement using a direct Arrhythm Electrophysiol. 2013;6:e7–e10.
apical approach. Magn Reson Med. 2006;56:958–964. 196. Weiss S, Wirtz D, David B, et al. In vivo evaluation and proof of
188. Yang X, Atalar E. MRI-guided gene therapy. FEBS Lett. radiofrequency safety of a novel diagnostic MR-electrophysiology
2006;580:2958–2961. catheter. Magn Reson Med. 2011;65:770–777.
189. Bennett MK, Gilotra NA, Harrington C, et al. Evaluation of the role of 197. Bhagirath P, van der Graaf M, Karim R, et al. Interventional cardiac
endomyocardial biopsy in 851 patients with unexplained heart failure magnetic resonance imaging in electrophysiology: advances toward
from 2000-2009. Circ Heart Fail. 2013;6:676–684. clinical translation. Circ Arrhythm Electrophysiol. 2015;8:203–211.
190. Rhode KS, Sermesant M, Brogan D, et al. A system for real-time XMR 198. Chubb H, Harrison J, Williams SE, et al. First in man: real-time
guided cardiovascular intervention. IEEE Trans Med Imaging. magnetic resonance-guided ablation of typical right atrial flutter using
2005;24:1428–1440. active catheter tracking. Europace. 2014;16:iii25.
191. Sermesant M, Coudière Y, Moreau-Villèger V, Rhode KS, Hill DLG, 199. Hilbert S, Sommer P, Gutberlet M, et al. Real-time magnetic resonance-
Razavi RS. A fast-marching approach to cardiac electrophysiology guided ablation of typical right atrial flutter using a combination of
simulation for XMR interventional imaging. Med Image Comput Comput active catheter tracking and passive catheter visualization in man: initial
Assist Interv. 2005;8:607–615. results from a consecutive patient series. Europace. 2015.
SECTION VII  Economics and Guidelines

49 
Cost-Effectiveness Analysis for
Cardiovascular Magnetic Resonance Imaging
Afshin Farzaneh-Far, Juerg Schwitter, and Raymond Y. Kwong

Explosive growth in medical imaging technology during the past few


BASIC TERMINOLOGY
decades has provided physicians with an unparalleled ability to diagnose
abnormalities of the cardiovascular system. However, in some cases, A detailed review of the technical methods of cost-effectiveness analysis
scientific enthusiasm and economic forces have helped diffuse new is beyond the scope of this chapter, and readers are referred elsewhere.3–6
technologies widely without careful assessment of their costs and benefits However, cardiovascular imaging physicians need to have a basic under-
to patient care. This growth in available imaging technologies has con- standing of the terminology used in this field. Cost-effectiveness analysis
tributed to the continuous increase in health care spending seen since tries to provide a framework to compare different management strate-
the 1970s. Despite a steady decline in deaths from coronary artery gies or treatments within the constraint of limited resources.4 It aims to
disease (CAD) in the United States, there has been an increase in health maximize health for a given budget. Although often used interchange-
care expenditures attributable to heart disease, with an estimated cost ably, the terms “cost-minimization,” “cost-benefit,” “cost-effectiveness,”
of $316 billion dollars in 2010.1 An analysis of Medicare claims between and “cost-utility” have distinct definitions (Fig. 49.2).
1999 and 2008 revealed that 78% of this growth in cardiovascular ser-
vices was attributed to noninvasive testing—primarily nuclear stress Cost-Minimization Analysis
imaging and echocardiography.2 When clinical equivalence is demonstrated through rigorous trial
Given these rising costs, there is increasing pressure worldwide from data then a cost-minimization analysis can be a useful economic tool.
government agencies, insurance companies and other stakeholders to The rationale being that if two therapies (or management strategies)
demonstrate proof of value for all medical expenditures. are equally effective, the cheaper one is favorable from an economic
Cost-effectiveness analyses compare the costs and outcomes for a perspective. In reality it is often very difficult to determine clinical
new intervention with an existing alternative treatment, strategy, or equivalence, so caution must be used when interpreting results from
intervention. The questions such analyses aim to answer are: how much such analyses.
does the new intervention cost compared with current practice and is
it more effective; and if so, how much more? Cost-effectiveness analyses Cost-Benefit Analysis
aim to provide the same information commonly used for making deci- In a cost-benefit analysis, both the cost and the benefit, which in this
sions about purchasing decisions in everyday life (Fig. 49.1). If a new case is improved health, are measured in monetary terms. This requires
strategy or potential purchase is more effective and less costly than the placing a dollar value on grades of health and duration of life. Although
currently available option, it is almost certainly worth doing, and in this methodology is appropriate in the analysis of many economic
general such a strategy is called “dominant.” Likewise, if the new strategy systems, placing a direct monetary value on human health and life
is less effective and more costly, no one is likely to use it. However, the extension can be problematic.
more usual outcome of a cost-effectiveness analysis of a health technol-
ogy is that the new technology may be more effective, but also more Cost-Effectiveness Analysis
costly. A judgment then needs to be made as to whether the benefit Due to the difficulty of assigning a dollar value on human health, many
obtained is worth the cost, and how certain we can be about that assess- health economists prefer cost effectiveness, which describes the relation-
ment. Cost-effectiveness analyses aim to provide a framework for making ship between cost and a measure of health relevant to the intervention
such decisions. being analyzed—such as life-years gained, disease free survival in a
It is clear that cost-effectiveness analysis is critical for the future of cancer treatment study, or reductions in blood pressure in a trial of an
cardiac imaging, given rising costs and increasing scrutiny from third- antihypertensive. The central metric of cost effectiveness is the cost-
party payers. In this chapter we will provide a brief overview of the effectiveness ratio with cost in the numerator (in dollars, euros, etc.)
techniques of cost-effectiveness analysis, as applied to cardiovascular and effectiveness in the denominator.7,8 In the context of a cost-
magnetic resonance (CMR) and review the current literature as it per- effectiveness analysis, the cost of a particular therapy is the sum of all
tains to CMR in both US and European contexts. resources consumed. This may include the direct cost of care (i.e.,

568
CHAPTER 49  Cost-Effectiveness Analysis for Cardiovascular Magnetic Resonance Imaging 569

New treatment
more costly

New treatment
Existing treatment
more effective
dominates
but more costly

New treatment New treatment


less effective more effective

New treatment
New treatment
less effective
dominates
but less costly

New treatment
less costly
FIG. 49.1  Types of economic evaluations used in health care.

Is effectiveness of interventions equal?

No Yes
Cost-minimization study

Yes
Can all outcomes be valued
Cost-benefit analysis
in monetary terms?

No
No
Cost-effectiveness analysis
Can outcomes be measured
as quality-adjusted life years?
Cost-utility analysis
Yes
FIG. 49.2  Cost-effectiveness plane. Most new treatments or technologies lie in the upper right corner.

hospital, drugs, treatments, diagnostic tests, physician fees), indirect Quality-Adjusted Life Year
costs (i.e., lost productivity from work, travel, day care), as well as In its most basic form, the “effectiveness” of a treatment can be rep-
intangible costs (i.e., pain, suffering). resented as the years of life gained. However, for most interventions
For a particular treatment or test, the cost-effectiveness ratio in improvements in quality of life are more prominent, giving rise to a
isolation is of little value, unless compared with an alternative treatment/ quality-adjusted life year (QALY). The QALY applies a weighting factor to
test (or no treatment/test). Cost-effectiveness analysis may therefore be account for varying degrees of health for each year of life gained or lost.
better characterized as an incremental cost-effectiveness (“comparative By convention, perfect health is assigned a value of 1, and death a value
effectiveness”) ratio as detailed in the formula below: of 0. For all of the health states in between, there is a deduction in QALY.
Incremental cost-effectiveness ratio Cost-Utility Analysis
Costnew strategy − Costcurrent practice A cost-utility analysis is a type of cost-effective analysis that uses QALY
=
Effectnew strategy − Effectcurrent practice to measure treatment effect. It therefore allows for the comparison of
570 SECTION VII  Economics and Guidelines

different treatments across different diseases such as dialysis in end- diagnostic test results, as well as ensuring strict protocol adherence to
stage kidney disease versus transcatheter aortic valve replacement for management strategies with minimization of patient crossover.
aortic stenosis. Cost-utility analyses are important from a public health Although imaging procedures share many of the features of other
policy and societal standpoint. diagnostic tests, there are several issues that may be unique to imaging:
(1) imaging results are often multidimensional (e.g., presence or absence,
Societal Perspective location, size, etc.) rather than one-dimensional (e.g., plasma creatinine
Depending on the specific health care system, there are many different level); (2) clear quantitative cut-off points are often not established (i.e.,
stakeholders: governments, insurance companies, hospitals, physicians, results are presented in terms such as “mild,” “moderate,” or “severe”);
employers, and patients. Each participant has a different perspective (3) images can incidentally reveal other diseases (e.g., an unsuspected
with regard to cost and effect. It is important to recognize that what lung mass); and/or (4) some imaging procedures may be associated with
may be cost-effective from a societal standpoint may not be from the increased risk of harm (e.g., radiation exposure or renal dysfunction).
perspective of an individual hospital or medical practice or institution. Historically, the performance of an imaging test has been evaluated
The societal perspective attempts to look at the aggregate costs and simply in terms of diagnostic accuracy and prognostic utility. However,
effects on all members and is the one most often employed in cost- the downstream decision-making and resource utilization occurring
effectiveness analysis.9 after a diagnostic test are very complex and involve numerous factors
that are difficult, if not impossible, to model or predict. For example,
Time Horizon Shaw et al. showed significant variation in clinical decision-making after
The time horizon is the period of time for which the analysis is con- presentation of the same nuclear perfusion imaging results to clinicians.12
ducted. It has significant impact on both costs and effects, because Finally, it should be noted that disease prevalence in the study popu-
over time the costs of an intervention may change (e.g., because of lation plays an important role in any assessment of cost effectiveness
emergence of other competing techniques, drop in clinical demand, because it affects diagnostic performance through Bayesian principles.
etc.) and effectiveness provided has a limited longevity. Short-term For example, cost-effectiveness analysis of CMR in symptomatic patients
versus long-term costs/effects may be very different and thus can alter with CAD cannot be applied to a broad population of asymptomatic
the cost-effectiveness ratio of a given therapy depending on the time patients without known CAD.
horizon used in the analysis.
COST-EFFECTIVE ANALYSES OF CARDIOVASCULAR
Cost-Effectiveness Thresholds
There are several commonly used thresholds seen in the literature, which
MAGNETIC RESONANCE
are sometimes used to define the boundaries of a cost-effective therapy. Differences in health care systems across the world greatly impact cost-
For example, in the United States, $50K/QALY is a commonly cited effectiveness analyses. To a large extent, the assumptions and conclusions
threshold, as compared with £20K/QALY in the United Kingdom. Such of these studies are limited to the country or region in which they were
thresholds are somewhat arbitrary and do not necessarily reflect a soci- performed. Therefore in the following sections of this chapter we will
ety’s willingness to pay, leading some to suggest alternative methods to attempt to separately review studies performed in the United States
determine what is cost-effective—such as using annual average income.10 and Europe.
These threshold values are therefore highly dependent on regional eco-
nomic and cultural forces. US Perspective
Cost-effectiveness studies of CMR within the US health care system
CHALLENGES IN CONDUCTING have been very limited to date (Table 49.1). This relates to significant
COST-EFFECTIVENESS ANALYSIS IN difficulties in ascertaining costs, compared with many European coun-
tries, which have universal health care systems; as well as the relatively
CARDIOVASCULAR IMAGING limited clinical adaptation of CMR in CAD, compared with other imaging
Cost-effectiveness analyses for diagnostic technologies differ from the modalities in the United States.
evaluation of therapeutic interventions in many respects.11 One of the It is extremely difficult to accurately assess actual costs in the US
most important and challenging differences is the indirect relationship health care system. In many instances, there is complex and opaque
between results of a diagnostic test and health outcomes. Diagnostic accounting—with varying cost-to-charge ratios resulting in differences
tests only provide intermediate results, often a surrogate of a disease between what is charged or billed and actual payments or costs received.
state, which may influence treatment decisions but not directly alter The “charge” is the price the consumer is billed by the health care facility
hard clinical health outcomes. for the service. This charge is highly variable depending on the hospital
Many factors can alter the chain of events between diagnostic test and other local factors, which can be very unclear. The actual payment
performance and ultimate outcomes. For example, the diagnostic test received may be a small fraction of the charge, and varies widely among
may be technically inadequate, or it may be technically adequate, but hospitals, states, and third-party payers. Consequently, investigators
the reader may fail to correctly interpret the images. The correct inter- frequently use cost estimates derived from the United States’ Centers for
pretation may not be accurately transmitted or received by the referring Medicare and Medicaid Services (CMS) average payments. These cost
physician, or the referring physicians may vary in their treatment deci- estimates published by the CMS can serve as an averaged metric for a
sions based on their clinical discretion. In addition, the patient may significant portion of the US population. They can be looked up online
choose not to comply with the recommended treatments. Perhaps most using the Healthcare Common Procedure Coding System (HCPCS)
importantly, ultimate outcomes are usually driven by the effectiveness or Current Procedural Technology (CPT) codes of the procedures
of the treatment(s) used. Therefore consideration needs to be given (http://www.cms.gov/apps/physician-fee-schedule/search/search
not only to the diagnostic imaging study, but also to the cost effectiveness -criteria.aspx).
of any subsequent therapies instituted as a result of imaging. This com- Within the US system, direct CMR costs include the cost of using
plicates study design because investigators must also account for the and maintaining the equipment (technical fee) and the cost of study
efficacy and cost of possible downstream therapies resulting from interpretation (professional fee). The costs of the medications used in
CHAPTER 49  Cost-Effectiveness Analysis for Cardiovascular Magnetic Resonance Imaging 571

TABLE 49.1  Summary of Major Cost-Effectiveness Studies for Cardiovascular Magnetic


Resonance Performed in the United States
Author N Study Population Study Conclusions
13
Miller et al. 110 Patients presenting to ED with chest pain and negative ECG/ Patients randomized to stress CMR had a significantly reduced
biomarkers randomized to stress CMR in an observation unit median hospitalization cost
vs. standard inpatient care
Miller et al.14 109 Patients presenting to ED with chest pain and negative ECG/ Cardiac-related costs at 1 year after discharge were significantly
biomarkers randomized to stress CMR in an observation unit lower for participants randomized to stress CMR than those
vs. standard inpatient care receiving standard inpatient care ($3101 vs. $4742 [P = .004])

CMR, Cardiovascular magnetic resonance; ECG, electrocardiogram; ED, emergency department; US, United States.

stress imaging and any contrast agent also factor into the overall costs. an economic point of view, first-line invasive coronary angiography
There is huge variability in price between different hospitals, counties, is a reasonable alternative to stress CMR for patients with a pretest
and states, as well as the insurance reimbursement. This cost variability likelihood greater than 60%. Similar results were obtained in a study
is a significant barrier to accurate and meaningful cost-effectiveness by Moschetti et al., where diagnostic performance of perfusion CMR,
analysis. These problems are further compounded by the variable uti- as observed in the European CMR registry, was used for comparison
lizations of downstream testing and procedures influenced by some with invasive coronary angiography.16 In this registry population with a
regional or local factors. CAD prevalence of 21%, perfusion CMR yielded the correct diagnosis
Despite these challenges, reports of cost-effectiveness analysis of of CAD, at lower costs than invasive coronary angiography when cal-
CMR in the United States are developing. Miller and colleagues inves- culated for the German health care system—which is consistent with
tigated the cost effectiveness of CMR for patients presenting with acute the earlier findings of Boldt et al.15 In the European CMR registry, costs
chest pain to a US hospital.13 They sought to determine whether stress were also calculated for the UK, Swiss, and US health care systems,
CMR in an observation unit would reduce costs among patients with with similar results demonstrating reduced costs by CMR in patient
emergent non-low-risk chest pain who otherwise would be managed diagnosis and management.16
with an in-patient care strategy. Emergency department patients (N = Moschetti et al. also compared ischemia detection by perfusion CMR
110) at intermediate or high probability for acute coronary syndrome with fractional flow reserve (FFR).17 To provide all information neces-
without electrocardiographic or biomarker evidence of a myocardial sary for a decision on revascularization, both strategies were coupled
infarction were randomized to stress CMR in an observation unit versus with invasive coronary angiography. Specifically, in the CMR+coronary
standard in-patient care. At 30 days, no subjects in either group expe- angiography arm, all patients with ischemia on perfusion CMR were
rienced an acute coronary syndrome. The stress CMR patients had a investigated by additional invasive coronary angiography, whereas in
reduced median hospitalization cost of $588, and 79% were managed the hypothetical coronary angiography+FFR arm, all patients under-
without hospital admission. In a follow-up study, they sought to compare went first-line invasive coronary angiography and patients with >50%
the direct cost of medical care and clinical events during the first year diameter stenosis were further studied by FFR. To calculate the fraction
after discharge.14 They used direct costs of cardiac-related care and of patients with >50% diameter stenosis, the stenosis–FFR relationship
clinical outcomes (myocardial infarction, revascularization, cardiovas- reported in the literature was used.18 Furthermore, it was assumed that
cular death). Cardiac-related costs at 1 year were significantly lower for the outcome would be the same for both strategies, given growing
participants randomized to stress CMR than those receiving conventional evidence demonstrating excellent prognosis in patients with either nega-
inpatient care ($3101 vs. $4742 [P = .004]). There was no significant tive perfusion CMR or negative FFR.19–21 These two strategies, that is
difference in occurrence of major cardiac events between the two groups CMR+coronary angiography and coronary angiography+FFR, were
at 1 year (6% vs. 9%, respectively [P = .72]). applied to a population with CAD prevalence rates ranging from 0%
to 100%. The CMR+coronary angiography strategy was more cost-
European Perspective effective than the coronary angiography+FFR strategy below a CAD
There are more published data regarding cost effectiveness of CMR in prevalence of 62%, 65%, 83%, and 82% for the Swiss, German, UK,
the European context—predominantly from Switzerland, Germany and and US health care systems, respectively. These numbers have impor-
the United Kingdom (Table 49.2). However, it is important to realize that tant implications. If one collects the percentage of patients undergo-
there are significant differences between different European countries ing revascularizations after invasive coronary angiography, the pretest
in terms of health care economics and delivery. Thus studies in one probability of CAD for patients undergoing coronary angiography can
country cannot necessarily be extrapolated to other countries in Europe. be estimated. Accordingly, for the Swiss, German, UK, and US health
Boldt and colleagues used a mathematical model based on Bayes’ care systems, the literature reports a pretest probability of significant
theorem to compare cost effectiveness and utility of stress CMR and CAD of 34% to 45%, 43%, 42%, and ~38%, respectively, all consider-
single-photon emission computed tomography (SPECT) versus inva- ably below the calculated thresholds needed for cost effectiveness of
sive coronary angiography as the standard of reference in a German the angiography+FFR strategy.16,17,20,22–24
population with CAD prevalence rates ranging from 0% to 100%.15 Walker and colleagues compared 8 different diagnostic pathways
From a third-payer perspective, stress CMR was more cost-effective than applying exercise treadmill testing, SPECT, or CMR in various combi-
SPECT for any CAD prevalence and this was true for costs per correct nations using patients from the UK CE-MARC study.23 Patients were
diagnosis of CAD as well as for costs per QALY gained. However, this assigned to different states such as true positive for patients who were
rank order changed in favor of invasive coronary angiography when correctly identified and revascularized, true negative for those without
CAD prevalence was above 60%. The authors concluded that, from significant stenosis, and false negative for patients who were misidentified
572 SECTION VII  Economics and Guidelines

TABLE 49.2  Summary of Major Cost-Effectiveness Studies for Cardiovascular Magnetic


Resonance Performed in Europe
Author Design Country N Study and Methods Study Conclusions
27
Pilz et al. Cost minimization Germany 250 Determination of costs between a strategy CMR perfusion resulted in cost savings
of CMR perfusion vs. coronary for all patients with the exception of
angiography using propensity matching patients at the highest risk of CAD
based on Morise scores. This cost
saving was achieved mainly through
reduction of invasive angiography.
Walker et al.23 Cost effectiveness, UK 752 Cost-effectiveness analysis using a CMR perfusion is more cost-effective
Markov-model decision analytic model to compare (lower cost/QALY) than SPECT
eight strategies for the diagnosis of
CAD based on CE-MARC data
Boldt et al.15 Cost effectiveness Germany — Based on Bayes’ theorem, a mathematical In patients with low to intermediate CAD
model was developed to compare the probabilities, CMR is more cost-
cost effectiveness and utility of CMR vs. effective (both lower cost/diagnosis and
SPECT in patients with suspected CAD cost/QALY) than SPECT. Direct coronary
angiography as first test is most
cost-effective at CAD prevalence >60%.
Thom et al.26 Cost effectiveness UK 898 Comparison of cost effectiveness and Noninvasive cardiac imaging can be used
outcomes of various initial imaging safely as the initial diagnostic test to
strategies in the management of stable diagnose CAD without adverse effects
chest pain in a long-term prospective on patient outcomes or increased costs,
randomized trial relative to angiography. Noninvasive
imaging avoided 20%–25% of invasive
angiography.
Petrov et al.28 Cost minimization Germany 1158 Determination of costs between a strategy Initial stress CMR strategy saved €12,466
of dobutamine CMR vs. coronary of hospital costs per life year
angiography. Propensity matched.
Moschetti et al.20 Cost minimization Germany, 3647 Comparison of the costs of a CMR-guided A CMR + CXA strategy for patients with
Switzerland, strategy vs. two invasive strategies in suspected CAD provides cost reduction
UK, US the EuroCMR registry population compared with a hypothetical CXA +
FFR strategy in patients with low to
intermediate disease prevalence in all
four countries
Pletscher et al.25 Cost effectiveness, Switzerland 752 Cost-effectiveness analysis using a CMR perfusion is more cost-effective
Markov-model decision analytic model to compare (lower cost/QALY) than other strategies
eight strategies for the diagnosis of using SPECT or direct coronary
CAD based on CE-MARC data angiography

CAD, Coronary artery disease; CE-MARC, Clinical Evaluation of MAgnetic Resonance imaging in Coronary heart disease; CMR, cardiovascular
resonance imaging; CXA, coronary angiography; FFR, fractional flow reserve; QALY, quality-adjusted life years; SPECT, single proton emission
computed tomography; UK, United Kingdom; US, United States.

and not revascularized or who died as a result of the mortality risks male with a prior likelihood of significant stenosis of 39.5% based on
associated with CAD. The proportion of patients in each state was CE-MARC data21) was also the most cost-effective strategy. They con-
dependent on the sensitivities and specificities of the various tests in cluded that the strategy of inconclusive stress testing followed by CMR
the diagnostic strategy, and each state was then linked to cardiovascular followed by coronary angiography (if CMR is positive or inconclusive)
events. They found only two strategies as potentially cost-effective for with all positive stress tests followed by coronary angiography (without
the diagnosis of CAD, both of which included CMR. In one strategy CMR testing) was the strategy with highest QALYs gained at lowest
CMR followed a positive or inconclusive stress test, followed by coronary costs.25 Similar to the UK study, the strategy of coronary angiography
angiography if CMR was positive or inconclusive (strategy 3). In the following a positive or inconclusive CMR was the strategy with the
other strategy, coronary angiography followed a positive or inconclusive second highest QALYs gained.25 Thus for both UK and Swiss health care
CMR (strategy 5). In this study, these two strategies appeared cost- systems, a CMR-based diagnostic strategy appears most cost-effective
effective at the lower and higher end of the threshold range used in in comparison with other noninvasive testing algorithms. Boldt et al.
the United Kingdom (i.e., at £20,000 and £30,000 per QALY gained), found similar results in the German system with CMR being superior
for a typical case patient (60-year-old male with a prior likelihood of to SPECT.15 However, another cost-effectiveness study performed in
significant stenosis of 39.5% based on CE-MARC data21). Similar mod- the United Kingdom found that cost effectiveness was similar between
eling was applied to the Swiss health care system by Pletscher et al.25 coronary angiography, CMR, SPECT, and stress echocardiography.26 This
In their study, a CMR-based work-up of a typical patient (60-year-old study has been criticized for being underpowered and having a very
CHAPTER 49  Cost-Effectiveness Analysis for Cardiovascular Magnetic Resonance Imaging 573

low percentage of avoided coronary angiography procedures (ranging and appropriate utilization. These studies will need to include large
between 20% and 25%) when starting with a noninvasive test.26 randomized patient populations comparing CMR-guided treatment
strategies with standard management, as well as registry data reflecting
real-world clinical practice. As the health care environment continues
CONCLUSION to evolve it is clear that fiscal constraints on cardiac imaging will likely
The field of CMR has developed over recent years on the basis of a increase, as the current rate of growth is unsustainable in many parts
large and growing body of published evidence showing excellent diag- of the world. Despite their limitations, cost-effectiveness analyses will
nostic and prognostic performance across a wide range of cardiovascular be pivotal in demonstrating the value of CMR and its impact on patient
conditions. However, given the rising costs of imaging, there is increasing outcomes.
pressure worldwide from government agencies, insurance companies,
and other stakeholders to demonstrate proof of value for all medical
expenditures. Therefore demonstrating the cost effectiveness of CMR
REFERENCES
across a spectrum of disease will be critical to ensure continued growth A full reference list is available online at ExpertConsult.com
CHAPTER 49  Cost-Effectiveness Analysis for Cardiovascular Magnetic Resonance Imaging 573.e1

REFERENCES the diagnostic work-up of coronary artery disease: application of the


European Cardiovascular Magnetic Resonance registry data to the
1. Writing Group Members, Lloyd-Jones D, Adams RJ, et al; American Heart German, United Kingdom, Swiss, and United States health care systems. J
Association Statistics Committee and Stroke Statistics Subcommittee. Cardiovasc Magn Reson. 2012;14:35.
Heart disease and stroke statistics—2010 update: a report from the 17. Moschetti K, Favre D, Pinget C, et al. Comparative cost-effectiveness
American Heart Association. Circulation. 2010;121:e46–e215. analyses of cardiovascular magnetic resonance and coronary angiography
2. Andrus BW, Welch HG. Medicare services provided by cardiologists in combined with fractional flow reserve for the diagnosis of coronary
the United States: 1999-2008. Circ Cardiovasc Qual Outcomes. artery disease. J Cardiovasc Magn Reson. 2014;16:13.
2012;5:31–36. 18. Toth G, Hamilos M, Pyxaras S, et al. Evolving concepts of angiogram:
3. Francis SA, Daly C, Heydari B, Abbasi S, Shah RV, Kwong RY. Cost- fractional flow reserve discordances in 4000 coronary stenoses. Eur Heart
effectiveness analysis for imaging techniques with a focus on J. 2014;35:2831–2838.
cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2013;15:52. 19. De Bruyne B, Pijls NH, Kalesan B, et al; FAME 2 Trial Investigators.
4. Weinstein MC, Stason WB. Foundations of cost-effectiveness analysis for Fractional flow reserve-guided PCI versus medical therapy in stable
health and medical practices. N Engl J Med. 1977;296:716–721. coronary disease. N Engl J Med. 2012;367:991–1001.
5. Torrance GW. Measurement of health state utilities for economic 20. Moschetti K, Petersen SE, Pilz G, et al. Cost-minimization analysis of
appraisal. J Health Econ. 1986;5:1–30. three decision strategies for cardiac revascularization: results of the
6. Diamond GA, Kaul S. Cost, effectiveness, and cost-effectiveness. Circ “suspected CAD” cohort of the European Cardiovascular Magnetic
Cardiovasc Qual Outcomes. 2009;2:49–54. Resonance Registry. J Cardiovasc Magn Reson. 2016;18:3.
7. Weinstein MC, Siegel JE, Gold MR, Kamlet MS, Russell LB. 21. Greenwood JP, Maredia N, Younger JF, et al. Cardiovascular magnetic
Recommendations of the Panel on Cost-Effectiveness in Health and resonance and single-photon emission computed tomography for
Medicine. JAMA. 1996;276:1253–1258. diagnosis of coronary heart disease (CE-MARC): a prospective trial.
8. Siegel JE, Weinstein MC, Russell LB, Gold MR. Recommendations for Lancet. 2012;379:453–460.
reporting cost-effectiveness analyses. Panel on Cost-Effectiveness in 22. Horstkotte D, Wiemer M, van Buuren F. Performance figures of invasive
Health and Medicine. JAMA. 1996;276:1339–1341. cardiology in Germany 2006 and 2007 focussing on coronary artery
9. Russell LB, Gold MR, Siegel JE, Daniels N, Weinstein MC. The role of disease. Clin Res Cardiol. 2011;100:187–190.
cost-effectiveness analysis in health and medicine. Panel on Cost- 23. Walker S, Girardin F, McKenna C, et al. Cost-effectiveness of
Effectiveness in Health and Medicine. JAMA. 1996;276:1172–1177. cardiovascular magnetic resonance in the diagnosis of coronary heart
10. Garber AM, Phelps CE. Economic foundations of cost-effectiveness disease: an economic evaluation using data from the CE-MARC study.
analysis. J Health Econ. 1997;16:1–31. Heart. 2013;99:873–881.
11. Gazelle GS, McMahon PM, Siebert U, Beinfeld MT. Cost-effectiveness 24. Patel MR, Peterson ED, Dai D, et al. Low diagnostic yield of elective
analysis in the assessment of diagnostic imaging technologies. Radiology. coronary angiography. N Engl J Med. 2010;362:886–895.
2005;235:361–370. 25. Pletscher M, Walker S, Moschetti K, et al. Cost-effectiveness of functional
12. Shaw LJ, Hachamovitch R, Heller GV, et al. Noninvasive strategies for the cardiac imaging in the diagnostic work-up of coronary heart disease. Eur
estimation of cardiac risk in stable chest pain patients. The Economics of Heart J Qual Care Clin Outcomes. 2016;2:201–207.
Noninvasive Diagnosis (END) Study Group. Am J Cardiol. 2000;86:1–7. 26. Thom H, West NE, Hughes V, et al; CECaT Study Group. Cost-
13. Miller CD, Hwang W, Hoekstra JW, et al. Stress cardiac magnetic effectiveness of initial stress cardiovascular MR, stress SPECT or stress
resonance imaging with observation unit care reduces cost for patients echocardiography as a gate-keeper test, compared with upfront invasive
with emergent chest pain: a randomized trial. Ann Emerg Med. coronary angiography in the investigation and management of patients
2010;56:209–219.e2. with stable chest pain: mid-term outcomes from the CECaT randomised
14. Miller CD, Hwang W, Case D, et al. Stress CMR imaging observation unit controlled trial. BMJ Open. 2014;4:e003419.
in the emergency department reduces 1-year medical care costs in 27. Pilz G, Patel PA, Fell U, et al. Adenosine-stress cardiac magnetic
patients with acute chest pain: a randomized study for comparison with resonance imaging in suspected coronary artery disease: a net cost
inpatient care. JACC Cardiovasc Imaging. 2011;4:862–870. analysis and reimbursement implications. Int J Cardiovasc Imaging.
15. Boldt J, Leber AW, Bonaventura K, et al. Cost-effectiveness of 2011;27:113–121.
cardiovascular magnetic resonance and single-photon emission computed 28. Petrov G, Kelle S, Fleck E, Wellnhofer E. Incremental cost-effectiveness of
tomography for diagnosis of coronary artery disease in Germany. J dobutamine stress cardiac magnetic resonance imaging in patients at
Cardiovasc Magn Reson. 2013;15:30. intermediate risk for coronary artery disease. Clin Res Cardiol.
16. Moschetti K, Muzzarelli S, Pinget C, et al. Cost evaluation of 2015;104:401–409.
cardiovascular magnetic resonance versus coronary angiography for
50 
Cardiac Positron Emission Tomography/
Magnetic Resonance
Felix Nensa and Thomas Schlosser

For decades, cardiovascular magnetic resonance imaging (CMR) and reducing the performance of the PET and the MR components. Uniform
positron emission tomography (PET) have been clinically established magnetic fields are of utmost importance in MR imaging. Thus any
imaging modalities in cardiovascular medicine. For several years, a new additional electronic circuits, which can distort the magnetic field, could
multimodality imaging system, PET/magnetic resonance (MR), using potentially deteriorate the accuracy and quality of MR images. Besides
sequential or even integrated scanner platforms has been available. This the interference with the magnetic field, conventional PET photo-
hybrid imaging technique is gradually being implemented into the clini- multiplier tubes (PMT) were not designed to be used inside strong
cal setting for cardiac imaging. electromagnetic fields and do not function properly in or near these
Because of its unique capabilities, CMR has become a key imaging fields. Consequently, the main challenge in combining PET and MR
modality in clinical cardiology practice and a widely accepted standard into one integrated system has been the development of MR-compatible
of reference for the quantification of left and right ventricular function, PET detector technology.4 Current integrated PET/MR systems are either
the assessment of global and regional wall motion abnormalities and based on avalanche photodiodes5 (APD; Siemens mMR Biograph) or
tissue characterization (scar, fibrosis, edema), as well as valve function. silicon photon multipliers6 (SiPM; GE Signa), where cross-interference
In contrast, PET is superb at quantification of myocardial perfusion with the MR is minimized.7
and coronary flow reserve as well as visualization and quantification
of particular metabolic processes at the molecular level.1 Combining Attenuation Correction in Positron Emission
both methods there is a range of complementary information, suggest- Tomography/Magnetic Resonance
ing the use of integrated cardiac PET/MR may be justified in routine For the attenuation correction of acquired PET data, modern PET and
setting for evaluation of different disease entities. However, the exact PET/CT systems use attenuation maps (µ-maps) that contain the
role and value of PET/MR for cardiovascular imaging has not yet been radiodensity of each body volume element for 511-keV photons. These
determined. Critical evaluation of cardiac PET/MR is needed regarding are typically calculated using transmission scans with external radio-
incremental value beyond diagnostic information provided by PET nuclide sources or coregistered computed tomography (CT) data, which
and CMR alone. Because cardiac PET/MR is still in its infancy, this needs an additional transformation to convert to the radiodensity for
chapter will be based on published studies and case reports, available 511-keV photons. However, for integrated PET/MR systems without a
evidence, and where not yet available, on personal experience and CT or external radionuclide source, new techniques for the creation of
expert opinion. attenuation maps are needed. One approach is based on tissue segmen-
tation using specialized computer algorithms to segment MR data into
TECHNICAL ASPECTS AND IMPLEMENTATION a fixed number of tissue types with a priori assigned coefficients of
radiodensity. The currently most common approach uses a multipoint
Positron Emission Tomography/Magnetic Resonance Dixon sequence for the segmentation into lung, fat, soft tissue, and
Scanners and Instrumentation background.8 However, one limitation of this method is that bones and
To generate fused PET/MR images, several approaches exist. In the calcifications are not assigned into a separate class but classified as soft
past the only practicable solution was to use software to register and tissue. Consequently, standardized uptake values of tissue close to bone
fuse separately acquired PET and MR data. Although this method might be significantly underestimated,9 which could particularly apply
works relatively well for body areas with little deformability such as to the retrosternal parts of the heart. However, underestimation seems
the head, imaging the heart poses problems with respect to coregis- to be rather small in cardiac imaging.8,10
tration as a result of patient breathing and cardiac motion, as well as To overcome this limitation, modified segmentation methods based
patient positioning.2 Therefore sequential PET/MR systems provide on ultrashort echo time (UTE) sequences can be used, segmenting
improvement of coregistration if the patient undergoes both scans tissue with very short T2* (such as bone) into a separate class. However,
in a row on a mobile table system without repositioning.3 Com- due to a rather small field of view, this technique is not yet usable in
pared with that, integrated PET/MR systems allow for a completely cardiac PET/MR imaging.
simultaneous data acquisition and in vivo observation of physiologic Because of the fact that all objects between the patient and the PET
processes. Moreover, this technique minimizes the likelihood of detector can potentially attenuate and thus compromise the acquired
movement-related misregistration and leads to a significant reduction of PET data, all instrumentation used in the PET field of view during PET
the scan time. data acquisition has to be optimized for PET transparency.11–13 This
However, for PET/MR the technical problem of system integration has particular significance for radiofrequency surface coils, due to their
is a major challenge attributed to the presence of magnetic fields. In unfavorable attenuation profiles. However, dedicated PET/MR surface
PET/MR the real goal has been to fully integrate both systems without coils with minimal attenuation for gamma quanta are commercially

574
CHAPTER 50  Cardiac Positron Emission Tomography/Magnetic Resonance 575

available. Alternatively, technical methods exist which allow for integra- artifacts or unexpected behavior of the segmentation algorithm can
tion of attenuation coefficients of standard coil systems into the attenu- cause more or less severe tissue misclassification, compromising the
ation map after these have been previously measured in a CT scanner; validity of attenuation-corrected PET data caused by wrong attenuation-
however, one limitation of this approach is that the expansion of the coefficients in the µ-map. Typical MR artifacts in cardiac imaging
attenuation map presumes knowledge of the exact coil position in the originate from foreign objects like implantable port systems, sternal
acquired image area.7 wires, artificial heart valves, or artificial joint replacement of the humerus.
Thus it is imperative that cardiac PET/MR reading should include visual
Motion Correction inspection of the underlying µ-maps. If significant errors are evident,
Electrocardiogram Gating findings in attenuation-corrected PET data should be interpreted with
To assign image data to a specific cardiac phase, electrocardiogram caution and correlation with uncorrected PET data should be performed.
(ECG)-based triggering is mandatory for MR and PET of the heart. If in doubt, an experienced reader should perform these tasks.
However, during MR, and thus also during integrated PET/MR, ECG Another typical attenuation-correction (AC)–related issue results
signals can be considerably distorted by the magnetic field and radio- from patient motion. In a typical setting, the MR-based µ-maps are
frequency pulses. Therefore special care is needed when applying the created in the beginning of the study and are later used for the AC of
electrodes and monitoring the signal. The comparably long cumulative PET data that get continuously acquired over time in list mode. If the
acquisition times of most CMR protocols allow for an extensive parallel patient changes body position following µ-map creation, this results
acquisition of PET signal, which typically compensates for the lost PET in misalignment between attenuation coefficients and PET data, which
data because of ECG gating, resulting in reconstructed PET images of can cause severe PET image artifacts and quantification bias. As a simple
high quality.14 workaround, it is recommended to perform repeated µ-map creation,
interleaved between main MR acquisitions. If patient motion is retro-
Magnetic Resonance-Based Motion Correction spectively detected, PET list-mode data can be truncated to an interval
One of the most promising, yet still experimental, technical new devel- before or after patient motion and an appropriate µ-map can be selected
opments of combined PET/MR is the advantage of motion detection for attenuation correction.
using ultrafast (“real-time”) three-dimensional (3D) MR acquisition
and tagging techniques. This allows for the MR-based estimation of Truncation of Field of View
motion-vector fields during PET acquisition, which can then be used Because of lack of space within the magnet and relatively long scan
to improve effective spatial resolution of PET and motion-induced times, cardiac PET/MR is usually performed with the patient’s arms
inaccuracies in PET quantification. Recent advances in this area suggest aligned along the body axis. Depending on the patient’s body habitus,
a relevant utility of this technology for simultaneous PET/MR cardiac this can result in parts of the arms being placed outside the MR field
imaging.15 of view. This can cause so-called truncation artifacts at the edges of
the attenuation maps, which can translate to errors in the attenuation-
Image Postprocessing, Visualization, and Quantification corrected PET data. These artifacts can at least partially be avoided by
To achieve a wide acceptance of complex cardiac imaging scans in extension of the MR field of view by the use of optimized readout
clinical practice, a semiautomated or even automated processing of gradients16 or by partial correction with PET emission data using
cardiac imaging data is required. Meanwhile, several software products maximum likelihood reconstruction of attenuation and activity
are available from commercial and academic sources. However, most (MLAA).17 However, the MLAA algorithm is restricted to radiotracers
of these software products focus on either CMR or PET, but a few that accumulate in the skin because it is based on contour detection
software packages analyzing both cardiac PET and MR data are avail- in PET images.
able as well (e.g., Munich Heart or syngo.via). For analysis of clinical
cardiac PET/MR scans, the respective software solutions should include Patient Preparation for Fludeoxyglucose Positron
tools for the assessment of ventricular function, viability, fibrosis and Emission Tomography/Magnetic Resonance Studies
scar, perfusion, flow quantification, and tissue composition (e.g., T1, Fludeoxyglucose (FDG) is still by far the most widely used tracer for
T2, T2* mapping) as well as dedicated postprocessing of cardiac PET cardiac PET. Normal myocardium is a very insulin-sensitive tissue that
data, including creation of bull’s eye plots and comparison of myocardial uses a variable mixture of glucose, lactate, ketone bodies, and free fatty
tracer uptake to normal databases. acids under uncontrolled metabolic conditions, with a preference for
fatty acids. Depending on the clinical question (inflammation/tumor
Common Pitfalls vs. viability), myocardial metabolism needs to be shifted to either free
Despite its complexity, integrated cardiac PET/MR has been demon- fatty-acid or glucose utilization in cardiac PET with FDG. Therefore
strated to be a robust and reliable imaging modality. This is based on careful patient preparation is of utmost importance. It is strongly rec-
our own experience and hundreds of scans performed in several spe- ommended to perform detailed patient interviews regarding the com-
cialized cardiovascular imaging centers and research institutes around pliance to the preparation protocol. This is ideally performed by a
the world. Given a certain expertise and knowledge of common pitfalls, nuclear medicine specialist before tracer injection. In cases of incompli-
cardiac PET/MR has shown to be reliable and robust, and thus suitable ance, the PET/MR scan can be postponed or certain countermeasures
for clinical routine imaging. Still, it is recommended that a team con- (e.g., insulin injection, unfractionated heparin injection, fatty-acid
sisting of a radiologist and a nuclear medicine specialist with expertise loading) can be taken. In addition to interviews, it is recommended to
in cardiac imaging should perform and interpret cardiac PET/MR, perform blood testing of glucose level and possibly insulin and free
supervising technicians with cardiac imaging experience. fatty-acid levels.
For cardiac FDG-PET of inflammation, infiltration, and tumors,
Segmentation and Misalignment Errors suppress glucose uptake into normal cardiomyocytes by means of low
The validity of attenuation-corrected PET data is directly dependent insulin levels and high levels of fatty acids to differentiate between
on the validity of the underlying MR tissue segmentation because the inflammatory infiltrates or tumor tissue and normal myocardium. This
creation of µ-maps is based on the MR data segmentation. MR image can be achieved using several techniques, including prolonged fasting,
576 SECTION VII  Economics and Guidelines

high-fat low-carbohydrate diet, fatty-acid loading, and additional injec- parallel acquisition and comparison of simultaneous PET and MR
tion of unfractionated heparin.18–20 However, fasting has been observed measurements of myocardial perfusion at rest and during pharmacologic
to be a major reason for patient discomfort, potentially contributing stress has been developed.27 A more recent study compared 13N-ammonia
to increased cancellation rates during cardiac PET/MR scans.14 In a PET/MR with single-photon emission computed tomography (SPECT)
recent study, we have described a high-fat low-carbohydrate protein- perfusion imaging and reported superior specificity and diagnostic
permitted diet without fasting that yields an 84% success rate regarding accuracy of PET/MR in patients with reversible ischemia.28
suppression of normal myocardial glucose uptake. Cancellation rate
was less than 3% and thus comparable with routine CMR scans.21 Acute Coronary Syndrome
For FDG-PET of myocardial viability it is necessary to significantly Infarct size is a strong predictor of outcome in patients with acute
raise insulin levels to favor the uptake of glucose by normal cardio- myocardial infarction (AMI). Both FDG-PET and LGE-CMR are clini-
myocytes. In principle, several techniques including hyperglycemic cally established techniques for the assessment of infarct size in the
clamping, administration of hypolipidemic agents (e.g., Acipimox), and chronic state. Initial PET/MR studies in the subacute phase after AMI
oral glucose loading are available. In our department, the following have also demonstrated moderate-to-good agreement between myo-
protocol for oral glucose loading has proved to be reliable, simple to cardial segments showing LGE and reduced FDG uptake14,29,30 (Figs.
perform, and well received by patients: patients fast before the scan, 50.1 and 50.2). Furthermore, CMR in the subacute phase of reperfused
which normally means skipping breakfast in the morning. The glucose AMI can differentiate between myocardial edema, microvascular obstruc-
level of diabetic patients should be below 150 mg/dL (8.3 mmol/L). tion, and intramyocardial hemorrhage, which is a potential indicator
Before the FDG injection, the patients orally receive 75 g glucose in a for reperfusion injury with additional prognostic impact also for the
preparation that is commercially available for glucose tolerance tests. right ventricle.31–33
Other studies have observed a certain mismatch between PET and
CMR. In cases of underestimation of infarct size using PET, this could
RADIATION EXPOSURE be explained by the higher spatial resolution of CMR and a higher
The effective radiation dose for patients undergoing cardiac FDG-PET sensitivity to detect subendocardial infarction. However, recently per-
usually ranges between 4 and 7 mSv.22 Because the overall PET/MR formed studies have identified patients with AMI where some myocardial
study duration is determined by the comparably long CMR acquisition segments demonstrated reduced FDG uptake but no LGE14,34 (Fig. 50.3).
time, this allows for a significant prolongation of the PET acquisition These myocardial segments had wall motion abnormalities comparable
interval, which might result in a significant reduction of the adminis- with that of infarcted (in terms of LGE) segments and showed only
tered activity and therefore the effective dose. In our own institution partial functional recovery after 6 months.30
the effective dose of cardiac FDG-PET/MR is approximately 2.5 ± 1.2 mSv, The discrimination between reversible and irreversible myocardial
yielding still significantly better PET image quality than our standard dysfunction in the subacute phase after AMI is of high clinical and
cardiac FDG-PET/CT scans and leaving room for further reduction of scientific interest. From the clinical perspective, reversible dysfunc-
the administered activity. Such radiation exposure goes significantly tion of myocardial segments will contribute to global left ventricular
below that reported for 64-slice coronary CT angiography23 and com- recovery. On the other hand, dysfunction of noninfarcted myocardial
petes with what is possible with third-generation dual-source CT.24 segments in periinfarct regions is associated with the salvage area, that
is, the difference between the area at risk and the final infarct size. A
CORONARY ARTERY DISEASE PET/MR study in patients with reperfused AMI has found that the
area of reduced FDG uptake correlates with the area at risk (as deter-
Suspected and Known Coronary Artery Disease mined by the endocardial surface area) and, in the absence of necrosis,
Published data on simultaneous PET/MR in patients with suspected is localized in the perfusion territory of the culprit artery.14 This has
or known coronary artery disease (CAD) is still sparse. The available been further substantiated by another study comparing the area at risk
data from PET/CT and MR studies indicate that simultaneous PET/ by FDG-PET to the area at risk by T2-mapping using simultaneous
MR has the potential to be a powerful tool for the detection of CAD PET/MR.35
and the assessment of myocardial viability. In integrated imaging pro- If finally confirmed, infarct size, area at risk, and salvage area could
tocols with PET tracers for perfusion imaging, viability imaging could be assessed with cardiac FDG-PET/MR and used as surrogate parameters
be performed using late gadolinium enhancement (LGE) CMR, which for the evaluation of strategies to reduce infarct size, such as precon-
seems not only to be the most sensitive modality for the detection of ditioning and postconditioning or remote conditioning.
myocardial scar, especially for small subendocardial scars, but would
also allow the omission of FDG imaging. This could greatly simplify
the PET scan because the complex task of a dual tracer study would
INFLAMMATORY HEART DISEASE
become obsolete. Moreover, myocardial perfusion PET has been dem- In recent years, FDG-PET/CT has attracted growing interest in the
onstrated to achieve the highest diagnostic accuracy in noninvasive detection and monitoring of inflammatory diseases; however, until now,
assessment of significant coronary artery stenosis and, in addition, allows no widespread clinical use of FDG-PET/CT has been observed for the
for the absolute quantification of myocardial blood flow.25 diagnosis of inflammatory heart disease.36 In contrast to PET, CMR is
In side-by-side comparison using sequential FDG-PET/CT and LGE an established imaging modality in the diagnostic workup and manage-
CMR in patients with known CAD, a close agreement between both ment of patients with cardiac inflammation.37–39 It is well known that
modalities was found for the detection of transmural myocardial scars. CMR can identify even small areas of myocardial damage using the
However, a significant number of segments with subendocardial LGE LGE technique or T1-mapping. In addition, CMR is a robust tool to
showed normal FDG uptake by PET, thus indicating a higher sensitivity accurately detect and even quantify regional and global wall motion
of LGE imaging for the detection of small myocardial scars.26 These abnormalities, to assess myocardial edema and hyperemia, as well as
findings have not yet been confirmed using simultaneous FDG-PET/MR. pericardial effusion. Thus a combination of multiparametric CMR with
Preliminary data show the feasibility of simultaneous PET/MR per- the high sensitivity and outstanding quantification capabilities of
fusion imaging using 13N-ammonia. In this context, a protocol for the FDG-PET could represent a very powerful imaging modality for cardiac
CHAPTER 50  Cardiac Positron Emission Tomography/Magnetic Resonance 577

A B

C D
FIG. 50.1  Fludeoxyglucose-positron emission tomography (PET)/magnetic resonance scan in a 59-year-old
male patient after reperfused acute occlusion of the left circumflex artery and a corresponding subendocardial
infarction of the lateral and inferior myocardial wall. (A) Four-chamber, (B) three-chamber, and (C) short-axis
views show the extent of the late gadolinium enhancement. (D) PET data mapped onto a polar plot and
overlaid with the 17-segment model of the left ventricle. (From Nensa F, Poeppel TD, Beiderwellen K, et al.
Hybrid PET/MR imaging of the heart: feasibility and initial results. Radiology. 2013;268:366–373.)

A B
FIG. 50.2  Fludeoxyglucose (FDG)-positron emission tomography/ magnetic resonance scan in a 53-year-old
male patient with reperfused acute occlusion of the proximal left anterior descending artery and consecutive
acute myocardial infarction. The short-axis late gadolinium enhancement scan shows large transmural infarc-
tion of the septal and anteroseptal wall with a hypointense area of microvascular obstruction (A, arrows). No
FDG uptake was observed in the infarct area (B). (From Nensa F, Poeppel TD, Beiderwellen K, et al. Hybrid
PET/MR imaging of the heart: feasibility and initial results. Radiology. 2013;268:366–373.)
578 SECTION VII  Economics and Guidelines

A C E G

B D F H
FIG. 50.3  Cardiac angiography and fludeoxyglucose positron emission tomography/magnetic resonance
(FDG-PET/MR) in a 44-year-old woman with an acute occlusion in segment three of the right coronary artery.
Cardiac angiograms show the culprit lesion before (A, arrow) and after (B) reperfusion (pain-to-balloon time
<40 min). PET/MR was performed 3 days after the cardiac event. Late gadolinium enhancement (LGE) polar
plot shows a complete absence of infarction (D), a finding that was also confirmed on the source LGE MR
image (C). Source PET image (E) and polar plot (F) show substantially reduced FDG uptake in the perfusion
territory of the right coronary artery (F, arrows). T2-weighted MR image shows myocardial edema in the
inferior wall (G), a finding that was in good agreement with the reduced FDG uptake seen on the fused
image (H). (From Nensa F, Poeppel T, Tezgah E, et al. Integrated FDG PET/MR imaging for the assessment
of myocardial salvage in reperfused acute myocardial infarction. Radiology. 2015;276:400–407.)

inflammation. In the near future, a number of non-FDG tracers against value of FDG-PET/MR and, most importantly, improved patient outcome
inflammation biomarkers, as recently demonstrated for Ga-68 pentixa- need to be performed.
for,40 could add significant clinical value to cardiac PET/MR.41
Cardiac Sarcoidosis
Myocarditis In patients with sarcoidosis, cardiac involvement is a strong predictor
To date, no published studies exist that evaluated PET/MR as a diag- of worse outcome, which often manifests in arrhythmia and heart failure.
nostic tool in patients with suspected or known myocarditis. An initial Thus early diagnosis and discrimination between active and chronic
case report has demonstrated the potential of FDG-PET/MR in a patient states are prerequisites for adequate treatment and can contribute to
with myocarditis caused by parvovirus B19.42 In this report, a typically the reduction of overall morbidity and mortality. Commonly, treatment
focal subepicardial LGE was observed which was closely matched by of cardiac sarcoidosis comprises both symptomatic management of
intense FDG uptake in this area and accompanied by myocardial edema cardiac dysfunction and immunosuppressive therapy. However, immu-
and hyperemia (Fig. 50.4). Although CMR would have been sufficient nosuppression needs to be carefully balanced against harmful side effects.
for the detection and diagnosis of myocarditis in this case, it highlights Hence, alongside early diagnosis, continuous monitoring of disease
the ability of FDG-PET to quantify inflammatory activity, particularly activity with the objective of guided dose adaptation is needed. Accurate
suitable for the use in disease monitoring. A similar case report was detection and monitoring of cardiac sarcoidosis still remain challenging,
published in a patient with myocarditis due to Epstein-Barr virus infec- especially as endomyocardial biopsy significantly suffers from sampling
tion, with a more diffuse FDG uptake in the lateral wall that closely error and can cause severe complications such as myocardial perfora-
matched LGE and myocardial edema.43 tion and pericardial tamponade. Therefore noninvasive techniques such
Neutrophils and the monocyte/macrophage family express high levels as CMR and FDG-PET represent promising alternatives in the clinical
of glucose transporters and hexokinase activity, which results in increased workup of patients with cardiac sarcoidosis.45 In fact, both CMR and
uptake and accumulation of FDG in inflammatory infiltrates. As such, FDG-PET are recommended for the assessment of cardiac sarcoidosis.45
FDG uptake is truly complementary to LGE (representing myocardial CMR has been demonstrated to predict death and other adverse events
necrosis), T2-weighted CMR (representing myocardial edema) and early in suspected cardiac sarcoidosis46 and a metaanalysis (7 studies, 164
gadolinium enhancement (representing myocardial hyperemia) and patients) reported a pooled 89% sensitivity and 78% specificity for
could be useful to extend the so-called Lake Louise Criteria in the FDG-PET in the detection of cardiac involvement in sarcoidosis.47 In
imaging assessment of suspected myocarditis.44 In future, FDG-PET a comparative study of CMR and FDG-PET, CMR provided a higher
could not only complement CMR in the diagnosis of myocarditis, but negative predictive value and thus might be superior for ruling out
could also allow for quantitative assessment of inflammation and moni- cardiac involvement.48
toring of disease activity or improve differentiation between acute and Also, there is some evidence that a combination of FDG-PET and
chronic/persistent myocarditis. This is particularly relevant because CMR might provide added value.49 In fact, both techniques visualize
more severe myocardial inflammation is thought to predict progression different pathologic correlates of cardiac sarcoidosis. Although the LGE
to dilated cardiomyopathy, increased risk of arrhythmias, and chronic technique can accurately detect myocardial necrosis, fibrosis, and scar-
heart failure. Nevertheless, further studies showing added diagnostic ring, FDG uptake is a quantitative surrogate parameter of increased
CHAPTER 50  Cardiac Positron Emission Tomography/Magnetic Resonance 579

A B C

60
50
Signal Intensity

40
30
20
10
0 10 20 30 40 50
D
Seconds
FIG. 50.4  Multiparametric fludeoxyglucose (FDG) positron emission tomography/magnetic resonance scan
in a patient with acute viral myocarditis caused by parvovirus B19. Subepicardial late gadolinium enhance-
ment (A) was found in the left ventricular (LV) anterior wall that was in excellent agreement with increased
FDG uptake (B). T2-weighted images revealed an edema in the LV apex (C). Dynamic perfusion imaging
revealed hyperemia in the LV anterior wall (D, plot at bottom right). (From Nensa F, Poeppel TD, Krings P,
et al. Multiparametric assessment of myocarditis using simultaneous positron emission tomography/magnetic
resonance imaging. Eur Heart J. 2014;35:2173.)

glucose metabolism, a hallmark of inflammation. Thus a combination response, as already discussed in the context of myocardial inflamma-
of CMR and FDG-PET has the potential to provide both accurate detec- tion. Until now, no studies on the use of FDG-PET/MR in infectious
tion and an assessment of disease activity. Several case reports have endocarditis have been published. However, one case report dem-
demonstrated the feasibility of integrated FDG-PET/MR in the detec- onstrates the feasibility of FDG-PET/MR in a patient with Loeffler
tion50,51 and therapy monitoring52 of cardiac sarcoidosis (Fig. 50.5). One endocarditis.55
study in 51 consecutive patients with cardiac sarcoidosis has found
improved diagnostic accuracy of combined FDG-PET/MR over FDG-PET
and CMR alone.53 Despite the sparseness of available studies, integrated
CARDIAC TUMORS
FDG-PET/MR holds great potential in the imaging of cardiac sarcoidosis. Given the role of CMR in the imaging of cardiac masses and the overall
However further evaluation, particularly with respect to clinical outcome significance of FDG-PET in oncologic imaging, a combination of both
of patients, is warranted. seems to be a powerful combination for the diagnostic workup of cardiac
tumors. Both modalities alone have already been demonstrated to yield
Endocarditis high diagnostic accuracy in the differentiation of benign and malignant
Infective endocarditis is a potentially lethal disease if not immediately tumors, which is, in most cases, the crucial question before treatment.
treated with antibiotics. In patients with prosthetic valves, complica- Nevertheless, in a small pilot study including 20 patients with cardiac
tions of inflammation include prosthetic-valve dehiscence, paravalvu- masses, integrated assessment using PET/MR yielded improved diag-
lar leaks, and abscesses. CMR is a robust imaging modality that can nostic accuracy over PET or MR only assessment56 (Fig. 50.6). Moreover,
visualize the infected valve, identify and quantify valve regurgitation, a Journal of the American College of Cardiology imaging vignette shows
and may even detect dislocation of the prosthetic valve.54 However, the potential of PET/MR in the diagnosis of cardiac and paracardiac
severe image artifacts frequently found in the vicinity of mechani- masses with histopathologic correlation.57 Considering the already strong
cal valves are a well-known limitation of CMR and can significantly diagnostic performance of PET/CT and CMR, high cost, and limited
compromise image quality and diagnostic accuracy. Review of nonat- availability of PET/MR scanners, integrated PET/MR imaging might
tenuated PET images is always mandatory because extinction artifacts be reserved for selected cases of cardiac tumors where true benefit can
in MR images lead to erroneous µ-maps causing underestimation of be expected. Such cases could include the planning of surgery in patients
tracer uptake in attenuation-corrected PET images. In addition to the with complex cardiac infiltration or the differentiation of scar tissue
morphologic imaging of CMR, FDG-PET allows for the assessment versus relapse in follow-up scans after surgery or radiation therapy of
of inflammatory activity and may be well suited to monitor therapy cardiac malignancies.56
580 SECTION VII  Economics and Guidelines

A B

C D
FIG. 50.5  Fludeoxyglucose positron emission tomography/magnetic resonance (FDG-PET/MR) scan in a
patient with general malaise, acute retrosternal chest pain, and palpitations. Baseline scan (A, B) showed
bilateral hilar lymphadenopathy (A, asterisks) and focal late gadolinium enhancement in the lateral left ven-
tricular wall (A, arrows). Both lymph nodes and myocardial lesions showed intense FDG uptake (B, arrows)
and sarcoidosis with myocardial involvement was diagnosed. After 4 weeks of treatment, PET still demon-
strated increased lymphonodular and myocardial (C, arrows) FDG uptake, whereas FDG uptake was signifi-
cantly reduced after 4 months of treatment (D, arrows). In contrast with FDG uptake, myocardial late gadolinium
enhancement remained constant in all three scans, and thus was not an indicator of treatment response.
(From Nensa F, Tezgah E, Poeppel T, et al. Diagnosis and treatment response evaluation of cardiac sarcoidosis
using positron emission tomography/magnetic resonance imaging. Eur Heart J. 2014;36:550.)

position of the patient’s arms (elevated vs. aligned to the body axis)
REPORTING
should be specified.
The practical implementation as well as the interpretation of cardiac Depending on the CMR sequences used, the report should provide
PET/MR scans should be performed with cooperation between the detailed information about the localization and extent of structural
radiology and nuclear medicine departments by experts in cardiac abnormalities of the myocardium including edema, perfusion deficits,
imaging. The documentation of the procedure should comprise a joint infarcts, microvascular obstruction, hemorrhage, and fibrosis. Usually,
consensus report by a radiologist and a nuclear medicine specialist. the report should include data of left and/or right ventricular function
All examination-related patient preparation should be carefully analysis, analysis of regional wall motion, and myocardial mass. It also
recorded (e.g., fasting or other dietary precautions, glucose loading). may include ventricular dimensions, which correlate to dimensions as
The name, dose, and route of administration of regulated nonradioac- measured in echocardiography to improve comparability.
tive drugs and contrast agents should also be stated. Blood glucose level For PET, the report should include a description of the location,
and, if available, free-fatty acids level before FDG administration and the extent, and the intensity of pathologic tracer accumulation (speci-
heparin injection should be reported. Study-specific details should fied as standardized uptake values) or reduction related to normal tissue.
include the radiopharmaceutical, the amount of injected activity in Extracardiac pathologies should be reported as well. If present, con-
megabecquerels (MBq), the route of administration (intravenous), and founding factors that might influence the sensitivity or specificity of
the date and time of administration. Furthermore, the used PET/MR the study should be mentioned such as ubiquitous and undesired high
system and instrumentation, such as coils, should be specified. or low tracer uptake in the myocardium (depending on the preparation
A description of the procedure should include the time interval and acquisition protocol).
between administration of the tracer and the start time of the acquisi- In the conclusion, the study should be identified as normal or abnor-
tion, as well as a brief description of the CMR protocol. Also, the mal. The question asked in the study requisition should be directly
CHAPTER 50  Cardiac Positron Emission Tomography/Magnetic Resonance 581

A B

C D
FIG. 50.6  Images of a patient with history of breast cancer (A, B). Images of a patient with anal cancer
(C, D). In both patients, large intracavitary masses were initially found in echocardiography. Arrows in (A)
and (C) show steady-state free precession (SSFP) images of large tubular masses in superior vena cava and
right atrium (A) and in right ventricular outflow tract (C). (B, D) Fusion images of SSFP and positron emission
tomography (PET) clearly demonstrating malignancy in the patient with breast cancer (B) and benign mass
in the patient with anal cancer (D). However, closer inspection of PET images revealed mediastinal lymph
node metastases in both patients (B, D; arrows). (From Nensa F, Tezgah E, Poeppel TD, et al. Integrated
18F-FDG-PET/MRI in the assessment of cardiac masses: a pilot study. J Nucl Med. 2014;56:255–260.)

addressed. If possible, a definite diagnosis should be stated. If necessary, definition of new compound biomarkers. Further technical improve-
recommendations for further imaging procedures should be given. Joint ments like MR-based PET motion correction will lead to higher spatial
approval from a radiologist and a nuclear medicine specialist should and temporal resolution, enabling advanced applications such as the
be performed. imaging of valves and coronary arteries. Finally, the translation of inno-
vative PET tracers from preclinical imaging into the clinical routine
will open up exiting new possibilities. Promising fields of application
CONCLUSION include suspected CAD and AMI, inflammatory heart diseases such as
Integrated PET/MR has been clinically available for a few years and myocarditis and cardiac sarcoidosis, and different cardiomyopathies,
available studies and case reports have shown cardiac PET/MR to be as well as cardiac tumors.
reliable and suitable for clinical use. The MR system is not affected by However, before the future clinical establishment of cardiac PET/
the integrated PET, and MR-based AC provides sufficient accuracy for MR, further studies need to demonstrate added value in comparison
most clinical applications. with current standard diagnostic procedures regarding diagnosis, moni-
PET/MR has the potential to provide a more accurate and earlier toring, and, most importantly, patient outcome, to justify the investments
diagnosis, as well as monitoring of cardiac diseases using the comple- in this rather complex and expensive technology.
mentary strengths of both techniques. The prospects of multiparametric
imaging need to be translated into diagnostic advantage. This requires
dedicated multiparametric imaging protocols, specialized software for
REFERENCES
the integrated reading of multiparametric PET/MR studies, and the A full reference list is available online at ExpertConsult.com
CHAPTER 50  Cardiac Positron Emission Tomography/Magnetic Resonance 581.e1

REFERENCES 18. Ishida Y, Yoshinaga K, Miyagawa M, et al. Recommendations for (18)F-


fluorodeoxyglucose positron emission tomography imaging for cardiac
1. Hendel RC, Berman DS, Di Carli MF, et al. ACCF/ASNC/ACR/AHA/ASE/ sarcoidosis: Japanese Society of Nuclear Cardiology recommendations.
SCCT/SCMR/SNM 2009 Appropriate Use Criteria for Cardiac Ann Nucl Med. 2014;28(4):393–403. doi:10.1007/s12149-014-0806-0.
Radionuclide Imaging: a report of the American College of Cardiology 19. Williams G, Kolodny GM. Suppression of myocardial 18F-FDG uptake by
Foundation Appropriate Use Criteria Task Force, the American Society of preparing patients with a high-fat, low-carbohydrate diet. AJR Am J
Nuclear Cardiology, the American College of Radiology, the American Roentgenol. 2008;190(2):W151–W156. doi:10.2214/AJR.07.2409.
Heart Association, the American Society of Echocardiography, the Society 20. Manabe O, Yoshinaga K, Ohira H, et al. The effects of 18-h fasting with
of Cardiovascular Computed Tomography, the Society for Cardiovascular low-carbohydrate diet preparation on suppressed physiological
Magnetic Resonance, and the Society of Nuclear Medicine. J Am Coll myocardial (18)F-fluorodeoxyglucose (FDG) uptake and possible
Cardiol. 2009;53(23):2201–2229. doi:10.1016/j.jacc.2009.02.013. minimal effects of unfractionated heparin use in patients with suspected
2. Nekolla SG, Martinez-Moeller A, Saraste A. PET and MRI in cardiac cardiac involvement sarcoidosis. J Nucl Cardiol. 2016;23(2):244–252.
imaging: from validation studies to integrated applications. Eur J Nucl doi:10.1007/s12350-015-0226-0.
Med Mol Imaging. 2009;36(suppl 1):S121–S130. doi:10.1007/ 21. Nensa F, Tezgah E, Schweins K, et al. Evaluation of a low-carbohydrate
s00259-008-0980-1. diet-based preparation protocol without fasting for cardiac PET/MR
3. Torigian DA, Zaidi H, Kwee TC, et al. PET/MR imaging: technical aspects imaging. J Nucl Cardiol. 2017;24(3):980–988. doi:10.1007/s12350-016
and potential clinical applications. Radiology. 2013;267(1):26–44. -0443-1.
doi:10.1148/radiol.13121038. 22. Flotats A, Knuuti J, Gutberlet M, et al. Hybrid cardiac imaging: SPECT/
4. Zaidi H, Ojha N, Morich M, et al. Design and performance evaluation of CT and PET/CT. A joint position statement by the European Association
a whole-body Ingenuity TF PET-MRI system. Phys Med Biol. of Nuclear Medicine (EANM), the European Society of Cardiac
2011;56(10):3091–3106. doi:10.1088/0031-9155/56/10/013. Radiology (ESCR) and the European Council of Nuclear Cardiology
5. Delso G, Fürst S, Jakoby B, et al. Performance measurements of the (ECNC). Eur J Nucl Med Mol Imaging. 2011;38(1):201–212. doi:10.1007/
Siemens mMR integrated whole-body PET/MR scanner. J Nucl Med. s00259-010-1586-y.
2011;52(12):1914–1922. doi:10.2967/jnumed.111.092726. 23. Hausleiter J, Meyer T, Hermann F, et al. Estimated radiation dose
6. Schaart DR, van Dam HT, Seifert S, et al. A novel, SiPM-array-based, associated with cardiac CT angiography. JAMA. 2009;301(5):500–507.
monolithic scintillator detector for PET. Phys Med Biol. doi:10.1001/jama.2009.54.
2009;54(11):3501–3512. doi:10.1088/0031-9155/54/11/015. 24. Mangold S, Wichmann JL, Schoepf UJ, et al. Automated tube voltage
7. Quick HH. Integrated PET/MR. J Magn Reson Imaging. 2013;39(2):243– selection for radiation dose and contrast medium reduction at coronary
258. doi:10.1002/jmri.24523. CT angiography using 3(rd) generation dual-source CT. Eur Radiol.
8. Martinez-Möller A, Souvatzoglou M, Delso G, et al. Tissue classification 2016;26(10):3608–3616. doi:10.1007/s00330-015-4191-4.
as a potential approach for attenuation correction in whole-body PET/ 25. Jaarsma C, Leiner T, Bekkers SC, et al. Diagnostic performance of
MRI: evaluation with PET/CT data. J Nucl Med. 2009;50(4):520–526. noninvasive myocardial perfusion imaging using single-photon emission
doi:10.2967/jnumed.108.054726. computed tomography, cardiac magnetic resonance, and positron
9. Samarin A, Burger C, Wollenweber SD, et al. PET/MR imaging of bone emission tomography imaging for the detection of obstructive coronary
lesions—implications for PET quantification from imperfect attenuation artery disease: a meta-analysis. J Am Coll Cardiol. 2012;59(19):1719–1728.
correction. Eur J Nucl Med Mol Imaging. 2012;39(7):1154–1160. doi:10.1016/j.jacc.2011.12.040.
doi:10.1007/s00259-012-2113-0. 26. Hunold P, Brandt-Mainz K, Freudenberg L, et al. [Evaluation of
10. Heusch P, Buchbender C, Beiderwellen K, et al. Standardized uptake myocardial viability with contrast-enhanced magnetic resonance
values for [18F] FDG in normal organ tissues: comparison of whole-body imaging—comparison of the late enhancement technique with positron
PET/CT and PET/MRI. Eur J Radiol. 2013;82(5):870–876. doi:10.1016/ emission tomography]. Rofo. 2002;174(7):867–873. doi:10.105
j.ejrad.2013.01.008. 5/s-2002-32697.
11. Delso G, Martinez-Möller A, Bundschuh RA, et al. Evaluation of the 27. Zhang HS, Rischpler C, Langwieser N, et al. Simultaneous measurement
attenuation properties of MR equipment for its use in a whole-body of myocardial perfusion by dynamic contrast enhancement MR and
PET/MR scanner. Phys Med Biol. 2010;55(15):4361–4374. ammonia PET. Proc Intl Soc Magn Reson Med. 2013;21:0576.
doi:10.1088/0031-9155/55/15/011. 28. Lau JMC, Laforest R, Zheng J, Woodard P. 13N-ammonia PET/MR
12. Tellmann L, Quick HH, Bockisch A, Herzog H, Beyer T. The effect of MR myocardial stress perfusion imaging early experience. J Nucl Med.
surface coils on PET quantification in whole-body PET/MR: results from 2014;55(suppl 1):242.
a pseudo-PET/MR phantom study. Med Phys. 2011;38(5):2795–2805. 29. Schlosser T, Nensa F, Mahabadi AA, Poeppel TD. Hybrid MRI/PET of the
doi:10.1118/1.3582699. heart: a new complementary imaging technique for simultaneous
13. Paulus DH, Braun H, Aklan B, Quick HH. Simultaneous PET/MR acquisition of MRI and PET data. Heart. 2012;99(5):351–352.
imaging: MR-based attenuation correction of local radiofrequency doi:10.1136/heartjnl-2012-302740.
surface coils. Med Phys. 2012;39(7):4306–4315. doi:10.1118/ 30. Rischpler C, Langwieser N, Souvatzoglou M, et al. PET/MRI early after
1.4729716. myocardial infarction: evaluation of viability with late gadolinium
14. Nensa F, Poeppel TD, Beiderwellen K, et al. Hybrid PET/MR imaging of enhancement transmurality vs. 18F-FDG uptake. Eur Heart J Cardiovasc
the heart: feasibility and initial results. Radiology. 2013;268(2):366–373. Imaging. 2015;16(6):661–669. doi:10.1093/ehjci/jeu317.
doi:10.1148/radiol.13130231. 31. Kandler D, Lücke C, Grothoff M, et al. The relation between hypointense
15. Petibon Y, Ouyang J, Zhu X, et al. Cardiac motion compensation and core, microvascular obstruction and intramyocardial haemorrhage in
resolution modeling in simultaneous PET-MR: a cardiac lesion detection acute reperfused myocardial infarction assessed by cardiac magnetic
study. Phys Med Biol. 2013;58(7):2085–2102. resonance imaging. Eur Radiol. 2014;24(12):3277–3288. doi:10.1007/
doi:10.1088/0031-9155/58/7/2085. s00330-014-3318-3.
16. Blumhagen JO, Ladebeck R, Fenchel M, Scheffler K. MR-based field-of- 32. Grothoff M, Elpert C, Hoffmann J, et al. Right ventricular injury in
view extension in MR/PET: B(0) homogenization using gradient ST-elevation myocardial infarction: risk stratification by visualization of
enhancement (HUGE). Magn Reson Med. 2012;70(4):1047–1057. wall motion, edema, and delayed-enhancement cardiac magnetic
doi:10.1002/mrm.24555. resonance. Circ Cardiovasc Imaging. 2012;5(1):60–68. doi:10.1161/
17. Nuyts J, Bal G, Kehren F, Fenchel M, Michel C, Watson C. Completion of CIRCIMAGING.111.967810.
a truncated attenuation image from the attenuated PET emission data. 33. Stillman AE, Oudkerk M, Bluemke D, et al; European Society of Cardiac
IEEE Trans Med Imaging. 2013;32(2):237–246. doi:10.1109/TMI Radiology. Assessment of acute myocardial infarction: current status and
.2012.2220376. recommendations from the North American Society for Cardiovascular
581.e2 SECTION VII  Economics and Guidelines

Imaging and the European Society of Cardiac Radiology. Int J Cardiovasc 47. Youssef G, Leung E, Mylonas I, et al. The use of 18F-FDG PET in the
Imaging. 2011;27(1):7–24. doi:10.1007/s10554-010-9714-0. diagnosis of cardiac sarcoidosis: a systematic review and metaanalysis
34. Nensa F, Poeppel T, Tezgah E, et al. Integrated FDG PET/MR imaging for including the Ontario experience. J Nucl Med. 2012;53(2):241–248.
the assessment of myocardial salvage in reperfused acute myocardial doi:10.2967/jnumed.111.090662.
infarction. Radiology. 2015;276(2):140564. doi:10.1148/radiol.2015140564. 48. Campbell P, Stewart GC, Padera RF, et al. Evaluation for cardiac
35. Bulluck H, White SK, Fröhlich GM, et al. Quantifying the area at risk in sarcoidosis—uncertainty despite contemporary multi-modality imaging.
reperfused ST-segment–elevation myocardial infarction patients using J Card Fail. 2010;16(8):S107. doi:10.1016/j.cardfail.2010.06.373.
hybrid cardiac positron emission tomography–magnetic resonance 49. Ohira H, Birnie DH, Pena E, et al. Comparison of (18)
imaging. Circ Cardiovasc Imaging. 2016;9(3):e003900. doi:10.1161/ F-fluorodeoxyglucose positron emission tomography (FDG PET) and
CIRCIMAGING.115.003900. cardiac magnetic resonance (CMR) in corticosteroid-naive patients with
36. Erba PA, Sollini M, Lazzeri E, Mariani G. FDG-PET in cardiac infections. conduction system disease due to cardiac sarcoidosis. Eur J Nucl Med Mol
Semin Nucl Med. 2013;43(5):377–395. doi:10.1053/j. Imaging. 2016;43(2):259–269. doi:10.1007/s00259-015-3181-8.
semnuclmed.2013.04.003. 50. White JA, Rajchl M, Butler J, Thompson RT, Prato FS, Wisenberg G.
37. Krieghoff C, Barten MJ, Hildebrand L, et al. Assessment of sub-clinical Active cardiac sarcoidosis: first clinical experience of simultaneous
acute cellular rejection after heart transplantation: comparison of cardiac positron emission tomography-magnetic resonance imaging for the
magnetic resonance imaging and endomyocardial biopsy. Eur Radiol. diagnosis of cardiac disease. Circulation. 2013;127(22):e639–e641.
2014;24(10):2360–2371. doi:10.1007/s00330-014-3246-2. doi:10.1161/CIRCULATIONAHA.112.001217.
38. Eitel I, Lücke C, Grothoff M, et al. Inflammation in takotsubo 51. Schneider S, Batrice A, Rischpler C, Eiber M, Ibrahim T, Nekolla SG.
cardiomyopathy: insights from cardiovascular magnetic resonance Utility of multimodal cardiac imaging with PET/MRI in cardiac
imaging. Eur Radiol. 2010;20(2):422–431. doi:10.1007/s00330-009-1549-5. sarcoidosis: implications for diagnosis, monitoring and treatment. Eur
39. Gutberlet M, Spors B, Thoma T, et al. Suspected chronic myocarditis at Heart J. 2014;35(5):312. doi:10.1093/eurheartj/eht335.
cardiac MR: diagnostic accuracy and association with 52. Nensa F, Tezgah E, Poeppel T, Nassenstein K, Schlosser T. Diagnosis and
immunohistologically detected inflammation and viral persistence. treatment response evaluation of cardiac sarcoidosis using positron
Radiology. 2008;246(2):401–409. doi:10.1148/radiol.2461062179. emission tomography/magnetic resonance imaging. Eur Heart J.
40. Rischpler C, Nekolla SG, Kossmann H, et al. Upregulated myocardial 2014;36(9):550. doi:10.1093/eurheartj/ehu473.
CXCR4-expression after myocardial infarction assessed by simultaneous 53. Wicks E, Menezes L, Pantazis A, et al. 135 novel hybrid positron emission
GA-68 pentixafor PET/MRI. J Nucl Cardiol. 2016;23(1):131–133. tomography—magnetic resonance (PET-MR) multi-modality
doi:10.1007/s12350-015-0347-5. inflammatory imaging has improved diagnostic accuracy for detecting
41. Wu C, Li F, Niu G, Chen X. PET imaging of inflammation biomarkers. cardiac sarcoidosis. Heart. 2014;100(suppl 3):A80. doi:10.1136/heart
Theranostics. 2013;3(7):448–466. doi:10.7150/thno.6592. jnl-2014-306118.135.
42. Nensa F, Poeppel TD, Krings P, Schlosser T. Multiparametric assessment 54. Pham N, Zaitoun H, Mohammed TL, et al. Complications of aortic valve
of myocarditis using simultaneous positron emission tomography/ surgery: manifestations at CT and MR imaging. Radiographics.
magnetic resonance imaging. Eur Heart J. 2014;35(32):2173. doi:10.1093/ 2012;32(7):1873–1892. doi:10.1148/rg.327115735.
eurheartj/ehu086. 55. Langwieser N, von Olshausen G, Rischpler C, Ibrahim T. Confirmation of
43. Olshausen GV, Hyafil F, Langwieser N, Laugwitz K-L, Schwaiger M, diagnosis and graduation of inflammatory activity of Loeffler
Ibrahim T. Detection of acute inflammatory myocarditis in Epstein Barr endocarditis by hybrid positron emission tomography/magnetic
virus infection using hybrid 18F-fluoro-deoxyglucose–positron emission resonance imaging. Eur Heart J. 2014;35(36):2496. doi:10.1093/eurheartj/
tomography/magnetic resonance imaging. Circulation. 2014;130(11):925– ehu148.
926. doi:10.1161/CIRCULATIONAHA.114.011000. 56. Nensa F, Tezgah E, Poeppel TD, et al. Integrated 18F-FDG-PET/MRI in
44. Friedrich MG, Sechtem U, Schulz-Menger J, et al. Cardiovascular the assessment of cardiac masses: a pilot study. J Nucl Med.
magnetic resonance in myocarditis: a JACC white paper. J Am Coll 2014;56(2):255–260. doi:10.2967/jnumed.114.147744.
Cardiol. 2009;53(17):1475–1487. doi:10.1016/j.jacc.2009.02.007. 57. Yaddanapudi K, Brunken R, Tan CD, Rodriguez ER, Bolen MA. PET-MR
45. Schatka I, Bengel FM. Imaging of cardiac sarcoidosis. J Nucl Med. imaging in evaluation of cardiac and paracardiac masses with
2013;55(1):99–106. doi:10.2967/jnumed.112.115121. histopathologic correlation. JACC Cardiovasc Imaging. 2016;9(1):82–85.
46. Greulich S, Deluigi CC, Gloekler S, et al. CMR imaging predicts death doi:10.1016/j.jcmg.2015.04.028.
and other adverse events in suspected cardiac sarcoidosis. JACC
Cardiovasc Imaging. 2013;6(4):501–511. doi:10.1016/j.jcmg.2012.10.021.
51 
Guidelines for Cardiovascular
Magnetic Resonance
Christopher M. Kramer and Michael Salerno

Cardiovascular magnetic resonance (CMR) has become an increasingly document regarding indications for CMR that was published in 2004
important tool in the armamentarium of the cardiovascular imager. It and developed through the ESC.7
has also had an increasingly prominent role in guidelines developed by CMR plays an important role in US multimodality guidelines
various societies, including the Society for Cardiovascular Magnetic with additional guidelines soon to be published. One crucial set of
Resonance (SCMR), American College of Cardiology (ACC), American expert documents are the appropriate use criteria (AUC) developed
Heart Association (AHA), European Society of Cardiology (ESC), and by the ACC, in conjunction with many other societies that outline
other societies. The role of guidelines is to direct the field, reduce unwar- which imaging tests are (A) appropriate, may be (M) appropriate, or
ranted variability, and improve the overall quality of the practice of are rarely (R) appropriate, given a particular clinical scenario. The
cardiovascular imaging. Because it often takes years from concept to first set of AUC guidelines for CMR was published in conjunction
publication for societal guideline recommendations to be developed, with computed tomography (CT) in 20068 and evaluated each single
they generally lag behind the state of the science and clinical trials and imaging modality against appropriateness. This document evaluated
therefore must be taken into context. This chapter will review the state only 33 clinical scenarios and identified 17 as appropriate for CMR,
of CMR in recent guidelines. 7 as uncertain (now termed may be appropriate), 9 as inappropriate
The SCMR has put out several guideline documents. The first was (now termed rarely appropriate). Appropriate indications for CMR
the 2008 document that created standardized protocols for performance included: (1) stress testing in patients who had intermediate pretest
of CMR.1 These protocols included most CMR procedures performed, probability of coronary artery disease (CAD) and were unable to
from stress imaging to evaluation of cardiomyopathies, pericardial dis- exercise or had an uninterpretable electrocardiogram (ECG); and
eases, and so on. Some of the CMR manufacturers used these guidelines (2) those with a stenosis identified by x-ray or CT angiography of
to create protocols on their scanners that could be used to follow these unclear significance. Most of the appropriate indications were in the
guidelines. These protocols were updated in 20132 and newer imaging structure and function category, such as postmyocardial infarction,
pulse sequences, such as T1 and T2 mapping, were incorporated into cardiomyopathies, myocarditis, complex congenital heart disease,
the protocols.2a These will continue to be updated as the field evolves. cardiac masses, pericardial disease, and preatrial fibrillation abla-
Neither of these documents covered congenital heart disease protocols tion. Myocardial viability and valvular assessments were also deemed
in sufficient depth and thus SCMR commissioned a separate protocol appropriate.
document to cover these diverse disease processes.3 The initial AUC documents were single modality. Since that time,
Soon after the initial protocols document was published, the SCMR the field has evolved toward the development of multimodality imaging
commissioned a document reviewing standardized reporting guide- documents. The first of this type was the 2014 publication regarding
lines.4 This document reviewed idealized reports for studies includ- the evaluation of patients with suspected or known ischemic heart
ing how to report stress tests, volumetric studies, and so on. Many disease.9 In the ischemic heart disease document, CMR stress testing
centers and report vendors have incorporated these guidelines into was rated appropriate in the same scenarios as listed previously for the
their reporting templates. Finally, the SCMR developed a document single modality document, but also in symptomatic patients with high
reviewing proper analysis techniques for various types of CMR images.5 pretest probability of CAD. CMR was also deemed appropriate in patients
These included guidance for the quantification of left ventricular (LV) with new systolic or diastolic heart failure and those with high-grade
volumes from cine imaging and scar quantification from late gadolinium ventricular arrhythmias. Additional appropriate indications for CMR
enhanced (LGE) images amongst others. This document set the stan- include patients with abnormal resting ECG and intermediate to high
dards for analysis that are being used in most CMR laboratories around global CAD risk, and those with abnormal ECG stress studies or stenosis
the world. identified on prior x-ray or CT coronary angiography. Uncertain ECG
In 2010, the ACC published a document that reviewed the state of stress findings or uncertain x-ray or CT coronary angiography results
the art in CMR including most of the standard clinical techniques and were also appropriate indications for CMR. Patients with new or wors-
the evidence behind them.6 This document was a consensus document ening symptoms and prior ECG stress testing, nonobstructive CAD on
with input from other societies including the AHA, SCMR, American x-ray angiography, obstructive CAD on CT coronary angiography, or
College of Radiology (ACR), and North American Society of Cardio- coronary calcium score >100 were thought to be appropriate. Postre-
vascular Imaging (NASCI). The document was quite comprehensive vascularization patients who were symptomatic with an ischemic equiva-
and served as an up-to-date review of the field at that time. However, lent were appropriate for CMR stress testing. Many other indications
it did not serve as a guideline because no recommendations were made were considered as may be appropriate depending on the individual
as to when a certain CMR procedure should or should not be used in patient’s clinical characteristics. AUC documents presently in prepara-
a given clinical scenario. This document replaced an older consensus tion are in the structure and function category and will cover both

582
CHAPTER 51  Guidelines for Cardiovascular Magnetic Resonance 583

myocardial and valvular diseases, which will likely include more of the ACC and ESC have led the way in the development of guidelines
key strengths of CMR. that cover specific disease processes and point out where CMR is of use
Another set of guidelines for the management of patients with stable in these diseases. The first such document was the 2008 ACC guideline
ischemic heart disease were published in 2012 by the ACC, in collabora- for the management of patients with congenital heart disease.13 CMR
tion with multiple other organizations.10 This document used slightly was recommended as an important modality to have available at a
different gradings from the previously mentioned AUC documents. center that specializes in caring for the congenital heart disease patient.
Recommendations are classified as class 1 (procedure should be per- CMR was noted to be useful in many clinical conditions involving
formed), class IIa (it is reasonable to perform the procedure), class IIb congenital heart disease, especially the postoperative patient, and was
(the procedure may be considered, or class III (procedure should not deemed complementary to ECG for many of these patients. In 2013, the
be performed). The level of evidence supporting the recommendations ACC and AHA led the development of guidelines for the management
is also considered (A being the highest level with multiple randomized of heart failure.14 CMR received a class IIa level of evidence C recom-
clinical trials or metaanalyses, B with a single randomized trial or non- mendation for evaluation of LV volumes and function as well as for
randomized studies, and C the lowest with only consensus opinions or evaluation of myocardial scar and infiltrative diseases (class IIa, level of
case studies). Stress CMR received a class IIa recommendation (level evidence B). By comparison, ECG received a Ia recommendation, level
of evidence B) for patients who are able to exercise with intermediate- of evidence C, for initial evaluation of the heart failure patient. Any
to-high pretest probability of obstructive CAD and an uninterpretable type of noninvasive evaluation (including CMR) was given a class IIa,
ECG. This was lower than the class I recommendations for exercise level of evidence C, for evaluation of ischemia and/or viability in this
stress echocardiography and stress single-photon emission computed setting, but with no discrimination between the various modalities. In
tomography (SPECT) in the same setting. Similarly, stress CMR was the valvular disease management guidelines published in 2014,15 CMR
classified as class IIa (level of evidence B) for patients in the same receives a class IA, level of evidence B, recommendation for evaluation
category but who are unable to exercise, which was again lower than of patients with moderate to severe aortic regurgitation or primary
the class I recommendation for pharmacologic stress echo and SPECT, mitral regurgitation who have inadequate ECG windows and a class IA,
likely because the latter techniques provide information about the level of evidence C, for evaluation of the aortic sinuses and ascending
patient’s exercise capacity. For patients with known stable ischemic aorta for patients with bicuspid aortic valves and known aortic dilata-
heart disease, CMR received a class IIa recommendation (level of evi- tion. CMR received a class IIb, level of evidence C, recommendation
dence B) for patients who are able to exercise but have an uninterpretable regarding its use in measuring right ventricular volumes in the setting
ECG or for those who are unable to exercise regardless of ECG inter- of severe tricuspid regurgitation. With emerging literature regarding
pretability. CMR received a class I (level of evidence B) recommendation the utility of CMR in many more common conditions, its role in many
in patients with stable ischemic heart disease who are being considered of these guidelines will likely expand in the future.
for revascularization in the setting of known coronary stenosis with CMR receives little mention in some of the ACC documents regard-
unclear physiologic significance. For patients with known stable ische- ing common clinical conditions.15a For example, in the document regard-
mic heart disease and worsening symptoms who are incapable of exercise, ing management of atrial fibrillation,15 CMR was mentioned only in
again CMR was rated IIa (level of evidence B) whereas stress echo and the data supplement as a potential method (with atrial LGE) to assess
SPECT were class I. In those with stable ischemic heart disease and left atrial wall fibrosis. In the 2014 guideline covering management of
silent ischemia who are unable to exercise, have an uninterpretable the patient with NSTEMI, it is only recommended in the patient with
ECG, or were incompletely revascularized, all stress imaging modalities stress (Takotsubo) cardiomyopathy.16 In the 2015 supraventricular
received a class IIa recommendation (level of evidence C). In summary, tachycardia guideline, CMR is only discussed as a method to assess the
CMR was rated lower than stress ECG and SPECT in many of the risk of arrhythmias in repaired tetralogy of Fallot patients.17 In the 2013
clinical scenarios, but this document is several years old and it did state AUC criteria regarding patient selection for ICD or cardiac resynchro-
that more evidence would be generated in regards to the utility of CMR nization therapy (CRT), CMR is discussed as one of several imaging
in these scenarios in the ensuing years. modalities to evaluate LV ejection fraction.18 An exception to this lack
In recent years, the ACC and ACR have partnered to develop two of discussion of CMR in disease-specific guidelines is the 2014 Heart
multimodality AUC documents that concern particular topics within Rhythm Society document on patients with myocardial sarcoidosis,19
cardiovascular medicine. The first such document covered imaging in which recommended CMR as a class IIA indication when a patient
heart failure and was published in 2013.11 This document covered with extracardiac sarcoidosis has symptoms, ECG, or echocardiographic
appropriateness of CMR, and all of the other major imaging modalities, findings suggestive of possible cardiac involvement.
in various clinical scenarios including new onset heart failure, ischemia, CMR indications are covered somewhat more frequently in current
and viability evaluation, evaluation for implantable cardioverter- ESC documents than in ACC and AHA guideline documents but remains
defibrillator (ICD) or biventricular pacemaker placement, and repeat limited.19a In the 2016 ESC guidelines in regards to management of
evaluation of heart failure. CMR was rated as appropriate in many of acute coronary syndromes,20 CMR is discussed as an excellent method
these indications, with the exception of post-ICD or pacer placement for the assessment of function, perfusion, and viability, but it does
because of the relative contraindication of CMR performance, as well not have a particular recommendation for use in given clinical sce-
as repeat evaluation of heart failure. The second ACC/ACR document narios other than making the diagnosis of stress (Takotsubo) cardio-
covering AUC dealt with imaging in the emergency department and myopathy. CMR was given a more prominent role in the guideline
was published in 2015.12 Clinical scenarios covered in this document document regarding the management of patients with sudden cardiac
included suspected ST elevation myocardial infarction (STEMI), chest death.21 For example, CMR or echocardiography was recommended
pain and non-STEMI (NSTEMI), suspected pulmonary embolus, and as an evaluation for family members of patients with sudden cardiac
suspected acute aortic syndrome. Appropriate roles for CMR in the death, and CMR is discussed as an alternative to echo for the evalua-
emergency department were the use of stress CMR in the evaluation tion of ventricular structure and function in patients with high-grade
of the chest pain patient with myocardial infarction excluded, as well ventricular arrhythmias. CMR was recommended in pediatric patients
as in aortic syndromes. In many other scenarios, CMR was rated as with frequent premature ventricular contractions and it received a class
may be or rarely appropriate. IIB recommendation for the evaluation of patients with myocarditis.
584 SECTION VII  Economics and Guidelines

In athletes with suspected structural heart disease, it was given a class patients with hypertrophic cardiomyopathy at their baseline assessment
1C recommendation. if local resources and expertise permit,” class IIa, level of evidence B.
CMR was discussed quite extensively in the 2016 ESC heart failure CMR with LGE received a class IIa, level of evidence C, for evaluation
guidelines.22 It received a recommendation of class I, level of evidence of apical hypertrophic cardiomyopathy and for patients before septal
C, for evaluation of LV structure and function in patients with poor myectomy or alcohol septal ablation. The document suggests repeating
echocardiographic windows and in patients with congenital heart disease. CMR every 5 years in stable disease or every 2 to 3 years in those with
For discrimination of ischemic and dilated cardiomyopathy, CMR progressive disease (class IIb, level of evidence C).
received a recommendation of class IIa, level of evidence C, and a class In summary, CMR is slowly and steadily gaining traction in cardiovascu-
I, level of evidence C, for evaluation of infiltrative cardiomyopathies lar guideline documents. Overall, at the present time, ESC guidelines include
such as amyloidosis, Anderson–Fabry disease, sarcoidosis, noncompac- a greater discussion of the role of CMR than ACC/AHA guidelines.15a,19a
tion, Chagas disease, and hemochromatosis. CMR and other noninvasive As more data become available from ongoing large multicenter clinical
stress modalities were given a class IIb, level of evidence B, recom- trials and registries, and with the ongoing expansion of CMR access, CMR
mendation in the evaluation of ischemia and viability in patients with will undoubtedly have an ever-increasing role in guidelines regarding the
ischemic cardiomyopathy before revascularization. In the 2014 Hyper- management of patients with cardiovascular disease.
trophic Cardiomyopathy guidelines,23 CMR is discussed as a method
to accurately measure increased wall thickness especially when some
segment(s) are unable to be visualized by echocardiography (class IIa,
REFERENCES
level of evidence C). It also states that “CMR should be considered in A full reference list is available online at ExpertConsult.com
CHAPTER 51  Guidelines for Cardiovascular Magnetic Resonance 584.e1

REFERENCES Cardiology Foundation Appropriate Use Criteria Task Force. J Am Coll


Cardiol. 2013;61(21):2207–2231.
1. Kramer CM, Barkhausen J, Flamm SD, Kim RJ, Nagel E. Standardized 12. Rybicki FJ, Udelson JE, Peacock WF, et al. 2015 ACR/ACC/AHA/AATS/
cardiovascular magnetic resonance imaging (CMR) protocols, society for ACEP/ASNC/NASCI/SAEM/SCCT/SCMR/SCPC/SNMMI/STR/STS
cardiovascular magnetic resonance: board of trustees task force on Appropriate Utilization of Cardiovascular Imaging in Emergency
standardized protocols. J Cardiovasc Magn Reson. 2008;10:35. Department Patients With Chest Pain: A Joint Document of the
2. Kramer C, Barkhausen J, Flamm S, et al. Standardized cardiovascular American College of Radiology Appropriateness Criteria Committee and
magnetic resonance (CMR) protocols 2013 update. J Cardiovasc Magn the American College-áof Cardiology Appropriate Use Criteria Task
Reson. 2013;15(1):91. Force. J Am Coll Cardiol. 2016;67(7):853–879.
2a.  Messroghli DR, Moon JC, Ferreira VM, et al. Clinical recommendations 13. Warnes CA, Williams RG, Bashore TM, et al. ACC/AHA 2008 Guidelines
for cardiovascular magnetic resonance mapping of T1, T2, T2* and for the Management of Adults With Congenital Heart Disease: A Report
extracellular volume: a consensus statement by the Society for of the American College of Cardiology/American Heart Association Task
Cardiovascular Magnetic Resonance (SCMR) endorsed by the European Force on Practice Guidelines (Writing Committee to Develop Guidelines
Association for Cardiovascular Imaging (EACVI). J Cardiovasc Magn on the Management of Adults With Congenital Heart Disease) Developed
Reson. 2017;19:75. in Collaboration With the American Society of Echocardiography, Heart
3. Fratz S, Chung T, Greil GF, et al. Guidelines and protocols for Rhythm Society, International Society for Adult Congenital Heart
cardiovascular magnetic resonance in children and adults with congenital Disease, Society for Cardiovascular Angiography and Interventions, and
heart disease: SCMR expert consensus group on congenital heart disease. Society of Thoracic Surgeons. J Am Coll Cardiol. 2008;52(23):e143–e263.
J Cardiovasc Magn Reson. 2013;15(1):1–26. 14. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA Guideline for the
4. Hundley WG, Bluemke D, Bogaert JG, et al. Society for Cardiovascular Management of Heart-áFailure: A Report of the American College of
Magnetic Resonance guidelines for reporting cardiovascular magnetic Cardiology Foundation/American Heart Association Task Force on
resonance examinations. J Cardiovasc Magn Reson. 2009;11:5. Practice Guidelines. J Am Coll Cardiol. 2013;62(16):e147–e239.
5. Schulz-Menger J, von Knobelsdorff-Brenkenhoff F, Bluemke DA, et al. 15. Nishimura RA, Otto CM, Bonow RO, et al. 2014 AHA/ACC Guideline for
Standardized post processing in cardiovascular magnetic resonance. J the Management of Patients With Valvular Heart Disease: Executive
Cardiovasc Magn Reson. 2013;15:35. Summary: A Report of the American College of Cardiology/American
6. Hundley WG, Bluemke DA, Finn JP, et al. ACCF/ACR/AHA/NASCI/ Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol.
SCMR 2010 Expert Consensus Document on Cardiovascular Magnetic 2014;63(22):2438–2488.
Resonance: a report of the American College of Cardiology Foundation 15a.  von Knobelsdorff-Brenkenhoff F, Pilz ZG, Schulz-Menger J.
Task Force on Expert Consensus Documents. J Am Coll Cardiol. Representation of cardiovascular magnetic resonance in the AHA/ACC
2010;55(23):2614–2662. guidelines. J Cardiovasc Magn Reson. 2017;19:70.
7. Pennell DJ, Sechtem UP, Higgins CB, et al. Clinical indications for 16. Amsterdam EA, Wenger NK, Brindis RG, et al. 2014 AHA/ACC Guideline
cardiovascular magnetic resonance (CMR): Consensus Panel report. Eur for the Management of Patients With non ST-Elevation Acute Coronary
Heart J. 2004;25(21):1940–1965. Syndromes: A Report of the American College of Cardiology/American
8. Hendel RC, Patel MR, Kramer CM, et al. ACCF/ACR/SCCT/SCMR/ Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol.
ASNC/NASCI/SCAI/SIR 2006 Appropriateness Criteria for Cardiac 2014;64(24):e139–e228.
Computed Tomography and Cardiac Magnetic Resonance Imaging: A 17. Page RL, Joglar JA, Caldwell MA, et al. 2015 ACC/AHA/HRS Guideline
Report of the American College of Cardiology Foundation Quality for the Management of Adult Patients With Supraventricular Tachycardia:
Strategic Directions Committee Appropriateness Criteria Working Group, A Report of the American College of Cardiology/American Heart
American College of Radiology, Society of Cardiovascular Computed Association Task Force on Clinical Practice Guidelines and the Heart
Tomography, Society for Cardiovascular Magnetic Resonance, American Rhythm Society. J Am Coll Cardiol. 2016;67(13):e27–e115.
Society of Nuclear Cardiology, North American Society for Cardiac 18. Russo AM, Stainback RF, Bailey SR, et al. ACCF/HRS/AHA/ASE/HFSA/
Imaging, Society for Cardiovascular Angiography and Interventions, and SCAI/SCCT/SCMR 2013 Appropriate Use Criteria for Implantable
Society of Interventional Radiology. J Am Coll Cardiol. Cardioverter-Defibrillators and Cardiac Resynchronization Therapy: a
2006;48(7):1475–1497. report of the American College of Cardiology Foundation Appropriate Use
9. Wolk MJ, Bailey SR, Doherty JU, et al. ACCF/AHA/ASE/ASNC/HFSA/ Criteria Task Force, Heart Rhythm Society, American Heart Association,
HRS/SCAI/SCCT/SCMR/STS 2013 Multimodality Appropriate Use American Society of Echocardiography, Heart Failure Society of America,
Criteria for the Detection and Risk Assessment of Stable Ischemic Heart Society for Cardiovascular Angiography and Interventions, Society of
Disease: A Report of the American College of Cardiology Foundation Cardiovascular Computed Tomography, and Society for Cardiovascular
Appropriate Use Criteria Task Force, American Heart Association, Magnetic Resonance. J Am Coll Cardiol. 2013;61(12):1318–1368.
American Society of Echocardiography, American Society of Nuclear 19. Birnie DH, Sauer WH, Bogun F, et al. HRS expert consensus statement
Cardiology, Heart Failure Society of America, Heart Rhythm Society, on the diagnosis and management of arrhythmias associated with cardiac
Society for Cardiovascular Angiography and Interventions, Society of sarcoidosis. Heart Rhythm. 2014;11(7):1304–1323.
Cardiovascular Computed Tomography, Society for Cardiovascular 19a.  von Knobelsdorff-Brenkenhoff F, Schulz-Menger J. Role of
Magnetic Resonance, and Society of Thoracic Surgeons. J Am Coll cardiovascular magnetic resonance in the guidelines of the European
Cardiol. 2014;63(4):380–406. Society of Cardiology. J Cardiovasc Magn Reson. 2016;18:6.
10. Fihn SD, Gardin JM, Abrams J, et al. 2012 ACCF/AHA/ACP/AATS/ 20. Roffi M, Patrono C, Collet JP, et al. 2015 ESC Guidelines for the
PCNA/SCAI/STS Guideline for the Diagnosis and Management of management of acute coronary syndromes in patients presenting without
Patients With Stable Ischemic Heart Disease: A Report of the American persistent ST-segment elevation. Eur Heart J. 2016;37(3):267–315.
College of Cardiology Foundation/American Heart Association Task 21. Priori SG, Blomström-Lundqvist C, Mazzanti A, et al. 2015 ESC
Force on Practice Guidelines, and the American College of Physicians, Guidelines for the management of patients with ventricular arrhythmias
American Association for Thoracic Surgery, Preventive Cardiovascular and the prevention of sudden cardiac death. Eur Heart J.
Nurses Association, Society for Cardiovascular Angiography and 2015;36(41):2793–2867.
Interventions, and Society of Thoracic Surgeons. J Am Coll Cardiol. 22. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the
2012;60(24):e44–e164. diagnosis and treatment of acute and chronic heart failure. Eur Heart J.
11. Patel MR, White RD, Abbara S, et al. 2013 ACCF/ACR/ASE/ASNC/SCCT/ 2016;37(27):2129–2200.
SCMR Appropriate Utilization of Cardiovascular Imaging in Heart 23. Elliott PM, Anastasakis A, Borger MA, et al. 2014 ESC Guidelines on
Failure: A Joint Report of the American College of Radiology diagnosis and management of hypertrophic cardiomyopathy. Eur Heart J.
Appropriateness Criteria Committee and the American College of 2014;35(39):2733–2779.
52 
Noncardiac Pathology
Muhammad Shahzeb Khan, Kiran Khurshid, and Faisal Khosa

Eyes do not see what the mind does not know (Anonymous). characterization of these noncardiac findings can pose a challenge to
the interpreting medical personnel who are reviewing and reporting
Cardiovascular magnetic resonance (CMR) has been increasingly incor- these studies.
porated into clinical practice as a noninvasive method that offers superior
structural and functional assessment of the heart, especially in patients
presenting with complex cardiac pathology, congenital cardiac diseases
WHAT IS AN INCIDENTAL NONCARDIAC FINDING?
and cardiomyopathies. Because CMR is a cross-sectional modality, it Incidental findings are defined as the previously undiagnosed medical
provides complementary information on noncardiac structures adjacent conditions that are discovered unintentionally and may be unrelated
to the heart, including the mediastinum, lungs, chest wall, and upper to the current medical condition which is being investigated or for
abdomen. The acquisition of imaging field data outside the heart offers which tests were being performed.
the opportunity for detection of these noncardiac findings. Location of the noncardiac incidental findings: in case of CMR, the
Indications for CMR: The American College of Cardiology Founda- organs or systems in which incidental findings may be identified include:
tion1 has listed many conditions considered appropriate for CMR. The • Liver and gall bladder
major indications include the following: • Kidneys and adrenal glands
1. Assessment of right ventricular (RV) and left ventricular (LV) systolic • Spleen
function in cases of: • Peritoneum
• Cardiac failure • Lungs and pleura
• Arrhythmias • Breast
• Pulmonary hypertension • Thyroid, esophagus, and vasculature
• Cardiomyopathy • Bones; involving ribs, vertebrae, and sternum
2. Assessment of myocardial perfusion:
• Ischemic heart diseases (angina) CLASSIFICATION OF INCIDENTAL NONCARDIAC
3. Evaluation of cardiac blood flow in the following conditions:
• Valvular heart diseases, for example, aortic regurgitation, mitral
FINDINGS BASED ON CLINICAL SIGNIFICANCE
regurgitation, aortic stenosis, mitral and so on The incidental findings encountered during CMR can be broadly divided
• Shunts: atrial septal defect, ventricular septal defect, patent ductus into the following categories:
arteriosus and so on 1. Clinically important incidental noncardiac findings: comprise find-
4. Inflammatory/neoplastic conditions: ings that require further clinical or radiological workup and/or an
• Pericarditis: constrictive, serous, inflammatory and so on intervention. Not effectively doing so in a timely manner can have
• Cardiac tumors; myxoma, rhabdosarcoma, carcinoids and so on negative consequences for the patient. Examples include:
• Thrombus • Pulmonary nodules
5. Congenital cardiac diseases • Solid/complex lesions of solid abdominal viscera
6. Coronary artery imaging • Malignant breast lumps
7. Assessment of myocardial viability: • Esophageal cancers
• After myocardial infarction • Thyroid masses
The anatomic region, or more accurately, the field of view covered • Aortic pathology including aneurysms or dissection, to name
during a routine CMR extends beyond the conventional boundaries of a few
the heart and associated major vessels to include all the thoracic viscera 2. Moderately important incidental findings: include the findings that
as well as the upper abdominal organs. Hence, it is not uncommon to may or may not have an effect on patient care depending on medical
encounter findings in the thoracic or upper abdominal viscera inci- history or symptomology. In other words, mortality and morbidity is
dentally while conducting a routine CMR. In fact, studies have shown not directly related to the prompt diagnosis and addressing of these
that the prevalence of the noncardiac findings on the routine CMR is findings. Examples include, but are not limited to, the following:
quite high2 and requires in certain cases prompt attention of not only • Liver adenoma or focal nodular hyperplasia
the radiologist but also of the attending physician. • Complex renal and adrenal lesions
An in-depth knowledge of entities affecting different organs in the • Aortic plaques
field of view and a keen eye to detect these noncardiac findings can 3. Clinically unimportant incidental noncardiac findings: these
lead to the reduction in morbidity, mortality in certain cases, and can are the findings that are benign and are inconsequential for the
also add up to screening value. In certain cases the detection and patient. Their presence however sometimes can point to disease

585
586 SECTION VII  Economics and Guidelines

pathology going on elsewhere in the body (e.g., gynecomastia, which Although taking the gender demographics into account the following
can point to a prolactinoma or cirrhotic liver disease). Examples points are noteworthy during the evaluation of incidental noncardiac
include: findings:
• Gynecomastia • Breast diseases, although not exclusive, are prevalent on the CMR
• Fibroadenoma of the breast studies done on women
• Simple cysts found in liver and kidneys • Aortic plaques and atherosclerosis are more common in men after the
age of 45 years, but the incidence is less common in premenopausal
IMPORTANCE OF THE INCIDENTAL women owing to the protective effect of the hormones;3 however,
the incidence becomes equal to men after the menopause
NONCARDIAC FINDINGS Similarly, the work history and social habits can also be incorporated
CMR examinations are not always interpreted by a radiologist. Therefore in the detection of incidental noncardiac findings. For example, the
it is important that the reviewing nonradiology physicians are well prevalence of lung nodules found as an incidental finding can also be
aware of the prevalence of these findings. Appropriate recognition and attributed to the social habit of smoking.
categorization of them by the interpreting physician can prevent mor-
bidity and mortality, and can circumvent unnecessary health care costs PREVALENCE OF INCIDENTAL FINDINGS
caused by delayed diagnosis. On the other hand, pursuing the workup
for the clinically unimportant findings can prove futile and can waste
IN THE LITERATURE
time, energy, and resources. Although the prevalence4–6 of incidental noncardiac findings on other
An important question from this discussion, then, is what are the imaging modalities such as coronary computed tomography (CT) angi-
incidental findings that one should consider while interpreting a CMR; ography, CT colonography, and renal magnetic resonance (MR) angi-
and how to prioritize and incorporate the management of incidental ography has been well described in literature, the prevalence of incidental
noncardiac findings in the final report? findings in CMR has only recently attracted attention. Before 2010,
The answer to both of these questions lies in the consideration of only two studies7,8 in the literature presented original data on prevalence
demographics. The important ones include the age and gender of the of incidental findings on CMR. However, more data has been recently
patient. Taking the age into consideration first, the incidental noncardiac published on this topic. Our literature review found 14 original studies7–20
findings that can be seen in patients over 45 years old include: that have reported the prevalence of incidental noncardiac findings in
• Aortic plaques and atherosclerotic changes CMR. Table 52.1 shows the demographic characteristic of these studies.
• Malignant processes such as lung and esophageal cancers All studies had more than 50% males, with McKenna et al.8 having
• Metastatic bone pathologies a sample population consisting of 96% males. Out of these 14 studies,
In people with an age range of 15 to 45 years, the incidental findings only two studies10,11 were prospective in design, with the remaining
mainly include: retrospective cohorts. Three studies9,15,16 reported the specific number
• Hemangiomas of children included in the sample. The largest cohort was of Chan
• Atelectasis et al,7 followed by Greulich et al.12
• Simple cysts The prevalence of noncardiac pathology differed immensely among
• Nodal diseases: Hodgkin and non-Hodgkin lymphoma these 14 studies. McKenna et al.8 reported an astonishingly high prevalence
In the pediatric population (5–15 years) the main incidental findings of 81%, whereas Chan et al.7 reported a prevalence rate of less than 10%.
to take into account include: The possible reasons explaining such a high rate of incidental findings
• Bone abnormalities: scoliosis in McKenna’s cohort could be their unusual sample population and
• Bone tumors: Ewing sarcoma methodology. All the CMRs were specially reviewed for incidental find-
• Congenital anomalies: diaphragmatic and hiatus hernia, trachea– ings in a sample population who underwent screening CMR for research
esophageal fistula and so on purposes. Thus the numbers from McKenna et al. cohort might not be

TABLE 52.1  Demographic Characteristics of the Studies That Have Reported Prevalence of
Incidental Findings in Cardiovascular Magnetic Resonance
First Author Study Year Study Design Study Duration (Months) Country Males (%) Age (Years)
Dewey 2007 Prospective NS Germany 80 (74%) 63 ± 9
McKenna 2008 Retrospective 8 USA 127 (96%) 74.2
Chan 2009 Retrospective 48 USA 951 (62%) 50 ± 15
Atalay 2010 Retrospective 21 USA 133 (55%) 54 ± 18
Khosa 2010 Retrospective 12 USA 279 (63%) 51 ± 16
May 2012 Retrospective 38 USA 210 (51%) 30
Wyttenbach 2012 Retrospective 36 Switzerland 262 (67%) 53 ± 17
Irwin 2012 Retrospective 13 England NS 54.5
Sohns 2013 Retrospective 40 Germany 542 (63.5%) 58 ± 12
Secchi 2013 Retrospective 6 Italy 192 (64%) 42 ± 22
Roller 2014 Retrospective 17 Germany 223 (55.8%) 52 ± 18
Greulich 2014 Retrospective 8 Germany 695 (64.7%) 58.4 ± 16.4
Dunet 2015 Prospective 25 Switzerland 515 (67.5%) 56 ± 18
Mahani 2016 Retrospective 58 USA 444 (60%) 9.7 ± 6.3
CHAPTER 52  Noncardiac Pathology 587

reflective of the real world clinical practice and should be considered in


the perspective of their study design. Similarly, Khosa et al.14 also reviewed PREVALENCE OF IMPORTANT INCIDENTAL
all clinical CMR specifically for noncardiac pathology. However, Khosa FINDINGS ON CARDIOVASCULAR
et al. reported a relatively lower prevalence (43%) of incidental findings
MAGNETIC RESONANCE
compared with McKenna et al. This could be attributed to the fact that
the Khosa et al. cohort was bigger and included patients undergoing Incidental findings may or may not represent serious underlying disease.
clinically indicated CMR instead of research subjects undergoing screen- Frequently, incidental findings are benign, having no clinical significance.
ing CMR. Studies by Dunet et al.11 and Atalay et al.9 also yielded similar The numbers that are of interest to the physicians are not the prevalence
prevalence to that of Khosa’s cohort. There are four other studies that of noncardiac pathology on CMR but the prevalence of important
reported the prevalence of incidental findings around 20%. incidental findings on CMR because this truly represents a situation
It is evident that the data on prevalence of noncardiac pathology where a patient might benefit from the diagnosis of an incidental finding.
on CMR is diverse. As alluded to earlier, these discrepancies could be Even with important incidental findings on CMR, the statistics are
attributed to many factors such as different patient population, different conflicting, with prevalence ranging from 4.4% to 47.6%.
imaging protocols, CMR reader expertise, different definitions of non- Again the major reason for such diverse data is the different defini-
cardiac pathology, and differences in study design. For example, some tions used by each cohort for labeling significant findings. The definition
studies9,12,18 did not include pathology of the ascending aorta and main of an important or major extracardiac finding has not been standard-
pulmonary trunk as incidental extracardiac findings because these ized, with some authors using “Finding that requires follow up” as the
structures are within the pericardium. Studies that did not include definition, whereas others employ “Finding that needs urgent follow-up
pathology of the great vessels as incidental noncardiac pathology had or treatment.” Some additional heterogeneity and confusion is also
lower prevalence rates. This brings us to the necessity of having a created when some authors include previously known findings among
consensus-reading nomenclature for noncardiac structures in CMR major incidental noncardiac pathology. Another important aspect is
similar to the one that is used in CT angiography. the referral bias. Irwin et al.13 noted that European studies differ con-
Based on the current literature, the overall prevalence of noncardiac siderably from US studies in terms of scan indication and patient
pathology found on clinical CMR referrals is 20% to 50%. We feel that the demographics. The main clinical indication for CMR in UK-based
data published by McKenna et al.8 and Chan et al.7 could be considered studies was ischemic heart disease, whereas for the United States, CMR
as outliers. Table 52.2 shows the results of the original studies, which referrals were for ventricular function and pulmonary vein assessment.
have documented the prevalence of noncardiac pathology on CMR. Hence, it is thought that differences in prevalence might also arise
Recently, Dunet et al.2 conducted a metaanalysis of literature on because of the different nature of local practice.13
incidental CMR findings. They incorporated 127–14,16–19 of the 14 studies Khosa et al.14 showed that 50% of noncardiac pathology was benign,
mentioned earlier in their statistical analysis. The pooled prevalence whereas the remaining 50% comprised the indeterminate and worri-
for all incidental findings was 35% (95% confidence interval [CI]: some, with only 4.6% of the findings labeled as truly worrisome. Similarly,
23%–47%). An important observation was made that pooled prevalence Chan et al., Irwin et al., and Atalay et al. also reported that less than
was higher in studies that were based on image review instead of CMR 10% of the incidental findings are clinically important or worrisome.
report review (45% [95% CI: 27%–62%] vs. 18% [95% CI: 9%–27%], As shown in Table 52.3, there were very few findings that changed the
P < .001). Similarly studies that used only a radiologist as an interpreter course of patient management. Apart from Sohns et al.19 and Dunet
had higher pooled prevalence compared with studies that had employed et al.,11 all studies reporting this outcome had fewer than 10 patients
a radiologist and/or cardiologist (48% [95% CI: 26%–69%] vs. 24% in which extracardiac findings actually changed patient management.
[95% CI: 14%–34%], P < .001). A similar trend was also seen with It is interesting that quite a few studies did not report this important
respect to major extracardiac findings. The pooled prevalence of major outcome. Moreover, hardly any study documented how many patients
incidental findings was calculated to be 12%.2 needed further invasive diagnostic imaging as a result of the incidental

TABLE 52.2  Prevalence of Incidental Findings Documented in Various Studies


CMR With Incidental No. of Incidental No. of Significant
First Author CMR Examinations Findings Findings Findings
Dewey — — 9 2 (22.2%)
McKenna 132 107 (81.1%) 224 25 (11.1%)
Chan 1534 116 (7.6%) 129 8 (6.2%)
Atalay 240 104 (58.3%) 162 16 (9.8%)
Khosa 495 212 (42.8%) 295 14 (4.7%)
May 408 86 (21.1%) 99 36 (36.3%)
Wyttenbach 401 — 250 84 (33.6%)
Irwin 714 154 (21.6%) 180 8 (4.4%)
Sohns 854 223 (26%) 286 49 (17.1%)
Secchi 300 19 (6.3%) 19 5 (26.3%)
Roller 400 25 (6.2%) — 25
Greulich 1074 235 (21.9%) 357 170 (47.6%)
Dunet 762 440 (57.7%) 735 129 (17.6%)
Mahani 849 140 (16.5%) 145 19 (13.1%)

CMR, Cardiovascular magnetic resonance.


588 SECTION VII  Economics and Guidelines

TABLE 52.3  Significant Findings That Changed Treatment


Field Magnetic Significant Findings Significant Findings
First Strength Resonance Image That Changed That Required Further
Author Used (T) or Report Review Interpreter Treatment Invasive Testing
Dewey 1.5 Image Radiologist 2 —
McKenna 1.5 Image Radiologist — —
Chan — Report Radiologist and/or cardiologist 6 3
Atalay 1.5 Image and report Radiologist 9 6
Khosa 1.5 Image Radiologist and/or cardiologist 7 —
May 1.5 Report Radiologist and/or cardiologist — —
Wyttenbach 1.5 Image Radiologist — —
Irwin 1.5 Report Radiologist and/or cardiologist 8 —
Sohns 1.5 Image Radiologist and/or cardiologist 13 8
Secchi 1.5 Image and report Radiologist — —
Roller 1.5 Image Cardiologist — —
Greulich 1.5 Report Radiologist and/or cardiologist 8 —
Dunet 1.5 and 3 Image Radiologist 11 —
Mahani — Report Radiologist — —

findings. A recent metaanalysis2 showed that the combined prevalence However, if the sample was categorized into <60 years and >60 years,
of major noncardiac pathology found on CMR that changed the patient a significant difference was noted not only in the prevalence of incidental
management was only 1% (95% CI: 1%–2%). This value increased to findings (33% vs. 59%, respectively) but also in important noncardiac
2% if anomalies of the great vessels were included. It is important to findings (1% vs. 12%). May et al.16 also commented that the positive
consider that none of the studies had a long-term (>3 years) follow-up correlation of age and frequency of noncardiac pathology was mainly
of patients. Hence, it is difficult to comprehend how much these inci- caused by increasing prevalence of incidental findings in people age
dental findings can impact patient mortality. older than 40 years. The literature suggests there is a sharp increase in
prevalence of incidental findings from the age group 40 to 60 years.
IMPACT OF PATIENT’S GENDER ON Thus it is vital that interpreting clinicians be on the lookout for non-
cardiac pathology, especially in the older population, considering it
INCIDENTAL FINDINGS may have impact on clinical management.
We found four studies7,9,14,17 that investigated the relationship of patient’s
gender with the number of incidental findings. None of the studies CARDIOLOGY AND RADIOLOGY REVIEWING OF
reported any significant correlation between gender and noncardiac
pathology except for Khosa et al.14 This was primarily due to the benign
CARDIOVASCULAR MAGNETIC RESONANCE
finding of gynecomastia in men, which they had considered as an inci- Few studies have reported original data on the impact of a radiologist
dental finding, compared with other studies that had not taken gyne- and cardiologist reading the CMR simultaneously. Khosa et al.14 was
comastia into account. Once gynecomastia was excluded from the analysis, one of the first to highlight this important issue. The results showed
even Khosa et al. found no significant difference in frequency of non- that a combined readout using a concomitant radiologist and cardiolo-
cardiac pathology among men and women (36% vs. 38%, respectively, gist lead to a significantly increased reporting of all noncardiac findings
P = .85). Moreover, Wyttenback et al.17 also explored the possible gender (P < .0001). A combined readout was used in around 80% (384/495)
difference between those who had significant extracardiac findings versus of all CMR performed in the study. The serial readout (independent
those who did not. The analysis revealed no statistically significant interpretation by a cardiologist and radiologist at separate reading ses-
difference (P = .27). Thus it can be implied that CMRs should be thor- sions) resulted in only 15% of all noncardiac findings being reported,
oughly studied for incidental findings in both the genders because they compared with 42% for combined readout. More importantly, this vast
have similar odds of having an important or unimportant extracardiac difference was also present when only the indeterminate and worrisome
pathology. findings were considered (56% for combined readout vs. 33% for serial
readout, P = .03). Hence the data showed that a concurrent readout
IMPACT OF PATIENT’S AGE ON may have an important impact on the detection of possible management-
altering extracardiac findings.
INCIDENTAL FINDINGS Similar results were published by Greulich et al.12 in 2014. Their
All the studies that investigated the link between patient’s age and fre- data showed that a joint approach reported 200 previously unknown
quency of noncardiac pathology found statistically significant trends. noncardiac findings in 138 patients versus 23 noncardiac findings in
Khosa et al.14 showed that the age of those with noncardiac findings 23 patients when the report was read by a cardiologist alone (P < .001).
was more advanced than those without extracardiac findings (56 vs. The trend remained significant (P < .0001), even when only the major
48 years, P < .001). Similar results were published by Chan et al.7 (54 extracardiac findings were considered. In contrast with Khosa et al. and
± 16 years vs. 49 ± 16 years, P < .001) and Wyttenback et al.17 (58.3 ± Greulich et al., Irwin et al.13 found no significant association between
14 years vs. 49.1 ± 17 years; P < .0001). Atalay et al.9 documented an detection of noncardiac pathologies and read-out by a cardiologist or
interesting trend showing that no significant difference existed between a radiologist (P = .38). The reporting rates by a cardiologist and radi-
middle age groups (40–59 years) and younger age groups (<40 years). ologist for noncardiac pathology were found to be 24.3% and 27.5%,
CHAPTER 52  Noncardiac Pathology 589

respectively. Also, for major incidental findings, the reporting rates were challenging to pool the data regarding different CMR sequences and
found to be similar (12.5% for radiologist vs. 13.0% for cardiologist, compare the available evidence.
P = .84).
COST EFFECTIVENESS OF REPORTING INCIDENTAL
IMPACT OF DIFFERENT CARDIOVASCULAR
NONCARDIAC PATHOLOGIES
MAGNETIC RESONANCE SEQUENCES IN
Although the cost effectiveness of CMR in the assessment of coronary
DETECTING INCIDENTAL PATHOLOGY artery disease (CAD) has been studied, to date there is no study that
Many studies have investigated the association between different has investigated the economic impact associated with these findings.
sequences of CMR and detection of incidental findings. McKenna et al.8 Roller et al.18 briefly touched on this topic by suggesting that additional
found no significant difference (P = .369) between the coronal localizer, axial bSSFP chest sequences do not offer any benefit in the visualizing
axial localizer, and short axis multislice sequences in detecting incidental of additional incidental findings and concluded that, in a world of
noncardiac findings. The three sequences were able to visualize 47%, severe time constraints, reviewing these additional sequences for non-
46%, and 41% of all noncardiac pathologies, respectively. Review of cardiac findings is not cost effective when high quality survey images
these three sequences had significantly (P = .013) higher chance of are already generated. However, they did not give any economic or cost
detecting noncardiac findings compared with transmitral flow cine, analysis/evaluation. Furthermore, no data exists on the possible negative
aortic pulsatility cine, and the single slice vertical long axis cine. Their impact of reporting incidental findings on patient’s anxiety and stress
study established that localizer sequences, owing to their larger field of levels. Apart from patient anxiety, such incidental findings can culminate
view, are critical in detecting noncardiac pathology on CMR. into potentially harmful interventions in the pursuit of a diagnosis that
Moreover, the study by Khosa et al.14 showed that among the various does not even exist. Hence, it is imperative that future research is geared
CMR sequences, the following were most revealing of incidental findings: toward characterizing the positive predictive value and specificity to
• Thoracic single-shot fast balanced steady-state free precession (bSSFP) avoid unnecessary financial and emotional implications in this cost-
scout sequences in the sagittal, coronal, and axial planes (field of conscious era.
view 450 × 450) We also feel that increased communication between different physi-
• Axial T1 weighted fast spin echo (FSE) sequence cians is vital to improve the relevance of noncardiac findings. The focus
The authors postulated that 99% of the management-changing incidental should not be on findings that can theoretically change management
noncardiac findings were detected on either one of these two sequences. but on the findings that can practically change management in the
Out of the total of 295 incidental noncardiac findings detected on the specific subset of patient population. The true significance of each
CMRs included in their study, 63% of the findings were picked up on finding can only be gauged when it is put into each individual patient’s
the thoracic bSSFP scout sequence, whereas 60% of the noncardiac clinical context. The interpreting clinician must be provided with impor-
incidental findings were detected by the axial T1-weighted fast spin tant aspects of the patient’s medical history to suspect a truly significant
echo sequence. The other views highlighted in the study included assess- finding. However, this approach of suspecting extracardiac findings
ment of LV and RV systolic functioning using bSSFP long axis, contigu- based on history removes the actual significance of incidental findings,
ous short axis, four-chamber, and LV outflow tract views and velocity which is to detect unsuspected findings. The real question remains how
encoded flow imaging of the proximal aorta and the proximal main wide we should open our eyes to detect these findings and how long
pulmonary artery. should we look for them on every image when a CMR study can have
These sequences served as complimentary in some cases to the bSSFP thousands of images.
and T1-weighted fast spin echo. In some instances, the other CMR Interphysician communication level can also reduce costs by com-
sequences detected the incidental findings that were already known paring incidental noncardiac findings with previous reports and providing
before the study. This study also reviewed the contrast enhanced CMR, clear follow-up recommendations. Moreover, many authors who are
which offered a very limited field of view, mainly of the heart and the in favor of reporting all incidental findings argue that physicians have
major blood vessels, and hence did not play any meaningful role in an ethical obligation and a medicolegal duty to report all pathology in
displaying the noncardiac incidental findings. the field of view. We believe that until cost-benefit analysis studies come
Atalay and his colleagues9 stated that out of the five principal pulse out, the most prudent way to approach the incidental findings will be
sequences in their study, axial stacked bright-blood and dark-blood to provide clear advice in the CMR report. The practice of writing
sequences had the highest sensitivity for displaying incidental noncardiac detailed and specific reports can be the key tool in preventing health
findings (73% and 70%, respectively). The remaining imaging sequences care dollars being spent on needless managements.
yield was less than 25%. On subanalysis of their results, it was found
that despite having lower overall detection rates for incidental findings, INCIDENTAL FINDINGS CLASSIFIED BY
multiplanar viability imaging had the best sensitivity for visualizing
abdominal noncardiac findings. Similarly, it was observed that bright-
BODY SYSTEM
blood methods and axial dark-blood were the most revealing of incidental The thorax and upper abdomen are the primary regions that are included
findings in the thorax. in the CMR field of view and hence where noncardiac pathology is
As shown by other authors, Wyttenbach et al.17 also demonstrated observed. Most of the studies had the highest number of incidental
that coronal, axial, and sagittal scout sequences are the best to detect findings in the thoracic region. Atalay’s9 study yielded a total of 114
nonsignificant and also potentially management-changing extracardiac noncardiac incidental findings limited to the chest territory out of the
findings. Late gadolinium enhancement (LGE) and cine bSSFP images total of 240 findings. The other studies showing remarkable relevance
were the next best with a detection rate of 47% and 34%, respectively. of finding noncardiac incidental pathology limited to the chest included
Because of difference in institutional policy, individual expertise, Khosa et al.,14 with the study yielding 162 chest findings in a total
and preference, there is great heterogeneity between the type and number of 495 cases. Other notable mentions include the studies conducted
of different sequences used by each of the studies. This not only impacts by Wyttenbach et al.,17 Irwin et al.,13 and Greulich et al.12 as shown in
the overall detection rate of noncardiac findings but also makes it Table 52.4.
590 SECTION VII  Economics and Guidelines

TABLE 52.4  Incidental Findings Classified by Body System


Significant
First Total Thorax Significant Thorax Total Abdomen Significant Abdomen Miscellaneous Miscellaneous
Author Findings Findings Findings Findings Findings Findings
Dewey 3 2/3 6 0/6 0 0/0
McKenna 95 15/95 115 9/115 14 1/14
Chan 90 8/90 36 0/36 3 0/3
Atalay 114 12/114 46 3/46 2 1/2
Khosa 162 14/162 66 0/66 67 0/67
May 38 16/38 22 8/22 39 12/39
Wyttenbach 106 56/106 128 21/128 16 7/16
Irwin 110 7/110 38 1/38 32 0/32
Sohns 116 31/116 157 10/157 13 8/13
Secchi — — — — — —
Roller — 21 — 3 — 1
Greulich 246 112/246 67 23/67 44 35/44
Dunet — — — — — —
Mahani 44 6/44 74 13/74 27 0/27

The metaanalysis by Dunet et al.2 revealed that almost half of the


minor extracardiac findings are observed in the thorax. The most
common minor thorax findings were pleural effusion (15.7%) and
parenchymal lung opacities or consolidation (11.8%). The abdomen,
which constituted ~40% of minor incidental findings, was primarily
affected by hepatic and renal cysts. Note that when considering only
the major incidental findings, the thoracic region is by far the most
common region where suspected lesions can be found (thoracic 72%
vs. abdominal 20.2%). In terms of organs, lung parenchyma (15%),
kidney (14.3%), and liver (16.4%) were the most commonly affected
overall. These findings indicate that CMR practitioners should pay close
attention especially to liver and lung parenchyma.
The most important incidental findings in the thorax region are
briefly discussed here:
• Lung nodule: The close proximity of the lungs to the heart, and
large field-of-view images on thoracic scout and axial T1-weighted
spin echo images results in the inclusion of the lung fields in all
CMR studies. A lung nodule is by far the most worrisome noncardiac
incidental finding on CMR. Although the prevalence of lung nodules
on routine CMR is quite low, ignoring or misinterpreting this finding
can culminate in dire consequences. In the study by Atalay et al.,9
5 new cancers (including 2 that were pulmonary) were diagnosed
FIG. 52.1  Axial fat saturation T2-weighted axial spin echo demonstrating
on CMR. In many instances, the lung nodule may be benign requir- a bright lung nodule. Subsequent evaluation determined this to be pul-
ing only periodic follow-up according to guidelines by the Fleischner monary carcinoid.
Society; but in some instances, the lung nodule can show a more
sinister pathology, morphing into lung cancer or metastasis (Fig.
52.1). Lung lesions are termed a lung nodule if the lesion is <3 cm higher in women, especially in the third trimester of pregnancy
or a lung mass if it is >3 cm. Because there is no effective universal or postpartum.
screening available for lung tumors at present, early detection and • Thyroid nodules: The axial scouts and axial T1 spin echo tho-
management of lung lesions detected incidentally on chest imaging racic stack often include images of the thyroid. Very few (4%–6%)
can have an important impact on the morbidity and mortality. Thus, of incidentally identified thyroid nodules are ultimately proven
it is vital that any lung nodule, diagnosed incidentally on CMR, be to represent thyroid malignancies. The vast majority are benign
compared with prior imaging (i.e., CT) or followed-up to ensure colloid nodules. The thyroid malignancy shows bimodal age dis-
stability. In addition, seemingly benign noncardiac lung findings tribution with the peaks occurring in the patients in their 20s and
such as atelectasis, pleural effusion, and air space disease can point 70s. Thyroid cancers have an excellent prognosis if caught early,
to a graver pathology. It is therefore advisable to follow-up these before the development of distant metastasis. Hence, their inciden-
noncardiac findings to resolution. tal finding necessitates follow-up. The studies conducted by May
• Aortic dissection: This is another less prevalent but potentially et al.,16 Greulich et al.,12 and a few others have laid special emphasis
lethal incidental finding requiring urgent diagnosis and treatment in observing the neck region while reporting CMR, to check for
(Fig. 52.2). Moreover, the risk of aortic dissection is significantly thyroid nodules.
CHAPTER 52  Noncardiac Pathology 591

FIG. 52.2  Axial T1-weighted spin echo image demonstrating a descend-


ing thoracic aortic dissection.

FIG. 52.3  Axial T1-weighted postcontrast image demonstrated an


enhancing mediastinal mass representing lymphadenopathy in a patient
• Lymphadenopathy: A relatively common noncardiac finding of the with lymphoma.
mediastinum and the neck region is enlarged lymph nodes. The
study by Khosa et al.14 showed lymphadenopathy in 4% of 495 over-
read CMRs, whereas Chan et al.7 showed lymphadenopathy as an
incidental noncardiac finding in <1% of 1534 of CMR reports.
Greulich et al.12 found 51 (5%) incidental lymph node findings in
1074 CMR studies. The reason why these studies identified nodal
pathology in such large numbers may be because of the simultane-
ous or coreporting of a radiologist and a cardiologist. There is a
multitude of causes of enlarged nodes, ranging from a reactive
lymphadenopathy as a result of infection to a more gruesome malig-
nant pathology. In cases of lymphoma, detecting lymphadenopathy
incidentally on a routine CMR study can help establish early diagnosis
and with proper referral may impact mortality. Fig. 52.3 shows an
enhancing mediastinal mass in a patient with known lymphoma.
• Pulmonary embolism: Few studies have commented on the impor-
tance of pulmonary embolism as an incidental noncardiac CMR
finding (Fig. 52.4). In the Dunet et al.11 cohort, pulmonary embolism
was the most frequent (N = 15, 3.1%) major extracardiac diagnosis.
Eight of those patients were further tested, of which five were con-
firmed for pulmonary embolism. According to the literature2 the
pooled prevalence of pulmonary embolism as a major noncardiac
finding is 1.2%.
• Breast lesions: The breasts are typically displayed in both scout images
and thoracic T1-weighted spin echo images. Breast cancer is the
most common cancer among women and if caught early can have FIG. 52.4  Oblique coronary cardiovascular magnetic resonance angio-
excellent prognosis. Lesions of the breast, especially in women over gram demonstrating a pulmonary embolism in the left main pulmonary
the age of 40 years should always be reported, compared with mam- artery.
mography (if available) and followed-up closely for malignancy. In
younger women, the CMR findings usually represent benign lesions
such as a breast fibroadenoma or fibrocystic changes. In any case, pathology, that is, pituitary tumor (prolactinoma), cirrhosis, or
it is best to further evaluate any breast pathology and to compare adrenal pathology.
with prior imaging (e.g., mammography). • Skeletal lesions: Any incidental bone lesions should be thoroughly
• Gynecomastia: The only study reporting gynecomastia as a noncar- evaluated for metastasis. May et al.16 in their research on pediatric
diac incidental finding was of Khosa et al.14 who reported 41 cases population found cases of syringomyelia as a clinically significant
of gynecomastia (Fig. 52.5). Gynecomastia is usually clinically unim- incidental finding on CMRs performed on patients aged younger
portant. In some cases, it may indicate important undiagnosed than 20 years. The other less prevalent vertebral lesions requiring
592 SECTION VII  Economics and Guidelines

• Hepatic cystic lesions: These are mostly benign liver lesions showing
predilection for the right lobe of the liver. In most cases, they do
not require follow-up or monitoring. The simple cysts are benign
developmental lesions that do not communicate with the biliary
tree and originate from hamartomatous tissue. The cysts can be
solitary or multiple, and seen in 2% to 7% of the population with
no age or gender preference.2 The studies conducted by Chan, Khosa,
and Wyttenbach reported hepatic cysts as noncardiac findings (11.6%,
5.8%, and 14.8%, respectively). Pooled analysis reveals that hepatic
cysts or hemangiomas are responsible for approximately 20% of all
minor noncardiac pathology. In terms of clinically significant find-
ings, hepatic masses and cirrhosis have frequency rates of 11.5%;
other lesions encountered may be adenoma, focal nodular hyperplasia,
and metastatic disease.2
• Gallstones: Widely prevalent in females 40 years of age and older,
gallstones are normally considered clinically unimportant because
they do not require any immediate medical attention. However,
a stone in the gall bladder has the potential to dislodge into the
FIG. 52.5  T1-weighted axial image demonstrating bilateral gynecomastia. common bile duct or pancreatic duct, causing choledocholithiasis
or pancreatitis.
• Hiatal hernia: In most cases, a hiatal hernia is a clinically unimport-
ant congenital or acquired anomaly discovered incidentally on CMR.
attention and follow-up include indeterminate bone lesions, degen- In almost all cases, it does not require any further radiological or
erative disc disease with herniation, and disc prolapse. Mild degrees clinical follow-up. Hiatal hernia represents 5% of all minor non-
of scoliosis usually do not require any follow-up. The study con- cardiac findings, whereas a diaphragmatic hernia constitutes only
ducted by Khosa et al.14 identified a few cases of scoliosis ranging 0.6% of all noncardiac pathology.2
from mild to severe degrees of deformity. Bony deformity of chest
wall (pectus) and adjoining ribs may be congenital or indicative of CONCLUSION: PROS AND CONS OF REPORTING
nutritional deficiency.
• Soft tissue lesions: mostly lipomas, are seen as an insignificant inci-
INCIDENTAL FINDINGS
dental finding in the chest as well in the abdomen. Almost all CMR The documentation and reporting of noncardiac findings on CMR has
studies reported lipomas as an incidental pathology in various loca- been a matter of much debate. Those who are in favor of reviewing
tions. In most cases, lipomas do not require follow-up. CMR images and reporting noncardiac findings claim that appropriate
Although the heart resides in the thorax, scout images and axial follow-up of these incidental findings in certain cases can serve as a
T1-weighted spin echo images frequently include the mid abdomen to screening tool for various diseases. Moreover, fatal diseases, such as
upper-abdomen in the field of view. Frequently seen noncardiac find- lung and breast cancers, can be detected by keen observation of non-
ings in abdomen include: cardiac pathology and patients may (although unproven) benefit from
• Renal cystic lesions: Most renal cysts are simple and prevalent among early interventions.
people of all ages, race, and gender. However, a minority of the cysts Those not in favor of reviewing CMRs for noncardiac pathology
can be complicated representing a more serious underlying pathology, base their argument on the fact that most of the noncardiac findings
such as malignancy or polycystic kidney disease. Complicated cysts are not clinically important or are benign and aggressive evaluation or
may show irregular margins, septations, calcification, or enhance- pursuit of these usually yields futile results and potential morbidity.
ment. Complicated cysts require further clinical follow-up, and in The follow-up can result in unnecessary financial strain on both the
some cases removal. The frequency of renal cysts found in various patient and the health care budget, resulting in a waste of resources. It
studies is similar and constitutes 10% to 20% of all noncardiac also causes false alarm and undue worry among the patients. Finally,
findings. It has been shown that complex renal cysts represent 7% no prospective study has demonstrated any clinical benefit of early
of all important noncardiac pathologies, whereas simple renal cysts detection of noncardiac findings.
contribute to 18% of all minor noncardiac pathology.2 Only a few
studies have commented on the presence of hydronephrosis as an
REFERENCES
incidental noncardiac finding. McKenna et al.8 and Atalay et al.9 all
detected a couple of cases of hydronephrosis. A full reference list is available online at ExpertConsult.com
CHAPTER 52  Noncardiac Pathology 592.e1

REFERENCES 10. Dewey M, Schnapauff D, Teige F, Hamm B. Non-cardiac findings on


coronary computed tomography and magnetic resonance imaging. Eur
1. Douglas PS, Khandheria B, Stainback RF, et al. ACCF/ASE/ACEP/AHA/ Radiol. 2007;17(8):2038–2043.
ASNC/SCAI/SCCT/SCMR 2008 appropriateness criteria for stress 11. Dunet V, Barras H, Boulanger X, et al. Impact of extracardiac findings
echocardiography: a report of the American College of Cardiology during cardiac MR on patient management and outcome. Med Sci Monit.
Foundation Appropriateness Criteria Task Force, American Society of 2015;21:1288–1296.
Echocardiography, American College of Emergency Physicians, American 12. Greulich S, Backes M, Schumm J, et al. Extra cardiac findings in
Heart Association, American Society of Nuclear Cardiology, Society for cardiovascular MR: why cardiologists and radiologists should read
Cardiovascular Angiography and Interventions, Society of Cardiovascular together. Int J Cardiovasc Imaging. 2014;30(3):609–617.
Computed Tomography, and Society for Cardiovascular Magnetic 13. Irwin RB, Newton T, Peebles C, et al. Incidental extra-cardiac findings on
Resonance endorsed by the Heart Rhythm Society and the Society of clinical CMR. Eur Heart J Cardiovasc Imaging. 2013;14(2):158–166.
Critical Care Medicine. J Am Coll Cardiol. 2008;51(11):1127–1147. 14. Khosa F, Romney BP, Costa DN, Rofsky NM, Manning WJ. Prevalence of
2. Dunet V, Schwitter J, Meuli R, Beigelman-Aubry C. Incidental noncardiac findings on clinical cardiovascular MRI. AJR Am J Roentgenol.
extracardiac findings on cardiac MR: systematic review and meta- 2011;196(4):W380–W386.
analysis. J Magn Reson Imaging. 2015 Sep 23. 15. Mahani MG, Morani AC, Lu JC, et al. Non-cardiovascular findings in
3. Jaffer FA, O’Donnell CJ, Larson MG, et al. Age and sex distribution of clinical cardiovascular magnetic resonance imaging in children. Pediatr
subclinical aortic atherosclerosis: a magnetic resonance imaging Radiol. 2016;46(4):473–482.
examination of the Framingham Heart Study. Arterioscler Thromb Vasc 16. May CW, Mansfield WT, Landes AB, Moran AM. Prevalence of
Biol. 2002;22(5):849–854. noncardiac findings in patients undergoing cardiac magnetic resonance
4. Ginnerup Pedersen B, Rosenkilde M, Christiansen TE, Laurberg S. imaging. ScientificWorldJournal. 2012;2012:474582.
Extracolonic findings at computed tomography colonography are a 17. Wyttenbach R, Médioni N, Santini P, Vock P, Szucs-Farkas Z. Extracardiac
challenge. Gut. 2003;52:1744–1747. findings detected by cardiac magnetic resonance imaging. Eur Radiol.
5. Glockner JF. Incidental findings on renal MR angiography. AJR Am J 2012;22(6):1295–1302.
Roentgenol. 2007;189:693–700. 18. Roller FC, Schneider C, Schuhback A, Rolf A, Krombach GA. Cardiac
6. Mueller J, Jeudy J, Poston R, White CS. Cardiac CT angiography after MRI: diagnostic gain of an additional axial SSFP chest sequence for the
coronary bypass surgery: prevalence of incidental findings. AJR Am J detection of potentially significant extracardiac findings in the cardiac
Roentgenol. 2007;189:414–419. MRI examination setting. Rofo. 2014;186:42–46.
7. Chan PG, Smith MP, Hauser TH, et al. Noncardiac pathology on clinical 19. Sohns JM, Schwarz A, Menke J, et al. Prevalence and clinical relevance of
cardiac magnetic resonance imaging. JACC Cardiovasc Imaging. extracardiac findings at cardiac MRI. J Magn Reson Imaging.
2009;2(8):980–986. 2014;39:68–76.
8. McKenna DA, Laxpati M, Colletti PM. The prevalence of incidental 20. Secchi F, Lanza E, Cannaò PM, Petrini M, Sconfienza LM, Sardanelli F.
findings at cardiac MRI. Open Cardiovasc Med J. 2008;2:20–25. Noncardiac findings in clinical cardiac magnetic resonance: prevalence in
9. Atalay MK, Prince EA, Pearson CA, Chang KJ. The prevalence and 300 examinations after blind reassessment. J Comput Assist Tomogr.
clinical significance of noncardiac findings on cardiac MRI. AJR Am J 2013;37(3):382–386.
Roentgenol. 2011;196(4):W387–W393.
APPENDIX A
CMR Screening Form: Beth Israel Deaconess
Medical Center (BIDMC)—CMR Center

&DUGLDF05&HQWHU 
(DVW&DPSXV*U\]PLVK  
7HOHSKRQH±  'DWHRI%LUWKBBBBBBBBBBBB
 )D[±  


'DWHBBBBBBB1DPHBBBBBBBBBBBBBBBBBBBBBBBBBBBBB+HLJKWBBBLQ:HLJKWBBBBBOEV

$Q05,LQYROYHVWKHXVHRIDYHU\VWURQJPDJQHW)RU\RXUVDIHW\WKHSUHVHQFHRIFHUWDLQPHWDOOLFREMHFWVPXVWEH
GHWHUPLQHGEHIRUH\RXHQWHUWKHH[DPURRP3OHDVHSODFHDFKHFNLQWKHDSSURSULDWHFROXPQIRUHDFKLWHPEHORZ

   <HV 1R  <HV 1R

 +DYH\RXKDGDQ05,EHIRUH"   'R\RXKDYHD3RUWDFDWKRU+LFNPDQGHYLFH"  


 
,I\HVGLG\RXUHFHLYHDFRQWUDVWLQMHFWLRQ"   ,I\HVLVLWDFFHVVHG"  

 3DFHPDNHU3DFHUZLUHV,PSODQWDEOHGHILEULOODWRU   $UH\RXRQGLDO\VLV"  


,I\HVKRZRIWHQBBBBBBBBBBBBBBBBBBBBBBBBBBB

 0HWDOOLFKHDUWYDOYHRUDQ\PHWDOOLFVWHQWV   3OHDVHOLVWDOOVXUJHULHV


 ,QWUDFUDQLDORUEUDLQDQHXU\VPFOLSEUDLQVXUJHU\   BBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBB

 %LRRUQHXURVWLPXODWRUHOHFWURQLFGHYLFHRULPSODQW   BBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBB


BBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBB
 7DWWRR V 7DWWRRHGH\HOLQHU   BBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBB
,I\HVORFDWLRQ V BBBBBBBBBBBBBBBBBBBBBBBBBBBBB

 %RG\SLHUFLQJ   3OHDVHFLUFOHLI\RXKDYHDQ\RIWKHIROORZLQJPHGLFDOFRQGLWLRQV


,I\HVORFDWLRQ V BBBBBBBBBBBBBBBBBBBBBBBBBBBB $VWKPD+D\IHYHU+HDUW'LVHDVH0XOWLSOH0\HORPD

 0HWDOLQMXU\WRWKHH\HUHTXLULQJPHGLFDODWWHQWLRQ   7K\URLG'LVHDVH3KHRFKURPRF\WRPD6LFNOH&HOO'LVHDVH

 6KUDSQHOJXQVKRW PHWDOLQERG\    < <


)(0$/(6 (
1
2
0$/(6 (
1
2
 (\HSURVWKHVLVVXUJHU\RQH\H  
6 6
 (DUSURVWKHVLVVXUJHU\RQHDU   3RVVLELOLW\RI   'R\RXKDYHD  

 /LPERUMRLQWUHSODFHPHQWRUSLQQLQJ   SUHJQDQF\" SHQLOHLPSODQW"

 7LVVXHH[SDQGHU HJEUHDVWLPSODQW     


,8' LQWUDXWHULQH ,I\HVPDNHDQGPRGHO
 ,PSODQWHGSXPS LQVXOLQSDLQPHGFKHPRWKHUDS\    GHYLFH RUGLDSKUDJP
 BBBBBBBBBBBBBBBBBBBBBBBBBBB

 $UH\RXZHDULQJDSDWFKWKDWGHOLYHUVPHGLFDWLRQ"   3HVVDU\ LQ   BBBBBBBBBBBBBBBBBBBBBBBBBBB


 'R\RXKDYHDKLVWRU\RIGLIILFXOW,9VWDUWV"   SHOYLV "

3HUVRQDOLWHPVVXFKDVWKHVHVKRXOGEHOHIWLQWKHORFNHUVSURYLGHG
&HOOSKRQH(OHFWURQLFGHYLFHV :DOOHW.H\V :DWFK-HZHOU\ &UHGLWDQG$70FDUGV +HDULQJDLGV 0HWDOKDLUFOLSVSLQVDFFHVVRULHV

593
&DUGLDF05&HQWHU
(DVW &DPSXV*U\]PLVK
7HOHSKRQH  'DWHRI%LUWKBBBBBBBBBBBB
)D[ 

(WKQLF2ULJLQDQG5DFH:HQHHGWKLVLQIRUPDWLRQWRDFFXUDWHO\FDOFXODWH\RXUNLGQH\IXQFWLRQ
‰+LVSDQLFRU/DWLQR‰1RW+LVSDQLFRU/DWLQR
(7+1,&,7< &KHFNRQH
5$&( &KHFNDOOWKDW$SSO\  ‰%ODFNRU$IULFDQ$PHULFDQ
‰:KLWH ‰1DWLYH+DZDLLDQRU2WKHU3DFLILF,VODQGHU
‰$VLDQ ‰$PHULFDQ,QGLDQRU$ODVND1DWLYH
&KR\NHTXHVWLRQQDLUH &KR\NH 3/7HFK8URO± <HV 1R
 +DYH\RXHYHUEHHQWROG\RXKDYHNLGQH\ SUREOHPV"
 +DYH\RXHYHUEHHQWROG\RXKDYHSURWHLQLQ\RXUXULQH"
 +DYH\RXHYHUKDG KLJKEORRGSUHVVXUH"
 'R\RXKDYHGLDEHWHV"
 +DYH\RXHYHUKDG JRXW"
 +DYH\RXHYHUKDGNLGQH\VXUJHU\"

 3OHDVH OLVW EHORZDQ\DOOHUJLHVWRPHGLFDWLRQVIRRGRUODWH[ ‰ 121(5HDFWLRQ

 3OHDVH OLVW EHORZDOOSUHVFULSWLRQDQGRYHUWKHFRXQWHUPHGLFDWLRQV\RXWDNH /DVW'RVH'DWH 7LPH


 
 
 
 
 
 
 
 
 
 
 

 $UH\RXWDNLQJDQ\RIWKHIROORZLQJPHGLFDWLRQV" ‰ 12
‰ 5HYDWLD ‰ $GFLUFD ‰ 9LDJUD‰ /HYLWUD‰ &LDOLV‰ Sildenafil‰ Vardenafil‰ 7DGDODILO /DVW'RVH BBDPSP BBBBBBB
7KHVHGUXJVFDQLQWHUIHUHZLWKFHUWDLQDVSHFWVRIVRPHFDUGLDF05, H[DPLQDWLRQV,I\RXWDNHDQ\ RIWKHVHGUXJVIRUHUHFWLOHG\VIXQFWLRQ\RXVKRXOG
UHIUDLQ IURPWDNLQJWKHVHPHGLFDWLRQVIRUKRXUV GD\V SULRUWR\RXUFDUGLDF05,,I\RXWDNHDQ\RIWKHVHGUXJVIRUSXOPRQDU\K\SHUWHQVLRQ
\RXVKRXOG127 VWRSWDNLQJ\RXUPHGLFDWLRQ EXW0867LQIRUPWKH&DUGLDF05&HQWHUVWDIIEHIRUH\RXUH[DPLQDWLRQ

3DWLHQW6LJQDWXUHBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBB 5HODWLRQVKLS LIQRWWKHSDWLHQW BBBBBBBBBBBB 'DWHBBBBBBB


6LJQDWXUHRI1XUVHRU7HFKQRORJLVWBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBBB 'DWHBBBBBBB
APPENDIX B
CMR Sequence Protocols in Use (2018) at
the Beth Israel Deaconess Medical Center
(BIDMC)—CMR Center

Congenital (Fellow, Gd)a

a
Also see Chapters 40 and 41 for disease-specific congenital protocols as well as Fratz S, Chung T, Greil GF, et al.
Guidelines and protocols for cardiovascular magnetic resonance in children and adults with congenital heart disease:
SCMR expert consensus group on congenital heart disease. J Cardiovasc Magn Reson. 2013;15:51.

Also see Kramer CK, Barkhausen J, Flamm SD, et al. Standardized cardiovascular magnetic resonance (CMR)
protocols 2013 update. J Cardiovasc Magn Reson. 2013;15:91.

595
C APPENDIX

Analogous CMR Terminology Used by


Various Vendors

Sequence Type Philips Siemens GE Hitachi Canon (Toshiba)


Spin echo SE SE SE SE SE
Multispin echo Multi SE Multi echo/MS SE SE Multi echo
Fast spin echo Turbo SE (TSE) Turbo SE (TSE) FSE (Fast SE) FSE (Fast SE) FSE (Fast SE)
Fast spin echo with DRIVE RESTORE FRFSE Driven T2 plus FSE
90-degree flip-back Equilibrium
pulse FSE
3D fast spin echo with VISTA SPACE CUBE isoFSE 3D MVOX
variable flip angle
Ultrafast spin echo (echo Single shot TSE HASTE SS-FSE Single shot FASE/SuperFASE
planar fast spin echo) FSE

Chemical shift fat Fat-sat CHESS Fat-sat


saturation
Combined chemical shift SPIR SPECIAL/SSRF
and inversion recovery
Combined chemical shift SPAIR SPAIR SPAIR
and adiabatic inversion
recovery
Dixon fat suppression mDixon Dixon IDEAL/FLEX fatSep WFOP
Water excitation ProSet WE SSRF PASTA/WET

Inversion recovery IR IR/IRM IR IR IR


Fast inversion recovery IR TSE Turbo IR/TIRM FSE-IR FIR Fast IR
Short T1 inversion STIR STIR STIR STIR STIR
recovery
Long Tau inversion FLAIR/FLAIR TSE FLAIR/Turbo FLAIR/Fast FLAIR FLAIR/Fast FLAIR/Fast FLAIR
recovery FLAIR FLAIR
True inversion recovery Real IR/PSIR Ture IR/PSIR T1 FLAIR/PSIR T1 FLAIR/PSIR PSIR
(phase-sensitive
inversion recovery)
Gradient recalled echo FFE GRE GRE GE Field Echo (FE)
Spoiled gradient echo T1-FFE FLASH SPGR RSSG T1-FFE/RF Spoiled
FE
Ultrafast gradient echo T1-TFE /T2-TFE TurboFLASH Fast GRE/Fast SPGR RGE Fast FE
THRIVE, eTHRIVE VIBE VIBRANT/FAME/LAVA SARGE RADIANCE/QUICK
3D

Ultrafast 3D gradient TFE/3D T1-TFE MP-RAGE 3D FGRE, 3D fast MP-RAGE


echo SPGR
Ultrafast gradient echo IR TFE T1/T2 IR-prepped, DE-SPGR, Fast FE
with magnetization TurboFLASH BRAVO
preparation
T1-weighted gradient THRIVE, eTHRIVE VIBE LAVA, LAVA XV, TIGRE QUICK 3D
echo with fat LAVA FLEX, FAME
suppression

596
APPENDIX C  Analogous CMR Terminology Used by Various Vendors 597

Sequence Type Philips Siemens GE Hitachi Canon (Toshiba)


Coherent gradient echo FFE FISP GRASS Rephased FE
SARGE
Coherent gradient echo T2-FFE PSIF SSFP Time-Reversed SSFP
with echo refocusing SARGE
Balanced gradient echo Balanced FFE TrueFISP FIESTA BASG True SSFP
(BFFE)/BTFE
Coherent balanced CISS FIESTA-C PBSG
gradient echo with dual
excitation
Echo planar EPI EPI EPI EPI EPI
Gradient recalled echo GRASE TGSE Hybrid EPI
plus spin echo (hybrid
echo)
Spoiled gradient echo M-FFE MEDIC MERGE
using combined
multiple free induction
decays
Spin echo black-blood Black-blood Dark-blood Double IR FSE w/
(cardiac) prepulse prepared blood suppression
TSE, HASTE
Spin echo fat-suppressed Black-blood TIRM Double IR FSE with
black-blood (cardiac) prepulse with blood suppression
SPIR or SPAIR
Single shot black blood Single shot TSE HASTE Single shot-FSE Single shot Fast advanced SE
(SSH-TSE)/ (SS-FSE) fast SE (FASE)
Ultrafast spin
echo (UFSE)

Time-of-flight MR Inflow MRA TOF TOF


angiography
Fluoroscopic triggering Bolus TRAK Care Bolus Fluoro trigger FLUTE VISUAL PREP
sequence
Time-resolved 4D-TRAK TWIST TRICKS
contrast-enhanced MR
angiography with
k-space manipulation
Contrast-enhanced MR CE Angio CE-MRA CE-MRA CE-MRA CE-MRA
angiography
Noncontrast angiography TRANCE/B- NATIVE Inhance Inflow IR VASC-ASL Time-SLIP
(inflow balanced SSFP TRANCE TrueFISP (IFIR)
with inversion recovery
saturation)
Contrast-enhanced MR MobiTrak, TimCT SmartStep
with moving table MobiFlex
(bolus-chase)
Real-time interactive scan Interactive CARE iDRIVE
Susceptibility-weighted Venous BOLD SWI SWAN
imaging

Parallel imaging SENSE iPAT ASSET RAPID


technique
Parallel imaging SENSE mSENSE SENSE SENSE SENSE
Parallel imaging with k GRAPPA ARC
space–based algorithm

3D, Three-dimensional; MR, magnetic resonance.


Prepared with the assistance of Patrick Pierce, RT(MR), and Tim Leiner, MD, PhD.

You might also like