Вы находитесь на странице: 1из 343

Clinical Cases in Cardiology

Alessandro Capucci
Editor

Clinical Cases in
Cardiology
A Guide to Learning and Practice

ERRNVPHGLFRVRUJ
Editor
Alessandro Capucci
Cardiology Clinic
Universit Politecnica delle Marche
Ospedali Riuniti Ancona
Ancona
Italy

ISBN 978-3-319-19925-2 ISBN 978-3-319-19926-9 (eBook)


DOI 10.1007/978-3-319-19926-9

Library of Congress Control Number: 2015949122

Springer Cham Heidelberg New York Dordrecht London


Springer International Publishing Switzerland 2015
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made.

Printed on acid-free paper

Springer International Publishing AG Switzerland is part of Springer Science+Business Media


(www.springer.com)
Preface

Knowledge in cardiovascular science has expanded widely in the last decades


and recent years. The proof of this development is the increasing number of
scientific articles published, and the frequent updates of the cardiological
guidelines. Cardiology now comprehends a wealth of subspecialties such as
hemodynamics, electrophysiology, heart failure management, the study of
syncope, and genetics of cardiac diseases, only to mention a few, and each of
these subspecialties counts on devoted specialists who focus their attention
and skillfulness basically on their specific field only. As a result, learning in
the cardiovascular field has become fragmentary, and some diagnostic and
therapeutic procedures artificially expand. In other words, in this day and age
there is an increasing need of a professional who should be able to follow the
patient from the diagnostic to the therapeutic phase, and therefore able to
interpret the role and meaning of every subspecialty approach. The figure I
am referring to is the clinical cardiologist, who should act as an interface
between the patient and the subspecialist, according to patient needs but
maintaining the ability to interpret single exam indications and responses,
and finally managing indications and contraindications for all the specific
therapies. The clinical cardiologist is the one who primarily should take care
of the patient by checking indications and results coming from more special-
ized fields in cardiology.
How is it possible to give adequate academic and professional preparation
to such an important medical figure, when a subspecialist is considered so
important nowadays and usually collects the majority of the resources devoted
to the health system? Specialized scientific articles and texts written for sub-
specialists are difficult learning tools for a cardiologist who is not dedicated
full time to a specific clinical topic, since it is often hard even to follow the
reported concepts. The majority of the medical texts are written for sub-
specialists and contribute to knowledge partition. Moreover, the current trend
of doctors in search for medical information goes towards internet, which on
one hand may help save time because one goes directly to a specific topic, but
on the other, a variety of different facets are missed, which are usually
contained in more encompassing and wide-ranging publications.
In this book we thought of a different approach, mainly aimed at reaching
the majority of professionals in cardiology. With this book we want to address
chiefly young cardiologists, cardiology residents in training, general practi-
tioners, and cardiologists that wish to maintain and improve their clinical
mission. Each chapter is devoted to a specific topic of cardiac diseases and is

v
vi Preface

constructed starting from a clinical case, that is thoroughly discussed, then


the description of the current referred knowledge and the guidelines in use.
One of our most important goals while writing this book is to invite the
readers to think primarily of the patient they have in front of them, when they
have to interpret symptoms and signs. At that stage, a correct and complete
anamnesis is mandatory and turns out to be the real basis for patient framing.
Second, but not less important, the patient should be visited considering all
the semeiotic aspects (not only the cardiological physical exam), then inter-
preting EKG. At that point, our clinical guess should be correctly addressed
and, when needed, we could rely on more specific exams, whose indications,
contraindications, adverse events, and possible interpretations we should
know well.
This book, by presenting to the reader clinical cases often encountered in
the daily practice, has the only goal to describe them and find the correct
clinical approach, always reminding the reader that semeiotic and clinical
evaluation keep a major role in our medical profession and allow an essential
link to the patients health and psychophysical recovery.
That was our intention and we hope we have achieved our goal.

Ancona, Italy Alessandro Capucci, MD


Contents

Part I Acute Ischemic Heart Disease

1 ST Elevation Related to the Site of Coronary


Occlusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Maria Vittoria Matassini and Matilda Shkoza
2 Non-ST-Segment Elevation Myocardial
Infarction (NSTEMI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Marco Marchesini, Marco Morelli, and Luca Piangerelli

Part II Mimicking Ischemic Heart Disease


3 Pericarditis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Alessia Urbinati and Marco Flori
4 Intracranial Bleeding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
Alessia Urbinati and Michela Brambatti
5 Takotsubo Cardiomyopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
Giulia Pongetti and Azzurra Fabbrizioli
6 Early Repolarization Pattern . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
Azzurra Fabbrizioli and Giulia Pongetti

Part III Heart Failure and Resynchronization Therapy


7 Acute Heart Failure and Pulmonary Edema . . . . . . . . . . . . . . . . 65
Andrea Romandini and Simone Maffei
8 Chronic Heart Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
Simona Masiero and Marco Morelli
9 Cardiogenic Shock . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
Maria Vittoria Matassini, Luca Piangerelli,
and Matilda Shkoza
10 Cardiac Resynchronization Therapy
and Possible Dysfunctions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
Laura Cipolletta and Michela Brambatti

vii

ERRNVPHGLFRVRUJ
viii Contents

Part IV Cardiomyopathy
11 Hypertrophic Cardiomyopathy . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Marco Marchesini and Erika Baiocco
12 Cardiac Amyloidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Alessandro Barbarossa and Erika Baiocco
13 Left Ventricular Non-compaction Cardiomyopathy . . . . . . . . . 141
Alessandro Maolo and Simona Masiero
14 Arrhythmogenic Right Ventricular Cardiomyopathy . . . . . . . . 153
Michela Brambatti and Matilda Shkoza

Part V Valvular Heart Disease


15 Aortic Stenosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
Lorena Scappini, Simone Maffei, and Alessio Menditto
16 Mitral Regurgitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
Marco Flori, Lorena Scappini, and Luca Piangerelli
17 Prosthetic Valve Dysfunctions . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
Alessia Urbinati, Marco Marchesini, and Ilaria Mazzanti

Part VI Rhythm Disorders: Tachyarrhytmias


18 Wide QRS Complex Tachycardias . . . . . . . . . . . . . . . . . . . . . . . 201
Alessandro Barbarossa, Daniele Contadini, and Marco Flori
19 Supraventricular Reentrant Tachycardias . . . . . . . . . . . . . . . . . 213
Daniele Contadini and Alessio Menditto
20 Atrial Fibrillation and Related Disorders. . . . . . . . . . . . . . . . . . 227
Jenny Ricciotti and Alessio Menditto
21 Atrial Flutter . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241
Erika Baiocco and Azzurra Fabbrizioli

Part VII Rhythm Disorders: Bradyarrhytmias


22 Sinoatrial Exit Block . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
Andrea Romandini and Lorena Scappini
23 Atrioventricular Blocks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
Andrea Romandini and Marco Morelli

ERRNVPHGLFRVRUJ
Contents ix

Part VIII Channelopathies


24 Brugada Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 277
Alessandro Barbarossa and Giulia Pongetti
25 Long QT Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
Maria Vittoria Matassini and Alessandro Maolo
26 Catecholaminergic Polymorphic Ventricular Tachycardia:
A Challenging Case of Epilepsy . . . . . . . . . . . . . . . . . . . . . . . . 299
Laura Cipolletta

Part IX Miscellanea
27 Sudden Cardiac Death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
Laura Cipolletta
28 The Role of Home Monitoring . . . . . . . . . . . . . . . . . . . . . . . . . . . 325
Laura Cipolletta and Jenny Ricciotti
29 Drug-Related Adverse Events . . . . . . . . . . . . . . . . . . . . . . . . . . . 337
Simone Maffei and Jenny Ricciotti
30 Difficult Interpretation of ECG: Small Clues May Make
the Difference. The Role of the P Wave. . . . . . . . . . . . . . . . . . . . 349
Alessandro Maolo and Daniele Contadini

ERRNVPHGLFRVRUJ
Part I
Acute Ischemic Heart Disease

ERRNVPHGLFRVRUJ
ST Elevation Related to the Site
of Coronary Occlusion 1
Maria Vittoria Matassini and Matilda Shkoza

1.1 Case Report Medical History and Cardiovascular


Risk Factors

Cardiovascular risk factors: systemic arterial


A 44-year-old man was referred to our ICU hypertension and dyslipidemia
at 1:30 AM from the medical first aid due to Family history: no family history of structural
intense, prolonged, non irradiated retroster- heart disease
nal pain, insurgent at 00:30 AM while 2005: stab wound in the midsternal area
sleeping, and profuse cold sweating. The
medical first aid recorded an ECG immedi-
ately, at their arrival, that showed sinus Allergies
rhythm, third-grade AV block, narrow QRS
escape rhythm with a heart rate of 33 bpm, None
ST elevation in the inferior leads (DII, DIII,
aVf) with specular ST depression in DI and
aVl, and QRS axis of 105. Blood pressure Medications
was 90/50 mmHg. Therapy with salicylic
acid 500 mg IV, unfractionated heparin None
4000 UI IV and clopidogrel 300 mg per os
therapy was administered. The patient was
immediately transferred to our ICU for Vital Signs
further evaluation and treatment.
Temperature: 36.5 C
Heart rate: 33 bpm
Arterial blood pressure: 85/50 mmHg
M.V. Matassini (*) M. Shkoza
Dipartimento di cardiologia,
Respiratory rate: 20 breaths/min
Clinica di Cardiologia e Aritmologia, Oxygen saturation: 97 %
Universit Politecnica delle Marche,
Via Conca, 71, Ancona 60126, Italy
e-mail: mavimatassini@yahoo.it;
matilda_shkoza@libero.it

Springer International Publishing Switzerland 2015 3


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_1

ERRNVPHGLFRVRUJ
4 M.V. Matassini and M. Shkoza

Physical Examination were placed, and the cath lab team was advised.
Therapy with crystalloid and dopamine 5 gamma/
General appearance: well developed, well kg/min infusion was practiced with blood pres-
nourished, alert but confused, suffering from sure increase to 120/70 mmHg, and morphine
retrosternal pain, with pallor and cold sweat was administered for the transcutaneous pac-
Lungs: dyspnea; clear to percussion and ing pain. At 1:45 AM the patient was transferred
auscultation without rhonchi, wheezing, or to the cath lab to perform coronary angiography
diminished breath sounds; presence of bibasi- and to position a temporary pacing via the right
lar rales femoral vein. The angiography showed right coro-
Cardiovascular: normal S1 and S2; no S3, S4, nary artery dominance with thrombotic occlusion
or murmurs; regular rhythm; no vascular of its middle tract; no other stenosis was pres-
murmurs ent. At 2:05 AM the patient underwent manual
Abdomen: positive bowel sounds, soft and non- thrombus aspiration (EXPORT) and PTCA with
distended, no guarding or rebound, no masses medicated self-expanding stent (STENTYS DES
3.54.5 17 mm) implantation via the right radial
artery. A bolus of abciximab 0.25 mg/kg was
Routine Laboratory Test administered, and the patient was shifted to prasu-
grel with a loading dose of 60 mg administered in
White blood cells 17,000 mmc, total cholesterol the cath lab. The residual stenosis was <20 % and
338 mg/dl, triglycerides 853 mg/ml, AST 398 TIMI flow was 3. Continuous unfractionated hepa-
U/L, ALT 117 U/L, LDH 689 U/L, GT 60 U/L, rin infusion was administered during the 12 h fol-
troponin I highly specific 116 ng/ml, CKMB lowing the procedure. The transvenous pacing was
222.8 ng/ml, and uric acid 8.4 mg/ml. The removed 48 h later being the patient again in stable
remaining laboratory tests were normal. sinus rhythm. Continuous electrocardiographic
monitoring during the first 6 days did not show any
dangerous tachyarrhythmia or bradyarrhythmia.
Instrumental Examination Therapy with ASA 100 mg od, prasugrel 10 mg
od, atorvastatin 80 mg od, losartan 12.5 mg od,
The ECG at entrance (Fig. 1.1a, b) confirmed the and pantoprazole 40 mg od was started since the
previous one, already described. first day. Metoprolol tartrate 25 mg bid was added
A complete echocardiographic examination the second day. The patient was transferred to our
was performed and showed normal dimensions semi-intensive cardiology unit on the third day and
of the cardiac chambers, reduced systolic left then discharged on the seventh day with a follow-
ventricle function (EF 45 %) due to hypokinesia up visit, ECG, and echocardiography programmed
of the inferior and posterolateral wall, normal 2 months later. Therapy at discharge was ASA
right ventricular function (TAPSE 21 mm), and 100 mg od, prasugrel 10 mg od for 12 months,
mild mitral and tricuspid regurgitation with nor- atorvastatin 80 mg od, losartan 12.5 mg od, panto-
mal pulmonary artery systolic pressure. prazole 40 mg, and metoprolol tartrate 25 mg bid.

Clinical Course and Therapeutic 1.2 ST Elevation Myocardial


Management Infarction (STEMI)

These findings all together were suggestive for Denition and Epidemiology
inferior ST elevation acute coronary syndrome
complicated by third-degree AV block. The last ESC guidelines published in 2012 define
As soon as possible, a transcutaneous pac- acute myocardial infarction (AMI) as the evi-
ing and a continuous electrocardiographic, blood dence of myocardial necrosis (elevation of
pressure, and oxygen saturation monitoring cardiac biomarkers, typical ECG alterations,

ERRNVPHGLFRVRUJ
1 ST Elevation Related to the Site of Coronary Occlusion 5

a b

Fig. 1.1 (a, b) Patient ECG: sinus rhythm, third-grade leads (DII, DIII, aVf) with specular ST-segment depres-
atrioventricular block, narrow QRS escape rhythm with a sion in DI and aVl, QRS axis of 105
heart rate of 33 bpm, ST-segment elevation in the inferior

imaging alterations, or autopsy evidence) in the The in-hospital mortality of unselected STEMI
presence of a clinical setting suggestive for myo- patients varies from 6 to 14 % [8]. Ejection frac-
cardial ischemia [1]. tion, the Killip class, age, time delay to treatment
Acute myocardial infarction with ST elevation and mode of treatment, prior myocardial infarc-
(STEMI) is a clinical syndrome characterized by tion history, renal dysfunction, diabetes mellitus,
the typical symptoms of myocardial ischemia and diseased coronary artery number are all fac-
with electrocardiographic ST elevation (persis- tors that influence mortality. Hospitals with a
tent for more than 20 min) and following release high clinical volume and high rate of invasive
of cardiac biomarkers [2]. procedures have lower mortality rates [11].
Coronary artery disease (CAD) is the most STEMI mortality significantly decreased thanks
common cause of death in the whole world. In to a frequency care increase [5, 7].
2012, 7.4 million people in Europe (which is
13.2 % of all deaths) died from CAD [3].
At present, up to 2540 % of AMI presenta- Pathology and Pathophysiology
tions are STEMI ones [47]. The incidence of
STEMI hospital admissions is different among Pathology
countries that belong to ESC [8]. In the last The causes of STEMI are different, but they can
decades there has been a STEMI incidence be divided into two principal groups:
decrease despite an NSTEMI incidence increase.
In the Sweden registry, which is probably the Coronary atherosclerosis complicated by cor-
most comprehensive STEMI registry, an inci- onary thrombosis, the main one
dence of 66 STEMI/100,000/year, similar to those Non-atherogenic forms that are rare such as
of other countries like the Czech Republic [9], arteritis, trauma to coronary arteries, coronary
Belgium [8], and the USA [10], has been reported. mural thickening with metabolic disease or

ERRNVPHGLFRVRUJ
6 M.V. Matassini and M. Shkoza

intimal proliferative disease, emboli to coro- Anatomically, two major types of MI can be
nary arteries, congenital coronary artery detected: transmural infarcts characterized by the
anomalies, myocardial oxygen demandsup- presence of a full ventricular wall thickness myo-
ply disproportion, hematologic, and miscella- cardial necrosis and nontransmural or subendo-
neous [12] cardial MI with necrosis involvement of the
subendocardium or intramural myocardium or
We will focus our attention on the first cause both. In the first case, there is a completely occlu-
of STEMI, the coronary atherosclerosis. sive thrombus of an epicardial coronary artery
A previous classification based on ECG evolu- that subtends the infarct area with a typical
tion divided patients with MI into two groups: ST-segment elevation. The transmural necrosis
patients with a Q-wave infarction, very often con- can cause a full wall thickness vital myocardial
sidered a transmural infarction, and patients with loss and subsequent fibrosis that is evidenced by
a non-Q-wave infarction. Currently, a new classi- Q-wave evolution in the leads overlying the
fication based on pathophysiology divides infarcted zone. In a few number of patients, there
patients into other two groups: those with STEMI is not a Q-wave evolution but an R-wave height
related to an acute thrombotic occlusion of an epi- reduction.
cardial coronary artery and those with NSTEMI/ There is a specific correlation between the
unstable angina, due to stenosis of a coronary coronary artery occluded, the myocardial area
artery without occlusive thrombi. When there is a developing necrosis, and the ECG derivation that
chronic total occlusion of the coronary artery, shows an ST elevation (Fig. 1.2).
patients do not always have an MI because of col-
lateral blood flow development and other factors.
The most important element on AMIs physio-
pathology is the atherosclerotic plaque. The
plaque evolution is an active process lasting years
that consists in intima lipoprotein accumulation,
lipoprotein oxidation and glycation, intima leu-
kocyte migration, foam cell development, intima
smooth cell migration with consequent extracel-
lular matrix accumulation, atherosclerotic plaque
growth, and a fibrofatty lesion formation with a
lipid core surrounded by an acellular fibrous cap-
sule. Cytokines and effector molecules like hypo-
chlorous acid and superoxide anion play an
important role in this process.
During this natural evolution, high-risk
plaques can undergo plaque disruption [13, 14] Fig. 1.2 Schematic representation of left ventricle
segments, Einthovens triangle with electrocardiogram
that is induced by stressors like intramural blood
derivations. LAD supplies segments nr 1, 2, 6, 7, 8, 12, 13,
pressure, coronary vasomotor tone, and tachycar- 14, 15, 16, and 17; LCX supplies segments nr 4, 5, 6, 10,
dia. So, thrombogenic substances are exposed 11, 12, 15, 16, and 17; RCA supplies segments nr 3, 4, 9,
with secondary activation and aggregation of 10, 11, 14, 15, 16, and 17. Areas of shared perfusion
between LAD, LCX, and RCA are shown in green. The
platelet; moreover, thrombin generation is pro-
infarct artery can be deduced identifying the leads with
moted with subsequent thrombus formation. ST-segment elevation and correlating these with the
There is seasonal and circadian variation of some segments that these leads explore and so with coronary
of these key physiologic variations, and that is arteries that supply these segments. For example, ST
segment elevated prominently in leads exploring seg-
why STEMI happens more frequently in the
ments 1, 2, 7, 8, 13, 14, and 17 which means that the
winter early morning hours and following natural occluded vessel is the LAD. LAD, left anterior descend-
disasters [15]. ing; RCA, right coronary artery; LCX, left circumflex

ERRNVPHGLFRVRUJ
1 ST Elevation Related to the Site of Coronary Occlusion 7

The earliest myocardial ultrastructural typical, and if the myocardial loss is >40 %, car-
changes occurring within the first 20 min are diogenic shock appears.
reversible. Changes become irreversible after Improvement is possible thanks to the recovery
20 min up to 120 min of ischemia [16]. After of the stunned (reversibly injured) myocardium
612 h of necrosis onset, myocardium gross after revascularization, but in some patients the
alteration can be identified. So, in the first 30 min infarcted LV may dilate causing LV remodeling.
after ischemia onset, myocardial injury is revers- This can be a compensatory mechanism restoring
ible; subsequently, a progressive viability loss a normal stroke at the expense of a reduced ejec-
occurs and usually completes at 612 h. That is tion fraction; however, dilation elevates afterload
the reason why the reperfusion therapy benefits (Laplaces law) that depresses LV stroke volume
are greatest when patients are treated early. and increments the consumption of myocardial
Generally, the right ventricle is less involved oxygen, intensifying myocardial ischemia. The
by infarction. It is interested in approximately infarct size, patency of the related coronary artery,
50 % of patients with transmural infarct of the and reninangiotensinaldosterone system
inferoposterior wall and posterior portion of the (RAAS) influence LV remodeling. For this rea-
septum [17]. The right ventricle shows an excel- son, LV remodeling can be reduced by an antago-
lent recovery of systolic function once reperfu- nist of RAAS. Even aldosterone inhibitors reduce
sion is restored [18]. collagen deposition and decrease ventricular
arrhythmia development [20].
Pathophysiology
When a coronary artery is occluded, immediate
myocardial contractile alteration occurs. There Clinical Features
are four sequential patterns of abnormal
contraction: Symptoms
The typical STEMI discomfort is a prolonged
Dyssynchrony (adjacent segments do not con- (more than 20 min), constricting, oppressing, or
tract at the same time) compressing pain of variable intensity. It has a
Hypokinesia (reduced contraction) retrosternal location and often radiates to the ulnar
Akinesia (cessation of contraction) side of the left arm, or rarely both arms; to the
Dyskinesia (the segment expansion is neck, jaw, and shoulders; and rarely to the
paradoxical) epigastrium or interscapular region. In some
patients, frequently those with an inferior STEMI,
As an acute compensation of these alterations, the location is the epigastrium simulating abdomi-
a hyperkinesia of the normal myocardium seg- nal disorders. In these patients nausea and vomit-
ments usually firstly develops. It results from ing may occur due to vagal reflex activation or LV
sympathetic nervous system activity increase receptor stimulation. Symptoms like cold perspi-
and FrankStarling mechanism and lasts up to ration, palpitations, profound weakness, dizziness,
2 weeks. and a sense of imminent death may be present.
If ischemic injury involves >15 % of the myo- In some cases there is an atypical presentation
cardium, the systolicdiastolic function of the LV of STEMI-like atypical location of the pain or
becomes depressed, and a decline of cardiac out- dyspnea, syncope, profound weakness, or acute
put, stroke volume, and blood pressure occurs. indigestion. Some patients are wholly asymp-
End-systolic volume and end-diastolic pressure tomatic, and STEMI can be unrecognized and
increase and diastolic dysfunction appears. The discovered in a subsequent routine electrocardio-
degree of end-systolic volume increase has been graphic examination. These patients have a simi-
shown to be an important predictor of STEMI lar prognosis of symptomatic ones.
mortality [19]. If ischemic injury involves >25 % In up to half of STEMI patients, a precipitat-
of the myocardium, clinical heart failure becomes ing factor like reduced oxygen supply to the

ERRNVPHGLFRVRUJ
8 M.V. Matassini and M. Shkoza

myocardium (hypotension, hypoxemia, pulmo- the first 2 weeks and most commonly on the sec-
nary embolism, etc.) or increased myocardial ond or third day.
oxygen demands (aortic stenosis, fever, agitation,
tachycardia, emotional stress, unusually heavy
exercise) can be identified. Diagnosis

Physical Examination The diagnosis of STEMI starts from symptom


STEMI patients may appear anxious and agi- assessment: history of chest pain lasting at least
tated. Heart rate varies from bradycardia to 20 min or more; not responding to nitroglycerine
tachycardia/tachyarrhythmia, and blood pressure is typical.
varies from hypotension (patient with right ven- The confirmation of diagnosis must be com-
tricle involvement or cardiogenic shock or low pleted as soon as possible with a 12-lead ECG,
blood pressure acute heart failure) to normoten- also considering the addition of posterior (V7
sion and even to hypertension due to adrenergic V8V9) or right leads (V4RV5R, V6R) in
activation. patients with high suspicion, respectively, of pos-
Fever is present in most patients and resolves terior or right ventricle infarction.
within 45 days. It is a nonspecific response to If available, a continuous ECG monitoring
tissue necrosis. should be initiated in all patients to detect life-
Patients with cardiogenic shock or right ventricu- threatening arrhythmias and allow defibrillation
lar infarction infarct have elevated jugular pressure. if required.
Carotid pulse in STEMI patients may be small The diagnostic electrographic sign is a new
due to reduced stroke volume. When LV failure ST-segment elevation measured at the J point in
develops, rales are audible. two contiguous leads with the following thresh-
The Killip classification is a prognostic clas- olds: 0.25 mV in men below the age of 40 years,
sification dividing STEMI patients according to 0.2 mV in men over the age of 40 years, or
the presence and severity of heart failure signs: 0.15 mV in women in leads V2V3 and/or
0.1 mV in other leads [1]. According to leads
Class I: no rales or third sound involved by ST elevation, the localization of the
Class II: rales in <50 % of pulmonary field ischemia is as follows:
with or without third sound
Class III: rales in >50 % of pulmonary field Anterior MI: V1V6
(pulmonary edema) Septal MI: V1V4
Class IV: cardiogenic shock Lateral MI: I, aVL, V5, V6
Inferior MI: II, III, aVF
A third and/or fourth sound may be heard in Posterior MI: V7V8V9 (high R in V1V3
STEMI patients with severe LV dysfunction that with ST depression V1V3)
determines elevation of LV filling pressure. Right ventricle MI: V1, V4RV5RV6R
When the fourth sound is heard, a corresponding
presystolic pulsation is present. Additional sys- Although not frequently seen, an earlier sign
tolic murmur (transient/persistent) due to mitral of ischemia could be the presence of hyperacute
regurgitation as a result of mitral valve apparatus T waves; later, the ECG alterations evolve in ST
dysfunction may be audible. Along the left and elevation in those leads that register the electrical
right sternal border, a holosystolic, prominent activity of the ischemic myocardium. ST eleva-
murmur, accompanied by a thrill, is audible in tion typically presents a concave configuration
the presence of interventricular septum rupture. but over time becomes more pronounced, more
Pericardial friction rubs can be present espe- convex, and rounded upward. In the absence of
cially in patients with large infarctions [21]. reperfusion strategies, the natural evolution of
They are audible along the left sternal border in ECG is as follows: the ST gradually returns to

ERRNVPHGLFRVRUJ
1 ST Elevation Related to the Site of Coronary Occlusion 9

isoelectric baseline, there is a reduction of troponin determination and echocardiography for


R-wave amplitude with the development of Q differential diagnosis and for the assessment of
waves, and T waves become inverted. The ECG the involved myocardium, left ventricular func-
changes usually may take place from few weeks tion, and mechanical complications.
to several hours from presentation.
Moreover, the initial ECG presentation of
acute coronary syndrome could be represented Therapy
by new or presumed new left bundle branch block
(LBBB) [1]. The following recommendations are based on
The electrocardiographic diagnosis could be currently accepted European guidelines [1].
more difficult in some categories of patients: Initial therapy of patients with acute coronary
syndrome with ST elevation is represented as
Patients with preexistent LBBB: in the pres- follows:
ence of intraventricular conduction delay, the
diagnosis could be suspected in the presence of Oxygen administration in the presence of
concordant ST elevation with QRS or in case of hypoxia or acute heart failure.
marked ST abnormalities. Two signs are highly Relief of pain and anxiety: IV opioids are very
specific: Cabreras sign, a prominent notching useful, although they must be used with cau-
in the ascending limb of S wave in leads V3 tion for their potential side effects, such as
V4, and Chapmans sign, a notching in the respiratory depression, nausea, vomiting,
ascending limb of R wave in V5V6 [22]. A hypotension, and bradycardia.
scoring system has been developed from the
GUSTO-1 trial called Sgarbossas criteria [23] The following steps of treatment are related to
but not providing diagnostic certainty. some crucial aspects: the first one is time from
Patients with paced rhythm: in case of clinical symptom onset and the second one, the
strong suspicion, the diagnosis should be con- availability of a primary PCI center.
firmed by angiography; reprogramming of the Patients with a diagnosis of STEMI within
pacemaker with the appearance of intrinsic 12 h from symptom onset should be considered
rhythm and the evaluation of ischemic ECG for mechanical or pharmacological reperfusion
changes may be considered when feasible. strategy, as soon as possible. Moreover, reperfu-
Patients with isolated posterior myocardial sion therapy should be taken into account in the
infarction: the involvement of the inferobasal presence of ongoing ischemia even if the onset of
portion of the heart may appear as an isolated pain dates back more than 12 h. Primary PCI may
ST depression 0.05 mV in leads V1 through also be done in stable patients presenting 1224 h
V3. The documentation of ST elevation after symptom onset.
0.05 mV in the posterior chest wall leads In case of reperfusion strategy, the choice
should be treated as STEMI. between mechanical and pharmacological meth-
Patients with left main coronary obstruction: ods depends on the availability of a primary PCI
the typical ECG signs are aVR ST elevation center with an experienced team or the time
and inferolateral ST depression; the presence needed to reach the PCI center. PCI strategy is
of ST depression in eight or more surface leads preferred to fibrinolysis when there is a primary
together with ST elevation in aVR and/or V1 PCI hospital or when the transfer to a PCI center
suggests ischemia due to multivessel disease could be realized within 120 min from symptom
or left main coronary artery obstruction. onset. In the other cases, fibrinolysis should be
undertaken and followed by consideration of res-
The following steps are not necessary for the cue PCI in case of treatment failure or by consid-
diagnosis; however, they complete the clinical eration of routine angiography in all stable
picture of patients with ACS: blood sampling for patients within 324 h.

ERRNVPHGLFRVRUJ
10 M.V. Matassini and M. Shkoza

In case of mechanical reperfusion, only the cul- guidelines suggest the administration of GP
prit lesion should be treated with PCI and stenting IIb/IIIa inhibitors for bailout therapy if there is
[24]. Primary multivessel revascularization in angiographic evidence of a massive thrombus,
addition to the supposed culprit lesion is indicated slow or no reflow, or thrombotic complication.
in case of cardiogenic shock or persistent isch- 2. Anticoagulant therapy in primary PCI-treated
emia, after the culprit lesion treatment [1, 25, 26]. patients may be achieved with unfractionated
In case of multivessel disease, staged multivessel heparin (UFH), enoxaparin, and bivalirudin,
PCI could be considered as recent meta-analysis with the aim to reduce acute vessel thrombosis
showed improvement in short- and long-term sur- risk. Anticoagulants should be started as soon
vival and reduction of repeated PCI [27]. However, as possible and stopped at the end of PCI pro-
other randomized trials should confirm the bene- cedure, except in the presence of other
fits of staged multivessel PCI in STEMI. conditions that require prolongation of antico-
agulants, such as atrial fibrillation or mechani-
Primary Percutaneous Coronary cal valve or left ventricular thrombosis.
Intervention: Pharmacotherapy Bivalirudin is a direct thrombin inhibitor,
The main stones of therapy in acute coronary nowadays, recommended by European guide-
syndrome are represented by antiplatelet and lines over unfractionated heparin and a GP IIb/
anticoagulant drugs. IIIa blocker (Ib) [1]. As previously reported,
when bivalirudin is preferred, addition of GP
1. Antiplatelet therapy consists of a combination IIb/IIIa blockers does not add adjunctive
of two antiplatelet agents: the first option is benefits, and bivalirudin alone is associated with
aspirin, both oral and IV, if patients are unable lower bleeding rates and reduced mortality [32].
to swallow, and the second one is an adenosine Enoxaparin (with or without routine GP
diphosphate (ADP) receptor blocker. For a long IIb/IIIa blocker) may be preferred over unfrac-
time, clopidogrel has been the only and pre- tionated heparin with a class II of indication,
ferred ADP blocker, while in the last years new level B; finally, unfractionated heparin with or
antiplatelet agents such as prasugrel and ticagre- without routine GP IIb/IIIa blocker must be
lor have been studied [28, 29] and currently used in patients not receiving bivalirudin or
accepted in guidelines and widely used in com- enoxaparin (I, C) [1].
mon clinical practice. Prasugrel and ticagrelor
present a more rapid onset of action and higher Fibrinolysis
efficacy when compared to clopidogrel. Fibrinolysis must be considered when mechani-
Therefore, clopidogrel is preferably used when cal revascularization is not available in recom-
prasugrel or ticagrelor is either not available or mended timelines [1]. Prehospital treatment with
contraindicated. Table 1.1 summarizes the main fibrinolytic drugs by emergency medical person-
features of old and new antiplatelet agents. nel is strongly suggested, when feasible: the ben-
The glycoprotein IIb/IIIa (GP IIb/IIIa) efits from fibrinolytic therapy are higher in the
antagonists are the most recent additions to the first 3 h from symptom onset and then rapidly
antiplatelet agents available that are given decline [33]. The delay from first patient contact
intravenously. Currently, three GPIIb/IIIa and initiation of fibrinolysis should be within
antagonists are available: abciximab, eptifiba- 30 min to improve treatment efficacy, in both
tide, and tirofiban. The role of these agents is hospital and prehospital settings.
debated: in the era of potent oral antiplatelet There are many absolute and relative contrain-
agents, the upstream use is uncertain; more- dications to fibrinolysis, as reported in Table 1.2.
over, if bivalirudin is chosen as an anticoagu- A fibrin-specific agent (tenecteplase, alteplase,
lant, its use does not add further benefits. On reteplase) is recommended (over non-fibrin-
the other side, if UFH or enoxaparin is specific agents).
administered, the association of GPIIb/IIIa Aspirin and clopidogrel must be administered
antagonists remains debatable. Current to treated patients. Parenteral anticoagulation is

ERRNVPHGLFRVRUJ
1 ST Elevation Related to the Site of Coronary Occlusion 11

Table 1.1 Old and new antiplatelet agent for acute coronary syndrome
Clopidogrel Ticagrelor Prasugrel
Trial CURE [30], PCI-CURE PLATO [28] TRITON-TIMI 38 [29]
[31]
Class Thienopyridine Nucleoside analog Thienopyridine
Doses 300600 mg loading 180 mg loading 60 mg loading
dose + 75 mg maintenance dose + 90 mg maintenance dose + 10 mg maintenance
dose dose bis in die dose
Mechanism of action Irreversible inhibition of Reversible and Irreversible inhibition of
P2Y12 subtype of ADP noncompetitive binding of P2Y12 subtype of ADP
receptors on the platelet P2Y12 subtype of ADP receptors on the platelet
surface receptors on the platelet surface
surface
Metabolism Prodrug activated through No metabolism for Prodrug with intestinal,
hepatic metabolism by activation serum, hepatic
cytochrome P450 metabolism (cytochrome
enzymes P450 enzymes)
Genetic variability in
enzyme function affects
drug efficacy
Onset of action Within 2 h Within 30 min Within 30 min
Excretion Renal (50 %), biliary Biliary Renal (68 %), biliary
(46 %) (27 %)
Side effect Dyspnea Increased rate of
Ventricular pauses bleedings
Increased rate of
bleedings
Contraindications Hypersensitivity Hypersensitivity Hypersensitivity
Active bleeding Active bleeding Active bleeding
Significant liver History of intracranial Prior stroke/transient
impairment hemorrhage ischemic attack
Cholestatic jaundice Moderate to severe Moderate to severe
hepatic impairment hepatic impairment
Concomitant use of
strong CYP3A4
inhibitors
Use with caution in Use with caution in Not recommended in
patients at high risk of patients at high risk of patients aged 75 years or
bleeding or with bleeding or with in patients with lower
significant anemia significant anemia body weight (<60 kg)
Use with caution in
patients at high risk of
bleeding or with
significant anemia

recommended until revascularization (if per- ischemia. Also patients with heart failure or car-
formed) or for the duration of hospital stay, from diogenic shock must undergo angiography [1].
at least 48 h up to 8 days. Enoxaparin is preferred
over UHF.
Patients treated with fibrinolysis must be Arrhythmia Management
transferred to a PCI-capable center: stable in Acute Phase
patients should be studied within 324 h. Rescue
PCI is indicated in case of treatment failure Arrhythmias are very frequent in the acute phase
(<50 % ST-segment resolution at 60 min), of STEMI, and every kind of arrhythmia could be
recurrence of ST-segment elevation, or recurrent seen [1].

ERRNVPHGLFRVRUJ
12 M.V. Matassini and M. Shkoza

Table 1.2 Contraindications to fibrinolysis bility or ongoing ischemia, urgent electrical car-
Absolute Previous intracranial hemorrhage, known dioversion is indicated, while pharmacological
cerebrovascular lesion or intracranial cardioversion could be achieved with amioda-
neoplasm, ischemic stroke within the last rone in stable patients with recent arrhythmia
6 months, aortic dissection, recent major
trauma/surgery/head injury, onset. For further details on atrial fibrillation
gastrointestinal bleeding within the past management, see Chap. 20.
month, known bleeding disorder, Ventricular arrhythmias (VA) vary from ven-
noncompressible punctures in the past tricular premature beats to non-sustained and
24 h
sustained ventricular tachycardias (VT) to ven-
Relative Poorly controlled hypertension
(Pas > 180 mmHg or Pad > 110 mmHg), tricular fibrillation. The prevalence of ventricular
transient ischemic attack in the last arrhythmias has been investigated in the fibrino-
6 months, current use of anticoagulant lytic era [35] and, later, in the primary percutane-
therapy, pregnancy or within 1 week
ous coronary intervention era [36]: VA remain
postpartum, advanced liver disease,
infective endocarditis, active peptic ulcer, fairly common even if the real incidence may be
prolonged or traumatic resuscitation underestimated because MI resulting in
prehospital sudden cardiac death could have not
been considered in evaluating studies.
As occurred in our patient, inferior myocar- Ventricular premature beats and non-sustained
dial infarction is frequently associated with VT are really common in the first days from
bradyarrhythmias, from sinus bradycardia to MI. Treatment is not recommended [1] unless non-
atrioventricular (AV) blocks. Sinus bradycardia, sustained VT causes hemodynamic instability.
first-degree AV block, and second-degree AV Sustained monomorphic VT are often not tol-
block type I usually do not require specific treat- erated, especially in the presence of worse left
ment; in case of symptoms or severe hypoten- ventricular dysfunction; moreover, they may pro-
sion, IV atropine should be administered, and if duce ischemia and degenerate in ventricular
not effective, temporary pacing should be fibrillation.
started. All medications interfering with In hemodynamically unstable patients, electri-
electrical conduction should be withheld. cal cardioversion is mandatory. In stable VT,
Second-degree, type II, and third-degree atrio- diagnosis and treatment should be prompt
ventricular blocks usually need temporary pac- because of the risk of rapid deterioration of clini-
ing, especially if concomitant hypotension or cal and hemodynamic conditions. If the patient is
heart failure is present. stable, IV amiodarone, sotalol, or lidocaine could
AV blocks developing during inferior infarc- be attempted, even if conversion rates are low.
tion are usually supra-Hisian and reversible with For polymorphic VT and ventricular fibrilla-
the restoration of coronary perfusion, and they tion, the first therapy consists of immediate defi-
carry a good prognosis. The ECG shows an brillation [37]. If polymorphic VT develop in the
escape rhythm with narrow QRS. In case of ante- setting of bradycardia, a temporary pacing at
rior MI, the presence of an AV block underlines higher rate should be started.
the existence of an extensive necrosis, interesting For further details in the management of ven-
electrical conduction ways; the block is usually tricular arrhythmias and long-term risk evalua-
infra-Hisian with a low escape, wide QRS tion for sudden death, see Chap. 18.
rhythm. A new bundle branch block or a new
hemiblock should be highly monitored because
they often precede a complete AV block. Long-Term Therapies for ST-Segment
Atrial fibrillation is also very common in Elevation Myocardial Infarction
STEMI patients, especially in the presence of
heart failure [34]. Anticoagulation should be Long-term management of patients with acute
started if not contraindicated, and effective rate coronary syndrome with ST elevation consists of
control achieved in order to reduce myocardial lifestyle changes, risk factor control, and long-
oxygen demand. In case of hemodynamic insta- term drug therapy.
ERRNVPHGLFRVRUJ
1 ST Elevation Related to the Site of Coronary Occlusion 13

Lifestyle changes are mainly represented by A, Knuuti J, Lenzen MJ, Mahaffey KW, Valgimigli
M, vant Hof A, Widimsky P, Zahger D (2012) ESC
interruption of smoking, diet and weight control,
Guidelines for the management of acute myocardial
and regular physical activity. Blood pressure infarction in patients presenting with ST-segment
should be regularly controlled with the following elevation. Eur Heart J 33(20):25692619
target: systolic pressure <140 mmHg. 2. OGara PT, Kushner FG, Ascheim DD, Casey DE,
Chung MK, de Lemos JA, Ettinger SM, Fang JC,
If available, exercise-based rehabilitation is
Fesmire FM, Franklin BA, Granger CB, Krumholz
strongly recommended; different studies showed HM, Linderbaum JA, Morrow DA, Newby LK, Ornato
positive effects of cardiac rehabilitation in terms JP, Radford MJ, Tamis-Holland JE, Tommaso CL,
of mortality, reinfarction, and quality of life [38]. Tracy CM, Woo YJ, Zhao DX (2013) ACCF/AHA
guideline for the management of ST-elevation myo-
Rehabilitation also favors achievement of a better
cardial infarction. J Am Coll Cardiol 61(4):e78e140
risk factor control and correct titration of accepted 3. WHO Fact sheet 310. Updated 2012, http://www.
therapy, as beta-blockers. who.int/mediacentre/factsheets/fs310/en/index2.html
Aspirin therapy is indicated indefinitely after 4. Mehta RH, Parsons L, Rao SV et al (2012) Association
of bleeding and in-hospital mortality in black and
STEMI, or if not tolerated, clopidogrel should be
white patients with ST-segment elevation myocar-
used. The combination of dual antiplatelet ther- dial infarction receiving reperfusion. Circulation
apy is recommended for up to 12 months after 125:17271734
STEMI, with a minimum of 1 month for patients 5. Fox KAA, Steg PG, Eagle KA et al (2007) Decline
in rates of death and heart failure in acute coronary
receiving PCI and bare-metal stents and 6 months
syndromes, 19992006. JAMA 297:18921900
for patients receiving PCI and drug-eluting stent. 6. Yeh RW, Sidney S, Chandra M et al (2010) Population
Dual antiplatelet therapy should be maintained trends in the incidence and outcomes of acute myo-
up to 1 year in patients with STEMI who did not cardial infarction. N Engl J Med 362:21552165
7. Mandelzweig L, Battler A, Boyko V et al (2006) The
receive a stent.
second Euro Heart Survey on acute coronary syn-
Oral beta-blockers must be introduced during dromes: characteristics, treatment, and outcome of
hospital stay and continued thereafter in all patients with ACS in Europe and the Mediterranean
patients without contraindications. High-dose Basin in 2004. Eur Heart J 27:22852293
8. Widimsky P, Wijns W, Fajadet J, de Belder M, Knot J,
statins early after admission are indicated in all
Aaberge L, Andrikopoulos G, Baz JA, Betriu A, Claeys
STEMI patients without contraindication or his- M, Danchin N, Djambazov S, Erne P, Hartikainen J,
tory of intolerance, regardless of initial choles- Huber K, Kala P, Klinceva M, Kristensen SD, Ludman
terol concentrations. A target value of <70 mg of P, Ferre JM, Merkely B, Milicic D, Morais J, Noc M,
Opolski G, Ostojic M, Radovanovic D, De Servi S,
LDL cholesterol must be reached, and regularly a
Stenestrand U, Studencan M, Tubaro M, Vasiljevic
lipid profile must be assessed. Z, Weidinger F, Witkowski A, Zeymer U (2010)
Angiotensin-converting enzyme, or if not tol- Reperfusion therapy for ST elevation acute myocar-
erated angiotensin receptor blocker, is indicated dial infarction in Europe: description of the current
situation in 30 countries. Eur Heart J 31:943957
in patients with an impaired ejection fraction
9. Widimsky P, Zelizko M, Jansky P, Tousek F, Holm F,
(EF < 40 %), with heart failure in the early phase, Aschermann M (2007) The incidence, treatment strat-
and with diabetes. However, they may be consid- egies, outcomes of acute coronary syndromes in the
ered in all STEMI patients for their modest effect reperfusion network of different hospital types in
the Czech Republic: results of the Czech evaluation
on mortality.
of acute coronary syndromes in hospitalized patients
Finally, aldosterone antagonists should be (CZECH) registry. Int J Cardiol 119:212219
considered for patients with at least mild left ven- 10. McManus DD, Gore J, Yarzebski J, Spencer F,
tricular dysfunction (EF 40 %) and heart failure Lessard D, Goldberg RJ (2011) Recent trends in the
incidence, treatment, and outcomes of patients with
or for diabetic patients, provided that no renal
STEMI and NSTEMI. Am J Med 124:4047
failure or hyperkalemia is present. 11. Birkhead JS, Weston C, Lowe D (2006) Impact of
specialty of admitting physician and type of hospi-
tal on care and outcome for myocardial infarction
in England and Wales during 20045: observational
References study. BMJ 332:1306
12. Cheitlin MD, McAllister HA, de Castro CM (1975)
1. Steg PG, James SK, Atar D, Badano LP, Blmstrom- Myocardial infarction without atherosclerosis. JAMA
Lundqvist C, Borger MA, Di Mario C, Dickstein 231:951
K, Ducrocq G, Fernandez-Aviles F, Gershlick AH, 13. Achenbach S (2008) Can CT detect the vulnerable
Giannuzzi P, Halvorsen S, Huber K, Juni P, Kastrati coronary plaque? Int J Cardiovasc Imaging 24:311
ERRNVPHGLFRVRUJ
14 M.V. Matassini and M. Shkoza

14. Fox JJ, Strauss HW (2009) One step closer to imaging Manitoba cardiogenic SHOCK Registry investigators.
vulnerable plaque in the coronary arteries. J Nucl Med Catheter Cardiovasc Interv 78:540548
50:497 26. Mylotte D, Morice MC, Eltchaninoff H, Garot J,
15. Manfredini R, Boari B, Salmi R et al (2005) Circadian Louvard Y, Lefvre T, Garot P (2013) Primary percu-
rhythms and reperfusion in patients with acute taneous coronary intervention in patients with acute
ST-segment elevation myocardial infarction. JAMA myocardial infarction, resuscitated cardiac arrest,
294:2846 and cardiogenic shock: the role of primary multi-
16. Schoen FJ (2010) The heart. In: Kumar V, Abbas vessel revascularization. JACC Cardiovasc Interv
AK, Fausto N (eds) Robbins & Cotran pathologic 6(2):115125
basis of disease, 8th edn. WB Saunders, Philadelphia, 27. Bainey KR, Mehta SR, Lai T, Welsh RC (2014)
pp 529587 Complete vs culprit-only revascularization for
17. Hamon M, Agostini D, Le Page O, Riddell JW (2008) patients with multivessel disease undergoing primary
Prognostic impact of right ventricular involvement percutaneous coronary intervention for ST-segment
in patients with acute myocardial infarction: meta- elevation myocardial infarction: a systematic review
analysis. Crit Care Med 36:2023 and meta-analysis. Am Heart J 167(1):114
18. Popescu BA, Antonini-Canterin F, Temporelli PL 28. Wallentin L, Becker RC, Budaj A, Cannon CP,
et al (2005) Right ventricular functional recovery after Emanuelsson H, Held C, Horrow J, Husted S, James
acute myocardial infarction: relation with left ven- S, Katus H, Mahaffey KW, Scirica BM, Skene A,
tricular function and interventricular septum motion. Steg PG, Storey RF, Harrington RA, Freij A, Thorsen
GISSI-3 echo substudy. Heart 91:484 M (2009) Ticagrelor versus clopidogrel in patients
19. Funaro S, La Torre G, Madonna M et al (2009) with acute coronary syndromes. N Engl J Med
Incidence, determinants, and prognostic value of 361:10451057
reverse left ventricular remodelling after primary per- 29. Wiviott SD, Braunwald E, McCabe CH, Montalescot
cutaneous coronary intervention: results of the Acute G, Ruzyllo W, Gottlieb S, Neumann FJ, Ardissino D,
Myocardial Infarction Contrast Imaging (AMICI) De Servi S, Murphy SA, Riesmeyer J, Weerakkody
multicenter study. Eur Heart J 30:566 G, Gibson CM, Antman EM (2007) Prasugrel ver-
20. Konstam MA (2008) Patterns of ventricular remodel- sus clopidogrel in patients with acute coronary syn-
ing after myocardial infarction: clues toward linkage dromes. N Engl J Med 357:20012015
between mechanism and morbidity. JACC Cardiovasc 30. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni
Imaging 1:592 G, Fox KK, Clopidogrel in Unstable Angina to
21. Dorfman TA, Aqel R (2009) Regional pericarditis: a Prevent Recurrent Events Trial Investigators (2001)
review of the pericardial manifestations of acute myo- Effects of clopidogrel in addition to aspirin in patients
cardial infarction. Clin Cardiol 32:115 with acute coronary syndromes without ST-segment
22. Lopes RD, Siha H, Fu Y, Mehta RH, Patel MR, elevation. N Engl J Med 345(7):494502
Armstrong PW, Granger CB (2011) Diagnosing acute 31. Mehta SR, Yusuf S, Peters RJ, Bertrand ME, Lewis
myocardial infarction in patients with left bundle BS, Natarajan MK, Malmberg K, Rupprecht H, Zhao
branch block. Am J Cardiol 108:782788 F, Chrolavicius S, Copland I, Fox KA (2001) Effects
23. Sgarbossa EB, Pinski SL, Barbagelata A, Underwood of pretreatment with clopidogrel and aspirin followed
DA, Gates KB, Topol EJ, Califf RM, Wagner GS by long-term therapy in patients undergoing percuta-
(1996) Electrocardiographic diagnosis of evolving neous coronary intervention: the PCI-CURE study.
acute myocardial infarction in the presence of left Lancet 358:527533
bundle-branch block. GUSTO-1 (Global Utilization 32. Mehran R, Lansky AJ, Witzenbichler B, Guagliumi
of Streptokinase and Tissue Plasminogen Activator G, Peruga JZ, Brodie BR, Dudek D, Kornowski R,
for Occluded Coronary Arteries) investigators. N Hartmann F, Gersh BJ, Pocock SJ, Wong SC, Nikolsky
Engl J Med 334:481487 E, Gambone L, Vandertie L, Parise H, Dangas GD,
24. Kornowski R, Mehran R, Dangas G, Nikolsky Stone GW (2009) Bivalirudin in patients undergoing
E, Assali A, Claessen BE, Gersh BJ, Wong SC, primary angioplasty for acute myocardial infarction
Witzenbichler B, Guagliumi G, Dudek D, Fahy M, (HORIZONS-AMI): 1-year results of a randomised
Lansky AJ, Stone GW (2011) Prognostic impact of controlled trial. Lancet 374:11491159
staged vs. one-time multivessel percutaneous inter- 33. Morrison LJ, Verbeek PR, McDonald AC, Sawadsky
vention in acute myocardial infarction: analysis from BV, Cook DJ (2000) Mortality and pre-hospital
the HORIZONS-AMI (Harmonizing Outcomes with thrombolysis for acute myocardial infarction: a meta-
RevascularIZatiON and stents in Acute Myocardial analysis. JAMA 283:26862692
Infarction) trial. J Am Coll Cardiol 58:704711 34. Schmitt J, Duray G, Gersh BJ, Hohnloser SH (2009)
25. Hussain F, Philipp RK, Ducas RA, Elliott J, Dzavik Atrial fibrillation in acute myocardial infarction: a sys-
V, Jassal DS, Tam JW, Roberts D, Garber PJ, Ducas J tematic review of the incidence, clinical features and
(2011) The ability to achieve complete revasculariza- prognostic implications. Eur Heart J 30:10381045
tion is associated with improved in-hospital survival 35. Newby KH, Thompson T, Stebbins A, Topol EJ,
in cardiogenic shock due to myocardial infarction: Califf RM, Natale A (1998) Sustained ventricular

ERRNVPHGLFRVRUJ
1 ST Elevation Related to the Site of Coronary Occlusion 15

arrhythmias in patients receiving thrombolytic ther- B, Silvers EP, Passman RS, White RD, Hess EP, Tang
apy: incidence and outcomes. The GUSTO investiga- W, Davis D, Sinz E, Morrison LJ (2010) Part 8: adult
tors. Circulation 98(23):2567 advanced cardiovascular life support: 2010 American
36. Mehta RH, Starr AZ, Lopes RD, Hochman JS, Heart Association Guidelines for Cardiopulmonary
Widimsky P, Pieper KS, Armstrong PW, Granger CB, Resuscitation and Emergency Cardiovascular Care.
APEX AMI Investigators (2009) Incidence of and Circulation 122(18 Suppl 3):S729
outcomes associated with ventricular tachycardia and 38. Lawler PR, Filion KB, Eisenberg MJ (2011) Efficacy
fibrillation in patients undergoing primary percutane- of exercise-based cardiac rehabilitation post-
ous coronary intervention. JAMA 301(17):1779 myocardial infarction: a systematic review and meta-
37. Neumar RW, Otto CW, Link MS, Kronick SL, Shuster analysis of randomized controlled trials. Am Heart J
M, Callaway CW, Kudenchuk PJ, Ornato JP, McNAlly 162(4):571584 e572

ERRNVPHGLFRVRUJ
Non-ST-Segment Elevation
Myocardial Infarction (NSTEMI) 2
Marco Marchesini, Marco Morelli,
and Luca Piangerelli

2.1 Case Report Allergies

None
A 66-year-old man with no previous car-
diovascular disease was admitted to the
emergency department for worsening chest Medications
pain and dyspnea. The patient referred the
presence of intermittent chest pain a month Ramipril 5 mg, atorvastatin 20 mg, pantoprazole
ago mainly at rest. No history of fever was 20 mg, and insulin
present.

Vital Signs

Medical History and Cardiovascular Temperature: 36.5 C


Risk Factors Heart rate: 95 bpm
Arterial blood pressure: 150/80 mmHg
Cardiovascular risk factors: type II diabetes Respiratory rate: 15 breaths/min
mellitus, systemic hypertension, and mild Oxygen saturation: 99 %
renal failure
Family history: no family history of structural
heart disease Physical Examination
2013: hospital admission for interstitial pneu-
monia complicated by respiratory failure General: alert, awake, and oriented; restless
Neck: no jugular venous distention, no lymph-
adenopathy, no carotid bruits
Cardiovascular: regular and tachycardic
rhythm, apical soft proto-mesosystolic mur-
mur (2/6 at the Levine scale)
M. Marchesini (*) M. Morelli L. Piangerelli
Clinica di Cardiologia e Aritmologia, Lungs: no rales, rhonchi, or wheezes to aus-
Universit Politecnica delle Marche, cultation, normal percussion
Via Conca, 71, Ancona, Italy Abdomen: no hepatomegaly or splenomegaly,
e-mail: marche9584@gmail.com; moro@abanet.it; no ascites, no masses, normal bowel sounds in
lucapiangerelli30@gmail.com

Springer International Publishing Switzerland 2015 17


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_2

ERRNVPHGLFRVRUJ
18 M. Marchesini et al.

all four quadrants, soft, non-distended/non- anterior hemiblock and ST depression (>0.1 mv)
tender, no rebound or guarding, no costoverte- in V3-6 and DII-avF with ST elevation in aVR
bral angle tenderness (Fig. 2.1).
Extremities: no cyanosis or clubbing, no Echocardiography: moderate concentric
peripheral edema hypertrophy (LV mass/BSA 135 g/m2, relative
wall thickness 0.45) with preserved LV global
function (estimated ejection fraction of 56 %)
Routine Laboratory Tests and hypokinesia of the middle and apical anterior
wall; normal dimension and function of the right
Complete blood count: normal ventricle (TAPSE 20 mm, FAC area 40 %);
Cholesterol (total, HDL, LDL) and TG: total impaired relaxation of the left ventricle
280 mg/dl, LDL 150 mg/dl (E/A < 0.75, E/E < 10); mild mitral and tricuspid
Hepatic function (GOT, GPT, -GT, ALP, regurgitation; systolic pulmonary artery pressure
total bilirubin, amylase, lipase): normal of 35 mmHg
Thyroid function (TSH, FT3, FT4): normal Chest x-ray showed the absence of pulmonary
Renal function (creatinine, BUN): creatinine congestion, lobe consolidation, or bronchograms.
1.5 mg/dl (eGFR 65 ml/min/1.73 mq)
Serum electrolytes: potassium 4.5 mEq/l,
sodium 139 mEq/l Clinical Course and Therapeutic
Biomarkers: troponin I 5.4 ng/ml, BNP 80 pg/ Management
ml, C-reactive protein 0.2 mg/dl, glycosylated
hemoglobin 81 mmol/l Clinical, instrumental, and laboratory data
allowed us to make diagnosis of SCA-NSTEMI:
ECG changes (ST depression >0.05 in more
Instrumental Examination than two contiguous leads), rise in cardiac bio-
marker levels, and normal left ventricle global
An ECG performed at patients admission function with regional hypokinesia no signs of
revealed a right bundle branch block with left myopericarditis.

Fig. 2.1 ECG of patients during chest pain

ERRNVPHGLFRVRUJ
2 Non-ST-Segment Elevation Myocardial Infarction (NSTEMI) 19

According to guidelines, the patient was was performed 5 days later without procedural or
assessed with established risk scores for progno- bleeding complications. The patient was then
sis and bleeding (GRACE 120, intermediate risk; transferred on day 12 to a postsurgery rehabilita-
CRUSADE 30, low risk of bleeding). Antiplatelet tion center with a progressive improvement in
therapy with aspirin and ticagrelor (P2Y12 inhib- functional capacity and subsequently dismissed
itor) with a loading dose of 300 mg and 180 mg, after 7 days. The therapy at discharge was dual-
respectively, and anticoagulant therapy with antiplatelet therapy (aspirin 100 mg and ticagre-
fondaparinux 2.5 mg/die were started. Intravenous lor 90 bid), ramipril 5 mg, atorvastatin 80 mg,
nitrate treatment and beta-blocker therapy (meto- metoprolol 50 mg bid, pantoprazole 20 mg, and
prolol 2.5 mg ev.) were administered due to per- insulin therapy.
sistent angina and tachycardia. ACE inhibitor
(ramipril 5 mg) was continued, and high-dose
statin therapy (atorvastatin 80 mg) was initiated. 2.2 Non-ST-Segment Elevation
The patient remained asymptomatic in the Myocardial Infarction
subsequent hours despite an increase in cardiac (NSTEMI)
biomarkers (troponin I 15 ng/ml) at the labora-
tory analysis (6 h after patient admission). Given Denition and Epidemiology
a GRACE score of 120, an ECG suggesting a left
main or multivessel coronary artery disease (ST Coronary artery disease (CAD) is one of the
depression in many leads with ST elevation in major causes of deaths and morbidity in devel-
avR) and the presence of high-risk criteria (sig- oped countries with a prevalence that increases
nificant rise in troponin) an early invasive strat- with age [1]. Nearly 17.3 million deaths in 2013
egy was performed. Thus, the patient underwent worldwide were related to cardiovascular disease
a coronary angiography (<24 h) that showed [2]. Acute coronary syndrome (ACS) represents
triple-vessel disease with a SYNTAX score >22 one of the most frequent and life-threatening clin-
(Fig. 2.2). ical presentations of CAD and is related to plaque
Considering the clinical status (asymptomatic rupture or an ischemic imbalance between myo-
patient, progressive lowering in cardiac biomark- cardial oxygen supply and demand. The extensive
ers: Tn I 10 ng/ml 12 h after patient admission) use of high-sensitive troponin assay may have led
and the unfavorable coronary anatomy (SYNTAX to more diagnosis of myocardial infarction (MI)
score >22), the patient was sent for coronary hiding a possible reduction in the incidence of MI
artery bypass grafting (CABG, class I A). [3]. According to the most recent guidelines [4],
Ticagrelor was then discontinued, and CABG the term myocardial infarction should be used in

Fig. 2.2 Coronarography showing multivessel disease

ERRNVPHGLFRVRUJ
20 M. Marchesini et al.

the presence of symptoms of ischemia and/or rise or erosion with acute thrombosis or vasoconstric-
in cardiac biomarkers (troponin I or T values tion leading to a sudden and critical reduction in
above the 99th percentile upper reference limit) blood flow. Inflammation plays a determinant role
with the following criteria: ST-segment or T-wave in plaque erosion and subsequent thrombus forma-
changes, imaging evidence of loss of viable myo- tion. Rarely ACS may be caused by other mecha-
cardium or regional motion abnormalities, and nisms such as arteritis, trauma, dissection,
intracoronary thrombus by angiography or congenital abnormalities, or drug abuse [8].
autopsy. Two different clinical presentations may
be encountered based on electrocardiogram
(ECG) findings and pathophysiology: Diagnosis

Myocardial infarction with persistent The diagnosis of NSTE is complex and more dif-
(>20 min) ST elevation (see Chap. 1). ficult than STEMI due to less obvious signs and a
Myocardial infarction without persistent ST wider differential diagnosis. Risk stratification
elevation includes ST-segment depression should be evaluated during the diagnostic phase
>0.05 mV in two contiguous leads, T-wave guiding the revascularization and treatment
inversion >0.1 mV in two contiguous leads, strategy.
pseudo-normalization of T wave, or no ECG
changes [4]. Two types of non-ST-elevation Clinical Presentation
ACS are recognized: NSTEMI or unstable The more frequent symptom of ACS is retroster-
angina (UA) based on the evaluation of car- nal chest pain irradiating to the left arm or to the
diac biomarkers. neck sometimes described as retrosternal pres-
sure or heaviness. Dyspnea, diaphoresis, or nau-
In the last years, it has been observed an sea is often associated. Atypical presentation is
increase in the rate of non-ST-elevation myocar- common in older patients, women, and patients
dial infarction (NSTEMI) in relation to with diabetes and may lead to a missed diagnosis
ST-elevation myocardial infarction (STEMI) [9]. Exacerbation of symptoms during physical
with an annual incidence of ~3 per 1,000 inhabit- exertion and reduction at rest is common. Relief
ants [5]. Hospital mortality is higher between of pain after administration of nitrates is also
STEMI patients, while in the long term death quite specific. UA is characterized by the pres-
rates are consistently larger in NSTEMI [6]. ence of new-onset angina, post-MI angina, or
NSTEMI patients are often older with significant crescendo angina. The presence of risk factors
comorbidities. These data suggest the need of should be carefully evaluated as it significantly
significant efforts both in the acute phase and in increases the probability of CAD diagnosis. The
long-term management to ensure better most common risk factors are male sex, family
outcomes. history of CAD, older age, peripheral artery dis-
ease, diabetes mellitus, renal failure, previous
cardiovascular disease, and dyslipidemia.
Pathophysiology
Physical Examination
Atherosclerosis is a multifocal disease caused by Physical examination shall evaluate the presence
lipid accumulation that affects large-sized and of NSTEMI complication such as heart failure
medium-sized arteries [7]. CAD is a dynamic pro- with pulmonary or systemic congestion and
cess that leads to a progressive reduction in the ves- establish the presence of precipitating factors
sel size due to plaque formation. NSTEMI is due to (i.e., anemia). Extracardiac (pneumonia, pneu-
complete or partial occlusion of a coronary artery mothorax, costochondritis) and nonischemic
or a mismatch between oxygen supply and demand. (valvular disease, pericarditis) causes of chest
ACS is generally precipitated by a plaque rupture pain may also be excluded.

ERRNVPHGLFRVRUJ
2 Non-ST-Segment Elevation Myocardial Infarction (NSTEMI) 21

Electrocardiogram Table 2.1 Possible causes of troponin rise


A 12-lead resting ECG must be obtained in Chronic or acute renal failure
10 min after the first medical contact. Additional Congestive heart failure
ECG must be obtained if the patient presents Hypertensive crisis
symptoms at (3 h) 6-9-24 h and immediately dur- Arrhythmias
ing symptoms [8]. Comparison with previous Pulmonary embolism
ECG recordings is recommended especially in Myocarditis
patients with known ST alterations (i.e., left ven- Stroke or subarachnoidal hemorrhage
tricular hypertrophy). A completely normal ECG Aortic dissection
does not exclude the presence of ACS. Typical Cardiac contusion, ablation, cardioversion
ECG findings are ST-segment depression or Takotsubo cardiomyopathy
T-wave inversion in at least two contiguous leads. Infiltrative disease (i.e., amyloidosis)
Drug toxicity
Cardiac Enzymes Sepsis or respiratory failure
Cardiac troponins are the reference markers of Rhabdomyolysis
MI because they are more specific and sensitive
than other markers [10]. The initial rise in tropo- phy (CT) permits visualization of coronary
nins occurs in approximately 4 h and may remain arteries and may be useful in excluding other
elevated up to 2 weeks. The diagnostic cutoff is a causes of chest pain such as aortic dissection or
value exceeding the 99th percentile of normal pulmonary embolism. Different studies reported
reference population with an assay with an high negative predictive value of this technique in
imprecision of <10 % [4]. High-sensitivity assays intermediate-risk patients [11].
have been introduced with higher sensitivity and
specificity. A single normal test is not sufficient Invasive Imaging
to exclude ACS in the presence of suggestive Coronary angiography remains the gold standard
symptoms and may be repeated. Relevant as it provides relevant diagnostic informations.
changes in troponin levels are also important Timing of angiography should be evaluated on
because they allow to make differential diagnosis the basis of risk assessment. It is recommended
between a relevant number of possible non-ACS- to prefer radial approach when feasible because it
related troponin elevations (Table 2.1). has lower risk of hematomas and bleeding [12].
Angiograms should be obtained after and before
Imaging the use of nitrates to exclude the vasoconstriction
due to ACS. In ambiguous lesions, intravascular
Noninvasive Imaging ultrasound (IVUS) of fractional flow reserve
Echocardiography is the tool of choice due to its (FFR) may help in the correct evaluation of ste-
availability and feasibility. Left and right ventric- nosis and treatment strategy.
ular systolic function may be assessed and repre-
sents a relevant prognostic factor. New-onset
regional wall hypokinesia or akinesia is also a Risk Stratication and Treatment
typical finding that suggests myocardial isch- Strategy
emia. Echocardiography may also exclude other
causes of chest pain such as aortic dissection, To date two alternative approaches have been
pulmonary embolism, aortic stenosis, pericardi- validated for the treatment of acute coronary syn-
tis, or hypertrophic cardiomyopathy. dromes (ACS) without ST-segment elevation
Stress echocardiography and nuclear myocar- (NSTE-ACS), such as unstable angina (UA) and
dial perfusion are II level tests that may lead to acute myocardial infarction without ST elevation
myocardial ischemia diagnosis especially in low- (NSTEMI). The two strategies differ in the tim-
risk patients. Multidetector computed tomogra- ing for cardiac catheterization.

ERRNVPHGLFRVRUJ
22 M. Marchesini et al.

The first option is the early invasive strategy, level of evidence A). Among all other patients
meaning that the patient is quickly sent to the with UA/NSTEMI, those who have at least one
cath lab for coronary angiography, followed by secondary high-risk criterion or recurrent symp-
the eventual PCI or surgical revascularization toms should be treated with an invasive strategy
based on the angiographic results. within 72 h (class I, level of evidence A).
The second option, called conservative strat-
egy, consists in medical therapy alone for the ini-
tial treatment, reserving cardiac catheterization Primary high-risk criteria:
only to those who have recurrent ischemia or (a) Relevant increase and fall of
other high-risk features. troponin
(b) Dynamic changes of the ST seg-
Conservative Strategy Versus Invasive ment and/or T wave
Strategy Secondary high-risk criteria:
Currently, the results of these alternative strate- (a) Diabetes mellitus
gies have been analyzed in several randomized (b) Renal impairment (eGFR <60 mL/
trials. A meta-analysis in which seven studies min/1.73 m2)
were included showed a significant benefit (c) Reduced left ventricular ejection
offered by the early invasive strategy in terms of fraction (<40 %)
reduction of 2-year mortality from all causes and (d) Early postinfarction angina
myocardial infarction (MI), with no increase in (e) Recent angioplasty (PCI)
adverse periprocedural events. Patients who have (f) Previous bypass surgery (CABG)
most benefited from an early invasive strategy (g) Intermediate- to high-GRACE-risk
were those with abnormal troponin values and score (108140)
high-risk features.
These results have been translated into the
European Society of Cardiology (ESC) NSTEMI
Guidelines published in 2011 that recommend a From this, it is clear that by definition NSTEMI
routine invasive strategy in almost the totality of ACS with elevation of myocardial necrosis mark-
patients with NSTEMI while highlighting the ers should be treated with an invasive strategy
crucial role of risk stratification. within 72 h, since the elevation of troponin alone
is considered a primary high-risk criterion.
NSTEMI Patients: Indications Therefore, a conservative strategy is now
in the Light of the Most Recent restricted to a narrow subgroup of patients: basi-
Guidelines cally not to those who have a NSTEMI, but only
Although, for ethics and safety reasons, patients to patients with unstable angina (then with nega-
with UA/NSTEMI at very high risk (refractory tive troponin) and without recurrent symptoms.
angina, severe heart failure, electrical and/or Moreover, even among patients with UA, only
hemodynamic instability) were excluded from those without high-risk characteristics should be
randomized clinical trials, it is universally approached with a conservative strategy, that is,
accepted that these patients must be sent within nondiabetics, those with normal global contrac-
2 h to the cath lab for an urgent invasive strategy, tile function of the left ventricle, those who have
regardless of risk stratification and the values of not been subjected in the past to PCI or CABG,
troponin (class I, level of evidence C). and those with a GRACE risk score <108. In this
Based on randomized clinical trials, patients small group of patients (low risk and without
at high risk who have a GRACE score >140 or recurrent symptoms), a noninvasive assessment
with at least one of the primary high-risk criteria of inducible ischemia is recommended (class I,
must be treated with an early invasive strategy, level of evidence A), which must be performed
within 24 h from the first medical contact (class I, before hospital discharge. Coronary angiography

ERRNVPHGLFRVRUJ
2 Non-ST-Segment Elevation Myocardial Infarction (NSTEMI) 23

should be performed only if the results of nonin- revascularization at both 1 month and 1 year.
vasive tests are positive for inducible ischemia. However, only 43 % of CABG patients could be
matched, and there was a strong trend for a higher
Revascularization Strategies rate of major adverse cardiac events (MACE) at
In approximately one-third of patients, angiogra- 1 year with PCI, compared with CABG (25.0 %
phy will reveal single-vessel disease, allowing ad vs. 19.5 %, respectively; p 0.05).
hoc PCI of the culprit lesion in most cases. In a Culprit-lesion PCI does not necessarily require
multivessel disease, the choice has to be made a case-by-case review by the Heart Team, when
between culprit-lesion PCI, multivessel PCI, the procedure needs to be performed ad hoc after
CABG, and a combined (hybrid) revasculariza- diagnostic angiography particularly in case of con-
tion [13, 14]. The distribution of PCI versus tinuing or recurrent ischemia, hemodynamic insta-
CABG in patients with multivessel disease suit- bility, pulmonary edema, recurrent ventricular
able for revascularization is approximately 80 % arrhythmias, or total occlusion of the culprit coro-
versus 20 % [15]. The revascularization strategy nary artery requiring urgent revascularization. For
in patients with multivessel CAD should be all other scenarios, revascularization should be
determined early by the Heart Team and based on discussed in a multidisciplinary setting. After cul-
the patients clinical status, as well as the severity prit-lesion PCI, patients should be discussed by
and distribution of the CAD and the characteris- the Heart Team, in the context of functional evalu-
tics of the lesion. The SYNTAX score has proven ation of the remaining lesions, patients comor-
to be strongly predictive of death, myocardial bidities, and individual characteristics.
infarction, and TVR [16]. Culprit-lesion PCI is
usually the first choice in most patients with Medical Therapy: The Importance
NSTE-ACS and multivessel disease; however, of Balancing the Ischemic
there are no prospective studies comparing and Hemorrhagic Risk
culprit-lesion PCI with early CABG. In stabi- The therapeutic management of NSTEMI
lized patients with multivessel disease and a high includes the use of anti-ischemic and antithrom-
SYNTAX score (>22), particularly when there is botic drugs in combination to coronary revascu-
no clearly identified culprit lesion, a strategy of larization. The timing and intensity of these
urgent CABG should be preferred. The strategy therapeutic interventions must be individualized
of multivessel PCI rather than culprit-lesion PCI for each patient in light of both the ischemic and
has not been evaluated in an appropriate random- the bleeding risk, since most of the antithrom-
ized clinical trial. In a large database including botic therapies increase the risk of bleeding. Risk
105,866 multivessel CAD patients with NSTE- assessment is an ongoing process that must begin
ACS, multivessel PCI was compared with single- with the first medical contact, until discharge
vessel PCI and was associated with lower from the hospital, because it can change the ther-
procedural success but similar in-hospital mortal- apeutic strategy at anytime.
ity and morbidity [17]. Complete revasculariza- Among the main ischemic risk factors, age and
tion at the time of the index procedure did not the presence of other comorbidity (anemia, diabe-
result in lower mortality rates over 3 years, as tes, etc.) have the greatest prognostic impact.
compared with a staged procedure strategy. Other factors consist in the characteristics and
However, incomplete revascularization appears mode of onset of pain (being worse angina at rest
to be associated with more 1-year adverse event and/or relapsing) and hemodynamic conditions.
rates. In the ACUITY trial, CABG was compared Other prognostic factors recognized and to which
with PCI among patients with multivessel disease attention should be paid are the electrocardio-
[15]. PCI-treated patients had lower rates of graphic changes and the elevation of a series of
stroke, myocardial infarction, bleedings, and biomarkers such as troponin, C-reactive protein
renal injury and similar 1-month and 1-year mor- (CRP), and the N-terminal fragment of the atrial
tality but significantly higher rates of unplanned natriuretic peptide (NT-proBNP).

ERRNVPHGLFRVRUJ
24 M. Marchesini et al.

From the integration of all these risk factors, Morphine 35 mg intravenously or subcutane-
several risk scores have been developed, among ously, if severe pain.
which the best known are the TIMI risk score and Patients who are taking chronic beta-blocker
the Global Registry of Acute Coronary Events therapy, admitted with ACS, should be contin-
(GRACE) score. The TIMI risk score is the most ued on beta-blocker therapy if not in Killip
used in the past due to its simplicity, but its dis- class III (I; C).
criminating power is significantly lower than that Oral beta-blocker treatment is indicated in all
of the GRACE score, basically because it does patients with LV dysfunction without contra-
not take into account parameters of crucial indications (I; B).
importance as hemodynamics. In contrast, the Calcium channel blockers are recommended
GRACE score includes the heart rate (HR), blood for symptom relief in patients already receiv-
pressure (BP), and the presence of heart failure, ing nitrates and beta-blockers (dihydropyri-
in addition to classical risk factors such as age, dine type) and in patients with
any prior MI or previous PCI, creatinine values, contraindications to beta-blockers (benzothi-
myocardial-specific enzymes, and ST changes on azepine or phenylethylamine type) (I; B).
ECG. Therefore, it provides a more accurate esti- Calcium channel blockers are recommended
mate of the risk both on admission and at dis- in patients with vasospastic angina (I; C).
charge. The calculation can be made online at Intravenous beta-blocker treatment at the time
http://www.outcomes-umassmed.org/grace/acs_ of admission should be considered for patients
risk/acs_risk_content.html. in a stable hemodynamic condition (Killip
To date, GRACE score remains the only isch- class <III) with hypertension and/or tachycar-
emic risk score considered in the ESC Guidelines. dia (IIa; C).
At the same time, it is crucial to perform an evalua- Nifedipine or other dihydropyridines are not
tion of the bleeding risk during the prognostic strati- recommended unless combined with beta-
fication. The CRUSADE bleeding score is a score blockers (III; B).
that is used to predict the risk of major bleeding in
light of eight parameters; patients with an increased
risk of bleeding are women, diabetics, and those Oral Antiplatelet Agents
with low hematocrit, with low values of creatinine
clearance, with signs of heart failure, with low (or ASA: loading dose of 150300 mg, mainte-
too high) blood pressure, with high heart rate, and nance dose of 75100 mg daily (I; A).
with history of other vascular disease. P2Y12 inhibitor should be added to aspirin
A score <21 identifies patients at very low risk, and maintained over 12 months (I; A).
between 21 and 30 configures a low risk, between Ticagrelor (180-mg loading dose, 90-mg twice
31 and 40 the risk is moderate, between 41 and 50 daily maintenance dose) is recommended for
the risk is high, and >50 configures a very high all patients at moderate-to-high risk of isch-
risk. The calculation can be made online at http:// emic events (I; B). Contraindications: previ-
www.crusadebleedingscore.org. The execution of ous hemorrhagic stroke, II or III degree
both scores is recommended by the ESC atrioventricular block, sick sinus syndrome,
Guidelines in class I, level of evidence B. bradycardia, syncope, and anticoagulant
therapy.
Prasugrel (60-mg loading dose, 10-mg daily
Anti-ischemic Agents maintenance dose) is recommended for
patients (especially diabetics) in whom coro-
Oxygen insufflation: (48 L/min) if oxygen nary anatomy is known and who are proceed-
saturation is <90 %. ing to PCI (I; B). Contraindications: previous
Nitrate treatment (sublingual, oral, or intrave- stroke or TIA, oral anticoagulant therapy,
nous) is indicated to relieve angina and/or in trauma or recent surgery, age >75 years, and
patients with signs of heart failure (I; C). low body weight <60 kg.

ERRNVPHGLFRVRUJ
2 Non-ST-Segment Elevation Myocardial Infarction (NSTEMI) 25

Clopidogrel (300-mg loading dose, 75-mg coagulant is fondaparinux, a single bolus of


daily dose) is recommended for patients who UFH (85 IU/kg adapted to ACT or 60 IU in
cannot receive ticagrelor or prasugrel (I; A). A the case of concomitant use of GP IIb/IIIa
600-mg loading dose of clopidogrel (or a sup- receptor inhibitors) should be added at the
plementary 300-mg dose at PCI following an time of PCI (I; B). It is contraindicated in
initial 300-mg loading dose) is recommended severe renal failure (CrCl <20 mL/min).
for patients scheduled for an invasive strategy Enoxaparin (1 mg/kg twice daily) is recom-
when ticagrelor or prasugrel is not an option mended when fondaparinux is not available (I;
(I; B). Genotyping and/or platelet function B). Dose reduction to 1 mg/kg once dialysis is
testing may be considered in selected cases indicated in the case of severe renal failure
when clopidogrel is used (IIb; B). (CrCl <30 mL/min).
A proton pump inhibitor (PPI; preferably not If fondaparinux or enoxaparin is not available,
omeprazole) in combination with DAPT is UFH with a target aPTT of 5070 s is indi-
recommended in patients with a history of cated (I; C). UFH infusion is recommended
gastrointestinal hemorrhage or peptic ulcer when CrCl is <30 mL/min or eGFR is <30 mL/
and appropriate for patients with multiple min/1.73 m2 with most anticoagulants
other risk factors (Helicobacter pylori infec- (fondaparinux <20 mL/min).
tion, age >65 years, anticoagulants, or steroids
therapie) (I; B). In a purely conservative strategy, anticoagula-
In patients pretreated with P2Y12 inhibitors tion should be maintained up to hospital dis-
who need to undergo non-emergent major sur- charge (I; A).
gery (including CABG), postponing surgery at Discontinuation of anticoagulation should be
least for 5 days after cessation of ticagrelor or considered after an invasive procedure unless
clopidogrel, and 7 days for prasugrel, if clini- otherwise indicated (IIa; C).
cally feasible and unless the patient is at high Crossover of heparins (UFH and LMWH) is
risk of ischemic events, should be considered not recommended (III; B).
(IIa; C). Ticagrelor or clopidogrel should be
considered to be (re)started after CABG sur-
gery as soon as considered safe (IIa; B). Treatment of Anemia
and Hyperglycemia

GP IIb/IIIa Receptor Inhibitors Treatment of elevated blood glucose should avoid


both excessive hyperglycemia (>180200 mg/
Among patients who are already treated with dL) and hypoglycemia (<90 mg/dL) (I; B).
DAPT, the addition of a GP IIb/IIIa receptor Blood transfusion may have deleterious
inhibitor (abciximab, tirofiban, eptifibatide) for effects, so it is recommended only in the case of
high-risk PCI (elevated troponin, visible throm- compromised hemodynamic status or hematocrit
bus) is recommended if the risk of bleeding is <25 % or hemoglobin level <7 g/dL (I; B).
low (I; B).

Secondary Prevention
Anticoagulants
Beta-blockers are recommended in all patients
Anticoagulation is recommended for all patients with reduced LV systolic function (LVEF <40 %)
(I; A). (I; A).
ACE inhibitors/ARB are indicated within 24 h
Fondaparinux (2.5 mg subcutaneously daily) in all patients with LVEF <40 % and in patients
is recommended as having the most favorable with heart failure, diabetes, hypertension, or
efficacy-safety profile (I; A). If the initial anti- CKD, unless contraindicated (I; A).

ERRNVPHGLFRVRUJ
26 M. Marchesini et al.

ACE inhibitors/ARB are recommended for all 8. Hamm CW et al (2011) ESC Guidelines for the man-
agement of acute coronary syndromes in patients pre-
other patients to prevent recurrence of ischemic
senting without persistent ST-segment elevation. Eur
events, with preference given to agents and doses Heart J 32:29993054
of proven efficacy (I; B). 9. Canto JG, Fincher C, Kiefe CI, Allison JJ, Li Q,
Aldosterone blockade with eplerenone is indi- Funkhouser E, Centor RM, Selker HP, Weissman NW
(2002) Atypical presentations among Medicare bene-
cated in patients after MI who are already being
ficiaries with unstable angina pectoris. Am J Cardiol
treated with ACE inhibitors and beta-blockers and 90:248253
who have an LVEF <35 % and either diabetes or 10. Wu AH, Feng YJ (1998) Biochemical differences
heart failure, without significant renal dysfunction between cTnT and cTnI and their significance for
diagnosis of acute coronary syndromes. Eur Heart J
(serum creatinine >2.5 mg/dL for men and
19(Suppl N):N25N29
>2.0 mg/dL for women) or hyperkalemia (I; A). 11. Chang SA, Choi SI, Choi EK, Kim HK, Jung JW,
Statin therapy with target LDL-C levels Chun EJ, Kim KS, Cho YS, Chung WY, Youn TJ,
<70 mg/dL initiated early after admission is rec- Chae IH, Choi DJ, Chang HJ (2008) Usefulness of
64-slice multidetector computed tomography as an
ommended (I; B).
initial diagnostic approach in patients with acute chest
Patients with NSTE-ACS and severe LV dys- pain. Am Heart J 156:375383
function should be considered after 1 month for 12. Jolly SS, Yusuf S, Cairns J, Niemela K, Xavier D,
device therapy (CRT and/or ICD) in addition to Widimsky P, Budaj A, Niemela M, Valentin V, Lewis
BS, Avezum A, Steg PG, Rao SV, Gao P, Afzal R,
optimal medical therapy (IIa; B).
Joyner CD, Chrolavicius S, Mehta SR (2011) Radial
versus femoral access for coronary angiography and
intervention in patients with acute coronary syn-
dromes (RIVAL): a randomised, parallel group, mul-
References ticentre trial. Lancet 377:14091420
13. Windecker S, et al. ESC/EACTS Guidelines on myo-
1. Nichols M, Townsend N, Scarborough P, Rayner M cardial revascularization (2014) Eur Heart J
(2014) Cardiovascular disease in Europe 2014: epide- 35:25412619
miological update. Eur Heart J 35:2950 14. Ben-Gal Y, Moses JW, Mehran R, Lansky AJ, Weisz
2. GBD 2013 Mortality and Causes of Death G, Nikolsky E, Argenziano M, Williams MR,
Collaborators (2015) Global, regional and national Colombo A, Aylward PE, Stone GW (2010) Surgical
age-sex specific all-cause and cause-specific mortality vs. percutaneous revascularization for multivessel dis-
for 240 causes of death, 19902013: a systematic ease in patients with acute coronary syndromes: anal-
analysis for the Global Burden of Disease Study 2013. ysis from the ACUITY (Acute Catheterization and
Lancet 385:117 Urgent Intervention Triage Strategy) trial. JACC
3. Parikh NI, Gona P, Larson MG et al (2009) Long-term Cardiovasc Interv 3(10):10591067
trends in myocardial infarction incidence and case 15. Palmerini T, Genereux P, Caixeta A, Cristea E,
fatality in the National Heart Lung and Blood Lansky A, Mehran R, Dangas G, Lazar D, Sanchez
Institutes Framingham Heart study. Circulation R, Fahy M, Xu K, Stone GW (2011) Prognostic
119:1203 value of the SYNTAX score in patients with acute
4. Thygesen K et al (2012) ESC/ACCF/AHA/WHF coronary syndromes undergoing percutaneous coro-
Expert Consensus Document: Third Universal nary intervention: analysis from the ACUITY (Acute
Definition of Myocardial Infarction. Eur Heart Journal Catheterization and Urgent Intervention Triage
33:22512567 StrategY) trial. J Am Coll Cardiol 57(24):23892397
5. Fox KA, Eagle KA, Gore JM, Steg PG, Anderson FA 16. Brener SJ, Milford-Beland S, Roe MT, Bhatt DL,
(2012) The Global Registry of Acute Coronary Events, Weintraub WS, Brindis RG (2008) Culprit-only or
1999 to 2009 GRACE. Heart 96:10951101 multivessel revascularization in patients with acute
6. Terkelsen CJ, Lassen JF, Norgaard BL, Gerdes JC coronary syndromes: an American College of
et al (2005) Mortality rates in patients with Cardiology National Cardiovascular Database
ST-elevation vs. non -ST elevation acute myocardial Registry report. Am Heart J 155(1):140146
infarction: observations from an unselected cohort. 17. Hannan EL, Samadashvili Z, Walford G, Jacobs AK,
Eur Heart J 26:1826 Stamato NJ, Venditti FJ, Holmes DR Jr, Sharma S,
7. Hamm C, Heeschen C, Falk E, Fox KAA (2006) Acute King SB 3rd (2013) Staged vs one-time complete
coronary syndromes: pathophysiology, diagnosis and revascularization with percutaneous coronary inter-
risk stratification. In: Camm AJ, Luescher TF, Serruys vention for multivessel coronary artery disease
PW (eds) The ESC textbook of cardiovascular medi- patients without ST-elevation myocardial infarction.
cine. Blackwell Publishing, Oxford, pp 333366 Circ Cardiovasc Interv 6(1):1220

ERRNVPHGLFRVRUJ
Part II
Mimicking Ischemic Heart Disease

ERRNVPHGLFRVRUJ
Pericarditis
3
Alessia Urbinati and Marco Flori

3.1 Case Report Medications

None
A 43-year-old man was admitted to our car-
diology department, reporting a progressive
chest pain which referred to the neck and Vital Signs
left shoulder within the last 7 days.
Temperature: 36 C
Heart rate: 82 bpm
Arterial blood pressure: 140/80 mmHg
Medical History and Cardiovascular Respiratory rate: 12 breaths/min
Risk Factors Oxygen saturation: 99 %

Previous medical history showed acute peri-


carditis 2 months ago and was treated with Physical Examination
ibuprofen 600 mg PO bid for 10 days.
Smoking (ten cigarettes a day) for 10 years. General: alert, awake, and oriented. Well
developed and well nourished
Neck: no jugular venous distention
Allergies Cardiovascular: Regular rate and rhythm, S1
and S2 are normal, mild systolic murmur at
Dust the apex, pericardial friction rub, no gallops,
point of maximal intensity non-displaced and
non-sustained, no hepatojugular reflux
Lungs: clear to auscultation
Abdomen: no pulsatile masses, normal bowel
sounds in all four quadrants, no high-pitched
or tinkling sounds, resonant to percussion,
A. Urbinati (*) M. Flori
Clinica di Cardiologia e Aritmologia, soft, non-distended/non-tender, no rebound or
Universit Politecnica delle Marche, guarding, no hepatosplenomegaly
Via Conca, 71, Ancona, Italy Extremities: no cyanosis or clubbing, no
e-mail: alessiaurbinati@hotmail.it; peripheral edema
mflori3001@gmail.com

Springer International Publishing Switzerland 2015 29


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_3

ERRNVPHGLFRVRUJ
30 A. Urbinati and M. Flori

Laboratory Tests Performed conduction, incomplete right bundle branch


block, and ST elevation in all leads but aVR.
Complete blood count: normal Cardiac echocardiography (Fig. 3.2) showed
Cholesterol (total, HDL, LDL) and TG: normal circumferential pericardial effusion and normal
Hepatic function (GOT, GPT, -GT, ALP, dimensions and function of the cardiac chambers,
total bilirubin, direct and indirect): normal mild mitral regurgitation, and mild inferior vena
Thyroid function (TSH, FT3, FT4): normal cava (IVC) dilatation that collapses with inspira-
Renal function (creatinine, BUN): normal tion. No signs of cardiac tamponade.
Serum electrolytes: potassium 4.4 mEq/l, The thoracic CT and the clinical history
sodium 141 mEq/l. showed no signs of malignancy.
Cardiac biomarkers (troponin, CK-MB): normal After 3 days, ECG findings changed (Fig. 3.3).
Markers of inflammation (CRP, ESR): CRP
1.3 ng/ml, ESR normal
Serologic testing for antinuclear antibody and Final Diagnosis
rheumatoid factor: negative
Mantoux test: negative Recurrent idiopathic acute pericarditis.
The patient was treated with colchicine in
combination with nonsteroidal anti-inflammatory
Instrumental Examination drugs (NSAID).

The first ECG (Fig. 3.1) revealed sinus rhythm


with a heart rate of 85 bpm, normal atrioventricular

Fig. 3.1 Rest ECG showing diffuse ST elevation

ERRNVPHGLFRVRUJ
3 Pericarditis 31

3.2 Pericarditis factor. Acute pericarditis is diagnosed in about


5 % of patients hospitalized to the emergency
Pericarditis is an inflammation of the pericar- department with nonischemic chest pain [2]. In
dium, often with fluid accumulation. Pericarditis developed countries, acute pericarditis is idio-
can be acute, chronic (defined as persistent after pathic in 8590 % of cases [3].
6 months), and recurrent. Acute pericarditis is a
common disorder with an annual incidence of
27.7 new cases per 100,000 inhabitants in Europe Etiology of Acute Pericarditis
[1]. Nevertheless, this disease may occur as an
isolated entity or manifestation of an underlying
Idiopathic
Infectious
Viral
Bacterial
Tuberculous
Fungal
Parasites
Neoplastic disease
Acute myocardial infarction (early peri-
carditis): within 1 or 3 days after a heart
attack
Fig. 3.2 Two-dimensional transthoracic echocardiogram Dressler syndrome
showing the parasternal long-axis view. A moderate-sized
pericardial effusion is present posteriorly

Fig. 3.3 Control ECG showing T-wave inversion in DI, aVL, V3, V4, V5, and V6

ERRNVPHGLFRVRUJ
32 A. Urbinati and M. Flori

pericarditis is still debated. Typical ECG altera-


Autoimmune disorders tions are divided into four stages [9]:
Uremia
Myocarditis
Trauma (severe injury to the chest) Stage 1: ST elevation in all standard
Radiotherapy (especially in breast can- leads except aVR and V1 with upward
cer and lung cancer patients) concavity. Elevation of more than 0.5 mV
Cardiac surgery is uncommon.
Adverse drug reaction (such as penicil- Stage 2: ST segment returns to the base-
lin and chemotherapy medicines) line and T-wave amplitude decreases.
Stage 3: Inversion of T waves, usually
not too deep.
Clinical History and Examination Stage 4: Return to normal.

Patients with pericarditis usually report a pro-


drome of fever, gastrointestinal disorders, or
myalgias. Typically, pericarditis comprises chest Depending on the time of presentation, some
pain, ECG alterations, and laboratory signs of stages might be absent, and different stages might
inflammation [4]. Absence of common inflam- be present in different leads at the same time.
mation markers (erythrocyte sedimentation rate Therefore, ST elevation and T-wave inversion
and C-reactive protein) could be present in up to can be observed simultaneously in different
20 % of cases, especially in the initial stage of the leads, but their coexistence in the same lead is
disease and with previous anti-inflammatory uncommon, and it should suggest an acute coro-
therapies [5]. Assessment of vital sign is nary syndrome. A typical alteration in ECG, dur-
important. The presence of hypotension, dys- ing pericarditis, is PR segment depression
pnea, reduced cardiac tones, and signs of elevated 0.5 mV compared to TP segment: these find-
venous tone (jugular distension, liver tenderness) ings are usually an early alteration, and it is
may indicate cardiac tamponade, which requires shown in almost 80 % of cases.
an urgent treatment. Chest pain is usually of Chest radiography is useful to exclude sec-
abrupt onset, stabbing or sharp, and substernal ondary causes of chest pain (pneumonia, pneu-
but may be radiated to arms, neck, or jaw. Chest mothorax, traumatism) and might show enlarged
pain is more intense in supine position and lighter heart in case of pericardial effusion.
in prone position. Pericardial rubs are due to the Echocardiography has a pivotal role for the
friction of inflamed pericardial layer and fibrin, assessment of the cardiac function and presence
auscultated usually in patients leaning frontward, and quantification of pericardial effusion. A large
at the left sternal border, throughout the whole pericardial effusion with diastolic collapse of the
cardiac cycle. The rub of pericarditis might be right atrium, respiratory variation of mitral inflow
transient, and it is important for the clinicians to (decrease in E wave of >20 % during inspiration),
auscultate the heart repeatedly [6]. and dilated inferior vena cava with reduced col-
Electrocardiogram might be normal during lapsibility may suggest cardiac tamponade.
pericarditis. Further, pericardial effusion can lead
to a decrease in the amplitude of the QRS voltage,
while the P waves are less frequently affected [7]. Differential Diagnosis
Nonetheless, it must be considered that low QRS
voltage is a weak predictor of pericardial effusion Acute pericarditis is usually benign but should be
and the correlation between the decrease in QRS promptly distinguished from the other life-
amplitude and the amount of fluid is poor [8]. threatening conditions. Ruling out an acute coro-
The etiology of changes in repolarization during nary syndrome (ACS) is of utmost importance.

ERRNVPHGLFRVRUJ
3 Pericarditis 33

Table 3.1 Different characteristics of pericarditis and acute coronary syndrome


Acute pericarditis Acute coronary syndrome
Clinical history CV risk factors Not correlated Correlated
and examination History of fever or Frequent No
GI disorders
Chest rubs Frequent No
Chest pain Hitching/stabbing; reduced Heaviness/constriction; no correlation
leaning frontward with movements
ECG ST elevation Diffuse, upward concavity Localized with specular ST depression
PR depression Frequent No
T-wave inversion Diffuse, uncommon with Diffuse/localized; ST elevation/
concomitant ST elevation/ depression may coexist
depression
Echocardiography Kinetics Normala Focal kinetic defect
Pericardial effusion Frequent Rare, may be present in cardiac rupture
Laboratory Troponin Negativea Positive; negative in UA
Inflammation Usually elevated Usually normal
markers
CV cardiovascular, GI gastrointestinal disorders, UA unstable angina
a
May be altered with concomitant myocarditis

Assessment of cardiovascular risk factors and pre- demanding physical activity help to minimize the
vious heart diseases may help in the diagnosis. symptoms.
Different characteristics of acute pericarditis and When pericardial effusion is present and clini-
ACS are shown in Table 3.1. Pneumothorax, acute cal examination and echocardiography suggest
pulmonary embolism, and acute aortic syndrome hemodynamic impairment of cardiac function
are other causes of acute chest pain with high (cardiac tamponade), pericardiocentesis should
mortality if undiagnosed and have to be excluded. be considered.
Other conditions that may simulate pericarditis Prognosis of acute pericarditis depends on eti-
are respiratory disorders (pneumonia, bronchitis, ology and comorbidities. Conditions such malig-
asthma), musculoskeletal diseases, herpes zoster, nancy, uremia, and HIV have the worse prognosis.
anxiety, and others, but ECG and cardiac bio- Viral or idiopathic pericarditis is usually a benign
marker alterations are rare in these conditions. condition with very low mortality.
Relapsing pericarditis is one of the most chal-
lenging complications of acute pericarditis, and
Therapy and Prognosis no optimal treatment for this disorder of the peri-
cardium has been definitively established. One or
Nonsteroidal anti-inflammatory drugs (NSAIDs) more recurrences arise up to 30 % of patients
are the first-line therapy in acute pericarditis. after an initial episode of acute pericarditis, usu-
High-dose aspirin, ibuprofen, or indomethacin ally within the first 18 months. Findings are simi-
may be used for 13 weeks, but no single NSAID lar to the initial episode, including pleuritic chest
appears to be more effective than the other [2]. pain, diffuse ST-segment elevation, and elevated
Colchicine has shown to be safe and effec- serum markers of inflammation [11]. Recurrences
tive in reducing recurrences [10]. Pericarditis must be differentiated from episodes of chest dis-
symptoms respond promptly to corticosteroid comfort with the patient reminiscent of prior
therapy. Nonetheless, corticosteroids are associ- pericardial pain but no other sign of recurrence
ated with increased recurrences and should only (no elevation of inflammatory markers, no ECG
be used when other therapies have shown to be alterations, pericardial rubs, or pericardial
ineffective. Concurrently, rest and avoidance of effusion).

ERRNVPHGLFRVRUJ
34 A. Urbinati and M. Flori

In the absence of specific clinical trials, 3. Zayas R, Anguita M, Torres F et al (1995) Incidence
of specific etiology and role of methods for specific
NSAIDs should be the first-line therapy of
etiologic diagnosis of primary acute pericarditis. Am
recurrences with a prolonged administration for J Cardiol 75(5):378382
24 weeks. Colchicine associated to NSAIDs 4. Maisch B, Ristic AD (2002) The classification of peri-
has shown to reduce episodes of recurrent peri- cardial disease in the age of modern medicine. Curr
Cardiol Rep 4:1321
carditis of additional 50 % [12]. If refractory
5. Imazio M et al (2011) Prevalence of C-reactive pro-
pericarditis is present, corticosteroid therapy tein elevation and time course of normalization in
should be considered using lower effective dose acute pericarditis. Circulation 123(10):10921097
for their side effects in long-term therapies. 6. Spodick DH (2003) Acute pericarditis: current con-
cepts and practice. JAMA 289:11501153
Immunosuppressors (methotrexate, azathioprine)
7. Holzmann M (1965) Klinische Elektrokardiographie.
have shown symptom improvement in a small Georg Thieme, Stuttgart, pp 511525
number of patients not respondent to steroids. 8. Casale PN, Deveraux RB, Kligfield P et al (1984)
Finally, few data of pericardiotomy in continuous Pericardial effusion: relation of electrocardiographic
finding. J Electrocardiol 17:115
relapsing pericarditis showed variable efficacy
9. Spodick DH (1974) The electrocardiogram in acute
with some patients reporting complete remission pericarditis: distributions of morphologic and axial
but others continuing to be plagued with ongoing changes in stages. Am J Cardiol 33:470474
symptoms [13]. 10. Imazio M et al (2005) Colchicine in addition to con-
ventional therapy for acute pericarditis. Circulation
112:20122016
11. Imazio M et al (2010) Controversial issues in the
References management of pericardial diseases. Circulation
121:916928
1. Imazio M, Cecchi E, Demichelis B et al (2008) 12. Imazio M, CORP (COlchicine for Recurrent
Myopericarditis versus viral or idiopathic acute peri- Pericarditis) Investigators et al (2011) Colchicine for
carditis. Heart 94(4):498501 recurrent pericarditis (CORP): a randomized trial.
2. Khandaker MH, Espinosa RE, Nishimura RA, Sinak Ann Intern Med 155:409414
LJ, Hayes SN, Melduni RM, Oh JK (2010) Pericardial 13. Khandaker MH et al (2012) Pericardiectomy vs medi-
disease: diagnosis and management. Mayo Clin Proc cal management in patients with relapsing pericardi-
85:572593 tis. Mayo Clin Proc 87:10621070

ERRNVPHGLFRVRUJ
Intracranial Bleeding
4
Alessia Urbinati and Michela Brambatti

4.1 Case Report Vital Signs

Heart rate: 80 bpm


A 40-year-old man was admitted to the Blood pressure: 108/80 mmHg
emergency room after a 2.5 m fall from Oxygen saturation: 98 %
a scaffold. He showed signs of impaired
consciousness and a scalp wound localized
in the left tempofrontal region. Personal Physical Examination
pathological history was unknown.
The neurological evaluation revealed a General: traumatic scalp wound 4 cm
Glasgow Coma Scale (GCS) score of 10:
eye response = 3 (eye opening to verbal
command), motor response = 6 (obeys
command), and verbal response = 1 (no
verbal response). The patient, after tra-
cheal intubation for airway protection,
underwent head computed tomography
(CT) scan, which revealed bifrontal and
left temporal subcortical-cortical bleeding
with subarachnoid hemorrhage (Fig. 4.1).
Subsequent chest x-ray, abdominal ultra-
sound, and chest-abdominal CT revealed
no further traumatic injuries.

A. Urbinati (*) M. Brambatti


Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche,
Via Conca, 71, Ancona 60126, Italy
e-mail: alessiaurbinati@hotmail.it; Fig. 4.1 Brain axial CT showing left temporal focal
michelabrambatti@gmail.com bleeding

Springer International Publishing Switzerland 2015 35


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_4

ERRNVPHGLFRVRUJ
36 A. Urbinati and M. Brambatti

Neurological: mental status was impaired. Laboratory Exams on Admission


Pupil exam showed pupil asymmetry <1 mm and Other Tests
and no dilatation or constriction; pupils were
reactive to light and accommodation bilater- Exams (including high-sensitivity cardiac tropo-
ally. No alterations in deep tendon reflexes. nin) were unremarkable except for a mild leuko-
Cardiovascular: regular rate and rhythm, S1 cytosis of 14,500/mm3.
and S2 normal, no murmurs, rubs, or gallops, After 2 days in the hospital, the patient had a
and no hepatojugular reflux. control CT of the brain that showed an increased
Lungs: no signs of respiratory effort on inspec- size of the left temporal subarachnoid hemor-
tion. No wheezes, no crackles or stridor, and rhage, with minimum brain compression and
no bronchial or vesicular breath sounds on appearance of posterior subcortical-cortical
auscultation over the lung fields bilaterally. bleeding. The patient was clinically stable, but
Abdomen: no lesions or signs of trauma. No ECG changed dramatically and showed sinus
pain or rebound on light and deep palpation, tachycardia; normal atrioventricular conduction;
no organomegaly, and normal bowel sounds in ST elevation in DII, DIII, aVF, V4, V5, and V6;
all four quadrants. and ST depression in aVR, aVL, V1, V2, and V3
(Fig. 4.2).
Routine ECG was performed after the hospital- Fast echocardiography was performed,
ization, showing sinus rhythm, normal atrioven- revealing no cardiac contractile abnormalities.
tricular conduction, incomplete right bundle branch Blood sample showed a rise in troponin level:
(RBBB), and no alterations in repolarization. 0.16 ng/ml.

Fig. 4.2 Rest ECG showing incomplete right bundle block and normal repolarization

ERRNVPHGLFRVRUJ
4 Intracranial Bleeding 37

What Are the Possible Causes for ST coronary artery. In the following 48 h, ST eleva-
Elevation? tion gradually resolved, and waves were inverted
in DII, DIII, aVF, V4, V5, and V6 (Fig. 4.3).
A repeated head CT scan revealed a partial
Acute ST-segment elevation myocardial reduction of the encephalic bleeding. The patient
infarction (STEMI) was extubated and transferred to a spoke inten-
Coronary spasm sive care unit.
Pericarditis
Left ventricular aneurysm
Brugada syndrome 4.2 ECG Correlation
Left bundle branch block and Intracranial Bleeding
Left ventricular hypertrophy
Early repolarization ECG Abnormalities
Drugs (e.g., cocaine, digoxin, quinidine,
tricyclics, and many others) In patients with acute cerebrovascular events,
Electrolyte abnormalities (e.g., especially intracranial hemorrhage (ICH) and
hyperkalemia) ischemic stroke, electrocardiographic repolariza-
Neurogenic factors (e.g., stroke, hemor- tion abnormalities are frequently observed
rhage, trauma, tumor, etc.) regardless of the presence or absence of previous
Metabolic factors (e.g., hypothermia, cardiac diseases. Repolarization abnormalities
hypoglycemia, hyperventilation) warrant attention as they increase the vulnerable
period during which an extrasystole is more
likely to result in ventricular tachycardia or fibril-
What Are the Possible Causes lation. Repolarization abnormalities thus may
of Increased Troponin Level? explain the higher risk of arrhythmias and sudden
death following acute neurological disorders
[14].
Acute coronary syndrome The exact incidence of ECG abnormalities is
High blood pressure in lung arteries still unclear but ranges from 50 to 98 % and
(pulmonary hypertension) includes QT prolongation, T wave inversion,
Blockage of a lung artery by a blood prominent U wave, ST segment elevation or
clot, fat, or tumor cells (pulmonary depression, peaked P waves, and transient patho-
embolus) logical Q waves [1]. A typical, but not pathogno-
Congestive heart failure monic, pattern is characterized by giant T
Myocarditis waves, abnormally wide T waves of increased
Prolonged exercise amplitude (mostly negative than positive); eleva-
Trauma that injures the heart tion or depression of the ST segment is less fre-
Weakening of the heart muscle quent [4].
(cardiomyopathy) However, as in our case, these ST changes are
Long-term kidney disease particularly important since they may simulate
acute myocardial ischemia and therefore mislead
physicians in the initial diagnosis. In fact, uncon-
In a setting of acute coronary syndrome and scious patients with ICH and ST segment eleva-
ST elevation, the patient has been promptly tion may be misdiagnosed with acute myocardial
treated with metoprolol and acetylsalicylic acid infarction and could be incorrectly treated with
intravenous and a percutaneous coronary angiog- antiplatelet and anticoagulative drugs with devas-
raphy was urgently performed which showed just tating consequences. Also, in the setting of cere-
a non-critical stenosis (<30 %) of the right brovascular events, changes in ST segment have

ERRNVPHGLFRVRUJ
38 A. Urbinati and M. Brambatti

Fig. 4.3 Control ECG showing ST elevation in DII, DIII, aVF, V4, V5, and V6 and ST depression in aVR, aVL, V1, V2,
and V3

generally the same polarity of T wave, and among patients who have developed brainstem or
rarely, ST elevation is diffuse simulating acute peri-insular hematomas [8].
pericarditis. As a consequence of repolarization abnormal-
The ECG abnormalities usually reverse after ities, especially with prolonged QT interval,
the neurological clinical recovery. malignant ventricular arrhythmias, including
ventricular tachycardia, torsades de pointes, and
ventricular fibrillation, may also be observed [9].
Arrhythmias So, it is very important to check for QTc prolon-
gation and to select drugs that do not prolong QT
In a prospective study, serious arrhythmia in the interval.
first 72 h after admission with acute stroke was
detected in 25 % of 501 patients [5]. The risk for
cardiac arrhythmia was highest during the first Mechanisms Leading to ECG
24 h of care and declined with time during the Abnormalities
first 3 days. In previous series, cardiac arrhyth-
mias were detected in 625 % of patients after The mechanism through which cerebrovascular
stroke [6, 7]. events lead to these ECG abnormalities is still
Atrial fibrillation is the most common cardiac unknown; however, autoptic findings reported
arrhythmia accounting for nearly 60 % of all that over 50 % of patients had cardiac muscle
events of arrhythmia [5]. However, transient contraction band necrosis. The most plausible
atrial fibrillation is more common among patients hypothesis suggests that ECG changes result as a
with ischemic stroke than with ICH; in this latter consequent alteration of the autonomic nervous
setting, atrial fibrillation occurs more frequently system control on cardiac electrophysiology that

ERRNVPHGLFRVRUJ
4 Intracranial Bleeding 39

results in catecholamine release at the terminal 3. Korpelainen JT, Sotaniemi KA, Huikuri HV (1997)
MyllylVV Circadian rhythm of heart rate variability
nerve at the cardiac myocyte [2, 3]. Increase of
is reversibly abolished in ischemic stroke. Stroke
adrenergic stimulus increases myocardial oxygen 28(11):2150
demand, reduces coronary perfusion time, and 4. Byer E, Ashman R (1947) Toth La Electrocardiograms
also has a direct cardiotoxic effect with general- with large, upright T waves and long Q-T intervals.
Am Heart J 33(6):796
ized spasm of the coronary arteries. Some evi-
5. Kallmnzer B, Breuer L, Kahl N, Bobinger T, Raaz-
dence suggests also that sympathetic Schrauder D, Huttner HB, Schwab S, Khrmann M
stroke-related activity causes calcium overload in (2012) Serious cardiac arrhythmias after stroke: inci-
ventricular myocytes and consequently arrhyth- dence, time course, and predictors--a systematic, pro-
spective analysis. Stroke 43(11):28922897. Epub
mias. Furthermore, the type and location of the
2012 Sep 6
cerebrovascular event may correlate with the type 6. Goldstein DS (1979) The electrocardiogram in stroke:
of arrhythmia as each cerebral hemisphere seems relationship to pathophysiological type and compari-
to have a different influence on cardiac functions. son with prior tracings. Stroke 10(3):253
7. Rem JA, Hachinski VC, Boughner DR, Barnett HJ
Indeed, injury of the right insula may cause bra-
(1985) Value of cardiac monitoring and echocardiog-
dycardia and vasodepressor effects, while the left raphy in TIA and stroke patients. Stroke 16(6):950
insular region may pose a higher risk for tachy- 8. Yamour BJ, Sridharan MR, Rice JF (1980) Flowers
cardia and hypertension [10, 11]. NC Electrocardiographic changes in cerebrovascular
hemorrhage. Am Heart J 99(3):294
9. Cruickshank JM, Neil-Dwyer G, Brice J (1974)
Electrocardiographic changes and their prognostic
References significance in subarachnoid haemorrhage. J Neurol
Neurosurg Psychiatr 37:755759. doi:10.1136/
1. Dimant J, Grob D (1977) Electrocardiographic jnnp.37.6.755
changes and myocardial damage in patients with 10. Hachinski VC, Oppenheimer SM, Wilson JX et al
acute cerebrovascular accidents. Stroke 8(4): (1992) Asymmetry of sympathetic consequences of
448455 experimental stroke. Arch Neurol 49:697
2. Hachinski VC (1993) The clinical problem of brain 11. Lane RD, Wallace JD, Petrosky PP et al (1992)
and heart. Stroke 24(12 Suppl):I1I2; discussion Supraventricular tachycardia in patients with right
I102 hemisphere strokes. Stroke 23:362

ERRNVPHGLFRVRUJ
Takotsubo Cardiomyopathy
5
Giulia Pongetti and Azzurra Fabbrizioli

5.1 Case Report Allergies

No allergy was referred by the patient.


A 65-year-old woman presented to the
emergency room for chest pain developed Social History
after a car accident, where she was
involved, persistent for at least 30 min. The She has two sons in good health. At present,
pain did not decrease by changing position. she is menopausal and retired.
It consisted of a retrosternal pressure or She drinks a glass of wine once in a while.
heaviness (angina) radiating to the neck, She walks for about 3 km three times a week.
accompanied by dyspnea and palpitations.
Medications

Sertraline 50 mg/day at 8:00 am


Medical History and Cardiovascular Valsartan 160 mg/day at 8:00 am
Risk Factors
Vital Signs
Familiar history of ischaemic cardiovascular
disease Temperature: 36 C
Current smoker (about ten cigarettes per day) Heart rate: 110 bpm
Arterial hypertension Blood pressure: 160/100 mmHg
Dyslipidemia Respiratory rate: 18 breaths per minute
Anxious depressive syndrome Oxygen saturation while breathing ambient
She doesnt refer any other previous cardio- air: 98 %
vascular disease
Physical Examination

G. Pongetti (*) A. Fabbrizioli General: agitated, alerted, awake, and oriented


Clinica di Cardiologia e Aritmologia, Head, eyes, ears, nose, and throat: normo-
Universit Politecnica delle Marche, cephalic, atraumatic, mucous membranes
Via Conca, 71, Ancona, Italy
e-mail: giuliapongetti@gmail.com; moist, extraocular muscles intact, pupils
Azzurra.fab@gmail.com equally round and reactive to light and

Springer International Publishing Switzerland 2015 41


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_5

ERRNVPHGLFRVRUJ
42 G. Pongetti and A. Fabbrizioli

accommodation bilaterally, bilateral tympanic


membrane intact, bilateral sclera anicteric, Fasting blood glucose: 156 mg/dl
and no conjunctival injection. Troponin I: 0.13 ng/ml (n.v. <0.08 ng/
Neck: supple, no jugular venous distension, no ml)
lymphadenopathy, and no carotid bruit. CK-MB: 1.7 ng/ml (n.v. <5 ng/ml)
Cardiovascular: regular heart rate, protodia- BNP 92 pg/ml (n.v. <100 pg/ml)
stolic gallop with S3 added, apical holosys- Inflammation index: VES 25 mm/h (n.v.
tolic murmur 2/6, no rubs, no hepatojugular <27 mm/h;) and CRP 0.5 mg/dl (n.v.
reflux, and capillary refill less than 2 s. <0.6 ng/ml)
Lungs: no rales on auscultation, no rhonchi or D-dimers: 180 ng/ml (n.v. <230 ng/ml)
wheezes, no egophony, no alterations in tactile
fremitus, and normal upon percussion.
Abdomen: plan, no pulsatile masses, normal Chest X-Ray
bowel sounds in all four quadrants, no high-
pitched or tinkling sounds, resonant on per- Normal heart size and volumes. No bone frac-
cussion, soft, nondistended/nontender, no tures and no signs of trauma or dissection of the
rebound or guarding, no costovertebral angle major mediastinal vessels. Absence of pulmo-
tenderness, and no hepatosplenomegaly. nary congestion, pleural effusion, or signs of
Extremities: no cyanosis or clubbing. No pneumothorax (not shown).
peripheral edema.
Neurological: no focal deficit.
Psychiatric: normal affect, no hallucinations, What Are the Possible Causes
normal speech, and no dysarthria. of Chest Pain in This Patient?
Skin: intact, no rashes, no lesions, and no
erythema.
Myocardial disease
Acute coronary syndromes without
Routine EKG at Rest persistent ST elevation (NSTE-ACS)
or unstable angina (UA)
Sinus tachycardia at 120 bpm, normal atrioven- Myocardial infarction secondary to
tricular conduction (PR 120 ms), complete right an ischemic imbalance for rise of
bundle branch block (QRS 140 ms), ST segment blood pressure or tachycardia
depression from V1 to V6 with inverted T waves, Coronary spasm
and QTc interval of 460 ms (Fig. 5.1). Cardiac contusion
Stress cardiomyopathy (Takotsubo
syndrome)
Routine Laboratory Tests Myocarditis, pericarditis, and
myopericarditis
Valvular heart disease
Complete blood count: normal Severe aortic stenosis
Renal function: creatinine 0.8 mg/dl Severe aortic regurgitation
(normal) Aortic disease
Hepatic function (GOT, GPT, y-GT, Traumatic aortic injury
ALP, total bilirubin, direct and indirect): Thoracic trauma and/or fractures
normal Pulmonary disease
Electrolytes: Na 137 mEq/L and K Pneumothorax
4.1 mEq/L (normal) Pulmonary embolism

ERRNVPHGLFRVRUJ
5 Takotsubo Cardiomyopathy 43

Fig. 5.1 EKG at rest while the patient was symptomatic

A pulmonary embolism is unlikely for the low angina) although it may consider a possible
pretest probability (the patient has never suffered posterior ST elevation with anterior ST depres-
previous pulmonary embolism or deep vein sion as a mirror image.
thrombosis; she didnt have previous surgery, A phlogistic damage (myocarditis, myopericar-
trauma, long immobilization, or cancer; and she ditis) is unlikely because the EKG modifications
never used hormone replacement therapy). are not typical and not widely represented in all the
Furthermore, the normal D-dimer level excludes EKG leads; moreover, the patient didnt report flu
this affection with high sensitivity. or gastrointestinal disease, and the laboratory tests
Pneumothorax and thoracic fractures can be did not show clear signs of a phlogistic state.
excluded because physical examination and chest The chest pain referred by the patient was
x-ray were normal. quite typical for angina or also could suggest
The EKG shows ST segment depression in an aortic damage caused by car accident. An
anterior leads which ruled out coronary spasm, echocardiography was done to evaluate those
usually characterized by ST elevation (Prinzmetal possible diagnostic alternatives.

ERRNVPHGLFRVRUJ
44 G. Pongetti and A. Fabbrizioli

Echocardiography (TAI) was also unlikely because of features of


the car accident suffered by the patient.
The aortic valve is trileaflet. Slight enlargement of The echocardiography findings showed mod-
the left atrium (LA diameter M-mode = 4.2 cm; erate reduction of systolic function for apical
area 4 c = 22 cm2). Mild functional mitral regurgi- and mid-left ventricular dilatation and aki-
tation caused by displacement of the papillary nesia with compensatory basal hyperkinesis
muscles. Normal right atrium (area 4 c = 12 cm2). compatible with ischemic injury. This pattern
Right ventricle also normal in size and function. of wall motion abnormality (apical balloon-
Normal tricuspid valve. Mild tricuspid regurgita- ing) is quite typical for stress cardiomyopa-
tion centrally directed (PASP = 35 mmHg). Normal thy (Takotsubo-like), but also an AMI cannot
pulmonic valve. The interatrial septum is normal. be excluded without a coronary angiography.
The left ventricle is slightly hypertrophic (LV mass
index 97 g/m2) with apical and mid-left ventricular While the patient has been prepared for coro-
dilatation; moderate reduction of systolic global nary angiography, he has been treated with:
function for complete akinesis of the apical and
mid-segments and compensatory basal hyperkine- Initial loading dose of aspirin (300 mg)
sis. The ejection fraction measured with Simpsons Initial loading dose of ticagrelor (180 mg)
biplane method was 40 %. Normal pericardium Fondaparinux 2.5 mg/day
and aorta (aortic root dimension = 3.0 cm; ascend- Perindopril 2 mg/day
ing aorta = 3.2 cm; aortic arch = 3.3 cm; thoracic Atorvastatin 80 mg/day
aorta = 2.9 cm; abdominal aorta = 2.0 cm). The Bisoprolol 2.5 mg/day
inferior vena cava was not dilated. Pseudonormal
diastolic pattern with increased filling pressure
(E/A 1.1, E/E 16, E dec time 170 m/s) (Fig. 5.2). Coronary Angiography and Left
Ventriculography
Conclusion
Severe valvular disease, hypertrophic cardio- A coronary angiography was performed within
myopathy pericardial effusion, and rupture of 24 h from admission showing right coronary
the wall or septum were ruled out. dominance and absence of significant coronary
Aortic root and aortic isthmus diameters were artery stenosis (Fig. 5.3).
normal without any intimal flap or thrombus The left ventriculography showed complete
along the aortic wall. A traumatic aortic injury apical and mid-ventricular akinesis with systolic

a b

Fig. 5.2 Echocardiographic picture recorded at rest dur- akinesis in the apical to mid-segments circumferentially
ing systole (a) and during diastole (b); typical LV balloon- of the left ventricle with relative compensatory hypercon-
ing during systole is visible, characterized by complete tractility in the basal segments

ERRNVPHGLFRVRUJ
5 Takotsubo Cardiomyopathy 45

a b

Fig. 5.3 Coronary angiography of left coronary artery (a) and right coronary artery (b); right coronary dominance and
absence of significant coronary artery stenosis are shown

a b

Fig. 5.4 Left ventriculography recorded during diastole (a) and systole (b); complete apical akinesis and mid-ventric-
ular akinesis are shown, with systolic apical ballooning and moderate systolic dysfunction

apical ballooning and moderate systolic dys- presentation and with low plasma levels of car-
function. No spasm was recorded (Fig. 5.4). diac biomarkers, disproportionate with the sever-
ity of ventricular dysfunction, could suggest the
Conclusion diagnosis of Takotsubo cardiomyopathy. This
Absence of significant culprit coronary artery diagnosis is confirmed by findings on imaging
stenosis or intracoronary thrombosis (echocardiography and ventriculography) of
Apical ballooning pattern transient apical- to mid-ventricular ballooning
with compensatory basal hyperkinesis, without
coronary stenosis.
Final Diagnosis Subsequent blood tests have shown normaliza-
tion of cardiac biomarkers (max values: troponin I
The clinical presentation that mimics an acute 2.45 ng/mL, CK MB 6.7 ng/mL), and echocar-
myocardial infarction triggered by emotional diography has displayed progressive recovery of
stress (car accident) with modest EKG changes at contractile function until complete normalization.

ERRNVPHGLFRVRUJ
46 G. Pongetti and A. Fabbrizioli

Subsequent EKGs performed during hospital- branch block (QTS 140 ms), and normal ST seg-
ization showed the typical evolution described in ment with deep inverted T waves from V1 to V6
TC with new onset of negative and deep T waves and DI, DII, AVL, and AVF (QTc interval of
and marked QTc interval prolongation and then 624 ms) (Fig. 5.5).
slow and complete regression of these abnormali- The patient was discharged with the following
ties after about 15 days. therapy:

Aspirin 100 mg/day


EKG Performed at Day 3 Perindopril 2 mg/day
Atorvastatin 40 mg/day
Sinus rhythm at 65 bpm, normal atrioventricular Bisoprolol 5 mg/day
conduction (PR 120 ms), complete right bundle

Fig. 5.5 EKG at rest during the third day of hospitalization while the patient was asymptomatic

ERRNVPHGLFRVRUJ
5 Takotsubo Cardiomyopathy 47

5.2 Takotsubo Syndrome Finally, the histological changes usually


observed in TC (coagulative myocytolysis and
Denition myofibrillar degeneration) are different from
those observed in ischemic disease (coagula-
Takotsubo syndrome (TS) is a reversible cardio- tive necrosis) [6].
myopathy, first described in 1990 by Sato et al. in Coronary microvascular dysfunction: This
Japan [1], also known as apical ballooning hypothesis has been initially proposed based
syndrome or stress cardiomyopathy, since it on findings of a diminished coronary flow
has become increasingly recognized worldwide. reserve [7] and on a significant improvement
It is characterized by signs and symptoms of of myocardial perfusion during adenosine
acute myocardial infarction (AMI) but without infusion in patients with TC but not with
demonstrable coronary artery stenosis. STEMI [8]. However, other trials didnt con-
firm the slow flow phenomenon in many
Takotsubo patients, so they supposed that this
Epidemiology impairment of microcirculation could be a
possible consequence of the myocardial dys-
TS accounts for about 12 % of all cases of sus- function caused by TC itself [9].
pected AMI [2, 3]. Catecholamine toxicity: Some authors found
It typically occurs in postmenopausal elderly markedly elevated catecholamine levels in
women (average age 68 years); men account for a patients with TC, suggesting that these might
minority of cases (413 %), and rarely also chil- be the main pathogenic factor [10]; however,
dren or young adults may be affected [4]. many subsequent studies have not uniformly
shown elevated plasma catecholamine levels
in TC patients [11]. So it is likely that circulat-
Pathogenesis ing catecholamines are somehow involved in
the pathogenic process but not the only ones
The exact pathogenesis of Takotsubo cardiomyopa- responsible.
thy (TC) is still unknown, but various hypotheses Cardiac sympathetic disruption: Recently a
have been suggested and discussed over the years. new hypothesis has been proposed regarding
the disruption of local cardiac sympathetic
Coronary artery spasm or transient acute nerve endings with local release of norepi-
artery thrombosis [3, 5]: These hypotheses nephrine into the myocardial tissue, which
have been actually withdrawn because the could damage it and lead to the systolic dys-
typical apical ballooning is incongruent to a function; moreover, it seems that the charac-
singular coronary artery supply region. The teristic circular pattern of ventricular wall
hypothesis of a multivessel coronary spasm motion abnormality follows the cardiac sym-
would not explain the discrepancy between pathetic supply system [12].
severe ventricular dysfunction and the only
slightly increased levels of cardiac enzymes
and why the recurrence could present different Clinical Features
myocardial localization in the same patient or
why some patients show the apical sparing The clinical features of TS are usually completely
variant [3]. superimposable with those of an acute myocar-
The long-lasting ST segment elevation in dial infarction (AMI).
Takotsubo patients challenges strongly the
hypothesis of a transient thrombosis which Symptoms: acute and prolonged anginal chest
usually presents rapid resolution of ST seg- pain, described like retrosternal pressure or
ment elevation [6]. heaviness, sometimes radiating to the left arm or

ERRNVPHGLFRVRUJ
48 G. Pongetti and A. Fabbrizioli

neck. When present, a typical aspect useful for lar name: it resembles a typical pot (called
diagnosis is that the chest pain usually occurs exactly Takotsubo) used for a long time in
after mental or physical stress, such as the unex- Japan to capture octopuses, which has a round
pected death of a relative or friend or receiving bottom and a narrow neck.
news of serious diagnosis or having a great Other patterns of wall motion abnormality
fright [13]. However, recently an international have been reported less frequently, involving
meta-analysis (COUNTS study) has shown that the basal-mid segment (apical sparing variant
these stressors have a limited impact occurring or inverted Takotsubo) or the right ventricle.
in about only 36 % of patients [14]. In men, Typically, the wall motion abnormality
physical stress rather than emotional stress is involves regions which are incongruent to a
much more associated with the occurrence [4]. particular coronary artery supply; instead dur-
Dyspnea, nausea, and diaphoresis can accom- ing an AMI, it is more common to find these
pany chest pain. hypokinetic/akinetic areas supplied by a par-
Comorbidities: A high number of patients ticular coronary artery.
affected by TS present cardiovascular risk fac-
tors like hypertension (54 %), dyslipidemia
(32 %), diabetes mellitus, obesity, and history Clinical Course and Complications
of smoking, similar to that seen in patients with
AMI [14]. Moreover, it has been noted that Clinical course of TC is usually characterized by
sometimes TS can occur in patients affected by the progressive lowering of chest pain and gradu-
comorbidities that are usually associated with ally complete resolution of the wall motion
an excess of catecholamine systemic produc- abnormality after some days; less frequently it
tion like obstructive pulmonary disease, sepsis, needs some weeks or months, but differently
thyroid (thyrotoxicosis), or neurological dis- from AMI, there arent permanent areas of hypo-
ease (cerebrovascular accidents, subarachnoid kinesis/akinesis of the left ventricle motion.
haemorrhage, etc.) [3], malignancy, or psycho-
logical disorders (22 %) [14]. EKG evolution: The EKG abnormalities show
12-Lead EKG: During the acute phase, it usu- a slower progression than during AMI, with a
ally shows ST segment depression or tran- specific evolution: gradually during the first
sient/persistent ST segment elevation in hours from symptom onset, there is the com-
anterior leads, perfectly mimic king acute plete resolution of ST segment elevation/
myocardial infarction [1518]. Sometimes it depression. Negative T waves usually occur
can show also T-wave inversion or Q waves. after 2472 h and then gradually deepen pro-
In subsequent days, the EKG abnormalities gressively during the days/weeks following
follow a distinctive time sequence [19]. [20]. Abnormal Q waves could be seen in pre-
Cardiac biomarkers: Troponin, creatinine cordial leads, but these are transient in most
kinase, and BNP are usually increased. patients and generally resolve within a few
Echocardiography: It plays a central role days to several weeks.
during diagnostic process because it shows a QTc interval becomes prolonged (>500 ms)
typical pattern of wall motion abnormality: progressively as the negative T wave deepens,
complete akinesis or hypokinesis in the apical and this could favor life-threatening ventricu-
to mid-segments circumferentially of the left lar arrhythmias such as torsades de pointes
ventricle with relative compensatory hyper- and ventricular fibrillation [18].
contractility in the basal segments (LV bal- After some weeks, there is usually the
looning during systole). During left complete restoration of the ECG pattern prior
ventriculography, this particular systolic left to the acute event [15].
ventricular shape is more evident and explains Recurrences: These may occur with a rate
the reason why this syndrome has this particu- range from 0 to 15 % after the first episode

ERRNVPHGLFRVRUJ
5 Takotsubo Cardiomyopathy 49

and may show the typical apical ballooning or first diagnostic criteria in 2004, which have been
even different patterns of wall motion abnor- modified recently (2008) and included [25, 26]:
malities [21].
Acute complication: Despite the most com- 1. Transient hypokinesis, dyskinesis, or akinesis
mon general favorable course, the in-hospital of the left ventricular midsegments, with or
death ranges from 0 to 8 % because of possi- without apical involvement; the regional wall
ble acute complications [22]. Congestive heart motion abnormalities extend beyond a single
failure is one of the most common acute com- epicardial vascular distribution, and a stress-
plications (approximately 346 %) occurring ful trigger is often, but not always, present.
more frequently in patients with right ventric- 2. Absence of obstructive coronary disease or
ular involvement [22]. angiographic evidence of acute plaque
As already mentioned, during the acute and rupture.
subacute phases, corrected QT (QTc) interval is 3. New ECG abnormalities (either ST segment
markedly prolonged (usually >500 ms), and this elevation or T-wave inversion) or modest ele-
is a potential risk factor for life-threatening vation in cardiac troponin.
arrhythmias such as torsades de pointes and 4. Absence of pheochromocytoma or
ventricular fibrillation that could require exter- myocarditis.
nal defibrillation [23]. Bradycardia, hypokale-
mia, hypomagnesemia, and the use of Patients were assigned this diagnosis when
antiarrhythmic drugs may favor arrhythmias they satisfied all these criteria.
when there is QTc prolongation, so they have to Japanese investigators have recently presented
be quickly corrected. The reversible nature of diagnostic guidelines [27].
the cardiomyopathy suggests that the use of sys- There is not a single shared diagnostic test so
tematic device implantation after a ventricular TC is currently a diagnosis of exclusion.
arrhythmias isnt recommended at the moment. Furthermore in addition to clinical features,
Apical thrombosis may occur during the acute EKG, and echocardiography abnormalities,
phase when the wall motion abnormalities are coronary angiography is mandatory to exclude
more evident, due to low blood flow within the coronary plaque rupture (culprit lesion) which
apical segment. This could become a potential would need angioplasty and stent implantation or
source of emboli especially when the resolu- multivessel epicardial coronary spasm. Most
tion of the apical akinesis/dyskinesis occurs patients have angiographically normal coronary
[24]. Prophylactic anticoagulation therapy arteries or mild atherosclerosis (considering
should be considered to prevent apical throm- advanced age and several coronary risk factors). In
bosis and embolic events until the resolution fact, many authors have attempted to find unique
of the apical ballooning. ECG criteria capable of distinguishing between
Hypotension occurs frequently and can result Takotsubo and STEMI patients during the acute
from LVOT obstruction associated with basal phase, but actually no criteria have been found,
hypercontractility, complicated by systolic and ECG keeps a limited diagnostic role [18, 20].
anterior movement of the mitral valve anterior Cardiac magnetic resonance is a suitable
leaflet and mitral regurgitation. method to establish TC diagnosis. It allows to
Cardiogenic shock and ventricular rupture are evaluate the global systolic function and the area
rare complications described in literature. of wall motion abnormalities; moreover, it shows
the area of myocardial edema, which is a typical
finding in TC, inflammation, and fibrosis.
Diagnosis Contrary to myocardial infarction or myocarditis,
there arent areas of delayed enhancement
There is yet no consensus on the diagnostic criteria because in TC there isnt an irreversible damage
for TC: researchers at the Mayo Clinic proposed [28, 29].

ERRNVPHGLFRVRUJ
50 G. Pongetti and A. Fabbrizioli

Treatment Heart J 27(13):15231529. doi:10.1093/eurheartj/


ehl032
4. Patel SM, Chokka RG, Prasad K, Prasad A (2013)
Medical treatment remains empirical. Distinctive clinical characteristics according to age
During the acute phase, therapy must be indi- and gender in apical ballooning syndrome (takot-
vidualized depending on the hemodynamic subo/stress cardiomyopathy): an analysis focusing on
men and young women. J Card Fail 19(5):306310.
situation.
doi:10.1016/j.cardfail.2013.03.007
In stable conditions, patients are often treated 5. Bybee KA, Kara T, Prasad A et al (2004) Systematic
with diuretics, angiotensin-converting enzyme review: transient left ventricular apical ballooning: a
(ACE) inhibitors, and -blockers to prevent syndrome that mimics ST-segment elevation myocar-
dial infarction. Ann Intern Med 141(11):858865
excessive sympathetic activation [13]. To reduce
6. Y-Hassan S (2014) Acute cardiac sympathetic dis-
the risk of thromboembolism, patients with ruption in the pathogenesis of the takotsubo syn-
marked apical ballooning should be treated with drome: a systematic review of the literature to date.
anticoagulant therapy until the contractility of the Cardiovasc Revasc Med 15(1):3542. doi:10.1016/j.
carrev.2013.09.008
apex is improved, unless there is a definite con-
7. Sadamatsu K, Tashiro H, Maehira N, Yamamoto K
traindication [24]. (2006) Coronary microvascular abnormality in the
In hemodynamically unstable patients, early reversible systolic dysfunction observed after noncardiac
administration of intra-aortic balloon pump disease. Jpn Circ J 64(10):789792. http://www.ncbi.
nlm.nih.gov/pubmed/11059622. Accessed 7 Jan 2015
counterpulsation should be considered in addi-
8. Galiuto L, De Caterina AR, Porfidia A et al (2010)
tion to cardiopulmonary circulatory support and Reversible coronary microvascular dysfunction:
continuous venovenous hemofiltration. There is a common pathogenetic mechanism in Apical
controversy on the use of cardiac stimulants Ballooning or Tako-Tsubo Syndrome. Eur Heart J
31(11):13191327. doi:10.1093/eurheartj/ehq039
because of increasing circulating catecholamines.
9. Virani SS, Khan AN, Mendoza CE, Ferreira AC, de
For patients with severe LV outflow tract obstruc- Marchena E (2007) Takotsubo cardiomyopathy, or
tion, treatment with -blocker and volume expan- broken-heart syndrome. Tex Heart Inst J 34(1):7679.
sion should be considered to reduce the http://www.pubmedcentral.nih.gov/articlerender.fcgi
?artid=1847940&tool=pmcentrez&rendertype=abstr
obstruction and increase the cardiac filling.
act. Accessed 11 Jan 2015
There is no consensus regarding long-term 10. Wittstein IS, Thiemann DR, Lima JAC et al (2005)
management of TC [30], although it could be rea- Neurohumoral features of myocardial stunning due to
sonable to treat patients with -blockers and ACE sudden emotional stress. N Engl J Med 352(6):539
548. doi:10.1056/NEJMoa043046
inhibitors during the ventricular recovery period;
11. Madhavan M, Borlaug BA, Lerman A, Rihal CS,
however, no data support the continuous use of Prasad A (2009) Stress hormone and circulating
these drugs for the prevention of recurrence and biomarker profile of apical ballooning syndrome
improvement of survival rate [21]. Physicians (Takotsubo cardiomyopathy): insights into the clini-
cal significance of B-type natriuretic peptide and tro-
may consider stopping those drugs after normal-
ponin levels. Heart 95(17):14361441. doi:10.1136/
ization of left ventricular function normalization. hrt.2009.170399
12. Y-Hassan S (2014) Pathophysiology of takotsubo syn-
drome: acute cardiac sympathetic disruption (ACSD)
syndrome. Cardiovasc Revasc Med 15(5):311312.
Bibliography doi:10.1016/j.carrev.2014.01.007
13. Sharkey SW, Windenburg DC, Lesser JR et al (2010)
1. Dote K, Sato H, Tateishi H, Uchida T, Ishihara M Natural history and expansive clinical profile of stress
(1991) [Myocardial stunning due to simultaneous (tako-tsubo) cardiomyopathy. J Am Coll Cardiol
multivessel coronary spasms: a review of 5 cases]. J 55(4):333341. doi:10.1016/j.jacc.2009.08.057
Cardiol 21(2):203214 14. Pelliccia F, Parodi G, Greco C et al (2015)
2. Akashi YJ, Goldstein DS, Barbaro G, Ueyama T (2008) Comorbidities frequency in takotsubo syndrome:
Takotsubo cardiomyopathy: a new form of acute, an international collaborative systematic review
reversible heart failure. Circulation 118(25):2754 including 1,109 patients. Am J Med. doi:10.1016/j.
2762. doi:10.1161/CIRCULATIONAHA.108.767012 amjmed.2015.01.016
3. Gianni M, Dentali F, Grandi AM, Sumner G, Hiralal 15. Kurisu S, Inoue I, Kawagoe T et al (2004) Time course
R, Lonn E (2006) Apical ballooning syndrome or of electrocardiographic changes in patients with tako-
takotsubo cardiomyopathy: a systematic review. Eur tsubo syndrome: comparison with acute myocardial

ERRNVPHGLFRVRUJ
5 Takotsubo Cardiomyopathy 51

infarction with minimal enzymatic release. Circ J 23. Madias C, Fitzgibbons TP, Alsheikh-Ali AA et al
68(1):7781 (2011) Acquired long QT syndrome from stress
16. Kosuge M, Kimura K (2014) Electrocardiographic cardiomyopathy is associated with ventricular
findings of takotsubo cardiomyopathy as compared arrhythmias and torsades de pointes. Heart Rhythm
with those of anterior acute myocardial infarc- 8(4):555561. doi:10.1016/j.hrthm.2010.12.012
tion. J Electrocardiol 47(5):684689. doi:10.1016/j. 24. Kurisu S, Inoue I, Kawagoe T et al (2011) Incidence
jelectrocard.2014.03.004 and treatment of left ventricular apical thrombosis in
17. Johnson NP, Chavez JF, Mosley WJ, Flaherty JD, Fox Tako-tsubo cardiomyopathy. Int J Cardiol 146(3):e58
JM (2013) Performance of electrocardiographic criteria e60. doi:10.1016/j.ijcard.2008.12.208
to differentiate Takotsubo cardiomyopathy from acute 25. Prasad A, Lerman A, Rihal CS (2008) Apical balloon-
anterior ST elevation myocardial infarction. Int J Cardiol ing syndrome (Tako-Tsubo or stress cardiomyopathy):
164(3):345348. doi:10.1016/j.ijcard.2011.07.029 a mimic of acute myocardial infarction. Am Heart J
18. Guerra F, Rrapaj E, Pongetti G et al (2013) Differences 155(3):408417. doi:10.1016/j.ahj.2007.11.008
and similarities of repolarization patterns during hos- 26. Prasad A (2007) Apical ballooning syndrome: an
pitalization for Takotsubo cardiomyopathy and acute important differential diagnosis of acute myocardial
coronary syndrome. Am J Cardiol 112(11):1720 infarction. Circulation 115(5):e56e59. doi:10.1161/
1724. doi:10.1016/j.amjcard.2013.07.036 CIRCULATIONAHA.106.669341
19. Mitsuma W, Kodama M, Ito M et al (2007) Serial elec- 27. Kawai S, Kitabatake A, Tomoike H (2007) Guidelines
trocardiographic findings in women with Takotsubo for diagnosis of takotsubo (ampulla) cardiomyopathy.
cardiomyopathy. Am J Cardiol 100(1):106109. Circ J 71(6):990992, http://www.ncbi.nlm.nih.gov/
doi:10.1016/j.amjcard.2007.02.062 pubmed/17527002. Accessed January 11, 2015
20. Duran-Cambra A, Sutil-Vega M, Fiol M et al (2014) 28. Kohan AA, Levy Yeyati E, De Stefano L et al
Systematic review of the electrocardiographic (2014) Usefulness of MRI in takotsubo cardio-
changes in the takotsubo syndrome. Ann Noninvasive myopathy: a review of the literature. Cardiovasc
Electrocardiol 20:16. doi:10.1111/anec.12228 Diagn Ther 4(2):138146. doi:10.3978/j.issn.2223-
21. Elesber AA, Prasad A, Lennon RJ, Wright RS, 3652.2013.10.03
Lerman A, Rihal CS (2007) Four-year recurrence 29. Eitel I, Behrendt F, Schindler K et al (2008)
rate and prognosis of the apical ballooning syndrome. Differential diagnosis of suspected apical balloon-
J Am Coll Cardiol 50(5):448452. doi:10.1016/j. ing syndrome using contrast-enhanced magnetic
jacc.2007.03.050 resonance imaging. Eur Heart J 29(21):26512659.
22. Kurisu S, Kihara Y (2012) Tako-tsubo cardiomy- doi:10.1093/eurheartj/ehn433
opathy: clinical presentation and underlying mecha- 30. Kurisu S, Kihara Y (2014) Clinical management of
nism. J Cardiol 60(6):429437. doi:10.1016/j. takotsubo cardiomyopathy. Circ J 78(7):15591566.
jjcc.2012.06.015 doi:10.1253/circj.CJ-14-0382

ERRNVPHGLFRVRUJ
Early Repolarization Pattern
6
Azzurra Fabbrizioli and Giulia Pongetti

6.1 Case Report Medical History and Cardiovascular


Risk Factors

A 38-year-old black man was admitted to Smoker (18 cigarettes/day).


the emergency medical service (EMS) for No familiar history of ischemic cardiovascular
some episodes of acute chest pain, started disease.
some days ago, exacerbated by deep inspi- About 6 months ago, the patient had been hospi-
ration and different movements and posi- talized in another hospital for an episode of
tions. The pain was intermittent, not acute chest pain developed after intense physi-
irradiated and not accompanied by other cal activity. The myocardial biomarkers werent
symptoms, and was independent of physi- elevated. He underwent coronary angiography,
cal exercise or food consumption. which showed normal coronary artery anatomy
The patient didnt refer flu or recent and absence of significant stenoses or thrombus.
infection neither traumatic events during He has been discharged with Cardioaspirin and
the previous weeks. beta blockers, but he stopped consumption
He has been admitted from EMS to our autonomously after some weeks.
department for further investigation. Migraine from some years.

Allergies

None

Medications

None

A. Fabbrizioli G. Pongetti (*) Vital Signs


Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche, Temperature: 36.7 C
Via Conca, 71, Ancona 60126, Italy
e-mail: giuliapongetti@gmail.com; Heart rate: 60 bpm
Azzurra.fab@gmail.com Blood pressure: 135/70 mmHg

Springer International Publishing Switzerland 2015 53


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_6

ERRNVPHGLFRVRUJ
54 A. Fabbrizioli and G. Pongetti

Respiratory rate: 16 breaths per minute


Oxygen saturation while breathing ambient Cardiomyopathy
air: 98 % Valvular disease
Glasgow Coma Scale (GCS): 15 Pulmonary causes
Pneumothorax
Pneumonia/pleuritis
Physical Examination Pulmonary infection
Pulmonary embolism
General: no acute distress, alert, awake, and Vascular causes
oriented. Well developed and well nourished. Aortic dissection
Head, eyes, ears, nose, throat: normocephalic, Aortic aneurysm
atraumatic, mucous membranes moist, extra- Gastrointestinal causes
ocular muscles intact, pupils equally round and Esophageal spasm
reactive to light and accommodation bilater- Esophagitis
ally, bilateral tympanic membrane intact, bilat- Peptic ulcer
eral sclera anicteric, no conjunctival injection.
Neck: supple, no jugular venous distention, no
lymphadenopathy, and no carotid bruit. A pulmonary disease is unlikely because the
Cardiovascular: Regular rate and rhythm, S1 chest pain was not accompanied by other sugges-
and S2 are normal; no murmurs, rubs or gal- tive symptoms like dyspnea, temperature, short-
lops; point of maximal intensity nondisplaced ness of breath, tachypnea, fatigue, tachycardia, or
and nonsustained; no hepatojugular reflux; hypotension and lung physical examination was
and capillary refill less than 2 s. normal. However, a chest x-ray was performed,
Lungs: no rales, rhonchus, or wheezes, no and it showed normal heart size and volumes, no
egophony, no alterations in tactil fremitus, and pulmonary congestion or pleural effusion, and no
normal percussion. signs of pneumothorax.
Abdomen: mild overweight, no pulsatile masses, A severe valvular disease is unlikely accord-
normal bowel sounds in all four quadrants, no ing to the absence of significant cardiac murmurs
high-pitched or tinkling sounds, resonant to per- during physical examination, the clinical features
cussion, soft, nondistended/nontender, no of chest pain, and the lack of other symptoms like
rebound or guarding, no costovertebral angle dyspnea, tachycardia, or fatigue. The chest pain
tenderness, no hepatosplenomegaly. could be also explained by gastrointestinal disor-
Extremities: no cyanosis or clubbing. Mild der like esophagitis or peptic ulcer, and a
peripheral edema. complete esophagogastroduodenoscopy (EGDS)
Neurological: cranial nerves II through XII could be required to exclude that possible cause
intact, no focal deficit. of symptoms.
Psychiatric: normal affect, no hallucinations, Owing to these observations, an EKG was
normal speech, no dysarthria. recorded, and an echocardiogram and routine
Skin: intact, no rashes, and no lesions. laboratory tests were performed.

What Are the Possible Causes EKG


of Acute Chest Pain in a Young Man?
Sinus rhythm, heart rate of 60 bpm, normal
atrioventricular conduction (PQ 200 ms), cardiac
Cardiac causes electrical axis +15, normal intrerventricular
Acute coronary syndrome (ACS) conduction, ST-segment elevation about 2 mm in
Myocarditis/pericarditis V2V4 and about 0.51 mm in V5V6, flat T
wave in D2, aVF. QTc 400 ms (Fig. 6.1).

ERRNVPHGLFRVRUJ
6 Early Repolarization Pattern 55

Fig. 6.1 EKG at rest while the patient was asymptomatic

Echocardiography (Not Shown) Routine Laboratory Tests

Normal both atria size (LA diameter Complete blood count: normal.
M-mode = 3.6 cm; area 4c = 18 cm2, RA area Cholesterol (total, HDL, LDL) and TG:
4c = 16 cm2). Normal size and wall thickness of normal.
the left ventricle (iLVEDV 42 ml/m2), which Hepatic function (GOT, GPT, -GT, ALP, total
shows a preserved systolic function (ejection bilirubin, direct and indirect): normal.
fraction with Simpsons method 0.67). Normal Thyroid function (TSH, FT3, FT4): normal.
size and global function of the right ventricle Renal function (creatinine, BUN): normal.
(TAPSE 23 mm). Electrolytes (Na+, K+, Ca++, Mg++, Cl):
Normal morphology of the cardiac valves; normal.
mild tricuspid regurgitation and normal systolic Fasting blood glucose: 102 m/dl
pressure gradient (PASP = 25 mmHg). Normal (5.67 mmol/L).
size of the aortic root (24 mm) and of the proxi- Troponin I-hs: 0.012 ng/ml (n.v < 0.055 ng/ml)
mal ascending aorta (28 mm); normal size of the Inflammation index: VES 35 mm/h (n.v < 27
aortic arch (26 mm) and the descending thoracic mm/h), CRP 0.2 mg/dl (n.v <0.6 ng/ml)
aorta (22 mm) in the explored segments too. The D-dimers: 150 ng/mL (n.v. < 280 ng/mL)
inferior vena cava has also a normal size (12 mm)
and physiological collapsing during inspiration. The echocardiographic findings are com-
Normal diastolic pattern without increased filling pletely normal, so we can rule out cardiomyopa-
pressure (E/A 0.9, E/E 6, E dec time 210 m/s). thies and any severe valvular disease as previously
Absence of pericardial effusion. supposed.
Conclusion: normal echocardiographic find- Similarly, the aortic size is normal, and there
ings for a 38-year-old man. are no signs of acute aortic dissection (intimal

ERRNVPHGLFRVRUJ
56 A. Fabbrizioli and G. Pongetti

flap) or intramural hematoma. Furthermore, the amplitude or disappear in the leads in which the
biomarkers are normal (D-dimer and troponin), ST elevation is observed (see Chap. 1).
and the symptoms referred by the patients are not The repolarization abnormalities in our patient
typical of acute aortic syndrome. Moreover, he were not typical of an acute pulmonary embolism,
didnt present any cardiovascular risk factor, with which is usually characterized by sinus tachycar-
the exception of the smoking habit. He did not dia, or other atrial arrhythmias, complete or incom-
have any familial history of arterial disease. plete right bundle branch block, pulmonary P
The EKG recorded while the patient was wave, ST-segment abnormalities, and/or inverted
asymptomatic showed ST-segment elevation in T wave in right precordial leads. Clinical signs and
precordial leads (V2V6), with positive T wave symptoms and the normal D-dimer level highly
and a concave pattern, without any reciprocal contribute to exclude pulmonary embolism.
ST-segment changes in the other leads. There were no peaked T waves (tended) in
right precordial leads as we could expect during
hyperkalemia; moreover, the laboratory tests
The Most Common Causes ruled out any electrolyte abnormality.
of ST-Segment Elevation The EKG pattern was also not typical of
in a Young Man Brugada type 1 pattern (ST-segment elevation
2 mm descending with an upward convexity,
inverted T wave in at least one right precordial
Acute coronary syndrome with ST- lead); the clinical history was also negative for
segment elevation (ACS-STEMI) with syncope or tachyarrhythmic episodes.
or without possible coronary spasm Acute pericarditis could be more likely, espe-
Myocarditis/pericarditis cially according to chest pain, early recurrence,
Early repolarization pattern and its characteristics. Usually in this pathology,
Acute pulmonary embolism the ST-segment elevation is diffuse in all the leads
Hyperkalemia and little pronounced; QRS complex generally
Brugada syndrome shows a reduction in amplitude, and PR can be
Acute aortic dissection depressed in many leads. In myocarditis, the most
common EKG finding is diffuse T wave inver-
sion, whereas ST-segment displacement is less
common. The absence of flu during the days/
The first hypothesis we have to exclude is weeks before, the normal values of the logistic
ACS-STEMI. Even though there was an index at the laboratory tests together with the
ST-segment elevation in more than one precor- absence of pericardial effusion or wall motion
dial lead, it should be 0.25 mV when measured abnormalities made those hypotheses unlikely.
at the J point in a man under 40 years. Moreover,
the upward concavity is not typical of STEMI;
the limb leads showed an essentially normal Final Diagnosis
repolarization pattern, and there were no recipro-
cal EKG changes. By considering also normal According to the clinical history, echocardio-
echocardiography and laboratory results and the graphic findings, and laboratory tests, we con-
clinical history (previous recent coronary angiog- cluded that the EKG abnormalities were due to
raphy was completely normal), we excluded the an early repolarization pattern.
ACS hypothesis. An exercise testing was performed showing a
The EKG pattern in possible coronary spasms complete normalization of the ST-segment eleva-
is also different because in addition to the tion during exercise with a slow and progressive
ST-segment modification the R wave may return during recovery (Fig. 6.2). That confirmed
become taller and the S wave may decrease in our diagnostic hypothesis.

ERRNVPHGLFRVRUJ
6 Early Repolarization Pattern 57

Fig. 6.2 EKG recorded during exercise testing showing the ER pattern at rest (a), normalization of the ST-segment
elevation during exercise (b); slow return to the baseline and then progressive return of the ER pattern (c, d)

ERRNVPHGLFRVRUJ
58 A. Fabbrizioli and G. Pongetti

Fig. 6.2 (continued)

ERRNVPHGLFRVRUJ
6 Early Repolarization Pattern 59

This EKG pattern is more frequent in black Epidemiology


men and does not require any pharmacological
treatment. ER affects the general population from 6 to 13 %
There is no common agreement if different [24] and is more frequent in men (75 %) and
ER patterns (commonly separated in benign and young athletes [4] and may disappear with aging
malignant forms, see below) are associated with a [5]. It is more common in black people [2, 5].
higher risk of SCD. Nevertheless, our patients
EKG showed a benign pattern because the
ST-segment elevation was present only in precor- Electrocardiographic Features
dial leads, with upper concavity, and the T waves
were large and positive. There was a J wave in The ER EKG pattern is characterized by J point
some leads (V4V5V6), but it was little and did elevation 1 mm in at least two contiguous leads
not show significant modifications. The complete (the J point is the junction between the end of the
repolarization pattern remained stable during the QRS complex and the ST segment) often associ-
hospitalization period without significant changes ated with J wave, also called Osborn wave [6],
at rest. Moreover, the patient did not present pro- that is a rare deflection similar to a P wave, situ-
arrhythmic conditions and did not refer familiar ated at the end of the QRS complex and usually
history of SCD. So he could be safely visible both in precordial and limb leads. It is a
discharged. typical finding during hypothermia, but may be
However, for completing diagnostic evalua- present also during hypercalcemia or in Brugada
tion, we decided to perform an EGDS, which syndrome [5]. His amplitude can increase with
showed a grade B gastroesophageal reflux dis- vagal tone [7, 8]. Actually, the presence of
ease (Los Angeles classification) with some ST-segment elevation is not necessary for ER
mucosal breaks that did not extend between the EKG diagnosis. Anyway, the ST-segment eleva-
tops of the two mucosal folds. Proton pump tion is characterized by upward concavity, more
inhibitor therapy has been prescribed, and a grad- frequently in precordial leads without any recip-
ual decrease of the chest pain was observed dur- rocal depression. It is usually <2 mm
ing the following days. Finally, the patient was (rarely > 5mm) in precordial leads (usually V4
discharged suggesting an additional gastroenter- V6) but may also be present in inferior or lateral
ological reassessment in the next weeks after full leads. In some cases, the J point elevation is asso-
therapy. ciated with horizontal/descending ST segment.
QT interval is always normal. The ER pattern
is intermittent and therefore not constant in all
6.2 Early Repolarization the recorded EKG.

The early repolarization (ER) pattern is a par-


ticular EKG aspect first described in 1936 by Pathogenesis
Shipley and Hallaran as a normal variant of
repolarization [1]. Subsequently, a new classi- The electrophysiological mechanism underlying
fication proposed by Haissaguerre et al. [2] the ER pattern is the presence of a voltage gradi-
defined ER pattern as the J point elevation ent between the epicardium and endocardium.
associated with a slurring (deflection in the Some epicardial regions have a faster repolar-
R-wave descent) or a notching (positive deflec- ization during phase 1 of the action potential than
tion with a secondary r wave) on the terminal the endocardial regions, due to an increased repo-
portion of QRS, called J wave, associated with larizing current or a decreased depolarizing cur-
ST-segment elevation or a horizontal/descend- rent. The more common ionic current involved in
ing ST segment. this modifications seems to be the K current

ERRNVPHGLFRVRUJ
60 A. Fabbrizioli and G. Pongetti

(Ito), which starts the repolarization phase [10, BRUGADA SYNDROME: in Brugada type 1
11]. In this way, there is a voltage gradient pattern, there is typical coved J wave- ST seg-
between the epicardium and endocardium which ment elevation 2 mV and an inverted T wave
produces the J wave on the surface EKG. When in at least one right precordial lead [11]. The J
this voltage gradient occurs later in the action wave may be accentuated by increased vagal
potential, it is responsible for the ST-segment tone and fever and may be unmasked by the
elevation. class I antiarrhythmic drugs (flecainide, ajma-
line, and procainamide) [11, 12].

Differential Diagnosis of ST-Segment


Elevation Exercise Test

One of the key points about ER is to discriminate Exercise or isoproterenol test may cause
this benign condition from other disorders regression of the ST-segment elevation, pre-
presenting ST-segment elevation and requiring sumably as a result of a diminished difference
specific medical treatment. between ventricular action potential durations
during sympathetic stimulations. This phe-
ACS-STEMI: the diagnosis of ACS-STEMI is nomenon may be clinically useful to diagnose
clinical and electrocardiographic. The typical ER pattern in patients with low suspect of
ischemic EKG changes usually show acute myocardial infarction or acute pericardi-
ST-segment elevation 0.25 mV in men below tis [9].
the age of 40 years and 0.2 mV in men over
40 years or 0.15 mV in women, when mea-
sured at the J point in at least two contiguous Clinical Features and Risk
leads, and reciprocal changes. The lack of Stratications
reciprocal changes (without changes except
aVR) and the isolated ST-segment elevation The main question about early repolarization is
reduce the positive predicted value for STEMI; its possible relation to an arrhythmic risk eventu-
anyway, it can be useful in the comparison ally causing sudden cardiac death (SCD) in
with previous EKG and the evaluation of young subjects.
symptoms, cardiovascular history, echocar- A first report in 1984 showed an association
diography, cardiac biomarkers, and serial between an ER pattern and ventricular fibrillation
EKG monitoring. (VF) [13]. More recently in 2008, Haissaguerre
Acute Pericarditis: the typical EKG modifica- et al. demonstrated in a large study that ER pat-
tions during an acute pericarditis are diffuse tern is six times more common in patients who
concave ST-segment elevation with positive T suffered previous VF than in matched controls;
waves and no reciprocal ST-segment depression moreover, these patients have more likely an
(except in aVR and V1). In about 50 % of cases, increased risk of recurrent SCD [2].
PR segment depression may be visible which In particular, it seems that the ER pattern in
usually occurs in all leads except aVR and V1, inferior leads is the one with the most increased
and it is pathognomonic of acute pericarditis. arrhythmic risk of SCD, especially when the J
To achieve the right diagnosis of acute point is >2 mm (three-fold risk increased), as
pericarditis, the comparison with previous demonstrated in a control study with over 10,000
EKG is useful, considering symptoms, pres- patients, followed for 30 years [4].
ence of pericardial rubbings, pericardial effu- In 2011, Tikkanen et al. analyzed the different
sion, positive cardiac biomarkers, and serial electrocardiographic phenotypes in ER and their
EKG monitoring to evaluate the typical time long-term outcome, distinguishing between
modification. benign and malignant forms. In the benign

ERRNVPHGLFRVRUJ
6 Early Repolarization Pattern 61

pattern, the ST segment and the T waves have a References


temporary stability, and T waves are generally
large, positive, and concordant with the 1. Shipley R, Hallaran W (1936) The four lead electro-
cardiogram in 200 normal men and women. Am Heart
ST-segment deviation from V2 to V4. In this J 11:325345
patient, the ER pattern is not a mortality risk fac- 2. Haissaguerre M, Derval N, Sacher F et al (2008)
tor, also if situated in inferior leads. Sudden cardiac arrest associated with early repolar-
On the other hand, the malignant pattern is ization. N Engl J Med 358:20162023
3. Klatsky AL, Oehm R, Cooper RA et al (2003) The
characterized by a notched J wave, a horizon- early repolarization normal variant electrocardiogram:
tal/descending ST elevation, and T waves dis- correlates and consequences. Am J Med. 115(3):
cordant to their respective ST-segment 171177.
deviation. The ST/T waves morphology may 4. Tikkanen JT, Anttonen O, Junttila MJ et al (2009)
Long-term outcome associated with early repolariza-
change in 24 h, particularly during the night. tion on electrocardiography. N Engl J Med 361:
This pattern seems to be associated with an 25292537
increased risk of all-cause mortality [14]. 5. Kambara H, Phillips J (1976) Long-term evaluation of
These data have been confirmed by a recent early repolarization syndrome (normal variant RS-T
segment elevation). Am J Cardiol 38:157161
meta-analysis where the ER pattern associated 6. Trevino A, Raza B, Beller BM (1971) The character-
with high SCD risk is present in <0.3 % of the istic electrocardiogram of accidental hypothermia.
general population [4, 15]. It is a common Arch Intern Med 127:470475
agreement that large and dynamic J waves are 7. Surawicz B, Knilans TK (2008) Chous electrocardi-
ography in clinical practice, 6th edn. Saunders/
more prevalent in patients with idiopathic Elsevier, Philadelphia, p 20
VF. Anyway, the risk of VF in general popula- 8. Junttila MJ, Sager SJ, Freiser M, McGonagle S,
tion is 0.03 % (3/10,000), and the higher risk Castellanos A, Myerburg RJ (2011) Inferolateral early
ER pattern may increase ten times SCD repolarization in athletes. J Interv Card Electrophysiol
31:3338
(30/10,000) [16]. There are, moreover, some 9. Goldman M (1953) RS-T segment elevation in
proarrhythmic conditions like acute ischemia, mid-and left precordial leads as a normal variant. Am
electrolytic disturbances, or depressed ventric- Heart J 46:817820
ular function which may trigger VF and unmask 10. Yan GX, Antzelevitch C (1996) Cellular basis for the
electrocardiographic J wave. Circulation 93:372379
an ER malignant pattern [9, 17]. 11. Antzelevitch C, Brugada P, Borggrefe M, Brugada J,
There is no consensus regarding the pharma- Brugada R, Corrado D, Gussak I, LeMarec H,
cological therapy for VF in these patients; amio- Nademanee K, Perez Riera AR, Shimizu W, Schulze-
darone or quinidine have been empirically used Bahr E, Tan H, Wilde A (2005) Brugada syndrome:
report of the second consensus conference: endorsed
in a clinical context [18]. by the Heart Rhythm Society and the European Heart
For patients resuscitated from SCD, it is Rhythm Association. Circulation 111:659670
mandatory to implant an ICD (implantable 12. Veltmann C, Schimpf R, Echternach C, Eckardt L,
cardioverter-defibrillator) whenever all the pos- Kuschyk J, Streitner F, Spehl S, Borggrefe M, Wolpert
C (2006) A prospective study on spontaneous fluctua-
sible reversible proarrhythmic triggers have been tions between diagnostic and non-diagnostic ECGs in
excluded [19]. Brugada syndrome: implications for correct pheno-
There are no clear recommendations for pri- typing and risk stratification. Eur Heart J
mary prevention in asymptomatic patients, and at 27:25442552
13. Otto CM, Tauxe RV, Cobb LA et al (1984) Ventricular
the moment, any pharmacological therapy and/or fibrillation causes sudden death in Southeast Asian
stopping physical activity is not recommended immigrants. Ann Intern Med 101:4547
even at competitive level. 14. Tikkanen JT, Junttila MJ, Anttonen O, Aro AL,
Patients with syncope and an ER pattern at Luttinen S, Kerola T, Sager SJ, Rissanen HA,
Myerburg RJ, Reunanen A, Huikuri HV (2011) Early
rest EKG should undergo common diagnostic repolarization: electrocardiographic phenotypes asso-
evaluation usually done for unexplained loss of ciated with favorable long-term outcome. Circulation
consciousness. 123:26662673
Currently, the ER pattern is still an object of 15. Wu SH, Lin XX, Cheng YJ, Qiang CC, Zhang J
(2013) Early repolarization pattern and risk for
study.

ERRNVPHGLFRVRUJ
62 A. Fabbrizioli and G. Pongetti

arrhythmia death: a meta-analysis. J Am Coll Cardiol syndrome. Pacing Clin Electrophysiol 35:e234e238.
61(6):645650. doi:10.1016/j.jacc.2012.11.023. doi:10.1111/j.1540-8159.2012.03460.x
Epub 2013 Jan 2 18. Kalla H, Yan GX, Marinchak R (2000) Ventricular
16. Rosso R, Kogan E, Belhassen B et al (2008) J-point fibrillation in a patient with prominent J (Osborn)
elevation in survivors of primary ventricular fibrilla- waves and ST segment elevation in the inferior elec-
tion and matched control subjects incidence and trocardiographic leads: a Brugada syndrome variant?
clinical significance. J Am Coll Cardiol 52: J Cardiovasc Electrophysiol 11:9598
12311238 19. The ACC/AHA/HRS (2008) guidelines for device-
17. Myojo T, Sato N, Nimura A, Matsuo A, Taniguchi O, based therapy of cardiac rhythm abnormalities: their
Nakamura H et al (2012) Recurrent ventricular fibril- relevance to the cardiologist, internist and family phy-
lation related to hypokalemia in early repolarization sician. J Invasive Cardiol 2009;21(5):234237.

ERRNVPHGLFRVRUJ
Part III
Heart Failure and Resynchronization
Therapy

ERRNVPHGLFRVRUJ
Acute Heart Failure
and Pulmonary Edema 7
Andrea Romandini and Simone Maffei

7.1 Case Report Social History

He used to smoke about 30 cigarettes/day some


A 64-year-old man was admitted to the years ago.
emergency room for acute dyspnea. The
patient reported fatigue and dyspnea for
minimum efforts during the days before. He Home Medications
reported no recent pathological findings.
Rosuvastatin 20 mg at 9.00 p.m., metformin
850 mg at 12.00 a.m and at 8.00 p.m.

Medical History and Cardiovascular


Risk Factors Vital Signs

Type 2 diabetes mellitus. Temperature: 36.5 C


2013: access to the emergency room for asthe- Heart rate: 126 beats per minutes
nia and dizziness; on that occasion, high levels Blood pressure: 170/100 mmHg
of glucose were found. Respiratory rate: 22/min
Oxygen saturation while breathing in ambient
air: 92 %
Allergies

None Physical Examination

A. Romandini (*) General: alert, awake, and oriented but slightly


Cardiology department Ospedali Riuniti di Ancona, agitated
Via Conca, 71, Ancona 60126, Italy Neck: slight jugular venous distention, no lymph-
e-mail: romandini.andrea@hotmail.it
adenopathy, and no carotid murmur
S. Maffei Cardiovascular: early diastolic gallop with S3
Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche,
and, systolic murmur 2/6 at the mesocardium
Via Conca, 71, Ancona 60126, Italy without radiation to the armpit, neck, or
e-mail: maffo-85@hotmail.it carotid vessels

Springer International Publishing Switzerland 2015 65


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_7

ERRNVPHGLFRVRUJ
66 A. Romandini and S. Maffei

Lungs: breath sounds diffusely decreased, in par- Chest X-Ray


ticular at the lung bases, and rales up to
medium shots bilaterally X-ray showed signs compatible with acute pul-
Abdomen: plain, no pulsatile masses, normal monary edema.
bowel sounds in all four quadrants, no high-
pitched or tinkling sounds, resonant to percus-
sion, soft, nondistended/nontender, no What Are the Possible
rebound or guarding, no costovertebral angle Causes of Worsening Acute
tenderness, hepatomegaly up to 2 cm from the Dyspnea and Orthopnea
costal margin, no splenomegaly, and Giordano in This Patient?
and Murphy signs negative
Neurological: negative cerebellar test, cranial There are several causes that may acutely unbal-
nerve intact, no focal deficit, and reflections ance the left ventricle function. There may be
normoexcitable cardiac, extracardiac, or iatrogenic triggers; how-
Psychiatric: normal ever, dyspnea can also be related to diseases
Skin: pale, cold, and sweaty with cyanosis of the affecting primarily the lungs. These are the pos-
extremities

Acute myocardial infarction


Routine Laboratory Tests Hypertensive crisis
Arrhythmias
Complete blood count: leukocytosis with neu- Acute myopericarditis
trophilia (WBC 10.760/mmc, 91.20 % neutro- Lung diseases (bacterial pneumonia)
phils), hemoglobin 13.5 g/dl, and platelets ARDS
248,000/mmc Pulmonary embolism
Inflammatory markers: ESR 29 mm/h and Valvular disease (acute mitral
CRP 0.6 mg/dl regurgitation)
Hepatic function: GOT normal, GPT with
slight increase (61 U/l), -GT 121 U/l, and
ALP, total bilirubin (direct and indirect), and
coagulation normal sible causes in this patient:
Normal renal function (creatinine 0.82 mg/dl, The patient was apyretic and CRP was nega-
BUN 38 mg/dl, eGFR 92.8 ml/min/1.73 m2) tive, although there was a mild leukocytosis with
Electrolytes (Na+, K+, Ca++, Mg++, Cl): normal neutrophilia.
Fasting blood glucose: 179 mg/dl
Myocardial necrosis markers: normal CK-MB
and Hs-TnI 0.059 ng/ml (n.v. 00.055) EKG
BNP: 744 pg/ml
Thyroid function: normal TSH and fT4 and EKG was negative for ischemic alterations, and
fT3 2.10 pg/ml (n.v. 2.24.2 pg/ml) hs-troponin I was minimally altered with normal
CK-MB.
The blood gas analysis performed in ambient According to these data, acute myocardial
air showed pH = 7.41, pO2 = 58 mmHg, infarction, bacterial pneumonia, arrhythmias, and
pCO2 = 40 mmHg, and p/F = 276. ECG showed a acute myopericarditis were initially excluded as
sinus tachycardia (heart rate was 126 beats per possible causes of dyspnea. The patient was then
minutes), normal atrioventricular and intraven- treated with furosemide bolus and infusion of
tricular conduction, and nonspecific alterations nitroglycerin to reduce high blood pressure ini-
of ventricular repolarization. tially encountered. A CPAP (continuous positive

ERRNVPHGLFRVRUJ
7 Acute Heart Failure and Pulmonary Edema 67

airway pressure) was positioned and was set a Therapy


FiO2 of 50 % and PEEP (positive end-expiratory
pressure) of 10 cmH2O. The patient showed After resolution of the acute phase, a specific
marked improvement in dyspnea, and blood gas therapy for heart failure was given to the patient
analysis showed a significant increase in pO2 (ACEI, beta-blockers, potassium sparing), and
(pO2 = 58 mmHg 139 mmHg). This favorable in a few days, a good cardiovascular compensa-
response to treatment could make us exclude a tion was restored (demonstrated also by the
noncardiogenic acute pulmonary edema (ARDS), gradual reduction of BNP: 744 pg/ml 250 pg/
which is characterized by severe hypoxemia ml). The absence of a compatible clinical his-
refractory to increased FiO2 and reduced lung tory, the slightest movement of hs-troponin
compliance. (0.059 ng/ml 0.061 ng/ml 0.033 ng/ml),
and the constant negativity of inflammatory
markers made the myopericarditis an unlikely
Echocardiography cause of the acute pulmonary edema. We
thought the hypertensive crisis was the cause of
An echocardiography was also recorded: tri- acute heart failure.
cuspid aortic valve with normal valve opening;
standard size of the aortic root and ascending
aorta with mild ectasia of the aortic arch. Mild Final Diagnosis
left atrial enlargement (44 ml/m2). Normal right
ventricle size and systolic function (TAPSE The final diagnosis was hypertensive crisis com-
22 mm). Slightly dilated left ventricle with plicated by acute pulmonary edema in patients
severe reduction of systolic global function (EF with hypokinetic-dilated cardiomyopathy with-
25 %) and diffuse hypokinesia; modest pericar- out hemodynamically significant stenosis After
dial effusion more evident close to the right 2 months, the patient was asymptomatic and in
sections and conditioning initial atrial collapse. good hemodynamic compensation. A new echo-
No significant gradients. Mild mitral insuffi- cardiogram showed the absence of pericardial
ciency, mild tricuspid regurgitation with high effusion and an improvement in ejection fraction
pulmonary arterial pressure (60 mmHg). (EF: 25 % 39 %).
Inferior vena cava dilated and hypo-collaps-
ing. Echocardiogram ruled out the presence of
significant valvular disease and dysfunction 7.2 Denition and Clinical
and dilatation of the right sections but showed Classication of Acute Heart
severe left ventricular dysfunction associated Failure Syndromes (AHFSs)
with mild pericardial effusion. At this diagnos-
tictherapeutic point, an underlying ischemic According to the latest European Society of
heart disease or a myopericarditis could not be Cardiology (ESC) guidelines, heart failure (HF)
excluded. can be defined as an anomaly of cardiac structure
or function impairing hearts ability to deliver
oxygen at a rate commensurate with the require-
Coronary Angiography ments of the metabolizing tissues despite normal
filling pressures or at the expense of increased fill-
An invasive coronary angiography documented ing pressures [1]. The clinical manifestations of
the absence of hemodynamically significant ste- heart failure result from the impaired forward car-
nosis, and an eco-color-Doppler of the lower diac output (forward failure) and/or elevated
limbs excluded the presence of a deep vein venous pressure related (backward failure) to the
thrombosis (to rule out thromboembolic pulmo- failing heart. The clinical syndrome of HF may
nary disease). result from disorders of any aspect of cardiac

ERRNVPHGLFRVRUJ
68 A. Romandini and S. Maffei

function including pericardial disease, myocar- gradual body weight gain [8]. Volume over-
dial disease, endocardial disease, valvular heart load is only one of the possible hemodynamic
disease, arrhythmias and conduction disorders, perturbations that may explain the elevated
congenital heart disease, high output state, or vol- filling pressure. Additional pathophysiologic
ume overload state. These patients may present mechanisms are afterload mismatch (increased
with reduced or preserved left ventricular (LV) afterload) and abnormal end-ventricular dia-
systolic function. LV ejection fraction (LVEF) is stolic pressure (related to ventricular diastolic
considered important in classification of patients dysfunction/abnormal compliance and valvular
with HF because of differing patient prognosis regurgitation). Pulmonary congestion, with or
and response to therapies and because most clini- without associated systemic signs, may be the
cal trials selected patients based on LV ejection results of two different pathways. The cardiac
fraction (LVEF). For this reason, patients with HF (central) pathway is the mechanism by which a
are broadly categorized (with some difference low cardiac output (usually an acute decrease
among major international guidelines) in HF with induced by a variety of precipitant mecha-
preserved EF (normal or mildly reduced LVEF nisms including ischemia, arrhythmia, inflam-
and LV not dilated) and HF with reduced EF matory activation or progression of underlying
(LVEF usually 3540 %). An episode of acute HF process induced by progressive myocardial
heart failure or acute heart failure syndrome dysfunction) leads to a further neurohormonal
(AHFS) is usually defined as a rapid or gradual activation, lower renal perfusion (cardiorenal
onset (or change) of symptoms and signs of heart syndrome), and fluid accumulation with sys-
failure (HF) requiring immediate medical atten- temic congestion (overload fluid retention) [9,
tion (unplanned hospitalization or office room 10]. The vascular (peripheral) pathway is related
visit). Patients with AHFS are generally classified to increased vascular stiffness/resistance with
into those presenting with HF for the first time (de acute afterload mismatch impairing systolic per-
novo AHF) and those with worsening chronic HF. formance and resulting in redistribution of fluid
from systemic to pulmonary circulation rather
than in general fluid retention [2, 9, 10]. Venous
Pathophysiology of AHFS volume mobilization of the splanchnic circula-
tion has also been proposed as complementary
Regardless of the underlying cause or superim- mechanism [11]. Although in most cases both
posed precipitating factor, pulmonary and sys- pathways are active during an AHF event, the
temic congestion (due to increased left- and/or magnitude of one pathway may predominate in
right-side heart filling pressures), with or without each patient and is usually suspected according
low cardiac output, is the unifying finding in the to AHF initial clinical presentation.
broad spectrum of hemodynamic models in
AHFS [2, 3]. Congestion and not low cardiac
output is the main cause for AHFS [47]. Precipitants of Acute Heart Failure
High left-side filling pressure results in pul-
monary hypertension (pulmonary congestion) Approximately 80 % of acute decompensated
with increased pulmonary capillary wedge pres- heart failure (ADHF) patients have a worsening
sure (PCWP) that preceded the subsequent clini- of chronic heart failure. In such patients with pre-
cal congestion with pulmonary interstitial and existing HF, one or more identifiable exacerbat-
alveolar edema. ing factors not necessarily related to the evolution
High right-side filling pressure results in of the underlying HF disease can be often identi-
systemic venous hypertension (systemic con- fied (up to 70 %) (Table 7.1). Detection and treat-
gestion) with increased central venous pres- ment of precipitating factors is necessary both for
sure (CVP) leading to jugular vein distension acute management of an episode of AHF and pre-
and often subsequent peripheral edema and vention of its recurrence.

ERRNVPHGLFRVRUJ
7 Acute Heart Failure and Pulmonary Edema 69

Table 7.1 Precipitants of acute heart failure Table 7.2 Common physical findings in HF
Precipitants and causes of acute heart failure syndromes Possible physical findings in heart failure
(AHFSs) More specific
Rapid deterioration Gradual deterioration Third heart sound (S3)
Rapid arrhythmias Arrhythmias Jugular venous distension
Acute coronary syndromes Infections (including Hepatojugular reflux
(ACSs) endocarditis)
Laterally displaced apical impulse
Mechanical complications Exacerbation of COPD/
Cardiac murmurs
of ACS asthma
Less specific
Acute pulmonary Anemia
embolism Pulmonary rales
Hypertensive crisis Renal failure Decreased breath sounds at lung bases (pleural
effusion)
Cardiac tamponade Use of drugs that
increase Na+ retention Peripheral edema and ascites
(steroids, NSAIDs, etc.) Hepatomegaly
Additional acute CV Nonadherence with HF Tachycardia
disorders (acute aortic medications or diet Tachypnea
dissection, myocarditis) regimen (including Irregular rhythm (ectopic beats or atrial fibrillation)
alcohol abuse)
Muscle wasting (cachexia)
Peripartum Poor controlled
cardiomyopathy hypertension Data from McMurray et al. [1]
Acute mechanical valve Endocrine abnormalities
dysfunction
On the basis of typical clinical and hemody-
Modified and reproduced with permission from McMurray namic characteristics, AHF patients may present
et al. [1]
with one of several distinct clinical profiles con-
sidering that some overlap between groups may
Clinical Proles at Presentation exist [8]. The main clinical profiles and relative
features are summarized in Table 7.3.
The two major classes of symptoms in HF are Another classification scheme has been previ-
those due to volume overload (dyspnea, orthop- ously proposed (Forrester classification) and is
nea, paroxysmal nocturnal dyspnea, cough, gas- based on the severity of disease at presentation
trointestinal symptoms) and those due to a rather than on the cause of HF [12, 13]. It is a
reduction in CO (fatigue and weakness). The simple strategy to classify patients into specific
most common are dyspnea and fatigue. Dyspnea hemodynamic profiles that may be helpful to
(at exertion or at rest) is related to complex physi- guide the initial management strategy.
ological mechanisms involving both pulmonary Accordingly, a patient presenting with AHFS
venous congestion and a buildup of lactic acid by may be classified into one of the four specific
working muscle increasing the ventilatory hemodynamic profiles based on the absence or
response to exercise. On the other hand, low car- presence of signs of congestion (wet or dry) and
diac output state often results in fatigue and the adequacy of peripheral perfusion (warm or
weakness due to reduced skeletal muscle perfu- cold): warm and dry, warm and wet, cold and dry,
sion or atrophy. Elevated systemic venous pres- and cold and wet.
sures like those occurring in volume overload or
right ventricular dysfunction states are responsi-
ble for abdominal discomfort (liver congestion Clinical Assessment and Diagnosis
and abdominal ascites), anorexia, and peripheral of AHF
edema. Common physical findings are summa-
rized in Table 7.2. The most common clinical Traditionally, the diagnosis of HF is a clinical
findings are dyspnea (approximately 90 %), rales, diagnosis combining characteristic symptoms
and peripheral edema (65 %). with physical findings, and still today no single

ERRNVPHGLFRVRUJ
70 A. Romandini and S. Maffei

Table 7.3 Common clinical profiles in AHFS


Clinical profiles Common clinical features
Hypertensive In many patients, LVEF is preserved (normal CI); relative rapid onset; prevalent pulmonary
(SBP oversystemic congestion
>160 mmHg)
Normal or Usual in patients with worsening HF (normal or low CI); gradual onset; mild-to-moderate
high-normal systemic congestion associated
blood pressure
Low blood Usual in patients with advanced or end-stage HF disease (severe reduced LVEF with low CI);
pressure (SBP many patients may have a low cardiac output with signs of organ hypoperfusion; intravascular
<90 mmHg) depletion due to aggressive diuretic therapy may play a role (ensure preload optimization);
gradual onset
Flash pulmonary Related to sudden rise in left-side filling pressure (with low or normal CI) induced by acute
edema precipitating factors (e.g., hypertensive crisis); rapid onset with respiratory distress
Cardiogenic Often complicated acute life-threatening condition inducing low CO state (acute MI, fulminant
shock myocarditis, acute valve dysfunction); rapid onset usually; evidence of signs of hypoperfusion
(altered mental status, cold skin, oliguria/anuria, etc.)
ACS and AHFS May be present in up to 25 % patients with ACS; rapid or gradual onset (depending on severity
of underlying LV dysfunction); possible resolution after efficacious myocardial
revascularization
Isolated Related to increased right-side filling pressure due to RV dysfunction or pulmonary
right-sided HF hypertension (if rapid-onset CI is usually low); onset may be rapid (e.g., RV infarct, acute
pulmonary embolism) or gradual (e.g., cor pulmonale, primary pulmonary hypertension,
cardiac mass/tumors); evidence of systemic (peripheral edema, hepatomegaly) over pulmonary
congestion
Perioperative Usually related to volume overload or myocardial injury during cardiac surgery; rapid or
AHFS gradual onset
High-output Related to conditions associated with high CI (septic shock, anemia, thyrotoxicosis, Pagets
failure disease, pregnancy); patients usually present tachycardia, warm extremities, variable degree of
pulmonary and systemic congestion
Data from Gheorghiade et al. [2]

tests can absolutely establish its presence or sions. Radiographic evidence of signs of pulmo-
absence. Unfortunately, signs and symptoms of nary congestion in a patient with dyspnea makes
HF often overlap with those of other common the diagnosis of heart failure more likely; how-
medical conditions (especially with chronic lung ever, the absence of radiographic pulmonary con-
disease), and those more specific are also less gestion does not exclude diagnosis of
common (like orthopnea and paroxysmal noctur- AHF. Patients with chronic heart failure, despite
nal dyspnea) or less reproducible (third heart AHF symptoms and elevated PCWP, may have
sound and jugular venous distension) so that sev- few radiographic signs because of enhanced lym-
eral ancillary tests, also contributing to determine phatic drainage. Electrocardiography (ECG) is
mechanisms underlying the AHF, are usually not useful for diagnosis but offers possible clues
needed to support the clinical diagnosis of AHF. to identify both specific treatable precipitating
A chest radiography should be performed ini- factors of AHF (acute myocardial ischemia and
tially because it may aid in diagnosis of HF as arrhythmias) and also possible etiology of HF
well as in ruling out other differential diagnoses (e.g., Q wave in ischemic cardiomyopathy).
(e.g., pneumonia). Findings suggestive of HF Laboratory tests (blood chemistry and hema-
include cardiomegaly (cardiac-to-thoracic width tological tests) are useful to guide initial therapy,
ratio above 50 %), upper zone vascular redistri- to detect reversible cause of HF (e.g., hypocalce-
bution (cephalization), interstitial edema with mia, thyroid dysfunction) and comorbidities
Kerley B-lines, alveolar edema, and pleural effu- (anemia), and to obtain prognostic information.

ERRNVPHGLFRVRUJ
7 Acute Heart Failure and Pulmonary Edema 71

Serial monitoring of myocardial necrosis bio- function, and pulmonary artery pressure). The
markers (troponin) is recommended initially for TD-derived E/Ea parameter is being used to non-
diagnostic (exclude acute coronary syndrome) invasively estimate LV filling pressures. In addi-
and prognostic purpose. Troponin elevation in tion, especially for those with hypotensive AHFS,
acute HF does not necessarily indicate the pres- echocardiographic assessment of inferior vena
ence of an acute coronary syndrome. A signifi- cava (IVC) diameter and its respiratory variation
cant number of patients with AHFS have aid to determine the patient volume status.
increased levels of troponin as a result of myocar-
dial injury during AHF episode resulting from
ischemic injury and myocyte apoptosis. Such tro- AHFS Management
ponin elevation is associated, however, with poor
long-term prognosis. The main goal of short-term therapy (hours to
Measurement of natriuretic peptide (NP) lev- days) for AHFS has been to achieve the lowest
els is helpful especially when the diagnosis is in left ventricular filling pressure possible without
question. Natriuretic peptides (BNP and decreasing cardiac output (especially renal perfu-
NT-proBNP) are a family of hormones released sion), increasing heart rate, or further activating
in increased amounts from myocytes (especially neurohormones because these factors have been
ventricular) secondary to myocardial stretch and associated with a worse prognosis [2]. The physi-
elevated end-diastolic filling pressure as occurs cians challenge is that many of the current medi-
in AHFS. Increased NP levels are indicators of cations that improve hemodynamics and
both the presence and severity of illness. symptoms may have potential deleterious effect
Accordingly, European guidelines recommend on such variables [8].
measurement of NP levels both to exclude alter- Currently, the use of available pharmacologi-
native causes of dyspnea and to obtain prognostic cal agents for the acute management of AHFS is
information. Patient presenting with acute onset largely empirical. None of the employed agents
or worsening of symptoms suggestive of HF with would meet todays standards for approval based
a plasma BNP level <100 pg/ml or NT-proBNP on evidence for clinical efficacy and safety.
<300 pg/ml is unlikely to have AHFS. For However matter, no major clinical practice guide-
patients presenting in nonacute way (slow onset lines include any therapeutic class I, level-of-
of symptoms), a lower exclusion NP cutoff point evidence A recommendations for the
should be used to avoid false-negative diagno- pharmacological treatment of AHFS [1].
sis (35 pg/ml for BNP and 125 pg/ml for Evaluation and management of AHFS include
NT-proBNP). Results of NP tests should be three main phases: the initial or early phase (sta-
always interpreted in the context of all available bilization phase), the in-hospital phase, and the
clinical data and should not be used in isolation discharge phase. The main goals of each phase
to diagnose HF. A variety of conditions associated are summarized in Table 7.4.
with myocardial stretch even in the absence of
AHF can still be associated with NP elevation
(e.g., atrial fibrillation, pulmonary hypertension, Initial Management Strategy
and pulmonary embolism). In addition, NP levels
are falsely increased in renal failure and tend to After treatment of life-threatening conditions,
be lower in obese patients. improving hemodynamics and correlated symp-
An initial bedside transthoracic echocardiog- toms are the key goals in early management. This
raphy is recommended both to support the diag- requires a basic understanding of pathophysio-
nosis of AHFS and to determine its etiology logic mechanisms underlying an episode of acute
through an assessment of cardiac anatomy and HF and how potential overt precipitants adversely
function (left and right ventricular systolic func- affect the cardiovascular system. These condi-
tion and wall motion, diastolic function, valvular tions and all HF precipitants should be targeted

ERRNVPHGLFRVRUJ
72 A. Romandini and S. Maffei

Table 7.4 Phases of AHFS management monary edema/congestion includes an initial


AHF management intravenous bolus of loop diuretics at time of pre-
Phases Goals sentation (usually furosemide 40 mg i.v. or 2.5
Early Ensure resuscitative supports and time the total outpatient oral loop diuretic dose)
stabilization appropriate timely interventions to and eventually an i.v. vasodilator if SBP
phase treat life-threatening conditions
>110 mmHg (class of recommendation II, level
eventually associated with AHFS
(such as hypoxia, unstable B) or an i.v. inotropic agent if SBP <85 mmHg
arrhythmias, STEMI, acute (class of recommendation II, level of evidence C)
mechanical valve dysfunction) as adjunctive therapy. Of note, in the American
Establish diagnosis
Guidelines (AHA 2013) on heart failure, no spe-
Determine patient clinical profile to
align initial treatment cific cutoff values exclude the use of a vasodilator,
Begin initial treatment to improve but its use is advocated generally in the absence of
congestive symptoms, cardiac symptomatic hypotension. Subsequently, patients
filling pressure, and/or CO
should be reevaluated (within 1 h) for adequate
Identify and treat reversible
precipitating factors adversely response. Response to treatment includes reduc-
affecting the CV system tion in dyspnea and adequate diuresis (>100 ml/h
In-hospital Start in-hospital monitoring (BP, urine production in first 2 h), accompanied by an
phase HR, O2 saturation, fluid balance, increase in oxygen saturation and usually reduc-
weight, laboratory tests)
tion in respiratory rate and heart rate. In the
Monitor signs/symptoms of
congestion for careful uptitration of absence of adequate response, all clinical-
decongestive therapy laboratory parameters should be reassessed (ECG,
Establish a proper workup to detect echocardiogram with hemodynamic measures,
and treat specific underlying cardiac
and principle laboratory tests) and several options
abnormalities or comorbidities that
cause or contribute to HF considered. The most common cause of inade-
progression (e.g., CAD, valvular quate response is, however, poor response to the
disease, arrhythmias, ventricular diuretic regimen utilized. Strategies to enhance
dyssynchrony, systemic or
diuretic efficacy will be discussed below in this
pulmonary arterial hypertension)
Initiation/uptitration of evidence- chapter. AHF patients unresponsive to diuretic
based therapy for chronic HF pharmacological therapy may be eventually con-
according to guidelines (beta- sidered for transient venovenous ultrafiltration
blockers, ACE inhibitors, ARBs,
(UF) that allows mechanical extracorporeal
MRA antagonists, electrical
devices) removal of plasma water. In patients with persis-
Discharge Ensure patient dry weight. tent hypotension (low CO) despite initial vasoac-
phase Congestive signs and symptoms tive therapy, other conditions like acute ischemic
should be reassessed (both at rest mechanical complications, severe valve dysfunc-
and during activity) and natriuretic
peptide levels measured
tion (particularly aortic stenosis), or alternative
Perform transition to oral diuretics diagnoses (e.g., pulmonary embolism) requiring
Assess functional capacity (6-min primary intervention rather than palliation of con-
walking test) sequences should be reconsidered. Pulmonary
Establish postdischarge planning
artery catheterization may be sometimes useful in
such unresponsive patients especially to ensure
and treated for optimal results. Aligning treat- that hypotension is not due to inadequate LV fill-
ment decision to initial patient clinical profile can ing pressure enabling more tailored vasoactive
yield to treat specific subgroups of patients with therapy (both inotropes and vasopressors). Finally,
more tolerable therapies. in unresponsive patients with persistent hypoten-
Taking the above consideration in mind, sion or cardiogenic shock with a rapid deteriora-
according to recommendations from ESC 2012 tion, a short-term mechanical circulatory support
Guidelines [1], early management of acute pul- (including intra-aortic balloon pump and ECMO)

ERRNVPHGLFRVRUJ
7 Acute Heart Failure and Pulmonary Edema 73

may be considered as a bridge to decision respiratory and metabolic improvement, its early
therapy. use can also prevent the need for endotracheal
Approximately 80 % of patients are hospital- intubation. However, in the recent Cardiogenic
ized with worsening of HF. For those with new- Pulmonary Oedema trial (3CPO), no differences
onset HF who stabilize after initial management, other than an improvement in dyspnea were seen
a chronic HF should be considered, and they in the rates of death or intubation between
should be treated according to recommendation patients treated with NIV compared to standard
of current guidelines. Initiation or implementa- oxygen therapy [16]. Currently, according to
tion of evidence-based pharmacological thera- ESC guidelines, NIV should be considered in
pies for chronic heart failure such as beta- blockers, such dyspneic patients generally when SBP is not
ACE inhibitors, aldosterone-blocking agents, below 85 mmHg (class of recommendation IIa,
ARB, and electrical device should occur soon level of evidence B).
during this phase after stabilization. This topic
will be extensively addressed in the chapter on
chronic heart failure. For the acute setting, it is Opiates
important to underline that the outpatient oral HF
medications should be always carefully reviewed Opiates such as morphine sulfate may be benefi-
at admission. Generally, HF therapy should be cial in pulmonary edema because it is thought to
continued at same doses during an AHFS episode induce mild venodilatation (thereby reducing
unless the patient has hypotension or contraindi- preload) and reduce anxiety and distress associ-
cations (such as hyperkalemia and severe renal ated with dyspnea.
failure for ACE inhibitors, angiotensin receptor Despite wide empiric use, small previous trial
blockers, aldosterone antagonists) that may raised concern about the safety of morphine,
require dose reduction or complete withholding. because its use has been associated with increased
Several reports have shown that continuation of need for invasive ventilation and greater in-
HF medical therapy with ACE inhibitors (or hospital mortality [17, 18]. At present, according
angiotensin receptor antagonists) and with beta- to the latest European guidelines, an i.v. opiate,
blockers for most patients is usually well toler- along with antiemetic medication, may be con-
ated and results in better outcomes [14, 15]. sidered in particularly restless and distressed
patients to relieve anxiety and improve breath-
lessness. Patients should be carefully monitored
Ventilation because opiates can induce respiratory
depression.
Oxygen supplementation should be titrated in
order to keep the patient comfortable achieving
arterial oxygen saturation above 90 %; caution is Diuretic Therapy
required in patients at risk of CO2 retention. In
the presence of significant respiratory distress, Diuretics are the mainstay of therapy in manag-
noninvasive positive-pressure ventilation (CPAP ing congestion in ADHS. Although safety and
or BiPAP) may be immediately considered to efficacy of diuretics have not been established in
relieve dyspnea and to improve hypoxia, meta- randomized controlled trials, long observational
bolic disturbance, and hemodynamic parameters experience has shown their efficacy in relieving
(reduced LV wall stress and cardiac work) in the congestive symptoms. By reducing intravascular
absence of contraindications (hypotension, vom- volume, diuretic therapy in ADHF lowers CVP
iting, depressed consciousness, pneumothorax). and PCWP reducing pulmonary and peripheral
Previous studies and meta-analysis support edema often increasing forward-stroke volume
the use of noninvasive ventilation (NIV) in car- and CO. In addition, when given intravenously,
diogenic pulmonary edema showing that besides loop diuretics may act as vasodilators (principally

ERRNVPHGLFRVRUJ
74 A. Romandini and S. Maffei

venodilators) with additional benefit on renal and Suggested maximum effective doses of loop
pulmonary congestion. Despite the demonstrated diuretics in heart failure and renal insufficiency
efficacy in managing congestion, the use of have been previously described [22]. Regarding
aggressive diuretic regimens may be associated the use of furosemide, in patients with HF and
with neurohormonal activation, worsening renal normal renal function, suggested maximal intra-
function, electrolyte abnormalities, and arrhyth- venous doses are 4080 mg, but in the presence
mias and so with adverse clinical outcomes [19, of renal insufficiency, larger staring doses may be
20]. required up to 160 or 200 mg in moderate and
Current guidelines recommend administering severe renal impairment, respectively. Doses of
the lowest dosage in order to achieve and main- 250 mg or above should be given by infusion
tain euvolemia avoiding volume depletion and over 4 h.
dehydration. From a pharmacokinetic and pharmacody-
Loop diuretics have the most rapid onset and namic perspective, there are potential benefits of
most powerful effect. In acute setting, like AHFS, continuous infusion versus intermittent bolus.
intravenous rather than oral administration is rec- Continuous infusion results in more constant
ommended because of greater drug bioavailabil- delivery of diuretics to the tubule with increased
ity and more rapid onset of action. Diuretic diuresis probably minimizing intermittent periods
dosing should be individualized and titrated of the known postsodium retention effect. After a
according to patient status and initial response. starting loading dose, suggested starting infusion
Considering the bolus therapy, usually the rate of loop diuretics varies with the level of renal
diuretic effect begins within 30 min with a peak function (GFR or creatinine clearance). For furo-
at 12 h [21]. Common suggested initial doses of semide, suggested doses are 10 mg/h with a
intravenous loop diuretics are 40 mg for furose- GFR > 75 ml/min, 1020 mg/h with a GFR
mide, 0.51 mg for bumetanide, and 510 mg for 2575 ml/min, and 2040 mg if GFR < 25 ml/min.
torsemide in patients who are not receiving loop If an adequate response has not occurred within
diuretics [22]. In patients who have been already 1h, a loading dose should be repeated and then the
taking a loop diuretic, the dose should be almost infusion rate uptitrated [22].
equal or greater (i.e., 2.5 times) than the mainte- However, existing data still does not allow
nance oral dose (in ESC guidelines, the greater definitive recommendations for clinical practice
dose is advocated) [1]. As discussed above, poor because even in a recent trial, no clear benefit or
response to the diuretic is a common cause of harms with intermittent bolus versus continuous
inadequate response to initial therapeutic infusion strategy have been demonstrated [24].
approach in AHFS. Now a widely accepted defi- Several strategies can be tried to overcome
nition of diuretic resistance in HF is still lacking. such diuretic resistance especially during in-
In HF patients, the diuretic doseresponse curve hospital phase. A common method for treating
may be shifted downward and to the right because diuretic resistance is the sequential nephron
of a reduction in both renal drug delivery and blockade by adding a thiazidic or thiazide-like
natriuretic response. So higher doses are required diuretic (DCT diuretics) [25, 26]. Metolazone
to achieve a given diuretic response, and the max- and hydrochlorothiazide are the most common
imal effect may be bunted [23]. Once a single molecules used in combination with loop diuret-
effective dose has been determined, it should be ics. Many clinicians prefer metolazone, a
administered multiple times per day (two or three thiazide-like diuretic, because it has a longer
times) according to the magnitude of diuresis half-life and a preserved efficacy in advanced
needed. If there is little or no response, experts renal failure (GFR below 20 ml/min) [27].
recommend dose doubling at 2 h intervals as However, such combination therapy is associated
needed (until effective diuresis is demonstrated) with significant increase in adverse effect espe-
up to the maximum effective doses (ceiling doses cially when high doses of DCT diuretics are used
over which no further diuresis will be achieved). compared to either therapy alone [28].

ERRNVPHGLFRVRUJ
7 Acute Heart Failure and Pulmonary Edema 75

It is advisable to start with lower dose of DCT with loop diuretics, the ultrafiltrate (or volume
diuretic (hydrochlorothiazide 12.525 mg or removed) is isotonic to plasma and therefore
metolazone 2.55 mg) and maintain a daily regi- removed more sodium (and less potassium). In
men for a short period with careful monitoring of addition, UF allows a better control of plasma
electrolyte balance and fluid depletion [29]. water removal rate that can be tuned to match the
putative refilling rate from the interstitium
(approximately 15 ml/min) avoiding intravascu-
Additional Strategy to Enhance lar depletion and the vicious cycle of further neu-
Diuresis rohormonal activation [31].

Dopamine infusion at low doses (3 g/kg/min)


may selectively activate dopamine receptors Intravenous Vasodilators
(DA1 and DA2) resulting in renal vasodilation
and increasing renal blood flow. However, a sig- By reducing both preload and afterload and
nificant benefit of standard use of dopamine has therefore cardiac filling pressure, vasodilators
not been confirmed in recent randomized studies may have beneficial hemodynamic effects in
[30]. Although there is uncertainty, in the latest ADHS by reducing pulmonary congestion and
2012 ESC guidelines, it is advocated to start infu- usually increasing CO. The mechanisms for
sion with low dose of dopamine (at 2.5 mcg/kg/ increased CO include left and right ventricular
min) in patients with poor response to diuretic afterload reduction, improved diastolic ventricu-
regimen. lar properties, reduced mitral regurgitation, and
Salt and fluid restriction is another strategy eventually reduction in myocardial ischemia.
that has also been commonly used during initial Currently approved intravenous vasodilators
management of AHF patients, although as noted in clinical practice are organic nitrates such as
in recent European guidelines no firm evidence nitroglycerin (NTG) and isosorbide dinitrate
exists to support this practice. By reducing (ISDN), inorganic nitrates such as sodium nitro-
sodium load at the nephron, postdiuretic sodium prusside (SNP), and nesiritide (currently not
retention may be reduced, especially when available in many European countries).
sodium intake is high. Generally, it is common to Despite that nitrates have been used to relieve
restrict sodium intake <2 g/day and fluid intake symptoms and improve hemodynamic acute HF
<1.52.0 L/day [1]. for many years, their use is still based on limited
evidence primarily from small, single-center
studies [32]. Currently, the use of intravenous
Ultraltration vasodilators is recommended (class II and level
of evidence B) to relieve symptoms and to reduce
AHF patients unresponsive to diuretic therapy pulmonary congestion in patients in AHFS with
may be considered for venovenous ultrafiltration intact blood pressure. In the latest 2012 ESC
(UF). UF allows mechanical extracorporeal guidelines [1], their use is advocated only in
removal of plasma water across a semipermeable patients with a BP greater than 110 mmHg, com-
membrane in response to a transmembrane pres- pared to ACCF/AHA guidelines where their use
sure gradient (convective transfer). Venovenous is limited only in the presence of symptomatic
UF is performed at bedside via a central or hypotension. Suggested intravenous doses of
peripheral vascular access using a transportable vasodilators in AHFS are indicated in Table 7.5.
UF console. With slow continuous UF usually Among organic nitrates, the most widely used
performed in HF patients, the amount of ultrafil- is NTG. At low modest doses, intravenous NTG
trate created is small (24 ml/min) and does not acts primarily through venodilatation, while at
require replacement with substitution fluid. higher doses (>40 mcg/min) the effect of arterio-
Compared to hypotonic urine output achieved lar dilatation begins to be apparent [33]. However,

ERRNVPHGLFRVRUJ
76 A. Romandini and S. Maffei

Table 7.5 Doses of intravenous vasodilators in AHFS the true incidence of clinically significant coro-
Suggested intravenous vasodilator doses in AHFS nary steal remains unknown.
Agent Doses
Nitroglycerine Start with 1020 mcg/min and
then increase up to 200 mcg/ Inotropes and Vasopressors
min
Isosorbide dinitrate Start with 1 mg/h and then
Despite the hemodynamic benefits in the short-
increase up to 10 mg/h
Nitroprusside Start with 0.3 mcg/kg/min and
term management, positive inotropic agents have
then increase up to 5 mcg/kg/ not demonstrated improved outcomes in patients
min with HF in both hospital and outpatient settings.
Modified and reproduced with permission from McMurray Rather, data from some registries and post hoc
et al. [1] analyses of RCT suggest an increased morbidity
and mortality with inotrope use in HF. In fact, as
opposed to hemodynamic benefits, inotropes
despite a graded doseresponse curve, a variable may cause sinus tachycardia and precipitate
interindividual response exists also related to myocardial ischemia and arrhythmias [3739].
baseline levels of systemic vascular resistance. In AHFS, the use of intravenous inotropic
A process of careful uptitration is always needed agents may be helpful to improve CO in patients
to avoid sudden BP reduction or hypotension. A with severe LV dysfunction with hypotension
significant drawback for intravenous nitrate par- (PAS < 85 mmHg) and/or low-output syndrome.
ticularly with NTG is the phenomenon of tachy- In such patients, the marginal systemic perfusion
phylaxis that may occur in 1530 % of patients may limit institution and adequate response to
within 24 h probably related to strong counter- the other pharmacological treatment like diuret-
regulatory neurohormonal activation that leads to ics. On the other hand, at the cost of increasing
sodium and water retention [34]. afterload and decreasing cardiac output, drugs
Sodium nitroprusside (SNP) is the sodium salt with arterial vasoconstriction action (e.g., norepi-
of a complex molecule that breaks down in the nephrine or dopamine at high doses) may be a
blood interacting with oxyhemoglobin and temporizing measure to redistribute CO from
directly releasing NO and cyanide into circula- extremities to vital organs, and their use is
tion. SNP is a potent vasodilator with the faster restricted to patients with persistent hypoperfu-
onset of action (within 6090 s). Its short half- sion despite optimization of cardiac filling pres-
life (approximately 2 min) facilitates early estab- sure and the concomitant use of inotropes. The
lishment in the intensive care unit of an individual use of inotropic/vasopressor agents will be
patients optimal level of vasodilation. Even low addressed in the chapter on the management
doses produce an equivalent venous and arterio- shock. In the 2102 ESC guidelines, the use of
lar dilatation resulting in balanced vasodilation inotropes is in class IIa of recommendation, level
of both sides of the circulation [35]. Because of evidence C, and the use of vasopressor in class
SNP can cause significant hypotension, it is usu- IIb, level of evidence C [1].
ally used in intensive care settings even with Calcium sensitizers such as levosimendan are
invasive arterial monitoring. It is postulated that a new category of inotropic agents that exert pos-
SNP may potentially increase the risk of a coro- itive inotropic effects by increasing the affinity of
nary steel phenomenon: as opposed to nitroglyc- troponin C for calcium. Levosimendan exertion
erins preferential effect on larger conductance also has a vasodilatory effect by blocking ade-
vessels, SNP dilates smaller resistance vessels nosine triphosphate-dependent potassium chan-
creating a low-pressure system distal to occluded nels in the vascular smooth muscle cells. Such
vessels that diverts critical pressure-dependent inotropic and vasodilator effects may result in
flow from ischemic areas [36]. The clinical sig- increased CO and reduced filling pressures in
nificance of these observations is uncertain, and AHF patients. Compared to the classic inotropic

ERRNVPHGLFRVRUJ
7 Acute Heart Failure and Pulmonary Edema 77

agents, calcium sensitizers have two major phar- 2. Gheorghiade M, De Luca L, Fonarow GC, Filippatos
G, Metra M, Francis GS (2005) Pathophysiologic tar-
macodynamic advantages: first, increase in con-
gets in the early phase of acute heart failure syn-
tractile force occurs without increasing calcium dromes. Am J Cardiol 96(6A):11G17G
loading that is associated with enhanced myocar- 3. Yancy CW (2008) Vasodilator therapy for decompen-
dial oxygen consumption, increased heart rate, sated heart failure. J Am Coll Cardiol 52(3):208210
4. Gheorghiade M, Vaduganathan M, Fonarow GC,
and arrhythmias; second, the inotropic effect is
Bonow RO (2013) Rehospitalization for heart failure:
not attenuated by concomitant treatment with problems and perspectives. J Am Coll Cardiol
beta-blockers. At present, the real risk/benefit 61(4):391403
ratio of levosimendan in AHFS is still debated in 5. Cleland JG, Swedberg K, Follath F et al (2003) The
EuroHeart Failure survey programmea survey on
light of less favorable outcomes observed in a
the quality of care among patients with heart failure in
recent study compared to placebo [40]. Currently, Europe. Part 1: patient characteristics and diagnosis.
levosimendan is approved in Europe as a second- Eur Heart J 24:442463
line agent for severe low-output HF refractory 6. Adams KF Jr, Fonarow GC, Emerman CL et al, for
the ADHERE Scientific Advisory Committee and
standard therapy, and according to ESC guide-
Investigators (2005) Characteristics and outcomes of
lines its use may be considered (recommendation patients hospitalized for heart failure in the United
class IIb, level of evidence C) especially in States: rationale, design, and preliminary observa-
patients with chronic beta-blocker therapy to tions from the first 100,000 cases in the Acute
Decompensated Failure National Registry
overcome the beta-blockade effect.
(ADHERE). Am Heart J 149:209216
7. Fonarow GC, Abraham WT, Albert NM et al (2004)
Organized program to initiate lifesaving treatment in
Thromboembolism Prophylaxis hospitalized patients with heart failure (OPTIMIZE-HF):
rationale and design. Am Heart J 148:4351
8. Gheorghiade M, Pang PS (2009) Acute heart failure
Several mechanisms like increased systemic syndromes. J Am Coll Cardiol 53(7):557573
venous pressure, low cardiac output, and proco- 9. Cotter G, Felker GM, Adams KF, Milo-Cotter O,
agulant blood changes may increase the risk of OConnor CM (2008) The pathophysiology of acute
heart failureis it all about fluid accumulation? Am
venous thromboembolism in patients with
Heart J 155(1):918
HF. For this reason, in the absence of contraindi- 10. Metra M, Felker GM, Zac V, Bugatti S, Lombardi C,
cation to anticoagulation, thromboembolism pro- Bettari L, Voors AA, Gheorghiade M, Dei CL (2010)
phylaxis (e.g., LMWH) is currently recommended Acute heart failure: multiple clinical profiles and
mechanisms require tailored therapy. Int J Cardiol
when HF patients are hospitalized (if not already
144(2):175179
anticoagulated) to reduce the risk of deep vein 11. Fallick C, Sobotka PA, Dunlap ME (2011)
thrombosis and pulmonary embolism (recom- Sympathetically mediated changes in capacitance:
mendation class I, level of evidence A) [1]. redistribution of the venous reservoir as a cause of
decompensation. Circ Heart Fail 4:669e75
12. Nohria A, Tsang SW, Fang JC, Lewis EF, Jarcho JA,
Mudge GH, Stevenson LW (2003) Clinical assess-
References ment identifies hemodynamic profiles that predict
outcomes in patients admitted with heart failure. J Am
1. McMurray JJ, Adamopoulos S, Anker SD, Auricchio Coll Cardiol 41(10):17971804
A, Bhm M, Dickstein K, Falk V, Filippatos G, 13. Forrester JS, Diamond G, Chatterjee K, Swan HJ
Fonseca C, Gomez-Sanchez MA, Jaarsma T, Kber L, (1976) Medical therapy of acute myocardial infarc-
Lip GY, Maggioni AP, Parkhomenko A, Pieske BM, tion by application of hemodynamic subsets (first of
Popescu BA, Rnnevik PK, Rutten FH, Schwitter J, two parts). N Engl J Med 295(24):13561362
Seferovic P, Stepinska J, Trindade PT, Voors AA, 14. Metra M, Torp-Pedersen C, Cleland JG et al (2007)
Zannad F, Zeiher A, ESC Committee for Practice Should beta-blocker therapy be reduced or withdrawn
Guidelines (2012) ESC guidelines for the diagnosis after an episode of decompensated heart failure?
and treatment of acute and chronic heart failure 2012: Results from COMET. Eur J Heart Fail 9:901
The Task Force for the Diagnosis and Treatment of 15. Fonarow GC, Abraham WT, Albert NM et al (2008)
Acute and Chronic Heart Failure 2012 of the European Influence of beta-blocker continuation or withdrawal
Society of Cardiology. Developed in collaboration on outcomes in patients hospitalized with heart fail-
with the Heart Failure Association (HFA) of the ure: findings from the OPTIMIZE-HF program. J Am
ESC. Eur Heart J 33(14):17871847 Coll Cardiol 52:190

ERRNVPHGLFRVRUJ
78 A. Romandini and S. Maffei

16. Gray A, Goodacre S, Newby DE, Masson M, Sampson Stehlik J, LeWinter MM, Konstam MA, Huggins GS,
F, Nicholl J, 3CPO Trialists (2008) Noninvasive ven- Rouleau JL, OMeara E, Tang WH, Starling RC,
tilation in acute cardiogenic pulmonary edema. Butler J, Deswal A, Felker GM, OConnor CM,
N Engl J Med 359(2):142151 Bonita RE, Margulies KB, Cappola TP, Ofili EO,
17. Hoffman JR, Reynolds S (1987) Comparison of nitro- Mann DL, Dvila-Romn VG, McNulty SE, Borlaug
glycerin, morphine and furosemide in treatment of BA, Velazquez EJ, Lee KL, Shah MR, Hernandez AF,
presumed pre-hospital pulmonary edema. Chest Braunwald E, Redfield MM, NHLBI Heart Failure
92:586593 Clinical Research Network (2013) Low-dose dopa-
18. Peacock WF, Hollander JE, Diercks DB et al (2008) mine or low-dose nesiritide in acute heart failure with
Morphine and outcomes in acute decompensated renal dysfunction: the ROSE acute heart failure ran-
heart failure: an ADHERE analysis. Emerg Med J domized trial. JAMA 310(23):25332543
25:205209 31. Felker GM, Mentz RJ (2012) Diuretics and ultrafiltra-
19. Domanski M, Norman J, Pitt B, Haigney M, Hanlon tion in acute decompensated heart failure. J Am Coll
S, Peyster E (2003) Diuretic use, progressive heart Cardiol 59(24):21452153
failure, and death in patients in the Studies Of Left 32. Piper S, McDonagh T (2014) The role of intravenous
Ventricular Dysfunction (SOLVD). J Am Coll Cardiol vasodilators in acute heart failure management. Eur J
42:705708 Heart Fail 16(8):827834
20. Hasselblad V, Gattis Stough W, Shah MR et al (2007) 33. Bayley S, Valentine H, Bennett ED (1984) The hae-
Relation between dose of loop diuretics and outcomes modynamic responses to incremental doses of intra-
in a heart failure population: results of the ESCAPE venous nitroglycerin in left ventricular failure.
trial. Eur J Heart Fail 9:10641069 Intensive Care Med 10:139145
21. Sagar S, Sharma BK, Sharma PL, Wahi PL (1984) A 34. Gupta D, Georgiopoulou VV, Kalogeropoulos AP,
comparative randomized double-blind clinical trial of Marti CN, Yancy CW, Gheorghiade M, Fonarow GC,
bumetanide and furosemide in congestive cardiac fail- Konstam MA, Butler J (2013) Nitrate therapy for
ure and other edema states. Int J Clin Pharmacol Ther heart failure: benefits and strategies to overcome tol-
Toxicol 22:473478 erance. JACC Heart Fail 1(3):183191
22. Brater DC (1998) Diuretic therapy. N Engl J Med 35. Elkayam U, Janmohamed M, Habib M, Hatamizadeh
339:387395 P (2008) Vasodilators in the management of acute
23. Ellison DH (2001) Diuretic therapy and resistance in heart failure. Crit Care Med 36(1 Suppl):S95S105
congestive heart failure. Cardiology 96(34): 36. Mann T, Cohn PF, Holman LB et al (1978) Effect of
132143 nitroprusside on regional myocardial blood flow in
24. Felker GM, Lee KL, Bull DA, Redfield MM, coronary artery disease: Results in 25 patients and
Stevenson LW, Goldsmith SR, LeWinter MM, Deswal comparison with NTG. Circulation 57:732738
A, Rouleau JL, Ofili EO, Anstrom KJ, Hernandez AF, 37. Elkayam U, Tasissa G, Binanay C et al (2007) Use
McNulty SE, Velazquez EJ, Kfoury AG, Chen HH, and impact of inotropes and vasodilator therapy in
Givertz MM, Semigran MJ, Bart BA, Mascette AM, hospitalized patients with severe heart failure. Am
Braunwald E, OConnor CM, NHLBI Heart Failure Heart J 153:98104
Clinical Research Network (2011) Diuretic strategies 38. Abraham WT, Adams KF, Fonarow GC, et al;
in patients with acute decompensated heart failure. ADHERE Scientific Advisory Committee and
N Engl J Med 364(9):797805 Investigators; ADHERE Study Group (2005) In- hos-
25. Ghose RR, Gupta SK (1981) Synergistic action of pital mortality in patients with acute decompensated
metolazone with loop diuretics. Br Med J 282: heart failure requiring intravenous vasoactive medica-
14321433 tions: an analysis from the acute decompensated heart
26. Fliser D, Schroter M, Neubeck M et al (1994) failure national registry (adhere). J Am Coll Cardiol
Coadministration of thiazides increases the efficacy of 46:5764
loop diuretics even in patients with renal failure. 39. OConnor CM, Gattis WA, Uretsky BF et al (1999)
Kidney Int 46:482488 Continuous intravenous dobutamine is associated
27. Dargie HJ, Allison ME, Kennedy AC et al (1972) with an increased risk of death in patients with
High dosage metolazone in chronic renal failure. Br advanced heart failure: insights from the Flolan
Med J 4:196198 International Randomized Survival Trial (first). Am
28. Oster JR, Epstein M, Smoller S (1983) Combined Heart J 138:7886
therapy with thiazide-type and loop diuretic agents for 40. Packer M, Colucci W, Fisher L, Massie BM, Teerlink
resistant sodium retention. Ann Intern Med 99: JR, Young J, Padley RJ, Thakkar R, Delgado-Herrera
405406 L, Salon J, Garratt C, Huang B, Sarapohja T, REVIVE
29. Ernst ME, Moser M (2009) Use of diuretics in patients Heart Failure Study Group (2013) Effect of levosi-
with hypertension. N Engl J Med 361(22): mendan on the short-term clinical course of patients
21532164 with acutely decompensated heart failure. JACC
30. Chen HH, Anstrom KJ, Givertz MM, Stevenson LW, Heart Fail 1(2):103111. doi:10.1016/j.
Semigran MJ, Goldsmith SR, Bart BA, Bull DA, jchf.2012.12.004. Epub 2013 Apr 1

ERRNVPHGLFRVRUJ
Chronic Heart Failure
8
Simona Masiero and Marco Morelli

8.1 Case Report opathy (confirmed by a coronary catheteriza-


tion). Since then he started a therapy with loop
diuretic, beta-blocker, ACE inhibitor, and
A 67-year-old man came to the emergency aldosterone antagonist.
room of our hospital for worsened exer- Since 2007 nowdays he underwent to several
tional dyspnea, orthopnea, increased fatigue hospital admissions for acute exacerbation of
during ordinary activities, swollen ankles heart failure.
and reduced urine output. He was affected In 2013 a CRT-D has been implanted because
by chronic nonischemic dilated cardiomy- of low ejection fraction (EF 25 %), LBBB
opathy. He reported on a progressively (left bundle branch block), and persistent
worsening of symptoms in the previous symptoms in NYHA functional class III
15 days and a weight gain of about 6 kg. despite optimized medical therapy. The ambu-
latory device follow-up highlighted some epi-
sodes of sustained ventricular tachycardia
treated with overdrive or shock and episodes
Medical History and Cardiovascular of atrial fibrillation. For these reasons he
Risk Factors started a therapy with amiodarone and VKA
(vitamin K antagonist: warfarin) therapies.
In 2007 the patient underwent to the first hos- Chronic renal failure.
pitalization for heart failure with a diagnosis Hypertension.
of nonischemic hypokinetic dilated cardiomy- Benign prostatic hypertrophy.

Medications

Furosemide 50 mg at 8:00 am, metolazone 5 mg


at 6:00 pm on alternate days, spironolactone
25 mg at 8:00 pm, metoprolol 25 mg at 8:00 am
S. Masiero (*) M. Morelli and 25 mg at 8:00 pm, warfarin according to
Department of Cardiovascular Sciences, INR, amiodarone 200 mg at 1:00 pm, dutasteride
Clinica di Cardiologia e Aritmologia,
0.5 mg at 8:00 am, and tamsulosin 0.4 mg at
Universit Politecnica delle Marche,
via conca 71, Ancona 60100, Italy 8:00 pm. Allopurinol 150 mg ore at 8:00 pm and
e-mail: simo_masiero@libero.it; moro@abanet.it pantoprazole 20 mg ore at 7:00 am
Springer International Publishing Switzerland 2015 79
A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_8

ERRNVPHGLFRVRUJ
80 S. Masiero and M. Morelli

Vital Signs Which Are the Possible Precipitants


for Heart Failure Decompensation?
Temperature: 36.4 C
Heart rate: 70 bpm
Blood pressure: 110/60 mmHg Inadequate dose/adherence to pre-
Respiratory rate: 20 breaths per minute (mild scribed therapies
tachypnea) Noncompliance with dietary restrictions
Oxygen saturation while breathing ambient (sodium and/or liquids, etc.)
air: 93 % Acute myocardial ischemia
Worsened valvular heart disease
Arrhythmias (bradyarrhythmias or
Physical Examination tachyarrhythmias)
Exacerbation of chronic obstructive pul-
General: fatigued, short of breath, alert, monary disease with or without
awake, and oriented; well developed and well pneumonia
nourished Infections (pneumonia, influenza, etc.)
Head, eyes, ears, nose, and throat: normoce- Renal dysfunction
phalic, atraumatic, mucous membranes moist, Endocrine disorders (diabetes mellitus,
extraocular muscles intact, pupils equally hypo-/hyperthyroidism)
round and reactive to light and accommoda- Anemia
tion bilaterally, bilateral tympanic membrane Medications (nonsteroidal anti-
intact, bilateral sclera anicteric, and no con- inflammatory drugs, drugs with nega-
junctival injection tive inotropic effect such as calcium
Neck: supple, no jugular venous distention, no channel blockers)
lymphadenopathy, and no carotid bruit
Cardiovascular: regular rate and rhythm,
laterally and down displaced apical impulse,
S1 and S2 normal, S3 present (gallop EKG
rhythm), 3/6 systolic murmur at the cardiac
apex and mesocardium, and no hepatojugu- A routine EKG at rest was performed (Fig. 8.1).
lar reflux Report: sinus rhythm, heart rate 72 bpm with
Lungs: rales at auscultation at the bases bilater- atrium-driven biventricular pacing, QRS duration
ally, mild wheezing, no rhonchi, no alterations 160 msec.
in tactile fremitus, and normal percussion Despite correct biventricular pacing (axis
Abdomen: mild overweight, no pulsatile directed upper right), the QRS complex is wide.
masses, normal bowel sounds in all four quad- Several lead configurations have been tried
rants, no high-pitched or tinkling sounds, res- assessing EKG and echocardiographic findings
onant to percussion, soft, non-distended/ in order to find the best stimulation without
non-tender, no rebound or guarding, and no success.
hepatosplenomegaly
Extremities: no cyanosis or clubbing; mild
peripheral edema of the ankles Routine Laboratory Tests
Neurologic: cranial nerves II through XII
intact and, no focal deficit Complete blood count: normal
Psychiatric: normal affect, no hallucinations, Cholesterol (total, HDL, LDL) and TG:
normal speech, and no dysarthria normal
Skin: intact, no rashes, no lesions, and no Hepatic function (GOT, GPT, -GT, ALP, bili-
erythema rubin): slight increase in transaminases

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 81

Fig. 8.1 (a, b) Routine EKG at rest. Despite correct tried assessing EKG and echocardiographic findings in
biventricular pacing (axis directed upper right), the QRS order to find the best stimulation without success
complex is wide. Several lead configurations have been

Thyroid function (TSH, FT3, FT4): normal Chest X-Ray


Renal function: creatinine 1.7 mg/dl (estimated
glomerular filtration rate with the Cockcroft A chest X-ray was performed too (Fig. 8.2).
Gault equation (GFR-CG) = 50 ml/min mod- Cardiac shadow was slightly enlarged for an
erate chronic kidney disease), BUN 55 mg/dl increase of cardiac transverse diameter. A left-
Electrolytes: mild hyponatremia 134 mEq/l sided pleural effusion was seen obliterating
and hypokalemia 2.1 mEq/l the costophrenic recess associated with a
Fasting blood glucose: 194 m/dl bilateral hilar enlargement with widespread
(10.78 mmol/L) bronchovascular marking and interstitial pul-
HbA1c: 6.8 % (50.8 mmol/mol) monary congestion was presence of right ven-
TnI-hs and CK-MB: normal tricular and atrial leads and coronary sinus
BNP: 1,100 pg/ml lead.

ERRNVPHGLFRVRUJ
82 S. Masiero and M. Morelli

The mitral ring was dilated with moderate val-


vular regurgitation.
There was evidence of massive tricuspid
regurgitation with pulmonary hypertension
(estimated PAPs of about 60 mmHg).

Clinical Course and Medical Therapy

Furosemide: 40 mg t.i.d. (three times a day)


intravenous boluses
Slow-release potassium chloride: 1,200 mg
b.i.d. (bis in die) per os
Canrenoate potassium: 100 mg o.d. (once
daily) intravenous boluses
Saline 0.9 % continuous infusion 40 cc/h + KCl
Fig. 8.2 Chest X-ray. The cardiac shadow is slightly 40 mEq
enlarged due to an increase of cardiac transverse diam-
Dobutamine (250 mg/50 ml): 2 g/kg/min
eter. A left-sided pleural effusion obliterates the costo-
phrenic recess and is associated with a bilateral hilar continuous infusion
enlargement with widespread bronchovascular marking Dopamine (200 mg/50 ml): 2 g/kg/min con-
and interstitial pulmonary congestion. The presence tinuous infusion
of right ventricular and atrial leads and coronary
Metoprolol: 25 mg b.i.d. per os
sinus lead
Ivabradine: 5 mg b.i.d. per os
Amiodarone: 200 mg o.d. per os
Warfarin: according to INR
Echocardiography Tamsulosin: 0.4 mg o.d. per os
Dutasteride: 0.5 mg o.d. per os
In order to complete the diagnostic process at Pantoprazole: 20 mg o.d. per os
admission, an echocardiography was performed.
C-PAP therapy was administered (PEEP
The left ventricle was severely dilated 7.5 mmHg, FiO2 0.5) with intermittent cycles of
(indexed left ventricular end-diastolic volume about 2 h.
(iLVEDV) 167 ml/m2) with severe reduction After 4872 h there was a significant loss of
of systolic function (ejection fraction mea- fluids supported by good diuresis. The patient
sured with Simpsons biplane method = 25 %; reported improvement in dyspnea, and we
stroke volume = 29.4 cc; cardiac output = 2.1 l/ observed a consistent reduction of ankle edema.
min; cardiac index = 1 l/m2) and restrictive The inotropic support was gradually discontin-
diastolic pattern. Moreover, filling pressures ued. The laboratory tests showed a progressive
were increased (E/E = 16). increase of potassium values and reduced levels
Right ventricle was slightly dilated and hypo- of BNP. The chest X-ray showed a reduced
kinetic (basal diameter RVD1 = 42 mm; tri- degree of pulmonary congestion, and the echo-
cuspid ring excursion TAPSE = 14 mm). cardiogram showed stable EF (0.25) associated
Severe dilatation of both atria (LA diameter with a small increase in cardiac index (1.3 ml/
M-mode = 53.5 cm; RA area A4C = 34 cm2). min/m2) and a slight reduction of the PAPs
No pericardial effusion. (50 mmHg).
The inferior vena cava was dilated without Once the patient reached an acceptable
inspiratory collapse. grade of compensation, he was proposed for the
The aortic valve was trileaflet and sclerotic. implantation of a left ventricular assist device as

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 83

a b

Fig. 8.3 (a, b) Chestabdomen CT with contrast, with completely normal appearance

destination therapy. For this purpose the patient signs (e.g., elevated jugular venous pressure, pul-
underwent to a new coronary angiography and monary crackles) resulting from an abnormality
chestabdomen CT with contrast which were of cardiac structure or function [1].
completely normal (Fig. 8.3a, b). Chronic heart failure (CHF) can be caused by
In order to perform a complete evaluation several types of cardiac dysfunction and is most
before the left ventricular assist device implant, commonly due to left ventricular dysfunction. An
we performed a right heart catheterization which isolated right ventricular (RV) dysfunction is
showed: very rare, and generally RV involvement is sec-
ondary to left ventricular (LV) dysfunction.
Normal pressures in the right atrium, right Demonstration of an underlying cardiac cause
ventricle, pulmonary artery, and wedge is essential to diagnose HF as the precise pathol-
Slight increase in total pulmonary resistance ogy determines the specific treatment used. More
Arteriolar resistance at the upper limit of normal recently, CHF has been classified into two cate-
gories: HF due to LV dysfunction even called HF
By the investigations carried out during the with reduced ejection fraction (HF-REF or sys-
hospitalization, the patient has been judged suit- tolic heart failure) and HF where only a diastolic
able for LVAD as a destination therapy and added dysfunction is detectable called HF with pre-
to the waiting list at a referral center for the served ejection fraction (HF-PEF or diastolic
implant. heart failure). 1 HF-REF is the best understood
Until the LVAD implant, on top therapy to type of HF in terms of pathophysiology and treat-
chronic heart failure including loop diuretic, ment and is the focus of this chapter.
aldosterone antagonist, ACE inhibitor, and beta- Furthermore, HF can present either as a
blocker, the patient underwent cyclic intravenous chronic condition or acutely, occurring de novo,
inotropic therapy. or as a decompensation of CHF. The purpose of
this chapter is to cover CHF, while acute heart
failure is discussed in another section of this
8.2 Heart Failure book.

Denition
Epidemiology
Heart failure (HF) can be clinically defined as a
syndrome in which patients have typical symp- CHF prevalence is 12 % of the population, and
toms (e.g., breathlessness, ankle swelling) and the prevalence increases to approximately 15 %

ERRNVPHGLFRVRUJ
84 S. Masiero and M. Morelli

in the elderly [2]. At least half of patients with symptoms of HF are due in part to compensatory
HF have a low EF, and approximately 50 % of mechanisms utilized by the body in an attempt to
patients with significant LV systolic dysfunction adjust for a primary deficit in cardiac output.
have no symptoms or signs of heart failure. HF Many of the processes involved in sustaining HF
occurs more frequently in male rather than are maladaptive which means that they were
female sex. originally designed to maintain blood pressure
and vital organ perfusion.

Etiology
Changes in Hemodynamics
The causes of CHF are listed in Table 8.1. There
is a geographical variation regarding the etiology Decrease of cardiac output leads to an increase of:
of CHF. In Western countries two-thirds are sec-
ondary to ischemic disease, and other important Left ventricular end-diastolic pressure
contributors are hypertension, valve disease, and Pulmonary capillary wedge pressure
alcohol. Rheumatic disease still remains the most
common cause of CHF in the developing coun- Based on the FrankStarling law, the initial
tries, while Chagas disease is frequent in South increase of left ventricular end-diastolic pressure
America. is initially compensated by an increase of con-
tractility, but as the increase persists, the myocar-
dium fails and cardiac output drops.
Pathophysiology

Left ventricular dysfunction is associated with Neurohumoral Changes


hemodynamic, autonomic, neurohumoral, and
immunological changes. Neurohumoral adaptations, such as activation of
The term systolic dysfunction refers to a the reninangiotensinaldosterone and sympa-
decrease in myocardial contractility and conse- thetic nervous systems by the low-output state,
quently a decrease in cardiac output. Signs and can contribute to maintenance of perfusion of
vital organs in two ways:
Table 8.1 Causes of chronic heart failure
Maintenance of systemic pressure by vaso-
Coronary artery disease
constriction, resulting in redistribution of
Hypertension
blood flow to vital organs
Valve disease
Restoration of cardiac output by increasing
Congenital heart disease
Infective: viral myocarditis, Chagas, HIV, Lyme
myocardial contractility and heart rate and by
disease expansion of the extracellular fluid volume
Alcohol
Toxins: anthracyclines or trastuzumab The principal neurohumoral systems involved
Deficiencies: beriberi, thiamine in the response to HF are the sympathetic nervous
Hemochromatosis system, the reninangiotensinaldosterone system
Idiopathic (RAAS), and antidiuretic hormone [3, 4, 5]. One of
Familiar the first responses to a decrease in cardiac output is
Peripartum activation of the sympathetic nervous system,
Tachycardia induced resulting in both increased release and decreased
Infiltrative states: amyloid, sarcoid, endomyocardial uptake of norepinephrine at adrenergic nerve end-
fibrosis, hypereosinophilic syndrome ings. The effects of high circulating concentrations
High output: AV fistulae, Pagets disease of epinephrine and norepinephrine include:

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 85

Increase in heart rate, blood pressure, and mones. The types of natriuretic peptide hormones
myocardial oxygen demand which circulate in high concentration in HF are:
A toxic damage on the myocardium leading to Brain natriuretic peptides
cell apoptosis
A downregulation of beta-1 receptors in the heart BNP, the active peptide
NT-proBNP, the inactive N-terminal fragment
The decrease of cardiac output leads to a reduc-
tion of renal afferent arteriolar blood flow causing Atrial natriuretic peptides (ANP and NT-ANP)
secretion of renin and, subsequently, production of These peptides cause:
angiotensinogen and angiotensin I. The angioten-
sin I is then converted by the ACE present in the Natriuresis
lung to angiotensin II. There is also evidence that Vasodilatation
angiotensin II can be synthesized locally at a vari- Offset in the activation of RAAS
ety of tissue sites including the kidney, blood ves-
sels, adrenal gland, and brain [6]. Plasma ANP levels rise early in the course of
Angiotensin II increases aldosterone release, the disease and have been used as a marker for
inducing systemic and renal vasoconstriction. the diagnosis of asymptomatic left ventricular
Furthermore, angiotensin II can act directly on dysfunction.
myocytes and in the myocardium to promote All these processes are responsible for
pathologic remodeling as myocyte hypertrophy, sodium and water retention and a progressive
re-expression of fetal protein isoforms, myo- depression of myocardial function. The last step
cyte apoptosis, and alterations in the interstitial of this fall is an adverse remodeling of the left
matrix. Aldosterone-mediated effects are ventricle involving myocyte hypertrophy, death,
sodium and water retention and hypokalemia and fibrosis.
resulting in pulmonary and peripheral edema
and increased afterload. Activation of the
carotid sinus and aortic arch baroreceptors by Diagnosis
the low cardiac output in heart failure leads to
enhanced release of antidiuretic hormone and Clinical Symptoms and Signs
stimulation of thirst. Elevated levels of ADH The three most common symptoms and signs of
may contribute to the increase in systemic vas- HF are:
cular resistance in HF via stimulation of the
V1A receptor, which is found on vascular Breathlessness
smooth muscle cells, and also promote water Fatigue
retention via the V2 receptor by enhancing Peripheral edema
water reabsorption in the collecting tubules.
The combination of decreased water excretion Breathlessness is induced by exercise, and
and increased water intake via thirst often leads only in case of advanced heart failure, it appears
to a fall in the plasma sodium concentration. at rest. Symptoms that are more specific (i.e.,
The degree of hyponatremia is an important orthopnea and paroxysmal nocturnal dyspnea)
predictor of survival in these patients. are less common, especially in patients with
milder form of HF and who are, therefore, insen-
sitive [7, 8].
The Natriuretic Peptide System Many of the symptoms of HF are nonspecific
and do not, therefore, help discriminate between
The increased LV and left atrium wall stretch due HF and other problems.
to raised left ventricular end-diastolic pressure Symptoms are used to assign NYHA class to
leads to secretion of the natriuretic peptide hor- patients as listed in Table 8.2.

ERRNVPHGLFRVRUJ
86 S. Masiero and M. Morelli

Table 8.2 Since many of the symptoms of HF are non- To diagnose HF both of the following criteria
specific and do not, therefore, help discriminate between
should be present:
heart failure and other problems, patients are usually
assigned an NYHA class
Symptoms and/or signs of heart failure
New York Heart Association functional classification
based on the severity of symptoms and physical activity Cardiac dysfunction at rest
Class No limitation of physical activity. Ordinary
I physical activity does not cause undue The presence of cardiac dysfunction must be
breathlessness, fatigue, or palpitations proved to make the diagnosis of heart failure.
Class Slight limitation of physical activity. As stated in the current guidelines 1, investi-
II Comfortable at rest, but ordinary physical
gations that should be considered in all patients
activity results in undue breathlessness,
fatigue, or palpitations are:
Class Marked limitation of physical activity.
III Comfortable at rest, but less than ordinary
physical activity results in undue
A 12-lead ECG, to determine heart
breathlessness, fatigue, or palpitations
rhythm, heart rate, QRS morphology,
Class Unable to carry on any physical activity
VI without discomfort. Symptoms at rest can be and QRS duration and to detect other
present. If any physical activity is undertaken, relevant abnormalities. HF is rare in the
discomfort is increased presence of a normal ECG. The predic-
tive value of the ECG is >90 %.
Measurement of natriuretic peptide
(BNP, NT-proBNP) to exclude alterna-
tive causes of dyspnea. If the value is
Physical signs can be related to the presence within a normal range, HF is very
of either fluid retention or poor cardiac output. unlikely. Natriuretic peptide measure-
They usually include: ment has an extremely high negative
predictive value (>98 %).
A chest radiograph (X-ray), to detect/
exclude certain types of lung disease
Elevated jugular venous pressure (e.g., cancer, asthma/COPD). It may
Orthopnea allow also to identify pulmonary con-
Hepatojugular reflux gestion/edema. A normal X-ray does not
Third heart sound (gallop rhythm) exclude a diagnosis of heart failure.
Laterally displaced apical impulse Transthoracic echocardiography is the
Cardiac murmur key exam in the diagnostic process of
Peripheral edema (ankle, sacral, HF. It allows to evaluate cardiac struc-
scrotal) ture and function, to measure LV ejec-
Wheezing, pulmonary crepitations tion fraction, and to understand the
Weight gain (>2 kg/week) cause of cardiac dysfunction.
Reduced air entry and dullness to per- Measurement of blood chemistry
cussion at lung bases (pleural effusion) (including sodium, potassium, calcium,
Tachycardia urea/blood urea nitrogen, creatinine/
Bloated feeling, irregular pulse estimated glomerular filtration rate,
Tachypnea (>16 breaths/min) liver enzymes and bilirubin, ferritin/
Hepatomegaly total iron blood capacity) and thyroid.
Ascites A complete blood count to detect ane-
Palpitations mia, which may be a cause or effect of
Tissue wasting (cachexia) CHF.

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 87

following factors as well as addressing underly-


Furthermore, only in case of clinical ing and concurrent cardiovascular disease.
suspicions, the following investigations Treatment should address systemic contribut-
can be considered 1: ing factors (e.g., thyroid dysfunction, infection,
uncontrolled diabetes), as well as comorbidities
Coronary angiography in patients with such as chronic obstructive pulmonary disease
angina pectoris to evaluate the coronary and sleep apnea.
anatomy Recommendations for lifestyle modification
Myocardial perfusion/ischemia imaging are:
(echocardiography, CMR, SPECT, or
PET) in patients thought to have CAD Cessation of smoking
and who are considered suitable for cor- Restriction of alcohol consumption
onary revascularization, to determine Salt restriction (<2 g/day in patients with
whether there is reversible myocardial symptomatic HF)
ischemia or viable myocardium Fluid restriction (1.52 L/day) in patients with
Exercise testing to detect reversible refractory HF, particularly those with
myocardial ischemia or as part of the hyponatremia
evaluation of patients for heart trans- Weight reduction in obese subjects
plantation and mechanical circulatory Daily weight monitoring recommended to
support, to aid in the prescription of detect fluid accumulation
exercise training, and to obtain prognos- Appropriate preventative care including pneu-
tic information mococcal vaccination and annual influenza
CMR imaging: recommended to evalu- vaccination
ate cardiac structure and function, to
measure LVEF, and to characterize car-
diac tissue, especially in subjects with Review of Drugs
inadequate echocardiographic images
or where the echocardiographic findings The drugs that now form the cornerstones in the
are inconclusive management of HF are:
Left and right heart catheterization as
part of evaluation process for heart Diuretics
transplantation or mechanical circula- Angiotensin-converting enzyme inhibitors
tory support, to estimate right and left (ACEi) and angiotensin receptor antagonists
heart function and pulmonary arterial (ARBs)
resistance Beta-adrenoceptor antagonists
Mineralocorticoid receptor antagonist (MRA)
Nitrate plus hydralazine
Digoxin
Management

The goals of HF therapy are clinical relief of Loop Diuretics


symptoms and a reduction in the risk of morbid-
ity (including the rate of hospitalization) and Loop diuretics are generally introduced first for
mortality. fluid control in patients in overt HF. The goal is
Management of HF begins with an accurate relief of signs or symptoms of volume overload,
assessment of the underlying etiology, contribut- such as dyspnea and peripheral edema. In general
ing factors, and severity of the syndrome. This is loop diuretic therapy is based on furosemide,
followed by a therapeutic regimen aimed at the bumetanide, or torasemide. The aim is to use the

ERRNVPHGLFRVRUJ
88 S. Masiero and M. Morelli

minimum dose necessary to render the patient Table 8.3 Suggested dosage for ACEi and ARBs with
proved clinical efficacy for HF based on randomized clini-
euvolemic. The most common side effects are
cal trials
renal dysfunction, hypokalemia, hyponatremia,
hyperglycemia, and gout. ACE inhibitors Starting dose Target dose
(mg) (mg)
The usual starting dose in outpatients with HF
Captopril 6.25 t.i.d. 50 t.i.d
is 2040 mg of furosemide or its equivalent.
Enalapril 2.5 b.i.d. 1020 b.i.d.
Subsequent dosing is determined by the diuretic Lisinopril 2.55.0 o.d. 2035 o.d.
response. In patients who are volume overloaded, Ramipril 2.5 o.d. 5 b.i.d.
a reasonable goal is weight reduction of 1.0 kg/ Trandolapril 0.5 o.d. 4 o.d.
day. If a patient does not respond, the diuretic ARB Starting dose Target dose
dose should initially be increased to find the sin- (mg) (mg)
gle effective dose, rather than giving the same Candesartan 4 or 8 o.d. 32 o.d.
dose twice a day. Valsartan 40 b.i.d. 160 b.i.d.
Intravenous diuretics (either as a bolus or a Losartan 50 o.d. 150 o.d.
continuous infusion) are more potent than their
equivalent oral doses and may be required for
unstable or severe disease.
Some patients can develop a loop diuretic over, ACEi can induce cough (510 %) and
resistance if they are treated for a long time. In angioedema. ARB can be used in case of ACEi
this category of patients, it is useful to add a thia- intolerance.
zide diuretic (i.e., metolazone 2.55 mg/day) These drugs are usually started at low doses
instead of increasing the dose of loop diuretics and then titrated. ACEi and ARBs with proven
(i.e., >80 mg b.i.d. furosemide) to block differing clinical efficacy in HF based on randomized clin-
sites in the nephron and overcome the ical trials with their suggested dosing are listed in
resistance. Table 8.3.

Angiotensin-Converting Enzyme Beta-Adrenoceptor Antagonists


Inhibitors and Angiotensin Receptor
Antagonists Beta-adrenoceptor antagonists are also manda-
tory in patients with HF. Beta-blockers are initi-
ACE inhibitors or, if not tolerated, ARBs are ated after the patient is stable on ACE inhibitors,
typically initiated during or after the optimiza- again beginning at low doses with titration to trial
tion of diuretic therapy. ACEi and ARBs were goals as tolerated. The beta-blocker trials in HF
first used because of their vasodilatory effect in were carried out in patients receiving therapy
the treatment of HF. It was subsequently under- with an ACE inhibitor; thus, the improvement in
stood that their beneficial effects arise above all survival is additive to that induced by ACE inhib-
from the antagonism of the renninangiotensin itors [9, 10]. They unequivocally reduce both
system. mortality and morbidity in clinical trials with a
These drugs are now first-line agents for all 35 % relative risk reduction on average. They can
patients with HF, and unless there are contraindi- be used in patients with COPD but are contrain-
cations, their use should be considered manda- dicated in patients with significant reversible air-
tory in all patients. It has been proved that they way obstruction. Furthermore, they should be
reduce both mortality and morbidity in large ran- used with caution in patients with peripheral vas-
domized clinical trials with a relative risk reduc- cular disease even if there is not an absolute
tion of 2025 %. contraindication.
ACEi and ARB must be used with caution in Drugs with proven efficacy in clinical trials
patients with significant renal dysfunction; more- and the approved doses are listed in Table 8.4.

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 89

Table 8.4 Beta-blockers with proven efficacy in clinical Table 8.5 Approved dosages for mineralocorticoid
trials and approved dosages receptor antagonists
Starting dose Target dose MRA Starting dose Target dose
Beta-blocker (mg) (mg) Eplerenone 25 o.d. 50 o.d.
Bisoprolol 1.25 o.d. 10 o.d. Spironolactone 25 o.d. 2550 o.d.
Carvedilol 3.125 b.i.d. 2550
b.i.d.
Nebivolol 1.25 o.d. 10 o.d.
Metoprolol succinate (CR/ 12.5/25 o.d. 200 o.d.
creatinine be checked 12 weeks after starting
XL) spironolactone or eplerenone and periodically
thereafter. Patients with poor renal function are
particularly at risk for hyperkalemia. For these
Mineralocorticoid Receptor reasons they should be used with caution in
Antagonist (MRA) elderly.
The approved doses are listed in Table 8.5.
Spironolactone and eplerenone, which compete
with aldosterone for the mineralocorticoid
receptor, prolong survival in selected patients Other Drug Options
with HF as demonstrated in randomized con-
trolled trials [11]. Ivabradine
MRAs are recommended to treat HF in It should be considered for patients in NYHA II
patients who have NYHA functional class II and IV heart failure with a heart rate 70/min in sinus
a left ventricular ejection fraction (LVEF) 30 % rhythm and LVEF 35 % despite treatment with
or NYHA functional classes III to IV and an evidence-based doses of:
LVEF <35 %, who can be carefully monitored for
serum potassium and renal function. MRAs are ACEi (or ARB)
also recommended for patients post ST elevation Beta-blocker
myocardial infarction who are already receiving MRA
therapeutic doses of ACE inhibitor, have an
LVEF 40 %, and have either symptomatic HF It may be also considered for patients in
or diabetes mellitus and who can be carefully NYHA IIIV heart failure with a heart rate 70/
monitored for serum potassium and renal func- min in sinus rhythm and LVEF 35 % who are
tion. The serum potassium should be <5.0 mEq/L unable to tolerate beta-blockers (true
and estimated glomerular filtration rate should be asthmatics).
30 mL/min per 1.73 m. The endocrine side
effects of spironolactone result from nonselective
binding to androgen and progesterone receptors; Hydralazine Plus Nitrates
eplerenone has greater specificity for the miner-
alocorticoid receptor and therefore has a lower In African-American population the addition of
incidence of endocrine side effects (1 versus hydralazine plus oral nitrate therapy is recom-
10 % in clinical trials). Although eplerenone is mended for patients with persistent NYHA
associated with fewer endocrine side effects than classes IIIIV and LVEF <40 % despite optimal
spironolactone (i.e., painful gynecomastia), this therapy including a beta-blocker, ACEi (or ARB),
advantage must be weighed against the marked MRA (if indicated), and diuretics. Although the
difference in cost between the two drugs. It may evidence of benefit is stronger in blacks, the addi-
be reasonable to begin with spironolactone (25 tion of hydralazine plus oral nitrate may be con-
50 mg/day) and switch to eplerenone (25 and sidered in non-blacks who have persistent NYHA
after 4 weeks 50 mg/day) if endocrine side effects class II or IV despite optimal conventional
occur. It is essential that serum potassium and therapy.

ERRNVPHGLFRVRUJ
90 S. Masiero and M. Morelli

Digoxin Implantable Cardiac Debrillators

Digoxin is given to patients with HF and sys- Implantable cardiac defibrillators (ICDs) have
tolic dysfunction to control symptoms (such as revolutionized the management of heart failure.
fatigue, dyspnea, and exercise intolerance) and An ICD is an advanced form of pacemaker that
to patients with atrial fibrillation to control the can detect and treat arrhythmias. Several types of
ventricular rate. As demonstrated in the DIG ICDs can be implanted: a single bipolar lead
trial, 12 digoxin therapy was associated with a placed in the right ventricular apex or a dual-
significant reduction in hospitalization for HF chamber device with a further atrial lead to
but no benefit in terms of overall mortality. improve detection of atrial from ventricular
Digoxin is indicated in patients with left ven- arrhythmias. ICDs use the following criteria to
tricular systolic dysfunction (LVEF <40 %) who distinguish ventricular tachycardia or fibrillation:
continue to have NYHA functional class II, III,
and IV symptoms despite appropriate therapy Rate detection zone.
including an ACE inhibitor, a beta-blocker, an Rate stability based on the principle that ven-
aldosterone antagonist if indicated, and an addi- tricular tachycardia is a stable rhythm without
tional diuretic if necessary for fluid control. The a significant inter-beat variation.
usual daily dose of digoxin is 0.125 mg or less, Sudden onset: ventricular arrhythmias usually
based upon renal function. Based upon the data have a sudden onset.
from the DIG trial, the recommended serum
digoxin concentration is maintained between According to the current guidelines 1, recom-
0.5 and 0.8 ng/mL. mendations for the use of ICDs in patients with
It is a useful drug in patients with atrial HF are resumed in Table 8.6.
fibrillation to reach an adequate rate control. It
should be avoided in patients with ventricular
arrhythmias. Resynchronization Therapy

Resynchronization therapy aims to address the


Device Therapy problem of ventricular dyssynchrony in
HF. Dyssynchrony is a complex phenomenon
Device therapy in heart failure addresses two which occurs at three different levels:
potential consequences of left ventricular
dysfunction: Electrical dyssynchrony considered as an
intra- or interventricular conduction delay
Malignant arrhythmias that can lead to sudden usually manifested as a left ventricular bundle
death branch block.
Ventricular dyssynchrony Structural dyssynchrony results from disrup-
tion of the myocardial collagen matrix impair-
Prevention of sudden death is an important ing and electrical conduction and mechanical
goal in HF because heart failure is a pro- efficiency.
arrhythmogenic condition arising from combina- Mechanical dyssynchrony manifests as
tion of structural heart disease and electrolyte regional wall motion abnormalities leading to
imbalance. Ventricular arrhythmias, from prema- increased workload and stress, paradoxical
ture ventricular beats to ventricular fibrillation, septal wall motion, presystolic mitral regurgi-
occur in 80 % of patients with heart failure and tation, and reduced diastolic filling times.
cardiomyopathy. Up to 50 % of heart failure
deaths are sudden cardiac death, usually Cardiac resynchronization (CRT) typically
arrhythmic. uses leads in the right atrium, right ventricular

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 91

Table 8.6 Current guideline recommendations for the use of ICDs in patients with HF [1]
Recommendations Recommendation class Recommendation level
Secondary prevention IA A
An ICD is recommended in a patient with a ventricular
arrhythmia causing hemodynamic instability, who is expected to
survive for >1 year with good functional status, to reduce the
risk of sudden death
Primary prevention IA A
An ICD is recommended in a patient with symptomatic HF
(NYHA classes IIIII) and an EF 35 % despite 3 months of
treatment with optimal pharmacological therapy, who is
expected to survive for >1 year with good functional status, to
reduce the risk of sudden death
(i) Ischemic etiology and >40 days after acute myocardial IA
infarction
(ii) Nonischemic etiology IB B

apex, and coronary sinus. CRT functioning will Extracorporeal ventricular support (useful for
be discussed more in detail in a separate section a short-term time support)
of this book.
Based on the current guidelines 1, the current Intra-aortic balloon pump
indications for CRT implant are listed in Table 8.7. Pulsatile ventricular assist device (VAD)
Non-pulsatile VAD

8.3 Management Extracorporeal membrane oxygenation


of Refractory HF (ECMO) useful for a short-term time support
Intracorporeal ventricular assist devices
Although the majority of patients with heart fail- useful for a long-term time support
ure due to systolic dysfunction respond to opti-
mal medical therapy, some patients do not Pulsatile VAD
improve and usually experience rapid recurrence Non-pulsatile VAD
of symptoms. These patients have symptoms at Total artificial heart
rest and often require repeated prolonged hospi-
talizations. Specialized strategies are generally The intentions to use these devices are
considered for these patients, including: resumed in Table 8.8.
Short-term devices are used in general in
Continuous or cyclic intravenous positive inotro- patients with acute heart failure or cardiogenic
pic therapy with dobutamine or levosimendan shock as BTD, BTR, or BTT, while long-term
Mechanical circulatory support devices are generally reserved as BTT, or in some
Cardiac transplantation countries, as Italy, they are approved as
DT. Continuous-flow VADs are by far the most
commonly used long-term devices.
Mechanical Circulatory Support

Mechanical circulatory support is a collection Cardiac Transplantation


of technologies that can be used to offer
short-or long-term ventricular assistance for Cardiac transplantation is the final intervention for
patients with heart failure. They can be classi- patients who remain symptomatic despite optimal
fied as follows: medical and device therapy. The patient selection

ERRNVPHGLFRVRUJ
92 S. Masiero and M. Morelli

Table 8.7 Current indications for CRT implant


Recommendations for the use of CRT where the evidence is strongpatients in sinus rhythm with NYHA
functional class III and ambulatory class IV heart failure and a persistently reduced ejection fraction, despite
optimal pharmacological therapy
LBBB QRS morphology I A
CRT-P/CRT-D is recommended in patients in sinus rhythm with a QRS duration of 120 ms, LBBB
QRS morphology, and an EF 35 %, who are expected to survive with good functional status for
>1 year, to reduce the risk of HF hospitalization and the risk of premature death
Non-LBBB QRS morphology II A
CRT-P/CRT-D should be considered in patients in sinus rhythm with a QRS duration of 150 ms,
irrespective of QRS morphology, and an EF 35 %, who are expected to survive with good
functional status for >1 year, to reduce the risk of HF hospitalization and the risk of premature
death
Recommendations for the use of CRT where the evidence is strongpatients in sinus rhythm with NYHA
functional class II heart failure and a persistently reduced ejection fraction, despite optimal pharmacological
therapy
LBBB QRS morphology I A
CRT, preferably CRT-D, is recommended in patients in sinus rhythm with a QRS duration of
130 ms, LBBB QRS morphology, and an EF 30 %, who are expected to survive for >1 year with
good functional status, to reduce the risk of HF hospitalization and the risk of premature death
Non-LBBB QRS morphology II A
CRT, preferably CRT-D, should be considered in patients in sinus rhythm with a QRS duration of
150 ms, irrespective of QRS morphology, and an EF 30 %, who are expected to survive for >1 year
with good functional status, to reduce the risk of HF hospitalization and the risk of premature death

Table 8.8 Terms describing various uses of mechanical circulatory support (MCS)
Bridge to decision Use of MCS in patients with drug-refractory acute circulatory collapse and at immediate
(BTD) risk of death to sustain life until a full clinical evaluation can be completed and additional
therapeutic options can be evaluated
Bridge to candidacy Use of MCS to improve end-organ function in order to make an ineligible patient eligible
(BTC) for transplantation
Bridge to Use of MCS to keep a patient at high risk of death before transplantation alive until a donor
transplantation organ becomes available
(BTT)
Bridge to recovery Use of MCS to keep patient alive until intrinsic cardiac function recovers sufficiently to
(BTR) remove MCS
Destination therapy Long-term use of MCS as an alternative to transplantation in patients with end-stage heart
(DT) failure ineligible for transplantation

for transplantation is very restrictive; indeed patients Lip GY, Maggioni AP, Parkhomenko A, Pieske BM,
Popescu BA, Rnnevik PK, Rutten FH, Schwitter J,
have to be able to face the major cardiac surgery and
Seferovic P, Stepinska J, Trindade PT, Voors AA,
the massive immunosuppressive regime. Cardiac Zannad F, Zeiher A, Task Force for the Diagnosis and
transplantation is not a cure but is the last Treatment of Acute and Chronic Heart Failure 2012 of
chance even because 1 year mortality is approxi- the European Society of Cardiology, Bax JJ,
Baumgartner H, Ceconi C, Dean V, Deaton C, Fagard
mately 17 % with a median survival of 10.9 years.
R, Funck-Brentano C, Hasdai D, Hoes A, Kirchhof P,
Knuuti J, Kolh P, McDonagh T, Moulin C, Popescu
BA, Reiner Z, Sechtem U, Sirnes PA, Tendera M,
Torbicki A, Vahanian A, Windecker S, McDonagh T,
References Sechtem U, Bonet LA, Avraamides P, Ben Lamin HA,
Brignole M, Coca A, Cowburn P, Dargie H, Elliott P,
1. McMurray JJ, Adamopoulos S, Anker SD, Auricchio Flachskampf FA, Guida GF, Hardman S, Iung B,
A, Bhm M, Dickstein K, Falk V, Filippatos G, Merkely B, Mueller C, Nanas JN, Nielsen OW, Orn S,
Fonseca C, Gomez-Sanchez MA, Jaarsma T, Kber L, Parissis JT, Ponikowski P, ESC Committee for

ERRNVPHGLFRVRUJ
8 Chronic Heart Failure 93

Practice Guidelines (2012) ESC guidelines for the 7. Davie AP, Francis CM, Caruana L, Sutherland GR,
diagnosis and treatment of acute and chronic heart McMurray JJ (1997) Assessing diagnosis in heart fail-
failure 2012: The Task Force for the Diagnosis and ure: which features are any use? QJM 90:335339
Treatment of Acute and Chronic Heart Failure 2012 of 8. Kelder JC, Cramer MJ, van Wijngaarden J, van
the European Society of Cardiology. Developed in Tooren R, Mosterd A, Moons KG, Lammers JW,
collaboration with the Heart Failure Association Cowie MR, Grobbee DE, Hoes AW (2011) The diag-
(HFA) of the ESC. Eur J Heart Fail 14:803869 nostic value of physical examination and additional
2. Mosterd A, Hoes AW (2007) Clinical epidemiology testing in primary care patients with suspected heart
of heart failure. Heart 93:11371146 failure. Circulation 124:28652873
3. Francis GS, Goldsmith SR, Levine TB et al (1984) 9. Exner DV, Dries DL, Waclawiw MA et al (1999)
The neurohumoral axis in congestive heart failure. Beta-adrenergic blocking agent use and mortality in
Ann Intern Med 101:370 patients with asymptomatic and symptomatic left ven-
4. Dzau VJ (1987) Renal and circulatory mechanisms in tricular systolic dysfunction: a post hoc analysis of the
congestive heart failure. Kidney Int 31:1402 Studies of Left Ventricular Dysfunction. J Am Coll
5. Benedict CR, Johnstone DE, Weiner DH et al (1994) Cardiol 33:916
Relation of neurohumoral activation to clinical vari- 10. Cleland JG, McGowan J, Clark A, Freemantle N
ables and degree of ventricular dysfunction: a report (1999) The evidence for beta blockers in heart failure.
from the Registry of Studies of Left Ventricular BMJ 318:824
Dysfunction. SOLVD Investigators. J Am Coll 11. Zannad F, McMurray JJ, Krum H et al (2011)
Cardiol 23:1410 Eplerenone in patients with systolic heart failure and
6. Dzau VJ, Colucci WS, Hollenberg NK, Williams GH mild symptoms. N Engl J Med 364:11
(1981) Relation of the renin-angiotensin-aldosterone 12. Digitalis Investigation Group (1997) The effect of
system to clinical state in congestive heart failure. digoxin on mortality and morbidity in patients with
Circulation 63:645 heart failure. N Engl J Med 336:525

ERRNVPHGLFRVRUJ
Cardiogenic Shock
9
MariaVittoriaMatassini, LucaPiangerelli,
andMatildaShkoza

9.1 Case Report


desaturation and hypotension. He required
prompt intubation and mechanical ventila-
tion. Inotropic support with dopamine was
A 60-year-old man with a known dilated
started, and the patient was immediately
valvular cardiomyopathy was admitted to
transferred to our ICU for further evalua-
the emergency department (ED) for wors-
tion and treatment.
ening dyspnea and fatigue. The patient
and his wife referred the onset of dyspnea
a week before, firstly exercise related and
then at rest. The patient also referred  edical History andCardiovascular
M
weight gain (4 kg in 5 days) despite usual Risk Factors
food consumption and water restriction
and decrease in urine output within the Cardiovascular risk factors: dyslipidemia and
last day. type II diabetes mellitus.
His vital signs were 80/45 mmHg and Family history: no family history of structural
125 beats per minute and his oxygen satu- heart disease.
ration was 89 %. 1999: diagnosis of severe mitral regurgitation
Few minutes after his arrival in the ED, due to rheumatic valvular disease. Concomitant
while waiting for medical visit, patients diagnosis of left ventricular dilatation and
condition rapidly deteriorated with further moderate left ventricular systolic dysfunction
(LVEF 40 %).
2000: hospital admission for acute pulmonary
edema during atrial fibrillation episode with
rapid ventricular response. After the acute
phase, the patient underwent an echocardio-
graphic evaluation that confirmed severe
mitral regurgitation together with the increase
M.V. Matassini (*) L. Piangerelli M. Shkoza
in systolic pulmonary artery pressure (PAP
Clinica di Cardiologia e Aritmologia, Universit 55 mmHg), severe left atrial enlargement, and
Politecnica delle Marche, moderate to severe left ventricular systolic
Via Conca, 71, Ancona, Italy dysfunction (LVEF 38 %). A coronary angi-
e-mail: mavimatassini@yahoo.it; lucapiangerelli30@
gmail.com; matilda_shkoza@libero.it
ography did not reveal any coronary disease.

Springer International Publishing Switzerland 2015 95


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI10.1007/978-3-319-19926-9_9

ERRNVPHGLFRVRUJ
96 M.V. Matassini et al.

The patient was evaluated and accepted for Cardiovascular: irregular and tachycardic
mitral valve surgery. rhythm, apical soft proto-mesosystolic mur-
2001: mitral valve replacement with mechani- mur (2/6 at Levine scale)
cal prosthesis. Surgical atrial fibrillation abla- Lungs: decreased tactile fremitus and dullness
tion was ineffective. to percussion at right pulmonary basis, bilat-
2006: hospital admission for hypotensive acute eral medio-basal rales
heart failure with renal and hepatic impairment Abdomen: moderate hepatomegaly, no spleno-
treated with IV diuretics and inotropes. The megaly, no ascites, no pulsatile masses, normal
echocardiogram revealed correct prosthesis bowel sounds in all four quadrants, no high-
function but further worsening of left ventricu- pitched or tinkling sounds, resonant to percus-
lar function (FEVS 30 %). The patient referred sion, soft, non-distended/non-tender, no rebound
an NYHA class III. The patient was discharged or guarding, no costovertebral angle tenderness
on optimal medical therapy with indication to Extremities: cold, mild cyanosis, peripheral
a follow-up visit after 3 months. edema
2007: cardiac resynchronization therapy with
defibrillator (CRT-D) implantation.
20102013: frequent hospital admissions for Laboratory Tests
acute heart failure. An evaluation for heart trans-
plantation was proposed, but the patient refused. White blood cells 8800/mmc, hemoglobin
10.5 g/l, hematocrit 32 %, platelets 138,000/
mmc, creatinine 2.4 mg/dl, blood urea nitrogen
Allergies 112 mg/dl, AST 288 U/L, ALT 204 U/L, GT
110 U/L, total bilirubin 2.1 mg/dl with direct bili-
None. rubin of 1.6 mg/dl, INR 4.2, uric acid 9.4 mg/ml,
potassium 4.8 mEq/l, sodium 134 mEq/l, magne-
sium 1.2 mg/dl, blood glucose 254 mg/dl
Medications

Furosemide 125 mg in the morning + 75 mg in Arterial Blood Gas Analysis


the evening, ramipril 2.5 mg o.d., bisoprolol
3.75 mg o.d., spironolactone 100 mg o.d., atorv- Before orotracheal intubation and mechanical
astatin 20 mg o.d., and metformin 500 mg b.i.d. ventilation: pH 7.28, PO2 55 mmHg, PCO2 52,
lactate 6.7 mmol/L with decreased serum bicar-
bonate (HCO3 18 mmol/L)
Vital Signs

Temperature: 36 C Instrumental Examination


Heart rate: 125 bpm
Arterial blood pressure: 85/55 mmHg The ECG (Fig. 9.1) revealed atrial fibrillation
Respiratory rate: 16 breaths/min with rapid ventricular response (130 bpm).
Oxygen saturation: 98 % The echocardiographic examination showed
severely dilated left ventricle (LV end-diastolic vol-
ume of 280 ml, LV end-diastolic diameter 78 mm),
Physical Examination impaired LV function with an estimated ejection
fraction (EF) of 20 % because of global hypoki-
General: intubated, sedated; cold sweats and nesia, correct function of the mitral prosthesis with
pallor a mild intraprosthetic regurgitation, right ventricle
Neck: jugular venous distention, no lymph- dilatation and dysfunction (TAPSE 12 mm, FAC
adenopathy, no carotid bruit area 25 %), and severe tricuspid regurgitation with

ERRNVPHGLFRVRUJ
9 Cardiogenic Shock 97

Fig. 9.1 ECG showing atrial fibrillation with rapid ventricular response

a b

Fig. 9.2 Echocardiographic images: apical four-chamber view (a) and parasternal short axis at papillary level (b) show-
ing chamber dilation, mitral valve prosthesis, and presence of lead in right chambers

systolic pulmonary artery pressure of 50 mmHg  linical Course andTherapeutic


C
(Fig. 9.2). The cardiac output and the cardiac index Management
as estimated by echocardiography were, respec-
tively, 2.8 l/min and 1.3 l/min/m2. The patient underwent insertion of a radial arte-
Urgent chest radiography was performed show- rial catheter for invasive measurement of blood
ing pulmonary vascular congestion with alveolar pressure and arterial blood gas testing, insertion
infiltrates and right moderate pleural effusion. of central venous catheter at right subclavian site

ERRNVPHGLFRVRUJ
98 M.V. Matassini et al.

for drug infusions, measurement of central Considering the end-stage HF despite optimal
venous pressure and blood sampling, and deter- pharmacological and device treatment and the
mination of central venous oxygen saturation. recent episode of cardiogenic shock, the possibil-
Clinical, instrumental, and laboratory data ity to an evaluation for advanced treatment
(hypotension <90 mmHg, tissue hypoperfusion options, as heart transplant or left ventricular
with increase in arterial lactate and renal and assistive device implantation, was offered to the
hepatic impairment, cardiac index of 1.3 l/min/ patient who agreed, and a visit in a national refer-
m2) allow us to make the diagnosis of cardiogenic ence hospital was therefore planned. The patient
shock in a patient with advanced heart failure. was discharged on day 25.
Inotropic support with dopamine (5 mcg/kg/
min) was continued, and also adrenaline was
started at a dose of 0.05 up to 0.08 mcg/kg/min 9.2 Cardiogenic Shock
because of persistent hypotension, increase in
arterial lactate (7.8 mmol/l), and oliguria. Definition andEpidemiology
Continuous infusion of loop diuretics (furose-
mide, 500 mg/24 h) was started with progressive Cardiogenic shock (CS) is a complex clinical
improvement in diuresis. Intravenous digoxin condition characterized by inadequate end-organ
was administered in the acute phase for AF rate perfusion due to the inability of the heart to pro-
control, and unfractionated heparin was intro- vide adequate flow. The tissue hypoperfusion, if
duced. Insulin infusion was started to correct prolonged, could result in end-organ damage and
hyperglycemia and on the following days finally in multiorgan failure. Cardiogenic shock
switched to bolus insulin injection. is a fatal condition if not early diagnosed and
The patients clinical status gradually treated. The in-hospital mortality approaches
improved, with normalization of arterial lactate 50 % and is related to the severity of hemody-
on day 2 and also the central venous oxygen satu- namic impairment, the promptness of diagnosis,
ration (from baseline 5663 % on day 2). On day and the type of management (medical therapy,
5, pulmonary congestion significantly improved mechanical support) [1]. Mortality decreased sig-
and the patient was extubated. The patient was nificantly during the last years because of the
gradually weaned from inotropes until interrup- wide use of revascularization.
tion on day 7. Therefore, an echocardiography The diagnosis of CS results from multipara-
was repeated confirming severe left ventricular metric evaluation and could be made in the pres-
dysfunction (FEVS 25 %) and mild to moderate ence of:
right ventricle dysfunction (TAPSE 15 mm, FAC
area 32 %), reducing pulmonary artery pressure Hypotension defined as systolic blood pres-
(35 mmHg). sure 90 mmHg or when vasopressors are
A beta-blocker and an angiotensin-converting required to maintain SBP 90 mmHg or mean
enzyme inhibitor were started on day 8 and up- arterial pressure is 30 mmHg lower than
titrated (ramipril 2.5 mg o.d., bisoprolol 3.75 mg baseline
o.d.). Loop diuretics were switched from IV to oral Evidence of organ hypoperfusion: resting tachy-
administration (furosemide 125 mg b.i.d., spirono- cardia, altered mental status, oliguria, poor cap-
lactone 100 mg o.d.). Oral digoxin was continued illary refill, cold/diaphoretic extremities
to achieve a better rate control. Oral anticoagula- A reduction in cardiac index (<1.8 L/min/mq
tion was reintroduced on day 9. Laboratory tests without support or <2.2 l/min/mq with sup-
showed improvement of kidney and hepatic func- port) with evidence of increase in pulmonary
tion (creatinine 1.4 mg/dl, total bilirubin 1.3 mg/dl, capillary wedge pressure (>18 mmHg)
normalization of AST, ALT, and GT).
Cardiac rehabilitation was started on day 12 with Any cause of severe left or right ventricle dys-
progressive improvement of functional capacity. function may cause cardiogenic shock; however,

ERRNVPHGLFRVRUJ
9 Cardiogenic Shock 99

acute coronary syndrome (ACS) with left ventricu- mechanisms. The initial response to a decrease in
lar failure is mostly involved. The incidence of CS blood pressure is mediated by arterial barorecep-
complicating ACS is approximately 7 % in ST ele- tors that cause an enhancement in sympathetic
vation myocardial infarction (STEMI) and 2.5 % in activity (via IX and X cranial nerves) with a con-
non-STEMI [2]. In those patients presenting with sequent increase in HR, myocardial contractility,
ACS and CS, mechanical complications as ventric- and SVR. More slowly acting mechanisms are
ular septal or free wall rupture and papillary muscle the activation of renin/angiotensin/aldosterone
rupture should be suspected and searched. system and fluid retention. The reduction in tis-
Other less frequent causes are acute myoperi- sue perfusion leads to a reduced oxygen delivery
carditis, stress-induced cardiomyopathy, acute with a shift to anaerobic metabolism and an
valvular regurgitation or prior severe valvular increase in lactate levels with a possible conse-
disease, hypertrophic cardiomyopathy, dilated quent metabolic acidosis.
cardiomyopathy, drugs and medications, arrhyth- Cardiogenic shock (CS) may be precipitated
mias, and traumatic cardiac injury. by different cardiac and extracardiac causes as
Cardiogenic shock due exclusively to right listed below:
ventricle involvement represents only 5 % of
cases, and it is characterized by a similar mortal- Cardiomyopathies: acute myocardial infarc-
ity to LV shock [3]. tion (MI) involving >40 % of the left ventricu-
In the ischemic setting, CS could be present lar myocardium or dilated cardiomyopathy
acutely or could develop later, within the first days. with cardiac pump failure.
It seems that later CS is associated with a higher Arrhythmias: supraventricular arrhythmias
mortality than earlier development of CS [4]. may cause cardiogenic shock through an
Different risk factors for CS in ischemic impairment of left ventricular filling.
patients have been recognized: anterior STEMI, Bradyarrhythmias or ventricular tachycardia/
multivessel disease, advanced age, female sex, fibrillation may reduce or abolish CO due to
previous diagnosis of diabetes and hypertension, an ineffective cardiac contraction.
prior cardiovascular disease, heart failure at Mechanical: valvulopathies (mitral or aortic
admission, systolic blood pressure <120 mmHg, regurgitation) or intracardiac shunt.
heart rate >90 bpm, and presence of left branch Extracardiac: any condition that causes a sig-
block [5, 6]. nificant reduction in preload or acute increase
in afterload (i.e., cardiac tamponade, pulmo-
nary embolism, tension pneumothorax, con-
Pathophysiology strictive pericarditis).

Systemic perfusion and blood pressure are related The CS pathophysiology is very complex with
to cardiac output (CO) and systemic vascular differences from patient to patient. Cardiogenic
resistance (SVR): shock evolves through different stages that repre-
sent a physiologic continuum from an initially
MAP ( mean arterial pressure ) = CO SVR compensated status (pre-shock or shock impend-
SVR =8hL / p r 4 ing) till multiorgan failure.
CO = SV HR Regardless of the precipitating cause, the main
feature consists of a reduction of cardiac output
= viscosity, L = vessel length, r = vessel radius, with consequent hypotension, unable to maintain
SV = stroke volume, HR = heart rate an adequate systemic perfusion (Fig. 9.3). The
reduction of blood pressure triggers the activa-
The stroke volume depends on the preload, tion of compensatory mechanisms through sym-
afterload, and myocardial contractility as pathetic system and renin/angiotensin/
explained by the FrankStarling and Hill aldosterone system with consequent tachycardia,

ERRNVPHGLFRVRUJ
100 M.V. Matassini et al.

SIRS Myocardial infraction

Myocardial dysfunction
Inotropes/
vasopressors Inflammatory cytokines Systolic Diastolic
Mechanical IL-6, TNF-
support: Cardiac output
IABP/LVAD stroke volume LVEDP
Pulmonary
iNOS/eNOS Hypotension congestion

Bleeding/ Systemic
perfusion Coronary
transfusion perfusion pressure Hypoxemia
NO
Peroxynitrite Ischemia

Perfusion:
PCI/CABG
Compensatory vasoconstriction Progressive
Vasodilation myocardial
SVR Fluid retention
dysfunction

MOF

Death

Fig. 9.3 Concept of CS pathophysiology. This is the (TNF)) production and consequent endothelial and induc-
downward spiral, induced by left ventricle (LV) systolic ible nitric oxide synthase (eNOS, iNOS) activation leads
dysfunction that leads to reduced stroke volume and car- to nitric oxide (NO) and peroxynitrite production that
diac output with consequent hypotension. Coronary blood causes reduced systemic vascular resistance (SVR) with
flow is therefore reduced with ischemia and further myo- vasodilatation and further myocardial depression.
cardial dysfunction. Even diastolic dysfunction with Bleeding complications and subsequent transfusions have
increased left ventricular end-diastolic pressure (LVEDP) a negative role in the shock spiral. If there is not a prompt
and pulmonary edema leads to hypoxemia and consequent intervention with treatment options shown in red (inotro-
ischemia. The reduced systemic perfusion activates com- pes/vasopressors, mechanical support with intra-aortic
pensatory mechanisms that cause vasoconstriction and balloon pumping (IABP) and left ventricular assist device
fluid retention, increasing left ventricle after- and preload (L-VAD), reperfusion by percutaneous coronary interven-
and aggravating myocardial dysfunction. The systemic tion (PCI) or coronary artery bypass (CABG)), the vicious
inflammatory response syndrome (SIRS) characterized by circle leads to multiorgan failure (MOF) and death
cytokine (interleukin-6 (IL-6) and tumor necrosis factor

increased contractility, a marked systemic vascu- Pump failure causes diastolic dysfunction
lar resistance (SVR) elevation increasing LV with increased ventricular diastolic pressure
afterload, and fluid retention with increase in pre- that further reduces coronary perfusion pres-
load. These compensatory mechanisms in the sure, worsening ischemia. The increased ven-
long term become maladaptive and result in a fur- tricular diastolic pressure increases left atrial
ther marked reduction in tissue perfusion. pressure which may cause pulmonary conges-
Hypotension, vasoconstriction, tachycardia, and tion leading to hypoxia, further exacerbating
increased myocardial contractility reduce myo- myocardial ischemia. Ischemia worsening
cardial perfusion and increase myocardial oxy- aggravates myocardial dysfunction and begins
gen demand, exacerbating ischemia. a vicious cycle that leads to progressive

ERRNVPHGLFRVRUJ
9 Cardiogenic Shock 101

end-organ hypoperfusion with multiorgan fail- Diagnosis


ure (MOF) and death when not interrupted
(Fig. 9.3). Reduced systemic perfusion leads to The diagnostic evaluation during cardiogenic
anaerobic metabolism and consequently to lac- shock must not delay resuscitation if needed and
tic acidosis that further depresses myocardial must be conducted at the same time. Diagnostic
function. efforts should be made to recognize the stage of
Some patients do not have elevated SVR, pre-shock to prevent loss of the compensatory
suggesting that the compensatory vasoconstric- mechanisms and progression to shock and multi-
tion is not universal, and moreover, a systemic organ dysfunction.
inflammatory response may be involved [7],
contributing to myocardial dysfunction and  edical History andPhysical
M
hypotension (via vasodilatation). In fact, in the Examination
presence of myocardial infarct, cytokines (inter- Medical history may be collected from the patient
leukin-6 (IL-6), tumor necrosis factor (TNF)) or relatives in case of advanced shock. The pres-
that activate inducible nitric oxide synthase ence of cardiovascular risk factors or the history
(iNOS) are released, leading to increased levels of chest pain may suggest acute myocardial
of nitric oxide (NO) with consequent vasodila- infarction (MI). Additional information about
tation and worsening hypotension [8]. Nitric comorbidities or allergies should be recorded.
oxide and superoxide lead to peroxynitrite pro- Cardinal findings on physical
duction that impairs myocardial contractility examination are:
[9]. The inflammatory mediators lead to micro-
circulatory abnormalities like regional hetero- Hypotension: defined as absolute (PAS
geneity in blood flow which plays a very <90 mmHg or PAM <65 mmHg) or relative
important role in organ failure pathogenesis. In (P > 30 mmHg). Prominent and persistent
fact data shows that IL-6 levels correlate with hypotension (>30 min), despite volemic cor-
organ failure and mortality [10]. rection, may require inotropes to ensure ade-
Even if severe LV failure is the principal cause quate systemic perfusion.
of CS, other factors, here below listed, may con- Oliguria: decreased urine output (diuresis
tribute to hypotension: <0.5 mg/kg/h), consequence of renal hypoper-
fusion related to reduced cardiac output and
Hypovolemia due to bleeding with a hemor- blood redistribution to other vital organs.
rhagic shock superimposition or due to Cool and clammy skin: compensatory vaso-
diuretic therapy constriction to redirect blood flow to vital
Septic shock superimposition organs, causes cold, mottled, or diaphoretic
Severe preexistent valvular heart disease like skin.
critical aortic stenosis or new-onset valvular Altered mental status: ranges from agitation to
disease like severe mitral regurgitation delirium and coma.
Important bradycardia that causes low cardiac
output and hypotension in patients with Dyspnea, chest pain with tachycardia, and
reduced LV function due to acute MI tachypnea are often present. On cardiac ausculta-
Atrial arrhythmias with rapid ventricular tion, gallop rhythm or new murmurs may be
response or ventricular tachycardia found. Pulmonary congestion with diffuse crack-
Drugs lowering blood pressure (nitrates, beta- les is also a typical finding but may lack in about
blocker, calcium antagonists, ACE inhibitors, one-third of patients at presentation [11]. Jugular
diuretics, and morphine) venous distension and hepatomegaly are clinical
signs related to an increased preload, especially
These factors should be promptly detected during right ventricular failure. A capillary refill
and, when possible, corrected. time >2 s is a frequent finding and should be

ERRNVPHGLFRVRUJ
102 M.V. Matassini et al.

associated with low mixed venous oxygen artery catheterization has never shown to
saturation. improve patients outcomes in clinical trials [2,
3, 15]. The diagnosis of cardiogenic shock is
Electrocardiogram confirmed in the presence of a reduced cardiac
Electrocardiogram (ECG) suggests the diagnosis index (<2.2 l/min with inotropic support or <1.8
of acute MI in the presence of ST-T alterations. without therapy), an increased PCWP (>15
Supraventricular and ventricular tachy- or brady- 18 mmHg), and/or a reduction in SvO2/SvcO2
arrhythmias may cause shock and can be diag- (<70 % and <65 %, respectively). A fall in SvO2
nosed by ECG monitoring. Shocked patients is suggestive for a reduced oxygen delivery or an
usually present sinus tachycardia. increase in oxygen consumption and may reflect
inadequate tissue perfusion even in a pre-shock
Echocardiogram stage. According to guidelines [12], an invasive
Echocardiography may confirm the diagnosis of hemodynamic monitoring is recommended in a
cardiogenic shock, showing marked depression patient with persistent hypotension refractory to
of left or right ventricular function with low pharmacological treatment with uncertain left
stroke volume and elevated filling pressures. It is filling pressures.
also useful in evaluating cardiac chambers,
regional wall motion, the pericardium, and Laboratory Evaluation
valves: it could detect causes or contributing fac- Laboratory tests are useful in identifying causes
tors as regional wall motion abnormalities, the of shock and in evaluation of organ failure. Basic
presence of cardiac tamponade, or severe mitral chemistry tests, complete blood count, liver and
or aortic regurgitation. In acute MI, echocardio- renal function tests, amylase and lipase, and arte-
gram should be repeated to exclude the presence rial blood gas should be evaluated. Cardiac bio-
of mechanical complications as ventricular sep- markers (troponin T/I, CK-MB) are useful in the
tal, free wall, or papillary muscle rupture [12]. diagnosis of acute MI and correlate with infarc-
Transthoracic echocardiography (TTE) is the tion extension. Arterial or venous lactates com-
first step, but transesophageal echocardiography plete the picture because an increased serum
(TEE) should be used when TTE images are sub- lactate level may correlate with a reduced oxygen
optimal especially in patients with mechanical delivery with a shift to anaerobic metabolism.
ventilation. Elevated lactate serum levels (>1.5 mmol/l at
TTE plays also a role as a less invasive tool for admission, >1 mmol/l after 24 h) are also associ-
evaluating hemodynamic parameters with ated with increased mortality [16].
Doppler-based methods. Small ventricles (kiss-
ing ventricles) usually suggest the use of fluid Coronary Angiography
challenge, while a dilated and hypokinetic right Coronary angiography should be performed
ventricle should be related to pulmonary early in patients with suspected acute MI.
embolism. Revascularization with percutaneous coronary
intervention (PCI) or coronary artery bypass graft
Hemodynamic Monitoring surgery (CABG) is also recommended without
Hemodynamic monitoring through a pulmonary any delay in patients with cardiac pump failure
artery catheter adds further details in the diagno- related to an ischemic cause [12].
sis, establishing cardiac output, pulmonary
artery occlusion pressure (PCWP), systemic vas-
cular resistance, and continuous mixed venous Differential Diagnosis
oxygen saturation (SvO2) [13]. These parameters
are also helpful in guiding inotropic/vasopressor The differential diagnosis for shock may be chal-
therapy or fluid resuscitation and in assessing lenging and must be focused on underlying
mechanical ventilation settings [14]. Pulmonary causes of inadequate tissue perfusion.

ERRNVPHGLFRVRUJ
9 Cardiogenic Shock 103

1. Hypovolemic shock hypovolemic shock is The principal therapeutic targets in these


related to an intravascular volume loss due to patients are:
hemorrhage or third-space loss with a
reduced preload that leads to a reduction in MAP >65 mmHg in order to restore tissue
CO and an increase in systemic vascular perfusion and to prevent multiorgan dysfunc-
resistance to maintain adequate perfusion. tion (MOF)
Dry mucous and decreased jugular and cen- Systemic vascular resistances: 800
tral venous pressure with low PCWP are 1000 dyn/s/cm5
typical findings. CI >2.5 l/min/m2
2. Distributive shock distributive shock is FC <110 bpm
related to vasodilatation with a consequent SVO2 >65 %
drop in SVR. The CO is increased as a com- Lactate <2 mmol/l
pensatory mechanism, while the PCWP
may be low or normal. Possible causes of These parameters should be monitored every
distributive shock are sepsis or systemic 90 min.
inflammatory response syndrome, anaphy- Reversible causes should be detected and
laxis, neurogenic disease, and toxic treated emergently, for instance, revasculariza-
problems. tion in acute coronary syndromes, surgery for
3. Cardiogenic shock as discussed previously, mechanical complications of MI or acute valvu-
cardiogenic shock is related to a cardiac pump lar disease, and pericardial drainage in
failure and may be divided in to four catego- tamponade.
ries in relation to etiology. In patients with evidence of ACS (ongoing
4. Combined shock different mechanisms con- ischemia, persistent ST elevation, new LBBB),
tribute to generate shock (i.e., septic shock early revascularization with either PCI or CABG
may coexist with a cardiac pump failure due must be considered. In the SHOCK trial, CS
to myocardial stunning related to sepsis). patients treated emergently with PCI or CABG
had an improved long-term survival than patients
who did not or underwent revascularization later
Management [17]. The CS is the only situation in which an
emergency multivessel revascularization can be
The initial approach consists of: performed [17, 18]. Fibrinolysis should be con-
sidered when PCI or CABG is not available [19].
Identification of patients with high risk to Indications for antiplatelet/antithrombin thera-
develop CS (they should be transferred to the pies are similar to those in STEMI patient with-
nearest tertiary center) out CS (see Chap. 1).
Early diagnosis (before a frank hypotension is When a reversible underlying cause is not
being manifested) with a consequent rapid present, the medical management role is primar-
stabilization ily supportive, serving as a bridge to mechanical
Identification/treatment of reversible causes circulatory support, heart transplantation, or
recovery, because there is lack of evidence that
Invasive blood pressure, heart rate, rhythm, the medical management alone improves
and oxygen saturation should be continuously survival.
monitored. Right heart catheterization is not Even if there are only a few clinical trials with
routinely recommended, but it can be very use- discording results, sympathomimetic inotropic
ful in a subgroup of patients (persistently hypo- and vasopressor agents are the mainstay of the
tensive patients or patients with uncertain LV medical first-line therapy. These agents interact
filling pressure) to guide optimal treatment with specific receptors (Table 9.1) and activate
decisions. adrenergic pathways, increasing myocardial

ERRNVPHGLFRVRUJ
104 M.V. Matassini et al.

contractility and modifying vascular tone. The dopamine as the first-line inotropic therapy
principal inotropes and vasopressors that we in CS.
commonly use in CS are (Table 9.2): Epinephrine (adrenaline): It is a potent agonist
Dobutamine: It is predominantly a of all adrenoreceptors. Its use results in HR, SV,
-adrenergic agonist with a 1/2 ratio of 3:1. It CO, and coronary blood flow increase. At low
increases HR, SV, and CO with a modest decrease doses, a passive pulmonary vessel stretching
in blood pressure and SVR [20]. It also has a mild accommodates CO increase, but at high doses, it
1-adrenergic agonism, and this is the reason determines a pulmonary vascular resistance
why vascular resistance decrease does not persist increase and so a right ventricle afterload increase.
at higher doses. These beneficial effects are lim- Even adrenaline increases myocardial oxygen con-
ited by myocardial oxygen consumption increase sumption due to increase in HR and stroke work. It
that worsens myocardial ischemia, precipitates has metabolic effects like increased plasma glucose
tachyarrhythmias, and increases mortality. The and lactate concentration. The lactate concentra-
ESC guidelines recommend dobutamine and tion increase seems not to be harmful.
Norepinephrine (noradrenaline): It is a potent
Table 9.1 Location and response of adrenergic -agonist that also stimulates 1 receptors, with
receptors an increase in blood pressure, SVR, and SV. Like
Receptor Location Activity adrenaline, it increases right ventricle afterload.
1 Vascular smooth Contraction Either cerebral circulation or coronary circulation
muscle Increase force of is protected to a certain extent from these vaso-
Heart contraction constrictor effects due to the relative paucity of
2 Vascular smooth Contraction the vascular adrenoreceptors, while pulmonary,
muscle
renal, splanchnic, and cutaneous blood flow is not
1 Heart Increase force of
contraction spared. The ESC guidelines recommend nor-
Increase AV nodal adrenaline as second-line therapy in CS patients.
conduction velocity Dopamine: It has a dose-dependent action.
2 Smooth muscle Relaxation At low doses (2 /kg/min), it activates dopami-
(vascular,
nergic receptors with splanchnic and renal vaso-
bronchial, GI, and
GU) dilatation. At medium doses (510 /kg/min), it
D Vascular smooth Relaxation activates 1 receptors with HR and CO increase.
muscle At intermediate doses (25 /kg/min), either dopa-
AV node atrioventricular node, GI gastrointestinal, GU minergic or 1 receptors are stimulated. At high
genitourinary doses, it predominates -adrenergic action with

Table 9.2 Inotropes and vasopressors used in CS


Medication Receptor/mechanism Doses BP HR CO SVR
Dobutamine 1 > 2 > 215 /kg/min
Milrinone PDE II inhibitor 0.3750.75 /kg/min
Levosimendan Ca sensitizer 0.050.2 /kg/min 0 0
Epinephrine 1 = 2 > 0.010.03 /kg/min, max
0.10.3 /kg/min
Norepinephrine > 1 > 2 0.010.03 /kg/min, max 0.1 /kg/ 0 or 0
min
Dopamine Moderate dose 510 /kg/min 0 or
Dopamine High dose 1020 /kg/min
Phenylephrine 1 6060 /min
Vasopressin V1 0.010.04 units/min 0 0
BP blood pressure, CO cardiac output, HR heart rate, SVR systemic vascular resistance

ERRNVPHGLFRVRUJ
9 Cardiogenic Shock 105

vasoconstriction and increase in SVR that may reverse beta-blocker effects if the last ones are
cause a CO decrease. De Backey et al. showed in thought to contribute to hypotension.
a subgroup analysis that in CS patients dopamine Phenylephrine: It is an 1-selective agonist
increased 28-day mortality rate compared with that causes an increase in SVR and blood pres-
norepinephrine, but in this study dopamine doses sure and a reflex bradycardia that determines a
in CS patients are not specified, and this may be decrease in CO. This is the reason why its utiliza-
a reasonable explanation of these results because tion in CS is very rare.
high-dose dopamine causes a CO decrease [21]. Vasopressin: It activates V1 vascular smooth
Milrinone: It inhibits phosphodiesterase-3 and receptors and causes vasoconstriction. In refrac-
prevents cyclic adenosine monophosphate tory CS, vasopressin has been utilized, increasing
(cAMP) degradation that activates protein kinase MAP without effects on CI, pulmonary capillary
A which results in increased calcium influx into wedge pressure, or urine output [24].
the cardiomyocyte with increased contractility. In Assessment and optimization of cardiac filling
the smooth muscle, elevated cAMP causes relax- pressure enhance hemodynamic improvement in
ation (vasodilatation) because it inhibits myosin CS. Hypovolemia should be treated with intrave-
light-chain kinase. Milrinone has a similar car- nous fluid replacement, and this should be guided
diovascular profile to dobutamine. In fact it by PCWP, systemic arterial pressure, arterial
increases HR, SV, and CO and decreases mean oxygen saturation, central venous pressure (tar-
blood pressure, SVR, and pulmonary artery resis- get value 810 mmHg if there is no right ventri-
tances, reducing preload and afterload and conse- cle (RV) dysfunction, 1012 mmHg if RV
quently ventricular wall stress. Although dysfunction), and cardiac output measurement.
milrinone affects hemodynamics, the OPTIME- When hypervolemia with pulmonary and periph-
CHF trial did not show a difference in days of eral edema is present, diuretics (loop diuretics or
hospitalization between decompensated heart combining loop with thiazide diuretic when patient
failure patients treated with 48 h administration becomes resistant to the first ones) should be used.
of milrinone and placebo. In this study, there was Actually the intra-aortic balloon pumping
not an increase in in-hospital mortality in the mil- (IABP) may be considered in patients with acute
rinone group [22]. Actually milrinone is recom- myocardial infarction complicated by CS. The
mended only for refractory CS patients. evidence does not support the IABP routine use
Levosimendan: It is a calcium-sensitizing because the IABP SHOCK II trial did not show a
agent that binds troponin C, at systolic calcium 30-day mortality difference between the IABP
concentrations, and prolongs myosinactin inter- and control groups in patients with CS compli-
action due to troponin I inhibition. So levosimen- cating MI, probably due to high rate of patient
dan does not increase cellular calcium shift from the control to the IABP group [25].
concentration and consequently does not impair In patients with refractory shock, LV mechan-
diastolic function and cardiac rhythm. It has phos- ical device may be considered. The percutaneous
phodiesterase III inhibitory effects and causes circulatory support devices can be distinguished
blood pressure decrease. Levosimendan has an in four categories:
active metabolite so its inotropic effects continue
even after infusion is stopped. The SURVIVE Mechanical left ventricle support that unloads
study did not show a 180-day mortality rate LV pressure (IABP)
through short-term levosimendan and dobutamine Mechanical left ventricle support that unloads
infusion in acute decompensated heart failure LV volume (TandemHeart and Impella
[23], but deaths in the first weeks were signifi- Recover 2.5 l/min or 4 l/min)
cantly fewer in the levosimendan group. There are Mechanical biventricular support (combina-
only few data (limited to case reports) on the role tion of right and left ventricle support)
of levosimendan in CS patients. The ESC guide- Mechanical biventricular support with mem-
lines recommend levosimendan infusion to brane oxygenation (ECMO)

ERRNVPHGLFRVRUJ
106 M.V. Matassini et al.

Oxygen or mechanical respiratory support is 12. OGara PT, Kushner FG, Ascheim DD et al (2013)
ACCF/AHA Guideline: 2013 ACCF/AHA Guideline
indicated according to clinical and blood gas
for the Management of ST-Elevation Myocardial
asset. Infarction: A Report of the American College of
Cardiology Foundation/American Heart Association
Task Force on Practice Guidelines. Circulation
128:1810
References 13. Mueller HS, Chatterjee K, Davis KB et al (1998) ACC
expert consensus document. Present use of bedside
1. Reynolds HR, Hochman JS (2008) Cardiogenic right heart catheterization in patients with cardiac dis-
shock: current concepts and improving outcomes. ease. J Am Coll Cardiol 32:840
Circulation 117:686697 14. Mimoz O, Rauss A, Rekik N et al (1994) Pulmonary
2. Holmes DR Jr, Berger PB, Hochman JS, Granger CB, artery catheterization in critically ill patients: a pro-
Thompson TD, Califf RM et al (1999) Cardiogenic spective analysis of outcome changes associated with
shock in patients with acute ischemic syndromes with catheter-prompted changes in therapy. Crit Care Med
and without ST-segment elevation. Circulation 22:573
100(20):20672073 15. Harvey S, Harrison DA, Singer M et al (2005)
3. Jacobs AK, Leopold JA, Bates E, Mendes LA, Sleeper Assessment of the clinical effectiveness of pulmonary
LA, White H, Davidoff R, Boland J, Modur S, Forman artery catheters in management of patients in inten-
R, Hochman JS (2003) Cardiogenic shock caused by sive care (PAC-MAN): a randomized controlled trial.
right ventricular infarction: a report from the SHOCK Lancet 366:472
registry. J Am Coll Cardiol 41:12731279 16. Smith I, Kumar P, Molloy S et al (2001) Base excess
4. Carnendran L, Abboud R, LA Sleeper R, Gurunathan and lactate as prognostic indicators for patients admit-
J, Webb G, Menon V, Dzavik V, Cocke T, Hochman ted to intensive care. Intensive Care Med 27:7483
JS, for the SHOCK Investigators (2001) Trends in 17. Hochman JS, Sleeper LA, Webb JG, Sanborn TA,
cardiogenic shock: report from the SHOCK Study. White HD, Talley JD, Buller CE, Jacobs AK, Slater
Eur Heart J 22:472478 JN, Col J, McKinlay SM, LeJemtel TH, Early revas-
5. Lindholm MG, Kober L, Boesgaard S et al (2003) cularization in acute myocardial infarction compli-
Cardiogenic shock complicating acute myocardial cated by cardiogenic shock. SHOCK Investigators
infarction: prognostic impact of early and late shock (1999) Should We Emergently Revascularize
development. Eur Heart J 24:258265 Occluded Coronaries for Cardiogenic Shock. N Engl
6. Goldenberg EJ, Frederisk AS, Gore JM, Lessard D, J Med 341:625634
Yarzebski J (2009) Thirty-year trends (1975 to 2005) 18. Hussain F, Philipp RK, Ducas RA, Elliott J, Dzavik
in the magnitude of management of, and hospital V, Jassal DS, Tam JW, Roberts D, Garber PJ, Ducas
death rates associated with cardiogenic shock in J (2011) The ability to achieve complete revascu-
patients with acute myocardial infarction. A larization is associated with improved in-hospital
population-based perspective. Circulation survival in cardiogenic shock due to myocardial
119:12111219 infarction: Manitoba cardiogenic SHOCK Registry
7. Kohsaka S, Menon V, Lowe AM et al (2005) Systemic investigators. Catheter Cardiovasc Interv 78:
inflammatory response syndrome after acute myocar- 540548
dial infarction complicated by cardiogenic shock. 19. Steg PG, James SK, Atar D, Badano LP, Blmstrom-
Arch Intern Med 165:16431650 Lundqvist C, Borger MA, Di Mario C, Dickstein
8. Shah AM (2000) Inducible nitric oxide synthase and K, Ducrocq G, Fernandez-Aviles F, Gershlick
cardiovascular disease. Cardiovasc Res 45:148155 AH, Giannuzzi P, Halvorsen S, Huber K, Juni P,
9. Ferdinandy P, Danial H, Ambrus I et al (2000) Kastrati A, Knuuti J, Lenzen MJ, Mahaffey KW,
Peroxynitrite is a major contributor to cytokine-induced Valgimigli M, vant Hof A, Widimsky P, Zahger
myocardial contractile failure. Circ Res 87:241247 D (2012) ESC Guidelines for the management of
10. Geppert A, Dorninger A, Delle-Karth G et al (2006) acute myocardial infarction in patients present-
Plasma concentrations of interleukin- 6, organ failure, ing with ST-segment elevation. Eur Heart J 33(20):
vasopressor support, and successful coronary revascu- 25692619
larization in predicting 30-day mortality of patients 20. Colucci WS, Wright RF, Jaski BE et al (1986)
with cardiogenic shock complicating acute myocar- Milrinone and dobutamine in severe heart failure: dif-
dial infarction. Crit Care Med 34:20352042 fering hemodynamic effects and individual patient
11. Menon V, White H, LeJemtl T et al (2000) The clini- responsiveness. Circulation 73:III175III183
cal profile of patients with suspected cardiogenic 21. De Backer D, Biston P, Devriendt J et al (2010)
shock due to predominant left ventricular failure; a Comparison of dopamine and norepinephrine in the
report from the SHOCK Trial Registry. Should we treatment of shock. New Engl J Med 362:779789,
emergently revascularize Occluded Coronaries in car- Clinical trial comparing two vasopressors in several
diogenic shock? J Am Coll Cardiol 36:1071 types of shock. Subgroup analysis suggests better

ERRNVPHGLFRVRUJ
9 Cardiogenic Shock 107

mortality in patients with cardiogenic shock treated 24. Jolly S, Newton G, Horlick E et al (2005) Effect of
with norepinephrine vasopressin on hemodynamics in patients with refrac-
22. Cuffe MS, Califf RM, Adams KF Jr et al (2002) tory cardiogenic shock complicating acute myocar-
Outcomes of a Prospective Trial of Intravenous dial infarction. Am J Cardiol 96:16171620
Milrinone for Exacerbations of Chronic Heart 25. Thiele H, Zeymer U, Neumann F-J, Ferenc M, Olbrich
Failure I. Short-term intravenous milrinone for acute H-G, Hausleiter J, Richardt G, Hennersdorf M,
exacerbation of chronic heart failure: a randomized Empen K, Fuernau G, Desch S, Eitel I, Hambrecht R,
controlled trial. JAMA 287:15411547 Fuhrmann J, Bhm M, Ebelt H, Schneider S,
23. Mebazaa A, Nieminen MS, Packer M et al (2007) Schuler G, Werdan K, for the IABP-SHOCK II Trial
Levosimendan vs dobutamine for patients with acute Investigators (2012) Intraaortic balloon support for
decompensated heart failure: the SURVIVE random- myocardial infarction with cardiogenic shock. N Engl
ized trial. JAMA 297:18831891 J Med 367:12871296

ERRNVPHGLFRVRUJ
Cardiac Resynchronization
Therapy and Possible 10
Dysfunctions

Laura Cipolletta and Michela Brambatti

10.1 Case Report Allergies

None
A 65-year-old man had been diagnosed
with congestive heart failure (HF) due to
dilated cardiomyopathy in 2013. He had Medications
been in chronic AF since 2012. He was
referred to the hospital with worsening HF Bisoprolol 5 mg, valsartan 40 mg b.i.d., furose-
in May 2014; however, his HF symptoms mide 40 mg t.i.d., spironolactone 50 mg q.d.,
remained New York Heart Association warfarin according to INR values
(NYHA) class III despite the administra-
tion of appropriate medical therapy. So he
was referred to our clinic for cardiac resyn- Vital Signs
chronization therapy (CRT).
Temperature: 36 C
Heart rate: 76 bpm
Arterial blood pressure: 130/75 mmHg
Medical History and Cardiovascular Respiratory rate: 14 breaths/min
Risk Factors Oxygen saturation: 96 %

No cardiovascular risk factor; he had a family


history of idiopathic dilated cardiomyopathy. Physical Examination
2013: diagnosis of nonischemic dilated
cardiomyopathy General Appearance: Well developed, well
nourished, alert, and cooperative
Lungs: Clear to auscultation and percussion,
L. Cipolletta (*) M. Brambatti several little rales at basal pulmonary fields,
Clinica di Cardiologia e Aritmologia, without rhonchi, wheezing, or diminished
Universit Politecnica delle Marche,
breath sounds
Via Conca, 71, Ancona 60126, Italy
e-mail: lauracipo2006@libero.it; Cardiovascular: Normal S1 and S2. S3, no S4,
michelabrambatti@gmail.com and 2/6 mitral systolic murmurs. Irregular

Springer International Publishing Switzerland 2015 109


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_10

ERRNVPHGLFRVRUJ
110 L. Cipolletta and M. Brambatti

rhythm. No peripheral edema, cyanosis, or Clinical Course and Therapeutic


pallor. Warm and well-perfused extremities Management
Abdomen: Positive bowel sounds. Soft, non-
distended. No guarding or rebound. No A CRT defibrillator (CRT-D) for primary preven-
masses tion of sudden cardiac death was implanted in
this high-risk patient in May 2014. Due to the
Routine laboratory test were normal (hemo- patients long-standing AF and severely dilated
globin 12.2 g/dl, white blood cells 7240/mmc, left atrium, we decided not to implant an atrial
creatinine 1.3 mg/dl, potassium 3.8 mEq/l, lead, which resulted in VVI biventricular pacing
sodium 138 mEq/l, magnesium 1.8 mg/dl). (Fig. 10.1). The interventricular interval was
determined at 0 ms guided by echocardiography.
Although the patients heart failure symptoms
Instrumental Examination improved from NYHA classes III to II and biven-
tricular pacing achieved a success rate of 95 %,
His 12-lead electrocardiogram (ECG) showed multiple episodes of high heart rate AF were
complete left bundle branch block with a QRS detected by the device one month after CRT-D
width of 167 ms. The chest X-ray showed a implantation, and oral amiodarone therapy consist-
severely enlarged cardiac silhouette. ing of 200 mg once daily for 5 days per week was
Two-dimensional echocardiography started. Three months after the initiation of amiod-
revealed a severely impaired left ventricular arone therapy in September 2014, the patients
(LV) function with a low LV ejection fraction heart failure symptoms worsened and blood sam-
(LVEF) of 28 % assessed according to the mod- ples showed an elevated plasma brain natriuretic
ified Simpson method. The patient also had a peptide (BNP). His ECG revealed atrial fibrillation
severely dilated left atrium with an anteroposte- with spontaneous QRS (Fig. 10.2).
rior diameter of 55 mm and left atrial volume of Since his heart failure symptoms were
45 mL/m2 . refractory to medical management, repeated

Fig. 10.1 12-lead ECG in atrial fibrillation with biventricular pacing

ERRNVPHGLFRVRUJ
10 Cardiac Resynchronization Therapy and Possible Dysfunctions 111

Fig. 10.2 12-lead ECG in atrial fibrillation with spontaneous QRS

echocardiography was performed that revealed 10.2 Cardiac Resynchronization


an increased left atrial diameter to 58 mm and Therapy (CRT)
a volume of 46 mL/m2 despite mild improve-
ment of the LVEF. The hypothesis of the worsen- Denition
ing clinical condition was that the exacerbation
of HF was due to high-rate AF or to displace- Cardiac resynchronization therapy (CRT) is a
ment of left ventricular (LV) lead; therefore, a specific type of pacemaker therapy that aims to
chest X-ray was performed and the imaging con- restore or improve ventricular contraction by
firmed the LV lead displacement (Fig. 10.3a, b). pacing both the right ventricle (RV) and the left
Figure 10.4a, b represents the coronary sinus ventricle (LV). Indeed, in addition to the right
angiography during the reimplantation of new lead, an extra catheter is usually implanted via
LV pacing lead. Two months after the reposition- the coronary sinus to pace the LV, most commonly
ing of biventricular pacing, echocardiography at the latest activated LV segment. This may be
demonstrated an increase in the LVEF from 28 carried out with or without the use of an implant-
to 38 % and a reduction in the left atrial diameter able cardioverter defibrillator (ICD), a device
from 58 to 56 mm and the left atrial volume from required in patients at risk for ventricular arrhyth-
46 to 45 mL/m2 compared with the values exam- mias [1].
ined in December 2014. Left bundle branch block (LBBB) is an elec-
The patients clinical course was satisfactory, trocardiographic sign of electrical dyssyn-
and his plasma BNP level decreased to 504 pg/mL chrony and, in association with poor LV
2 months after the repositioning of the LV lead. function, is an independent risk factor for

ERRNVPHGLFRVRUJ
112 L. Cipolletta and M. Brambatti

a b

Fig. 10.3 (a) Posteroanterior chest X-ray showing the left ventricular lead displacement. (b) Latero-lateral chest X-ray
confirming the left ventricular lead displacement

a b

Fig. 10.4 (a) Coronary sinus angiography on right anterior oblique projection. (b) Coronary sinus angiography on left
anterior oblique projection

mortality [2]. LBBB often correlates with chrony can further worsen the pump function of
mechanical dyssynchrony also called interven- a failing LV. CRT, through the reestablishment
tricular delay, a nonphysiological timing of LV of a more physiological ventricular activation,
and RV contraction. There may also be abnor- improves cardiac performance and reduces HF
mal contraction of individual segments of the symptoms and also determines a reverse remod-
LV, causing intraventricular delay. In an HF eling of the LV along with mortality and hospi-
patient, the inter- and intraventricular dyssyn- talization reduction [37].

ERRNVPHGLFRVRUJ
10 Cardiac Resynchronization Therapy and Possible Dysfunctions 113

CRT is usually set in DDD or DDDR mode if Indications


the patient is in sinus rhythm. Atrial events trig-
ger an atrioventricular interval that should be The 2013 European Society of Cardiology (ESC)
long enough in order to allow the optimal atrial guidelines [2] on cardiac pacing and CRT
contribution to ventricular filling but short included the following indications for CRT in:
enough to allow ventricular pacing for most of
the time. As specified in the ESC guidelines [1], 1. Chronic HF patients in sinus rhythm with left
the goal of CRT should be to achieve BiV pacing ventricular ejection fraction (LVEF 35 %):
as close to 100 % as possible since the reduction NYHA functional class II, III, or ambula-
in mortality and hospitalization is strongly asso- tory IV despite adequate medical treatment
ciated with an increasing percentage of BiV pac- and LBBB with QRS duration >150 ms
ing. In addition, CRT pacing often results in a (Class I with level of evidence A).
QRS complex that is narrower than the native NYHA functional class II, III, or ambula-
QRS complex because of fusion between the two tory IV despite adequate medical treatment
paced signals. and LBBB with QRS duration 120150 ms
(Class I with level of evidence B).
NYHA functional class II, III, or ambula-
Clinical Trials tory IV despite adequate medical treatment
and non-LBBB with QRS duration >150 ms
CRT has been clinically evaluated in more than (Class IIa with level of evidence B).
4000 patients in randomized controlled trials NYHA functional class II, III, or ambulatory
(Table 10.1). Early studies were positive in terms IV despite adequate medical treatment and
of morbidity, including improvements in quality non-LBBB with QRS duration 120150 ms
of life, functional status, and exercise capacity. (Class IIb with level of evidence B).
Finally, the large trial COMPANION [3], along CRT is not recommended in patients with
with the CARE-HF trial [4], provided the demon- chronic HF and QRS duration <120 ms.
stration of mortality benefit in HF patients, 2. Chronic HF patients with permanent atrial
defined as LVEF <35 %, QRS duration 120 ms, fibrillation:
and NYHA functional class III or ambulatory NYHA functional class II, III, or ambula-
class IV HF. Recently, three major randomized tory IV despite adequate medical treat-
trials [MADIT-CRT [5], RAFT [6], and ment, LVEF 35 %, and QRS duration
REVERSE [7]] have demonstrated that CRT can 120 ms, provided that a biventricular pac-
provide functional improvement and decrease the ing as close to 100 % as possible can be
heart failure events in patients with reduced LVEF achieved (Class IIa with level of
and NYHA class I or II HF and QRS 120 or evidence B). Otherwise, atrioventricular
130 ms. CRT-D has also been shown to decrease junction ablation should be added in case
the mortality rate for patients with class II but not of incomplete biventricular pacing (Class
class I HF. Further subgroup analyses of data IIa with level of evidence B).
from MADIT-CRT, REVERSE, and RAFT trials In patients with uncontrolled heart rate who
demonstrated that a CRT implant in patients with are candidates for atrioventricular junction
a QRS duration 150 ms yielded a greater sur- ablation, CRT should be considered in
vival benefit compared to a QRS duration of patients with reduced LVEF who are candi-
120 ms. Also meta-analyses using aggregate data dates for AV junction ablation for rate con-
from randomized trials confirmed that CRT was trol (Class IIa with level of evidence B).
effective in reducing adverse clinical events in 3. Chronic HF patients with conventional pace-
patients with baseline QRS duration 150 ms and maker indication:
proposed that CRT might not reduce events in Upgrade from conventional pacemaker or
patients with a QRS <150 ms [8]. ICD is indicated in patients with LVEF

ERRNVPHGLFRVRUJ
114 L. Cipolletta and M. Brambatti

Table 10.1 Summary of major studies of CRT


Patients, NYHA LVEF, QRS, Primary end Secondary end
Trials no. class % ms points points Main findings
COMPANION [3] 1520 III or 21 159 (1) All-cause (2) All-cause CRT-P/CRT-D
IV mortality or mortality and reduced (1) and (2)
hospitalization cardiac mortality
CARE-HF [4] 813 III or 25 160 (1) All-cause (2) All-cause CRT-P reduced (1)
IV mortality or mortality and (2)
hospitalization
REVERSE [7] 610 I or II 27 7 153 (1) HF clinical (2) LVESVi, (3) Primary end point
composite HF hospitalization, NS CRT-P/CRT-D
score (4) all-cause reduced (2) and (3)
mortality but not (4)
MADIT-CRT [5] 1820 I or II 24 5 162 (1) HF or death (2) All-cause CRT-D reduced (1)
mortality (3) and (3) but not (2)
LVESVi
RAFT [6] 1798 II or 23 5 158 (1) All-cause (2) All-cause CRT reduced (4)
III mortality or HF mortality, (3)
hospitalization cardiac mortality,
(4) HF
hospitalization

35 % and high percentage of ventricular However, in order to avoid implantation-


pacing who remain in NYHA class III and related complications and additional costs, CRT
ambulatory IV despite adequate medical nonresponders should be accurately identified.
treatment (Class I with level of evidence B). Several factors have been shown to influence the
De novo CRT should be considered in efficacy of CRT, including chronic renal insuffi-
patients with reduced EF and expected ciency, hemodynamic abnormalities such as pre-
high percentage of ventricular pacing in capillary pulmonary hypertension, and ischemic
order to decrease the risk of worsening HF. HF [9, 10]. Patients with ischemic HF represent a
challenge because coronary disease is progres-
sive, and upcoming ischemic events may be non-
Denition of Nonresponders associated to CRT response. In particular ischemic
patients with extensive scar, usually measured by
Despite advances in procedural expertise and MRI, have been demonstrated to a lower response
experience, about one-third of patients cur- rate to resynchronization therapy. End-stage HF
rently do not respond to CRT [2]. There is a patients, including those who are inotropic depen-
lack of a universally accepted definition to dent, appear to have a poor response rate as well.
classify a patient as responder or nonresponder With regard to sex differences, women appear to
to CRT. Typically, clinical trials evaluate have a more favorable response from CRT than
event-driven outcome such as HF hospitaliza- men, particularly in patients with LBBB and even
tions and mortality as primary clinical deter- at a QRS duration less than 150 ms. Contrarily,
minant of response to CRT and measures of no significant interaction between age and CRT
cardiac function and functional status as sec- effect was found [11].
ondary end points. In real life, patients over-
all well-being is a more significant outcome.
Also, taking into account that HF is a progres- 10.3 CRT Malfunctions
sive disease, the prevention of the progress of
the illness should be considered as a benefit Biventricular pacing systems are susceptible
deriving from CRT and thus a positive response to similar system malfunctions of other types
to CRT therapy. of pacemakers; however, in patients with CRT,

ERRNVPHGLFRVRUJ
10 Cardiac Resynchronization Therapy and Possible Dysfunctions 115

long-term complications are more frequent Phrenic Stimulation


than in single- or dual-chamber pacemaker/
ICD systems since these patients have Phrenic nerve (PN) stimulation occurs in approx-
advanced heart failure and implantation is imately 15 % of CRT patients and can limit
more difficult [12]. biventricular pacing. The left phrenic runs on the
anterior wall and can be irritated by anterolateral,
lateral, or posterolateral epicardial stimulation of
Infections the LV; the diaphragm can be directly stimulated
by electrocatheters placed in posteroinferior
Infection risk of CRT averages between 1 and areas of the same LV. The occurrence of phrenic
3 % yearly and is associated with prolonged nerve stimulation after LV lead implant may be a
hospitalization stay and increased cost. This risk sign of LV lead migration, in some cases without
is increased by some factors such as renal fail- significant changes in the chest X-ray [12]. The
ure, device failure, device replacements, malnu- growing use of quadripolar left ventricular leads
trition, prolonged therapy with steroids, diabetes is overcoming the problem.
mellitus, device size, and re-interventions. The
most common germ in infections related to
devices is the Staphylococcus (7295 %). In Oversensing
cases of latent infection, the most responsible
microorganism results to be the coagulase-nega- Oversensing is defined as the detection of
tive Staphylococcus epidermidis. The infection an inappropriate electrical signal. Ventricular
can be evident several years after the implant. In oversensing may result in inhibition of ventricu-
most patients with CRT infection, the complete lar pacing and loss of CRT. In a BiV pacing sys-
system including all leads should be explanted, tem, oversensing of atrial activity on the
although there is a major risk related to the lead ventricular channel results in ventricular inhibi-
burden and the underlying condition of the tion and loss of CRT, as well as in misdiagnosis
patient. of ventricular tachyarrhythmias.

Loss of Capture Mechanical Lead Failure

Non-capture is defined as the release of an All major malfunctions, including loss of cap-
atrial or ventricular pacing stimulus without ture, undersensing, and oversensing, can be asso-
capture. The left ventricular lead of CRT sys- ciated with a rupture in lead insulation or
tem is separated from the myocardium by the conductor failure. A marked drop in the mea-
vein wall and by any epicardial fat that may sured stimulation impedance is a sign of lead
coat the vein. When loss of capture occurs, the insulation break, while a significant increase in
capture threshold of the left ventricular lead impedance conductor is a sign of lead fracture.
may be higher than values measured at the
implantation procedure. In modern CRT
devices, the right and left ventricular leads can
References
be assessed independently if loss of capture is
suspected. Widening of the QRS complex or a 1. European Society of Cardiology (ESC), European
more elusive change in QRS morphology may Heart Rhythm Association (EHRA), Brignole M,
be a sign of loss of capture of LV lead. However, Auricchio A, Baron-Esquivias G, Bordachar P,
Boriani G, Breithardt OA, Cleland J, Deharo JC,
only pacemaker interrogation will confirm the
Delgado V, Elliott PM, Gorenek B, Israel CW,
issue or a chest X-ray in presence of a signifi- Leclercq C, Linde C, Mont L, Padeletti L, Sutton R,
cant displacement. Vardas PE (2013) 2013 ESC guidelines on cardiac

ERRNVPHGLFRVRUJ
116 L. Cipolletta and M. Brambatti

pacing and cardiac resynchronization therapy: the cardiac resynchronization in mildly symptomatic
task force on cardiac pacing and resynchronization heart failure patients and in asymptomatic patients
therapy of the European Society of Cardiology (ESC). with left ventricular dysfunction and previous
Developed in collaboration with the European Heart heart failure symptoms. J Am Coll Cardiol 52(23):
Rhythm Association (EHRA). Europace 15(8):1070 18341843
1118. doi:10.1093/europace/eut206, Epub 2013 Jun 8. Sipahi I, Carrigan TP, Rowland DY, Stambler BS,
24 Fang JC (2011) Impact of QRS duration on clinical
2. Shamim W, Francis DP, Yousufuddin M, Varney S, event reduction with cardiac resynchronization ther-
Pieopli MF, Anker SD, Coats AJ (1999) apy: meta-analysis of randomized controlled trials.
Intraventricular conduction delay: a prognostic Arch Intern Med 171(16):1454
marker in chronic heart failure. Int J Cardiol 70(2):171 9. Delgado V, van Bommel RJ, Bertini M et al (2011)
3. Bristow MR, Saxon LA, Boehmer J, Krueger S, Kass Relative merits of left ventricular dyssynchrony, left
DA, De Marco T, Carson P, DiCarlo L, DeMets D, ventricular lead position, and myocardial scar to pre-
White BG, DeVries DW, Feldman AM, Comparison dict long-term survival of ischemic heart failure
of Medical Therapy, Pacing, and Defibrillation in patients undergoing cardiac resynchronization ther-
Heart Failure (COMPANION) Investigators (2004) apy. Circulation 123:7078
Cardiac-resynchronization therapy with or without an 10. Kass DA (2003) Predicting cardiac resynchronization
implantable defibrillator in advanced chronic heart response by QRS duration: the long and short of it. J
failure. N Engl J Med 350(21):2140 Am Coll Cardiol 42:21252127
4. Cleland JG, Daubert JC, Erdmann E, Freemantle N, 11. European Heart Rhythm Association, European
Gras D, Kappenberger L, Tavazzi L, Cardiac Society of Cardiology, Heart Rhythm Society, Heart
Resynchronization-Heart Failure (CARE-HF) Study Failure Society of America, American Society of
Investigators (2005) The effect of cardiac resynchro- Echocardiography, American Heart Association,
nization on morbidity and mortality in heart failure. N European Association of Echocardiography, Heart
Engl J Med 352(15):1539 Failure Association, Daubert JC, Saxon L, Adamson
5. Moss AJ, Hall WJ, Cannom DS, Klein H, Brown PB, Auricchio A, Berger RD, Beshai JF, Breithard O,
MW, Daubert JP, Estes NA, Foster E, Greenberg H, Brignole M, Cleland J, Delurgio DB, Dickstein K,
Higgins SL, Pfeffer MA, Solomon SD, Wilber D, Exner DV, Gold M, Grimm RA, Hayes DL, Israel C,
Zareba W, MADIT-CRT Trial Investigators (2009) Leclercq C, Linde C, Lindenfeld J, Merkely B, Mont
Cardiac-resynchronization therapy for the prevention L, Murgatroyd F, Prinzen F, Saba SF, Shinbane JS,
of heart-failure events. N Engl J Med 361(14):1329 Singh J, Tang AS, Vardas PE, Wilkoff BL, Zamorano
6. Tang AS, Wells GA, Talajic M, Arnold MO, Sheldon JL (2012) 2012 EHRA/HRS expert consensus state-
R, Connolly S, Hohnloser SH, Nichol G, Birnie DH, ment on cardiac resynchronization therapy in heart
Sapp JL, Yee R, Healey JS, Rouleau JL, failure: implant and follow-up recommendations
Resynchronization-Defibrillation for Ambulatory and management. Heart Rhythm 9(9):15241576.
Heart Failure Trial Investigators (2010) Cardiac- doi:10.1016/j.hrthm.2012.07.025
resynchronization therapy for mild-to-moderate heart 12. Biffi M, Moschini C, Bertini M, Saporito D, Ziacchi
failure. N Engl J Med 363(25):2385 M, Diemberger I, Valzania C, Domenichini G, Cervi
7. Linde C, Abraham WT, Gold MR, St. John Sutton M, E, Martignani C, Sangiorgi D, Branzi A, Boriani G
Ghio S, Daubert C, REVERSE (REsynchronization (2009) Phrenic stimulation: a challenge for cardiac
reVErses Remodeling in Systolic left vEntricular dys- resynchronization therapy. Circ Arrhythm
function) Study Group (2008) Randomized trial of Electrophysiol 2:402410

ERRNVPHGLFRVRUJ
Part IV
Cardiomyopathy

ERRNVPHGLFRVRUJ
Hypertrophic Cardiomyopathy
11
Marco Marchesini and Erika Baiocco

11.1 Case Report Medical History and Cardiovascular


Risk Factors

A 57-year-old man presented to the emer- The family history revealed that his mother
gency room complaining palpitations that was affected by hypertrophic cardiomyopathy
started suddenly few hours before, during a (HCM). There was not any case of sudden car-
moderate walking. Palpitations were diac death in the family.
accompanied by dyspnea and mild dizzi- At 20 years old, HCM was incidentally diag-
ness. Symptoms lasted for about 10 min, nosed during a sport screening and since pre-
forcing the patient to rest. The episode participation screening and since then he was
interrupted abruptly and spontaneously, and regularly followed up.
the patient did not remember if heartbeat He was a smoker.
was regular during the episode. He never
had arrhythmias before and he denied syn-
cope or angina. Allergies
The patient presented completely
asymptomatic to the emergency room. None

Medications

Verapamil 80 mg BID

Vital Signs

Temperature: 36.3 C
Resting heart rate: 58 bpm
M. Marchesini (*) E. Baiocco
Clinica di Cardiologia e Aritmologia, Universit Blood pressure: 125/90 mmHg
Politecnica delle Marche, Respiratory rate: 18 breaths per minute
Via Conca, 71, Ancona 60126, Italy Oxygen saturation while breathing in room
e-mail: marche9584@gmail.com; erikabaiocco@ air: 99 %
libero.it

Springer International Publishing Switzerland 2015 119


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_11

ERRNVPHGLFRVRUJ
120 M. Marchesini and E. Baiocco

Physical Examination Routine Laboratory Tests

The patient appeared in good clinical condition. Complete blood count: normal
At physical examination, the relevant findings Cholesterol (total, HDL, LDL) and TG: normal
were the following: Fasting blood glucose: 78 m/dl (4.33 mmol/L)
Cardiovascular: Apical precordial impulse Hepatic function (GOT, GPT, -GGT, ALP, total
was forceful but not displaced laterally. Regular bilirubin, direct and indirect): normal
rate and rhythm; S1 and S2 were normal with an Thyroid function (TSH, FT3, FT4): normal
adjunctive S4. Systolic ejection late-peaking Renal function (creatinine, BUN): normal
murmur was best heard between the apex and left Electrolytes (Na+, K+, Ca++, Mg++, Cl):
sternal border without radiation to the neck, and normal
it was increased by Valsalva maneuver. Mild dia-
stolic decrescendo murmur was detected in Erb
auscultator focus. EKG
Lungs: No rales at auscultation neither rhon-
chi nor wheezes bilaterally. A routine EKG at rest was performed (Fig. 11.1).
Abdomen: Plain and tractable; no The initial EKG showed sinus rhythm at 58 bpm,
hepatosplenomegaly. normal atrioventricular conduction (PR interval 160
Extremities: No lower limb edema. s), normal QRS duration (0.08 s), absent Q wave,

Fig. 11.1 12-lead ECG

ERRNVPHGLFRVRUJ
11 Hypertrophic Cardiomyopathy 121

and horizontal QRS axis (+15). Increased ampli-


tude and duration (>0.04 s) of the terminal negative
portion of P wave in V1. High-voltage QRS com-
plexes. T-wave inversion in the left precordial leads,
reciprocal ST-segment elevation, and tall T wave in
the right precordial leads. ST-segment depression
and T-wave inversion in lead I and aVL.
The Sokolow diagnostic criteria for left ven-
tricular hypertrophy (LVH) were satisfied: R
wave in V6 + S wave in V1 >3.5 mV.
Asymmetrical configuration of inverted T
waves suggested a nonischemic origin.
Conclusion: sinus rhythm, normal conduction,
possible left atrial enlargement, and left ventricu-
Fig. 11.2 Parasternal long-axis view shows septal
lar hypertrophy with secondary anomalies of hypertrophy
repolarization
We hypothesized that the described palpitations
were provoked by a hyperkinetic arrhythmia (supra-
ventricular or ventricular), and we explored the dif-
ferent secondary causes of arrhythmic events:

Hyperthyroidism
Fever
Anxiety
Anemia
Use of medications containing stimu-
lant, caffeine, or nicotine
Strenuous exercise, poor training

He did not report any specific stress condition or Fig. 11.3 Parasternal short-axis ventricle view
recent changes in his lifestyle. He was walking
slowly, when the arrhythmia occurred, which there-
fore can be excluded a physiologic activity response
and poor training. According to physical examina-
tion, fever was excluded, and the laboratory tests
did not show any anemia and thyroid dysfunction.
The most probable cause of symptoms was
spontaneous arrhythmia.

Transthoracic
Echocardiography (TTE)

A TTE showed (Figs. 11.2, 11.3, and 11.4):

Left ventricular volumes at lower normal lim-


its (iLVEDV 35 mL/m2) with massive Fig. 11.4 Apical four-chamber view

ERRNVPHGLFRVRUJ
122 M. Marchesini and E. Baiocco

asymmetrical hypertrophy with septal wall point to a hemodynamic distress that is often
thickness that reached 35 mm; the posterior related to sustained ventricular tachycardias.
wall was 15 mm with a septum to posterior In adult patients with HCM, most recent data
wall ratio of 2.4. report on an annual incidence of cardiovascular
Ejection fraction with the biplane Simpson death near 12 % with sudden cardiac death
method was 65 % without regional wall being (SCD) the most common.
motion abnormalities.
Diastolic dysfunction grade II with normal
Major clinical features associated with an
estimated filling pressure (E/E 6).
increased risk of SCD are:
Severe left atrial enlargement (LA diameter
55 mm, iLAV >40 mL/m2). Young age
Systolic anterior motion (SAM) of the ante- Non-sustained ventricular tachycardia
rior mitral valve leaflet, without LVOT (NSVT)
obstruction at rest, but with mild flow Maximum left ventricular wall thickness
acceleration during Valsalva (peak gradient Family history of sudden cardiac death
15 mmHg). Syncope
Normal size and function of right atrium and Left atrial diameter
ventricle. Left ventricular outflow tract obstruction
Normal aortic tricuspid valve with mild cen- Exercise blood pressure drop
tral regurgitation
Normal tricuspid and pulmonic valves.
Normal dimension of inferior vena cava (IVC) The patient had no family history of SCD and
with >50 % inspiratory collapse. denied syncope, but the risk assessment com-
No pericardial effusion. prised also of a 24-h ambulatory ECG and an
exercise test.
According to ESC guidelines, HCM is defined
as a wall thickness >15 mm in one or more LV
myocardial segments, as measured by any imag- Exercise Testing with Treadmill
ing technique (echocardiography, cardiac mag-
netic resonance imaging (CMR), or computed Exercise testing was terminated for asthenia and
tomography (TC)), that is not explained solely by muscular weakness at a heart rate corresponding
chronic loading conditions. to 93 % of maximal heart rate predicted for age.
Echocardiographic findings were suggestive Neither symptoms nor ST-segment depression or
of hypertrophic cardiomyopathy: the patient had elevation occurred. During exercise, arrhythmias
no history of hypertension or valve disease with were not detected except for two isolated poly-
elevated afterload. Infiltrative cardiomyopathy morphic ventricular ectopic beats (VE) and one
was excluded, because the ventricular thickening VE couple. Systolic blood pressure and heart rate
is usually concentric with characteristic pattern response were normal.
of granular sparkling and different degrees of
pericardial effusion.
In a patient with HCM, a sustained episode of 24-h Ambulatory ECG
palpitation lasting more than few minutes is often
caused by supraventricular arrhythmias, espe- 24-h ambulatory ECG was performed to detect
cially in the presence of left atrium enlargement; atrial or ventricular arrhythmias. The total num-
atrial fibrillation is the most common arrhythmia ber of beats analyzed was 82,105. Sinus rhythm
in this population. In our patient, we could not at average heart rate of 64 bpm, with minimum of
exclude the hypothesis of a ventricular origin, 50 bpm and a maximum of 95 bpm. Two hundred
particularly because the symptoms associated fifty total ventricular ectopic (VE) beats of differ-
with palpitation (dyspnea and dizziness) could ent morphologies with six VE couples and one

ERRNVPHGLFRVRUJ
11 Hypertrophic Cardiomyopathy 123

NSVT (four beats at 150 bpm). Forty-three total


supraventricular ectopic beats with one short run
of 12 beats. No significant pauses.
NSVT is defined as 3 consecutive ventricu-
lar beats at 120 bpm lasting <30 s, so only one
episode was detected.
In the absence of sustained arrhythmia, the
electrophysiological study (EPS) is not specifi-
cally recommended.

Clinical Course

According to HCM guidelines, ICD implantation


should be considered in patients with an esti-
mated risk of SCD 6 % and a life expectancy of
>1 year, and it may be considered in patients with Fig. 11.5 Chest X-ray anterior-posterior (AP)
an estimated risk between 4 and <6 %, while it projection
is not recommended in patients with an estimated
risk <4 % unless they have clinical features that
are of proven prognostic importance.
Our patients estimated risk of sudden cardiac
death at 5 years was 5 % (intermediate), based on
severe cardiac hypertrophy and NSVT; that sup-
ported the indication for ICD implant in primary
prevention.
The patient was informed about his SCD risk
and the necessity of an ICD implant. He was
made also aware on the risk of inappropriate
shocks, implant complications, and the social and
occupational implications of an ICD implant.
According to young age, primary prevention
indication, good AV conduction, and patients
preference, we scheduled a subcutaneous ICD
(S-ICD) implantation.
At the preimplantation screening test, the
patient presented the anatomical and electrocar-
diographic features ideal for a suitable subcuta-
neous sensing. An S-ICD was then implanted
without complications. Fig. 11.6 Chest X-ray lateral view

Therapy and Discharge


Chest X-Ray
Because the episode of prolonged palpitation was
Chest X-ray was performed the day after implan- his first and an S-ICD was implanted, we decided
tation (Figs. 11.5 and 11.6). not to give antiarrhythmic therapy and main-
Conclusion: Correct position of the implanted tained the calcium channel blockers (verapamil)
S-ICD system in order to control LVOT gradient.

ERRNVPHGLFRVRUJ
124 M. Marchesini and E. Baiocco

Moreover, at discharge, the patient was disorders due to inborn errors of metabolism
advised to abstain from competitive athletic (e.g., Pompe disease, Fabry disease) or deposi-
activity and strenuous physical exertion and was tion of anomalous misfolded proteins (different
given clinical and echocardiographic follow-up types of amyloidosis). Other genetic causes could
appointments. be mithocondrial diseases, neuromuscular disor-
ders (Friedreichs ataxia), or malformative syn-
dromes like Noonan or LEOPARD [4].
11.2 Hypertrophic The true hypertrophic cardiomyopathy is a
Cardiomyopathy genetic disease with an autosomal dominant trait
caused by mutations in cardiac sarcomere protein
Introduction and Epidemiology genes. In general, patients with a sarcomere pro-
tein mutation present earlier and report a higher
The most recent expert consensus on cardiomy- prevalence of family history of HCM and sudden
opathies has adopted a new classification system cardiac death (SCD) than those without a muta-
no more based on primary or secondary involve- tion. They also tend to have more severe hypertro-
ment of the heart but in which cardiomyopa- phy, microvascular dysfunction, and myocardial
thies are defined by specific morphological and fibrosis.
functional phenotypes as they present for the
first time to the observer: mainly hypertrophic,
dilated, arrhythmogenic cardiomyopathy and Incidence
restrictive phenotype. Only in the second time,
cardiomyopathies are grouped into familial/ A number of studies worldwide report a preva-
genetic and nonfamilial/nongenetic subtypes, lence of HCM in the range of 0.020.23 % in
irrespective of the presence of extra-cardiac dis- adults. In pediatric registries, the prevalence of
ease [1, 2, 3]. HCM in children is unknown, but population-
based studies report an annual incidence of 0.3
0.5 per 100,000. Most studies report a small male
Hypertrophic cardiomyopathy (HCM) is preponderance, while the prevalence in different
defined by the presence of increased left racial groups is similar.
ventricular (LV) wall thickness that is not
solely explained by abnormal loading con-
ditions. Many are the secondary causes of Diagnosis and Denition
hypertrophy (in particular left ventricular
hypertrophy) that should be considered in In an adult, HCM is defined by a wall thick-
differential diagnosis: ness 15 mm in one or more LV myocardial
segmentsas measured by any imaging tech-
1. Athletes heart nique such as echocardiography, cardiac mag-
2. Hypertensive cardiomyopathy netic resonance imaging (CMR), or computed
3. Valve diseases imposing increased tomography (CT)that is not explained
afterload (mainly aortic stenosis) solely by loading conditions.
4. Isolated basal septal hypertrophy in In children as in adults, the diagnosis of HCM
elderly people requires a LV wall thickness more than two
standard deviations greater than the predicted
mean z-score.
Moreover hypertrophic phenotype (variable The clinical diagnosis of HCM in first-degree
grade and distribution of ventricular wall thick- relatives of patients with unequivocal disease
ening) could represent a common picture of dif- (LVH 15 mm) is based on the presence of
ferent pathologic conditions such as infiltrative otherwise unexplained increased LV wall

ERRNVPHGLFRVRUJ
11 Hypertrophic Cardiomyopathy 125

thickness 13 mm in one or more LV myocar- and Danon disease in adolescent males. Different
dial segments. degrees of concentric hypertrophy with left ven-
tricular systolic impairment is a clue to infiltra-
tive diseases, because the hypokinetic end-stage
Variants phases of a true HCM more often pass through a
dilation of left ventricle.
A particular variant is apical hypertrophic cardio-
myopathy (AHCM) that is a rare form of HCM,
which usually involves the apex of the left ventricle A New Diagnostic Tool: Cardiac
and rarely involves the right ventricular apex or Magnetic Resonance
both. Historically, this condition was thought to be
confined to the Japanese population, but it is also Cardiac magnetic resonance (CMR) is particu-
found in other populations. Of all the HCM patients larly useful for characterizing the presence, loca-
in Japan, the prevalence of AHCM was 15 %, tion, and extent of LV hypertrophy, which can be
whereas in the USA and Europe, the prevalence limited to one or two left ventricle (LV) seg-
was only 3 %. The diagnostic criteria for AHCM ments. In those cases, CMR offers a superior
included demonstration of asymmetrical LV hyper- visualization and a higher diagnostic accuracy
trophy, confined predominantly to the LV apex, respect to 2D echocardiography, particularly
with an apical wall thickness 15 mm and a ratio when the only segments involved are the basal
of maximal apical to posterior wall thickness 1.5. anterolateral free wall or the apex.
In contrast with the common variant of HCM, up Recent study showed that diffuse hypertrophy,
to 54 % of patients with AHCM are symptomatic. involving >50 % of the left ventricle and 8 or
This entity should be well known because of its more segments, is present in 54 % of patients
difficult recognition (the apical position of hyper- with HCM, whereas only 10 % of patients pres-
trophic segments represents a limitation for a rou- ent with single segment involvement [6, 7].
tine 2D echocardiography) and its high prevalence During an exam directed to distinguishing the
of complications such as atrial fibrillation, myocar- possible origin of a hypertrophic phenotype found
dial infarction, apical aneurysm, embolic events, with echocardiography, CMR could add diagnos-
and congestive heart failure [5]. tic clues, demonstrating a constellation of sugges-
tive features of genetic HCM: such as anomalies
in papillary muscles, right ventricle, subclinical
Differential Diagnosis features, and particularly tissue characterization.
Papillary muscle involvement in HCM con-
Genetic and nongenetic disorders causing hyper- sists in apical displacement of its insertion and
trophic phenotype can present with lesser the presence of multiple or bifid papillary mus-
degrees of wall thickening (1314 mm); in these cles with increased mass. All these variants could
cases, the diagnosis of true HCM requires evalu- favor SAM and outflow tract obstruction perturb-
ation of other features including family history, ing the normal activity of mitral valve apparatus.
noncardiac symptoms and signs, electrocardio- In 1/3 of patients with HCM, right ventricular
gram (ECG) abnormalities, laboratory tests, and wall thickness and/or mass is increased, includ-
multimodality cardiac imaging. In any situation, ing about 10 % of patient with extreme right ven-
the age of presentation is a fundamental clue to tricle (RV) wall hypertrophy (>10 mm). Finally
the differential diagnosis. Severe (maximal in preclinical (genotype [+]/phenotype [])
thickness more than 30 mm or equivalent in chil- patients with HCM, CMR may show the pres-
dren) and concentric ventricular hypertrophy in ence of crypts. Myocardial crypt is a deep fissur-
a child, adolescent, or young adult should rise ing of the muscle orthogonal to the endocardial
suspicion of metabolic or storage disorders, in border (often visualized also in angiography),
particular Pompe disease in the infantile period localized predominately in the inferior septum,

ERRNVPHGLFRVRUJ
126 M. Marchesini and E. Baiocco

although the etiology of these structural abnor- The latter group can be further divided into those
malities remains uncertain. who develop outflow tract obstruction and those
Moreover, CMR is able to accurately define who do not.
ventricular volume and function, being the gold Outflow tract obstruction at rest with exer-
standard for ejection fraction measurement. tional limitations is present in 25 % of all affected
Frequently in patients with HCM, the ventricular patients; an additional 25 % present inducible
volumes are reduced, and the hyperkinetic outflow tract obstruction; other groups of symp-
appearance of systolic contraction translates into tomatic patients are those with restrictive physi-
a supernormal ejection fraction. This is true until ology and frequent tachyarrhythmias and who
the end stage of cardiomyopathy is reached, may experience exertional limitation because of
when the diastolic dysfunction, present from the diastolic dysfunction and those who are at risk or
beginning and related to myocardial thickness have already experienced ventricular arrhythmias
and rigidity, is accompanied by a reduced sys- and sudden cardiac death. At last, there is a small
tolic thickening [8, 9, 10]. proportion of patients (up to 5 %) who may
In 3-chamber view with cine imaging, CMR is develop the end-stage phase of hypertrophic car-
able to elucidate the precise mechanism of outflow diomyopathy with left ventricular dilation and
tract obstruction demonstrating turbulent flow systolic impairment.
generated by systolic movement of anterior mitral
leaflet, chordae, and papillary muscle toward the Left Ventricular Outow Tract
interventricular septum [11]. Obstruction
Contrast-enhanced CMR with LGE Treatment of outflow obstruction should be
sequences can detect areas of focal abnormality restricted to patients who exhibit the associated
in approximately 5080 % of patients. There is symptoms. Recognition of obstruction-related
no specific pattern of LGE characteristic for symptoms may be made challenging by both a
HCM, although the distribution of LGE in HCM patients restriction in physical activity and by
does not correspond to a coronary vascular terri- the presence of latent obstruction (obstruction
tory. LGE is most often located in the most not present at rest but only under provocative
hypertrophied segment with an intramyocardial conditions such as exercise, Valsalva maneuver).
distribution (focal spot or linear deposit). First-line therapy consists in pharmacologic
Moreover, recent studies have demonstrated a approach with -blockers or disopyramide to
significant association between the presence of reduce left ventricular inotropism and to prolong
LGE and ventricular tachyarrhythmias on ambu- diastolic filling time.
latory 24-h Holter electrocardiography. Patients who cannot tolerate or who are refrac-
However, it is not clear whether the presence of tory to medical therapy are candidates for surgi-
LGE provides a strong predictive value in iden- cal or catheter-based treatment of outflow
tifying patients with HCM at risk for sudden obstruction. In experienced centers, both proce-
death, so much so the last ESC guidelines, pub- dures are associated with low rates of complica-
lished in 2014, do not include LGE in risk strati- tions and high successful rate. There is debate
fication algorithm [1217]. over which procedure is best, but concerns are
emerging about the potential for creation of an
arrhythmogenic focus with percutaneous septal
Treatment ablation, as well as the increased risk of complete
heart block with that procedure. The routine per-
Treatment depends on disease expression, which formance of CMR after septal reduction therapy
can differ greatly among individuals, even within is not recommended, but it can be of value when
a single family. The natural history of hypertro- questions arise about LV residual function or
phic cardiomyopathy includes those who remain when gradients do not resolve or recur late after
asymptomatic and those who develop symptoms. the procedure.

ERRNVPHGLFRVRUJ
11 Hypertrophic Cardiomyopathy 127

Table 11.1 Major clinical features associated with an increased risk of sudden cardiac death in adults according to the
2014 ESC guidelines on diagnosis and management of hypertrophic cardiomyopathy
Risk factor Comment
Age The effect of age on SCD has been examined in a number of studies, and two have shown a
significant association, with an increased risk of SCD in younger patients
Some risk factors appear to be more important in younger patients, most notably, NSVT severe
LVH and unexplained syncope
Non-sustained NSVT occurs in 2030 % of patients during ambulatory ECG monitoring and is an independent
ventricular predictor of SCD
tachycardia There is no evidence that the frequency, duration, or rate of NSVT influences the risk of SCD
Maximum left The severity and extent of left ventricular hypertrophy measured by TTE are associated with the
ventricular wall risk of SCD
thickness Several studies have shown the greatest risk of SCD in patient with a maximum wall thickness
of 30 min, but there are few data in patients with extreme hypertrophy (35 mm)
Family history While definitions vary, a family history of SCD is usually considered clinically significant when
of sudden one or more first-degree relatives have died suddenly aged <40 years with or without a
cardiac death at diagnosis of HCM or when SCD has occurred in a first-degree relative at any age with an
a young age established diagnosis of HCM
Left atrial Two studies have reported a positive association between LA size and SCD. There are no data
diameter on the association between SCD and LA area and volume. Measurement of LA size is also
important in assessing the risk of AF
Left ventricular A number of studies have reported a significant association with LVOTO and SCD. Several
outflow tract unanswered questions remain, including the prognostic importance of provable LVOTO and the
obstruction impact of treatment (medical or invasive) on SCD
Exercise blood Approximately one third of adult patients with HCM have an abnormal systolic blood pressure
pressure response to exercise characterized by progressive hypotension or a failure to augment the
response systolic blood pressure that is caused by an inappropriate drop in systemic vascular resistance
and a low cardiac output reverse
Various definitions for abnormal blood pressure response in patients with HCM have been
reported; for the purposes of this guideline, an abnormal blood pressure response is defined as a
failure to increase systolic pressure by at least 20 mmHg from rest to peak exercise or fall of
>20 mmHg from peak pressure
Abnormal exercise blood pressure response is associated with a higher risk of SCD in patient
aged 40 years, but its prognostic significance in patients aged >40 years is unknown
HCM hypertrophic cardiomyopathy, LA left atrium, LVH left ventricular hypertrophy, LVOT left ventricular outflow
tract obstruction, NSVT non-sustained ventricular tachycardia, SCD sudden cardiac death, TTE transthoracic
echocardiography

Ventricular Arrhythmias and Sudden ful risk factor. Multiple risk factors in an
Cardiac Death individual strengthen the case for an implantable
Ventricular arrhythmias and sudden cardiac death defibrillator, as stated by the most recent interna-
remain dreaded outcomes of HCM, occurring in tional guidelines (Table 11.1).
young, otherwise healthy individuals. Well-known
clinical risk factors for sudden cardiac death allow
clinicians to target implantable cardioverter defi- References
brillator therapy to those who are at the highest
risk. Not all risk factors predict this outcome 1. Elliot PM et al (2014) 2014 ESC guidelines on diagno-
equally, and placement of this type of device in sis and management of hypertrophic cardiomyopathy:
young patients is associated with an important the task force for the diagnosis and management of
hypertrophic cardiomyopathy of the European Society
lifetime risk of complications [1822]. of Cardiology (ESC). Eur Heart J 35(39):27332779
A personal history of cardiac arrest or sus- 2. Maron BJ, McKenna WJ et al (2003) American col-
tained ventricular arrhythmia is the most power- lege of cardiology/european society of cardiology

ERRNVPHGLFRVRUJ
128 M. Marchesini and E. Baiocco

clinical expert consensus document on hypertrophic 13. Elliott PM, Anastasakis A et al (2014) ESC guidelines
cardiomyopathy. Eur Heart J 24:19651991 on diagnosis and management of hypertrophic cardio-
3. Rapezzi C, Arbustini E et al (2013) Diagnostic work- myopathy. Eur Heart J. doi:10.1093/eurheartj/ehu284
up in cardiomyopathies: bridging the gap between 14. Ellims AH, Iles LM et al (2014) A comprehensive
clinical phenotypes and final diagnosis. A position evaluation of myocardial fibrosis in hypertrophic car-
statement from the ESC working group on myocardial diomyopathy with cardiac magnetic resonance imag-
and pericardial diseases. Eur Heart J 34:14481458 ing: linking genotype with fibrotic phenotype. Eur
4. Nagueh SF (2014) Anderson-fabry disease and other Heart J Cardiovasc Imaging 15:11081116
lysosomal storage disorders. Circulation 15. Kellman P, Hansen MS (2014) T1-mapping in the
130:10811090 heart: accuracy and precision. J Cardiovasc Magn
5. Yusuf SW, Bathina JD et al (2011) Apical hypertro- Reson 16:120
phic cardiomyopathy. World J Cardiol 3(7):256259 16. Maron SM (2012) Clinical utility of cardiovascular
6. Nagueh SF, Bierig SM et al (2011) American society magnetic resonance in hypertrophic cardiomyopathy.
of echocardiography clinical recommendations for J Cardiovasc Magn Reson 14:13
multimodality cardiovascular imaging of patients 17. Shiozaki AA, Kim RJ (2007) Cardiovascular mag-
with hypertrophic cardiomyopathy. J Am Soc netic resonance in hypertrophic cardiomyopathy. Arq
Echocardiogr 24:473498 Bras Cardiol 88(2):216221
7. To ACY, Dhillon A et al (2011) Cardiac magnetic 18. Christiaans I, van Engelen K et al (2010) Risk stratifi-
resonance in hypertrophic cardiomyopathy. JACC cation for sudden cardiac death in hypertrophic car-
Cardiovasc Imaging 4(10):11231137 diomyopathy: systematic review of clinical risk
8. Noureldin RA, Liu S et al (2012) The diagnosis of markers. Europace 12:313321
hypertrophic cardiomyopathy by cardiovascular mag- 19. Bruder O, Wagner A et al (2010) Myocardial scar
netic resonance. J Cardiovasc Magn Reson 14:17. visualized by cardiovascular magnetic resonance
http://www.jcmr-online.com/content/14/1/17 imaging predicts major adverse events in patients with
9. Pedrotti P (2013) La risonanza magnetica cardiaca hypertrophic cardiomyopathy. J Am Coll Cardiol
nella cardiomiopatia ipertrofica. Cardiol Sci 56:875887
11:7081 20. Maron MS, Appelbaum E et al (2008) Clinical profile
10. Hundley WG, Bluemke DA et al (2010) ACCF/ACR/ and significance of delayed enhancement in hypertro-
AHA/NASCI/SCMR 2010 Expert Consensus phic cardiomyopathy. Circ Heart Fail 1:184191
Document on Cardiovascular Magnetic Resonance: A 21. OHanlon R, Grasso A et al (2010) Prognostic signifi-
Report of the American College of Cardiology cance of myocardial fibrosis in hypertrophic cardio-
Foundation Task Force on Expert Consensus myopathy. J Am Coll Cardiol 56:867874
Documents. Circulation 121:24622508 22. Rubinshtein R, Glockner JF et al (2010) Characteristics
11. Ibrahim M, Rao C et al (2012) Modern management and clinical significance of late gadolinium enhance-
of systolic anterior motion of the mitral valve. Eur J ment by contrast-enhanced magnetic resonance imag-
Cardiothorac Surg 41(6):111 ing in patients with hypertrophic cardiomyopathy.
12. Maron MS, Houser TH et al (2007) Right ventricular Circ Heart Fail 3:5158
involvement in hypertrophic cardiomyopathy. Am J
Cardiol 100:12931298

ERRNVPHGLFRVRUJ
Cardiac Amyloidosis
12
Alessandro Barbarossa and Erika Baiocco

12.1 Case Report Medical History and Cardiovascular


Risk Factors

A 67-year-old man presented to the family Smoker


doctor complaining of productive cough No familial history for cardiovascular disease
and dyspnea on exertion during ordinal
physical activity. Symptoms arose about 3
weeks ago, associated with atypical chest Allergies
pain (punctory, not correlated to exercise);
the patient denied palpitation and fever. Nonsteroidal anti-inflammatory drugs (NSAIDs)
Those symptoms were diagnosed by the
GP as an upper respiratory tract infection
and treated with i.m. antibiotic therapy Medications
(ceftriaxone 2g once daily). Despite ther-
apy, the patient remained strongly symp- None
tomatic and his family doctor decided to
program a chest X-ray that showed bi-basal
pleural effusion, more evident to the right Vital Signs
side.
The patient was referred to a cardiolo- Temperature: 36.5 C
gist for consulting; meanwhile, he starts Heart rate: 70 bpm
oral diuretic therapy (furosemide) with Blood pressure: 120/80 mmHg
consequent improvement of dyspnea and Respiratory rate: 10 breaths per minute
pleural effusion reduction. Oxygen saturation while breathing ambient
air: 98 %

A. Barbarossa (*) E. Baiocco


Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche,
Via Conca 71, Ancona 60126, Italy
e-mail: Alessandro.barbarossa@hotmail.it;
erikabaiocco@libero.it

Springer International Publishing Switzerland 2015 129


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_12

ERRNVPHGLFRVRUJ
130 A. Barbarossa and E. Baiocco

Physical Examination
Heart diseases
General: fatigued, no acute distress, alert, Heart failure (HF)
awake, and oriented. Well developed and well Tachyarrhythmias and bradyarrhythmias
nourished Pulmonary embolism
Head, eyes, ears, nose, and throat: normoce- Anemia
phalic, atraumatic, mucous membranes moist, Poor training
extraocular muscles intact, pupils equally round
and reactive to light and accommodation bilater-
ally, bilateral tympanic membrane intact, bilateral Symptoms arose quite recently and therefore
sclera anicteric, and no conjunctival injection we can exclude a poor training condition.
Neck: supple, no jugular venous distention, no The patient does not refer fever, and chest
lymphadenopathy, and no carotid bruit X-ray didnt show pulmonary consolidations, so
Cardiovascular: regular rate and rhythm; S1 the hypothesis of a pneumonitis seems also
and S2 are normal; no murmurs, rubs, or gal- unlikely.
lops; point of maximal intensity nondisplaced According to the physical exam (bi-basal rales),
and nonsustained; no hepatojugular reflux; the most probable cause is heart failure. COPD
and capillary refill less than 2 s exacerbation is also unlikely (chest X-ray), but
Lungs: rales to auscultation at the bases bilat- cannot be excluded because of his smoking habit.
erally, no rhonchi or wheezes, no egophony, Pulmonary embolism and anemia could not be
no alterations in tactile fremitus, and normal excluded only according to physical exam.
percussion
Abdomen: mild overweight, no pulsatile masses,
normal bowel sounds in all four quadrants, no Routine EKG at Rest (Fig. 12.1)
high-pitched or tinkling sounds, resonant to per-
cussion, soft, non-distended/non-tender, no Conclusions: sinus rhythm, normal atrioventricu-
rebound or guarding, no cost vertebral angle lar conduction, counterclockwise rotation on
tenderness, and no hepatosplenomegaly horizontal axis, low voltages on limb leads, and
Extremities: no cyanosis or clubbing, with diffuse nonspecific alterations in repolarization
mild peripheral edema
Neurological: cranial nerves II through XII
intact, no focal deficit Routine Laboratory Tests
Psychiatric: normal affect, no hallucinations,
normal speech, and no dysarthria Complete blood count: normal
Skin: intact, no rashes, no lesions, and no Cholesterol (total, HDL, LDL) and TG:
erythema normal
Hepatic function (GOT, GPT, -GGT, ALP,
total bilirubin, direct and indirect bilirubin):
Which Are the Possible Causes normal
for Dyspnea on Exertion and Atypical Thyroid function (TSH, FT3, FT4): normal
Chest Pain? Renal function (creatinine, BUN): normal
Electrolytes (Na+, K+, Ca++, Mg++, Cl):
normal
Lung diseases Fasting blood glucose: 78 m/dl (4.33 mmol/L)
Chronic obstructive pulmonary Troponin I-hs: 0.16 ng/ml (n.v. < 0.055 ng/ml)
disease (COPD) BNP: 1195 pg/ml (n.v. <100 pg/ml)
Pneumonitis Inflammation index: VES 3 mm/h (n.v. < 27 mm/h),
Bronchitis CRP 0.3 mg/dl (n.v. <0.6 ng/ml)
D-dimers: 213 ng/ml (n.v. <230 ng/ml)

ERRNVPHGLFRVRUJ
12 Cardiac Amyloidosis 131

Fig. 12.1 Rest EKG performed in hospital

Pulmonary Function Tests


Cardiomyopathy
No alteration in pulmonary volumes and, normal Familial (hypertrophic, dilated,
Tiffeneau index, with mild reduction in DLCO. ARVD, restrictive, left ventricular
Normal D-dimers, showed at routine labora- non-compaction)
tory tests, ruled out pulmonary embolism. Myocarditis (infective, immune medi-
According to EKG bradyarrhythmias could be ated, chemotherapy, cocaine, alcohol,
excluded and normal red blood cells count heavy metals)
excluded the presence of anemia. Finally, COPD Endocrine/nutritional (pheochromocy-
was excluded by pulmonary function tests. toma, vitamin deficiency, hypophospha-
The diagnosis of heart failure is now very temia, hypocalcemia)
likely according to symptoms, physical exam, Pregnancy
chest radiography, and laboratory tests. Infiltration (amyloidosis,
malignancy)
Valvular heart disease
Which Are the Possible Causes Mitral
of Heart Failure? Aortic
Tricuspid
Pulmonary
Myocardial disease Pericardial disease
Coronary artery disease Constrictive pericarditis
Hypertension Pericardial effusion

ERRNVPHGLFRVRUJ
132 A. Barbarossa and E. Baiocco

Acute myocarditis remains a valid possibility


Congenital heart disease (rapid onset of symptoms, presence of cough and
Arrhythmias dyspnea) and also pericarditis (atypical chest
Tachyarrhythmias (atrial, ventricular) pain and low voltages, albeit in only limb leads,
Bradyarrhythmias (sinus node dys- at basal EKG). In addition infiltrative cardiomy-
function, atrioventricular block) opathy cannot be excluded.
High-output states The next step was to record an echocardio-
Anemia gram in order to evaluate function and morphol-
Sepsis ogy of the left ventricle.
Thyrotoxicosis
Pagets disease
Arteriovenous fistula Echocardiography Results
Volume overload (Fig. 12.2a, b)
Renal failure
Iatrogenic (postoperative fluid infusion) Bi-atrial enlargement (LA diameter M-mode = 50
cm; area 4c = 26 cm2, RA area 4c = 20 cm2). Small
size of the left ventricle (iLVEDV 44 ml/m2),
increased wall thickness (IVSDd = 1.4 cm) with
HF may be the final result of different patho- granular sparkling appearance (particularly evi-
logical pathways. However, some possibilities dent at the level of the inter ventricular septum)
and reduced systolic function (ejection fraction
can be excluded just considering medical history,
with Simpsons rule 0.45). Normal size of the right
physical examination, and EKG. Our patient was ventricle, with increased free wall thickness and
smoker, but did not have other risk factors for reduced systolic function (TAPSE 11 mm). Inter
coronary artery disease (CAD); a diagnosis of atrial septum thickening.
Mitral valve leaflets are thickened with mild
ischemic cardiomyopathy was unlikely. A heart
regurgitation.
valvular disease was excluded by the physical Tricuspid valve leaflets are also thickened with
examination. The patient had not any familiar his- mild regurgitation and normal systolic pressure
tory for genetic cardiomyopathies (ARVD, hyper- gradient (PASP = 30 mmHg).
Normal aortic and pulmonic valves.
trophic, non-compaction, dilated), albeit this is
The inferior vena cava is slightly dilated (26
not an absolute exclusion criteria. The basal EKG mm). There is less than 50% inspiratory collapse
did not show brady- or tachyarrhythmias and the of the IVC.
patient did not complain palpitations. Finally, Small circumferential pericardial effusion, not
hemodynamically significant.
normal routine laboratory tests excluded anemia,
Restrictive diastolic pattern with increased fill-
thyrotoxicosis, renal failure, and sepsis. ing pressure (E/A 4, E/E 29, E dec time 147 m/sec).

a b

Fig. 12.2 (a, b) PLAX and apical 4-CH short axis showed left ventricular hypertrophy with granular sparkling
appearance and pericardial effusion

ERRNVPHGLFRVRUJ
12 Cardiac Amyloidosis 133

Conclusion: mild biventricular systolic aspect and severe diastolic dysfunction) in


dysfunction. Biventricular wall thicknesses with
addition to low electrocardiographic limb lead
granular sparkling aspect. Bi-atrial enlargement.
Severe left ventricular diastolic dysfunction. Mild voltages makes an infiltrative cardiac disease
pleural effusion. Not significant valve regurgita- more likely.
tion or gradients.

The echocardiography findings were compat- Cardiac RMN


ible with hypertrophic/hypertensive or infiltrative
cardiomyopathy. In order to acquire further diagnostic elements
The patient had no history of hypertension and about ventricle morphology of the ventricles, a
arterial pressure values were normal during cardiac RMN was performed (Fig. 12.3a, b).
physical examination, so a hypertensive cardio- [] bi-ventricular parietal hypertrophy and systolic
myopathy was unlikely. dysfunction (LVEF 41%, RVEF 47%). Bi-atrial
enlargement (left > right). Mild mitral and tricuspid
Distinguishing between hypertrophic and regurgitation. No myocardial edema. Post contrasto-
infiltrative, only by echocardiography, is an graphic diffuse subendocardial late enhancement.
easy task. The ventricle involvement is usually Moderate circumferential pericardial effusion.
concentric in the infiltrative forms and eccentric Conclusion: pattern suggestive of cardiac
amyloidosis
in the hypertrophic forms. The presence of
dynamic obstruction of left ventricular output There are three main types of cardiac
tract is more frequent in hypertrophic forms. A amyloidosis: AL-related amyloidosis, ATTRm
pericardial effusion and the typical pattern of amyloidosis, and ATTRwt amyloidosis.
granular sparkling of the myocardium are sug- According to familial history negative for
gestive of an infiltrative form. A severe diastolic cardiovascular disease, the patients age, and
dysfunction (grade III) may be found in both epidemiology, the most likely diagnosis is
diseases. AL-related amyloidosis.
In our case the typical echocardiographic pat- The next step to confirm this hypothesis is to
tern (wall thickening with granular sparkling evaluate the presence of a high production of

Fig. 12.3 (a, b) Magnetic resonance showed areas of delayed enhancement (DA), diagnostic for amyloidosis and
pleuropericardial effusion

ERRNVPHGLFRVRUJ
134 A. Barbarossa and E. Baiocco

Red Flags

Some characteristic features, founded


during diagnostic work up, could help to
reach the final correct diagnosis minimiz-
ing the time and use of inappropriate exams:

Clinical history substantially negative


for cardiovascular risk factors
Cardiac physical examination negative
for murmurs; presence of signs of high
Fig. 12.4 Monoclonal proliferation at serum
electrophoresis central venous pressure
Low voltages in limb leads at standard
EKG
Monoclonal peak in the gamma zone at
immunoglobulin light chains. For this purpose serum electrophoresis
we performed a serum electrophoresis, which Typical echocardiographic appearance
showed a monoclonal peak in the gamma zone (concentric hypertrophy with granular
immunofixation that demonstrates an increase sparkling pattern, mild pericardial effusion,
of light-chain quantity in blood and urine valve and interatrial septum thickening)
(Fig. 12.4).

Bone marrow biopsy (BOM) confirmed the


diagnosis of multiple myeloma (MM), and 12.2 Amyloidosis
finally, abdomen fat biopsy confirmed the diag-
nosis of amyloidosis. Classication

According to the literature there are three most


frequent type of amyloidosis [1]:
Final Diagnosis
Acquired monoclonal immunoglobulin
Cardiac-acquired monoclonal immunoglobulin light-chain amyloidosis (AL): characterized
light-chain (AL) amyloidosis. by clonal plasma cells in the bone marrow that
The patient was treated with i.v. diuretic with produce the immunoglobulin light chains of
consequent progressive resolution of the the fibrillary deposits. The most common
peripheral edema, lung congestion, and plasma dyscrasia associated with this form is
improvement of dyspnea and NYHA functional multiple myeloma (MM).
class. Because of low potassium serum level, an Hereditary transthyretin-related form
oral potassium-sparing diuretic was added. (ATTRm): can be caused by >100 mutations of
Other heart failure-specific drugs (that were transthyretin, a transport protein synthesized
introduced during diagnostic work up) such as mainly by the liver.
renin-angiotensin-aldosterone antagonist Wild-type (nonmutant) TTR-related amyloido-
(ACE-I and ARBs) and beta-blockers were sis (ATTRwt): nonhereditary senile systemic
interrupted according to scientific evidences. amyloidosis, which affects mainly the hearts
Patient, after clinical stabilization, was of elderly men caused by cardiac deposition of
referred to a hematologist for implementation of amyloid derived from wild-type transthyretin
the amyloidosis-specific therapy. (with a normal amino acid constitution) [2].

ERRNVPHGLFRVRUJ
12 Cardiac Amyloidosis 135

Pathophysiology About 50 % of patients with AL-related amy-


loidosis present with rapidly progressive symp-
Cardiac amyloidosis is a manifestation of one of toms such as dyspnea, almost always associated
several systemic diseases known as amyloido- with evidence of elevated right-sided filling pres-
sis. The common feature of this group of dis- sure and peripheral edema, and in late-stage dis-
eases is the extracellular deposition of a ease, ascites is not uncommon. Also for ATTRm
proteinaceous material that, when stained with and ATTRwt forms, heart failure is the main
Congo red, demonstrates apple-green birefrin- clinical manifestation, albeit, differently from AL
gence under polarized light and that has a distinct form, progression is slower but also inexorable.
color when stained with sulfated Alcian Blue [3].
Cardiac involvement may be the predominant Small Vessel Disease
feature or may casually be found during investi- Vascular involvement may manifest as purpura,
gation of a patient presenting with another major claudication, or angina. The presence of perior-
organ involvement. Its predominance varies bital purpura, often occurring with coughing,
regarding amyloidosis type: senile systemic amy- sneezing, or very minor trauma, suggests capil-
loidosis and some forms of transthyretin amyloi- lary involvement, and its presence in a patient
dosis invariably affect the heart, whereas cardiac with unexplained heart failure strongly suggests
involvement ranges from absent to severe in amy- amyloidosis, almost always of the AL type. The
loidosis derived from a light-chain precursor. presence of claudication (principally leg and
Otherwise, secondary form of amyloidosis jaw), angina, or both is suggestive of vascular
almost never affects the heart in a clinically sig- amyloidosis. Typically, the involved vessels are
nificant manner [4]. small and intramyocardial; as a result, coronary
Regardless of the underlying pathogenesis of angiography is usually normal or shows only
amyloid production, cardiac amyloidosis is a minor abnormalities [5, 6].
myocardial disease characterized by extracellu-
lar amyloid infiltration throughout the heart. Syncope
Amyloid deposits are located in the ventricles Syncope or presyncope is a common finding in
and atria, perivascularly (particularly in the patients with cardiac amyloidosis and may be
small vessels), and in the valves. It may also due to multiple additional factors. These include
involve the conduction system. Thus, it may postural or exertional hypotension due to exces-
result in initially diastolic dysfunction of both sive diuresis or autonomic neuropathy. Moreover,
ventricles, followed by deterioration of systolic an exercise syncope may represent an inability to
function and precipitating heart failure. augment cardiac output; when that occurs a high
Moreover, symptoms and signs of myocardial mortality rate is expected in 3 months, often due
ischemia may occur because of the presence of to sudden cardiac death.
vascular infiltration and conduction distur- Documented ventricular arrhythmias are an
bances (such as atrioventricular blocks and bun- infrequent cause of syncope, maybe because the
dle branch blocks) with conduction system amyloid heart reacts poorly to hypoperfusion and
involvement [3]. any severe tachyarrhythmia causing loss of con-
sciousness is usually fatal [7].

Clinical Features Conduction System Disease


Despite a widespread involvement of the conduc-
Heart Failure tion system, high-degree atrioventricular block is
Patients with cardiac amyloidosis usually com- an unusual feature in AL-related amyloidosis,
plain symptoms of biventricular heart failure and symptomatic sinus node dysfunction appears
with some differences between the three main uncommon. In contrast a progressive conduction
forms. system disease often occurs in the ATTR amyloi-

ERRNVPHGLFRVRUJ
136 A. Barbarossa and E. Baiocco

dosis (both senile and familial), and a pacemaker Nuclear imaging: scintigraphy with 99 mTc-
implantation is often required [8]. 3,3-diphosphono-1,2-propanodicarboxylic
acid (99mTc-DPD) may be useful in differen-
Extracardiac Manifestation tial diagnosis between AL and ATTR amyloi-
Neurological (peripheral and autonomic neurop- dosis [12].
athy, carpal tunnel syndrome), dermatological Serum electrophoresis: the presence of a serum
(periorbital purpura, nail dystrophy), macroglos- or urine monoclonal paraprotein in the setting of
sia (typical of AL form). a typical echocardiogram is suggestive of
Senile cardiac amyloidosis (ATTRwt), which AL-related amyloidosis, but it alone does not
is different from AL and ATTRm amyloidosis, firmly establish the diagnosis. In fact, as demon-
usually is not associated with other major organ strated in some studies, older males with clini-
involvements [3]. cally isolated cardiac involvement and a small
monoclonal gammopathy may have senile car-
diac amyloid and unrelated MGUS [13].
Diagnosis Tissue biopsy: the final diagnosis of cardiac
amyloidosis requires demonstration of amyloid
Physical examination: signs and symptoms deposits in the heart with endomyocardial biopsy
of heart failure (rales, high jugular venous or, in patients with appropriate imaging cardiac
pressure, hepatomegaly); usually systolic findings, demonstration of amyloid deposits at
BP is low (<100 mmHg); purpura may be histologic examination of other tissues, like the
present. abdominal fat pad, rectum, or kidney.
EKG: low voltage in the limb leads is one of Abdominal fat pad aspirate staning with Congo
the most common electrocardiogram (EKG) red will result positive in more of 70 % of
abnormalities in AL cardiac amyloidosis patients with AL-related amyloidosis, but it is
(occurring in approximate 50 %), although it subject to false positive especially in center
is less common in the other forms of cardiac with low expertise. Instead, bone marrow
amyloidosis, being reported in about 25 % of biopsy should be the test of choice to confirm
patients with familial disease (ATTRm) and in the suspect of AL-related amyloidosis, because
about 40 % of patients with senile cardiac it demonstrates evidence of a plasma cell dys-
amyloidosis (ATTRwt) [1, 3]. crasia in >80 % of patients and shows amyloid
Echocardiography: thickness of the atrium deposits in about 60 % [14].
and ventricular walls (which may or may not
be associated with increased echogenicity) as
well as thickened valve leaflets and interatrial Prognosis and Therapy
septum. Diastolic dysfunction and reduction
in longitudinal systolic function precede AL-related cardiac amyloidosis is associated
reduction in left ventricular ejection fraction. with a worse prognosis and more rapid progres-
Atrial dysfunction is frequently present and sion of heart failure (mainly due to diastolic dys-
may be associated with appendage thrombus function) compared with the other forms [1],
even in the absence of a history of atrial fibril- with a median survival of 6 months without spe-
lation [911]. cific therapy [3]. ATTRm forms, differently, have
Cardiovascular magnetic resonance: cardio- a better long-term survival [15].
vascular magnetic resonance (CMR) imaging
can provide evidence strongly suggestive of
amyloid cardiomyopathy, particularly a dis- Treatment
tinctive pattern of global left ventricular late
gadolinium enhancement (LGE) rarely seen in Treatment requires a twofold approach: manage-
other cardiomyopathies. ment of cardiac-related complications due to

ERRNVPHGLFRVRUJ
12 Cardiac Amyloidosis 137

amyloid deposition (which is similar regardless of amyloid cardiomyopathy, free from other comor-
the specific type of amyloid) and treatment of the bidities [20, 21].
underlying disease to suppress new amyloid for-
mation (which is tailored for each specific form). Specic Therapy
AL-related amyloidosis: the specific therapy
Cardiac-Related Therapy aims to stop the production of abnormal light
Standard drugs used for the treatment of conges- chains by the plasma cells. This can be achieved
tive heart failure are not useful and poorly toler- with a specific chemotherapy. Because each
ated or in some cases may be dangerous in patient is different, the dosage and choice of
patients with amyloidosis. drug require assessment by a hematologist in
Beta-blockers (BBs) and calcium channel order to minimize any adverse side effects.
blockers (CCBs) are deleterious because they Measuring the free light chains in the blood
decrease heart rate, which is the only mechanism assesses the response to AL-related amyloido-
that can maintain cardiac output in these patients sis treatment. The levels will normalize in suc-
with severe diastolic dysfunction; moreover, they cessful treatment, usually before there is a
may also aggravate autonomic dysfunction like definite improvement in the symptoms. The
angiotensin-converting enzyme inhibitors (ACE- initial therapy is often changed if there is no
i) and angiotensin receptor blockers (ARB) [16]. clear response over the first 23 cycles [18].
Moreover, these drugs may increase the risk of ATTRm (familial amyloidosis): because the
hypotension. main source of mutant transthyretin is the
Digitalis has been shown to accumulate in car- liver, transplantation is currently the treatment
diac amyloid deposits in in vitro studies; for this of choice in carefully selected patients.
reason its use may be harmful in patients with Intensive investigation is underway to develop
cardiac amyloidosis [17]. and test drugs that can prevent the production
Loop diuretics (e.g., furosemide), given at of amyloid in patients with the abnormal
high dosage in patients with severe fluid reten- genes [18].
tion, are the mainstay of therapy for ATTRwt (senile amyloidosis): no specific ther-
amyloid-related symptoms, although they dont apy currently exists.
reduce mortality. When fluid retention is severe,
a booster diuretic (metolazone) can be used inter- Conclusion
mittently. Furthermore, daily use of spironolac- Although cardiac amyloidosis is uncommon,
tone in addition to loop diuretics could help to knowledge of typical clinical and instrumental
keep normal potassium level [18]. features is essential for the diagnosis in order
In case of arrhythmias, amiodarone should be to start the correct treatment and to avoid
considered as the first-line therapy. unnecessary, and sometimes harmful,
Anticoagulation therapy is mandatory in therapies.
patients with supraventricular arrhythmias and The distinction between the major amyloi-
may be considered in patients with sinus rhythm dosis forms is fundamental because specific
but with severe contractile atrial dysfunction. therapies and prognosis differ greatly from
Pacemaker implantation may be indicated in one another. Moreover, early diagnosis is criti-
patients with symptomatic bradycardia or con- cal because patients with advanced disease are
duction disorders, while the utility of implantable usually too compromised for intensive
cardioverter defibrillators (ICDs) is still contro- chemotherapies.
versial because electromechanical dissociation Despite recent progress in knowledge of
seems to be the more frequent cause of sudden the disease mechanism, cardiac amyloidosis is
cardiac death in these patients [19]. a severe disease with a poor prognosis and few
Finally, heart transplantation could be a solu- therapeutic possibilities. For this reason fur-
tion in selected young patients, with advanced ther studies and investigation are necessary.

ERRNVPHGLFRVRUJ
138 A. Barbarossa and E. Baiocco

References Serial Doppler echocardiographic follow-up of left


ventricular diastolic function in cardiac amyloidosis.
J Am Coll Cardiol [Internet] 16:11351141. [cited
1. Rapezzi C, Merlini G, Quarta CC, Riva L, Longhi S,
2014 Dec 11]. Available from: http://www.ncbi.nlm.
Leone O, Salvi F, Ciliberti P, Pastorelli F, Biagini E,
nih.gov/pubmed/2229760
Coccolo F, Cooke RMT, Bacchi-Reggiani L,
11. Patel AR, Dubrey SW, Mendes LA, Skinner M,
Sangiorgi D, Ferlini A, Cavo M, Zamagni E, Fonte
Cupples A, Falk RH, Davidoff R (1997) Right ven-
ML, Palladini G, Salinaro F, Musca F, Obici L, Branzi
tricular dilation in primary amyloidosis: an indepen-
A, Perlini S (2009) Systemic cardiac amyloidoses:
dent predictor of survival. Am J Cardiol [Internet]
disease profiles and clinical courses of the 3 main
80:486492. [cited 2014 Dec 11]. Available from:
types. Circulation [Internet] 120:12031212. [cited
http://www.ncbi.nlm.nih.gov/pubmed/9285663
2014 Mar 19]. Available from: http://www.ncbi.nlm.
12. Perugini E, Guidalotti PL, Salvi F, Cooke RMT,
nih.gov/pubmed/19752327
Pettinato C, Riva L, Leone O, Farsad M, Ciliberti P,
2. Westermark P, Sletten K, Johansson B, Cornwell GG
Bacchi-Reggiani L, Fallani F, Branzi A, Rapezzi C
(1990) Fibril in senile systemic amyloidosis is derived
(2005) Noninvasive etiologic diagnosis of cardiac
from normal transthyretin. Proc Natl Acad Sci U S A
amyloidosis using 99mTc-3,3-diphosphono-1,2-
[Internet] 87:28432845. [cited 2014 Dec 4].
propanodicarboxylic acid scintigraphy. J Am Coll
Available from: http://www.pubmedcentral.nih.gov/
Cardiol [Internet] 46:10761084. [cited 2014 Jun 9].
articlerender.fcgi?artid=53787&tool=pmcentrez&ren
Available from: http://www.ncbi.nlm.nih.gov/
dertype=abstract
pubmed/16168294
3. Falk RH (2005) Diagnosis and management of the
13. Lachmann HJ, Booth DR, Booth SE, Bybee A,
cardiac amyloidoses. Circulation [Internet] 112:2047
Gilbertson JA, Gillmore JD, Pepys MB, Hawkins PN
2060. [cited 2014 Dec 1]. Available from: http://www.
(2002) Misdiagnosis of hereditary amyloidosis as AL
ncbi.nlm.nih.gov/pubmed/16186440
(primary) amyloidosis. N Engl J Med [Internet]
4. Dubrey SW, Cha K, Simms RW, Skinner M, Falk RH
346:17861791. [cited 2014 Dec 11]. Available from:
(1996) Electrocardiography and Doppler echocar-
http://www.ncbi.nlm.nih.gov/pubmed/12050338
diography in secondary (AA) amyloidosis. Am J
14. Swan N, Skinner M, OHara CJ (2003) Bone marrow
Cardiol [Internet]. 77:313315. [cited 2014 Dec 2].
core biopsy specimens in AL (primary) amyloidosis.
Available from: http://www.ncbi.nlm.nih.gov/
A morphologic and immunohistochemical study of
pubmed/8607418
100 cases. Am J Clin Pathol [Internet] 120:610616.
5. Al Suwaidi J, Velianou JL, Gertz MA, Cannon RO,
[cited 2014 Dec 11]. Available from: http://www.ncbi.
Higano ST, Holmes DR, Lerman A (1999) Systemic
nlm.nih.gov/pubmed/14560572
amyloidosis presenting with angina pectoris. Ann
15. Dubrey SW, Cha K, Skinner M, LaValley M, Falk RH
Intern Med [Internet] 131:838841. [cited 2014 Dec
(1997) Familial and primary (AL) cardiac amyloido-
11]. Available from: http://www.ncbi.nlm.nih.gov/
sis: echocardiographically similar diseases with dis-
pubmed/10610629
tinctly different clinical outcomes. Heart [Internet]
6. Mueller PS, Edwards WD, Gertz MA (2000)
78:7482. [cited 2014 Dec 4]. Available from: http://
Symptomatic ischemic heart disease resulting from
www.pubmedcentral.nih.gov/articlerender.fcgi?artid
obstructive intramural coronary amyloidosis. Am J
=484868&tool=pmcentrez&rendertype=abstract
Med [Internet] 109:181188. [cited 2014 Dec 11].
16. Bouhour JB, Haddak M, Lefevre M (1986) Risks of
Available from: http://www.ncbi.nlm.nih.gov/
beta-blockers and calcium inhibitors in amyloid cardi-
pubmed/10974179
opathy. Presse Med [Internet] 15:981. [cited 2014
7. Chamarthi B, Dubrey SW, Cha K, Skinner M, Falk
Nov 19]. Available from: http://www.ncbi.nlm.nih.
RH (1997) Features and prognosis of exertional syn-
gov/pubmed/2874550
cope in light-chain associated AL cardiac amyloido-
17. Rubinow A, Skinner M, Cohen AS (1981) Digoxin
sis. Am J Cardiol [Internet] 80:12421245. [cited
sensitivity in amyloid cardiomyopathy. Circulation
2014 Dec 11]. Available from: http://www.ncbi.nlm.
[Internet] 63:12851288. [cited 2014 Nov 19].
nih.gov/pubmed/9359565
Available from: http://www.ncbi.nlm.nih.gov/
8. Mathew V, Olson LJ, Gertz MA, Hayes DL (1997)
pubmed/7014028
Symptomatic conduction system disease in cardiac
18. Quarta CC, Kruger JL, Falk RH (2012) Cardiac amy-
amyloidosis. Am J Cardiol [Internet] 80:14911492.
loidosis. Circulation [Internet] 126:e178e182. [cited
[cited 2014 Dec 11]. Available from: http://www.ncbi.
2014 Dec 2]. Available from: http://www.ncbi.nlm.
nlm.nih.gov/pubmed/9399732
nih.gov/pubmed/22988049
9. Sedlis SP, Saffitz JE, Schwob VS, Jaffe AS (1984)
19. Lin G, Dispenzieri A, Kyle R, Grogan M, Brady PA
Cardiac amyloidosis simulating hypertrophic cardio-
(2013) Implantable cardioverter defibrillators in
myopathy. Am J Cardiol [Internet] 53:969970. [cited
patients with cardiac amyloidosis. J Cardiovasc
2014 Dec 11]. Available from: http://www.ncbi.nlm.
Electrophysiol [Internet] 24:793798. [cited 2014
nih.gov/pubmed/6538383
Dec 11]. Available from: http://www.ncbi.nlm.nih.
10. Klein AL, Hatle LK, Taliercio CP, Taylor CL,
gov/pubmed/23489983
Kyle RA, Bailey KR, Seward JB, Tajik AJ (1990)

ERRNVPHGLFRVRUJ
12 Cardiac Amyloidosis 139

20. Mignot A, Varnous S, Redonnet M, Jaccard A, Epailly 21. Sattianayagam PT, Gibbs SDJ, Pinney JH, Wechalekar
E, Vermes E, Boissonnat P, Gandjbakhch I, Herpin D, AD, Lachmann HJ, Whelan CJ, Gilbertson JA,
Touchard G, Bridoux F (2008) Heart transplantation Hawkins PN, Gillmore JD (2010) Solid organ trans-
in systemic (AL) amyloidosis: a retrospective study of plantation in AL amyloidosis. Am J Transplant
eight French patients. Arch Cardiovasc Dis [Internet] [Internet] 10:21242131. [cited 2014 Nov 19].
101:523532. [cited 2014 Nov 19]. Available from: Available from: http://www.ncbi.nlm.nih.gov/pub
http://www.ncbi.nlm.nih.gov/pubmed/19041836 med/20883547

ERRNVPHGLFRVRUJ
Left Ventricular Non-compaction
Cardiomyopathy 13
Alessandro Maolo and Simona Masiero

13.1 Case Report Medical History and Cardiovascular


Risk Factors

A 50-year-old man referred to the family Type 2 diabetes mellitus


doctor for worsening dyspnea and fatigue Hypertension
during ordinary physical activities appear- No previous or current pathology
ing 3 months earlier. From that time on, No fever episode in the last 3 years
symptoms arise every time the patient
walks a little longer than usual even at flat
level. Allergies
At anamnesis, he reported two syncopes
preceded by dyspnea and dizziness without No allergy is referred by the patient.
any feeling of palpitation or sweating. The
first one occurred while he was walking
and was complicated by concussion. The Social History
second episode occurred while he was
watching television. In both cases, he He works as a photographer.
recovered rapidly without neurological He smokes 45 cigarettes per day.
signs. He never used illicit drugs.
The family doctor referred the patient to He drinks a glass of wine only in particular
the hospital for further investigation. occasions.

Medications

Sitagliptin 50 mg + metformin 850 mg at


8:00 a.m. and 12:00 a.m., metformin 850 mg at
A. Maolo (*) S. Masiero 20:00 p.m.
Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche,
Via Conca 71, Ancona 60126, Italy
e-mail: alessandro.maolo@alice.it;
simo_masiero@libero.it

Springer International Publishing Switzerland 2015 141


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_13

ERRNVPHGLFRVRUJ
142 A. Maolo and S. Masiero

Vital Signs What Are the Possible Causes


for Dyspnea on Exertion and Fatigue
BMI: 27 in This Case?
Temperature: 36.7 C
Heart rate: 75 bpm
Blood pressure: 130/80 mmHg The following hypotheses were considered:
Respiratory rate: 16 breaths per minute
Oxygen saturation while breathing ambient Flu
air: 98 % Lung diseases
COPD
Pneumonitis
Physical Examination Bronchitis
Heart diseases
General: fatigued, no acute distress, alert, Heart failure
awake, and oriented. Well developed and well Bradyarrhythmias
nourished Tachyarrhythmias
Head, eyes, ears, nose, throat: normocephalic, Poor training
atraumatic, mucous membranes moist, extra- Recent weight gain
ocular muscles intact, pupils equally round
and reactive to light and accommodation bilat-
erally, bilateral tympanic membrane intact,
bilateral sclera anicteric, no conjunctival The patient denies a recent weight gain (his
injection BMI was 27 at the last family doctor visit). The
Neck: supple, no jugular venous distention, no symptoms arose 3 months ago that makes a poor
lymphadenopathy, no carotid bruit training unlikely considering the recent emer-
Cardiovascular: regular rate and rhythm, S1 gence of the dyspnea.
and S2 are normal, S3 present, no murmurs or The patient doesnt refer fever, cough, or other
rubs, point of maximal intensity nondisplaced disorders related to flu, so the hypothesis of a
and non-sustained, no hepatojugular reflux, pneumonitis or bronchitis seems unlikely, too.
capillary refill less than 2 s According to the physical exam (rales at the
Lungs: rales at auscultation at the bases bilat- bases bilaterally and S3 present), heart failure is
erally, no rhonchi or wheezes, no egophony, the most likely cause.
no alterations in tactile fremitus, normal
percussion ECG
Abdomen: mild overweight, no pulsatile masses, A routine ECG at rest was performed (Fig. 13.1):
normal bowel sounds in all four quadrants, no sinus rhythm, normal atrioventricular and intra-
high-pitched or tinkling sounds, resonant to per- ventricular conduction, and no abnormalities in
cussion, soft, non-distended/non-tender, no repolarization.
rebound or guarding, no costovertebral angle
tenderness, no hepatosplenomegaly Chest X-Ray
Extremities: no cyanosis or clubbing, mild A chest x-ray was performed too (Fig. 13.2):
peripheral edema cardiac shadow was slightly enlarged with an
Neurologic: cranial nerve II through XII increase of cardiac transverse diameter with
intact, no focal deficit left ventricular preponderance. A left-sided
Psychiatric: normal affect, no hallucinations, pleural effusion was seen obliterating the costo-
normal speech, no dysarthria phrenic recess and was associated to a bilateral
Skin: intact, no rashes, no lesions, no hilar enlargement with widespread bronchovas-
erythema cular signs.

ERRNVPHGLFRVRUJ
13 Left Ventricular Non-compaction Cardiomyopathy 143

Fig. 13.1 Routine ECG. Notice the normal sinus rhythm without abnormalities in conduction or repolarization

bilirubin, direct and indirect): normal


Thyroid function (TSH, FT3, FT4): normal
Renal function (creatinine, BUN): normal
Electrolytes (Na+, K+, Ca++, Mg++, Cl):
normal
Fasting blood glucose: 154 m/dl (8.56 mmol/L)
HbA1c: 7.1 % (54 mmol/mol)
TnI-hs and CK-MB: normal
BNP: 673 pg/ml

What Are the Possible Causes


of Heart Failure?

Myocardial disease
Coronary artery disease
Fig. 13.2 Chest x-ray performed for the worsening dys-
pnea. The bronchovascular marking is evident Hypertension
Cardiomyopathy
Routine Laboratory Tests Familial (hypertrophic, dilated,
Complete blood count: normal ARVD, restrictive, left ventricular
Cholesterol (total, HDL, LDL) and TG: normal non-compaction)
Hepatic function (GOT, GPT, -GT, ALP, total

ERRNVPHGLFRVRUJ
144 A. Maolo and S. Masiero

ventricle was normal in size with normal function


Myocarditis (infective, immune- (TAPSE 22 mm). There was a mild tricuspid
mediated, chemotherapy, cocaine, regurgitation centrally directed. There was no evi-
alcohol, heavy metals) dence of atrial septal defects. The left ventricle
Endocrine/nutritional (pheochro- was slightly dilated (indexed left end-diastolic vol-
mocytoma, vitamin deficiency, ume 80 ml/m2) with severe reduction of systolic
hypophosphatemia, hypocalcemia) function and areas of prominent trabeculae in the
Pregnancy apex and the side wall. Ejection fraction measured
Infiltration (amyloidosis, with Simpsons biplane method was 30 %. The
malignancy) aorta was normal (aortic root dimension, 2.7 cm;
Valvular heart disease ascending aorta, 3.1 cm; aortic arch, 3.1 cm; tho-
Mitral racic aorta, 2.8 cm; abdominal aorta, 1.8 cm). IVC
Aortic was slightly dilated with an inspiratory collapse
Tricuspid <50 % (RAP =10 mmHg and PAPS = 35 mmHg).
Pulmonary Conclusions: There is severe reduction of left
Pericardial disease ventricular function. Left ventricle walls are homo-
Constrictive pericarditis geneously hypokinetic. Morphologic alterations of
Pericardial effusion the apex and side wall of the left ventricle compati-
Congenital heart disease ble with non-compaction of the myocardium.
Arrhythmias Because of that morphologic pattern of the left
Tachyarrhythmias (atrial, ventricular) ventricle, a cardiac RMN was requested
Bradyarrhythmias (sinus node dys- (Fig. 13.4af). [] Prominent endocardic
function, atrioventricular block) apical-subapical trabeculae, extended to the side
High output states wall and the middle of LV, compared to a com-
Anemia pact epicardial layer (Noncompaction/compac-
Sepsis tion ratio 2.3). Those findings can be related to
Thyrotoxicosis areas of myocardial non-compaction.
Pagets disease The patient was diagnosed with left ventricu-
Arteriovenous fistula lar non-compaction cardiomyopathy.
Volume overload The patient was monitored, and an intrave-
Renal failure nous diuretic therapy was introduced with clini-
Iatrogenic (postoperative fluid infusion) cal benefit. Symptoms resolved and dyspnea and
fatigue disappeared.

The diagnosis of heart failure is now very


likely according to symptoms, physical exam, Now the Heart Failure Is Explained.
chest radiography, and laboratory tests. But What About the Syncope? What
Are the Possible Causes?
Echocardiography
Echocardiography was also performed to evalu-
ate function and morphology of cardiac valves Neurally mediated syncope (vasovagal,
and the left ventricle (Fig. 13.3a, b). situational, carotid sinus syncope)
Aortic valve was normal and trileaflet. Normal Orthostatic hypotension (primary auto-
dimensions of the left atrium (LA diameter nomic failure as Parkinsons disease or
M-mode, 3.9 cm; area 4c, 13 cm2). The mitral secondary autonomic failure as diabetes
valve, tricuspid valve, and pulmonic valve had or amyloidosis, drug-induced, volume
normal function and morphology; the right atrium depletion)
was also normal (area 4c = 13 cm2). The right

ERRNVPHGLFRVRUJ
13 Left Ventricular Non-compaction Cardiomyopathy 145

a b

Fig. 13.3 Echocardiogram: (a) apical four-chamber view. The marked trabeculae of the apex can be noticed. (b) Apical
two-chamber view. Apex trabeculae are visible also in this view

Cardiac syncope (bradyarrhythmias, 13.2 Left Ventricular


tachyarrhythmias as supraventricular or Non-compaction
ventricular tachycardia, drug-induced,
structural disease as aortic stenosis) Left ventricular non-compaction (LVNC) is a rare
congenital cardiomyopathy characterized by a
spongy feature of the myocardium. A thin, com-
pacted epicardial layer under an extensive non-
Episodes of hypoglycemia could be a possible compacted endocardial layer can be noticed with
cause too, but in that case, the syncope has differ- the common cardiac image techniques. Prominent
ent characteristics, and in addition, it can be trabeculation and deep recesses that communicate
excluded, thanks to the fasting glucose measured with the left ventricular cavity can be visible too.
by the patient at home and the laboratory tests The spongy morphologic feature is not charac-
that indicate an elevated HbA1c. teristic only of the left ventricle but can also involve
In this case, considering the left ventricular the right ventricle. This is why talking about non-
dysfunction and the morphologic abnormalities compaction cardiomyopathy seems to be more
of the myocardium, the most probable and dan- correct. The non-compaction of the myocardium is
gerous cause of syncope could be ventricular a consequence of an abnormal embryonic develop-
arrhythmias. ment of the ventricular myocardium. During the
Indeed, during hospitalization, a non-sustained eighth week of gestation, when the interventricular
ventricular tachycardia lasting 23 s was recorded septum is complete, the increase of ventricular vol-
at the ECG monitoring symptomatic for dizzi- umes is responsible of compression against the tra-
ness and profound weakness but without beculae of the ventricular walls. This lead to the
syncope. growth of the compact layer and the organization
of some trabeculae in the papillary muscle.
At the same time, intramyocardial vascular-
Final Diagnosis ization develops from the coronary arteries fol-
lowing an epicardial to endocardial, base to apex,
The final diagnosis was non-sustained ventricular and septum to side wall direction [1]. For this
tachycardia symptomatic of dizziness in patient reason, the non-compaction morphology is more
with left ventricular non-compaction and heart common in the apex, side, and inferior walls of
failure. the left ventricle.

ERRNVPHGLFRVRUJ
146 A. Maolo and S. Masiero

a b

d
c

e f

Fig. 13.4 (af) Cardiac RMN, short-axis view of the left ventricle in different levels (T1-weighted black blood imag-
ing). In these images, trabeculas extension to the side wall is visible

ERRNVPHGLFRVRUJ
13 Left Ventricular Non-compaction Cardiomyopathy 147

Epidemiology Clinical Manifestations

The overall prevalence of LVNC in the general Clinical manifestation of LVNC is very heteroge-
population is not well known, but it seems to be neous. The major clinical manifestations of LVNC
ranged between 1 and 3 % [1, 2]. This percentage are three, and they can appear simultaneously or
is probably an underestimate. The described separately: heart failure, supraventricular or ven-
prevalence of LVNC is 34 % among patients tricular arrhythmias, and thromboembolic events.
with heart failure [3, 4]. In the pediatric population, heart failure is the pre-
dominant clinical manifestation, while in the
adult population, there is an overlap between the
Genetics three clinical manifestations as first clinical pre-
sentation without a clear predominance of one
Non-compaction cardiomyopathy is a heteroge- over the other [6].
neous disease from the genetic point of view
which includes either familiar or sporadic forms
[5]. Familiar forms are more frequent in the male Diagnosis
sex, while sporadic forms are equally distributed
in both sexes. The most common form of inheri- ECG
tance is autosomal dominant compared to either ECG is often abnormal in patients with LVNC,
X-linked or autosomal recessive forms [6]. but changes of the ECG are not enough spe-
So far, have been reported mutation in several cific to allow an accurate screening of the pop-
genes in patients with LVNC and the most frequent ulation. The most common changes that can be
mutations involve genes encoding: proteins of the seen include right or left bundle branch block,
cytoskeleton, sarcomere, and mitochondria [7]. fascicular block, atrial fibrillation, ventricular
tachycardia, increased left ventricular volt-
Tafazzin Mutations in the tafazzin gene (aka ages, and left axial deviation. Wolff
G4.5 or TAZ) lead to cardiolipin deficiency in the ParkinsonWhite syndrome has been described
mitochondrial membrane and are responsible for in up to 18 % of pediatric patients affected by
Barth syndrome [1, 810], an X-linked disorder LVNC [14].
which usually causes cardiomyopathy, neutrope-
nia, and skeletal myopathy. Echocardiography
Although there is no consensus on the diagnostic
Alpha-Dystrobrevin (DTNA) Alpha-dystrobrevin criteria, echocardiography still remains the main
is a cytoskeleton protein. A mutation in the gene diagnostic tool. The echocardiographic criteria
coding for alpha-dystrobrevin has been identified are illustrated in Table 13.1.
in patients with LVNC, occasionally related to The first diagnostic criteria were published by
congenital heart disease [8, 11]. Chin et al. in 1990. They defined for the first time
the ratio between X/Y 0.5 as fundamental for
Sarcomeric Protein Genes Mutations related to the diagnosis of LVNC where X is the distance
sarcomeric proteins seem to be linked to several dis- from the epicardial surface to the base of the tra-
tinct cardiomyopathies, including LVNC, with a becular recess and Y is the distance from the epi-
common molecular base. The pE96K in the tropo- cardial surface to peak of trabeculation. These
nin T gene is more frequent, and it was described criteria, based on the observation of eight
for the first time in a family with LVNC and with patients, can be applied only at the level of left
reduced ejection fraction [12]. The E101K mutation ventricular apex on the subxiphoid or apical four-
in the alpha-cardiac actin gene (ACTC) has been chamber views at end diastole [5].
described in families with LVNC, septal defects, Afterwards, Jenni et al. [15] in 2001 pro-
and apical hypertrophic cardiomyopathy [13]. posed the following criteria that can be applied

ERRNVPHGLFRVRUJ
148 A. Maolo and S. Masiero

Table 13.1 Diagnostic criteria for LVNC


Chin et al. criteria 1990 Jenny et al. Criteria 1999 Stllberger et al. 2002
Views: parasternal short axis Views: parasternal short axis and apical Views: apical four chambers
and apical
Measured in end diastole Measured in end systole
LVNC is defined by a ratio of A two-layer structure, with a thin compacted More than three trabeculations
X/Y 0.5 layer and a thick non-compacted layer protruding from the left
X = distance from the epicardial measured in end systole at the parasternal ventricular wall, apically to the
surface to the trough of the short-axis views papillary muscles, visible in a
trabecular recess LVNC is defined by a ratio of N/C > 2 where single image plane
Y = distance from the epicardial N = non-compacted layer of the myocardium Intertrabecular spaces perfused
surface to peak of trabeculation C = compacted layer of the myocardium from the ventricular cavity,
Absence of coexisting cardiac structural visualized on color Doppler
abnormalities imaging
Numerous, excessively prominent
trabeculations and deep intratrabecular
recesses
Recesses supplied by intraventricular blood
on color Doppler

only in patients without coexisting cardiac CMR features in seven patients with LVNC with
abnormalities: 170 healthy volunteers, athletes, or patients with
dilated or hypertrophic cardiomyopathy, hyper-
1. A maximum ratio of non-compacted to com- tensive heart disease, or aortic stenosis. They
pacted myocardium >2:1 at end systole in the found that the best distinguishing feature for
parasternal short-axis view LVNC was a maximum ratio in diastole of non-
2. Color Doppler evidence of flow within the compacted to compacted myocardial thickness of
deep intertrabecular recesses >2.3 as assessed in three long-axis views (sensi-
tivity 86 % and specificity 99 %). In another
Stllberger et al. [16] proposed different crite- study, Jacquier [18] evaluated the diagnosis of
ria to diagnose LVNC based on the hypertrabecu- LVNC based on the ventricular mass: a volume
lation of the myocardial tissue. of the non-compacted tissue >20 % of the total
myocardial tissue is a criterion to diagnose
1. More than three trabeculations protruding from LVNC. Another important aspect related to CMR
the left ventricular wall, apically to the papil- is the possibility to use gadolinium which per-
lary muscles, visible in a single image plane mits to identify delayed enhancement areas
2. Intertrabecular spaces perfused from the ven- which are expression of myocardial fibrosis. The
tricular cavity, visualized on color Doppler number and the distribution of these areas seem
imaging to be related to the severity of the disease.
Furthermore, it is not rare to find delayed
Cardiovascular Magnetic enhancement areas far from the trabeculae, which
Resonance (CMR) may suggest that LVNC is a widespread cardio-
The importance of CMR as a diagnostic tool for myopathy involving all the myocardial tissue.
LVNC is increasing in the last few years. CMR
offers a good spatial resolution of the left
ventricular apex and lateral wall, and it permits Prognosis
better visualization of trabeculations and recesses.
CMR criteria to diagnose LVNC are slightly LVNC is associated with high rates of morbidity
different from the above-described echocardio- and mortality in children and adults. Outcomes
graphic criteria. Petersen et al. [17] compared are more closely related to the severity of the

ERRNVPHGLFRVRUJ
13 Left Ventricular Non-compaction Cardiomyopathy 149

disease at clinical presentation than to the diagno- increased mortality, especially if they involve an
sis per se. Sudden cardiac death is relatively com- already disfunctional left ventricle [21, 22].
mon among patients with LVNC, and it causes half For this reason, only bicameral ICD should be
of all deaths that occur in the adult population. implanted in this setting because they allow a
reliable discrimination between atrial and ven-
tricular arrhythmias. Regarding CRT-D, due to
Therapy the lack of randomized studies in LVNC popula-
tion, the indications for their implantation are the
There is no specific therapy for LVNC, even same as the current guidelines [20].
because data on treatment of LVNC are limited
and there are not guidelines. Medical manage- Prevention of Thromboembolism
ment varies with the clinical manifestations: Patients with LVNC with or without atrial fibril-
heart failure, the presence or absence of arrhyth- lation are at high risk for thromboembolism in
mias, and perceived risk of thromboembolism. the presence of impaired left ventricular function
or symptomatic presentation [23]. Therefore,
Heart Failure anticoagulation with warfarin is recommended in
Patients with LVNC and heart failure symptoms patients with LVNC without atrial fibrillation
should be treated according to the current guide- with LVEF <40 %. Patients with LVNC and atrial
lines [19]. Patients with LVNC and heart failure fibrillation who meet standard criteria for antico-
should receive the standard medical therapy used agulation should be anticoagulated according to
in heart failure with LV dysfunction that is mainly standard guidelines.
based on ACE inhibitors, beta-blockers, miner-
alocorticoid receptor antagonist, and diuretics.
Clinical Course and Therapeutic
Arrhythmias Management of the Clinical Case
Since patients with LVNC are at risk for atrial
and ventricular arrhythmias, an annual Holter For the specific clinical case discussed above, the
monitoring to detect asymptomatic arrhyth- medical therapy for heart failure was adminis-
mias is advisable. Patients with LVNC should tered including ramipril 5 mg a day, bisoprolol
receive ICD therapy according to standard indi- 2.5 mg a day, furosemide 25 mg 2 mg twice daily,
cations for ICD therapy in patients with non- and eplerenon 25 mg once daily.
ischemic cardiomyopathy. Patients with history According to the findings described above in
of sustained ventricular tachycardia or sudden this patient, we thought that a ventricular tachy-
cardiac arrest (SCA) should receive an implant- cardia was the most probable cause of the syn-
able cardioverterdefibrillator (ICD) therapy for cope and therefore the patient was at sudden
secondary prevention of SCA. ICD implantation death danger. A bicameral endocavitary ICD was
for primary prevention is indicated in patients implanted. The classical ICD was preferred
with LVNC with an LVEF 35 % and NYHA because of the possibility in upgrading to CRT-D
class II to III HF [20]. The efficacy of receiv- in case of a worsening of the cardiomyopathy
ing an ICD for the prevention of SCA is not despite an optimal medical therapy.
well proved because in patients with LVNC,
the arrhythmic substrate is increased due to the
dysfunction of the microvasculature and is not
confined to the areas with trabeculations, but References
often involves all the endocardium. Therefore,
1. Pignatelli RH, McMahon CJ, Dreyer WJ et al (2003)
the high prevalence of supraventricular arrhyth- Clinical characterization of left ventricular noncom-
mias in these patients implies a high percent- paction in children: a relatively common form of car-
age of inappropriate shocks, which can lead to diomyopathy. Circulation 108:2672

ERRNVPHGLFRVRUJ
150 A. Maolo and S. Masiero

2. Stanton C, Bruce C, Connolly H et al (2009) Isolated ciation with additional cardiac abnormalities and
left ventricular noncompaction syndrome. Am J neuromuscular disorders. Am J Cardiol 90:899
Cardiol 104:1135 17. Petersen SE, Selvanayagam JB, Wiesmann F, Robson
3. Kovacevic-Preradovic T, Jenni R, Oechslin EN et al MD, Francis JM, Anderson RH, Watkins H, Neubauer
(2009) Isolated left ventricular noncompaction as a S (2005) Left ventricular non-compaction: insights
cause for heart failure and heart transplantation: a from cardiovascular magnetic resonance imaging. J
single center experience. Cardiology 112:158 Am Coll Cardiol 46:101105
4. Patrianakos AP, Parthenakis FI, Nyktari EG, Vardas 18. Jacquier A, Thuny F, Jop B, Giorgi R, Cohen F,
PE (2008) Noncompaction myocardium imaging with Gaubert JY, Vidal V, Bartoli JM, Habib G, Moulin G
multiple echocardiographic modalities. (2010) Measurement of trabeculated left ventricular
Echocardiography 25:898 mass using cardiac magnetic resonance imaging in the
5. Chin TK, Perloff JK, Williams RG, Jue K, Mohrmann diagnosis of left ventricular noncompaction. Eur
R (1990) Isolated noncompaction of left ventricular Heart J 31:10981104
myocardium. A study of eight cases. Circulation 19. McMurray JJ, Adamopoulos S, Anker SD, Auricchio
82:507513 A, Bhm M, Dickstein K, Falk V, Filippatos G,
6. Bleyl SB, Mumford BR, Brown-Harrison MC, Fonseca C, Gomez-Sanchez MA, Jaarsma T, Kber L,
Pagotto LT, Carey JC, Pysher TJ, Ward K, Chin TK Lip GY, Maggioni AP, Parkhomenko A, Pieske BM,
(1997) Xq28-linked noncompaction of the left ven- Popescu BA, Rnnevik PK, Rutten FH, Schwitter J,
tricular myocardium: prenatal diagnosis and Seferovic P, Stepinska J, Trindade PT, Voors AA,
pathologic analysis of affected individuals. Am J Med Zannad F, Zeiher A; Task Force for the Diagnosis and
Genet 72:257265 Treatment of Acute and Chronic Heart Failure 2012 of
7. Oechslin E, Jenni R (2011) Left ventricular non- the European Society of Cardiology, Bax JJ,
compaction revisited: a distinct phenotype with Baumgartner H, Ceconi C, Dean V, Deaton C, Fagard
genetic heterogeneity? Eur Heart J 32:14461456 R, Funck-Brentano C, Hasdai D, Hoes A, Kirchhof P,
8. Ichida F, Tsubata S, Bowles KR et al (2001) Novel Knuuti J, Kolh P, McDonagh T, Moulin C, Popescu
gene mutations in patients with left ventricular non- BA, Reiner Z, Sechtem U, Sirnes PA, Tendera M,
compaction or Barth syndrome. Circulation 103:1256 Torbicki A, Vahanian A, Windecker S, McDonagh T,
9. Bleyl SB, Mumford BR, Thompson V et al (1997) Sechtem U, Bonet LA, Avraamides P, Ben Lamin HA,
Neonatal, lethal noncompaction of the left ventricular Brignole M, Coca A, Cowburn P, Dargie H, Elliott P,
myocardium is allelic with Barth syndrome. Am J Flachskampf FA, Guida GF, Hardman S, Iung B,
Hum Genet 61:868 Merkely B, Mueller C, Nanas JN, Nielsen OW, Orn S,
10. Probst S, Oechslin E, Schuler P et al (2011) Sarcomere Parissis JT, Ponikowski P; ESC Committee for
gene mutations in isolated left ventricular noncom- Practice Guidelines (2012) ESC guidelines for the
paction cardiomyopathy do not predict clinical phe- diagnosis and treatment of acute and chronic heart
notype. Circ Cardiovasc Genet 4:367 failure 2012: the Task Force for the Diagnosis and
11. Xing Y, Ichida F, Matsuoka T et al (2006) Genetic Treatment of Acute and Chronic Heart Failure 2012 of
analysis in patients with left ventricular noncompac- the European Society of Cardiology. Developed in
tion and evidence for genetic heterogeneity. Mol collaboration with the Heart Failure Association
Genet Metab 88:71 (HFA) of the ESC. Eur J Heart Fail 14:803869
12. Jenni R, Wyss CA, Oechslin EN, Kaufmann PA 20. Brignole M, Auricchio A, Baron-Esquivias G,
(2002) Isolated ventricular noncompaction is associ- Bordachar P, Boriani G, Breithardt OA, Cleland J,
ated with coronary microcirculatory dysfunction. J Deharo JC, Delgado V, Elliott PM, Gorenek B, Israel
Am Coll Cardiol 39:450454 CW, Leclercq C, Linde C, Mont L, Padeletti L, Sutton
13. Monserrat L, Hermida-Prieto M, Fernandez X et al R, Vardas PE; ESC Committee for Practice Guidelines
(2007) Mutation in the alpha-cardiac actin gene asso- (CPG), Zamorano JL, Achenbach S, Baumgartner H,
ciated with apical hypertrophic cardiomyopathy, left Bax JJ, Bueno H, Dean V, Deaton C, Erol C, Fagard
ventricular non-compaction, and septal defects. Eur R, Ferrari R, Hasdai D, Hoes AW, Kirchhof P, Knuuti
Heart J 28:1953 J, Kolh P, Lancellotti P, Linhart A, Nihoyannopoulos
14. Stollberger C, Finsterer J, Blazek G (2002) Left ven- P, Piepoli MF, Ponikowski P, Sirnes PA, Tamargo JL,
tricular hypertrabeculation/noncompaction and asso- Tendera M, Torbicki A, Wijns W, Windecker S;
ciation with additional cardiac abnormalities and Document Reviewers, Kirchhof P, Blomstrom-
neuromuscular disorders. Am J Cardiol 90:899902 Lundqvist C, Badano LP, Aliyev F, Bnsch D,
15. Jenni R, Oechslin E, Schneider J et al (2001) Baumgartner H, Bsata W, Buser P, Charron P, Daubert
Echocardiographic and pathoanatomical characteris- JC, Dobreanu D, Faerestrand S, Hasdai D, Hoes AW,
tics of isolated left ventricular non-compaction: a step Le Heuzey JY, Mavrakis H, McDonagh T, Merino JL,
towards classification as a distinct cardiomyopathy. Nawar MM, Nielsen JC, Pieske B, Poposka L,
Heart 86:666 Ruschitzka F, Tendera M, Van Gelder IC, Wilson CM
16. Stllberger C, Finsterer J, Blazek G (2002) Left ven- (2013) 2013 ESC Guidelines on cardiac pacing and
tricular hypertrabeculation/noncompaction and asso- cardiac resynchronization therapy: The Task Force on

ERRNVPHGLFRVRUJ
13 Left Ventricular Non-compaction Cardiomyopathy 151

cardiac pacing and resynchronization therapy of the 22. Kobza R, Jenni R, Erne P, Oechslin E, Duru F (2008)
European Society of Cardiology (ESC). Europace Implantable cardioverter-defibrillators in patients
15:10701118 with left ventricular noncompaction. Pacing Clin
21. Sweeney MO, Sherfesee L, DeGroot PJ, Wathen MS, Electrophysiol 31:461467
Wilkoff BL (2010) Differences in effects of electrical 23. Pitta S, Thatai D, Afonso L (2007) Thromboembolic
therapy type for ventricular arrhythmias on mortality complications of left ventricular noncompaction: case
in implantable cardioverter-defibrillator patients. report and brief review of the literature. J Clin
Heart Rhythm 7:353360 Ultrasound 35:465

ERRNVPHGLFRVRUJ
Arrhythmogenic Right Ventricular
Cardiomyopathy 14
Michela Brambatti and Matilda Shkoza

14.1 Case Report In 2002, one episode of wide complex tachy-


cardia (left bundle branch block morphology
and top axis at 200 bpm) resolved with precor-
dial thump.
A 54-year-old man with a long history of In 2012, the patient was hospitalized in a
brief episodes of palpitations was referred spoke hospital for frequent non-sustained ven-
by a spoke hospital to our cardiology tricular arrhythmias. An echocardiogram
department due to two episodes of hemo- revealed normal left ventricle and moderate
dynamically tolerated wide complex tachy- dilatation of right ventricle with hypokinesis
cardia treated with amiodarone infusion. in the mid- and apical segments. The coronary
Two days before, the patients complaint of angiogram showed noncritical stenosis. He
palpitations occurred while he was underwent cardiac magnetic resonance (MRI)
running. that was not conclusive for arrhythmogenic
right ventricular dysplasia. The patient was
recommended to stop physical activity and
was discharged on sotalol 80 mg t.i.d.
Medical History and Cardiovascular Over 3 years, the patient was stable and
Risk Factors asymptomatic and refused any further cardi-
ologist follow-up.
Cardiovascular risk factors: systemic arterial Allergies: none.
hypertension.
Family history: one brother died for sudden
cardiac death (SCD) at age 23. Medications
At age 23, episodes of exercise-induced palpi-
tations, of variable duration (minutes up to Sotalol 80 mg t.i.d
1 h).

Vital Signs
M. Brambatti (*) M. Shkoza
Clinica di Cardiologia e Aritmologia, Universit
Politecnica delle Marche, Temperature: 36.3 C
Via Conca, 71, Ancona 60126, Italy Heart rate: 60 bpm
e-mail: michelabrambatti@gmail.com; matilda_ Arterial blood pressure: 130/70 mmHg
shkoza@libero.it

Springer International Publishing Switzerland 2015 153


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_14

ERRNVPHGLFRVRUJ
154 M. Brambatti and M. Shkoza

Respiratory rate: 17 breaths/min observed. The left ventricle was mildly enlarged
Oxygen saturation: 99 % with an ejection fraction of 50 % without regional
wall motion abnormalities.
In order to better evaluate the right ventricular
Physical Examination size, motility, and ejection fraction, the patient
underwent cardiac MRI that revealed right ven-
General appearance: well developed, well tricular dilatation with wall thickening and mod-
nourished, alert, and cooperative erate dyskinesia in the mid- and apical segments.
Lungs: clear to auscultation bilaterally Right ventricular ejection fraction was deeply
Heart: regular rate and rhythm without mur- reduced, nearly 35 %. Left ventricle was mildly
murs, gallops, or rubs dilated with normal ejection fraction (Fig. 14.2).
Extremities: no peripheral edema, cyanosis or Unfortunately, the patient refused gadolinium
pallor. Warm and well perfused injection.
Abdomen: Nontender, nondistended, normo-
active bowel sounds throughout, no hepato-
splenomegaly, and no bruits Clinical Course and Therapeutic
Management

Laboratory Tests These imaging and clinical findings were strongly


suggestive for ARVC.
Tests were within normal limits (hemoglobin According to the last AHA/ACC/ESC guide-
14 g/dl, white blood cells 7,320/mmc, creatinine lines [1], taking into account the history of wide
0.88 mg/dl, potassium 4.3 mEq/l, sodium complex tachycardia suggestive for hemodynam-
140 mEq/l, magnesium 1.7 mg/dl). ically stable sustained VTs and severe right ven-
tricle dysfunction, the patient was judged to be at
moderate risk for SCD and underwent bicameral
Instrumental Examination implantable cardioverter defibrillator (ICD)
implant without major and minor complications.
The patient underwent electrocardiogram (ECG) The decision to implant a bicameral ICD was
and echocardiogram to evaluate the right and left based on the presence of first-degree AV block,
ventricle function. anterior fascicular block, and incomplete right
Surface 12-lead ECG revealed sinus rhythm ventricular block, so the patient was judged to be
(heart rate 56 bpm), first-degree atrioventricular at risk of developing high-degree AV block in the
block, anterior fascicular block, incomplete right future. Sotalol was suspended and QTc interval
ventricular block, the characteristic epsilon waves became normal (440 ms). Therapy with a
in leads V1V4, and also inverted T-waves in -blocker was administered because of the recur-
inferior lead and leads V2V6, with slightly pro- rent sustained VTs. Also, a genetic test for
longed QT interval (corrected QT QTc = 463 ms). ARVC-related mutations was performed.
The right bundle was very abnormal with frac- On the 9th day, the patient was discharged on
tionation throughout the right bundle in keeping metoprolol tartrate 50 mg daily. Patient was
with a cardiomyopathic process (Fig. 14.1). advised to avoid competitive sport.
Echocardiogram revealed right ventricular All the family members were recommended
enlargement with hypokinesis in the mid- and to undergo cardiologic visit, ECG and echocar-
apical segments and mild tricuspid regurgitation. diography evaluation, and eventually genetic
Also, right outflow dilatation (23 mm/m2) with screening once the mutation is identified in the
normal systolic pulmonary pressure was patient.

ERRNVPHGLFRVRUJ
14 Arrhythmogenic Right Ventricular Cardiomyopathy 155

Fig. 14.1 ECG sinus rhythm (heart rate 56 bpm), first- inferior lead and leads V2V6, with prolonged QT
degree atrioventricular block, anterior fascicular block, interval (corrected QTQTc = 463 ms). The right bundle
incomplete right ventricular block, characteristic epsilon is very abnormal with fractionation throughout the right
waves in leads V1V4, and also inverted T-waves in bundle in keeping with a cardiomyopathic process

a b

Fig. 14.2 Two axial FIESTA images (a, diastolic, and b, systolic) clearly depict a dilation of the four cardiac cham-
bers, especially right ones. Wall of RV is prominently thinned

ERRNVPHGLFRVRUJ
156 M. Brambatti and M. Shkoza

14.2 Arrhythmogenic Right Pathogenesis


Ventricular Cardiomyopathy
The pathogenesis of ARVC is mostly related to
The arrhythmogenic right ventricular cardiomy- reduced function of desmosomes which are
opathy (ARVC), also known as arrhythmogenic intercellular adhesion complexes that provide
right ventricular dysplasia (ARVD), is a genetic mechanical connections between cardiac myo-
disorder characterized clinically by malignant cytes. In fact, it has been hypothesized that
ventricular arrhythmias and an increased risk of impaired cell adhesion may cause destruction
sudden cardiac death (SCD), especially in young and degeneration of cardiomyocytes, especially
adults and athletes [2]. when subjected to mechanical stress (e.g.,
The ARVC is characterized by loss of myo- intense physical exercise) [2]. Because of the
cytes and progressive fibroadipose replacement limited regeneration capacity of the myocar-
resulting in structural and functional changes of dium, the repair process results in fibroadipose
the right ventricle. However, considering the replacement which proceeds from the epicar-
recent data showing a frequent involvement of dium to endocardium. These process is com-
the left ventricle, the ARVC is currently consid- pletely absent at birth and likely begins during
ered as a genetic disease of both ventricles and the puberty [2].
most properly called arrhythmogenic cardiomy- Anatomical fibrofatty replacement results in
opathy [2]. the free wall weakness and progressive ventricu-
The adult prevalence in general population is lar dilatation with systolic dysfunction and aneu-
about 1: 1,0005,000 and is considered to be rysm formation in the thinnest portions of the
more common in individuals of Italian and Greek right ventricle, the so-called triangle of dysplasia
origin; however, the real prevalence in general (the apex, the inflow tract, and the outflow tract)
population is likely to be underestimated [3]. [5]. Instead, fibrofatty replacement of the left
ventricle generally involves the posterolateral
wall, relatively thin, and rarely the interventricu-
Genetics lar septum [2].

Data from genetic studies suggest that 3050 %


of cases are familial. The most common pattern Clinical Manifestations
of inheritance is an autosomal dominant form
with incomplete penetrance and variable expres- The clinical presentation is more frequent
sivity. The autosomal recessive inheritance form between the second and the fourth decades.
is less common and associated with cutaneous Because of genetic transmission with associated
manifestations and wooly hair (Naxos disease, reduced penetrance, the severity of the clinical
Carvajal syndrome). phenotype is somewhat variable.
To date, eight genes involved in the patho- In the natural history of ARVC, four stages are
genesis of CVAD have been identified; these identified: (1) concealed phase, characterized
genes encode for the desmosomal proteins by the absence of symptoms and minor structural
[JUP (plakoglobin), DSP (desmoplachina), abnormalities (the SCD can be the first and the
Pkp2 (placofilina2), Gene (desmoglein-2), only manifestation of the disease); (2) overt
Dsc2 (desmocollina-2)], the transforming electrical instability, characterized by ventricu-
growth factor (TGF 3), the transmembrane lar arrhythmias and morphofunctional cardiac
proteins (TMEM43), and the ryanodine receptor abnormalities; (3) right ventricular failure, with
(RYR2)] [4]. severe systolic dysfunction of the right ventricle

ERRNVPHGLFRVRUJ
14 Arrhythmogenic Right Ventricular Cardiomyopathy 157

and initial or absent left ventricular abnormali- Classic: primarily affects the right ventricle
ties; and (4) biventricular failure, with severe and, only in advanced stages, the left ventricle
systolic dysfunction of both ventricles similar to (39 %).
a cardiomyopathy [2]. Dominant left: characterized by early and
severe involvement of the left ventricle and
Ventricular/Supraventricular relatively mild disease of the right ventricle. It
Tachyarrhythmias and Sudden is very insidious variant as fibroadipose
Cardiac Death replacement involves initially only the epicar-
VTs in patients with ARVC range from single dium of the left ventricle without causing wall
extra beats to complex VT, symptomatic and not, motion abnormalities. The use of MRI con-
and the frequency appears to be proportional to trast medium allows the identification of non-
the severity of the disease. The most common VT transmural scar.
is generally monomorphic with origin from the Biventricular: characterized by a parallel
right ventricle and left bundle branch block mor- involvement of both ventricles.
phology. The arrhythmic episodes may origin
from the apex, the inflow tract, or the outflow
tract; when the site of origin is the right outflow Diagnosis
tract, a differential diagnosis with idiopathic VT
is required [2]. The diagnosis of ARVC is complex and
Unfortunately, the SCD may be the first mani- requires a high degree of clinical suspicion. To
festation of the disease. The estimated mortality date, no single diagnostic test is enough sensi-
rate for SCD varies from 0.08 to 9 % per year [1]. tive and specific to confirm or rule out the dis-
Exercise, especially the endurance sport, is ease; thus multiple parameters need to be
considered a precipitating factor for arrhythmias considered.
in patients with ARVC. This trigger effect is The Task Force of the Working Group on
likely related to the increased right ventricular Myocardial and Pericardial Disease of the
stimulation by catecholamine exposure; in addi- European Society of Cardiology has developed
tion, data from literature suggest that the exer- the ARVC diagnostic criteria that take into
cise itself contributes to the right ventricle account structural, histologic, arrhythmic, and
dilatation and consequently to the disease pro- genetic features. The original version of 1994 [8]
gression [2]. that the revised version of 2010 [9] includes
Supraventricular tachyarrhythmia (SVT), major and minor criteria collected in six main
such as atrial fibrillation, atrial tachycardia, and categories (Table 14.1). Based on this classifica-
atrial flutter, in association with ventricular tion, definite diagnosis of ARVC requires two
arrhythmias, is present in up to 25 % of patients major criteria or one major and two minor or
with ARVC; less commonly, SVT may be the four minor criteria from different categories
only arrhythmia present [6]. (Table 14.1).
In the suspicion of ARVC, as a general
Left Ventricular Involvement approach, it is recommended to go through a
The wider use of cardiac MRI has allowed to careful personal and family medical history, a
appreciate a more common involvement of the 12-lead ECG, and a transthoracic echocardiogra-
left ventricle in ARVC [7]. phy in all patients. Additional tests should be per-
In a study of 200 patients undergoing MRI, formed according to the clinical scenario or when
three patterns of disease expression were the results of initial tests are not conclusive.
identified: Recommended tests are as follows:

ERRNVPHGLFRVRUJ
158 M. Brambatti and M. Shkoza

Table 14.1 Task Force Criteria [9]


2010 Task Force Criteria
Definite = 2 major or 1 major + 2 minor or 4 minor from different categories
Borderline = 1 major + 1 minor or 3 minor
Possible = 1 major or 2 minor
1. Global/regional dysfunction/structural alterations
Major By 2D echo:
Regional RV akinesia, dyskinesia, or aneurysm and 1 of the following (end diastole):
PLAX RVOT 32 mm (PLAX/BSA 19 mm/m2)
PSAX RVOT 36 mm (PSAX/BSA 21 mm/m2)
Fractional area change 33 %
By MRI:
Regional RV akinesia or dyskinesia or dyssynchronous RV contraction and 1 of the following:
RVEDV/BSA 110 mL/m2 (male) or 100 mL/m2 (female)
RV ejection fraction 40 %
By RV angiography:
Regional RV akinesia, dyskinesia, or aneurysm
Minor By 2D echo:
Regional RV akinesia or dyskinesia and 1 of the following (end diastole):
PLAX RVOT 29 to <32 mm (PLAX/BSA 16 to <19 mm/m2)
PSAX RVOT 32 to <36 mm (PSAX/BSA 18 to <21 mm/m2)
Fractional area change >33 to 40 %
By MRI:
Regional RV akinesia or dyskinesia or dyssynchronous RV contraction and 1 of the following:
Ratio of RVEDV to BSA 100 to <110 mL/m2 (male) or 90 to <100 mL/m2 (fem)
RV EF >40 to 45 %
2. Tissue characterization of wall
Major Residual myocytes <60 % by morphometric analysis (or <50 % if estimated), w/ fibrosis replacement of
RV free wall myocardium in 1 sample, w/ or w/o fatty replacement of tissue on endomyocardial biopsy
Minor Residual myocytes 6075 % by morphometric analysis (or 5060 % if est.) w/ fibrous replacement of the
RV free wall in 1 sample, w/ or w/o fatty replacement of tissue on endomyocardial biopsy
3. Repolarization abnormalities
Major TWI (V1, V2, V3) or beyond; >14 years of age; in the absence of complete RBBB QRS 120 ms
Minor TWI in V1 and V2; >14 years of age; in the absence of complete RBBB or in V4, V5, or V6
TWI in V1V4; >14 years of age; in the presence of complete RBBB
4. Depolarization conduction abnormalities
Major Epsilon wave in right precordial leads (V1V3)
Minor Late potentials by SAECG in 1 of 3 parameters in the absence of QRS duration of 110 ms on ECG:
Filtered QRS duration (fQRS) 114 ms
Duration of terminal QRS <40 V 38 ms
Root mean square voltage of terminal 40 ms 20 V
Terminal activation 55 ms measured from nadir of S-wave to end of QRS, including R, in V1, V2, or
V3, in the absence of complete RBBB
5. Arrhythmias
Major NSVT or sustained VT, LBBB morphology with superior axis
Minor NSVT or sustained VT of RV outflow configuration, LBBB with inferior or of unknown axis
>500 PVCs per 24 h on Holter monitoring
6. Family history
Major ARVC/D in first-degree relative who meets Task Force Criteria
ARVC/D confirmed pathologically at autopsy or surgery in first-degree relative
Identification of the pathogenic mutation (associated or probably associated w/ ARVC/D) in the patient
under evaluation
(continued)

ERRNVPHGLFRVRUJ
14 Arrhythmogenic Right Ventricular Cardiomyopathy 159

Table 4.1 (continued)


Minor History of ARVC in first-degree relative in whom it is not possible to determine if the family member
meets Task Force Criteria
Premature SCD (<35 years of age) due to suspected ARVC/D in first-degree relative
ARVC/D confirmed pathologically or by current Task Force Criteria in second-degree relative
RVOT right ventricular outflow tract, PLAX parasternal long axis, PSAX parasternal short axis, BSA body surface area,
RV right ventricle, RVEDV right ventricle end-diastolic volume, EF ejection fraction, TWI T-wave inversion, RBBB right
bundle branch block, LBBB left bundle branch block, SAECG signal-averaged electrocardiography, NSVT non-sus-
tained ventricular tachycardia, VT ventricular tachycardia, ARVC/D arrhythmogenic ventricular cardiomyopathy/dys-
plasia, SCD sudden cardiac death, PVC premature ventricular complex

Signal-averaged ECG (SAECG) low-frequency cutoff of 0.05 Hz and a


24 h ECG Holter high-frequency cutoff greater than
Stress testing 150 HZ in adults and 250 Hz in children.
Cardiac MRI with gadolinium T-wave inversion in right precordial
Right ventriculography leads (V1, V2, V3) in the absence of
Endomyocardial biopsy complete right bundle branch block.
Electrophysiologic study and three- This ECG feature, which can be a nor-
dimensional electroanatomic mapping mal variant in children under 14 years of
Genetic testing age, is present in 87 % of subjects with
ARVC. The extension of the T-wave
inversion also correlates with the degree
of right ventricular dilatation and
Electrocardiography (ECG) arrhythmic risk. T-wave inversion in
In all patients with suspected diagnosis of ARVC, leads V5V6, instead, is an indirect sign
a 12-lead ECG is recommended. The sensitivity of left ventricular involvement.
is suboptimal, because 4050 % of patients have
a normal ECG submission. However, reflecting
the progressive nature of the disease, most
patients develop characteristic ECG abnormali- Late Potentials
ties years after the presentation, such as [10]: Signal-averaged ECG abnormalities are common
and have an excellent sensitivity and specificity
for ARVC; however, they are not predictive of
QRS prolongation, especially in lead V1 arrhythmic risk.
versus V6. Late potential abnormalities are considered as
Incomplete/complete right bundle minor criteria if they meet at least one of the fol-
branch block. lowing three conditions, without a QRS duration
Prolonged QRS terminal activation 110 ms [9]:
(55 ms measured from nadir of S-wave
to end of QRS, including R, in V1, V2, Filtered QRS duration 114 ms
or V3, in the absence of complete Terminal QRS duration <40 microV and
RBBB). 38 Ms
Epsilon waves in right precordial leads, Root mean square voltage of terminal 40 ms
particularly in V1 identified in 30 % of 20 microV
patients with ARVC. In order to detect
epsilon waves and avoid ST distortion, Echocardiography
ECG filters should be set with a A transthoracic echocardiogram should be per-
formed in all patients with the clinical suspicion

ERRNVPHGLFRVRUJ
160 M. Brambatti and M. Shkoza

of ARVC. Echocardiography is a noninvasive Electrophysiologic Testing


method and readily available and easily allows and Electroanatomic Voltage Mapping
right ventricle visualization in most patients. The Electrophysiologic testing is of limited role in the
echocardiographic findings frequently observed diagnostic workup of patients with suspect
are [9]: ARVC. However, electrophysiologic testing has
a role in the differential diagnosis between idio-
Right ventricular dilation, especially the right pathic outflow tract and the ARVC-related VT
outflow tract (RVOT) [11]. In fact, contrarily to VT in ARVC patients,
Reduction of right ventricular fractional area the idiopathic outflow tract VT is not associated
change with ECG abnormalities, is more frequent in
Regional and global wall motion abnormali- women, and is induced by isoprenaline infusion
ties of the right ventricle rather than programmed ventricular stimulation.
Morphological abnormalities of the right ven- Electroanatomic voltage mapping is an inno-
tricle (trabecular derangement, hyperreflective vative tool useful to identify and quantify areas of
moderator band, and aneurysms) fibrosis by detection of low-voltage local right
ventricle electrograms. Despite electroanatomic
Left ventricular echocardiographic abnormali- mapping increases accuracy in diagnosing, the
ties are seen less frequently. technique is operator dependent, invasive, and
costly.
Cardiovascular Magnetic Resonance
Imaging Genetic Testing
In patients with suspected ARVC and no diag- The ARVC is a genetic disease and a desmo-
nostic results with other imaging modalities, somal gene mutation identified in approximately
cardiac MRI is strongly recommended. Cardiac 3050 % of cases. However, given multiple muta-
MRI allows evaluation of right ventricle dila- tions causing the disease and the variable pene-
tion, global and regional wall motion abnor- trance, genetic testing should not be performed in
malities, intramyocardial fat, focal wall all patients suspected for ARVC. Genetic tests
thinning, and late gadolinium enhancement in are recommended for patients who meet the Task
fibrosis areas. Force Criteria [9] for a definitive or borderline
The main limitations of cardiac MRI are ARVC diagnosis and all first-degree relatives fol-
related to the limited experience of the centers lowing the identification of the ARVC-causative
and significant interobserver variability resulting mutation in an index case [12].
in high rate of false positives. In addition, intra-
myocardial fat infiltration of the right ventricular Endomyocardial Biopsy
wall should be interpreted with caution, as fat Endomyocardial biopsy is an invasive procedure
infiltration is frequently in healthy subjects, espe- with poor sensitivity and specificity; thus, it is not
cially if elderly [2]. recommended for the initial diagnostic workup.
When endomyocardial biopsy is performed,
Right Ventriculography one major concern is related to the sampling
Ventriculography may be useful to assess the error. The interventricular septum, which is the
structure and function of the right ventricle in usual and low-risk biopsy site, is not generally
patients with strong suspect for ARVC and useful in ARVC patients since it is not generally
nondiagnostic results with other imaging affected by the pathology. In addition, the isto-
modalities. Regional right ventricle akinesia, fibrofatty replacement cannot be considered a
dyskinesia, or aneurysms are generally observed pathognomonic ARVC feature as it is also
[9]. However, since ventriculography is an inva- observed in healthy subjects, especially elderly.
sive method, it is rarely used. In the light of these data, in ARVC patients,

ERRNVPHGLFRVRUJ
14 Arrhythmogenic Right Ventricular Cardiomyopathy 161

endomyocardial biopsy is a class IIb recommen- diagnosis of ARVC. In contrast, -blockers are
dation in the diagnostic workup for ARVC [13]. not usually recommended in mutation carriers
who do not express the ARVC phenotype.
Antiarrhythmic medications, in association
Differential Diagnosis for ARVC with -blockers or alone, may be useful to reduce
ventricular arrhythmias frequency and symptoms
Dilated cardiomyopathy: biventricular dilated in patients with ARVC. Although there is no evi-
and congestive heart failure may be similar to dence to suggest a specific antiarrhythmic treat-
ARVC with left ventricle involvement. ment, class III drugs as amiodarone and sotalol
Uhl anomaly: total lack of right ventricular are the drugs of choice [1]. However, any antiar-
myocardium. In ARVC, the myocardium is rhythmic medication has demonstrated to reduce
not completely absent but replaced by a vari- the risk of SCD; as a result, antiarrhythmic drug
able degree of fibrosis. therapy should not be considered an equivalent
Idiopathic VT: Left bundle branch block mor- alternative to ICD implantation in ARVC patients.
phology VT in a structurally healthy heart and
absence of SCD familiarity. ICD Implantation and Arrhythmic Risk
Stratication
In patients with ARVC and documented sus-
Therapy and Prognosis tained ventricular fibrillation or VT (PV), ICD
implantation for secondary prevention is strongly
The therapeutic approach in ARVC patients is recommended. In these patients, appropriate ICD
addressed to SCD prevention and relief of symp- intervention occurs in about 10 % of cases per
toms of arrhythmias related, as well as improve- year, and the estimated survival benefit is about
ment in terms of functional capacity and survival 2530 % at 3 years follow-up [2].
in patients who develop heart failure. To achieve On the contrary, due to the relatively low prev-
these goals, available treatment options include alence of the disease and lack of randomized con-
lifestyle changes, pharmacological therapy with trolled trials, the indications for ICD implantation
-blockers, antiarrhythmic and heart failure for primary prevention are still object of intense
drugs, transcatheter ablation, ICD implantation, debate among experts.
and cardiac transplantation. Prognostic stratification and indications for
ICD implantation for primary prevention are cur-
Lifestyle Changes rently based on several arrhythmic risk factors,
Given the well-known association between mostly identified from retrospective studies or
endurance sports and five-time increased risk of autopsy data, such as unexplained syncope; pro-
SCD, patients with a definitive diagnosis of band status; young age; physical activity; family
ARVC and asymptomatic mutation carriers history for SCD, QRS, and QT dispersion; severe
should not participate in any competitive activity right ventricular dysfunction; left ventricular
[14]. No absolute contraindications in practicing involvement; and inducible VT [15]. However,
low-intensity physical activity are established, not all these risk factors should be taken into
especially for ARVC patients treated with account when an ARVC patient is evaluated for
-blockers. ICD implant.
An interesting pyramidal representation of the
-Blockers and Antiarrhythmic Therapy relationship between the arrhythmic risk and cur-
With the assumption that the adrenergic stimula- rent ICD implant indications for ARVC patients
tion during exercise triggers arrhythmias, treat- was proposed by Corrado and colleagues [4]. At
ment with -blockers currently represents a the apex of the pyramid, there are the high-risk
first-choice therapy in patients with a definitive patients who are most likely to benefit from an

ERRNVPHGLFRVRUJ
162 M. Brambatti and M. Shkoza

Table 14.2 Our approach to patients with ARVC according to the arrhythmic risk
Subgroups Risk markers Recommendations ICD indication
High risk (810 %/ Aborted SCD Avoid competitive sport Recommended
year) Hemodynamically unstable -blockers
sustained VT
Moderate risk Unexplained syncope Avoid competitive sport Consider
(12 %/year) Hemodynamically stable -blockers
sustained VT Annually FU including:
Non-sustained VT ECG
Cardiac imaging (ECHO vs.
CMR)
Holter
Exercise stress testing
Moderate risk (?%/ Severe dilatation and/or Avoid competitive sport Consider
year) dysfunction of RV, LV, or -blockers
both Annually FU including:
Early onset structurally ECG
severe disease (age < 35 Cardiac imaging (ECHO vs.
years) CMR)
Holter
Exercise stress testing
Low risk (<1 %/ Asymptomatic mutation Reduce physical exercise Not recommended
year) carriers Avoid competitive sport
ARVD patients without risk Annually FU including:
factors ECG
Cardiac imaging (ECHO vs.
CMR)
Holter
Exercise stress testing
ARVD arrhythmogenic right ventricular dysplasia, FU follow-up, CMR cardiac magnetic resonance, LV left ventricle,
RV right ventricle, SCD sudden cardiac death, VT ventricular tachycardia, ICD internal cardiac defibrillator

ICD such as those with previous heart attack, Radiofrequency Ablation


unstable hemodynamic VT, and unexplained The radiofrequency ablation is not a definitive
syncope. By contrast, at the bottom, there are therapy for ventricular arrhythmias and should
patients with definitive diagnosis of ARVC but not be considered an equivalent alternative to
without arrhythmic risk factors and patients who ICD implantation in high-risk patients for
are mutation carriers who do not express the SCD. This therapeutic approach may be appro-
ARVC phenotype. For these low-risk patients, priate in selected ARVC patients who are not
ICD implantation is not generally recommended. candidates for ICD implantation or who are ICD
Patients with intermediate risk, linked to the recipients with frequent VT episodes and ICD
detection of non-sustained VT or demonstration shocks despite antiarrhythmic therapy [1]. The
of a moderate-severe ventricular dysfunction, are ablative technique may include a multiple epicar-
represented in the middle of the pyramid. In these dial and endocardial mapping approach due to
patients, the indication to implantation of the the multifocal nature of the disease.
ICD must be evaluated taking into account both
the arrhythmic profile and the risk of potential Cardiac Transplantation
implant complications, costs, and the psychologi- In rare cases, patients with ARVC who develop
cal impact, especially for the young. According heart failure symptoms or refractory ventricular
to these stratification risks, our approach in arrhythmias may be candidates for heart
patients with ARVC is described in Table 14.2. transplantation.

ERRNVPHGLFRVRUJ
14 Arrhythmogenic Right Ventricular Cardiomyopathy 163

Conclusions novel insights into patterns of disease expression.


Circulation 115(13):17101720
ARVC is a genetic disease of the myocardium 8. McKenna WJ et al (1994) Diagnosis of arrhythmo-
with hereditary transmission in half of cases genic right ventricular dysplasia/cardiomyopathy.
and associated with an increased risk of Task Force of the Working Group Myocardial and
SCD. The diagnosis is complex and, if missed, Pericardial Disease of the European Society of
Cardiology and of the Scientific Council on
can be fatal. The therapy goal is the SCD Cardiomyopathies of the International Society and
prevention. Although the benefit of the Federation of Cardiology. Br Heart J 71(3):215218
implantation of the ICD in a patient who sur- 9. Marcus FI et al (2010) Diagnosis of arrhythmogenic
vived to a cardiac arrest is universally recog- right ventricular cardiomyopathy/dysplasia: proposed
modification of the task force criteria. Circulation
nized, the role of the ICD for primary 121(13):15331541
prevention is still being debated. 10. Marcus FI, Zareba W (2009) The electrocardiogram
in right ventricular cardiomyopathy/dysplasia. How
can the electrocardiogram assist in understanding the
pathologic and functional changes of the heart in this
References disease? J Electrocardiol 42(2):136.e15
11. ODonnell D et al (2003) Clinical and electrophysio-
1. Zipes DP et al (2006) ACC/AHA/ESC 2006 guide- logical differences between patients with arrhythmo-
lines for management of patients with ventricular genic right ventricular dysplasia and right ventricular
arrhythmias and the prevention of sudden cardiac outflow tract tachycardia. Eur Heart J 24(9):801810
death: a report of the American College of Cardiology/ 12. Ackerman MJ et al (2011) HRS/EHRA expert con-
American Heart Association Task Force and the sensus statement on the state of genetic testing for the
European Society of Cardiology Committee for channelopathies and cardiomyopathies this document
Practice Guidelines (Writing Committee to Develop was developed as a partnership between the Heart
Guidelines for Management of Patients With Rhythm Society (HRS) and the European Heart
Ventricular Arrhythmias and the Prevention of Sudden Rhythm Association (EHRA). Heart Rhythm
Cardiac Death). J Am Coll Cardiol 48(5):e247e346 8(8):13081339
2. Basso C et al (2009) Arrhythmogenic right ventricular 13. Cooper LT et al (2007) The role of endomyocardial
cardiomyopathy. Lancet 373(9671):12891300 biopsy in the management of cardiovascular disease:
3. Thiene G et al (1997) Arrhythmogenic right ventricu- a scientific statement from the American Heart
lar cardiomyopathy a still underrecognized clinic Association, the American College of Cardiology, and
entity. Trends Cardiovasc Med 7(3):8490 the European Society of Cardiology Endorsed by the
4. Corrado D et al (2011) Molecular biology and clinical Heart Failure Society of America and the Heart
management of arrhythmogenic right ventricular Failure Association of the European Society of
cardiomyopathy/dysplasia. Heart 97(7):530539 Cardiology. Eur Heart J 28(24):30763093
5. Marcus FI et al (1982) Right ventricular dysplasia: a 14. Pelliccia A et al (2006) Recommendations for partici-
report of 24 adult cases. Circulation 65(2):384398 pation in competitive sport and leisure-time physical
6. Camm CF et al (2013) Prevalence of atrial arrhyth- activity in individuals with cardiomyopathies,
mias in arrhythmogenic right ventricular dysplasia/ myocarditis and pericarditis. Eur J Cardiovasc Prev
cardiomyopathy. Heart Rhythm 10(11):16611668 Rehabil 13(6):876885
7. Sen-Chowdhry S et al (2007) Clinical and genetic 15. Basso C et al (2012) Arrhythmogenic right ventricular
characterization of families with arrhythmogenic cardiomyopathy. Circ Arrhythm Electrophysiol
right ventricular dysplasia/cardiomyopathy provides 5(6):12331246

ERRNVPHGLFRVRUJ
Part V
Valvular Heart Disease

ERRNVPHGLFRVRUJ
Aortic Stenosis
15
LorenaScappini, SimoneMaffei,
andAlessioMenditto

15.1 Case Report A more recent (4 months ago) acute coronary


syndrome NSTEMI treated by percutaneous
coronary intervention and implantation of two
A 75-year-old man was admitted to the car- DES on LAD 12 (intrastent) and on LAD 2.
diology department from the emergency A noncritical lesion on the right coronary
room for chest pain. He reported the occur- remained.
rence of constrictive chest pain after mild Peripheral artery disease (carotid plaques).
exertion; this pain did not vary with breath- Chronic kidney disease (fifth stage) on dialy-
ing, with posture, or with movement and sis (11 ml/min/1.73 m2).
was relieved with rest until disappeared Gouty arthritis.
completely.

Allergies

 edical History andCardiovascular


M No allergy is referred by the patient.
Risk Factors

Type 2 diabetes mellitus. Social History


Arterial hypertension.
Dyslipidemia. Smoked about 15 cigarettes/day for 5 years
Stable coronary artery disease treated by per- Never used illegal drugs
cutaneous coronary intervention (PTCA) with
two drug-eluting stent (DES) implantations on
the proximal middle part of the left anterior Medications
descending (LAD 12) and on the first obtuse
marginal (OM 1) (1 year ago). Pantoprazole 40 mg at 8:00 a.m., clopidogrel
75 mg at 8:00 a.m., furosemide 25 mg at
L. Scappini (*) S. Maffei A. Menditto
8:00 a.m., repaglinide 0.5 mg at 8:00 a.m. and at
Clinica di Cardiologia e Aritmologia, Universit 8:00 p.m., repaglinide 1 mg at 12:00 a.m.,
Politecnica delle Marche, ivabradine 5 mg at 8:00 a.m. and at 8:00 p.m.,
Via Conca, 71, Ancona 60126, Italy doxazosin 2 mg at 8:00 a.m. and at 8:00 p.m.,
e-mail: lorena.scappini@gmail.com; maffo-85@
hotmail.it; alessiomenditto@hotmail.it
nitroglycerin transdermal 10 mg at 8:00 a.m.,

Springer International Publishing Switzerland 2015 167


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI10.1007/978-3-319-19926-9_15

ERRNVPHGLFRVRUJ
168 L. Scappini et al.

aspirin 100 mg at 12:00 a.m., allopurinol 150 mg The routine laboratory tests were performed:
at 3:00 p.m., amlodipine 5 mg at 8:00 p.m., and
ezetimibe/simvastatin 10/20 mg at 10:00 p.m Complete blood count: normal (hemoglobin
13 g/dl)
Inflammatory markers (ESR, CRP): normal
Vital Signs Hepatic function (GOT, GPT, -GT, ALP, total
bilirubin, direct and indirect): normal
Temperature: 36 C Impaired renal function (creatinine 6.61 mg/
Heart rate: 53 bpm dl, BUN 146 mg/dl)
Blood pressure: 140/80 mmHg Electrolytes (Na+, K+, Ca++, Mg++, Cl):
Respiratory rate: 16/min normal
Oxygen saturation while breathing in ambient Fasting blood glucose: 55 mg/dl
air: 97 % Myocardial necrosis markers (CK-MB and
Hs-TnI): negative in two different samples

Physical Examination A routine rest 12-lead EKG showed sinus bra-


dycardia (heart rate was 53 bpm) and normal
General: alert, awake, and oriented atrioventricular and intraventricular conduction.
Head, eyes, ears, nose, and throat: normoce- Left ventricular hypertrophy was observed. ST
phalic, atraumatic, mucous membranes moist, segment depression was present in I, aVL, and
extraocular muscles intact, pupils equally V5V6 leads (Fig. 15.1).
round and reactive to light and accommoda-
tion bilaterally, bilateral tympanic membrane
intact, bilateral sclera anicteric, and no con-  hat Are thePossible Causes
W
junctival injection ofChest Pain inThis Patient?
Neck: supple, no jugular venous distention, no
lymphadenopathy, and bilateral carotid murmur
Cardiovascular: regular rhythm, S1 normal, Acute coronary syndrome (unstable
S2 low intensity, and aortic diamond-shaped angina)
(crescendo/decrescendo) systolic murmur 3/6 Valvular disease (aortic stenosis)
with radiation to carotid vessels Acute pericardial disease
Lungs: no rales, rhonchi, or wheezes, no Chest wall pain
egophony, no alterations in tactile fremitus, Lung diseases
and normal percussion
Abdomen: overweight, no pulsatile masses,
normal bowel sounds in all four quadrants, no
high-pitched or tinkling sounds, resonant to The characteristics of chest pain were sug-
percussion, soft, non-distended/non-tender, gestive for typical angina, the patient was apy-
no rebound or guarding, no costovertebral retic, the inflammatory biomarkers were
angle tenderness, and no hepatosplenomegaly negative, there werent clinical and instrumental
Extremities: no cyanosis or clubbing, and no signs of respiratory disease, and therefore, acute
edema pericardial disease, chest wall pain, and lung
Neurologic: cranial nerves I through XII diseases from differential diagnosis were
intact, and no focal deficit excluded. The presence of systolic heart mur-
Psychiatric: normal affect, no hallucinations, mur and of STT downslope on rest ECG sup-
and normal speech ported the hypothesis of ischemic heart disease
Skin: intact, no rashes, and no lesion or of aortic stenosis.

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 169

Fig. 15.1 ECG shows ST


segment depression in I, aVL
and V5-V6 leads

Fig. 15.2 Echocardiography. Concentric left ventricular hypertrophy (left side). Severe aortic valve stenosis with high
trans-aortic valve gradient (right side)

Echocardiography severe stenosis (peak velocity 4.3 m/s, medium


gradient 36 mmHg, calculated aortic valve area
Echocardiography was performed to better 0.8 cm2, index calculated aortic valve area
evaluate the aortic area (Fig. 15.2) and showed 0.43 cm2/m2). Mild mitral regurgitation. Mild
the following results: Concentric left ventricu- tricuspid regurgitation with high pulmonary
lar hypertrophy with normal left ventricle arterial pressure (40 mmHg).
global and regional systolic function (EF 57 %).
Severe dilatation of the left atrium (index vol-
ume 66 ml/m2). Pseudo-normal diastolic func- Final Diagnosis
tion. Normal right ventricle size and systolic
function (TAPSE 17 mm). Mild dilatation of The final diagnosis is severe aortic stenosis with
the right atrium. No pericardial effusion. effort angina in patient with known coronary
Severely calcified tricuspid aortic valve with artery disease.

ERRNVPHGLFRVRUJ
170 L. Scappini et al.

a b

Fig. 15.3 Coronary angiography: right coronary (a); left coronary (b)

Symptomatic severe aortic stenosis has sur- Calcific aortic valve disease is the most com-
gery correction indication; although the last mon cause of AS among adults in the Western
PTCA was done in the previous 4 months, an world [2]. Prevalence increases with age [1, 3],
invasive coronary angiography was performed and severe AS, if left untreated, is fatal a few
because the angina symptoms show a LAD 70 % years from symptom onset [4].
diameter stenosis (intrastent), severe stenosis of
the first diagonal branch (D1) and second diago-
nal branch (D2), and a severe stenosis of the pos- Etiology
terior descending artery (PDA) (Fig. 15.3a, b).
The patient was admitted to the cardio-surgery AS is a hereditary or acquired disease that causes
department to replace the aortic valve and for the a progressive obstruction to left ventricular
heart to be revascularized through coronary outflow. Obstruction may be valvular, subvalvu-
artery bypass graft (CABG). A bioprosthetic aor- lar, or supravalvular.
tic valve was implanted, and an artery graft with Valvular stenosis is the most common cause of
the left internal mammary artery to LAD and a aortic obstruction. Its etiology may be age
saphenous vein graft to PDA were performed. related: patients younger than 20 usually have a
congenital abnormality.
Patients aged 4060 usually have a calcified
15.2 Aortic Stenosis bicuspid valve or a valve previously damaged by
rheumatic heart disease.
Epidemiology The most common cause of AS in patients 7080
years old is degenerative AS; this is an active pro-
Valvular aortic stenosis (AS) is the second most cess characterized by lipid accumulation, inflam-
prevalent adult valve disease in the USA. The mation, and calcification of aortic valve cusps that
prevalence of AS increases with age: it occurs in provokes a valve degeneration [57].
4 % of patients older than 75 years [1] and Subvalvular AS occurs in less than 10 % of all
approximately in 2 % of population aged 65 patients with obstruction of left ventricular outflow
years old. and is frequently associated with aortic regurgitation

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 171

due to valve damage. This kind of obstruction can point, both the high afterload and the intrinsic
be due to a subvalvular ridge or diffuse tunnellike myocardial disease significantly increase wall
narrowing of the entire outflow tract. If severe stress, and a vicious cycle of deterioration in ven-
hypertrophy of the left ventricle is present, addi- tricular function ensues.
tional dynamic outflow obstruction from systolic A death spiral may occur: if systemic hypo-
anterior motion of the mitral valve may occur. tension will occur (due to either drugs or a vaso-
Supravalvular AS is an uncommon (less than vagal reaction), perfusion of the coronary arteries
1 % of patients with aortic stenosis) congenital may decrease; this increases the myocardial oxy-
abnormality, consisting of narrowing of the gen supplydemand mismatch and results in
ascending aorta (immediately over the aortic myocardial ischemia. The myocardial ischemia,
valve) secondary to a single stenosis or a long in turn, reduces forward cardiac output, and aor-
tubular lesion of the entire ascending aorta. This tic diastolic pressure decreases, further decreas-
type of stenosis is often associated with some ing coronary perfusion pressure. Unless
congenital abnormalities like coronary dysplasia, immediate steps are taken to increase perfusion
elfin facies, mental retardation, coarctation of the pressure, progressive hemodynamic deterioration
aorta, hypercalcemia, peripheral pulmonary ste- and death may occur.
nosis, and Williams syndrome.

Clinical Manifestation
Physiopathology
The clinical presentation of AS varies. Many
AS represents a continuum disease: (1) an increase patients will remain asymptomatic for decades.
in afterload, (2) a decrease in systemic and coro- The diagnosis of AS is usually made in these
nary blood flow from obstruction, and (3) progres- patients on the basis of a systolic murmur on aus-
sive hypertrophy. These mechanisms result in the cultation and confirmed by echocardiography.
classic symptom triad of dyspnea, angina, and syn- Symptoms usually consist of one or more of
cope. Left ventricular cavity size and systolic func- the classic triad of exertional dyspnea, angina,
tion are initially maintained; in fact, the increase in and syncope. The cause of syncope may include
left ventricular wall thickness acts as a compensa- ventricular arrhythmias, a sudden decrease in
tory mechanism to normalize wall stress. So at the systemic flow caused by the obstruction, or
beginning, hypertrophy is a beneficial adaptation. abnormal vasodepressor reflexes caused by the
However, this phenomenon may cause a high left ventricular intracavitary pressure. Albeit
reduced coronary flow reserve and oxygen sup- rare, sudden death may be the initial manifesta-
plydemand mismatch [8]. Hypertrophic hearts tion of aortic stenosis.
are more sensitive to diffuse subendocardial isch- Uncommonly, patients with end-stage AS and
emic injury, which may result in both systolic concomitant left ventricular dysfunction present
and diastolic dysfunctions. The latter occurs from with anasarca and cardiac cachexia.
prolonged ventricular relaxation and decreased The development of symptoms is a critical
compliance and is caused by myocardial isch- point in the natural history of patients with AS. In
emia, a thick noncompliant left ventricle, and fact, after symptom onset, the average survival is
increased afterload [9, 10]. 23 years.
The so-called reversible phase is characterized by
an obstruction progression to a higher level that pro-
vokes a decrease of left ventricle systolic function. Physical Examination
If afterload excess was continued, myocyte
degeneration and fibrosis would occur (irrevers- The physical examination of a patient with AS
ible left ventricular systolic dysfunction). At this reveals classic, characteristic findings that are

ERRNVPHGLFRVRUJ
172 L. Scappini et al.

important for diagnosis and estimation of sever- T wave inversion, or a left bundle block can be
ity of this valvular disease. present.
The characteristics of the carotid upstroke are
a reliable indicator of the severity of AS in most
patients. There will be both parvus (small pulse) Chest Radiography
and tardus (slow upstroke) in patients with severe
AS. These typical findings couldnt be present on Chest radiography may demonstrate enlargement
carotid palpation in older patients with a of the ascending aorta and a left ventricular pre-
noncompliant vasculature. When there is a thrill dominance. Aortic calcification is frequently seen
felt in the right carotid artery but not in the left on lateral chest radiographs (i.e., porcelain aorta).
carotid artery, the diagnosis of supravalvular AS This find implies high risk for operation [11].
should be suspected. This phenomenon is due to
the high-velocity jet of blood directed toward the
innominate artery. How toAssess Aortic Stenosis
A sustained bifid left ventricular impulse indi-
cates concomitant left ventricular hypertrophy. A Echocardiography
systolic thrill, if present, indicates the presence of In addition to physical examination, electrocardi-
severe AS (mean gradient >50 mmHg). ography, and chest radiography, various imaging
Accurate auscultation is an essential compo- modalities may be used to confirm the presence
nent of evaluating patients with AS. An absent of aortic stenosis.
aortic component of the second heart sound indi- Transthoracic echocardiography is the gold
cates severe calcification of the aortic valve. A standard modality for initial diagnosis and subse-
delayed murmur apex is also present in severe AS. quent evaluation of aortic stenosis; with this
A fourth heart sound is frequently audible. The ultrasound technology, the physician can also
turbulence across the aortic valve always produces determine the level of obstruction (supravalvular,
a systolic ejection murmur. It is not necessarily the valvular, or subvalvular), the number of aortic
intensity but the timing of the murmur that deter- cusps, and the degree of cusp fusion.
mines the severity of AS. In patients with mild AS, The severity of aortic stenosis cannot be
the murmur is characterized by an early peak, and determined by visualization of valve motion
the duration ends before the second heart sound. alone, and Doppler echocardiography must be
As AS becomes more severe, the peak intensity of used to further assess the severity of aortic valve
the murmur occurs in mid-to-late systole, and the disease [12].
murmur extends into the second heart sound. The The main hemodynamic parameters recom-
murmur of AS must be differentiated from that of mended for clinical evaluation of AS severity
hypertrophic obstructive cardiomyopathy or mitral with transthoracic echocardiography are [13]:
regurgitation due to a flail posterior leaflet. A
Valsalva maneuver may be of help in this case. 1. AS jet velocity
So we can say that severe AS is characterized 2. Mean transaortic gradient
by (1) a dampened upstroke of the carotid artery, 3. Valve area by continuity equation
(2) a sustained bifid left ventricular impulse, (3)
an absent A2, and (4) a late-peaking systolic ejec- The antegrade systolic velocity across the nar-
tion murmur. rowed aortic valve, or aortic jet velocity, is mea-
sured using continuous-wave (CW) Doppler
(CWD) ultrasound [1416].
Electrocardiography AS jet velocity is defined as the highest veloc-
ity signal obtained from any window after a care-
The electrocardiography usually shows sinus ful examination. Color Doppler is also helpful to
rhythm with left ventricular hypertrophy and left avoid recording the CWD signal of an eccentric
atrial enlargement. An ST interval depression, a mitral regurgitation (MR) jet.

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 173

Fig. 15.4 Continuous-wave


Doppler of aortic stenosis

A smooth velocity curve with a dense outer The aortic valve area can be calculated by
edge and clear maximum velocity should be Doppler echocardiography using the continuity
recorded. The maximum velocity is measured at equation.
the outer edge of the dark signal. To get the veloc- From the parasternal long-axis view, the
itytime integral (VTI) for the continuity equa- operator must get the left ventricular outflow
tion and the mean gradient, it is necessary to tract (LVOT) diameter and convert it to the
draw the outer edge of the dark envelope of the LVOT area. The LVOT velocity is obtained and
velocity curve (Fig. 15.4) [13]. traced to derive the timevelocity integral (TVI)
To distinguish the level and severity of from an apical approach with pulsed-wave
obstruction, the shape of the CW Doppler veloc- Doppler.
ity is helpful; the maximum velocity occurs later Continuity equation
in systole, and the curve is more rounded in shape
with more severe obstruction [13].
( LVOTTVI ) ( LVOTarea )
Aortic valve area =
To determine the fixity or the dynamism of ( AVTVI )
obstruction, the shape of the CWD velocity curve
also can be helpful. A characteristic late-peaking Significant errors in the area calculation may be
velocity curve is shown by a dynamic obstruc- caused by small errors in diameter measurement;
tion, with a concave upward curve in early sys- for this reason the calculation of aortic valve area
tole (Fig. 15.5), in a patient with severe anemia. with continuity equation does require a skilled
Transaortic pressure gradient (P) is calcu- echocardiographist to ensure accurate measure-
lated from velocity (v) using the Bernoulli equa- ment of the LVOT diameter.
tion as When LVOT diameter is squared for calcula-
tion of cross-sectional area, it becomes the great-
DP = 4v 2
est potential source of error in the continuity
The maximum gradient is calculated from maxi- equation [13].
mum velocity To reduce the errors associated with the
measurements of the LVOT, this ratio can be
DPmax = 4v 2 max
used:
and the mean gradient is calculated by averaging
VLVOT
the instantaneous gradients over the ejection Velocity ratio =
period [13]. VAV

ERRNVPHGLFRVRUJ
174 L. Scappini et al.

Fig. 15.5 An example of


dynamic outflow obstruction.
Note the different shapes of
the velocity curves and the
later maximum velocity with
dynamic obstruction

This dimensionless velocity ratio expresses the Table 15.1 Recommendations for classification of AS
size of the valvular effective area as a proportion severity
of the cross-sectional area of the LVOT [13]. In Aortic
the absence of valve stenosis, the velocity ratio sclerosis Mild Moderate Severe
approaches 1. If the velocity ratio is 0.25 or less, Aortic jet 2.5 2.62.9 3.04.0 >4.0
severe stenosis is present [17]. velocity
(m/s)
Aortic valve area planimetry may be an
Mean <20 2040b >40b
acceptable alternative when Doppler estimation gradient (<30a) (3050a) (>50a)
of flow velocities is unreliable. When valve calci- (mmHg)
fication causes shadows or reverberations limiting AVA (cm2) >1.5 1.51.0 <1.0
identification of the orifice, planimetry may be Index AVA >0.85 0.850.6 <0.6
inaccurate. (cm2/m2)
Severe aortic stenosis can be diagnosed if a Velocity >0.50 0.50 <0.25
ratio 0.25
patient has these features:
ESC guidelines
a

AHA/ACC guidelines
b
Clinical findings consistent with severe aortic
stenosis
A mean gradient greater than 40 mmHg
AVA less than 1.0 cm2 (<0.6 cm2/m2 when Aortic valve effective orifice area (EOA)
indexed for body surface area) <1.0 cm2 or <0.6 cm2/m2 when indexed for
body surface area
Recommendations for classification of AS Low mean transvalvular gradient (i.e.,
severity are listed in Table 15.1. <40 mmHg)
Low LVEF (<40 %), causing an LF state [18]
 ow-Flow, Low-Gradient Aortic
L
Stenosis withLow Left Ventricular The diagnostic problem in this kind of pathol-
Ejection Fraction (LVEF) ogy is to distinguish true severe from pseudose-
Low LVEF, LF-LG severe AS is characterized by vere aortic stenosis. In true severe AS, the LV
the combination of these features: dysfunction is a secondary or concomitant

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 175

phenomenon, while the primary culprit is deemed resulting in low cardiac output (i.e., stroke vol-
to be the valve disease. Instead, the main factor in ume index <35 ml/m2) and lower than expected
pseudosevere AS is deemed to be myocardial dis- transvalvular gradients (i.e., <40 mmHg); this
ease, and AS severity is overestimated due to clinical entity was defined as paradoxical
incomplete opening of the valve in relation to the LF-LG AS [26, 27].
LF state. Distinction between these two entities is The prevalence of this entity increases with
essential: patients with true severe AS generally older age, female gender, and concomitant pres-
will benefit from aortic valve replacement (AVR), ence of systemic arterial hypertension.
whereas those with pseudosevere AS may not Paradoxical LF-LG AS is characterized by a
benefit [18]. restrictive physiology, whereby left ventricular
Dobutamine infusion is helpful in differentiat- pump function and thus stroke volume are mark-
ing pseudosevere from true severe AS. edly reduced despite a preserved LVEF. Other
True severe AS is characterized by little or distinctive features are:
absent increase in effective orifice area and an
increase in gradient that is congruent with the Marked reduction in intrinsic LV systolic
relative increase in flow; pseudosevere AS shows function, not evidenced by the LVEF.
an increase in effective orifice area (EOA) and Longitudinal shortening is reduced to a larger
relatively little increase in gradient in response to extent in these patients due to more advanced
increasing flow [18]. fibrosis in the subendocardial layer.
Some parameters and criteria have been pro- More pronounced LV concentric remodeling
posed to identify patients with pseudosevere AS and myocardial fibrosis both contributing to
during dobutamine stress echocardiography: reduce the size, compliance, and filling of the
LV [26, 2830].
Peak stress mean gradient <30 or <40 mmHg
(depending on studies) Several studies reported that these patients
Peak stress EOA >1.0 or 1.2 cm2 have a worse prognosis than those with moderate
An absolute increase in EOA >0.3 cm2 AS or normal flow severe AS [26].
Characteristics of low-flow low-gradient AS,
The optimal cutoff values remain to be deter- paradoxical low-flow low-gradient, and pseudos-
mined. The prevalence of pseudosevere AS is evere AS are shown in Table 15.2.
reported to be between 20 and 30 % [1924].
A high plasma B-type natriuretic peptide
(BNP) level (>550 pg/ml) appears to be a pow- Natural History
erful predictor of mortality in patients with
LF-LG AS regardless of treatment (medical vs. Calcific AS is a chronic disease. Patients may be
surgical) or the presence and/or absence of flow asymptomatic for a long period of time [19, 22,
reserve [25, 38]. 36, 37], and the duration of the asymptomatic
phase varies a lot between individuals. Sudden
Low-Flow, Low-Gradient cardiac death seems to be rare in the truly asymp-
ASwithNormal LVEF (Paradoxical tomatic (<1 % per year), even in very severe AS,
Low-Flow Low-Gradient Aortic while it is a frequent cause of death in symptom-
Stenosis) atic patients. In asymptomatic patients with
Some authors [26] reported that a substantial severe AS, reported average event-free survival at
proportion of patients with severe aortic steno- 2 years ranged from 20 % to more than 50 % [19,
sis (EOA <1.0 cm2 and/or indexed EOA 22, 32, 36, 37].
<0.6 cm2/m2) and a preserved LVEF (i.e., There are clinical and echocardiographic pre-
>50 %) might develop a restrictive physiology, dictors of symptom development and adverse

ERRNVPHGLFRVRUJ
176 L. Scappini et al.

Table 15.2 Characteristics of low-flow low-gradient AS, paradoxical low-flow low-gradient, and pseudosevere AS
Mean gradient
EOA (cm2) Index EOA (cm2/m2) (mmHg) LVEF
Low-flow low-gradient aortic <1.0 cm2 <0.6 cm2/m2 <40 mmHg <40 %
stenosis
Paradoxical low-flow <1.0 cm2 <0.6 cm2/m2 <40 mmHg >50 % with
low-gradient aortic stenosis stroke volume
index < 35 ml/m2
Pseudosevere aortic stenosis Peak stress Absolute increase in Peak stress mean
(during dobutamine stress EOA > 1.0 or EOA > 0.3 cm2 gradient <30 or
echocardiography) 1.2 cm2 40 mmHg

outcomes in asymptomatic patients: older age, [31, 35, 40]. Treadmill testing may be appropri-
presence of atherosclerotic risk factors, peak aor- ate for stratifying patients with asymptomatic
tic jet velocity [19, 22, 36, 37], LVEF [22], rate of severe aortic stenosis, but it is contraindicated in
hemodynamic progression, increase in gradient a symptomatic patient.
with exercise, excessive LV hypertrophy, and
abnormal tissue Doppler parameters of systolic
and diastolic LV function [33, 34]. Indications forIntervention
When the symptoms appear, the prognosis of
severe AS is poor, with survival rates of only  ortic Valve Replacement
A
1550 % at 5 years. Early valve replacement should be recommended
in all patients with severe aortic stenosis and
 ymptomatic andAsymptomatic
S symptoms. As long as the mean gradient remains
Patients >40 mmHg, there is virtually no lower EF limit
Patients with severe aortic stenosis and symp- for surgery [22]. Paradoxical low-flow, low-
toms should be considered for aortic valve gradient aortic stenosis requires special attention.
replacement (AVR). Concomitant coronary In such cases, surgery should be performed only
artery bypass grafting should be performed for when symptoms are present and if comprehen-
coronary atherosclerosis when epicardial lesions sive evaluation suggests significant valve obstruc-
are >50 % [40], and age should not be consid- tion. The management of patients with
ered a contraindication to surgery [39]. The asymptomatic aortic valve stenosis still remains a
treatment of the asymptomatic patient with topic of discussion. Recent studies do not provide
severe aortic stenosis is more controversial. convincing data to support the general recom-
Surgery is reasonable to consider in asymptom- mendation of early AVR, even in patients with
atic patients when there is critical aortic asymptomatic, very severe AS [22].
stenosis:
Balloon Valvuloplasty
Gradient >60 mmHg In patients who will be undergoing surgery or
Valve area <0.6 cm2 TAVI, balloon valvuloplasty may be considered
Expected operative mortality <1.0 % as a bridge to these types of intervention. It may
also be considered in patients with symptomatic
If there is evidence of a high probability of severe AS who require urgent major noncardiac
rapid progression (Doppler peak velocity surgery, in hemodynamically unstable patients
increases by >0.3 m/s per year or when the valve who are at high risk for surgery, and as a pallia-
area decreases by >0.1 cm2 per year), aortic tive measure in selected individual cases when
valve replacement may also be considered for surgery is contraindicated and TAVI is not an
adults with severe asymptomatic aortic stenosis option [22].

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 177

 ranscatheter Aortic Valve


T tricular pacing (by percutaneous transvenous
Implantation (TAVI) pacing wire in the right ventricle), the balloon is
Since few years, transcatheter aortic valve implan- inflated.
tation is the treatment of choice in patients with The transfemoral TAVI requires good femo-
severe aortic valve stenosis who are not candidates ral access. Through a small cut in the thigh, a
for surgery. Contraindications for surgery, or risks catheter is inserted into the body into the femo-
of surgery, are defined by clinical judgment, STS ral artery (rarely the procedure requires the sur-
score >10 %, EuroSCORE (logistic) >20 % and/or gical preparation of the femoral vessel). Once
porcelain aorta, history of thoracic irradiation, inserted into the aorta, the guidewires are
severe thoracic deformity, and patent coronary advanced and placed across the aortic arch and
bypass. TAVI should only be performed in hospi- through the stenosed aortic valve. During this
tals with cardiac surgery on-site and with a heart retrograde pathway, it is important to prevent
team that assesses individual patients risks [22]. calcium dislodgment from a diseased aortic
It should be identified contraindications, both wall.
clinical and anatomic. Eligible patients should Therefore, the valve is advanced over the
have a life expectancy of more than 1 year and guidewire into the aortic position. CoreValve
should also be likely to gain improvement in their auto-expands after the removal of the delivery
quality of life (see indications and contraindica- system, whereas SAPIENTM is expanded using
tions below). the rapid inflation of the inner catheter balloon
During TAVI, a biological valve prosthesis under rapid cardiac pacing (heart beat rate
will be positioned within the calcific native aortic >180 bpm).
valve. The TAVI is performed through a left anterolat-
There are two types of valves used for this eral mini-thoracotomy through the fifth or the
procedure: sixth intercostal space. Once the cardiac apex is
identified (through the use of transthoracic echo-
Medtronic CoreValve System: this is a self- cardiography), a 3 cm incision is made over the
expandable nitinol stent with an inner porcine top of the rib to avoid trauma to the neurovascular
pericardial valve that is designed to sit into bundle. The left lung does not usually interfere
the aortic root and to anchor into the aortic with the exposure of the left ventricular apex.
annulus. The valve is available in two sizes Therefore, a transapical sheath is placed into the
(26 and 29 mm) and is approved only for ret- left ventricle, and a stiff guidewire is placed into
rograde applications [41]. the ascending aorta via the sheath. The valve is
Edwards SAPIENTM: this stent valve is a tubelike advanced over the stiff wire in an anterograde
stainless steel balloon-expandable stent with manner, and it has to be ideally placed 1/3 below
an inner bovine pericardial valve; this valve is the base of aortic sinuses. It is extremely impor-
available in two sizes (23 and 26 mm) and is tant to ensure accurate positioning of the valve
approved for transfemoral and transapical prior to its deployment to avoid malpositioning,
(TA) applications [41]. embolization, and significant perivalvular leak
[42]. Echocardiography and fluoroscopy can be
Two approaches are being mainly used for the valuable aids to confirm the correct positioning of
installation of these biological valve prostheses: the valve.
transfemoral (TF-TAVI) retrograde and transapi- This procedure is however not free from a
cal (TA-TAVI) antegrade approaches. series of complications, more or less serious and
It is important to note that before the position- with different implications on outcome and qual-
ing of the prosthetic valve, a balloon aortic valvu- ity of life (Table 15.3).
loplasty should be performed. An angiogram is Regarding the outcome of patients undergoing
performed to confirm proper positioning of the TAVI, in 2013, data from a 3-year follow-up of
balloon, and during a short period of rapid ven- the PARTNER trial were published.

ERRNVPHGLFRVRUJ
178 L. Scappini et al.

Table 15.3 Complications of post-TAVI Symptom improvement was similar in both


Possible complications of post-TAVI groups and maintained for 3 years.
1. Stroke At 3 years, strokes were similar in TAVI and
1a. Early surgery patients, despite increased periproce-
Periprocedural hypotension dural neurologic events in TAVI patients
Embolism of debris during valve implantation (stroke risk TAVI vs. SAVR, 7.7 vs. 4.9 %
1b. Delayed (periprocedural) 8.2 vs. 9.3 %; p = 0.783 (at
Aggregation of platelets and fibrin on valve leaflet 3 years) [43].
Incomplete endothelialization
TAVI hemodynamic performance was main-
2. Paravalvular leak
tained with similar valve gradients and areas
3. New-onset atrial fibrillation
compared with surgery.
4. Atrioventricular block (with subsequent implant of
PMK) Both TAVI and surgery resulted in significant
Edwards valve: rate of pacemaker implantation less LVEF improvement and LV mass regression.
than 10 % at 30 days Paravalvular leak or total aortic regurgitation
CoreValve: rate of pacemaker implantation was associated with increased late mortality.
exceeding 20 % at 30 days (this could be due to
the nitinol structure of this valve which exerts a The effect was proportional to the severity of
high and persistent radial force on the the regurgitation, and even mild aortic
interventricular septum, causing major changes regurgitation was associated with an increased
on the atrioventricular system) rate of late death.
5. Other major adverse events
Major ventricular tachyarrhythmia
Myocardial infarction The authors concluded that TAVI should be
Cardiac tamponade considered an alternative to surgery with similar
Conversion to surgery mortality and similar other major clinical out-
Valve-in-valve procedure
comes; periprocedural stroke concerns after
Aortic dissection/perforation
TAVI have diminished with longer-term follow-
up, and periprocedural regurgitation (even mild)
The PARTNER trial is the largest randomized has emerged as a predictor of late mortality [43].
trial on TAVI (a first-generation Edwards
SAPIEN valve was used), and two cohorts of
patients in this study were randomized: Follow-Up
PARTNER cohort A compared treatment with
TAVI versus traditional cardiac surgery in high- The wide variability of the rate of progression of
risk patients; PARTNER cohort B compared AS requires that patients be carefully educated
treatment with TAVI versus optimal medical about the importance of follow-up and reporting
therapy in inoperable patients. symptoms as soon as they develop [22]. Follow-up
This study demonstrated the superiority of visits should include echocardiography with
TAVI in terms of 1-year mortality in patients who attention on hemodynamic progression, LV func-
could not undergo surgery (PARTNER cohort B, tion and hypertrophy, and the ascending aorta.
inoperable patients), and non-inferiority com- Every 6 months asymptomatic severe AS
pared traditional surgery (PARTNER cohort A, should be reevaluated for the occurrence of
patients with high surgical risk). symptoms, change in exercise tolerance, and
At 3 years, in patients with symptomatic change in echo parameters. Also measurement of
severe AS who were high-risk candidates for sur- natriuretic peptides may be considered.
gical aortic valve replacement, data have shown: Patients should be reevaluated yearly if there
are mild and moderate AS with significant calci-
No difference in all-cause mortality between fication. Intervals may be extended to 23 years
TAVI and surgery (TAVI vs. SAVR, 44.2 % vs. [22] in younger patients with no significant calci-
44.8 %; p = 0.483) [43]. fication and with mild AS.

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 179

Appendix Severe valve calcification and a rate of


peak transvalvular velocity progression
I ndications forAortic Valve 0.3 m/s per year
Replacement inAortic Stenosis [22]
Class IIB, Level C
Class I, Level B AVR may be considered in symptomatic
AVR is indicated in patients with severe AS patients with severeAS low-flow, low-gradient,
and any symptoms related to AS. and LV dysfunction without flow reserve.
AVR may be considered in asymptomatic
Class I, Level C patients with severe AS, normal EF, and none
AVR is indicated in asymptomatic patients of the abovementioned exercise test abnor-
with severe AS and abnormal exercise test malities, if surgical risk is low, and one or
showing symptoms on exercise clearly related more of the following findings are present:
to AS. Markedly elevated natriuretic peptide lev-
AVR is indicated in patients with severe AS els confirmed by repeated measurements
undergoing CABG and surgery of the ascend- and without other explanations
ing aorta or another valve. Increase of mean pressure gradient with
AVR is indicated in asymptomatic patients exercise by >20 mmHg
with severe AS and systolic LV dysfunction Excessive LV hypertrophy in the absence
(LVEF <50 %) not due to another cause. of hypertension

Class IIa, Level B


AVR should be considered in high-risk  ecommendations fortheUse
R
patients with severe symptomatic AS who are ofTranscatheter Aortic Valve
suitable for TAVI, but in whom surgery is Implantation [22]
favored by a heart team based on the indi-
vidual risk profile and anatomic suitability. Class I, Level C
AVR should be considered in asymptomatic TAVI should only be undertaken with a
patients with severe AS and abnormal exercise multidisciplinary heart team including car-
test showing fall in blood pressure below diologists and cardiac surgeons and other
baseline. specialists if necessary.
AVR should be considered in patients with TAVI should only be performed in hospitals
moderate AS undergoing CABG and surgery with cardiac surgery on-site.
of the ascending aorta or another valve.
AVR should be considered in symptomatic Class I, Level B
patients with low-flow, low-gradient TAVI is indicated in patients with severe
(<40 mmHg) AS with normal EF only after symptomatic AS who are not suitable for AVR
careful confirmation of severe AS. as assessed by a heart team and who are
AVR should be considered in symptomatic likely to gain improvement in their quality of
patients with severe AS; low-flow, low-gradient life and to have a life expectancy of more than
with reduced EF; and evidence of flow reserve. 1 year after consideration of their
AVR should be considered in asymptomatic comorbidities.
patients, with normal EF and none of the
abovementioned exercise test abnormalities, if Class IIa, Level B
the surgical risk is low, and one or more of the TAVI should be considered in high-risk
following findings are present: patients with severe symptomatic AS who
Very severe AS defined by a peak transval- may still be suitable for surgery, but in whom
vular velocity >5.5 m/s TAVI is favored by a heart team based on

ERRNVPHGLFRVRUJ
180 L. Scappini et al.

the individual risk profile and anatomic 8. Julius BK, Spillmann M, Vassalli G et al (1997)
Angina pectoris in patients with aortic stenosis and
suitability.
normal coronary arteries. Mechanisms and patho-
physiological concepts. Circulation 95:892
9. Gaasch WH, Levine HJ, Quinones MA et al (1976)
 ontraindications forTranscatheter
C Left ventricular compliance: mechanisms and clinical
implications. Am J Cardiol 38:645653
Aortic Valve Implantation [22] 10. Hess OM, Ritter M, Schneider J et al (1984) Diastolic
stiffness and myocardial structure in aortic valve dis-
Absolute contraindications: Estimated life expec- ease before and after valve replacement. Circulation
tancy <1 year, improvement of quality of life 69:855865
11. Schreiber C, Lange R (2006) Porcelain aorta: thera-
by TAVI unlikely because of comorbidities,
peutical options for aortic valve replacement and con-
inadequate annulus size (<18 mm, >29 mm), comitant coronary artery bypass grafting. Ann Thorac
thrombus in the left ventricle, active endocar- Surg 82:381
ditis, plaques with mobile thrombi in the 12. Wang A, Bashore TM (2009) Valvular heart disease.
Humana Press, Dordrecht/New York
ascending aorta or aortic arch, and inadequate
13. Baumgartner H, Hung J, Bermejo J et al (2008)
vascular access (vessel size, calcification, Echocardiographic assessment of valve stenosis:
tortuosity). EAE/ASE recommendations for clinical practice.
Relative contraindications: Bicuspid or noncalci- Eur J Echocardiogr 10:125
14. Currie PJ, Seward JB, Reeder GS et al (1985)
fied valves, untreated coronary artery disease
Continuous-wave Doppler echocardiographic assess-
requiring revascularization, and LVEF <20 % ment of severity of calcific aortic stenosis: a simulta-
hemodynamic instability. neous Doppler catheter correlative study in 100 adult
patients. Circulation 71:11621169
15. Smith MD, Kwan OL, DeMaria AN (1986) Value and
limitations of continuous wave Doppler echocardiog-
raphy in estimating severity of valvular stenosis.
References JAMA 255:31453151
16. Burwash IG, Forbes AD, Sadahiro M et al (1993)
1. Nkomo VT, Gardin JM, Skelton TN et al (2006) Echocardiographic volume flow and stenosis severity
Burden of valvular heart diseases: a population-based measures with changing flow rate in aortic stenosis.
study. Lancet 368:10051011 Am J Physiol 265(5 Pt 2):H1734H1743
2. Kurtz CE, Otto CM (2010) Aortic stenosis: clinical 17. Oh JK, Taliercio CP, Holmes DR Jr et al (1988)
aspects of diagnosis and management, with 10 illus- Prediction of the severity of aortic stenosis by Doppler
trative case reports from a 25-year experience. aortic valve area determination: prospective Doppler-
Medicine (Baltimore) 89:349379 catheterization correlation in 100 patients. J Am Coll
3. Lindroos M, Kupari M, Heikkila J et al (1993) Cardiol 11:12271234
Prevalence of aortic valve abnormalities in the elderly: 18. Pibarot P, Dumesnil JG (2012) Low-flow, low-
an echocardiographic study of a random population gradient aortic stenosis with normal and depressed
sample. J Am Coll Cardiol 21:12201225 left ventricular ejection fraction. J Am Coll Cardiol
4. Schueler R, Hammerstingl C, Sinning JM et al (2010) 60(19):18451853. Qubec City, Qubec, Canada
Prognosis of octogenarians with severe aortic valve 19. Vahanian A, Alfieri O, Andreotti F et al (2012)
stenosis at high risk for cardiovascular surgery. Heart Guidelines on the management of valvular heart dis-
96:18311836 ease (version 2012): the Joint Task Force on the
5. OBrien KD, Reichenbach DD, Marcovina SM et al Management of Valvular Heart Disease of the
(1996) Apolipoproteins B, (a), and E accumulate in European Society of Cardiology (ESC) and the
the morphologically early lesion of degenerative European Association for Cardio-Thoracic Surgery
valvular aortic stenosis. Arterioscler Thromb Vasc (EACTS). Eur Heart J 33:24512496
Biol 16:523532 20. De Filippi CR, Willett DL, Brickner E et al (1995)
6. Rajamannan NM, Subramaniam M, Rickard D et al Usefulness of dobutamine echocardiography in dis-
(2003) Human aortic valve calcification is associated tinguishing severe from nonsevere valvular aortic ste-
with an osteoblast phenotype. Circulation nosis in patients with depressed left ventricular
107:21812184 function and low transvalvular gradients. Am J
7. Olsson M, Dalsgaard CJ, Haegerstrand A et al (1994) Cardiol 75:191194
Accumulation of T lymphocytes and expression of 21. Schwammenthal E, Vered Z, Moshkowitz Y et al
interleukin-2 receptors in nonrheumatic stenotic aor- (2001) Dobutamine echocardiography in patients
tic valves. J Am Coll Cardiol 23:11621170 with aortic stenosis and left ventricular dysfunction:

ERRNVPHGLFRVRUJ
15 Aortic Stenosis 181

predicting outcome as a function of management 31. Rosenhek R, Binder T, Porenta G et al (2000)


strategy. Chest 119:17661777 Predictors of outcome in severe, asymptomatic aortic
22. Monin JL, Quere JP, Monchi M et al (2003) Low- stenosis. N Engl J Med 343:611617
gradient aortic stenosis: operative risk stratification 32. Pellikka PA, Sarano ME, Nishimura RA et al (2005)
and predictors for long-term outcome: a multicenter Outcome of 622 adults with asymptomatic, hemody-
study using dobutamine stress hemodynamics. namically significant aortic stenosis during prolonged
Circulation 108:319324 follow-up. Circulation 111:32903295
23. Nishimura RA, Grantham JA, Connolly HM et al 33. Rosenhek R, Zilberszac R, Schemper M et al (2010)
(2002) Low-output, low-gradient aortic stenosis Natural history of very severe aortic stenosis.
in patients with depressed left ventricular systolic Circulation 121:151156
function: the clinical utility of the dobutamine chal- 34. Bergler-Klein J, Klaar U, Heger M et al (2004)
lenge in the catheterization laboratory. Circulation Natriuretic peptides predict symptom-free survival
106:809813 and postoperative outcome in severe aortic stenosis.
24. Zuppiroli A, Mori F, Olivotto I et al (2003) Therapeutic Circulation 109:23022308
implications of contractile reserve elicited by dobuta- 35. Monin JL, Lancellotti P, Monchi M et al (2009) Risk
mine echocardiography in symptomatic, low-gradient score for predicting outcome in patients with asymp-
aortic stenosis. Ital Heart J 4:264270 tomatic aortic stenosis. Circulation 120:6975
25. Bergler-Klein J, Mundigler G, Pibarot P et al (2007) 36. Kvidal P, Bergstrom R, Horte LG et al (2000)
B-type natriuretic peptide in low-flow, low-gradient Observed and relative survival after aortic valve
aortic stenosis: relationship to hemodynamics and replacement. J Am Coll Cardiol 35:747756
clinical outcome. Circulation 115:28482855 37. Wang A, Bashore TM, Sorajja P et al (2009) Valvular
26. Hachicha Z, Dumesnil JG, Bogaty P et al (2007) heart disease aortic stenosis. Valvular heart disease
Paradoxical low flow, low gradient severe aortic ste- aortic stenosis. Humana Press, a part of Springer
nosis despite preserved ejection fraction is associated Science. doi:10.1007/978-1-59745-411-7
with higher afterload and reduced survival. Circulation 38. Tribouilloy C, Lvy F, Rusinaru D et al (2009)
115:28562864 Outcome after aortic valve replacement for low-flow/
27. Dumesnil JG, Pibarot P, Carabello B (2010) low-gradient aortic stenosis without contractile
Paradoxical low flow and/or low gradient severe aor- reserve on dobutamine stress echocardiography. J Am
tic stenosis despite preserved left ventricular ejection Coll Cardiol 53:18651873
fraction: implications for diagnosis and treatment. Eur 39. Brown ML, Pellikka PA, Schaff HV et al (2008) The
Heart J 31:281289 benefits of early valve replacement in asymptomatic
28. Cramariuc D, Cioffi G, Rieck AE et al (2009) Low- patients with severe aortic stenosis. J Thorac
flow aortic stenosis in asymptomatic patients: val- Cardiovasc Surg 135:308315
vular arterial impedance and systolic function from 40. Kang DH, Park SJ, Rim JH et al (2010) Early surgery
the SEAS substudy. J Am Coll Cardiol Img 2: versus conventional treatment in asymptomatic very
390399 severe aortic stenosis. Circulation 121:15021509
29. Lancellotti P, Donal E, Magne J et al (2010) Impact of 41. Swiss Med Wkly. 2010;140:w13127
global left ventricular afterload on left ventricular 42. Anson C, Kevin ML (2012) Illustrated techniques for
function in asymptomatic severe aortic stenosis: a transapical aortic valve implantation. Ann
two-dimensional speckle-tracking study. Eur J Cardiothorac Surg 1(2):231239
Echocardiogr 11:537543 43. Thourani VH (2013) Three-year outcomes after trans-
30. Herrmann S, Stork S, Niemann M et al (2011) Low- catheter or surgical aortic valve replacement in high-
gradient aortic valve stenosis: myocardial fibrosis and risk patients with severe aortic stenosis. American
its influence on function and outcome. J Am Coll College of Cardiology Scientific Session/i2, - cardio-
Cardiol 58:402412 letter.ch

ERRNVPHGLFRVRUJ
Mitral Regurgitation
16
MarcoFlori, LorenaScappini, andLucaPiangerelli

16.1 Case Report No previous or current pathology, but


about 15 years ago, he was told to have a
cardiac murmur, without further
A 68-year-old man is referred to his family investigation.
doctor for dyspnea and fatigue for ordinary A mild cough without expectoration was pres-
activities; symptoms arose 3 months before. ent in the last month without fever.
At the beginning, dyspnea was mild and
was present only after a moderate to intense
physical activity. In the last 2 weeks, the Allergies
symptoms worsened until dyspnea was evi-
dent during moderate physical activity (fast None
walking or after few steps of stairs) with
even one episode of orthopnea at night that
resolved after 30 min. No chest pain at rest Social History
or during exertion was referred; there were
no syncope or dizziness and no palpitations. Retired teacher
Smokes 10 cigarettes per day since youth
Never used illicit drugs
Doesnt drink alcohol or coffee
 edical History andCardiovascular
M
Risk Factors
Medications
Hypercholesterolemia LDL-C 145 mg/dl.
Hypertension treated with drugs in the last 10 Amlodipine 5 mg/day at 20:00
years.

M. Flori (*) L. Scappini L. Piangerelli Vital Signs


Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche, Temperature: 36.7 C
Via Conca, 71, Ancona 60126, Italy Heart rate: 85 bpm
e-mail: mflori3001@gmail.com;
lorena.scappini@gmail.com; Blood pressure: 120/80 mmHg
lucapiangerelli30@gmail.com Respiratory rate: 16 breaths per minute

Springer International Publishing Switzerland 2015 183


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI10.1007/978-3-319-19926-9_16

ERRNVPHGLFRVRUJ
184 M. Flori et al.

Physical Examination The patient didnt refer fever and cough was
mild and without expectoration. Furthermore the
General: no acute distress, alert, awake, and symptoms arose 3 months before, so the hypothe-
oriented. Well developed and well nourished sis of a pneumonitis or bronchitis seemed unlikely.
Head, eyes, ears, nose, and throat: normal Myocardial ischemia was possible even with-
Neck: supple, mild jugular venous distention, out chest pain; dyspnea could have been an angi-
no lymphadenopathy, and no carotid bruit nal equivalent.
Cardiovascular: regular rate and rhythm, S1 According to the physical exam (rales at the
and S2 normal, third tone audible, blowing bases bilaterally, peripheral edema, mild jugular
crescendo-decrescendo holosystolic murmur distension, third tone, and cardiac murmur), the
grade IV in Levine scale, and best audible at most probable cause seemed to be heart failure.
the fifth left midclavicular intercostal space
and irradiated to the armpit
Lungs: harsh breath sounds with rales on  hat Kind ofDisease Is Suggested
W
auscultation at the bases bilaterally, no rhon- fromCardiac Physical Examination?
chi or wheezes, no alterations in tactile fremi-
tus, and normal upercussion Aortic stenosis
Abdomen: mild overweight, no pulsatile Aortic regurgitation
masses, normal bowel sounds in all four quad- Mitral stenosis
rants, no abdominal rebound or guarding, no Mitral regurgitation
costovertebral angle tenderness, and no
hepatosplenomegaly There was a left apical at the fifth left intercos-
Extremities: no cyanosis or clubbing. Mild tal space toward the anterior axillary line. Systolic
peripheral pitting edema at ankle murmur was located in the mitral region and radi-
Neurologic: cranial nerves II through XII ated to the armpit. Its quality (holosystolic, blow-
intact and no focal deficit ing) suggested a mitral regurgitation over aortic
Psychiatric: normal stenosis.
Skin: intact, no rashes, no lesions, and no
erythema
What Should theFamily Doctor Do?

 hat Are thePossible Causes


W Treat heart failure with appropriate drugs
forDyspnea onExertion andFatigue? according to guidelines.
Refer the patient to a cardiologist.
Refer the patient to the emergency room.
Flu
Lung diseases The diagnosis of heart failure was done, and a
COPD myocardial ischemia should be ruled out.
Pneumonia The patient was referred to a cardiologist as a
Bronchitis correct choice and also an EKG and echocardiog-
Heart diseases raphy assessment was requested. Short-term
Heart failure delay should be suggested and visit undertaken
Myocardial ischemia within a week.
Bradyarrhythmias The patient was stable at rest and symptoms
Tachyarrhythmias were stable in the last week; therefore, there was no
reason for urgent access to the emergency room.

ERRNVPHGLFRVRUJ
16 Mitral Regurgitation 185

After 5 days, the patient was seen by a cardi- quence of either hypertension or mitral insuffi-
ologist which confirmed the diagnosis of MR; a ciency. These alterations led to the development
routine EKG at rest (resulting normal) and echo- of heart failure. The presence of cardiac remodel-
cardiography were performed. ing suggested a chronic valve disease, and even if
the symptoms arose 3 months before, severe
insufficiency was probably present since several
Echocardiography (Fig.16.1) months or years. Myxoid degeneration of valve
apparatus (Barlow disease) was the probable
Normal and trileaflet aortic valve. Normal aorta. mechanism of valve disease. No alterations sug-
Moderate dilatation of the left atrium (LA diam- gesting endocarditis were found. Ventricle was
eter M-mode = 4.5 cm; area 4c = 26 cm2). dilated, but the extent of dilatation wasnt suffi-
Eccentric hypertrophy of the left ventricle (with cient to consider valve insufficiency as
end diastolic diameter 60 mm). Normal systolic secondary.
function (ejection fraction 60 % measured with
Simpsons biplane) without focal hypokinesia or
akinesia. Thickened mitral leaflet with severe Final Diagnosis
prolapse of the posterior leaflet and mild prolapse
of the anterior leaflet. Severe eccentric mitral Congestive heart failure due to a severe mitral
regurgitation (vena contracta 7.5 mm, PISA valve prolapse and regurgitation
1.1 cm, effective regurgitant orifice area (EROA)
0.5 mm2). Normal right atrium (area 4c = 17 cm2).
Restrictive mitral inflow pattern (grade III dia- What Should theCardiologist Do?
stolic dysfunction). Normal tricuspid valve. Mild
tricuspid regurgitation which was centrally Refer the patient for cardiac surgery.
directed with an estimated PASP = 50 mmHg. Treat the patient for heart failure.
Normal pulmonic valve. Normal interatrial sep- Treat the patient with drugs and reevaluate
tum. Normal pericardium. Normal aorta. Slightly after 3 months.
dilated inferior vena cava. Less than 50 % inspi-
ratory collapse of the IVC. Cardiac surgery in primary mitral regurgitation
Mitral regurgitation had major hemodynamic is curative and the occurrence of symptoms is a
consequences in this patient and led to atrial major indication for surgery. The presence of
enlargement due to chronic volume overload. increased PASP is a further indication for surgery.
Ventricular hypertrophy was probably the conse- Medical therapy may alleviate symptoms, but

Fig. 16.1 Echocardiography, four-chamber view. From swirling toward interatrial septum and reaching the
left to right: two-dimensional imaging shows thickened pulmonary veins (Coand effect, arrow point). Nyquist
mitral leaflet with posterior leaflet prolapse (arrow). Color limit is shifted downward to detect proximal isovelocity
Doppler shows severe eccentric mitral regurgitation surface area (PISA) >1 cm (star)

ERRNVPHGLFRVRUJ
186 M. Flori et al.

reduction of valve regurgitation is unlikely (which and the left ventricular wall [2]. When one of
is possible in secondary MR). Hence evaluation these portions of the apparatus becomes abnor-
for cardiac surgery, including coronary angiogra- mal, MR may develop. Also left ventricle dilata-
phy for the detection of asymptomatic coronary tion or papillary muscle displacement can
artery disease, is the right choice. Concurrent provoke the dysfunction of the mitral valve
medical therapy for heart failure, including diuret- apparatus.
ics, beta-blockers, and ACE inhibitors or ARBs, As seen above, the etiology of MR can be
is important until cardiac surgery is undertaken. stratified into nonischemic and ischemic.
Nonischemic etiologies include:

16.2 Mitral Regurgitation (a) Degenerative MR includes myxomatous


degeneration of the mitral valve with resul-
Definition andClassification tant mitral valve prolapse with or without
ruptured chordae, mitral valve leaflet sclero-
Mitral regurgitation (MR) is characterized by a sis or calcification, and mitral annulus
systolic blood flow reversal from the left ventricle calcification.
(LV) to the left atrium (LA). MR can be classified (b) Rheumatic MR is usually due to commissure
based on etiology and mechanisms. The etiology fusion in chronic type, while in pure active
of MR is generally stratified as ischemic that rheumatic MR, annulus dilatation and ante-
occurs in up to 40 % of patients affected by rior leaflet prolapse from chordal elongation
myocardial infarction [1] and nonischemic. are described [3].
Mechanisms of MR are stratified as functional MR etiology is predominantly degenera-
(mitral valve is structurally normal and MR is due tive in developed countries and rheumatic in
to valve deformation caused by ventricular remod- developing countries:
eling) or as organic (intrinsic valve lesions). A fur- (c) Infective endocarditis (5 % of severe MR)
ther classification of MR, according to Carpentier, that causes MR mainly through tissue
is by observation of leaflet movement: type I (nor- destruction.
mal valve movement such as annular dilatation, (d) Congenital lesions (isolated cleft mitral valve
perforation), type II (excessive movement), and or associated with persistent atrioventricular
type III (restrictive movement: IIIa, diastolic canal, corrected transposition with or with-
restriction such as rheumatic disease; IIIb, systolic out Ebsteins abnormality of the left atrio-
restriction as in functional MR) (Table 16.1). ventricular valve).
(e) Iatrogenic (radiation/drugs).
(f) Cardiac tumors.
Etiology (g) Other causes (connective tissue diseases,
systemic lupus erythematosus, antiphospho-
The mitral valve apparatus is made of the leaflets, lipid antibody syndrome, cardiac trauma,
the annulus, the chordae, the papillary muscles, hypertrophic cardiomyopathy or sarcoidosis,

Table 16.1 Severity of mitral regurgitation in adults


Mild Moderate Severe
LV and LA size Normal size LV LV and LA enlarged
Vena contracta <3 36.9 >7
PISAr (Nyquist limit of 40 cm/s) <0.4 cm >1 cm
Regurgitant volume <30 ml/bpm 3059 ml/bpm >60 ml/bpm
Regurgitant fraction <30 % 3049 % >50 %
Regurgitant orifice area <0.20 cm2 0.200.39 cm2 >0.40 cm2

ERRNVPHGLFRVRUJ
16 Mitral Regurgitation 187

eosinophilic syndromes, endocardial fibro- reduction or improved contractility. Its change is


elastosis, carcinoid tumors). independent of rate [8]. The consequences of vol-
Ischemic etiologies include papillary muscle ume overload of the LA and LV are different
(PPM) displacement, leaflet tethering, and depending on the fact that regurgitation is acute
annular dilatation. Functionally the leaflets or chronic. In acute severe MR, the left ventricle
present a restricted systolic motion due to responds to the sudden volume overload with
tethering forces that displace the coaptation increased sarcomere stretch and augmented left
surface toward the left ventricle (LV) apex [4]. ventricular stroke volume (Frank-Starling mech-
According to the classification of the mecha- anism). The larger volume increases left ventric-
nisms of MR, it is possible to distinguish two ular diastolic pressure, which in turn increases
types of pathologies: functional MR and organic left atrial pressure. However, left atrial compli-
MR. While the former includes all forms of MR ance is normal in the acute state and the large
that are not secondary to alterations of the valve regurgitant volume markedly increases left atrial
per se, the latter is due to primary mitral valve pressure. This phenomenon causes pulmonary
disease (fusion of commissures, annulus dilata- congestion, edema, and dyspnea. Otherwise,
tion, leaflet prolapse, chordal elongation, retrac- chronic MR progresses slowly, so the LA has
tile fibrosis, etc.). enough time to remodel and accommodate the
Even if etiology and mechanism are not syn- RVol with a near normal LA pressure. As a result,
onymous (a single etiology may generate MR by clinical tolerance may be excellent [10]. In these
different mechanisms), the mechanism of isch- patients, the Frank-Starling mechanism contin-
emic and functional MR is similar; in fact mitral ues to augment total stroke volume. The left
leaflets are intrinsically normal, but their coapta- atrium dilates, thus increasing its compliance.
tion is incomplete [5] because of annular and left The blood that reflows from a dilated left ventri-
ventricular dilatation due to ischemia, previous cle to a dilated atrium provokes only a small rise
myocardial infarction and scarring, aneurysm of filling pressures, with minimal symptoms of
formation, cardiomyopathy, or myocarditis. pulmonary congestion (chronic compensated
stage).
LV dysfunction (chronic decompensated
Pathophysiology stage) is a frequent complication of severe MR
that occurs if the disease is not treated properly
Anatomic malcoaptation of mitral leaflets during during the reversible phase [11, 12]. It is associ-
systole results in a regurgitant orifice (ERO), ated with myocardial structural damage as inter-
which under the influence of the pressure gradi- stitial fibrosis and reduction in myofiber content
ent between the LV and LA allows abnormal that provokes decrease in myofiber contractility
regurgitant flow into the LA. The systolic pres- [13] and increase in left ventricular filling pres-
sure gradient between the LV and LA begins sures and finally pulmonary congestion. A down-
when mitral valve closes (S1) and persists after ward cycle is now activated: increased ventricular
aortic valve closure (S2) up to mitral opening [6]. pressure further dilates the left ventricle, increas-
Thus, regurgitant flow is typically holosystolic. ing systolic wall stress and afterload, which in
The regurgitant flow throughout systole is the turn reduces ventricular systolic function. The
regurgitant volume (RVol) accumulated in the time during which patients progress from com-
LA, which reenters the LV during the next dias- pensated to decompensated MR depends on some
tole, causing a volume overload of LA and LV. variables as the severity of the regurgitation,
Determinants of this volume overload are the afterload, and ventricular contractility.
area of ERO [7], the regurgitant gradient, and In ischemic/functional MR, blood regurgita-
duration of regurgitation. In both organic MR [8] tion is provoked by tethering and tenting of the
and functional MR [9], ERO increases when leaflets and loss of coaptation surface [14, 15] due
afterload increase and decreases with afterload to localized LV deformation with apical and

ERRNVPHGLFRVRUJ
188 M. Flori et al.

posterior displacement of the papillary muscles. Physical Examination


The RVol in ischemic MR is usually less than in MR is usually diagnosed by auscultation. S1 may
organic MR [16], and LV dilatation and LA dila- be diminished due to the failure in the closure of
tation are in excess to the degree of MR [7], due to the mitral valve. A soft systolic murmur, best
the presence of a dilatated, poor performant heart. heard at the apex and radiated to the axilla, is the
most frequent finding, but its timing and intensity
depend on severity and mechanisms of MR. The
Clinical Manifestations murmur has little variations with respiration and
becomes louder when the preload (i.e., raising
Chronic MR is a slow process that allows a grad- legs) or afterload (i.e., squatting) increases.
ual LA enlargement and a consequent increase in Sometimes the systolic murmur isnt audible
LA compliance that permits to maintain a normal even in the presence of moderate or severe MR
or near normal LA pressure. For this reason, (silent MR), especially in ischemic MR, owing to
patients may remain asymptomatic for years a low pressure gradient between the left ventricle
despite severe MR. However, once symptoms and atrium (low systemic blood pressure with
arise, fatigue and generalized weakness predomi- elevated LA pressure). A systolic click may sug-
nate early. As left ventricular function deterio- gest a valve prolapse, while a thrill or S3 suggests
rates, exertional dyspnea, orthopnea, and a severe regurgitation.
paroxysmal nocturnal dyspnea become more
prominent. Frank pulmonary edema or hemopty- Electrocardiogram
sis may also develop and is usually triggered by Electrocardiogram usually shows left atrial
atrial fibrillation or increase in the degree of enlargement (notched P wave >0.12 s in DII).
MR. Sudden death as the initial presentation of Left ventricular enlargement is less frequent [17]
MR is possible [17]. and right ventricular hypertrophy rare. Fibrillation
is a common arrhythmia in these patients due to
atrial enlargement and increased shear stress. Q
Diagnosis wave or ST-T abnormalities may be found in
ischemic MR.
Symptoms
Clinical manifestations of mitral regurgitation Echocardiography
(MR) are related to the severity of regurgitation, Transthoracic echocardiography (TTE) is a sen-
the rate of progression, and the associated sitive technique in initial evaluation and longitu-
cardiopathy. dinal assessment of mitral valve disease. TTE
Acute MR due to chordal rupture (flail), allows to detect the presence of regurgitation and
endocarditis, or trauma leads to an increase in hemodynamic consequences. A careful evalua-
left atrial pressure due to a volume overload in tion of mitral structures with the determination of
a noncompliant left atrium. Pulmonary edema mechanisms and etiology is required. Assessment
or cardiogenic shock may be the presenting of left ventricular and atrial size and function is
symptoms owing to elevated filling pressures mandatory as it provides information about the
and low cardiac output. The dramatic increase mechanism and helps in defining a correct surgi-
of pulmonary pressure may also cause right cal timing.
heart failure. Transesophageal echocardiography (TOE) is
Chronic MR leads to a progressive left atrial useful when TTE quality is suboptimal or when a
enlargement without elevated left atrial pressure. more accurate definition of valvular lesions is
Patients usually remain asymptomatic for years required like in reparative surgery. TOE plays
unless there is left ventricular (LV) failure. also a role in intraoperative assessment.
Dyspnea and fatigue gradually worsening over Three-dimensional echocardiography is an
the years are the most frequent symptoms. emerging technique in assessing MR mechanisms

ERRNVPHGLFRVRUJ
16 Mitral Regurgitation 189

and etiologies providing more accurate evalua- Grading Severity


tion on leaflet motions and multiple images simi- Based on the 2014 ESC Guidelines [18] for the
lar to surgeon view. Management of Patients With Valvular Heart
Disease, the easiest approach is the measurement of
Primary Mitral Regurgitation the narrowest part of the jet on Doppler color flow
Primary MR is due to intrinsic valvular disease (vena contracta) [19]. More accurate echocardio-
with a large spectrum of possible valve lesions. graphic evaluations may be obtained by calculation
Progressive thickening of the valve with fusion of of regurgitant volume, regurgitation fraction, and
commissures and doming of the anterior leaflet effective regurgitant orifice area (EROA) based on
may suggest a rheumatic disease, with vegeta- the evaluation of the convergence signal during
tions or abscess points at endocarditis. When the color flow imaging [20]. EROA should be calcu-
etiology is chordal or papillary rupture, a flail lated by dividing the flow rate through the regurgi-
leaflet may be observed. tant orifice (product of the area of the hemisphere
Degenerative MR is the most common cause and aliasing velocity) by the peak velocity of the jet:
of surgical repair and ranges from billowing to
complete prolapse or floppy valve (>5 mm dia- EROA = ( 2r 2 * Va ) / PkVreg
stolic excursion in left atrium). The presence of
redundant and thickened leaflets may suggest where 2r = surface area of hemisphere,
myxoid infiltration by Barlows disease. Mitral Va = aliasing velocity, and PkVreg = peak V of
calcifications are a frequent finding in older regurgitant jet.
patients and may be a limitation to surgical repair The presence of systolic flow reversal in the
when too extended. pulmonary vein is a significant marker of severity
but may not be present in the absence of sinus
Functional Mitral Regurgitation rhythm [21]. Left atrial and ventricular enlarge-
Functional MR is related to alterations in LV ments are also typical findings and often reflect
geometry due to ischemic or dilated cardiomyop- the history of the disease.
athy with distortion in normal spatial relationship Sometimes in ischemic MR, the regurgitant
between mitral valve apparatus. The dispropor- flow may be trivial at rest and worsens under exer-
tion between closing forces (LV contractility, cise due to papillary muscle dysfunction related to
annular contraction, papillary muscle synchrony) the hypokinesis of left ventricular walls.
and tethering forces (LV and annular dilatation
with displacement of papillary muscles) leads to Cardiac Catheterization
incomplete coaptation and MR. The regurgitant Cardiac catheterization for the evaluation of
flow during systole usually varies with an early intracardiac pressures or left ventricular angiog-
systolic peak and a progressive decrease. raphy is not recommended in routine evaluation
Ischemic MR may result from a restricted sys- of MR. Coronary angiography should be per-
tolic motion of the leaflet leading to inadequate formed when coronary disease is suspected on
apposition particularly in posterior infarction the basis of symptoms or the presence of reduced
(asymmetric pattern). A mitral incompetence due LV systolic function and risk factors [18].
to a deficit in coaptation of both leaflets (sym- Right heart catheterization may demonstrate
metric pattern) may be found in more complex the presence of significant v wave due to the
infarction or dilated cardiomyopathy [4]. regurgitant blood volume in the left atrium.
The LV is usually spherical with multiple
regional dysfunctions. Tenting distance should be
measured as the distance between annulus plane Treatment
and coaptation point, while the tenting area is the
area enclosed between annulus plane and Severe MR imposes a significant overload to LV
leaflets. leading to progressive hypertrophy and dilatation.

ERRNVPHGLFRVRUJ
190 M. Flori et al.

Acute MR carries a poor prognosis and in most replacement and ring annuloplasty with excellent
cases requires surgery after hemodynamic stabi- outcomes in experienced centers [23]. The major
lization. Chronic severe MR, even if asymptom- benefits in valvular repair are the preservation of
atic, reduces the patients prognosis to 5 years subvalvular apparatus and the absence of
[18]. The management of MR largely depends on prosthetic valves. Preoperative echocardiography
pathophysiology, severity, and the presence of is needed to correctly evaluate the anatomy of the
indications to surgery. Two mitral surgery tech- regurgitant valve and to choose the appropriate
niques are available: valve repair and valve surgical approach. TOE is also recommended
replacement. Recently a percutaneous edge-to- intraoperatively to enhance surgery outcomes.
edge procedure has been proposed with good The durability of mitral repair is a potential
clinical results [22] and may be performed in limitation. In experienced centers with appropri-
inoperable patients who meet the echocardio- ate patients screening, the rate of freedom from
graphic criteria. reoperation is similar to valve replacement.
Extensive calcifications, prolapse of more than
 rimary Mitral Regurgitation
P one-third of the leaflet, or complete muscle rup-
Acute MR due to papillary or chordal rupture is ture usually suggests replacement over repair.
an emergency and often requires surgical treat-
ment after medical stabilization with an intra- Mitral Valve Replacement
aortic balloon pump and inotropes or vasodilators. Mitral valve replacement requires the use of a
According to the most recent guidelines on val- prosthetic valve. The choice between mechanical
vular disease, mitral valve repair should be pre- or bioprosthetic valves and issues about prosthetic
ferred when feasible [18]. In chronic MR, surgical valves will be best developed in Chap. 17.
treatment is indicated in symptomatic patients According to ESC guidelines [18], a bioprosthetic
with no contraindications for surgery. Indications valve must be preferred in patients with contrain-
for surgery as suggested by the guidelines are dication to warfarin or >65 years. Chordal preser-
reported in Table 16.2. vation should be preferred if possible (i.e.,
subvalvular scarring or calcification) due to the
Mitral Valve Repair negative impact of left ventricular function.
Valve repair should be the preferred surgical
treatment in primary MR when feasible. Current Medical Therapy
techniques include the use of artificial chord Management of acute MR with hemodynamic
instability should be pointed on reduction of fill-
ing pressures and reduction of the regurgitant
Table 16.2 Indications for mitral valve surgery in
primary MR according to the 2012 ESC guidelines blood volume. Sodium nitroprusside should be
used to reduce afterload and regurgitant fraction
Class I evidence or agreement that mitral valve
surgery is indicated as intra-aortic balloon pump (IABP). IABP and
Patients with severe MR undergoing CABG and LVEF inotropes may be added in case of hypotension or
>30 % shock. Theres no evidence about the use of vaso-
Class IIa weight of evidence favors mitral surgery dilators in chronic MR.
Patients with moderate MR undergoing CABG
Patients with severe MR, LVEF <30 %, and the need  unctional Mitral Regurgitation
F
for revascularization with evidence of myocardial In functional or secondary MR, mitral valve
viability
apparatus is normal and the regurgitant volume
Class IIb weight of evidence less well established
results from a disproportion between closing and
Symptomatic severe MR with LVEF >30 % despite
medical treatment, low comorbidity, and no need for tethering forces. The ischemic etiology carries a
revascularization poor prognosis. Severe tricuspidal regurgitation
LVESD left ventricular end systolic diameter, LVEF left is often associated, and right ventricular dysfunc-
ventricular ejection fraction tion may be a predictor of worse outcome [24].

ERRNVPHGLFRVRUJ
16 Mitral Regurgitation 191

Medical Therapy Cardiac Resynchronization Therapy


In functional MR with heart failure, the role of Cardiac resynchronization therapy (CRT) is a
medical therapy must be emphasized. Optimal pacing modality that provides synchronized
medical therapy, according to guidelines [18], biventricular stimulation to improve cardiac
including ACE inhibitor/ARB, beta-blocker, and function (discussed in Chap. 10). CRT may also
aldosterone antagonist, is the first step in these reduce the degree of MR and left ventricular vol-
patients with a benefit in improving cardiac func- umes by reducing papillary dyssynchrony and
tion and overall survival. Some data suggest a increasing closing forces [26]. Ischemic MR
possible role of medical therapy in reducing the seems to have a worse response to CRT, so a
degree of functional MR [25]. Diuretics and/or careful evaluation is needed in this population.
nitrates may be added to reduce volume overload Benefits gained by CRT disappear with therapy
and treat symptoms. discontinuation [27].

Mitral Valve Repair Percutaneous Mitral Repair


The role of mitral surgery remains controversial A percutaneous edge-to-edge mitral valve repair
as it carries a higher operative mortality than pri- has been recently proposed in patients with func-
mary MR and worse long-term prognosis. When tional MR already judged inoperable by a Heart
indicated (Table 16.3), the preferred technique Team and who fulfill echocardiographic criteria
should be mitral annuloplasty with an undersized (IIb, level of evidence C).
ring that ensures low surgical risk but a high
recurrence of MR.
References
1. Kumanohoso T, Otsuji Y, Yoshifuku S, Matsukida K,
Koriyama C, Kisanuki A, Minagoe S, Levine RA, Tei
Table 16.3 Indications for mitral valve surgery in C (2003) Mechanism of higher incidence of ischemic
functional MR according to the 2012 ESC guidelines mitral regurgitation in patients with inferior myocar-
dial infarction: quantitative analysis of left ventricular
Class I evidence or agreement that mitral valve and mitral valve geometry in 103 patients with prior
surgery is indicated myocardial infarction. J Thorac Cardiovasc Surg
Acute severe MR 125:135143
Severe MR in symptomatic patients with LVESD 2. Otto CM (2001) Evaluation and management of
<55 mm and LVEF >30 % chronic mitral regurgitation. N Engl J Med 345:740
Asymptomatic patients with LV dysfunction (LVESD 3. Marcus RH, Sareli P, Pocock WA, Barlow JB (1994)
>45 mm and/or LVEF <60 %) The spectrum of severe rheumatic mitral valve disease
in a developing country. Correlations among clinical
Class IIa weight of evidence favors mitral surgery presentation, surgical pathologic findings, and hemo-
Asymptomatic severe MR with preserved LV function dynamic sequelae. Ann Intern Med 120:177183
and new onset of atrial fibrillation or pulmonary 4. Tibayan FA, Rodriguez F, Zasio MK, Bailey L, Liang
hypertension (defined as systolic pulmonary pressure D, Daughters GT, Langer F, Ingels NB, Miller DC
at rest >50 mmHg) (2003) Geometric distortions of the mitral valvular-
Asymptomatic severe MR with preserved LV function ventricular complex in chronic ischemic mitral regur-
with high likelihood of surgical repair and low surgical gitation. Circulation 108(Suppl 1):II116II121
risk and/or flail leaflet 5. Izumi S, Miyatake K, Beppu S et al (1987) Mechanism
Severe MR with LV dysfunction (LVEF <30 % or of mitral regurgitation in patients with myocardial
LVESD >55 mm) with high likelihood of surgical infarction: a study using real-time two-dimensional
repair and low comorbidity Doppler flow imaging and echocardiography.
Circulation 76:777785
Class IIb weight of evidence less well established
6. Yellin EL, Yoran C, Sonnenblick EH, Gabbay S,
Severe MR in patients with severe LV dysfunction Frater RW (1979) Dynamic changes in the canine
(LVEF <30 % or LVESD >55 mm) with low mitral regurgitant orifice area during ventricular ejec-
likelihood of surgical repair and low comorbidity tion. Circ Res 45:677683
CABG coronary artery bypass grafting, LVEF left ven- 7. Enriquez-Sarano M, Seward JB, Bailey KR, Tajik AJ
tricular ejection fraction (1994) Effective regurgitant orifice area: a noninvasive

ERRNVPHGLFRVRUJ
192 M. Flori et al.

Doppler development of an old hemodynamic con- severe mitral regurgitation. Am J Cardiol


cept. J Am Coll Cardiol 23:443451 70:10881092
8. Yoran C, Yellin EL, Becker RM, Gabbay S, Frater 18. Lancellotti P et al (2013) Recommendations for the
RW, Sonnenblick EH (1979) Dynamic aspects of echocardiographic assessment of native valvular
acute mitral regurgitation: effects of ventricular vol- regurgitation: an executive summary from the
ume, pressure and contractility on the effective regur- European Association of Cardiovascular Imaging.
gitant orifice area. Circulation 60:170176 Eur Heart J Cardiovasc Imaging 14:611644
9. Keren G, Bier A, Strom JA, Laniado S, Sonnenblick 19. Tribouilloy C, Shen WF, Qur JP, Rey JL, Choquet
EH, LeJemtel TH (1986) Dynamics of mitral regurgi- D, Dufoss H et al (1992) Assessment of severity of
tation during nitroglycerin therapy: a Doppler echo- mitral regurgitation by measuring regurgitant jet
cardiographic study. Am Heart J 112:517525 width at its origin with transesophageal Doppler color
10. Braunwald E, Awe WC (1963) The syndrome of flow imaging. Circulation 85:12481253
severe mitral regurgitation with normal left atrial 20. Enriquez-Sarano M, Miller FA Jr, Hayes SN, Bailey
pressure. Circulation 27:2935 KR, Tajik AJ, Seward JB (1995) Effective mitral
11. Enriquez-Sarano M, Tajik AJ, Schaff HV et al (1994) regurgitant orifice area: clinical use and pitfalls of the
Echocardiographic prediction of left ventricular func- proximal isovelocity surface area method. J Am Coll
tion after correction of mitral regurgitation: results Cardiol 25:703709
and clinical implications. J Am Coll Cardiol 21. Enriquez-Sarano M, Dujardin KS, Tribouilloy CM,
24:15361543 Seward JB, Yoganathan AP, Bailey KR et al (1999)
12. Crawford MH, Souchek J, Oprian CA et al (1990) Determinants of pulmonary venous flow reversal in
Determinants of survival and left ventricular perfor- mitral regurgitation and its usefulness in determining
mance after mitral valve replacement. Department of the severity of regurgitation. Am J Cardiol
Veterans Affairs Cooperative Study on Valvular Heart 83:535541
Disease. Circulation 81:11731181 22. Mauri L et al (2013) 4-year results of a randomized
13. Urabe Y, Mann DL, Kent RL et al (1992) Cellular and controlled trial of percutaneous repair versus surgery
ventricular contractile dysfunction in experimental for mitral regurgitation. J Am Coll Cardiol 62(4):317
canine mitral regurgitation. Circ Res 70:131147 328. doi:10.1016/j.jacc.2013.04.030
14. He S, Fontaine AA, Schwammenthal E, Yoganathan 23. Suri RM, Schaff HV, Dearani JA et al (2006) Survival
AP, Levine RA (1997) Integrated mechanism for advantage and improved durability of mitral repair for
functional mitral regurgitation: leaflet restriction ver- leaflet prolapse subsets in the current era. Ann Thorac
sus coapting force: in vitro studies. Circulation Surg 82:819
96:18261834 24. Enriquez-Sarano M, Akins CW, Vahanian A (2009)
15. Otsuji Y, Handschumacher MD, Schwammenthal E Mitral regurgitation. Lancet 373:13821394
et al (1997) Insights from three-dimensional echocar- 25. Seneviratne B, Moore GA, West PD (1994) Effect of
diography into the mechanism of functional mitral captopril on functional mitral regurgitation in dilated
regurgitation: direct in vivo demonstration of altered heart failure: a randomised double blind placebo con-
leaflet tethering geometry. Circulation 96:19992008 trolled trial. Br Heart J 72:63
16. Enriquez-Sarano M, Tajik AJ, Bailey KR, Seward JB 26. Van Bommel RJ et al (2011) Cardiac resynchroniza-
(1993) Color flow imaging compared with quantita- tion therapy as a therapeutic option in patients with
tive Doppler assessment of severity of mitral regurgi- moderate-severe functional mitral regurgitation and
tation: influence of eccentricity of jet and mechanism high operative risk. Circulation 124:912
of regurgitation. J Am Coll Cardiol 21:12111219 27. Brandt RR, Reiner C, Arnold R et al (2006) Contractile
17. Glick BN, Roberts WC (1992) Usefulness of total response and mitral regurgitation after temporary
12-lead QRS voltage in diagnosing left ventricular interruption of long-term cardiac resynchronization
hypertrophy in clinically isolated, pure, chronic, therapy. Eur Heart J 27:187

ERRNVPHGLFRVRUJ
Prosthetic Valve Dysfunctions
17
Alessia Urbinati, Marco Marchesini,
and Ilaria Mazzanti

17.1 Case Report Physical Examination

Physical examination revealed an extra sound


A 73-year-old man was admitted to the (S3) and a holosystolic murmur at the apex radia-
hospital with dyspnea and fatigue. Past ing toward the left axilla. There were crackles in
medical history revealed a previous inferior both lungs and ankles are swelling. The liver was
myocardial infarction complicated by rup- palpable 1 cm below the right lower costal mar-
ture of the posteromedial papillary muscle gin, in the midclavicular line; the spleen tip was
resulting in severe mitral regurgitation and not palpable below the left costal margin neither
cardiogenic shock. The patient was suc- were the lymph nodes. The patient showed mildly
cessfully treated with a mechanical bileaf- icteric sclerae.
let mitral valve replacement (St. Jude 25)
and concomitant coronary artery bypass
grafting. Since the surgery, the patient was Cardiovascular Risk Factors
regularly on warfarin.
Hypertension
High blood cholesterol
Chronic kidney disease
Vital Signs

Heart rate: 68 bpm EKG on Admission


Blood pressure: 115/70 mmHg
Respiratory rate: 12 breaths per minute Sinus rhythm, normal atrioventricular and intra-
Oxygen saturation: 98 % ventricular conduction, no alterations in
repolarization.

Laboratory Exams on Admission


A. Urbinati (*) M. Marchesini I. Mazzanti
Clinica di Cardiologia e Aritmologia, Universit
Politecnica delle Marche, Complete blood cell count revealed a hemoglo-
Via Conca, 71, Ancona 60126, Italy bin level of 6.7 g/dL (mean corpuscular volume,
e-mail: alessiaurbinati@hotmail.it; marche9584@ 86.9 fL; mean corpuscular hemoglobin, 25.8 pg),
gmail.com; ilaria_mazzanti@libero.it

Springer International Publishing Switzerland 2015 193


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_17

ERRNVPHGLFRVRUJ
194 A. Urbinati et al.

a hematocrit of 22.6 %, white blood cell count of Transesophageal Echocardiography


7.98 109/L, and platelets of 386 109/L. Serum
iron, total iron-binding capacity, and ferritin level Dehiscence of the prosthetic valve. Severe mitral
were 71 ug/dL, 247 ug/dL, and 35 ug/dL, respec- regurgitation. Two physiologic jets due to lavage
tively. Basic metabolic panels were 137 mmol/L volume (Fig. 17.1) and a perivalvular leakage due
for sodium, 3.9 mmol/L for potassium, to dehiscence of the prosthesis (Fig. 17.2). Severe
104 mmol/L for chloride, and 2.36 mg/dL for pulmonary hypertension. Normal left ventricular
creatinine (previous value of 1.6 mg/dl). Serum volume and normal ejection fraction.
bilirubin was 1.57 mg/dL (direct bilirubin,
0.31 mg/dL). Aspartate aminotransferase was
95 IU/L and alanine aminotransferase was Final Diagnosis
24 IU/L. Lactate dehydrogenase (LDH) level was
2006 U/L, and the haptoglobin level was 1 mg/ Acute heart failure caused by severe mitral regur-
dL. The hematological features were suggestive gitation due to dehiscence of the prosthetic valve.
of hemolytic anemia that may occur in patients Hemolytic anemia due to the mechanical disrup-
with prosthetic valve. tion of red blood cells.
The patient has been recommended to undergo
further surgery, but he refused and asked to be
Transthoracic Echocardiography discharged. After 3 weeks, he experienced the
same symptoms and was admitted again at the
While the exam was performed, the patient hospital. Since the early clinical decompensation,
experienced an episode of paroxysmal atrial the patient agreed with the previous plan. He
flutter (ventricular rate approximately underwent a successful mechanical valve
150 bpm). replacement.
Normal left ventricular volume, wall thick- The patient was keen to switch from warfarin
ness, and mass, with normal ejection fraction to one of the new oral anticoagulants (NOACs).
(about 60 %). Normal motion of the moving parts Unfortunately, while NOACs were proved to be
of the mitral prosthesis. No leaflet calcifications more effective and safer than vitamin K antago-
or abnormal echo density attached to the valve. nist in prevention of stroke in patient with non-
Apparently normal valve sewing ring integrity valvular atrial fibrillation, they failed in the
and motion. No occluder movement. Mild mitral setting of valvular atrial fibrillation especially in
regurgitation. Mild aortic regurgitation. Moderate patients with mechanical prosthetic valves and
tricuspid regurgitation. Severe pulmonary hyper- severe mitral stenosis. Warfarin is, at the present
tension (75 mmHg).

Chest X-Ray

Widened vascular pedicle width. Perihilar haze,


septal lines, and accentuated interstitium.
Bilateral pleural effusion.
The patient, after a red blood cell transfusion,
showed a hemoglobin level of 11 g/dl. However,
there was just a mild improvement in the
dyspnea.
A transesophageal echocardiogram was then
performed for a better evaluation of the prosthetic Fig. 17.1 Transesophageal echocardiography showing
valve. two physiologic jets due to lavage volume

ERRNVPHGLFRVRUJ
17 Prosthetic Valve Dysfunctions 195

time, its important to keep in mind that regurgi-


tation of a prosthetic valve can be physiologic.
There are two different types of physiologic
regurgitations: The first is the closure volume,
that is, the volume of blood pulled back by the
closure of the occluders and is confined just after
the valve closure; the second is the lavage volume
that consists in the blood flow that passes between
the occluders and the prosthetic ring throughout
the closing state of the valve and is functional to
the correct washing of the valve elements. A
caged ball valve realizes small and variable jets
Fig. 17.2 Transesophageal echocardiography showing along the whole circumference of the ball. A tilt-
the perivalvular leakage due to dehiscence of the ing disk valve presents jets, one at each side of
prosthesis
the occluder; in the bileaflet valve, we can see
three regurgitant jets: one central and two near
the hinges; making a distinction from perivalvu-
time the only available anticoagulant for patients lar leakages could be very difficult [4].
with prosthetic valvular atrial fibrillation. Since Pathologic intra-valvular regurgitation could
that, no switch was possible for the patient. be secondary to the blockage of an occluder in
opening position due to thrombosis, vegetation,
or pannus, that is, a fibrous overgrowth of the
17.2 Prosthetic Valve endocardial tissue over the valve elements. When
Dysfunctions facing a perivalvular or suspected perivalvular
regurgitation, we are called to distinguish a
Prosthetic valve dysfunctions may be due to lavage volume passing near the hinges of the
regurgitations or obstructions. Sometimes, in occluders from an anomalous regurgitation due
case of high velocity flow rate, it is possible to to an initial prosthetic valve detachment.
encounter hemolytic anemia due to the mechan- Pathologic perivalvular regurgitations are bigger
ical disruption of red blood cells. It ranges from than physiologic ones and show an extensive
a mild level to a critical hemolysis requiring aliasing. Grading criteria are the same of regurgi-
blood transfusions. This happens mainly in tation in native valves although the eccentricity
perivalvular regurgitations, but also in normal and the valve masking phenomenon could make
or accelerated anterograde flow in a mechanical a purely quantitative approach misleading [6].
valve.
There are several types of regurgitations that
must be differentiated. The most important clas- Biologic Prosthetic Valve
sification is between pathologic and physiologic
regurgitations [15]. Regarding biologic prosthetic valve, physiologic
regurgitation rarely occurs 2 years before the
implant, but when present has a central origin, a
Mechanical Prosthetic Valves short length, and low velocities without aliasing.
It is important to bear in mind that biologic stent-
In mechanical prosthetic valves, color Doppler is less valves could present physiologic central
the mainstay in identifying, localizing, and grad- regurgitation also just after the implant.
ing the severity of regurgitation. The knowledge Pathologic regurgitations of different origin,
of the specific model and type of the prosthetic direction, and nature, either intra- or perivalvular,
valve we are facing is mandatory. At the same can occur due to calcific degeneration and

ERRNVPHGLFRVRUJ
196 A. Urbinati et al.

endocarditis that could damage a biologic valve the commissures and at the base of the leaflets,
even more frequently than a native one. reducing the excursion movement.
Prosthetic valve detachment is the most dra- A different scenario is the patient-prosthesis
matic complication that should be monitored mismatch, more frequently viewed with aortic
after the recognition of a true perivalvular regur- stented bioprosthesis, where the increased after-
gitation. It is commonly due to an infective endo- load of the left ventricle is due to a normal func-
carditis, frequently with an early onset within 1 tioning valve that is however undersized in
year from the implant and related to the implant relation to the patients body surface area [7].
itself. The infective process disrupts the sewing This situation occurs mainly in patient with a
ring directly generating a perivalvular regurgita- small native aortic annulus in which it is possible
tion or creating a periprosthetic aneurysm that to lodge only a small prosthesis. The clinical sce-
could be silent until its emptying, when a loss of nario consists of a patient with a valvular stenosis
tissue occurs and the valve anchorage is lost. that is not solved [812].
Beside regurgitation that could range from mod- For patients with prosthetic valves, regardless
erate to severe leading to heart failure manifesta- of the type or model, most recent international
tions, we can see the rocking movement of the guidelines and experts consensus recommend a
valve that is a sign of a more extensive detach- visit, a chest X-ray, a 2D echocardiography
ment with a more urgent indication to surgery. within 4 weeks from the implant in order to
The most dreadful consequence is prosthetic obtain a picture in a moment in which the valve is
valve embolization. thought to work at best, and be able to register
On the opposite side, we can face prosthetic any modifications from this optimal condition.
valve obstruction generating valve stenosis of Then the follow-up consists of an annual cardiol-
different severity [3]. In this setting, knowing the ogy visit, with adjunctive controls with echocar-
exact type (mechanical, single disk, bileaflet or diography in case of symptoms variations or new
biologic, stented/stentless, etc.), model, and cardiac murmur discovery [2, 8]. If any dysfunc-
dimension of the valve is of paramount impor- tion is detected, a 2D echocardiography should
tance because the normal values reported by the be performed every 3 or 6 months in relation to
manufacturers differ in relation to these factors. the severity of the valve dysfunction. On the other
Moreover, it is essential, at every follow-up, to side, to perform an echocardiogram every 12
know the early post-implant gradient values in months, as a routine in a patient with stable clini-
order to avoid an incorrect diagnosis of prosthetic cal conditions, is not recommended [1320].
valve dysfunction. In mechanical valves, the
obstruction could be suspected in the presence of
a gradient higher than expected for the specific
prosthesis. Sometimes the cause is evident such References
as the presence of a voluminous mass (thrombus
or vegetation) occupying space and then reducing 1. Pibarot P, Dumesnil JG (2009) Valvular heart disease:
changing concepts in disease management.
the effective orifice area or blocking an occluder Circulation 119:10341048
in close position; sometimes the cause consists of 2. Vesey JM, Otto CM (2004) Complications of pros-
a small pathologic process that interferes with the thetic heart valves. Curr Cardiol Rep 6:106111
hinges that is not directly visible with 2D echo- 3. Roudaut R et al (2007) Thrombosis of prosthetic heart
valves: diagnosis and therapeutic considerations.
cardiography. In those cases, we must look for an Heart 93:137142
anomalous or incomplete opening of the occlud- 4. Ruel M et al (2004) Late incidence and determinants
ers, although it is often difficult to see without the of reoperation in patients with prosthetic heart valves.
use of fluoroscopy. Eur J Cardiothorac Surg 25:364370
5. ORourke DJ et al (2001) Outcome of mild peripros-
In biologic prosthesis, the main clue of suspi- thetic regurgitation detected by intraoperative trans-
cion is the presence of increased thickness esophageal echocardiography. J Am Coll Cardiol
(>3 mm) and extensive calcification, beginning at 38:163166

ERRNVPHGLFRVRUJ
17 Prosthetic Valve Dysfunctions 197

6. Habets J et al (2012) Imaging of prosthetic heart valve 13. Dumesnil JG et al (1990) Validation and applications
dysfunction. J Am Coll Cardiol Img 5(9):956961 of indexed aortic prosthetic valve areas calculated by
7. Botzenhardt F et al (2005) Hemodynamic perfor- Doppler echocardiography. J Am Coll Cardiol
mance and incidence of patient-prosthesis mismatch 16:637643
of the complete supraannular Perimount Magna bio- 14. Pibarot P, Dumesnil JG (2006) Prosthesis-patient mis-
prosthesis in the aortic position. Thorac Cardiovasc match: definition, clinical impact, and prevention.
Surg 53:226230 Heart 92:10221029
8. Vahanian A et al (2007) Guidelines on the management 15. Jamieson WR et al (2004) Surgical management of
of valvular heart disease: the Task Force on the valvular heart disease 2004. Can J Cardiol 20(Suppl
Management of Valvular Heart Disease of the European E):7E120E
Society of Cardiology. Eur Heart J 28:230268 16. Heras M et al (1995) High risk of thromboemboli
9. Butchart EG et al (2005) Working Groups on Valvular early after bioprosthetic cardiac valve replacement. J
Heart Disease, Thrombosis, and Cardiac Am Coll Cardiol 25:11111119
Rehabilitation and Exercise Physiology, European 17. Davila-Roman VG (2004) et all. Prevalence and
Society of Cardiology. Recommendations for the severity of paravalvular regurgitation in the Artificial
management of patients after heart valve surgery. Eur Valve Endocarditis Reduction Trial (AVERT) echo-
Heart J 26:24632471 cardiography study. J Am Coll Cardiol
10. Pibarot P, Dumesnil JG (2000) Hemodynamic and 44:14671472
clinical impact of prosthesis-patient mismatch in the 18. Akins CW et al (2005) Early and late results of the
aortic valve position and its prevention. J Am Coll surgical correction of cardiac prosthetic paravalvular
Cardiol 36:11311141 leaks. J Heart Valve Dis 14:792799
11. Kunadian B et al (2007) Meta-analysis of valve hemo- 19. Verheugt FWA (2013) The new oral anticoagulants in
dynamics and left ventricular mass regression for atrial fibrillation: an update. Neth Heart J
stentless versus stented aortic valves. Ann Thorac 21:480484
Surg 84:7378 20. Eikelboom JW, Connolly SJ, Brueckmann M (2013)
12. Dumesnil JG et al (1990) Validation and applications Dabigatran versus warfarin in patients with mechani-
of mitral prosthetic valvular areas calculated by cal heart valves. N Engl J Med 369:12061214
Doppler echocardiography. Am J Cardiol
65:14431448

ERRNVPHGLFRVRUJ
Part VI
Rhythm Disorders: Tachyarrhytmias

ERRNVPHGLFRVRUJ
Wide QRS Complex Tachycardias
18
Alessandro Barbarossa, Daniele Contadini,
and Marco Flori

18.1 Case Report between ascending aorta and left anterior


descending artery (LAD).
Unstable angina (UA) 3 years ago. The patient
A 72-year-old man was presented to the ER was subjected to coronary arteriography that
for an episode of palpitations, fatigue, and documented a total occlusion of the sequential
dyspnea lasting more than 6 h. VG to RC and LMA 1 and a stenosis of 95 %
of the VG between aorta and LAD. For this
reason, another CABG surgery consisting of
left internal mammary artery (LIMA) to LAD
Medical History and Cardiovascular was performed.
Risk Factors Benign prostatic hypertrophy.

Arterial hypertension.
Hypercholesterolemia (LDL 98 mg/dl on drug Allergies
therapy).
Stage 3 chronic kidney disease (GFR 40 ml/ None
min).
Previous inferior myocardial infarction
(STEMI) 9 years ago with consequent coro- Social History
nary artery bypass graft (CABG) surgery:
sequential venous graft (VG) between ascend- He never smoked.
ing aorta and right coronary artery (RC) and He never used illicit drugs.
first left marginal artery (LMA1) and VG He has a sedentary lifestyle.

Home medications: pantoprazole 20 mg at


8:00 AM, ramipril 2.5 mg at 8:00 AM and 2.5 mg
A. Barbarossa (*) D. Contadini M. Flori
Clinica di Cardiologia e Aritmologia, at 8:00 PM, metoprolol 50 mg at 8:00 AM and
Universit Politecnica delle Marche, 50 mg at 8:00 PM, aspirin 100 mg at 12:00 AM,
Via Conca 71, Ancona 60126, Italy furosemide 50 mg at 8:00 AM and 25 mg at
e-mail: Alessandro.barbarossa@hotmail.it; 4:00 PM, simvastatin 40 mg at 10:00 PM, tamsu-
daniele.contadini@gmail.com;
Mflori3001@gmail.com losin 0.4 mg at 10:00 PM

Springer International Publishing Switzerland 2015 201


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_18

ERRNVPHGLFRVRUJ
202 A. Barbarossa et al.

Vital Signs at ER Entrance Hepatic function (GOT, GPT, -GT, ALP, total
bilirubin, direct and indirect): normal
Temperature: 36.7 C Renal function: creatinine 1.71 mg/dl, BUN
Heart rate: 120 bpm 25.2 mg/dl
Blood pressure: 105/70 mmHg Electrolytes (Na+, K+, Ca++, Mg++, Cl):
Respiratory rate: 16 breaths/min normal
Oxygen saturation while breathing in ambient Thyroid function (TSH, fT3, fT4): normal
air: 91 % Fasting blood glucose: 131 mg/dl
HbA1C: 7.0 % (53 mmol/mol)
Physical Examination Hs-TnT: 12 pg/ml (highest value)

General: fatigued, sweaty, alert, awake, and


oriented ECG
Head, eye, ear, nose, and throat: normocephalic,
atraumatic, mucous membranes moist, extraoc- A standard 12-lead ECG, at rest, was performed.
ular muscles intact, pupils equally round and The ECG (Fig. 18.1) showed a wide QRS tachy-
reactive to light and accommodation bilaterally, cardia. Heart rate was 120 bpm (RR 500 ms).
bilateral tympanic membrane intact, bilateral QRS axis was about 160. QRS duration was
sclerae anicteric, no conjunctival injection 200 ms. The QRS morphology was right bundle
Neck: supple, no jugular venous distention, no branch block type (Rr in lead V1). P waves
lymphadenopathy, no carotid bruit were not clearly visible. ST segment was not
Cardiovascular: regular rhythm, tachycardia assessable.
rate, S1 and S2 are normal, holosystolic mur- What are the possible types of wide QRS
mur III/VI on Levine scale at the apex and at complex tachycardia?
the anterior axillary area
Lungs: rales only at the pulmonary bases. No Ventricular tachycardia (VT)
rhonchi or wheezes, no egophony, no altera- Supraventricular tachycardia with aberrant AV
tions in tactile fremitus, normal percussion in conduction
remaining areas Atrial fibrillation
Abdomen: flat, no pulsatile masses, normal Atrial flutter
bowel sounds in all four quadrants, no high- Focal atrial tachycardia
pitched or tinkling sounds, resonant to percus- Atrioventricular reciprocating tachycardia
sion, soft, non-distended/non-tender, no (ortho- or antidromic)
rebound or guarding, no costovertebral angle Atrioventricular nodal reciprocating
tenderness, no hepatosplenomegaly tachycardia
Extremities: no cyanosis or clubbing, mild Pre-excited tachycardia (with anterograde
edema at both ankles conduction on the accessory pathway)
Neurologic: cranial nerves I through XII intact, Atrial fibrillation
no focal deficit Atrial flutter
Psychiatric: normal affect, no hallucinations Focal atrial tachycardia
and normal speech
Skin: intact, sweaty, no rushes, no lesion The presence of a regular rhythm excludes the
hypothesis of atrial fibrillation and atrial flutter/
Routine Laboratory Tests tachycardia with variable AV conduction.
In the present ECG, there are not any visible P
Complete blood count: normal (hemoglobin waves (or F wave); the P wave could be inside the
12.9 g/dl) terminal part of the ventricular complex or inside
Inflammatory markers: ESR 46 mm/h, CRP the T wave. So, a clear atrioventricular dissocia-
1.3 mg/dl tion (suggestive for VT) is not demonstrable.

ERRNVPHGLFRVRUJ
18 Wide QRS Complex Tachycardias 203

Fig. 18.1 (ad) Rest ECG showing a wide complex tachycardia

ERRNVPHGLFRVRUJ
204 A. Barbarossa et al.

Moreover, the heart rate (120 bpm) is too slow At echocardiography: mild dilatation of the
for a 2:1 atrial flutter. Even if P wave is not visi- left atrium. Moderate dilatation of the left ventri-
ble, hypothesis of a sinus tachycardia is unlikely cle (iEDV 92 ml/m2). Severe left ventricular sys-
because there were not physiological reasons for tolic dysfunction (EF 40 %). Akinetic and thinned
having an elevated rest heart rate. inferior wall of the left ventricle. Normal dimen-
There are not fusion or capture beats (diagnos- sions of the right ventricular dimension and sys-
tic for VT). tolic function (TAPSE 19 mm). II grade diastolic
There was hemodynamic stability, and there- dysfunction. Moderate mitral regurgitation. Mild
fore we performed carotid sinus massage that did tricuspid regurgitation with normal pulmonary
not modify the tachycardia cycle. The QRS com- arterial pressure. No pericardial effusion.
plexes in the precordial leads were not concor- There were not any clues attributable to acces-
dant: V1 positive and negative from V2 to V6 sory AV pathways or to channelopathies (e.g.,
(concordance is high suggestive of ectopic ven- Brugada syndrome or long and short QT syn-
tricular beats, if negative specifically for VT). We drome). Also, cardiomyopathies like hypertro-
measured the distance from the onset of QRS to phic cardiomyopathy, arrhythmogenic right
the nadir of S wave in a precordial lead (we chose ventricular dysplasia, or non-compacted myocar-
V2), and it is about 120 ms (when this interval is dium were excluded. Finally, the presence of an
more or equal to 100 ms, you should consider inferior wall necrosis was confirmed.
VT). Finally, our ECG analysis ends with QRS Because of the low EF unknown in previous
morphological criteria: we are in front of a controls, we repeated a coronary arteriography
RBBB-type wide QRS tachycardia. In V1 the that showed occlusion of VG between RC and
QRS complex morphology is Rr and in V6 LMA1, occlusion of VG between aorta artery
rS. This morphological pattern is suggestive for and LAD, and a good flow on LIMA to LAD.
VT. The QRS axis at 160 and the QRS duration Amiodarone therapy was started (400 mg/die for
of 200 ms are other adjuvant criteria for VT. the 1st week then 200 mg/die) to prevent recur-
Owing these criteria, we postulated the diag- rences. During hospitalization at ECG monitor, a lot
nosis of ventricular tachycardia. of premature ventricular beats (PVBs), couple of
The patient, after sedation with midazolam PVBs, non-sustained VT (nsVT), and another VT
5 mg IV, was successfully treated with electric (which required cardioversion with DC shock) were
cardioversion (DC shock 200 J) with an immedi- recorded. All these ventricular arrhythmias had the
ate return to sinus rhythm (Fig. 18.2). same QRS morphology of the first VT episode.
At sinus rhythm, the QRS complex had a com- An invasive electrophysiological study (EPS)
plete different morphology. The signs of the pre- was performed and documented:
vious inferior MI were visible. [] easy induction by programmed ventricular
pacing of a ventricular tachycardia coming from
the inferior wall of left ventricle. A catheter abla-
Admission to the Arrhythmology tion of the site of origin of ventricular tachycardia
was performed successfully with no more induc-
Department tion of arrhythmia after programmed ventricular
pacing of the area. []
The patient was then admitted to our cardiology
and arrhythmology department to seek the cause The final diagnosis was ventricular tachycar-
of arrhythmia. dia starting from the inferior wall of the left ven-
Electrolytec imbalance, hyper- or hypothyroid- tricle due to macro-reentrant circuits secondary
ism, and coronary acute syndrome were excluded. to a myocardial scar.
ECG: sinus rhythm, normal AV conduction, VT did not return during hospitalization.
QRS axis +30, Q wave in inferior leads as a pre- One month after discharge, we performed a
vious inferior myocardial infarction, normal 7-day-long ECG-Holter monitoring that did not
repolarization. document any VT recurrence.

ERRNVPHGLFRVRUJ
18 Wide QRS Complex Tachycardias 205

Fig. 18.2 (a, b) ECG performed after electrical cardioversion

ERRNVPHGLFRVRUJ
206 A. Barbarossa et al.

Fig. 18.2 (continued)

ERRNVPHGLFRVRUJ
18 Wide QRS Complex Tachycardias 207

18.2 Ventricular Tachycardia cardia (VT) has to be taken into account. The
mainstay of differential diagnosis is the ECG
Denition itself, but there are other elements that may help
the diagnosis.
VT is a tachycardia that rises from the ventricles
and lasts at least 3 beats. Non-sustained VT ECG in Wide QRS Tachycardias
(nsVT) is a VT that lasts less than 30 s; contrarily,
a VT that lasts longer than 30 s is called sustained
ventricular tachycardia (sVT). Wide QRS complex tachycardias are one
of the most challenging questions of
ECG. A regular wide QRS complex tachy-
Epidemiology and Etiology cardia may represent one of the following
rhythms:
VTs are associated with a structural heart dis-
ease, but can occur also in its absence. Ischemic
heart disease is the most common cause of
VT. During the acute phase of ischemia, a poly- Aberrant atrioventricular conduction of
morphic VT or a ventricular fibrillation (VF) is supraventricular tachycardia (SVT)
the principal cause of sudden cardiac death Ventricular tachycardia (VT)
(SCD). Preexisting right (RBBB) or left (LBBB)
Monomorphic VT is usually a consequence of bundle branch block with SVT
a myocardial scar zone that became a reentrant Anterograde conduction over the bypass
substrate in patients with structural heart disease. tract of atrioventricular connection in
The scars can derive from an old myocardial patients with Wolff-Parkinson-White
infarction but also from nonischemic cardiomy- (WPW) syndrome
opathies, including idiopathic dilated cardiomy-
opathy, hypertrophic cardiomyopathy (HCM),
infiltrative heart disease (e.g., amyloidosis), and
arrhythmogenic right ventricular dysplasia Irregular wide QRS complex tachycar-
(ARVD). dia may represent one of the following
Moreover, some genetic syndromes (chan- rhythms:
nelopathies) are mainly associated with polymor-
phic ventricular arrhythmias: Brugada syndrome,
long and short QT syndromes, and catecholamin-
ergic polymorphic ventricular tachycardia (see Atrial fibrillation with aberrant ventric-
specific chapters). ular conduction
Finally, a VT that occurs in the absence of Atrial fibrillation with ventricular
structural heart disease and genetic conditions is pre-excitation
referred to as idiopathic VT. This form of VTs Torsade de pointes
usually originates from the right ventricular out- Polymorphic VT
flow tract (RVOT) and is associated with good
prognosis [1].

Differentiating ventricular tachycardia from


ECG Diagnostic Criteria other rhythms is of ut most importance for prog-
nosis and therapeutic management. The follow-
In the presence of a tachyarrhythmia with wide ing elements have been suggested for helping the
QRS, a possible diagnosis of ventricular tachy- differential diagnosis [2].

ERRNVPHGLFRVRUJ
208 A. Barbarossa et al.

ECG During Sinus Rhythm Morphology of QRS Complex


The presence of conduction defect during sinus The abovementioned findings are a strong evi-
rhythm in a previous or subsequent ECG and the dence of VT but available only in a small number
comparison with the QRS complex during tachy- of cases. Most of the times, there are no clues for
cardia may guide the diagnosis. Furthermore, the the diagnosis other than the QRS itself. Hence,
presence of PVCs with the same morphology of QRS morphology analysis is of ut most impor-
tachycardia may orientate to VT. If the onset of tance. During aberrant conduction, the ventricle
tachycardia is recorded, the initiation by a prema- is activated through the left or right bundle, and
ture P wave suggests SVT, whereas initiation by VT is suspected when QRS complexes do not
PVC may be either SVT or VT. If the wide QRS resemble typical LBBB or RBBB. The first por-
tachycardia starts with shorter PR interval tion of the QRS complex is called intrinsicoid
between last sinus P and first wide QRS complex, deflection or R wave peak time and represents the
VT should be suspected. conduction through the His bundle. Since aber-
rant conducted impulses follow the His pathway,
Atrioventricular Dissociation During the intrinsicoid deflection remains narrow or less
Tachycardia wide during aberrance (about <0.04 s) but
AV dissociation, when present, is diagnostic for enlarges during VT. Exception may occur
VT. Detection of P waves may be challenging. depending on the underlying heart disease (MI
Notches and irregularities repeated cyclically scar, ventricular remodeling, or drug treatment)
may be the only indication of underlying P and origin of arrhythmias (the closer the arrhyth-
waves. AV dissociation also may be seen in junc- mia is to His bundle, the narrower the intrinsicoid
tional tachycardias with retrograde second- deflection is). Wide QRS tachycardias have been
degree block, but this condition is rare [3]. classically divided in RBBB morphology depend-
Conversely a 1:1 AV correspondence is possible ing on the polarity of the main QRS deflection in
even during VT as 1:1 retrograde conduction lead V1 (positive deflection is RBBB and negative
may be present. deflection is LBBB). Table 18.1 shows the mor-
phology criteria for VT diagnosis, based on intra-
Precordial Concordance cardiac studies [4, 5].
When in all the six standard precordial leads the
QRS complexes have the same polarity (all posi- Diagnostic Elements Other than ECG
tive or all negative), that so-called concordance In diagnosis of wide-complex tachycardias, there
suggests VT diagnosis. Exceptions may occur: a are few elements other than ECG that may help
negative concordance has a higher specificity; a the diagnosis. AV dissociation may be diagnosed
positive concordance may be also observed in during tachycardia with echocardiography: pulse
WPW during conduction through a left lateral Doppler of mitral inflow shows dissociation
pathway. between A wave and QRS complexes. Clinically
AV dissociation is suggested by first-tone inten-
Captures and Fusions sity variability as clinical sign, but this finding
During tachycardia a sinus beat may be con- has poor sensitivity.
ducted to the ventricle, resulting in a normal
(capture) or a hybrid narrower (fusion) QRS Table 18.1 ECG findings indicating VT in wide complex
complex. This finding is a strong evidence of tachycardia
VT. Exceptionally, fusion beats may be present Lead V1 Lead V6
during aberrant supraventricular tachycardia LBBB R wave >30 ms, onset of Any Q
when in presence of BBB a contralateral PVC morphology QRS to nadir of S wave wave
(i.e., PVC from the right ventricle during RBBB) >60 ms
results in pseudonormalization of the QRS RBBB Monophasic R or rS,
complex. morphology biphasic qR, QR, RS QR, qr

ERRNVPHGLFRVRUJ
18 Wide QRS Complex Tachycardias 209

Response to vagal maneuvers and adenosine Management of VT


may help the diagnosis. The arrhythmia interrup-
tion or variation in AV ratio with unmasking of Acute Management
underlying P or flutter waves favors VT. The first step in the management of VTs is to
assess the hemodynamic stability of the patient:
What May Be Misleading
Age should not be considered. Older patients Unstable patient: hemodynamic compromise
have higher probability to have both VT may occur with any VT, regardless of the eti-
and SVT. ology. Furthermore, patients who initially
Heart rate should not be used for absolute appear stable may deteriorate rapidly.
diagnosis, but at a regular frequency of 150 bpm,
atrial flutter with 2:1 VA conduction should be Unstable patients but still responsive and with
suspected. In this setting, vagal maneuvers or an arterial pulse should undergo emergent syn-
adenosine may unmask flutter waves through chronized cardioversion (100200 j in monopha-
transient AV block. sic shock or 50100 j in biphasic shock) with
Although the QRS tends to be wider during uptitration of energy as needed [6]. If the QRS
VT than during SVT, width itself should not be complex and T wave cannot be distinguished
used as a diagnostic element. accurately, a synchronized shock could not be
Last, the hemodynamic tolerance of VT and possible. Such patients should be treated with
SVT relies on several factors and is of most clini- immediate defibrillation (unsynchronized shock
cal importance but is not useful for differential using 360 J in monophasic shock or 200 J in
diagnosis. biphasic shock).
Use of intravenous sedatives may be appropri-
What If the Diagnosis Remains ate but must be balanced against the risks of fur-
Unclear? ther hemodynamic deterioration.
Despite accurate ECG analysis, differentiation Patients who become unresponsive or pulsel-
between VT and SVT is not always clear. The ess should be managed according to ACLS resus-
patient should be treated as VT when the diagno- citation algorithms, with immediate high-energy
sis remains unclear. defibrillation [7].

Stable patients: in hemodynamically stable


patients, additional time can be given for the
Clinical Presentation differential diagnosis between VT and SVT,
and therapy may be targeted to the specific
Clinical symptoms and signs of VT presentation arrhythmia substrates.
may have a wide variability. VT can be classi-
fied as hemodynamically stable and unstable. Ventricular Tachycardia or Uncertain
The patient with a hemodynamically stable VT Diagnosis (Should Be Treated as a VT) [7]
can be totally asymptomatic or contrariwise Elective electrical cardioversion:
show symptoms such as palpitations, dyspnea, Synchronized cardioversion (100200 j in
chest pain, fatigue, dizziness, pre-syncope, and monophasic shock or 50100 j in biphasic
syncope. The hemodynamically unstable VT is shock) with uptitration of energy as
characterized by systolic hypotension and all needed.
the signs of organ hypoperfusion up to pulseless Pharmacological cardioversion [7]:
VT and cardiogenic shock. Another important Procainamide (1015 mg/kg): proved to be
complication of high-rate VT is its degeneration superior to lidocaine (1.5 mg/kg) for termi-
into ventricular fibrillation, a condition that nation of hemodynamically stable mono-
without a rapid treatment can cause patients morphic VT. It can be administered at a rate
death. of 2050 mg/min until the arrhythmia is

ERRNVPHGLFRVRUJ
210 A. Barbarossa et al.

suppressed. It should be avoided in patients In atrial fibrillation, atrial flutter, or atrial


with long QT or heart failure or low EF. tachycardia, a strategy of rate or rhythm control
Sotalol (100 mg IV over 5 min): also more may be chosen according to clinical indications.
effective than lidocaine in patients with
sustained monomorphic VT. It should be Chronic Management
avoided in patients with long QT.
Amiodarone (150 mg IV over 10 min up to Ventricular Tachycardia
1.2 g/24 h) is useful in recurrent monomor- In patients who survived a sudden cardiac arrest/
phic or refractory VTs. VT, the first step is to exclude possible transient
Lidocaine (11.5 mg/kg IV bolus): should reversible causes. In the presence of sign of acute
be considered the second-line antiarrhyth- myocardial ischemia, myocardial revascularization
mic therapy for monomorphic VT and only (PTCA, CABG) may be performed. Myocardial
in ischemic setting. revascularization may be sufficient in patients sur-
viving VF in association with myocardial ischemia
Supraventricular Tachycardia (normal ejection fraction and no history of MI).
Management is similar to an SVT with a normal Instead, sustained monomorphic VT with prior MI
QRS duration. is unlikely to be affected by revascularization [8].
In SVT due to a reentrant circuit: Electrolyte abnormalities should be also excluded
as the cause of the arrhythmias.
Vagal maneuvers may be considered such as In sudden cardiac arrest/VT without remov-
carotid sinus pressure (if no carotid bruits are able cause, randomize controlled trials showed
present) or Valsalva maneuver as the initial ICD superiority compared with antiarrhythmic
intervention. drug therapy in prevention of sudden death [8].
Adenosine (6 mg IV over 12 s): highly effec- 2012 AHA/ACC/HRS guidelines
tive in terminating many SVTs (e.g., AVNRT, recommended:
AVRT), and for others (e.g., AF, atrial flutter),
adenosine may facilitate the diagnosis by slow-
ing the ventricular response to allow clearer ICD therapy is indicated in patients who
assessment of atrial activity. If the initial dose is are survivors of cardiac arrest due to VF or
ineffective, a 12 mg dose may be given and hemodynamically unstable sustained VT
repeated once if necessary. Adenosine has a after evaluation to define the cause of the
very short half-life (less than 10 s), reducing the event and to exclude any completely revers-
risk of an untoward reaction. ible causes. (Class I, Level of Evidence: A)
Calcium channel blockers or beta-blockers: ICD therapy is indicated in patients with
intravenous verapamil (2.55 mg IV) or structural heart disease and spontaneous
beta-blockers (e.g., metoprolol 510 mg IV) sustained VT, whether hemodynami-
may be given if the SVT persists after ade- cally stable or unstable. (Class I, Level
nosine administration. These medications of Evidence: B)
can terminate AVNRT as well as some atrial ICD implantation is reasonable for
tachycardias. If the specific SVT diagnosis patients with sustained VT and normal
remains unknown, these drugs may slow the or near-normal ventricular function.
ventricular response and facilitate diagnosis. (Class IIa, Level of Evidence: C)
Finally, calcium channel blockers and beta- ICD therapy is not indicated for patients
blockers should not be used in AVRT. with ventricular tachyarrhythmias due to
Cardioversion is rarely necessary in patients a completely reversible disorder in the
with a stable SVT. However, if AVNRT or absence of structural heart disease (e.g.,
AVRT persist after the above interventions, electrolyte imbalance, drugs, or trauma).
synchronized cardioversion is usually effec- (Class III, Level of Evidence: B)
tive in restoring sinus rhythm.

ERRNVPHGLFRVRUJ
18 Wide QRS Complex Tachycardias 211

Antiarrhythmic drug therapy may be consid- 7. Neumar RW, Otto CW, Link MS, Kronick SL, Shuster
M, Callaway CW, Kudenchuk PJ, Ornato JP, McNally
ered as the second choice in patients who cannot
B, Silvers SM, Passman RS, White RD, Hess EP, Tang
receive ICD therapy or in patients with recurrent W, Davis D, Sinz E, Morrison LJ (2010) Part 8: adult
VT/VF associated with ICD shocks [9]. advanced cardiovascular life support: 2010 American
Heart Association Guidelines for Cardiopulmonary
Resuscitation and Emergency Cardiovascular Care.
Supraventricular Tachycardia
Circulation [Internet] 122:S729S767. [cited 2013
WCTs due to SVT should be treated as SVT with Nov 6]. Available from: http://www.ncbi.nlm.nih.gov/
narrow QRS complex (see Chap. 19). pubmed/20956224
8. Epstein AE, DiMarco JP, Ellenbogen KA, Estes NAM,
Freedman RA, Gettes LS, Gillinov AM, Gregoratos G,
Hammill SC, Hayes DL, Hlatky MA, Newby LK,
Page RL, Schoenfeld MH, Silka MJ, Stevenson LW,
Sweeney MO, Tracy CM, Darbar D, Dunbar SB,
References Ferguson TB, Karasik PE, Link MS, Marine JE,
Shanker AJ, Stevenson WG, Varosy PD (2013) 2012
1. Koplan BA et al (2009) Ventricular tachycardia and ACCF/AHA/HRS focused update incorporated into
sudden cardiac death. Mayo Clin Proc 84(3):289297 the ACCF/AHA/HRS 2008 guidelines for device-
2. Surawicz B, Knilans T (2008) Chous electrocardiog- based therapy of cardiac rhythm abnormalities: a
raphy in clinical practice, 6th edn, Saunders (elsevier). report of the American College of Cardiology
pp 440455 Foundation/American Heart Association Task Force
3. Bauernfeind RA et al (1978) Retrograde block during on Practice Guide. J Am Coll Cardiol [Internet] 61:e6
dual pathway atrioventricular nodal reentrant paroxys- e75. [cited 2014 Nov 21]. Available from: http://www.
mal tachycardia. Am J Cardiol 42:499 ncbi.nlm.nih.gov/pubmed/23265327
4. Wellens HJ et al (1978) The value of electrocardio- 9. Blomstrom-Lundqvist C, Scheinman MM, Aliot EM
gram in the differential diagnosis of a tachycardia with et al (2003) ACC/AHA/ESC guidelines for the man-
widened QRS complex. Am J Med 64:27 agement of patients with supraventricular arrhythmias:
5. Kindwall K et al (1988) Electrocardiographic criteria executive summary: a report of the American College of
for ventricular tachycardia in wide complex left bun- Cardiology/American Heart Association Task Force on
dle branch block morphology tachycardia. Am J Practice Guidelines and the European Society of
Cardiol 61:1279 Cardiology Committee for Practice Guidelines (Writing
6. Gupta AK, Thakur RK (2001) Wide QRS complex Committee to Develop Guidelines for the Management
tachycardias. Med Clin North Am [Internet] 85:245 of Patients With Supraventricular Arrhythmias) devel-
266, ixx. [cited 2015 Jan 26]. Available from: http:// oped in collaboration with NASPE-Heart Rhythm
www.ncbi.nlm.nih.gov/pubmed/11233948 Society. J Am Coll Cardiol 42:14931531

ERRNVPHGLFRVRUJ
Supraventricular Reentrant
Tachycardias 19
Daniele Contadini and Alessio Menditto

19.1 Case Report Social History

The patient does regular physical activity, smoked


A 61-year-old man presented to the emer- about 30 cigarettes/day until 7 years ago, and
gency room for an episode of malaise char- never used illicit drugs.
acterized by palpitations, sweating,
dizziness, and chest pain that lasted for
more than an hour. Medications

Pantoprazole at 8:00 AM, telmisartan/hydrochlo-


rothiazide 80/12.5 mg at 8:00 AM, aspirin
Medical History and Cardiovascular 100 mg at 12:00 AM, metformin 500 mg at
Risk Factors 12:00 AM and at 8:00 PM, and atorvastatin
20 mg at 10:00 PM
Type 2 diabetes mellitus
Arterial hypertension
Dyslipidemia Vital Signs
Overweight
Post-traumatic subdural hematoma which Temperature: 35.4 C
required neurosurgical drainage about 1 year Heart rate: 200 beats per minute
ago Blood pressure: 115/80 mmHg
Respiratory rate: 18/min
Oxygen saturation while breathing in ambient
Allergies air: 98 %

No allergy is referred by the patient.


Physical Examination
D. Contadini A. Menditto (*)
Clinica di Cardiologia e Aritmologia, General: fatigued, sweaty, alert, awake, and
Universit Politecnica delle Marche, oriented
Conca, Ancona 60100, Italy Head, eye, ear, nose, and throat: normoce-
e-mail: daniele.contadini@gmail.com; phalic, atraumatic, mucous membranes moist,
alessiomenditto@hotmail.it

Springer International Publishing Switzerland 2015 213


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_19

ERRNVPHGLFRVRUJ
214 D. Contadini and A. Menditto

extraocular muscles intact, pupils equally were not clearly visible. ST segment depression
round and reactive to light and accommoda- was present in I, II, aVF, and V3V6 leads. ST seg-
tion bilaterally, bilateral tympanic membrane ment elevation was present in aVR and V1 leads.
intact, bilateral sclera anicteric, and no con-
junctival injection
Neck: supple, no jugular venous distention, no What Are the Possible Types
lymphadenopathy, no carotid bruit of Supraventricular Arrhythmias?
Cardiovascular: regular rhythm, tachycardia
rate, S1 and S2 normal, and no murmurs
Lungs: no rales, rhonchi, or wheezes, no Sinus tachycardia
egophony, no alterations in tactile fremitus, Reentrant supraventricular tachycardias
and normal percussion Atrioventricular reciprocating
Abdomen: overweight, no pulsatile masses, tachycardia
normal bowel sounds in all four quadrants, no Atrioventricular nodal reciprocating
high-pitch or tinkling sounds, resonant to per- tachycardia
cussion, soft, non-distended/non-tender, no Focal atrial tachycardia
rebound or guarding, no costovertebral angle Atrial flutter
tenderness, and no hepatosplenomegaly Atrial fibrillation
Extremities: no cyanosis or clubbing and no
edema
Neurologic: cranial nerves I through XII intact
and no focal deficit The presence of a regular rhythm excludes the
Psychiatric: normal affect, no hallucinations, hypothesis of atrial fibrillation. There isnt a
and normal speech clearly visible P wave, probably because it is
Skin: intact, sweaty, and no rushes, and no lesion inside the terminal part of the ventricular com-
plexes. The atrioventricular ratio is 1:1. Heart
rate (200 bpm) is too low for a 1:1 atrial flutter, so
Routine Laboratory Tests this hypothesis is unlikely. Even if the morphol-
ogy of the P wave is not assessable, we can
Complete blood count: normal (hemoglobin exclude the hypothesis of a sinus tachycardia
13 g/dl) because there were no physiological causes for
Inflammatory markers (ESR, CRP): normal having high heart rate at rest. RP interval was
Hepatic function (GOT, GPT, -GT, ALP, total shorter than PR and longer than 70 ms. The most
bilirubin, direct and indirect): normal likely diagnoses left are therefore atrioventricular
Renal function (creatinine, BUN): normal reciprocating tachycardia, atrioventricular nodal
Electrolytes (Na+, K+, Ca++, Mg++, Cl): reciprocating tachycardia, and atrial tachycardia.
normal Vagal maneuver response may allow to distin-
Thyroid function (TSH, fT3, fT4): normal guish between the different forms of supraven-
Fasting blood glucose: 93 mg/dl tricular tachycardias. Atrial tachycardia generally
HbA1C: 7.0 % (53 mmol/mol) is conducted with a transitory high-grade atrio-
Hs-TnT: 120 pg/ml (highest value) ventricular block. Reentrant supraventricular
tachycardias end suddenly. In this case, the sinus
carotid massage caused a sudden tachycardia
Routine 12-Lead ECG at Rest (Fig. 19.1) interruption (Fig. 19.2).
Therefore, the diagnosis was reentrant supra-
The ECG showed a narrow QRS complex tachy- ventricular tachycardia. The high heart rate
cardia. Heart rate was 200 beats per minute (RR (about 200 bpm) was more suggestive for an
300 ms). The QRS axis is about + 20. P waves

ERRNVPHGLFRVRUJ
19 Supraventricular Reentrant Tachycardias 215

Fig. 19.1 ECG shows a narrow QRS tachycardia

Fig. 19.2 Tachycardia termination during sinus carotid massage

ERRNVPHGLFRVRUJ
216 D. Contadini and A. Menditto

atrioventricular reciprocating tachycardia, but What Is the Meaning of ST Segment


this is not a definite clue. Depression During Supraventricular
The patient was admitted to the cardiology Paroxysmal Tachycardia? Can It
department to perform an invasive electrophysi- Be a Sign of Atherosclerotic Coronary
ological investigation with subsequent catheter Disease?
ablation of the tachycardia.
This patient had a high cardiovascular risk profile
(type 2 diabetes mellitus, arterial hypertension,
Invasive Electrophysiological dyslipidemia, overweight, past smoking status),
Investigation (Fig. 19.3) and he was symptomatic for chest pain during
tachycardia. Echocardiography and a treadmill
[] eccentric type ventriculo-atrial retrograde con- test were performed.
duction. Early retrograde conduction in left postero-
Echo: Normal atrial size. Normal left ventricle
lateral region (CS 56). Induction by programmed
atrial pacing of an orthodromic atrioventricular size and systolic function (EF 60 %). Normal
reciprocating tachycardia (concealed accessory right ventricle size and systolic function (TAPSE
pathway). During tachycardia a catheter ablation of 19 mm). Normal diastolic function. Mild mitral
the posterolateral accessory pathway with termina-
regurgitation. Mild tricuspid regurgitation with
tion of tachycardia was performed. []
normal pulmonary arterial pressure. No pericar-
The final diagnosis was atrioventricular recip- dial effusion.
rocating tachycardia from posterolateral con- The exercise test was interrupted at the end of
cealed accessory pathway. Bruces protocol 5th stage. The patient reached

Fig. 19.3 Electrophysiological study. Endocavity ECG during tachycardia. A atrium, H His, V ventricle

ERRNVPHGLFRVRUJ
19 Supraventricular Reentrant Tachycardias 217

the peak exercise with a heart rate of 154 bpm nodal pathways (dual AV nodal pathways). The
(96 % of theoretical maximum heart rate) and a fast pathway, which conducts more rapidly (PR
systolic blood pressure of 170 mmHg (double interval 100150 ms and AH <220 ms) and has a
product: 26,180). Heart rate and blood pressure long refractory period, appears to be located near
showed a normal behavior. There were no symp- the apex of Kochs triangle. The slow pathway,
toms of ischemic ECG changes. which conducts more slowly (PR interval
We conclude for nonischemic ST alteration. >220 ms) and has a short refractory period,
The abnormal repolarization may be the conse- extends inferoposterior to the compact AV node
quence of a distinct pattern of retrograde atrial tissue and extends along the septal margin of the
activation through accessory pathway. This phe- tricuspid annulus at the level of the coronary
nomenon (ST depression >2 mm) is more fre- sinus. The fast pathway constitutes the normal,
quent in AV reentrant tachycardia than in AV physiological, AV conduction axis [6].
nodal reentrant tachycardia. Indeed, in ortho- AVNRT has been categorized into typical or
dromic AV tachycardia, atrial retrograde activa- atypical. This categorization is based on the ret-
tion, as assessed by intracardiac electrograms, rograde limb of the circuit: if it is the fast path-
occurs during the ST segment on the surface way, it is defined typical; if it is the slow pathway,
ECG. In contrast, in AV nodal reentrant tachycar- it is atypical. Typical atrioventricular nodal reen-
dia, the retrograde atrial activation is most fre- trant tachycardia is the more common type
quently simultaneous with the QRS complex and (90 %) and involves the slow pathway for ante-
therefore does not interfere with repolarization grade conduction and the fast pathway for retro-
[14]. grade conduction (slow-fast variant). Atypical
atrioventricular nodal reentrant tachycardia is
less common (10 %) and includes fast-slow and
19.2 Atrioventricular Nodal slow-slow variants [6].
Reentrant Tachycardia An atrial premature complex that is blocked in
(AVNRT) the fast pathway and is conducted through the
slow pathway generally initiates typical
Denition and Epidemiology AVNRT. If the fast pathway has recovered excit-
ability, the impulse can conduct retrogradely over
Atrioventricular nodal reentrant tachycardia is the fast pathway, resulting in an atrial echo. If the
the most common form of regular paroxysmal impulse reenters into the slow pathway, typical
supraventricular tachycardia. Although it was AVNRT may be initiated. Seldom it is initiated
classically described more often in the young and by a ventricular premature complex (Fig. 19.4).
women, it can be detected at any age and sex. The Atypical AVNRT (fast-slow variant) initiates
overall prevalence of atrioventricular nodal reen- with a ventricular premature complex that is
trant tachycardia is 23 cases per 1000 persons. blocked in the fast pathway. The impulse then
Atrioventricular nodal reentrant tachycardia is conducts to the atrium via the slow pathway and
not usually associated with structural heart dis- returns to the ventricle via the fast pathway.
ease [57]. Seldom an atrial premature complex initiates
tachycardia (Fig. 19.5).
In AVNRT, neither the atrium nor the ventricle
Physiopathology and Classication has a critical role in the reentrant circuit [6].

The reentrant circuit is confined not only in the


compact AV node, but the perinodal atrial tissue Clinical Presentation and Diagnosis
has an important role too. Atrioventricular nodal
reentrant tachycardia is related to the presence of AVNRT usually has a sudden onset and termina-
two functionally and anatomically distinct AV tion, without a warm-up period. Its symptoms

ERRNVPHGLFRVRUJ
218 D. Contadini and A. Menditto

Atrium S
E
Slow Fast

SP
AV node FP

Ventricle

Fig. 19.4 Typical AVNRT (slow-fast) mechanism. FP fast pathway, SP slow pathway, S sinus beat, E extrasystoles

Atrium S

Slow Fast

SP
AV node FP

Ventricle E

Fig. 19.5 Atypical AVNRT (fast-slow) mechanism. FP fast pathway, SP slow pathway, S sinus beat, E extrasystoles

include palpitations, neck pulsations, dizziness, way so the atrium and ventricle are activated
and in some cases chest pain or dyspnea. simultaneously during tachycardia; the retro-
Tachycardia can last a few minutes or hours, and grade P wave is either hidden within the QRS or
the termination can be spontaneous or not. embedded in the terminal portion of the QRS,
Sometimes precipitating factors (drugs, physical resulting in RP < PR (usually RP is less or equal
and psychological stress, menstruation, anemia, to 70 ms) with pseudo-r waves in lead V1 and
hyperthyroidism, etc.) can be found [57]. pseudo-s waves in inferior leads. Comparison
A 12-lead ECG is fundamental for the diagno- with the QRS in sinus rhythm can help in identi-
sis. ECG, during AVNRT, shows in most cases a fying pseudo-r and pseudo-s waves. In atypical
regular narrow QRS complex tachycardia; some- AVNRT, the conduction to the atrium is through
times the QRS complex can be wide with the the slow pathway; the atrium is activated late
typical bundle branch block morphology (LBBB relative to the ventricle, so the retroconducted P
or RBBB). Rate of tachycardia is more often wave is away from the previous QRS and it is
between 140 and 250 per minute. The AV rela- near the following QRS (RP > PR) [6].
tionship is practically in all cases 1:1. Atrial acti- Invasive electrophysiological investigation
vation is concentric type because the (EPS) allows to detect the presence of dual AV
retroconducted stimulus comes from the AV nodal pathways, to induce and identify AVNRT
node, so the retroconducted P wave is narrow and (typical or atypical), and to perform catheter
negative in inferior leads. In typical AVNRT, the ablation of tachycardia. Dual pathway can be
impulse conducts to the atrium via the fast path- demonstrated with delivery of a premature atrial

ERRNVPHGLFRVRUJ
19 Supraventricular Reentrant Tachycardias 219

complex that results in a jump in AH conduc- QRS complex, and orthodromic AVRT ends
tion curve (defined as an increase >50 ms in AH with a retroconducted by accessory pathway P
interval with a 10 ms increase in atrial extrastim- wave [5, 6].
ulus prematurity). This jump is due to ante-
grade block in the fast pathway with the
conduction through the slow pathway. The elec- Treatment
trophysiological characteristic of AVNRT is the
concentric retrograde activation of the atrium, Acute treatment: patients with hemodynamic
with a short ventriculoatrial (VA) interval or with compromise should be electrically cardioverted
a A on V (simultaneous activation of the atrium without delay. Usually, AVNRT does not cause
and ventricle) in typical AVNRT or with a long significant hemodynamic compromise. Vagal
VA time in atypical (fast-slow) AVNRT [6]. maneuvers (Valsalva maneuver, dive reflex,
carotid sinus massage) can be used to terminate
the tachycardia. These maneuvers increase the
Differential Diagnosis vagal tone resulting in slow pathway conduction
blockage. If vagal maneuvers are ineffective,
Surface ECG can make differential diagnosis medications known to prolong refractoriness of
between AVNRT and the other narrow QRS com- the slow pathway of the AVNRT circuit can be
plex tachycardia during tachycardia, by the used. These agents include adenosine, calcium
induction and termination of tachycardia and by channel blocker (verapamil or diltiazem), and
the response to vagal maneuvers [5]. beta-blockers (metoprolol, atenolol, propranolol,
ECG features to assess are shown in Fig. 19.6. esmolol). Intravenous administration of these
The response to vagal maneuvers or to ade- drugs should be performed with continuous ECG
nosine can help to distinguish between the differ- monitoring. These agents are very effective in
ent types of supraventricular tachycardia. Gradual terminating the AVNRT [5, 6].
slowing then reacceleration of rate is typical of Long-term treatment: in patients with recur-
sinus tachycardia or focal atrial tachycardia. rent sustained episodes of AVNRT, we have two
Sudden termination is typical of AVNRT and strategies of long-term treatment, catheter abla-
AVRT. Persisting tachycardia with transient high tion and oral drug therapy. For oral therapy, the
grade of AV block is typical of atrial flutter or first choice includes AV nodal blocking agents
atrial tachycardia [5]. like nondihydropyridine calcium channel block-
Termination of tachycardia is due to block in ers (verapamil, diltiazem) or beta-blockers. The
slow pathway in AVNRT and in AV node in use of other antiarrhythmic agents like class IC
AVRT. Thus, typical AVNRT ends with a retro- (flecainide, propafenone) or class III (sotalol,
conducted P wave, atypical AVNRT ends with a amiodarone) is not common for treating AVNRT

1st: assess RR RR regular RR irregular

2nd: assess P wave P not


and P/QRS ratio P: QRS = 1:1 P:QRS>1 no P waves F waves Different P waves
visible

RP<PR
3rd: assess RP and PR RP>PR
RP<70ms RP>70ms
ST; AT; Aflu with
4th: most probable diagnosis atypical Aflu; AT AF different AV
Typical AVNRT AVRT; AT MAT
AVNRT; conduction
PJRT

Fig. 19.6 ECG features to assess. AVNRT atrioventricu- tachycardia, PJRT permanent junctional reciprocating
lar node reentrant tachycardia, AVRT atrioventricular tachycardia, Aflu atrial flutter, AF atrial fibrillation, MAT
reentrant tachycardia, AT atrial tachycardia, ST sinus multifocal atrial tachycardia

ERRNVPHGLFRVRUJ
220 D. Contadini and A. Menditto

today; these are used only when the AV nodal syndrome. The expression WPW pattern defines
blocking agents have failed to prevent recurrence the ventricular preexcitation in the absence of
of tachycardia and patient refuses catheter abla- tachycardias symptoms and signs [5, 7].
tion. Radiofrequency catheter ablation is the non- The prevalence in the general population is
pharmacologic treatment option. Success rates about 13 per 1000 people. Men have an higher
for this technique are approximately 90 %. The incidence of ventricular preexcitation than
major procedural risk is the AV complete block. women (M/F = 2.2:1), and there is a higher inci-
The ablation of slow pathway, rather than the dence of multiple accessory pathways in men,
ablation of the fast pathway, reduces the risk of too. Some cases of WPW are inherited. Parents
AV complete block (1 % vs 8 %) and results in who have accessory pathways may pass them
the absence of hemodynamic consequences of onto their children. The incidence of preexcita-
marked prolongation of PR interval that derives tion in first-degree relatives could be as high as
from fast pathway ablation. Therefore, slow path- 5.5 per 1000 people. In a recent clinical study, a
way ablation represents the first choice today [5]. mutation to the gene that codifies for PRKAG2
(-2 regulatory subunit of AMP-activated protein
kinase) was identified to be responsible for a syn-
Prognosis drome associated with ventricular preexcitation
and early onset of atrial fibrillation and conduc-
Atrioventricular nodal reentrant tachycardia has tion disease. About 720 % of patients with
a good prognosis, and it is not usually associated WPW also have congenital defects within the
with structural heart disease. In some patient, heart [57].
AVNRT coexists with atrial fibrillation, and
recent reports suggest that it may serve as a trig-
ger for atrial fibrillation and its eliminations can Accessory Pathways Anatomy
help to treat atrial fibrillation [6].
The anatomical substrate of ventricular preex-
citation is an accessory pathway. There are four
19.3 Ventricular Preexcitation different types of them. The most common acces-
and Atrioventricular sory pathway is the bundle of Kent (more of 96 %
Reentrant Tachycardia of cases). This is an atrioventricular bypass: the
(AVRT) electrical impulse from the atria directly reaches
the ventricles. The bundle of Kent is made of
Denition and Epidemiology common myocardial cells. They are sodium-
dependent cells, and for this reason, they have
Ventricular preexcitation is a congenital alteration a faster conduction of the electrical impulse than
of the normal conduction of the electrical impulse the specialized cells of the heart conduction sys-
in the heart. In this pathology, an accessory tem that are calcium dependent. The electrical
pathway is present through which the electrical conduction through the bundle of Kent is often
impulse can activate an area of the myocardium frequency-independent and occurs with an all-or-
more rapidly bypassing the normal conduction nothing mode (non-decremental conduction). It
system. In 1930, Louis Wolff, John Parkinson, means that an electrical impulse can be conducted
and Paul Dudley White described a series of through the accessory pathway or blocked, and
young patients who experienced paroxysms of when conducted, it isnt slowed down. The bundle
tachycardia and had characteristic abnormali- of Kent can conduct the electrical impulse in dif-
ties on electrocardiography (ECG) of ventricular ferent ways: both directions (anterograde and ret-
preexcitation. When ventricular preexcitation is rograde, about 68 % of cases), only in retrograde
associated to symptoms and signs of tachycardias, direction (from ventricles to atria, about 21 % of
it is defined as Wolff-Parkinson-White (WPW) cases), or only in anterograde direction (from atria

ERRNVPHGLFRVRUJ
19 Supraventricular Reentrant Tachycardias 221

to ventricles, about 11 % of cases). In addition to normal pathways. Indeed, a larger preexcita-


the bundle of Kent, there are three other types of tion will be the consequence of a rapid con-
accessory pathways grouped under the name of duction through the accessory pathways. This
fibers of Mahaim: atriofascicular tract (from fact depends on the anatomical position of the
atria to one of the bundle branches, sometimes also accessory pathway: one, near to the sinus-atrial
called Brechenmacher tract), nodoventricular tract node thus achievable rapidly from the electri-
(from AV node to one of the bundle branches or cal impulse, will show a large preexcitation;
to ventricles), and fasciculoventricular tract (from another, far from the sinus-atrial node thus
the bundle of His or one of the bundle branches). achievable slowly from the electrical impulse,
Finally, there are some rare cases where multiple will show a little preexcitation.
accessory pathways can be found [8, 9]. 4. Alterations of repolarization: secondary ST
segment and T-wave changes. The altered
sequence of ventricular activation in patients
Ventricular Preexcitations ECG with the WPW pattern results in secondary
Pattern repolarization abnormalities. Most commonly,
the direction of the ST segment displacement
The WPW ECG pattern with full preexcitation and the T-wave polarity is opposite to the
contains the following elements: direction of the delta wave and the major
deflection of the QRS complex. In some cases,
1. Short PR: a PR interval equal or less than 0.12 any abnormalities are seen. The alteration of
s, with a normal P wave. When the electrical repolarization can persist after disappearance
activation from the atria reaches the ventricles of preexcitation, too. This is the phenomenon
through the bundle of Kent (by a faster path- of the heart electrical memory. They com-
way), as a consequence, the PR interval pletely regress within several weeks after abla-
reduces its length. tion of the accessory pathway [8, 9] (Fig. 19.7).
2. Wide QRS complex: abnormally wide QRS
complex with a duration of 0.11 s or more.
The QRS complex represents a fusions beat Classication
made of two wavefronts: one from the normal
pathway and the other from the accessory. Ventricular preexcitation can be classified by the
Each of them depolarizes a portion of the ven- direction (anterograde, retrograde, or both) of the
tricular mass. accessory pathways and by the presence or
3. Delta wave: the presence of initial slurring of absence of signs of WPW on the ECG:
the QRS complex. This is the most important
finding in WPW pattern. Delta wave is in the Manifest preexcitation: the basal ECG shows
initial part of the QRS complex and is visible the WPW pattern. The direction of the con-
as a very slow initial deflection. The duration duction through the accessory pathways must
of the delta wave varies between 0.02 and be necessarily anterograde, but can be possi-
0.07 s. Theoretically, the delta wave should be bly retrograde conduction too.
present in all leads, but it may become isoelec- Not manifest preexcitation: the basal ECG
tric and be easily overlooked in the leads with doesnt show the WPW pattern. The direction
the lead axis, which is nearly perpendicular to of the conduction through the accessory path-
the initial QRS forces. The end of delta wave ways must be necessarily anterograde, but can
joins to the QRS complex (sometimes a little be possibly retrograde conduction too.
notch can be found between the end of the Concealed preexcitation: the basal ECG
delta wave and the QRS complex). The entity doesnt show the WPW pattern. The direction
of the preexcitation depends on the difference of the conduction through the accessory path-
of the times of conduction of the accessory and ways must be only retrograde [8, 9].

ERRNVPHGLFRVRUJ
222 D. Contadini and A. Menditto

Fig. 19.7 ECG WPW pattern.


Short PR interval, delta wave,
wide QRS complex, ST-T
alteration

Localization of Accessory Pathway myocardium in the anterograde direction,


Using ECG returning to the atrium via accessory pathway
in the retrograde direction. In this arrhythmia,
Sometimes with the standard 12-lead ECG, it is the delta wave is not visible during the tachy-
possible to identify the position of the accessory cardia, and the duration of QRS complex is
pathway on the patients heart. An almost correct normal (narrow). The average heart rate is
localization can be reached only when delta 140250 beats/min. Tachycardia is often initi-
wave is visible on the ECG. A perfect localiza- ated by premature atrial or ventricular com-
tion can be made only with EPS. The possible plexes. Diagnostic criteria on the ECG are (1)
localization of accessory pathway and their char- not sinus P wave (negative in DI), (2) PR lon-
acteristics on the ECG is listed in Table 19.1 ger than RP, (3) P wave after the QRS com-
(Fig. 19.8a, b). plex (at least after 0.07 s), (4) occasional
occurrence of electrical alternation of differ-
ent voltages of QRS complex, and (5) P/QRS
Physiopathology of Arrhythmias ratio always 1:1; an AV block during tachycar-
dia rules out the AV reentrant one (Fig. 19.9).
Paroxysmal tachycardias are the most important Antidromic atrioventricular reentrant tachy-
clinical manifestations in patients with WPW cardia: the tachycardia is based on an electri-
syndrome. They were recorded in about 47 % of cal loop made up of the atrium, accessory
patients with the WPW pattern. The possible pathway, and ventricular myocardium in the
arrhythmias in the patient with an accessory path- anterograde direction, returning to the atrium
way are: via the His-Purkinje system and AV node. In
this arrhythmia, the QRS complex is fully
Orthodromic atrioventricular reentrant tachy- preexcited and completely made of delta
cardia: the tachycardia is based on an electri- wave. The duration of the QRS complex is
cal loop made up of the atrium, AV node, increased (wide complex tachycardia).
His-Purkinje system, and ventricular Tachycardia is often initiated by premature

ERRNVPHGLFRVRUJ
19 Supraventricular Reentrant Tachycardias 223

Table 19.1 Main accessory pathway ECG features


Delta waves Delta wave in Delta wave in Transition on
axis QRSs axis V1 V6 precordial leads
Lateral left +90 to +150 +90 to +150 + V1
Posterolateral 150 to 90 150 to 90 + + V1
Lateral right 30 to +45 +45 to 45 + V3V4
Anteroseptal +30 to +90 0 to +90 + V3V4
Posteroseptal left 30 to 90 30 to 90 + + V1
Posteroseptal right 30 to 90 30 to 90 + V2

a b

Fig. 19.8 Accessory pathway axis. Peripheral leads (a) and precordial leads (b). LL lateral left, PostL posterolateral, P
posteroseptal, PL posteroseptal left, PR posteroseptal right, LR lateral right, A anteroseptal

Atrium S
AP E

NAV
HIS bundle
AP H

Ventricle

Fig. 19.9 Orthodromic AVRT mechanism. NAV atrioventricular node, AP accessory pathway, S sinus beat, E extrasys-
toles, H His bundle

atrial or ventricular complexes. Diagnostic not sinus P wave, when visible; and (4) P/
criteria on the ECG are (1) wide QRS regular QRS ratio always 1:1; an AV block during
tachycardia; (2) QRS morphology identical tachycardia rules out the AV reentrant one
to that of the fully preexcited complex; (3) (Fig. 19.10).

ERRNVPHGLFRVRUJ
224 D. Contadini and A. Menditto

S
Atrium
E
AP
NAV
HIS bundle AP H

Ventricle

Fig. 19.10 Antidromic AVRT mechanism. NAV atrioventricular node, AP accessory pathway, S sinus beat, E extrasys-
toles, H His bundle

Preexcited atrial fibrillation/atrial flutter: Clinical Presentation


atrial fibrillation and flutter are less common
than AV reentrant tachycardia in the WPW Patients with WPW can remain completely
syndrome. Atrial impulses during atrial fibril- asymptomatic, and the preexcitation can be dis-
lation and atrial flutter are conducted to the covered with an ECG made for other reason.
ventricles through the accessory pathway. Usually, patients become symptomatic when par-
The result is a wide complex tachycardia with oxysmal arrhythmias occur. In this case, common
irregular RR in the case of atrial fibrillation symptoms are palpitations, fatigue, lightheaded-
and regular in the case of atrial flutter. This ness, chest discomfort, dyspnea, presyncope, or
tachycardia may be as rapid as 220360 syncope.
beats/min. This high rate is possible in the Arrhythmic episodes usually have sudden
presence of a short, effective refractory period onset and termination. The episode may have dif-
of the accessory bundle. Because of the rapid ferent duration, from few minutes to several
ventricular response during atrial fibrillation, hours. It is important to investigate all the symp-
patients with the WPW syndrome may toms, number of episodes, duration, frequency,
develop ventricular fibrillation that results in mode of onset, and possible triggers [5, 7, 10].
sudden death. In the case of atrial flutter, the
AV conduction ratio may be 2:1 or 1:1
(Fig. 19.11). Diagnosis and Differential Diagnosis
Permanent junctional reciprocating tachycardia
(Coumel type): the tachycardia is based on an The diagnosis can be suggested in the presence of
electrical loop made up of the atrium, AV node, symptoms of the paroxysmal arrhythmias
His-Purkinje system, and ventricular myocar- referred by the patient (previously discussed).
dium in the anterograde direction, returning to The 12-element ECG is crucial for the diagnosis.
the atrium via accessory pathway in the retro- Indeed, in case of preexcitation, the ECG shows
grade direction. In this case, the accessory the typical features: short PR, wide QRS com-
pathway has retrograde decremental conduc- plex, delta wave, and alterations of repolariza-
tion properties. The accessory pathway is tion. In the case of strong clinical suspect, an EPS
located near to the His bundle (para-Hisian). It is recommended in order to identify the acces-
is usually a long-lasting tachycardia [5, 6, 8, 9]. sory pathway and/or to evocate possible arrhyth-

ERRNVPHGLFRVRUJ
19 Supraventricular Reentrant Tachycardias 225

AF
Atrium
AP
H
NAV AP
HIS bundle

Ventricle

Fig. 19.11 Preexcited atrial fibrillation mechanism. NAV atrioventricular node, AP accessory pathway, AF atrial fibril-
lation, H His bundle

mias. Invasive EPS allows overstimulating the In fact a P wave at the end of the tachycardia is sug-
atria to identify if there is another way (in addi- gestive for AVRT or AVNRT. Continuation of
tion to the AV node) for the electrical impulse to tachycardia with AV block rules out AVRT. In case
reach the ventricles and overstimulating the all the maneuvers for the termination of the tachy-
ventricles to identify if there is another way to cardia are not effective, electrical cardioversion
reach the atria. In addition, studying the first acti- should be considered. In patients with a regular
vated and the last activated area is possible to wide QRS complex tachycardia, if a differential
localize exactly the position of the accessory diagnosis of ventricular tachycardia (VT) is
pathway. In some cases, it is possible to induce unclear, the arrhythmia should be treated as VT. In
tachycardia, too [57, 10]. case of preexcited tachycardias, AV nodal blocking
The differential diagnosis has already been agents should be used cautiously. It is preferable to
discussed in the AVNRT paragraph. treat preexcited AF with IV ibutilide, flecainide, or
procainamide.
Chronic treatment: in patients with WPW
Treatment syndrome, the first-line therapy is catheter abla-
tion of the accessory pathway. This approach is
Acute treatment: if the patient, during the arrhyth- recommended in patients with preexcitation and
mia, is hemodynamically unstable, an immediate symptomatic arrhythmias, mostly in the case of
electrical cardioversion is mandatory. In the case of preexcited tachycardias or when the arrhythmic
a regular narrow QRS complex tachycardia, in a episodes are frequent or poorly tolerated. In
hemodynamically stable patient, it is important to these patients, when catheter ablation is not pos-
do vagal maneuvers (Valsalva, carotid sinus mas- sible, a prophylactic antiarrhythmic therapy
sage, and facial immersion) to terminate the should be considered. The advised drugs are fle-
arrhythmia. If these fail, IV antiarrhythmic drugs cainide or propafenone or sotalol or amiodarone.
should be administered. Adenosine or nondihydro- AV-blockers (nondihydropyridine calcium chan-
pyridine calcium channel antagonists are the drugs nel antagonists or beta-blockers) should not be
of choice (the first one should be avoided in patients used as single therapy in patients with preexcited
with severe bronchial asthma). Second-choice tachycardia (especially AF) because there is con-
drugs are IV calcium channel blockers or beta- crete risk to favor the conduction through the
blockers. An ECG should be recorded during the accessory pathway. In case of rare episodes of
maneuvers for the termination of the tachycardia. AVRT without preexcitation or in asymptomatic

ERRNVPHGLFRVRUJ
226 D. Contadini and A. Menditto

patients with preexcitation, catheter ablation 3. Nelson SD, Kou WH, Annesley T et al (1988)
Significance of ST segment depression during parox-
should be considered. In such cases, patients
ysmal supraventricular tachycardia. J Am Coll Cardiol
choice should be taken into consideration. In this 12(2):383387
case, a pharmacological pill in the pocket 4. Rivera S, Conde D et al (2014) The retrograde P-wave
therapy (taking an antiarrhythmic drug only at theory: explaining ST segment depression in supra-
ventricular tachycardia by retrograde AV node con-
the onset of a tachycardia episode) could be
duction. Pacing Clin Electrophysiol 37:11001105
made with verapamil, diltiazem, or beta-blocker. 5. Blomstrom-Lundqvist C, Scheinman MM, Aliot EM
Sometimes none therapy could be another strat- et al (2003) ACC/AHA/ESC guidelines for the man-
egy, too [5, 6, 10]. agement of patients with supraventricular arrhyth-
miasexecutive summary: a report of the American
College of Cardiology/American Heart Association
Task Force on Practice Guidelines and the European
Prognosis Society of Cardiology Committee for Practice
Guidelines (writing committee to develop guidelines
for the management of patients with supraventricular
The risk of sudden cardiac death due to the pres-
arrhythmias). Circulation 108:18711909
ence of accessory pathway has been estimated to 6. Lee KW, Badhwar N, Scheinman MM (2008)
range from 0.15 to 0.39 % over 310 years fol- Supraventricular tachycardia part I. Curr Probl
low-up. If the risk of sudden cardiac death after Cardiol 33:467546
7. Bonow RO, Mann DL, Zipes DP, Libby P (2012)
accessory pathway ablation is the same or less is
Braunwalds heart disease: a textbook of cardiovascu-
debated yet [5, 10]. lar medicine, 9th edn. Saunders Elsevier, Philadephia,
pp 799850
8. Surawicz B, Knilans T (2008) Chous electrocardiog-
raphy in clinical practice, 6th edn. Saunders Elsevier,
References Philadelphia, pp 481508
9. Oreto G (2010) Lelettrocardiogramma: un mosaico a
1. Riva S, Della Bella P, Tondo C et al (1996) Value of 12 tessere, 1st edn. Edi. Ermes, Milano, pp 223242
ST segment changes during tachycardia in determin- 10. Cohen MI, Triednman JK et al (2012) PACES/HRS
ing type of narrow QRS complex tachycardia. J Am expert consensus statement on the management of the
Coll Cardiol 27(6):14801485 asymptomatic young patient with a Wolff-Parkinson-
2. Gle S, Ertab F et al (1999) Value of ST-segment white (WPW, ventricular preexcitation) electrocardio-
depression during paroxysmal supraventricular graphic pattern. Heart Rhythm 9(6):10061024
tachycardia in the diagnosis of coronary artery dis-
ease. Am J Cardiol 83:458460

ERRNVPHGLFRVRUJ
Atrial Fibrillation and Related
Disorders 20
Jenny Ricciotti and Alessio Menditto

20.1 Case Report Medications

No home medications
A 51-year-old man was admitted to the
emergency room for exertional dyspnea,
orthopnea, and edemas at lower limbs. He Vital Signs
reported a constrictive chest pain at rest
lasting almost 2 h. He denied any recent Temperature: 36.4 C
episodes of fever or infection diseases. Heart rate: irregular, about 130 beats per
There is negative anamnesis for previ- minutes
ous diseases. Cardiovascular risk factors Blood pressure: 120/95 mmHg
were absent. Respiratory rate: 24/min
Oxygen saturation while breathing in ambient
air: 93 %

Allergies
Physical Examination
No allergy was referred.
General: alert, awake, and oriented
Head, eye, ear, nose, and throat: normoce-
Social History phalic, atraumatic, mucous membranes moist,
extraocular muscles intact, pupils equally
The patient worked in a bakery, never smoked, round and reactive to light and accommoda-
and did not use illicit drugs. tion bilaterally, bilateral tympanic membrane
intact, bilateral sclera anicteric, and no con-
junctival injection
Neck: supple, jugular venous distention, and
no lymphadenopathy
J. Ricciotti A. Menditto (*)
Clinica di Cardiologia e Aritmologia, Cardiovascular: irregular rhythm, about
Universit Politecnica delle Marche, 130 beats per minutes, and no murmurs
Conca, Ancona 60100, Italy Lungs: bilateral rales at medium-basal lung
e-mail: jen.ricciotti@tiscali.it; fields, no rhonchi or wheezes, no egophony,
alessiomenditto@hotmail.it

Springer International Publishing Switzerland 2015 227


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_20

ERRNVPHGLFRVRUJ
228 J. Ricciotti and A. Menditto

no alterations in tactile fremitus, and normal He was treated with a diuretic (furosemide)
upon percussion and rate-control agents (digoxin and metoprolol)
Abdomen: no pulsatile masses, normal bowel with an improvement of symptoms after 24 h.
sounds in all four quadrants, no high-pitched Echocardiography was recorded and showed
or tinkling sounds, resonant on percussion, the following findings: normal-sized left ventri-
soft, non-distended/nontender, no rebound or cle (60 ml/m2) with severe depression of global
guarding, no costovertebral angle tenderness, systolic function (EF less than 20 %) due to dif-
and no hepatosplenomegaly fuse hypokinesia. Severe dilatation of the left
Extremities: no cyanosis or clubbing, with atrium (index volume 40 ml/m2). High filling
lower limb edema pressure. Normal right ventricle size with reduced
Neurologic: cranial nerves I through XII systolic function (TAPSE 12 mm). Moderate dil-
intact, no focal deficit atation of the right atrium. No pericardial effu-
Psychiatric: normal affect, no hallucinations, sion. No gradients. Mild to moderate mitral
and normal speech regurgitation. Moderate to severe tricuspid regur-
Skin: intact, no rashes, and no lesion gitation with high pulmonary arterial pressure
(40 mmHg) (Fig. 20.3).

Routine Laboratory Tests


What Are the Possible Causes of This
Complete blood count: normal (hemoglobin Severe Depression of Left Ventricular
14.8 g/dl) Systolic Function?
Inflammatory markers (ESR, CRP): normal
Hepatic function (GOT, GPT, -GT, ALP, total Possible alternatives:
bilirubin, direct and indirect): normal
Renal function: normal (creatinine 1.2 mg/dl)
Electrolytes (Na+, K+, Ca++, Mg++, Cl): normal Ischemic heart disease
Elevated myocardial necrosis markers (Hs- Idiopathic dilated cardiomyopathy
TnI 4.21 ng/ml, CK-MB 21.7 ng/ml) Myocarditis
Elevated BNP (872 pg/ml) Tachycardiomyopathy
Normocapnic hypoxemia at arterial blood gas
analysis (pH 7.38, PaO2 63 mmHg, PCO2
31 mmHg, HCO3 18 mEq/l) An invasive coronary angiography was per-
formed in order to exclude a coronary disease
(chest pain, elevated myocardial necrosis markers)
Routine 12-Lead ECG at Rest (Fig. 20.1) but didnt show any coronary lesion (Fig. 20.4a, b).
The patient had heart failure and therefore an
ECG showed atrial fibrillation (AF) with high ven- electrical cardioversion was indicated.
tricular rate (130 beats per minute). There were Because AF was not datable, a transesopha-
diffuse and nonspecific repolarization alterations. geal echocardiography (TEE) was done to
exclude atrial thrombi before going to electrical
cardioversion. TEE didnt show atrial thrombi
Chest X-Ray (Fig. 20.2) and a successful electrical cardioversion was
performed. The sinus rhythm was restored after a
A standard chest X-ray demonstrated the pres- single synchronized biphasic shock at
ence of bilateral lung interstitial edema and heart 150 J. Amiodarone was there introduced for
enlargement. rhythm control together with warfarin to prevent
The patient was admitted to the cardiology thromboembolic events (Fig. 20.5).
department with the initial diagnosis of heart An acute myocarditis was also later excluded
failure in patient with AF at high ventricular rate. by magnetic resonance imaging.

ERRNVPHGLFRVRUJ
20 Atrial Fibrillation and Related Disorders 229

Fig. 20.1 ECG shows atrial fibrillation with high ventricular rate

dismissed after 3 days in good clinical state


with the following medical therapy: amioda-
rone 200 mg at 8.00 a.m., losartan 25 mg at
8.00 a.m., furosemide 50 mg at 8.00 a.m. and
at 6.00 p.m., warfarin at 4.00 p.m. (variable
dose to maintain INR from 2.0 to 3.0), eplere-
none 25 mg at 6.00 p.m., and metoprolol 50 mg
at 8.00 a.m. and at 8.00 a.m.
After 1 month, another echocardiogram
was recorded. It showed a great biventricular
systolic function improvement (EF 57 %;
TAPSE 17 mm) and a reduction of mitral and
tricuspid regurgitation (now mild). Also pul-
monary arterial pressure returned to normal.
ECG confirmed sinus rhythm maintenance
(Fig. 20.6a, b).
The full systolic function recovery conse-
Fig. 20.2 Chest X-ray quent of a stable sinus rhythm confirmed the
previous diagnosis of tachycardiomyopathy.
The final diagnosis was severe biventricu- Warfarin was stopped after 1 month from cardio-
lar systolic dysfunction secondary to tachycar- version because patient had not any stroke risk
diomyopathy (high-rate AF). The patient was factor (CHA2DS2-VASc = 0).

ERRNVPHGLFRVRUJ
230 J. Ricciotti and A. Menditto

Fig. 20.3 Echocardiography.


Severe depression of left
ventricular global systolic
function. LVEDV left
ventricular end-diastolic
volume, LVESV left ventricu-
lar end-systolic volume,
LVEF left ventricular ejection
fraction

a b

Fig. 20.4 Coronary angiography. Right coronary angiography (a) and left coronary angiography (b)

Fig. 20.5 Electrical cardioversion. Synchronized biphasic shock at 150 J restores sinus rhythm

ERRNVPHGLFRVRUJ
20 Atrial Fibrillation and Related Disorders 231

Fig. 20.6 Echocardiography. Normal left ventricular global systolic function (a). ECG with sinus rhythm (b)

20.2 Atrial Fibrillation (AF) common sustained arrhythmia in clinical practice


and is the leading cause of hospitalization for
Denition and Epidemiology arrhythmias.
The prevalence of AF in the general popula-
Atrial fibrillation (AF) is a supraventricular tion is about 12 % and increased with age; 70 %
tachyarrhythmia characterized by chaotic and of patients with atrial fibrillation are more than
irregular atrial electrical activity. Its the most 65 years old [1, 2].

ERRNVPHGLFRVRUJ
232 J. Ricciotti and A. Menditto

More than six million in Europe have this episodes of AF long enough (>524 h) are associ-
form of arrhythmia, and its prevalence is expected ated with an increased risk of thromboembolism
to double over the next 50 years with the aging [8, 9].
population. The European Heart Rhythm Association had
proposed a new classification for assessing symp-
toms during AF (EHRA score) [10, 11]: class I,
Classication related to no symptoms; class II mild symptom,
with no impairment of normal daily activities;
AF is classified according to the modality of pre- class III, severe symptoms that affected normal
sentation, its duration and the possibility of resto- daily activity; and class IV, disabling symptoms
ration of sinus rhythm into: with disruption of normal daily activity.
The diagnosis of AF requires an ECG per-
formed during the arrhythmia. ECG provides
1. First diagnosed AF, in patients present- information not only on heart rhythm but also
ing with the arrhythmia for the first time on the presence of ventricular hypertrophy, ven-
2. Paroxysmal AF, in general, self-termi- tricular pre-excitation, bundle branch block,
nating within 48 h, but with a maximum myocardial necrosis, and concomitant
duration up to 1 week arrhythmias.
3. Persistent AF, when an episode of AF Transthoracic echocardiography (TTE) is
lasts more than 7 days or requires car- extremely useful for assessing the existence of an
dioversion to restore sinus rhythm underlying heart disease; it helps to define the
4. Long-standing persistent AF if AF lasts presence of a valvular disease and its severity to
more than a year when an attempt is evaluate chamber dimension and left ventricular
made to restore the rhythm systolic and diastolic function and estimate pul-
5. Permanent AF if it is chronic and no monary pressure.
more attempts to restore normal heart Transesophageal echocardiography (TEE) is
rhythm will be made [3] used to assess the presence of e.g., left atrial
appendage thrombi before cardioversion of non-
datable AF.
Chest X-ray is useful in particular in cases of
Clinical Presentation and Diagnostic AF associated to symptoms of heart failure for
Evaluation the evaluation of the state of pulmonary circula-
tion. It can reveal the presence of concomitant
The most common symptoms in patients with AF AF-associated pulmonary diseases (e.g.,
are palpitations (54 %), which are more common COPD).
in paroxysmal forms, and dyspnea (44 %), which Laboratory tests to be performed are tests for
is prevalent in permanent type. Other common thyroid hormones (TSH, FT4), serum electro-
symptoms are easy fatigability and asthenia lytes, blood counts, and renal and hepatic func-
(14 %), chest pain (10 %), and dizziness and syn- tions that help in choosing antiarrhythmic and
cope (10 %) [4, 5]. anticoagulant drugs.
AF may be also asymptomatic and conse- ECG monitoring system as ambulatory ECG
quently silent; the frequency of silent FA varies Holter or external loop recorder may be used in
depending on the method used for recording, case of palpitations suspicious for paroxysmal
reaching up to 5174 % if considering the memo- AF but without previous electrocardiographic
ries of pacemakers and ICD [6, 7]. documentation of arrhythmia and for the diagno-
The high prevalence of asymptomatic AF has sis of asymptomatic episodes of AF.
implications on the need to start the oral antico- Performing other imaging studies should be
agulant therapy; various trials showed that only evaluated case by case.

ERRNVPHGLFRVRUJ
20 Atrial Fibrillation and Related Disorders 233

Treatment Gault formula, less than 30 ml/min) and in valvu-


lar AF (prosthetic valves and rheumatic valvular
In AF treatment, we have to consider two aspects: disease, in particular mitral stenosis).
(1) prevention of thromboembolism and (2) treat- In patients with important contraindications to
ment of the arrhythmia. lifelong anticoagulant therapy, it is possible to
perform the occlusion of the left atrial appendage
Antithrombotic Therapy (surgical or percutaneous). This is a non-
Patients, during prolonged (more than 48 h) AF pharmacological method to reduce stroke risk.
episode, have to do anticoagulant therapy to The rationale of this technique is that the left
reduce cardio embolic stroke risk. Indication to atrial appendage is the main (but not the only)
lifelong oral anticoagulant therapy in patients site of thrombi formation.
with previous AF episodes (regardless of current
rhythm) is based on a stroke risk score (CHA2DS2- Treatment of Arrhythmia
VASc score). CHA2DS2-VASc score [congestive Acute Therapy: In unstable patient, an immediate
heart failure (1 point), hypertension (1 point), age electrical cardioversion is mandatory. A stable
more than 75 (2 points), diabetes (1 point), previ- patient with a rapid ventricular response needs an
ous stroke or TIA (2 points), vascular disease (1 acute therapy to control the ventricular rate (beta-
point), age more than 65 (1 point), sex category blockers, non-dihydropyridine calcium-channel
female (1 point)] greater than or equal to 1 identi- antagonist, digoxin, or a combination of them).
fies patients at risk of stroke who have indication In case of slow ventricular rate AF, a therapy with
to do lifelong oral anticoagulant therapy. The atropine or temporary pacemaker may be useful
stroke rate in AF patients increases progressively in symptomatic patient.
from 1.3 %/year (CHA2DS2-VASc = 1) to 15.2 %/ If AF is of recent onset (less than 48 h), it is
year (CHA2DS2-VASc = 9). HAS-BLED score possible to try to restore sinus rhythm by electrical
[hypertension (1 point), abnormal renal and liver or pharmacological cardioversion. Electrical car-
functions (1 point each), previous stroke (1 dioversion consists of a synchronized electrical
point), bleeding (1 point), labile INRs (1 point), shock delivery in sedated patients, and it is more
elderly more than 65 years (1 point), drug or effective than pharmacological cardioversion. For
alcohol abuse (1 point each)] is used to assess the pharmacological cardioversion, the choice is
bleeding risk but not to contraindicate the oral between class IC antiarrhythmic drugs (flecainide
anticoagulant therapy. When HAS-BLED score or propafenone for patients without structural
is greater than or equal to three, the bleeding risk heart disease and amiodarone for patient with
is high and a close follow-up of anticoagulated structural heart disease). Class IC antiarrhythmic
patients is necessary. drugs can be administered intravenously or with
The oral anticoagulant therapy can be prac- oral high dose (pill-in-the-pocket approach),
ticed with vitamin K antagonist (warfarin) dosing and their efficacy length is about 24 h.
to obtain INR values from 2.0 to 3.0 or with the Pharmacological cardioversion does not require
new oral anticoagulants (NOAC) (dabigatran, a sedation of patient.
direct factor II inhibitor; apixaban; rivaroxaban; If AF onset is prolonged upto 48 h, the cardio-
and edoxaban, a direct factor X inhibitor). NOAC version can be made only after a TEE that
are fixed-dose drugs (twice daily for dabigatran excludes left atrium or left atrium appendage
and apixaban, once daily for rivaroxaban and thrombi or after at least 3 weeks of correct
edoxaban) that do not require periodical coagula- anticoagulation (INR 2.03.0 or constant intake
tion assessment; neither the dose has to be varied of NOAC). If TEE reveals thrombi, cardioversion
in response to changes in laboratory coagulation is not indicated and a new TEE has to be recorded
parameter (INR or aPTT). NOAC are not indi- after at least 3 weeks of correct anticoagulation
cated in patients with severe renal impairment before cardioversion to assess if thrombi are still
(creatinine clearance, assessed by Cockcroft- present. In this case, long-term oral anticoagulant

ERRNVPHGLFRVRUJ
234 J. Ricciotti and A. Menditto

therapy is indicated regardless of CHA2DS2- persistent AF despite rhythm-control therapy,


VASc score and a rate-control strategy is catheter ablation (usually pulmonary vein isola-
recommended. tion) is an alternative strategy to maintain sinus
In patients with AF more than 48 h, 4 weeks of rhythm or to reduce recurrences. Pulmonary vein
anticoagulation therapy is indicated after cardio- isolation can be made by radiofrequency or by
version independently from CHA2DS2-VASc cryo ablation. Radiofrequency ablation technique
score. is based on the production of several lesion lines
Long-Term Therapy: there are two strategies to separate electrically the left atrium from the
for long-term management of AF, rhythm control pulmonary veins. In cryo ablation technique, the
and rate control. pulmonary vein isolation is obtained by a circular
Rhythm-control strategy is based on prevent- continuous lesion made by a cryo-balloon posi-
ing AF recurrence and maintaining sinus rhythm tioned in the pulmonary vein ostia. The effective-
by the administration of antiarrhythmic drugs. ness of AF ablation to prevent recurrence after
There are different antiarrhythmic drugs used to single procedure is about 6075 % for paroxys-
try to maintain sinus rhythm (flecainide, propafe- mal AF and about 4560 % for persistent AF in
none, sotalol, dronedarone, amiodarone). Class the first year. Multiple procedures increase the
IC antiarrhythmic drugs (flecainide and propafe- effectiveness to about 10 %.
none) can be safely used to maintain sinus The rate-control strategy is based on getting a
rhythm in patient without structural heart dis- normal heart rate during AF to reduce patients
ease. Amiodarone is better than other antiar- symptoms. Drugs used for rate control (beta-
rhythmic drugs to maintain sinus rhythm (the blockers, non-dihydropyridine calcium-channel
number of patients needed to be treated is three antagonist, digoxin, amiodarone, or a combina-
for amiodarone, four for flecainide, five for tion of them) act by reducing the atrioventricular
propafenone, and eight for sotalol), but it is gen- node conduction and consequently the ventricu-
erally used in patients with AF recurrences lar rate. The choice between different drugs has
despite other drug therapies because it has sev- to be made considering patients associated dis-
eral side effects (thyroid dysfunctions, vision eases: in patients with heart failure, the first
problems, liver disease, lung disorder, etc.). choice is beta-blockers, followed by digoxin; in
Dronedarone is less effective than amiodarone, patients with asthma or COPD, non-dihydropyri-
but it can be used in patients experiencing amio- dine calcium-channel antagonist and digoxin are
darone toxicity or side effects. preferred over beta-blockers.
The presence and type of structural heart dis- The rest ventricular frequency target is less
ease are important for choosing among different than or equal to 110 beats per minutes (lenient
antiarrhythmic drugs (Fig. 20.7). rate control). A more intensive rate control (rest
In patients without structural heart disease and heart rate 80/min and heart rate during moder-
recurrent symptomatic episode of paroxysmal or ate exercise 110/min) is necessary when patient

Structural heart disease


HT HF
Minimal or
no heart
CAD NYHA III-IV or
disease Stable
unstable NYHA
(NYHAI-II)
No LVH LVH II

First choice F, P, D, S F, P, D, S D S, D D A
Second choice A A A A A

Fig. 20.7 Antiarrhythmic drugs for rhythm control. HT hypertension, CAD coronary artery disease, HF heart failure,
F flecainide, P propafenone, D dronedarone, S sotalol, A amiodarone

ERRNVPHGLFRVRUJ
20 Atrial Fibrillation and Related Disorders 235

remains symptomatic or if tachycardiomyopathy the other side, the increased filling pressures in
occurs despite lenient rate control. If the rate con- heart failure favor wall stress and atrial dilatation
trol has failed, despite maximum tolerated medi- and fibrosis; the increase in adrenergic tone and
cal therapy, the ablate and pace (radiofrequency neurohumoral activation typical of chronic heart
ablation of atrioventricular node and pacemaker failure determines changes in the electrical sub-
implant) is a non-pharmacological strategy to strate, which are responsible for AF onset.
obtain rate control. Therefore the coexistence of AF and heart fail-
There are no differences in terms of mortality, ure leads to increased mortality and quality of life.
quality of life, and stroke rate between rhythm-
and rate-control strategies; the choice depends on
symptoms, structural heart disease, patients age, AF and Stroke
number of previous episodes, and comorbidity [3].
Stroke is the most important complication in
patients with AF. It is estimated that up to 25 %
20.3 Atrial Fibrillation of strokes in the elderly are a result of AF, which
and Related Disorders increases its risk fivefold [14].
Cardioembolic strokes due to AF are the result
AF is correlated with an increased risk of mortal- of emboli originating from the left atrium and the
ity, ischemic events, and heart failure. left auricle; they are often very large, with a
In the Framingham Heart Study, AF resulted in higher risk of disability [15] and an increased
an increased risk of mortality of 1.5-fold in men poststroke mortality than other types of strokes;
and 1.9-fold in women, independently from the moreover AF is an independent risk factor for
presence of concomitant cardiovascular disease stroke severity and recurrence [16].
[12]. Moreover, AF has a significant impact on the Therefore proper stratification and prevention
quality of life, with a lower score of 1630 % of of thromboembolic risk are a very important part
all the parameters commonly examined. The dete- in the management of AF patients. Actually ESC
rioration of the quality of life in AF patients is AF guidelines recommend to use the CHADS2
similar or even more pronounced than in patients score and more recently the CHA2DS2-VASc
with myocardial infarction or heart failure and score that consider various major and minor clin-
patients undergoing coronary angioplasty [13]. ical risk factor predictors of stroke for evaluating
the indication for anticoagulation treatment (see
Antithrombotic Therapy).
AF and Heart Failure Several randomized clinical trials and meta-
analyses have shown that oral anticoagulation
AF and heart failure are two conditions that often therapy is highly effective in the primary and sec-
coexist. About a third of AF patients have a his- ondary prevention of stroke in patients with AF;
tory of congestive heart failure, and 10 to 30 % of anticoagulation therapy reduced the relative risk
patients with heart failure have a history of of stroke by 64 %, corresponding to an absolute
AF. The prevalence of AF in heart failure increases annual risk reduction in all strokes (2.7 %) [17].
with the progression of the NYHA functional
class (from 4 % in class I to 50 % in class IV).
These two conditions (AF and heart failure) AF and Tachycardiomyopathy
share the same risk factors, and the presence of
one predisposes to the development of the other: Tachycardiomyopathy or tachycardia-induced
the loss of atrial contribution to ventricular fill- cardiomyopathy (TIC) is defined as a condition
ing, the high heart rate, the irregularity of cardiac of systolic and/or diastolic dysfunction second-
cycles, and the reduced duration of diastole dur- ary to chronic or frequent recurrent tachyarrhyth-
ing AF lead to a reduction in cardiac output; in mias. It is characterized by ventricular dilatation

ERRNVPHGLFRVRUJ
236 J. Ricciotti and A. Menditto

and signs and symptoms of heart failure, com- rhythmic drugs. The return to sinus rhythm
pletely or partially reversible after normalization improves cardiac output, ejection fraction, and
of heart rate and/or heart rhythm abnormality. ventricular volumes and also restores AV syn-
TIC may result from various types of chronic chrony and the atrial contribution to ventricular
or frequent paroxysmal supraventricular and ven- filling.
tricular tachyarrhythmias, even if the most fre- In general, the choice of pharmacological
quent etiology is AF. therapy depends on the type of the arrhythmia.
The incidence of TIC is not exactly known but Class IC antiarrhythmic drugs should be used
it can occur at any age. In selected studies of with caution in these patients because of their
patients with AF, based on retrospective series, it negative inotropic effect. Beta-blockers and class
was reported that 25 to 50 % of patients with left III antiarrhythmic drugs (amiodarone, sotalol) are
ventricular dysfunction and AF can have some the drugs of choice in patients with atrial fibrilla-
degree of TIC [18]. tion, ventricular tachycardia, and frequent PVCs.
The time needed to the onset of ventricular However when the medical treatment is not
dysfunction and its extent vary from subject to effective, a non-pharmacological approach must
subject; many variables must be considered as the be considered; these include catheter radiofre-
type of arrhythmia, its rate and duration, the age quency ablation by electrical isolation near the
of the patient, drugs, the presence of pre existing ostia of the pulmonary veins and ablate and pace
heart disease, or other comorbidities [19]. strategy with radiofrequency ablation of the AV
The diagnosis of TIC is often made retrospec- node with pacemaker implantation.
tively, after the recovery of ventricular function The recovery of contractile function may be
once the normalization of cardiac rhythm and/or complete, partial, or totally absent. This can
heart rate is obtained. result from the prolonged duration of the arrhyth-
Diagnostic tests include an EKG that may mia, which causes irreversible histopathological
show the responsible arrhythmia. Chest X-ray alterations, and from the concomitant presence of
can reveal a dilated cardiac silhouette and inter- an underlying heart disease. The time required
stitial edema in the pulmonary circulation. for recovery of left ventricular function is very
Echocardiogram shows dilatation of the left and variable; in experimental animal models, the
right ventricles with systolic and/or diastolic dys- range is about 2 weeks after the end of the cardiac
function and signs of elevated filling pressure; stimulation; in clinical studies, the time neces-
mitral insufficiency secondary to valvular annu- sary for the improvement of ventricular function
lus dilatation is often present. can range from 1 month up to a maximum of 68
Other causes of hypokinetic dilated cardiomy- months.
opathy must be excluded, as idiopathic, ischemic,
and post-myocarditis types. TIC can occur also in
patients with underlying heart disease and worsen AF and Acute Coronary Syndromes
the preexistent systolic function.
The treatment of TIC aims to control heart AF is common in patients with ischemic heart
rate and/or normalization of the rhythm. This can disease, and its a frequent complication in acute
be achieved through non-pharmacological and coronary syndrome (ACS), with a reported inci-
pharmacological approaches. dence of new-onset AF in patients hospitalized
In patients with AF, the initial approach is the for ACS varying from to 2 to 23 % [20].
control of the heart rate, which can be achieved The prevalence of AF in the setting of ACS
with drugs that affect the AV node slowing the reduced in the last years, as a consequence of the
ventricular response (beta-blockers, digitalis, increasing use of revascularization strategies
calcium-channel blockers). (PCI in the acute phase).
Subsequently restoration of sinus rhythm can Several risk factors are associated with inci-
be achieved with DC cardioversion or antiar- dence of AF in ACS patients, including advanced

ERRNVPHGLFRVRUJ
20 Atrial Fibrillation and Related Disorders 237

age, infarct size and severity (ST-segment eleva- have also been postulated as responsible of tropo-
tion myocardial infarction), higher Killip class nin and BNP release during increased heart rates
and left ventricular dysfunction, female gender, [22, 23].
and poorer renal function [21]. Some trials reported that troponin elevation
In the management of patients with AF during may help in the risk stratification in patients with
ACS first of all in case of severe hemodynamic AF. In the biomarker substudies of two recent
instability, if an adequate rate control cant be and larger AF trials, RE-LY (randomized evalua-
obtained with pharmacological therapy, urgent tion of long-term anticoagulant therapy) and
DCC is indicated. In the choice of antiarrhythmic ARISTOTLE (apixaban for reduction in stroke
drugs, patient underlying comorbidities must be and other thromboembolic events in atrial fibril-
considered; current guidelines recommend the lation), including a total of 21,081 patients, ele-
use of amiodarone, sotalol, and dronedarone in vated troponin levels were associated with an
the presence of ischemic heart disease; class I increased risk of stroke or systemic embolism
antiarrhythmic drugs are contraindicated during and of cardiovascular death, independently from
acute ischemia. other clinical characteristics [24, 25].
In AF patients undergoing revascularization
(PCI) during ACS with stent implant, triple ther-
apy (warfarin, aspirin, and clopidogrel) must be AF and Cognitive Dysfunction
administered in the initial period (36 months)
and subsequently replaced by warfarin plus clop- It has suggested that AF may be a risk factor for
idogrel up to 12 months; after that, warfarin can the development of cognitive impairment.
be continued alone. For patients at high bleeding In the prospective, population-based
risk, bare metal stents instead of drug-eluting Rotterdam Scan Study [26], dementia and
stents must be considered to reduce the duration Alzheimers disease were correlated with the
of antiplatelet therapy [3]. presence of AF in the elderly, and a subsequent
In patients with ACS complicated with AF, an substudy [27] revealed an association between
increase in hospital and long-term mortality and silent brain infarcts with the risk of dementia
an increased risk of ischemic stroke were and decline in cognitive function in older patients.
observed; in the large multinational GRACE reg- Other possible causes are concomitant heart fail-
istry, a decline in short-term mortality over the ure and microcirculatory dysfunction, secondary
study period in AF patients (between 2000 and to hypertension.
2007), as a reflex of improvements in ACS ther- In a recent review [28], the incidence of cogni-
apy, was reported. tive impairment and/or dementia in patients with
and without AF was compared. It was observed
that patients with AF had a greater risk of cogni-
AF and Troponin Elevation tive impairment and a 2.3-fold (95 % CI 1.43.7)
increased risk of dementia, compared with
In patients presenting with AF and high heart patients in sinus rhythm.
rate, mild elevation of troponin is frequent, also
in the absence of other signs and symptoms of
ischemic heart disease. AF and Pericarditis
Potential mechanisms of troponin elevation in
AF patients, other than coronary syndromes, are A great number of studies have reported that
related to supply-demand mismatch secondary to inflammation has an important role in AF etiol-
increased heart rate, with shorter diastolic filling ogy. AF is observed between 2 and 3 days after
time, that can lead to a reduction in coronary flow cardiac surgery; this time coincides with the peak
and possible subendocardial ischemia. Increased of inflammatory proteins (C-reactive protein,
left ventricular wall strain and myocardial stretch IL-6). In acute pericarditis, the inflammation of

ERRNVPHGLFRVRUJ
238 J. Ricciotti and A. Menditto

pericardial sack can induce AF. In this case, AF is and the control of heart rate may be difficult in
secondary to inflammation and generally resolves this setting. In case of AF during an acute respira-
after the acute phase [29]. tory decompensation, treatment of underlying
A different mechanism is responsible for the pulmonary disease is the primary target, as resto-
onset of AF in constrictive pericarditis. Altered ration of sinus rhythm may be ineffective until
filling pressure for a long time leads to remodel- respiratory failure is corrected.
ing of heart chamber and atrial dilatation. This
atrial dilatation increases the possibility of AF
onset; about one-third of patients with constric- References
tive pericarditis develop AF [30].
1. Go AS, Hylck EM, Phillips KA et al (2001) Prevalence
of diagnosed atrial fibrillation in adults. National
AF and Vagal Tone implications for rhythm management and stroke pre-
vention: the Anticoagulation and Risk factors In
Atrial fibrillation (ATRIA) study. JAMA 285:
Vagally mediated AF is not a clinical entity, 23702375
with paroxysmal episodes that occur generally 2. Feinberg WM, Blackshear JL, Laupacis A, Kronmal
in healthy young males at night when there is R, Hart RG (1995) Prevalence, age distribution, and
gender of patients with atrial fibrillation. Analysis and
an increase in vagal tone. An increase in vagal
implications. Arch Intern Med 155:469473
tone can induce AF by shortening the atrial 3. Camm AJ, Kirchhof P, Lip GY, Schotten U, Savelieva
refractory period; this can facilitate premature I, Ernst S, Van Gelder IC, Al-Attar N, Hindricks G,
beats and reentry. This type of AF is more fre- Prendergast B, Heidbuchel H, Alfieri O, Angelini A,
Atar D, Colonna P, De Caterina R, De Sutter J, Goette
quent in endurance sports athletes [31]. An AF
A, Gorenek B, Heldal M, Hohloser SH, Kolh P, Le
onset during the night does not imply a vagal Heuzey JY, Ponikowski P, Rutten FH; ESC Committee
AF [32]. for Practice Guidelines, Vahanian A, Auricchio A,
Bax J, Ceconi C, Dean V, Filippatos G, Funck-
Brentano C, Hobbs R, Kearney P, McDonagh T,
Popescu BA, Reiner Z, Sechtem U, Sirnes PA,
AF and Chronic Obstructive Tendera M, Vardas PE, Widimsky (2010) Guidelines
Pulmonary Diseases (COPD) for the management of atrial fibrillation: the Task
Force for the Management of Atrial Fibrillation of the
European Society of Cardiology (ESC). Europace
COPD are associated with a high incidence of
12:13601420
atrial fibrillation and atrial flutter, with a 1015 % 4. Miyasaka Y, Barnes ME, Gersh BJ et al (2006)
of AF patients having COPD; pulmonary dis- Secular trends in incidence of atrial fibrillation in
eases and sleep-disordered breathing are emerg- Olmsted County, Minnesota, 1980 to 2000, and impli-
cations on the projections for future prevalence.
ing risk factors for the development of AF. Patients
Circulation 114:119125
in which the two conditions coexist have a worse 5. Nieuwlaat R, Capucci A, Camm AJ et al; European
prognosis with higher mortality. Heart Survey Investigators (2005) Atrial fibrillation
The presence of hypoxia and hypercapnia management: a prospective survey in ESC member
countries: the Euro Heart Survey on Atrial Fibrillation.
with respiratory acidosis in chronic lung diseases
Eur Heart J 26:24222434
is responsible for autonomic system alterations 6. Flaker GC, Belew K, Beckman K, Vidaillet H, Kron J,
and increase in intrathoracic pressures; these Safford R, Mickel M, Barrell P (2005) Asymptomatic
hemodynamic modifications may induce repolar- atrial fibrillation: demographic features and prognos-
tic information from the Atrial Fibrillation Follow-Up
ization abnormality, atrial stretch, and structural
Investigation of Rhythm Management (AFFIRM)
remodeling leading to AF. study. Am Heart J 149:657663
Reduced lung function and pulmonary hyper- 7. Kerr C, Boone J, Connolly S et al (1996) Follow-up of
tension are independent risk factors for AF in atrial fibrillation: the initial experience of the
Canadian Registry of Atrial Fibrillation. Eur Heart J
patients with COPD exacerbation [33].
17(Suppl C):4851
Drugs used to treat bronchospasm, as theoph- 8. Glotzer TV, Hellkamp AS, Zimmerman J, MOST
yllines and adrenergic agonists, may trigger AF Investigators et al (2003) Atrial high rate episodes

ERRNVPHGLFRVRUJ
20 Atrial Fibrillation and Related Disorders 239

detected by pacemaker diagnostics predict death and Society Of Cardiology Working Group on Myocardial
stroke: report of the Atrial Diagnostics Ancillary and Pericardial Diseases. Eur Heart J 29:270276
Study of the Mode Selection Trial (MOST). 20. Schmitt J, Duray G, Gersh BJ, Hohnloser SH (2009)
Circulation 107:16141619 Atrial fibrillation in acute myocardial infarction: a sys-
9. Capucci A, Santini M, Padeletti L, et al; Italian AT500 tematic review of the incidence, clinical features and
Registry Investigators (2005) Monitored atrial fibril- prognostic implications. Eur Heart J 30:10381045
lation duration predicts arterial embolic events in 21. Lau DH, Huynh LT, Chew DP et al (2009) Prognostic
patients suffering from bradycardia and atrial fibrilla- impact of types of atrial fibrillation in acute coronary
tion implanted with antitachycardia pacemakers. J syndromes. Am J Cardiol 104:13171323
Am Coll Cardiol 46:19131920 22. van den Bos EJ, Constantinescu AA, van Domburg
10. Glotzer TV, Daoud EG, Wyse DG et al (2009) The RT, Akin S, Jordaens LJ, Kofflard MJ (2011) Minor
relationship between daily atrial tachyarrhythmia bur- elevations in troponin I are associated with mortality
den from implantable device diagnostics and stroke and adverse cardiac events in patients with atrial
risk: the TRENDS study. Circ Arrhythm Electrophysiol fibrillation. Eur Heart J 32:611617
2:474480 23. Gupta K et al (2014) Clinical utility and prognostic
11. Kirchhof P, Auricchio A, Bax J, Crijns H, Camm J, significance of measuring troponin I levels in patients
Diener HC, Goette A, Hindricks G, Hohnloser S, presenting to the emergency room with atrial fibrilla-
Kappenberger L, Kuck KH, Lip GY, Olsson B, tion. Clin Cardiol 37(6):343349
Meinertz T, Priori S, Ravens U, Steinbeck G, 24. Hijazi Z, Oldgren J, Andersson U et al (2012) Cardiac
Svernhage E, Tijssen J, Vincent A, Breithardt G biomarkers are associated with an increased risk of
(2007) Outcome parameters for trials in atrial fibrilla- stroke and death in patients with atrial fibrillation: a
tion: executive summary. Recommendations from a randomized evaluation of long-term anticoagulation
consensus conference organized by the German Atrial therapy (RE-LY) substudy. Circulation
Fibrillation Competence NETwork (AFNET) and the 125(13):16051616
European Heart Rhythm Association (EHRA). Eur 25. Hijazi Z, Wallentin L, Siegbahn A et al (2014) High
Heart J 28:28032817 sensitivity troponin-T and risk stratification in atrial
12. Benjamin EJ, Wolf PA, DAgostino RB, Silbershatz fibrillation during treatment with apixaban or warfa-
H, Kannel WB, Levy D (1998) Impact of atrial fibril- rin. J Am Coll Cardiol 63:5261
lation on the risk of death: the Framingham Heart 26. Ott A, Breteler MM, de Bruyne MC, van Harskamp F,
Study. Circulation 98:946952 Grobbee DE, Hofman A (1997) Atrial fibrillation and
13. Dorian P, Jung W, Newman D et al (2000) The impair- dementia in a population-based study. The Rotterdam
ment of health-related quality of life in patients with Study. Stroke 28(2):316321
intermittent atrial fibrillation: implications for the 27. Vermeer SE, Prins ND, den Heijer T, Hofman A,
assessment of investigational therapy. J Am Coll Koudstaal PJ, Breteler MM (2003) Silent brain
Cardiol 36:13031309 infarcts and the risk of dementia and cognitive decline.
14. Lloyd-Jones D, Adams RJ, Brown TM et al (2010) N Engl J Med 348(13):12151222
Executive summary: heart disease and stroke statis- 28. Udompanich S, Lip GY, Apostolakis S, Lane DA
tics 2010 update: a report from the American Heart (2013) Atrial fibrillation as a risk factor for cognitive
Association. Circulation 121:948954 impairment: a semi-systematic review. QJM
15. Simpson JR, Zahuranec DB, Lisabeth LD et al (2010) 106(9):795802
Mexican Americans with atrial fibrillation have more 29. Zang Y, Wang YT et al (2015) Role of inflammation
recurrent strokes than do non-Hispanic whites. Stroke in the initiation and maintenance of atrial fibrillation
41(10):21322136, ISSN 00392499 and the protective effect of atorvastatin in a goat
16. Penado S, Cano M, Acha O et al (2003) Atrial fibril- model of aseptic pericarditis. Mol Med Rep 11:
lation as a risk factor for stroke recurrence. Am J Med 26152623
114(3):206210, ISSN 00029343 30. Rezaian GR et al (2009) Atrial fibrillation in patient
17. Hart RG, Pearce LA, Aguilar MI (2007) Meta- with constrictive pericarditis: the significance of peri-
analysis: antithrombotic therapy to prevent stroke in cardial calcification. Ann Noninvasive Electrocardiol
patients who have nonvalvular atrial fibrillation. Ann 14(3):258261
Intern Med 146:857867 31. De Castro RRT et al (2006) Parasympathetic-mediated
18. Redfield MM, Kay GN, Jenkins LS, Mianulli M, atrial fibrillation during tilt test associated with
Jensen DN, Ellenbogen KA (2000) Tachycardia- increased baroreflex sensitivity. Europace 8:349351
related cardiomyopathy: a common cause of ventricu- 32. Coccagna G, Capucci A, Bauleo S, Boriani G,
lar dysfunction in patients with atrial fibrillation Santarelli A (1997) Paroxysmal atrial fibrillation in
referred for atrioventricular ablation. Mayo Clin Proc sleep. Sleep 20(6):396398
75:790795 33. Terzano C et al (2014) Atrial fibrillation in the acute,
19. Elliot P et al (2008) Classification of the cardiomy- hypercapnic exacerbations of COPD. Eur Rev Med
opathies: a position statement from the European Pharmacol Sci 18:29082917

ERRNVPHGLFRVRUJ
Atrial Flutter
21
Erika Baiocco and Azzurra Fabbrizioli

21.1 Case Report Allergies

No allergy is referred by the patient.


A 66-year-old man presented to the family
doctor complaining of abrupt onset of palpita-
tion, initially associated with mild dizziness, Social History
occurred a few days ago (probably 3 days)
while he was working. He had a recent epi- He works as a bank employee.
sode of acute bronchitis treated with empirical He never smoked.
antibiotic therapy (amoxicillin) with slow He never used illegal drugs.
resolution. During physical examination, the He occasionally drinks one glass of wine and
physician detected a rhythmic tachycardia a couple of beers a week.
pulse. The patient was at rest and was not agi-
tated. By suspecting the presence of an
arrhythmia, the doctor referred the patient to Medications
the emergency room for further assessment.
Valsartan 160 mg/day

Medical History
and Cardiovascular Risk Factors Vital Signs

Arterial hypertension Temperature: 36.5 C


Dyslipidemia Heart rate: 150 bpm
Overweight Blood pressure: 100/80 mmHg
Hiatal hernia Respiratory rate: 22 breaths per minute
No previous cardiovascular events Oxygen saturation while breathing ambient
air: 98 %
E. Baiocco A. Fabbrizioli (*)
Clinica di Cardiologia e Aritmologia,
Universit Politecnica delle Marche, Physical Examination
Via conca, 71, Ancona 60126, Italy
e-mail: erikabaiocco@libero.it; General: awake, oriented, and cooperative
Azzurra.fab@gmail.com

Springer International Publishing Switzerland 2015 241


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_21

ERRNVPHGLFRVRUJ
242 E. Baiocco and A. Fabbrizioli

Head, eyes, ears, nose, and throat: normoce- Differential Diagnosis for Narrow
phalic, atraumatic, mucous membranes moist, QRS Complex Tachycardia
pupils equally round and reactive to light and
accommodation bilaterally, normal sclera and (see Chap. 19)
conjunctiva, and normal tympanic membranes
and external auditory canal
Neck: movable without resistance, no jugular Sinus tachycardia
venous distention, no lymphadenopathy, and Reentrant supraventricular tachycardias
no carotid bruit. No thyroid enlargement Atrioventricular reciprocating
Cardiovascular: regular rhythm tachycar- tachycardia
dia; S1 and S2 normal in intensity; no mur- Atrioventricular nodal reciprocating
murs, rubs, or gallops; point of maximal tachycardia
intensity non-displaced and non-sustained; Focal atrial tachycardia
no hepatojugular reflux; and capillary refill Atrial flutter
less than 2 s Atrial fibrillation
Lungs: no rales on auscultation in the lung
bases bilaterally, no rhonchi or wheezes, no
egophony, no alterations in tactile fremitus,
and normal upon percussion The regular RR interval excludes atrial fibril-
Abdomen: mild overweight, no pulsatile lation. The presence of an organized atrial rhythm
masses, normal bowel sounds in all areas, no at a rate of 300 bpm, with no isoelectric segment
high-pitched or tinkling sounds, resonant on between the typical sawtooth waves, best seen
percussion, soft, non-distended/non-tender, in inferior leads, suggests the diagnosis of typical
no rebound or guarding, no costovertebral atrial flutter.
angle tenderness, and no hepatomegaly or The two-to-one conduction makes difficult to
splenomegaly recognize the atrial waves superimposed on the
Extremities: no cyanosis, clubbing, or edema QRS complex or T waves, but the cardiologist per-
Neurologic: cranial nerves IIXII intact and formed vagal maneuvers (carotid massage) to
no focal deficit reveal atrial waves increasing AV block (Fig. 21.2).
Psychiatric: normal affect, no hallucinations, The presence of AV block and the ineffective-
normal speech, and no dysarthria ness of carotid massage to terminate the tachy-
Skin: normal in appearance, texture, and cardia exclude the hypothesis of supraventricular
temperature reentrant tachycardia. The atrial tachycardia may
be excluded because of higher atrial rate and the
absence of isoelectric baseline between regular
Routine EKG at Rest (Fig. 21.1) atrial activations.
At the end, the cardiologist concluded a diag-
Conclusions: regular tachycardia, heart rate nosis of typical atrial flutter.
150 bpm, incomplete right bundle branch block
(100 ms), QRS axis +10, apparent atrial
rhythm at a rate of 300 bpm and 2:1 AV con- Routine Laboratory Tests
duction, ST stretch segment from V4 to V6
with flat T waves, and normal corrected QT Complete blood count: normal (Hgb
segment duration 14.6 g/ dl)
The cardiologist was called to analyze the Cholesterol (total, HDL, LDL) and TG:
arrhythmia. normal
The presence of narrow QRS (100 ms) com- Hepatic function (GOT, GPT, -GT, ALP, total
plex indicates supraventricular tachycardia. bilirubin, direct and indirect): normal

ERRNVPHGLFRVRUJ
21 Atrial Flutter 243

a b

c d

Fig. 21.1 Clockwise-type typical atrial flutter with 2:1 ventricular response

Fig. 21.2 ECG strip of leads I, II, and III shows carotid sinus massage with progressive increase in atrioventricular
block

ERRNVPHGLFRVRUJ
244 E. Baiocco and A. Fabbrizioli

Thyroid function (TSH, FT3, FT4): normal abnormalities. Normal right ventricle size with
Renal function (creatinine, BUN): normal normal function. Mild mitral and tricuspid regur-
Electrolytes (Na+, K+, Ca++, Mg++, Cl): gitation with normal pulmonary arterial pressure
normal (PAPs = 30 mmHg). No pericardial effusion.
Fasting blood glucose: 90 mg/dl Slightly dilated inferior vena cava (24 mm) with
HbA1c: 5.0 % >50 % respiratory collapse. Monophasic dia-
TnI-hs and CK-MB: 0.25 ng/ml (normal stolic flow pattern with initial increase of esti-
<0.055 ng/ml) mated filling pressure.
BNP: 127 pg/ml (normal 1100 pg/ml) Conclusions: hypertensive cardiomyopathy
(HCM). The absence of any signs of ventricular
The routine laboratory did not show any dysfunction excludes a tachycardiomyopathy.
reversible proarrhythmic trigger such as electro- The patient was a young active person and he
lyte imbalance, anemia, and thyroid dysfunction. had never had heart rhythm problems before; in fact
The troponin I-hs value was slightly increased this episode of arrhythmia was the first in his life.
due to the tachyarrhythmia. The high myocardial Furthermore it could be triggered by bronchitis.
rate may cause an imbalance between myocardial For these reasons, and in consideration of the
oxygen supply and demand by enhancing myo- presence of only mild left atrium enlargement,
cardial oxygen requirements and reduces the dia- the cardiologist chose a rhythm-control strategy.
stolic coronary filling time. Also the BNP value The patient was electrically cardioverted.
was slightly increased as a result of initial atrial Catheter ablation is indicated for recurrent,
increased filling pressure. symptomatic, or drug-refractory arrhythmia and
therefore was not performed in this case.
The exact duration of atrial flutter was
Chest X-Ray unknown and lasted longer than 48 h. The patient
had a CHA2DS2-VASc score of 2, suggesting a
Both lung fields and costophrenic angles are considerable risk of stroke.
clear. The mediastinum is within normal limits. Many risks and complications of cardiover-
Cardiac size and shape are normal. sion are associated with thromboembolic events,
In agreement with patients symptoms and so effective anticoagulation for at least 3 weeks is
physical examination, chest X-ray did not show mandatory for AFL of >48 h or AFL of unknown
radiographic signs of pulmonary stasis. duration, unless the patient was hemodynami-
cally unstable (LGAF).
At this point, the cardiologist proposed to per-
Echocardiography Performed form a transesophageal echocardiography (TEE)
to Assess Cardiac Function as an alternative to 3 weeks of adequate pre-
and the Presence of Structural Heart cardioversion anticoagulation.
Disease In the meantime, a good control of ventricular
rate was achieved by administrating a calcium-
Normal trileaflet aortic valve and normal aorta channel blocker (verapamil).
dimension (aortic root dimension = 2.7 cm; Transesophageal echocardiography was
ascending aorta = 3.1 cm; aortic arch = 3.1 cm; performed.
thoracic aorta = 2.8 cm; abdominal aorta = 1.8 cm). Conclusion: TEE excluded the presence of
Mild enlarged left atrium (LA diameter M-mode thrombus in the left atrium or left atrial appendage.
41 mm; LA indexed volume 36 ml/m2). Normal The patient was fully informed about the risk
right atrium. No atrial septal defect. Mild left of cardioversion procedure.
ventricular hypertrophy with normal systolic He was sedated with a light anesthetic (mid-
function (mass indexed volume 70 ml/m2; EF azolam). Two electrode paddles were placed on
0.65) in the absence of regional wall motion his chest: the anterior patch was placed under the

ERRNVPHGLFRVRUJ
21 Atrial Flutter 245

Fig. 21.3 Electrical cardioversion. The atrial flutter is converted to sinus rhythm after one synchronized biphasic elec-
trical shock at 50 J

Fig. 21.4 Sinus rhythm, 85 bpm, normal atrio and inter- ventricular conduction, normal ripolarization

right clavicle and the apical patch was placed at patient had a considerable risk of thromboem-
the apex (anterior-apical paddle position). bolism (CHADS2VASc = 2) and a low risk of
Electrical cardioversion was performed during a bleeding (HAS-BLED = 1). In this case, oral
close oxygen level, blood pressure, and heart anticoagulant (OAC) therapy is indicated not
rhythm monitoring. Atrial flutter was success- only for 4 weeks after cardioversion (atrial
fully converted to sinus rhythm after one syn- stunning-related risk), but it should continue
chronized biphasic electrical shock at 50 J, lifelong irrespective of an apparent mainte-
without complications (Figs. 21.3 and 21.4). nance of sinus rhythm. The cardiologist
explained pros and cons of the OAC therapy
and following the patients preferences pre-
21.2 Atrial Flutter scribed a new oral anticoagulant (NOAC).
Catheter ablation and chronic prophylactic
Atrial flutter (AFL) management includes anti- antiarrhythmic therapy were not recommended
thrombotic and antiarrhythmic therapy, correc- because it was the patients first episode of AFL
tion of possible underlying causes, and and his AFL was probably due to a respiratory
treatment of associated comorbidities. The trigger (bronchitis). It was also well tolerated.

ERRNVPHGLFRVRUJ
246 E. Baiocco and A. Fabbrizioli

Catheter ablation is indicated in case of poorly Physiopathology


tolerated or recurrent symptomatic AFL.
Hypertension is a risk factor for atrial flutter AFLs comprise a heterogeneous group of atrial
and its related complications. The patient referred arrhythmias produced by abnormalities of
optimal blood pressure control with systolic impulse conduction that are underpinned by
blood pressure <140 mmHg and diastolic blood macro-reentrant circuits. The mechanism of
pressure <90 mmHg; the antihypertensive ther- reentry consists of a repetitive excitation of a
apy was not changed. specific myocardial region (excitable gap) with
consequent conduction of the electrical impulse
across a defined circuit, around a fixed obstacle.
Denition The macro-reentry circuit is supported by con-
ditions of slowed conduction constrained by
The atrial flutter is a common supraventricular barriers that may be anatomical, functional, or
arrhythmia caused by an intra-atrial macro- both. The development of a unidirectional con-
reentrant circuit. It is characterized by a rapid and duction block starts the arrhythmias, and it is
organized atrial depolarization at a rate between usually a consequence of an acceleration of the
250 and 350 bpm and a different degree of AV heart rate or block of a premature impulse,
nodal blocking [1]. while for its maintenance, a slow conduction is
required [4].
Different types of macro-reentrant are possible.
Epidemiology Atrial flutter and atrial fibrillation may be
present in the same person. A common trigger is
The Marshfield Epidemiological Study Area recognized such as repetitive premature impulse
(MESA) shows an incidence of atrial flutter of originating from pulmonary veins or from both
88/100,000 person-years with 200,000 new atria. Furthermore the AF originating from the
cases [2]. Overall, atrial flutter represents left atrium may invade the right atrium following
approximately 10 % of supraventricular tachy- macro-reentrant circuit triggering AFL. In addi-
cardia, and it is much less common than atrial tion atrial remodeling consequently to AFL may
fibrillation [3]. However atrial flutter occurs in promote the onset of AF [5].
approximately 2535 % of patients with atrial
fibrillation. The incidence is greater in men
than in women and increased markedly with Atrial Flutter Classication
age [4].
In 60 % of cases, atrial flutter occurs with a The classification of atrial flutter proposed by
specific precipitating event such as thyroid dys- the European Society of Cardiology and the
function, respiratory infections, pulmonary North American Society of Pacing and
embolism, or recent major surgery, while in Electrophysiology is based on both anatomical
remaining cases, atrial flutter is usually associ- features and electrophysiological mechanisms,
ated with chronic comorbid conditions such as determined at the time of electrophysiological
heart failure, valve abnormalities (especially testing [6]. Atrial flutter is categorized into typi-
mitral valve), congenital defects, hypertension, cal or atypical.
chronic lung disease, alcoholism, or use of stimu-
lants such as cocaine, amphetamines, diet pills, Typical Atrial Flutter
and even caffeine. More rarely (only 1.7 % of
cases), atrial flutter can occur in the absence of
structural heart disease, like idiopathic form Typical atrial flutter is subdivided into two
(lone atrial flutter) [4]. The possibility of a genetic subtypes:
predisposition is unclear.

ERRNVPHGLFRVRUJ
21 Atrial Flutter 247

Counterclockwise atrial flutter is the Right cavo tricuspid isthmus-dependent


most common type of AFL (90 %). The atrial flutter
reentrant circuit is located in the right Counterclockwise or typical atrial
atrium, whose fundamental component flutter
is the isthmus, a posteroseptal part of Clockwise or reverse typical atrial
slow conduction bounded by the inferior flutter
vena cava, Eustachian ridge, coronary Double-wave reentry
sinus, and the tricuspid valve annulus. Right non-cavo tricuspid isthmus-
The impulse activation rotates in a coun- dependent atrial flutter
terclockwise direction through the atrial Scar-related atrial flutter
septum, by the superior vena cava, and Upper loop reentry
then inferiorly down the right atrial free Critical flutter circuits
wall, through the isthmus to reenter the Left atrial flutter
atrial septum. Mitral valve annulus atrial flutter
Clockwise atrial flutter is less prevalent Scar-related atrial flutter
(10 %). It is supported by the identical Left membranous circuits
reentrant circuit to those in counter-
clockwise atrial flutter but with a clock-
wise direction of rotation around the Surface ECG Atrial Flutter
tricuspid annulus. Characteristics

AFL is characterized by a rapid regular atrial


rhythm at the rate of 250350 bpm [1]. Atrial rate
Atypical Atrial Flutter lower than 250 bpm is generally caused by anti-
The reentrant circuit is not cavo tricuspid isthmus arrhythmic drugs (IA and IC sodium channel
dependent, and it can be located either in the right or blocking) that slow down macro-reentrant circuit
left atrium. The circuit curves around a scar or sur- and may cause one-to-one conduction of
gical incision, especially after surgical repair of AFL. Also patients with markedly enlarged atria,
congenital heart defect, atrial patch implantation or which increases duration of flutter circuit, have a
radiofrequency, and cryotherapy catheter ablation. slower atrial rate. Digitalis may increase atrial
The most common forms of right atrial atypi- rate and induce AF. Commonly the ventricular
cal flutter can show a macro-reentry circuit response in the absence of treatment is due to the
located in the right atrial free wall or around the functional atrioventricular (AV) block; the most
superior vena cava (upper loop atrial flutter) common is 2:1 but may be 4:1, 3:1, or 3:2 or
[79]. second-degree AV block. A third-degree AV
Left AFLs are usually associated with block is possible for impaired AV conduction or
hypertension, mitral valve disease, LA dila- drug treatment, and it is visible on surface ECG
tion, and cardiac failure. The circuits may be for the presence of flutter waves without constant
located around the mitral valve annulus, around relation to the regular QRS complexes [1, 5].
the region of scar, and the ostia of the pulmo-
nary veins [10].
ECG and Atrial Flutter Circuit
Location
The following are the classification of atrial
flutter based on specific atrial circuits pro- Typical counterclockwise atrial flutter elec-
posed by Scheinman et al. [11]: trocardiographically shows a characteristic
sawtooth pattern in leads II, III, and aVF,

ERRNVPHGLFRVRUJ
248 E. Baiocco and A. Fabbrizioli

with a dominant negative flutter wave char- response. Patients with AFL are usually more
acterized by a sharp steep ascent followed by symptomatic than those with just AF because the
a gradual descent. Lead V1 shows positive heart rate is more rapid during AFL and because
flutter wave, with an isoelectric component AF is usually associated with increased AV nodal
between the oscillations. The positive deflec- penetration and slower ventricular responses.
tion progressively becomes inverted across Atrial flutter with one to one AV conduction may
the precordium, and the flutter wave is com- be life-threatening. Rapid ventricular rate may be
pletely negative in V6. This type of AFL is caused by antiarrhythmic treatment (IA and IC)
characterized by a rate of 250350 bpm [4, that slows the atrial rate or precipitated by
1214]. increased sympathetic stimulation such as during
Typical clockwise atrial flutter shows sym- exercise or induced by anesthesia. Another high-
metrical flutter waves in the inferior leads, risk condition is present in patient with accessory
with the same ascent and descent segments AV pathways capable of rapid conduction and
without isoelectric line between oscillations. excessive ventricular rate. Moreover, in patients
Lead V1 is characterized by negative deflec- with impaired cardiac function, the hemodynamic
tions, and the flutter waves show a gradually instability may be due to loss of regular atrial rate
transition across the precordium to an upright and diastolic atrial contribution even if the ven-
deflection in V6 [4, 9, 14, 15]. tricular rate is not excessively rapid.
Sometimes thromboembolism with transient
Some people may present both typical AFLs ischemic attack or stroke can be the first mani-
with either counterclockwise or clockwise right festation of arrhythmia. Polyuria is another typi-
atrial macro-reentrant circuit. cal symptom; it is caused by the release of atrial
Atypical atrial flutter shows a more variable natriuretic peptide in response to increased
surface ECG appearance depending on macro- atrial pressure during atrial contraction against a
reentry anatomical location, direction of rotation, closed AV valve, and that indicates a sustained
and the presence of more loop circuits, some- arrhythmia [4].
times mimicking a focal atrial tachycardia. In the Atrial flutter typically has a sudden onset. As
inferior leads, the morphology of flutter waves is atrial fibrillation, this arrhythmia can be clini-
not typically sawtooth and usually the isoelec- cally distinguished into [4]:
tric line is present. The surface ECG findings are
often similar for different underlying substrates,
making the localization within the LA based on Paroxysmal form: when it is transient
the ECG difficult. The flutter wave usually shows and ends spontaneously
a prominent positive deflection in lead V1 and Persistent form: when AFL is present
uncommonly is flat or isoelectric. The flutter and either lasts longer than 7 days or
waves in leads II, III, and aVF may be upright but requires termination by cardioversion
are frequently of low amplitude. LA AFL may (either with drugs or by direct current
sometimes mimic a focal atrial tachycardia [14]. cardioversion)
Permanent form: when the presence of
the arrhythmia is accepted by the patient
Clinical Presentation and the physician and the rhythm-con-
trol strategy is, by definition, not carried
Patients with atrial flutter can be asymptomatic, out
but more commonly, they present with palpita- Long-standing persistent AFL form:
tions, shortness of breath, dizziness, and fatigue. when AFL is present for at least 1
Patients with underlying heart or lung disease year when it is decided to pursue a
may complain also of syncope and chest pain. The rhythm-control strategy
hemodynamic stability depends on ventricular

ERRNVPHGLFRVRUJ
21 Atrial Flutter 249

Diagnosis Tachycardiomyopathy
AFL with persistent high ventricular rate (120
The surface EKG usually allows to diagnose 130 bpm), if untreated, may induce reversible
AFL; it is essential to obtain documentation of ventricular dysfunction (ventricular tachycardio-
the arrhythmia for the choice of treatment. When myopathy) [4].
the ventricular response is one to one or two to
one, EKG analysis may be difficult, because flut-
ter waves are superimposed on the QRS complex Treatments
and T waves. In these cases, vagal maneuvers or
intravenous adenosine administration can be The treatment of atrial flutter should be directed
used to unmask atrial activity. When arrhythmia to relief symptoms, prevention of complications,
is only suspected, it is necessary to perform achieving control of ventricular rate, conversion
Holter ECG monitoring (24 h to 7 days) or to to sinus rhythm, and maintenance of sinus
implant a loop recorder to establish the diagnosis rhythm.
and to choose the strategies of treatment.
Sometimes, the surface EKG is limited to define Acute Therapy
anatomical localization of macro-reentrant cir- Acute management of patients with atrial flutter
cuits, so the endocardial mapping can be used to depends on clinical presentation. The main goals
identify precise anatomical circuit. are to reduce symptoms and to prevent AFL-
related complications. It includes the management
of antithrombotic therapy, achieving control of
Differential Diagnosis ventricular rate through the administration of AV
nodal-blocking agent, conversion to sinus rhythm
See Chap. 19. through electrical or chemical cardioversion, or
atrial overdrive pacing.
If the patient is hemodynamically compro-
Complications mised or complains of significant symptoms (chest
pain, congestive heart failure), like often occurred
Thromboembolic Events in case of one-to-one AV conduction, urgent rate
AFL presents an increased risk for the develop- control or DC cardioversion is required.
ment of thromboembolic events. In non-antico- There are four options possible for the acute
agulated patients, the incidence of atrial treatment of AFL:
echo-dense material or clots increases with atrial
flutter duration longer than or equal to 48 h, and Emergent synchronized external DC cardio-
the risk of embolism was 2.2 % [16]. This is due version (CV), performed under anesthesia, is
to the high atrial rate that determines the paraly- the treatment of choice because of very high
ses of either the RA or LA with consequent blood likelihood of success (terminates AFL in
stasis and development of thrombus. Clots are >90 % of episodes) and safety (class I with
more frequently observed in the appendage level of evidence C). DC cardioversion is very
where the blood velocity is lower. effective because of rapid homogeneous depo-
Risk factors related to these complications are larization of the entire atrium. The necessary
similar to those described for atrial fibrillation, energy to revert AFL to sinus rhythm is less
and the CHADS2VASc score remains the sim- than 50 J using monophasic shocks and with
plest scheme of stroke risk stratification [15]. less energy using biphasic shocks [4, 17].
Furthermore AF and AFL often coexist in the Unsuccessful conversion is usually associated
same patient (2030 %), so the same manage- with prolonged period of the tachycardia, and
ment of antithrombotic therapy is recommended a higher CV energy is necessary that may
(see Chapter 20 AF and related disorders). compromise left ventricular function or

ERRNVPHGLFRVRUJ
250 E. Baiocco and A. Fabbrizioli

increase left atrial size. One option is pretreat- acting (about 30 min), and it is highly effec-
ing patient with an antiarrhythmic agent (ibu- tive in slowing ventricular rate response but
tilide is the most effective) and then the lower may provoke hypotension (10 %) especially in
CV energy may be sufficient. patients with significant left ventricular dys-
Atrial overdrive pacing by transesophageal function. Verapamil is just effective as diltia-
route or with atrial electrodes should be con- zem for rate control but with higher incidence
sidered as an option for conversion to sinus of symptomatic hypotension in particular in
rhythm, mainly in patients with implantable patients with left ventricular dysfunction. The
devices and after cardiac surgery, because beta-blockers are effective as class IV drugs,
these patients frequently have epicardial atrial but they should be avoided in patients at risk
pacing wires. It does not require anesthesia. for reactive airway disease exacerbation.
Nevertheless, high-frequency atrial pacing Digoxin and amiodarone are favored for rate
can induce sustained atrial fibrillation. control in patients with significant congestive
Pretreatment with antiarrhythmic agents may heart failure since other drugs (classes II and
reduce the risk of inducing AF with atrial IV) may worsen cardiac decompensation.
overdrive [4]. This method has a success rate
of about 80 %. Overdrive atrial pacing is pre- However, these are not a permanent solution
ferred in patients who have a history of sick and the arrhythmia may come back.
sinus syndrome with risk for significant bra- If the patient is hemodynamically stable and
dycardia after conversion. atrial flutter presents higher grades of AV block,
Chemical cardioversion is another method to conversion to sinus rhythm should be deferred,
terminate AFL without the need for anesthe- especially with arrhythmia of more than 48 h in
sia. The effect is not instantaneous but the suc- duration, because in these cases, anticoagulant
cess is recorded within 1 h after start of drug therapy is a priority. When a patient is adequately
infusion. The drug necessary to pharmacolog- anticoagulated, the DC cardioversion is the first
ical CV should prolong the refractory period choice (class I with level of evidence B).
within the tachycardia circuit, inhibiting con-
tinuation of the circulating wave front. Class Prevention of Recurrence and Chronic
III agents, such as amiodarone and ibutilide, Therapy
are typically used because they prolong refrac- Chronic pharmacological treatments in patient
toriness and may terminate the arrhythmia with AFL include:
because the circulating wave front encounters
tissue that is refractory. Intravenous amioda- For pharmacological prevention of recurrence,
rone has longer latency period. Ibutilide is class IA drugs such as quinidine and class III
more effective (70 % of success) and rapid drugs are used because they can prevent atrial
(about 30 min), but it may produce torsades de premature beats, reduce the excitable gap, and
pointes arrhythmias in 25 % of cases, and increase atrial refractoriness. Class IC drugs
ECG monitoring for at least 6 h is necessary. alone are usually ineffective and dangerous
But ibutilide should not be used in patients and should be combined with AV nodal-
with structural cardiac diseases, prolonged QT blocking agents [5].
interval, or underlying sinus node disease [5].
The administration of drugs to slow AV nodal When currently available antiarrhythmic
conduction and ventricular response (rate con- drugs for the prevention of AFL are not efficient
trol) is another pharmacological strategy. It and poorly tolerated (in particular IC or sotalol),
should be achieved with class II, III, and IV dronedarone can be used that decreases the risk
drugs [4, 5]; also if in AFL, the rate control of arrhythmic recurrence [18].
may be more particularly difficult to achieve In 60 % of patients, AFL is triggered by exac-
than in AF. Intravenous diltiazem (IV) is rapid erbation of pulmonary disease, postoperative

ERRNVPHGLFRVRUJ
21 Atrial Flutter 251

cardiac or pulmonary surgery, or during acute electrodes that form a Cartesian reference system
myocardial infarction. In this clinical condition, (X-Y-Z) are used. The myocardial virtual anatom-
the use of chronic therapy is not required [5]. ical map is created by moving the electrophysio-
The preferred therapies of poorly tolerated logical catheter in the heart chamber and recording
(also first episode) and recurrent or persistent the precise point of the electrophysiological cath-
atrial flutter is radiofrequency and cryotherapy eters electrodes rather than the surface electrodes,
catheter ablation (class I with level of evidence B) thanks to the recorded voltage gradient. The sys-
[4]. The typical flutter can be mapped with multi- tem analyzes the voltage of each myocardial point
polar catheter (e.g., halo) located in the anterolat- and identifies the sequence of activation com-
eral area of RA that allows to record the direction pared with the reference electrodes recording
of the cavo tricuspid isthmus macro-reentrant cir- more points of the same camera (electric map of
cuit and to distinguish between typical clockwise voltage and activation). The sequence of activa-
and counterclockwise AFL circuits. Diagnosis by tion time is shown with color map and is inte-
this endocavity electrophysiological study is sim- grated with the scar areas recorded with voltage
ple and usually more complex techniques such as map. The macro-reentrant circuit identified by
NavX are not necessary. Radiofrequency ablation these techniques can be interrupted by linear RF
of CTI-dependent AFL involves creating a linear lesion. For pharmacological prevention of recur-
lesion from the inferior vena cava (IVC) to the rence, class IA drugs such as quinidine and class
tricuspid ring to eliminate the critical isthmus in III drugs are used because they can prevent atrial
the macro-reentrant circuit. Successful ablation premature beats, reduce the excitable gap, and
interrupts macro-reentrant circuit and it is demon- increase atrial refractoriness. Class IC drugs alone
strated by bidirectional conduction block. For are usually ineffective and dangerous and should
diagnosis of atypical atrial flutter, typical flutter in be combined with AV nodal-blocking agents [5].
RA is necessary to exclude and to identify the Long-term anticoagulation is advised for
location of macro-reentrant circuit. This is not patients with persistent or paroxysmal atrial flut-
possible with conventional electrophysiological ter, whereas after successful catheter ablation,
study, and it is necessary to use CARTO or NavX anticoagulation can be stopped 46 weeks later if
techniques. The CARTO technique records an sinus rhythm is still present.
electroanatomic map of the RA and LA in sinus For several reasons, follow-up is very impor-
rhythm showing scar area. This system uses a tant for patients with AFL. The risk profile can
magnetic field generated around the patients change as well as the indication for anticoagula-
chest which allows to identify the specific tip tion, for example, when a new diagnosis of diabe-
position of the electrophysiological catheter in the tes or hypertension will be done or when a renal
RA or LA showing three-dimensional anatomical dysfunction occurred. Because of the potential
reconstruction of the heart chambers (anatomical side effects and proarrhythmic risk related to
virtual map). At the same time, the system records chronic antiarrhythmic therapy, patients that
the electric signal from each point using color receive these drugs should be monitored through
code (electric map). Then the two maps are inte- 12-lead ECG, laboratory tests, and echocardio-
grated. Moreover the activation and propagation gram at regular intervals [15].
map recorded with a color scale identifies the
macro-reentrant circuit, the direction, and the
point with slower conduction target for ablation.
The NavX technique uses an electric current References
between two surface electrodes that generates a
voltage gradient between the two electrodes. This 1. Surawicz B, Knilans T (2008) Chous electrocardiog-
raphy in clinical practice, Adult and pediatric. Sixth
voltage gradient is recorded by a standard electro- Edition. Elsevier Health Sciences, London
physiological catheter located in the heart cham- 2. DeStefano F et al (1996) Epidemiologic research in
bers. Usually, in NavX technique, three surface an integrated regional medical care system: the

ERRNVPHGLFRVRUJ
252 E. Baiocco and A. Fabbrizioli

Marshfield Epidemiologic Study Area. J Clin electrocardiographic characteristics of right and left
Epidemiol 49(6):643652 atrial flutter. Circulation 108:6066
3. Wellens HJJ (2002) Contemporary management of 11. Scheinman MM et al (2004) Atrial flutter: part II
atrial flutter. Circulation 106:649665 nomenclature. Pacing Clin Electrophysiol
4. Blomstrm-Lundqvist C, Scheinman MM et al (2003) 27(4):504506
ACC/AHA/ESC guidelines for the management of 12. SippensGroenewegen A, Lesh MD, Roithinger FX,
patients with supraventricular arrhythmias. American Ellis WS, Steiner PR, Saxon LA et al (2000) Body
College of Cardiology Foundation, American Heart surface mapping of counterclockwise and clockwise
Association, Inc., and European Society of Cardiology typical atrial flutter: a comparative analysis with
5. Delise P et al (2008) Aritmie. Fisiopatologia e diag- endocardial activation sequence mapping. J Am Coll
nosi: dallecg al mappaggio tridimensionale. Cardiol 35:12761287
Sciantifica Internazionale, CESI 13. Cosio FG, Arribas F, Lopez-Gil M, Gonzalez HD
6. Saoudi N et al (2001) Classification of atrial flutter (1996) Radiofrequency ablation of atrial flutter.
and regular atrial tachycardia according to electro- J Cardiovasc Electrophysiol 7:6070
physiologic mechanism and anatomic bases: a state- 14. Medi C, Kalman JM (2008) Prediction of the atrial
ment from a joint expert group from the Working flutter circuit location from the surface electrocardio-
Group of Arrhythmias of the European Society of gram. Review. Europace 10:786796. doi:10.1093/
Cardiology and the North American Society of Pacing europace/eun106
and Electrophysiology. J Cardiovasc Electrophysiol 15. ESC/EHRA 2011 (2010) Guidelines for the manage-
12(7):852866 ment of atrial fibrillation. Eur Heart J 31:23692429.
7. Yang Y, Cheng J, Bochoeyer A, Hamdan MH, Kowal doi:10.1093/eurheartj/ehq278
RC, Page R et al (2001) Atypical right atrial flutter 16. Pengo V, Rampado E, Iliceto S (2006) La terapia anti-
patterns. Circulation 103:30923098 coagulante nel flutter atriale. G Ital Aritmol Cardiostim
8. Tai CT, Liu TY, Lee PC, Lin YJ, Chang MS, Chen SA 3:151156
(2004) Non-contact mapping to guide radiofrequency 17. Stec S, Kryski T, Kuakowski P (2011) Efficacy of
ablation of atypical right atrial flutter. J Am Coll low energy rectilinear biphasic cardioversion for reg-
Cardiol 44:10801086 ular atrial tachyarrhythmias. Cardiol J 18(1):3338
9. Ricard P, Imianitoff M, Yaici K, Coutelour JM, 18. Guerra F, Capucci A et al (2014) Efficacy and safety
Bergonzi M, Rinaldi JP et al (2002) Atypical atrial of dronedarone in patients previously treated with
flutters. Europace 4:229239 other antiarrhythmic agents. Wiley Online Library
10. Bochoeyer A, Yang Y, Cheng J, Lee RJ, Keung EC, 37(12):717724
Marrouche NF et al (2003) Surface

ERRNVPHGLFRVRUJ
Part VII
Rhythm Disorders: Bradyarrhytmias

ERRNVPHGLFRVRUJ
Sinoatrial Exit Block
22
Andrea Romandini and Lorena Scappini

22.1 Case Report


was treated according to the protocol for
hyperkalemia with infusion of NaHCO3,
A 77-year-old man was referred to the hydration with glucose and insulin 10 IU,
cardiology department from the emergency and administration of furosemide
room of our hospital due to dyspnea, asthe- IV. Echocardiography demonstrated upper
nia, and dizziness which arose a few hours limits of normality for LVM (left ventriccle
before. The patient also reported that he dimension) index with slightly reduced
had taken up to 2 days before treatment ejection fraction and mild mitral regurgita-
levofloxacin for bronchopneumonia. ECG tion. All echocardiographic findings were
revealed the presence of mild bradycardia already present in a previuos echocardio-
(58 bpm) with type II sinoatrial block gram performed elsewhere. No abnormali-
(Fig. 22.1); it was also observed that bipha- ties were observed in other heart valves, not
sic T waves in some leads (V4, V5, V6) and even on the right ventricle.
negative T waves in III and aVF were pres- The patient was then transferred to our
ent. Urgent laboratory blood test showed a arrhythmology and cardiology clinic for
high level of serum creatinine (2.3 mg/dl) further evaluation and treatment.
and hyperkalemia (7 mEq/l). All other
blood tests were normal, including the val-
ues of troponin I and CK-MB. The patient

Medical History and Cardiovascular


Risk Factors

1. Cardiovascular risk factors: hypertension,


dyslipidemia, overweight body, and family
history of cardiovascular disease
A. Romandini L. Scappini (*) 2. In 2011 the patient had a non-ST-elevation
Clinica di Cardiologia e Aritmologia, Universit myocardial infarction treated with PCI + stent-
Politecnica delle Marche, ing of the first segment of the left anterior
Via Conca, 71, Ancona 60126, Italy
e-mail: romandini.andrea@hotmail.it; lorena. descending coronary. The angiography did
scappini@gmail.com not reveal significant stenoses of other

Springer International Publishing Switzerland 2015 255


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_22

ERRNVPHGLFRVRUJ
256 A. Romandini and L. Scappini

Fig. 22.1 The figure shows the patients ECG when he arrived at the emergency room: it was characterized by type II
sinoatrial block; biphasic T waves were present in V4, V5, and V6, and negative T waves in III and aVF

coronaries. Since then, the patient underwent Physical Examination


annual cardiology visit with EKG and echo-
cardiography, the last of which dated back General appearance: alert and cooperative and
about 1 month before he entered the emer- slightly dyspneic
gency room. No abnormalities were found Lungs: clear to percussion and minimal rales on
during that visit. The echocardiogram reported pulmonary bases
the same alterations listed above. Moreover, Cardiovascular system: normal S1 and S2.
he denied angina, dyspnea, or syncope before Murmur 2/6 at the apex. Bradycardic rhythm.
that day. No peripheral edema, cyanosis, or pallor.
Warm and well-perfused extremities
Abdomen: positive bowel sounds. Soft and non-
Medications distended. No guarding or rebound. No masses

ASA 1 cp, ramipril 2.5 mg 1 cp bid, potassium


canrenoate 100 mg 1 cp, bisoprolol 2.5 mg 1 cp, Routine Laboratory Test
atorvastatin 80 mg 1 cp, pantoprazole 20 mg 1 cp,
tamsulosin chlorhydrate 1 cp, and allopurinol 1 cp Potassium 6.1 mEq/l, sodium 136 mEq/l, and
creatinine 2 mg/dl. Normal in other blood tests

Vital Signs
Instrumental Examination
Temperature 36.5 C, heart rate 35 bpm, arterial
blood pressure 95/55 mmHg, respiratory rate A complete echocardiogram was performed and
20 breaths/min, and oxygen saturation 98 % showed upper limits of normality for LVM index

ERRNVPHGLFRVRUJ
22 Sinoatrial Exit Block 257

(LV diastolic volume/BSA 75 ml/m2) with a A follow-up visit was performed 1 month
slightly reduced ejection fraction (EF 48 %) due later. The patient was completely asymptomatic.
to hypokinesia of interventricular septum and Physical examination was normal. The ECG
apical segments of the anterolateral wall. Normal showed a sinus rhythm (heart rate of 62 bpm).
dimensions and function of right cardiac cham- Holter monitoring did not record any arrhythmia.
bers, a mild mitral regurgitation, a calcification of Laboratory blood tests were normal, including
the aortic valve without regurgitation nor steno- the values of potassium (4 mEq/l) and creatinine
sis, a slightly enlarged tubular ascending aorta (1.4 mg/dl). Blood pressure level was
(38 mm), and a mild tricuspid regurgitation with 145/95 mmHg, so it was decided to resume
32 mmHg pulmonary artery systolic pressure administration of ramipril 2.5 mg cp bid. It was
were already shown by the echocardiogram that also recommended to perform periodic checks of
the patient underwent 1 month before. renal function and echocardiographic follow-up
A chest radiograph was done and disclosed of the tubular ascending aortic dimensions.
normal cardiac and pulmonary findings.

22.2 Electrocardiographic
Clinical Course and Therapeutic Diagnosis of Sinoatrial
Management Exit Block

These findings altogether were suggestive of The major electrocardiographic expressions of


symptomatic sinoatrial block. All possible sinus node dysfunction (SND) are the presence
removable causes of sinoatrial block were of abnormal sinus bradycardia or the occurrence
checked: a strict control of kalemia was per- of sinus pause.
formed, and the patient continued to be treated by The term sinus pause is usually used to
hydration with glucose and insulin 10 IU and describe the occurrence on ECG of a sudden
administration of furosemide and Kayexalate. pause in the underlying sinus rhythm: one or
Moreover, administration of bisoprolol, ramipril, more sinus beats fail to appear at the expected
and potassium canrenoate was interrupted. A time and a pause occurs (long P-P interval). After
continuous telemetric monitoring was placed and the pause, the resumption of the basic sinus
showed a restoration of sinus rhythm when the rhythm can be slower for some cycles (temporary
level of potassium reached values of 5.5 mEq/l. rate suppression); some rescue rhythms (escape
The protocol for hyperkalemia was continued beats or rhythm) may also appear from ectopic
until the values of potassium in the blood were subsidiary supraventricular sites (perisinus,
normal. The patient became asymptomatic dys- atrial, or junctional).
pnea, asthenia, and dizziness. During the hospi- The occurrence of a sinus pause can be due to
talization he underwent an ECG stress test that the failure of a sinoatrial (SA) node to initiate
showed a normal chronotropic competence and (sinus arrest) or to conduct the impulse (sinus
no ischemia signs. On the 5th day, he was dis- exit block). Some authors dont distinguish the
charged with a diagnosis of symptomatic sino- sinus pause from the sinus arrest, restricting the
atrial block secondary to iatrogenic acute renal latter in case of prolonged sinus inactivity, even if
failure and hyperkalemia in patient with ischemic no precise definition exists [1]. For practice pur-
cardiomyopathy. Since the ECG was unchanged poses in this text, we refer to sinus pause in its
from the previous, myocardial enzymes were broadest sense.
normal, and there was no evidence of ischemia at Electrocardiographically the sinus impulse
the ECG stress test, it was then decided to not is only recorded once it has left the sinus node
perform a coronarography. Before the discharge, and activated the atrium, thereby resulting in
a patch-type long-term external Holter recorder the P wave. The presence of delay or block in
was applied to the patient. conduction at the SA junction can be detected

ERRNVPHGLFRVRUJ
258 A. Romandini and L. Scappini

by calculation of certain rhythmic sequences pause. The pause generated in a Wenckebach


involving the pause and the P-P intervals [2]. series is twice the sinus cycle minus the sum of
In sinus arrest there is a failure of impulse for- the increments of conduction delay at each cycle.
mation within the sinus node, disappointing the However, the P-P interval doesnt accurately
timing of SA node discharge. In this case the reflect the sinus interval, as it has been modified
underlying rhythm would not resume on time by varying SA conduction times.
after the pause; the long cycle has no mathemati- For this reason, the duration of presumed
cal relation with the basic P-P interval but is ran- sinus cycle may be calculated on the basis of
dom in duration, often interrupted by escape Wenckebach principle periodicity, by dividing
beats or rhythm. the total duration of the period by the number of
In sinus exit block, an electrical impulse is ini- visible cycles + 1 (sinus impulses which are con-
tiated by the SA node but not conducted to the ducted after a long pause will probably have SA
atria because it is blocked within the SA node or intervals of similar length, i.e., they are corre-
the sinoatrial junction. The dropped beats may sponding impulses. The isoconduction interval
occur sporadically or in regularly recurring pat- between these corresponding impulses will be
terns as fixed block. In the presence of a regular the same at both sinus and atrial levels) [2].
basic sinus rhythm, SA block can almost always Type II SA block shows a failed conduction of
be distinguished from sinus arrest because the a sinus impulse without previous prolongation of
sinus block generating pause is mathematically SCAT. For this reason the pattern of type II block
related to the baseline P-P interval (see later in is characterized by the following: (1) a P-P inter-
the text). val following the dropped SA impulse is not sig-
Sinoatrial exit block can be divided into three nificantly larger than the interval preceding it
types (first, second, and third degrees), analogous (this is the most important criterion for diagno-
to those occurring at the atrioventricular (AV) sis) [3], (2) the P-P interval including a blocked
node. Being characterized by a prolongation of SA impulse is an exact multiple of the P to P
the sinoatrial conduction time (SACT), the first cycle length, and (3) the P-P intervals until the
SA exit block cannot be recognized on the sur- pause are constant.
face ECG. In the presence of concomitant sinus arrhyth-
The second-degree exit block is further classi- mia causing beat to beat variations, it could be
fied into type I (SA block with Wenckebach con- impossible to distinguish between sinus arrest
duction) and type II (SA Mobitz II). Type I block and SA exit block because of the presence of the
is characterized by the decremental progressive variable and unpredictable long intervals without
lengthening of the conduction between the sinus mathematical relation to short P-P intervals
node and the atria, until a sinus impulse is not (which are not constant). In this case the ECG
conducted to the atria. The classical criteria for panel may be categorized as a pathological or
the diagnosis of SA Wenckebach conduction are: chaotic sinus arrhythmia usually associated with
(1) The P-P interval following the dropped SA sinus bradycardia and representing another sign
impulse is larger than the interval preceding it. of underlying sinus node dysfunction [4].
This phenomenon is common to all Wenckebach Sinoatrial 2:1 block is a pattern that may be
forms, even in the most atypical, and it is due to suspected in the presence of severe sinus brady-
the fact that the lengthening of SA conduction is cardia on ECG (usually <40 bpm). This pattern
present at the beginning of the sequence (the sec- of SA exit block resembles a severe sinus brady-
ond beat in typical series) [3]. (2) The P-P inter- cardia but may be distinguishable from it because
val including a blocked SA impulse is shorter in SA 2:1 block, the acceleration and decelera-
than double (or multiple in case of more than one tion of the sinus rhythm do not occur progres-
consecutively dropped beat) the distance of the sively. For diagnostic purposes, it is useful to
P-P interval preceding it. (3) There is a progres- have a more prolonged ECG recording to follow
sive diminution of the P-P intervals until the the onset/termination of the bradycardia or the

ERRNVPHGLFRVRUJ
22 Sinoatrial Exit Block 259

observation of a sudden heart rate doubling dur- with a collection of cardiac rhythm disturbances
ing atropine test. marked by sinus node inability (sinus node dys-
High-degree type II sinoatrial block occa- function) to generate an atrial rate matching the
sionally more than one sinus impulse in bodys physiologic requirements [5]. When SND
succession fails to reach the atrium, and two or is associated with symptoms or prolonged asys-
three successive P waves are dropped, giving a tole, it is referred as SSS. Sick sinus syndrome is
3:1 or 4:1 exit block and a long pause. The diag- not a disease with a single etiology and
nosis is possible only in the presence of pause pathogenesis.
exactly multiple of an underlying regular sinus Abnormalities encompassed in this syndrome
cycle. Usually it is difficult because this long include various bradyarrhythmias such as sinus
pause is interrupted by the intervention of escaped bradycardia, sinus arrest with or without escape
beats/rhythm. rhythm, or sinus exit block, associated or not
The third-degree (or complete) SA exit block is with supraventricular tachyarrhythmias.
a term used to describe a complete absence of P When complicating supraventricular arrhyth-
waves because no SA node impulse is conducted mias are present, the condition is termed the
to the atria. ECG shows an escape rhythm from a syndrome of alternating bradycardia and tachy-
lower pacemaker site after a sinus pause, and so it cardia or simply the bradycardia-tachycardia
is not distinguishable from a sinus arrest. syndrome (BTS) [6]. This syndrome is more
The occurrence of a sinus pause (or a long P-P common in older patients with advanced sick
interval) on ECG may result also from mecha- sinus syndrome. The most common tachyar-
nisms that should be differentiated: marked sinus rhythmias are atrial fibrillation and atrial flutter
arrhythmia, sinus suppression after ectopic pre- [7, 8]. It is argued that different electrophysiolog-
mature (often atrial) impulses even blocked or ical mechanisms may be responsible for BTS:
conducted, overdrive suppression after ectopic bradycardia associated with SND may favor
tachycardia, and single reciprocating echo P reentrant beats and related tachycardias, while
waves. many evidences suggest that atrial fibrillation and
Respiratory sinus arrhythmia can be easily atrial flutter can lead to atrial remodeling and
recognized because the cyclic lengthening of P-P subsequent SND [9, 10].
interval is usually gradual and phasic; on the Clinically, SSS may produce a variety of ECG
other hand, non-respiratory sinus arrhythmia manifestations consisting in inappropriate sinus
presents variable and unpredictable long P-P bradycardia, SA exit block or sinus arrest, pro-
intervals with different durations and no mathe- longed sinus arrest with failing ectopic pace-
matical relationship with short P-P intervals. maker, persistent atrial or atrioventricular escape
rhythm, episodes of alternating supraventricular
tachyarrhythmias with bradyarrhythmias, and
Clinical Presentation: The Sick Sinus long pause following cardioversion of atrial
Syndrome tachyarrhythmia.
Chronotropic incompetence (CI) is another
SA exit block may be chronic but often occurs as manifestation of SSS. It is defined as a failure to
intermittent episodes, so patients may have nor- achieve 85 % of the maximum predicted heart
mal sinus rhythm for several days or weeks rate during exercise testing.
between episodes. Furthermore, patients with SA
block usually have additional rhythm disturbance Prevalence of Sinus Node Dysfunction
because this arrhythmia is included in a broader and SSS
clinical scenario, the so-called sinus node dys- Bradycardia is frequently seen in healthy indi-
function or sick sinus syndrome. viduals, especially in trained subjects, with a
Sick sinus syndrome (SSS) is a term used to waking hour rate usually >40 bpm [11]. In highly
describe the clinical manifestations associated trained athletes, a slower heart rate (3035 bpm)

ERRNVPHGLFRVRUJ
260 A. Romandini and L. Scappini

may be seen, so that only profound sinus brady- symptoms for months or years like dizziness,
cardia and/or marked sinus arrhythmia (heart rate light-headedness, confusion, exertional fatigue,
less than 30 bpm and/or pauses 3 s during wake dyspnea, weakness, nocturnal wakefulness, and
hours) needs to be distinguished from sinus node vague digestive disturbance. Patients with BTS
disease [12]. may have also palpitations or peripheral throm-
Heart rate between 30 and 35 bpm, asymp- boembolic accidents related to atrial flutter or
tomatic sinus pauses lasting between 2 and 3 s, atrial fibrillation episodes.
escape junctional beats or rhythms (with func-
tional AV dissociation), and first- and second- Causes of SSS
degree atrioventricular nodal block are normal Sometimes SND is associated with abnormalities
variants during sleep [11]. intrinsic to the sinus node that include degenera-
Even if asystole longer than 3 s and heart rate tive age-related fibrosis (idiopathic SSS),
<20 bpm are usually secondary to SND [13], this ischemia-induced fibrosis (e.g., coronary artery
condition per se is not sufficient to define a clini- disease, arteritis), infiltrative disease process
cal disorder like SSS (a pause of at least 6 s is (amyloidosis, sarcoidosis, connective tissue dis-
necessary to cause symptoms) [14, 15]. ease, hemochromatosis, tumors), remodeling of
The exact incidence of SND is unknown. It the SA node associated with heart failure or atrial
may develop at any age, but it is primarily fibrillation, primary ion channel dysfunction
diseased in the elderly with the average age of (familiar or congenital form), and surgical injury.
occurrence being about 68 years [16, 17]. In Moreover, many extrinsic factors, usually
young patients it is often related to underlying transient and reversible, can alter SA node func-
heart disease or previous cardiac surgery. The tion, mimicking the SSS (or exacerbating an
natural history of SND may be highly variable, underlying subclinical intrinsic SA node dys-
although it tends to be progressive. SSS approxi- function): autonomic dysfunction of vagal reflex
mately affects 1 in 600 cardiac patients older than (vasovagal, situational, carotid sinus hypersensi-
65 years [18] and for this reason is more preva- tivity), increased vagal tone (training, sleep,
lent in countries with longer life expectancies. anesthesia, and surgical interventions), metabolic
Currently, in Europe, SSS represents the second disturbances (electrolyte abnormalities like
leading cause of permanent pacemaker implanta- hypo-/hyperkalemia, hypocalcemia, hypother-
tion after AV block [1924]. mia, hypoxia, hypothyroidism), obstructive sleep
apnea, drugs (beta-blockers, calcium channel
Sign and Symptoms blockers, digoxin, antipsychotic agents,
All symptoms would be uncommon in the early membrane-active antiarrhythmics like class I and
disease course. They are related to low cardiac preferentially IA and IC, amiodarone, sympatho-
output that occurs with brady- and/or tachyar- lytic medication, morphine), increased intracra-
rhythmias, causing acute end-organ hypoperfu- nial pressure, and temporal lobe seizure.
sion or worsening preexisting organ failure (heart
failure, angina pectoris, or cerebral vascular acci- Clinical Diagnosis: Diagnostic Workup
dent). The extent of bradycardia or length of The cornerstone for the diagnosis of SSS is the
pause that results in symptoms however varies correlation of end-organ perfusion symptoms with
among individuals because stroke volume, the occurrence of bradyarrhythmias, with or with-
peripheral resistance, and local vascular patency out tachycardia. The presence of the sinus node
also contribute to the extent of regional blood dysfunction should always trigger a proper workup
flow at any heart rate. to rule out reversible extrinsic causes before the
Cerebral hypoperfusion is the most common definitive diagnosis of SSS is made. For this pur-
manifestation often causing abrupt symptoms pose, a full review of systemic conditions and
like near fainting or syncopal episodes. However, medications used is necessary as these conditions
many patients may have mild and nonspecific could be potential reversible causes of SSS.

ERRNVPHGLFRVRUJ
22 Sinoatrial Exit Block 261

Since manifestations of SSS may be erratic steadily stored, with the oldest ECG samples
especially in the early stages, the resting ECG being overwritten by the newest (typically
could not reveal abnormal heart rhythm. 1020 min in duration). When activated
Furthermore, symptoms are often nonspecific (patient activated or by auto-trigger algo-
(e.g., dizziness, fatigue, weakness, or heart fail- rithm), data are stored for a programmable
ure) and in common with other disorders occur- fixed amount of time before and after the acti-
ring with progressive aging. vation. Recently, a new generation of wear-
Depending on the severity of symptoms and able garment-inserted ECG system has been
the overall clinical risk stratification, this can be developed (Nuubo system).
done by in-hospital telemetry or by outpatient Mobile cardiac outpatient telemetry (MCOT)
cardiac monitoring devices. systems are devices made up of an external
The true challenge in SSS is to capture a loop recorder with a portable receiver that
recording of the cardiac rhythm during symp- allows wireless transmission of ECG data
toms, so that the type of device that has to be cho- (non-real time or in real time) to a remote
sen depends on the frequency of symptoms. operating center or to a dedicated website
Ambulatory Holter monitoring that allows via the mobile telephone line or Internet.
recording for 2448 h may be sufficient for Typical duration use is 14 weeks. However,
patients with almost daily symptoms. In other nowadays appropriate use guidelines for this
patients with intermittent symptoms, longer mon- type of ECG monitor have not yet been
itoring is often necessary. Recently, several new developed.
external long-term devices have been developed:
Implantable loop recorders (ILR) are leadless
Patch-type long-term external Holter recorders subcutaneously implanted arrhythmia monitor-
using comfortable adhesive patches that enable a ing devices requiring a minor invasive procedure
continuous single-lead monitoring up to 14 through a small skin incision of about 2 cm in the
days. left precordial region. A single-lead ECG signal
Event recorders (post-event/symptom) are is recorded through two electrodes within the
small portable devices applied to the patients device. The memory loop is activated by the
skin and manually activated whenever symp- patient through an external activator or even
toms are experienced. These devices usually automatically (auto-trigger algorithm). ILR can
provide one-lead electrocardiographic record- be used by patients for a long period of time (up
ing for 3090 s (with up to 610 min of mem- to 3 years with the newest devices) until a diag-
ory storage capacity) that needs to be nosis is reached or the battery runs down. This
transmitted transtelephonically or via mail to type of monitoring is considered when all other
a central monitoring site (healthcare provider) investigation results are inconclusive.
for validation and analysis. Recently, a new Other tests to identify the presence of SND
generation of event recorder smartphone- may include the exercise testing and intracardiac
based device has been launched for this pur- electrophysiologic tests.
pose (AliveCor). However, this type of Exercise testing may confirm the diagnosis of
monitoring has several limitations because its chronotropic incompetence but is not useful for
useless in patients with syncope or short detecting the arrhythmias that occur in SSS.
arrhythmias and has no pre-event data. Electrophysiological study (EPS) may be
External loop recorders (looping memory sys- sometimes helpful in patients with highly sus-
tem or continuous loop recorder) are devices pected SSS and no symptom-rhythm correlation
connected continuously to the patient by demonstration after prolonged cardiac monitor-
means of skin ECG electrodes and equipped ing. It is no longer routinely recommended for
with a memory loop. The device has a built-in diagnosis because of its poor sensitivity and
looping memory in which the ECG signal is specificity (about 50 %) [25]. The two most

ERRNVPHGLFRVRUJ
262 A. Romandini and L. Scappini

common EPS tests for sinus node function severity and frequency of symptoms and the
measure are: overall clinical risk stratification.
D. In a symptomatic patient in whom ECG
1. The sinus node recovery time (SNRT), i.e., the events compatible with SSS fail to correlate
time taken for the sinus rhythm to resume with symptoms, consider implantable loop
after 30 s or 1 min of overdrive atrial pacing. recorder or additional laboratory testing
This interval is measured in the high lateral (exercise stress testing, electrophysiological
right atrium from the last paced beat to the study, carotid sinus massage, tilt test).
first spontaneous sinus beat. This test is based
on the assumption that automaticity sup-
pressed by overdrive recovers less rapidly Treatment
when the SA node is dysfunctional, so the lon-
ger the sinus node takes to recover, the more Nowadays there are no medications that reliably
abnormal sinus node function is evident. Its increase the heart rate in patients with bradyar-
poor diagnostic yield may be due to the fact rhythmias. Cardiac pacing therapy through an
that sinus recovery time is a more complex artificial pacemaker is the only choice.
event and many factors besides automaticity As discussed above, early identification of a
are involved. A delay of longer than 1500 ms potential reversible cause is the first step of
is abnormal. The corrected value (CSNRT) treatment. Discontinuation or dose reduction of
can be determined by subtracting the intrinsic nonessential concomitant drugs affecting sinus
sinus cycle length from the SNRT value: val- node function is recommended to see whether
ues longer than 550 ms suggest sinus node normal sinus function returns. All other medical
dysfunction. conditions which may precipitate the SSS should
2. The other EPS test for sinus node function be treated.
measure is the sinoatrial conduction time If bradyarrhythmia persists, indications for
(SACT) used for detecting delayed conduc- cardiac pacing therapy should be evaluated. The
tion between the sinus node and surrounding cause-effect correlation between symptoms and
atrial tissue. This technique involves resetting bradyarrhythmias is an essential critical step
the sinus node by an eight-beat period of atrial when positive effects of pacemaker placement
pacing at rates just above sinus (Narula proto- are evaluated. Indeed, there is no evidence that
col): the difference between the post-pacing cardiac pacing prolongs survival in patients with
pause and the sinus cycle length would equal SND [2628]. The primary goals of pacemaker
to the SACT. SACT values greater than placement are symptom relief and improved
120 ms are considered abnormal. quality of life.
However, in clinical practice some patients
Our approach to the diagnosis of SSS is sum- affected by sinus node disease may not have a
marized as follows: clear symptom-ECG correlation because of the
slow and erratic course of syndrome and frequent
A. Perform a comprehensive history and physi- comorbidities. In such circumstances, in the
cal examination. absence of other symptoms clearly attributable to
B. Carefully review for systemic conditions and SND, the mechanism of intermittent clinical
medications used as potential remediable significant events like syncope remains uncertain
causes for apparent SSS. as other competitive causes (e.g., autonomic dys-
C. Obtain a resting ECG and if nondiagnostic function) may play an important role. From a
proceed with in-hospital telemetry or ambu- practical perspective, when a competitive
latory ECG monitoring to identify episode of diagnosis can be ruled out (after head-up tilt
bradycardia and eventually correlated symp- testing, carotid sinus massage, and eventually an
toms. The type of monitoring depends on the EPS excluding a very prolonged csnrt, i.e.,

ERRNVPHGLFRVRUJ
22 Sinoatrial Exit Block 263

Table 22.1 The following table is a summary of the main recommendations in the European Society of Cardiology
guidelines on cardiac pacing (ESC guidelines on cardiac pacing 2013)
Recommendations Class Level
Pacing is indicated when symptoms can clearly be attributed to persistent bradycardia or to I B
documented (intermittent) sinus arrest or sinoatrial block
Pacing may be indicated when symptoms are likely to be due to bradycardia, even if the IIb C
evidence is not conclusive
Pacing is not indicated in patients with bradycardia which is asymptomatic or due to III C
reversible causes
Dual-chamber pacing mode with preservation of spontaneous AV conduction is indicated I A (vs. VVI)
for reducing the risk of AF and stroke, avoiding PM syndrome and improving quality of life B (vs. AAI)

>800 ms), cardiac pacing may be reasonable in 4. Oreto G (1997) I disordini del ritmo cardiaco. 2nd
patients with syncope and intrinsic node disease edition. Torino: Centro Scientifico Editore
5. Rubenstein JJ et al (1972) Clinical spectrum of the
with documentation of significant asymptomatic sick sinus syndrome. Circulation 46(1):513
pauses (>6 s). 6. Short DS (1954) The syndrome of alternating brady-
Table 22.1 lists the latest practice recommen- cardia and tachycardia. Br Heart J 16:208
dations with level of evidence from the European 7. Gomes JA, Kang PS, Matheson M et al (1981)
Coexistence of sick sinus rhythm and atrial flutter-
Society of Cardiology task force on cardiac fibrillation. Circulation 63:8086
pacing (2013) in patients with sinus node 8. Ferrer MI (1968) The sick sinus syndrome in atrial
disease [25]. disease. JAMA 206:645646
In SSS dual-chamber pacing mode with 9. Elvan A, Wylie K, Zipes DP (1996) Pacing-induced
chronic atrial fibrillation impairs sinus node function
preservation of spontaneous AV conduction (AV in dogs. electrophysiological remodeling. Circulation
delay management algorithm) is the first choice. 94(11):29532960
A recent study [29] suggests that dual-chamber 10. Sparks PB, Jayaprakash S, Vohra JK, Kalman JM
pacing mode, compared to AAI and VVI pacing (2000) Electrical remodeling of the atria associated
with paroxysmal and chronic atrial flutter. Circulation
mode, confers a modest reduction in AF and 102(15):18071813
stroke (but not in hospitalization and death) and 11. Mangrum JM, DiMarco JP (2000) The evaluation and
reduces the risk of PM syndrome (compared to management of bradycardia. N Engl J Med
VVI pacing mode) which is associated with a 342:703709
12. Corrado D, Pelliccia A, Heidbuchel H et al (2010)
reduction in quality of life. Furthermore, patients Section of Sports Cardiology, European Association
with sick sinus syndrome have also a slightly of Cardiovascular Prevention and Rehabilitation.
increased risk of developing atrioventricular Recommendations for interpretation of 12-lead
block (0.61.9 %/year) [2932]. This result electrocardiogram in the athlete. Eur Heart J
31:243259
supports the advantages of dual-chamber over 13. Northcote RJ, Canning GP, Ballantyne D (1989)
single-chamber pacing mode in this condition. Electrocardiographic findings in male veteran endur-
ance athletes. Br Heart J 61:155160
14. Wieling W, Thijs RD, van Dijk N et al (2009)
Symptoms and signs of syncope: a review of the link
References between physiology and clinical clues. Brain 132
15. Menozzi C, Brignole M, Lolli G et al (1993)
Follow-up of asystolic episodes in patients with car-
1. Surawicz B, Knilans TK (2008) Chous electrocardi-
dioinhibitory, neurally mediated syncope and VVI
ography in clinical practice, 6th edn. Saunders,
Philadelphia pacemaker. Am J Cardiol 72:11521155
2. Schamroth L, Dove E (1966) The Wenckebach phe- 16. Adn V, Crown LA (2003) Diagnosis and treatment of
nomenon in sino-atrial block. Br Heart J 28:350358 sick sinus syndrome. Am Fam Physician 67:17251732
3. Cabeen WR Jr, Roberts NK, Cbild JS (1978) 17. Jensen PN, Gronroos NN, Chen LY et al (2014)
Recognition of the Wenckebach phenomenon (clini- Incidence of and risk factors for sick sinus syndrome
cal notes in diagnostic cardiology). West J Med in the general population. J Am Coll Cardiol
129:521526 64(6):531538

ERRNVPHGLFRVRUJ
264 A. Romandini and L. Scappini

18. Rodriguez RD, Schocken DD (1990) Update on sick collaboration with the European Heart Rhythm
sinus syndrome, a cardiac disorder of aging. Geriatrics Association (EHRA). Eur Heart J 34:22812329
45:2630 26. Alboni P, Menozzi C, Brignole M et al (1997) Effects
19. Coma Samartin R, Sancho-Tello de Carranza MJ, of permanent pacemaker and oral theophylline in sick
Ruiz Mateas F et al (2011) Spanish pacemaker regis- sinus syndrome the THEOPACE study: a randomized
try. Eighth official report of the Spanish Society of controlled trial. Circulation 96(1):260266
Cardiology Working Group on Cardiac Pacing (2010). 27. Shaw DB, Holman RR, Gowers JI (1980) Survival in
Rev Esp Cardiol 64:11541167 sinoatrial disorder (sick-sinus syndrome). Br Med J
20. Cunningham D, Charles R, Cunningham M, de Lange 280:139141
A (2010) Cardiac rhythm management: UK National 28. Sutton R, Kenny RA (1986) The natural history of
Clinical Audit. http://www.ucl.ac.uk/nicor/audits/car- sick sinus syndrome. Pacing Clin Electrophysiol
diacrhythmmanagement/publicreports/pdfs/ 9:11101114
Heartrhythm10 29. Nielsen JC, Thomsen PE, Hojberg S et al (2011) A
21. Markewitz A (2010) The German Pacemaker Register. comparison of single-lead atrial pacing with dual-
Herzschrittmacherther Elektrophysiol 21:248255 chamber pacing in sick sinus syndrome. Eur Heart J
22. Proclemer A, Ghidina M, Gregori D et al (2010) 32:686696
Trend of the main clinical characteristics and pacing 30. Andersen HR, Thuesen L, Bagger JP et al (1994)
modality in patients treated by pacemaker: data from Prospective randomised trial of atrial versus ventricu-
the Italian Pacemaker Registry for the quinquennium lar pacing in sick-sinus syndrome. Lancet
200307. Europace 12:202209 344:15231528
23. Swedish ICD and Pacemaker Register (2010) Annual 31. Castelnuovo E, Stein K, Pitt M et al (2005) The effec-
statistical report 2010. www.pacemakerregistret.se tiveness and cost-effectiveness of dual-chamber pace-
24. Tuppin P, Neumann A, Marijon E et al (2011) makers compared with single-chamber pacemakers
Implantation and patient profiles for pacemakers and for bradycardia due to atrioventricular block or sick
cardioverter defibrillators in France (20082009). sinus syndrome: systematic review and economic
Arch Cardiovasc Dis 104 evaluation. Health Technol Assess 9:iii, xixiii, 1246
25. Brignole M, Auricchio A, Baron-Esquivias G et al 32. Nielsen JC, Kristensen L, Andersen HR et al (2003) A
(2013) Guidelines on cardiac pacing and cardiac randomized comparison of atrial and dual-chamber
resynchronization therapy: the task force on car- pacing in 177 consecutive patients with sick sinus
diac pacing and resynchronization therapy of the syndrome: echocardiographic and clinical outcome. J
European Society of Cardiology (ESC). Developed in Am Coll Cardiol 42:614623

ERRNVPHGLFRVRUJ
Atrioventricular Blocks
23
Andrea Romandini and Marco Morelli

23.1 Case Report 1992: aortic valve replacement with a mechan-


ical prosthesis Starr-Edwards n.27.
1996: transient ischemic attack (TIA).
A 69-year-old man was referred to our 2009: admitted to cardiology department
cardiology department from the ER due to fainting. In that circumstance, an
because of the 4-day appearance of asthe- electrophysiological study showed a second-
nia and dyspnea (NYHA III), associated degree atrioventricular block induction located
with sporadic precordial pains. At the above the His bundle; therefore, it was consid-
entrance, ECG revealed a total third-degree ered benign.
atrioventricular block with a ventricular 2010: access to the emergency room for a par-
escape rhythm of 35 bpm. There was atrio- oxysmal tachycardia associated with asthenia.
ventricular dissociation with atrial rate At ECG frequent ventricular premature beats
faster than the ventricular one and left bun- were recorded, and the patient was addressed
dle branch block (LBBB) QRS type. to take amiodarone therapy.

Allergies
Medical History and Cardiovascular
Risk Factors None

Risks factors: arterial hypertension, cigarette


smoking, dyslipidemia, and impaired glucose Medications
tolerance.
VKA, ARB, ASA 100 mg od, amiodarone
200 mg od, and furosemide 25 mg od
A. Romandini (*)
Cardiology department Ospedali Riuniti di Ancona,
Via Conca, 71, Ancona 60126, Italy Vital Signs
e-mail: romandini.andrea@hotmail.it
M. Morelli Temperature, 36.5 C; heart rate, 35 bpm;
Clinica di Cardiologia e Aritmologia, arterial blood pressure, 115/70 mmHg; respi-
Universit Politecnica delle Marche,
Via Conca, 71, Ancona 60126, Italy ratory rate, 16 breaths/min; and oxygen satu-
e-mail: moro@abanet.it ration, 99 %
Springer International Publishing Switzerland 2015 265
A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_23

ERRNVPHGLFRVRUJ
266 A. Romandini and M. Morelli

Physical Examination Instrumental Examination

General appearance: well developed, well ECG (Fig. 23.1a, b) shows a third-degree
nourished, alert, and cooperative atrioventricular block with slow ventricular
Lungs: clear to auscultation and percussion escape rhythm (35 bpm); There is a clear atrio-
without rales, rhonchi, wheezing, or dimin- ventricular dissociation with atrial rate faster
ished breath sounds than the ventricular. QRS has a left bundle branch
Cardiovascular: Normal S1 and S2. No S3, block (LBBB) pattern.
S4, or murmurs. Regular bradycardic rhythm.
No peripheral edema, cyanosis, or pallor. Echocardiogram showed normal dimensions
Warm and well-perfused extremities and function of cardiac chambers (ejection frac-
Abdomen: Positive bowel sounds. Soft, non- tion 5560 %), aortic valve prosthesis normally
distended. No guarding or rebound. No masses functioning, mild mitral regurgitation, and mild
tricuspid regurgitation with a normal pulmonary
artery systolic pressure.
Routine Laboratory Tests

Tests were substantially normal (hemoglobin Clinical Course and Therapeutic


13.6 g/dl, white blood cells 7840/mmc, creati- Management
nine 1.05 mg/dl, potassium 4.6 mEq/l, sodium
140 mEq/l, magnesium 1.6 mg/dl). Creatine A continuous electrocardiographic monitoring was
kinase-MB and troponin I were within normal placed, and we started isoproterenol infusion that
limits, while brain natriuretic peptide (BNP) was increased HR up to 44 bpm (0.02 mcg/kg/min).
slightly increased (499 pg/ml). After a complete diagnostic workup aimed to

a b

Fig. 23.1 (a, b) A third-degree atrioventricular block the ventricular rate is clearly visible. QRS has a left
with slow ventricular escape rhythm (35 bpm) is shown. bundle branch block (LBBB) pattern
Atrioventricular dissociation with atrial rate faster than

ERRNVPHGLFRVRUJ
23 Atrioventricular Blocks 267

a b

Fig. 23.2 (a) Atrial flutter with third-degree atrioventricular block and junctional escape rhythm. (b) The same patient
after cardioversion: sinus rhythm with AV dissociation is shown

the pacemaker leads, and no complications related


to the surgery procedure.
On the 3rd day, the patient was discharged.
A follow-up visit was performed 1 month
later. The patient was completely asymptomatic.
Physical examination was normal. ECG showed
regular rhythm with paced QRS following sinusal
P waves (VDD mode). PMK was interrogated
and did not show any malfunction.

23.2 Atrioventricular Blocks


Fig. 23.3 Chest X-ray showing normal cardiac and pul-
monary dimensions, correct positioning of the pacemaker An AV block is present when the atrial impulse is
leads, and no complications related to the surgery conducted with delay or is not conducted at all to
procedure the ventricle. The block can occur within the AV
node, His bundle, or bundle branches. In the first-
exclude secondary reversible causes for AV block, degree heart block, conduction time is prolonged
a dual-chamber pacemaker was implanted the day but all impulses are conducted. Second-degree
after hospital admission. After implantation ECG heart block is characterized by occasional or
showed sinusal P waves followed by paced QRS repetitive sudden block of conduction of an
complexes (Fig. 23.2a, b). The chest X-ray showed impulse. In third-degree AV block, no impulses
the following things: the normal cardiac and pul- are conducted from atria to ventricles. The term
monary dimensions, and a correct positioning of advanced heart block indicates blockage of two

ERRNVPHGLFRVRUJ
268 A. Romandini and M. Morelli

or more consecutive impulses. Retrograde con- group) is longer than the cycle preceding the
duction can occur in the presence of anterograde blocked impulse [5].
AV block [1, 2]. Type II AV block is often a precursor of syn-
cope and complete AV block, because it is more
likely to be sub-Hisian. Type I AV block with a
First-Degree AV Block normal QRS complex generally does not prog-
ress to more advanced forms of AV conduction
During first-degree AV block, every atrial impulse disturbances. In elderly people, type I AV block
conducts to the ventricles and a regular ventricu- may be associated with a clinical course similar
lar rate is present. The prolonged PR interval to that seen in type II.
exceeds 200 ms in adults. The conduction delay In the acute coronary syndromes (ACS), type
can be located within the AV node (AH interval I AV block generally develops with inferior
prolonged), in the HisPurkinje system (HV infarction, is transient, and does not require tem-
interval prolonged), or both. If the QRS complex porary pacing [4].
has a normal width, the AV delay most likely Type II AV block occurs usually in the setting
resides within the AV node and rarely within the of an anterior myocardial infarction, can require
His bundle. On the contrary, if the QRS has a temporary or permanent pacing, and is associated
bundle branch block pattern, the conduction with a high mortality rate.
delay may be within the AV node or HisPurkinje A high-degree AV block can occur also in
system as well. Enhancement of vagal tone by patients with acute inferior myocardial infarction
carotid massage can cause first-degree AV nodal (MI) and is associated with an extended myocar-
block to progress to type I second-degree AV dial damage and a higher mortality rate than
block [2]. those without AV block. All types of AV blocks
in the setting of an acute MI can be reversible
within 1421 days.
Second-Degree AV Block In type I AV block with a normal QRS dura-
tion, the conduction delay is likely to be at the AV
In second-degree AV block, the nonconducted P node level proximal to the His bundle. A type I
wave can be intermittent and the preceding PR intra-Hisian block is uncommon. Type II AV
interval duration may be fixed or prolonged. In block, particularly in association with a bundle
this type of block, the PQRS relationship is not branch block, may be localized within or below
random. Mobitz type I (LucianiWenckebach) is the HisPurkinje system. Type I AV block in a
characterized by progressive PR prolongation patient with a bundle branch block can be caused
until a nonconducted P wave. In contrast, in by a block in the AV node or in the HisPurkinje
Mobitz type II, the PR interval remains constant system as well [2].
before a P wave is blocked [3, 4]. Type 2:1 AV block can be due to type I or type
During Mobitz type I block, the conduction II AV block.
time delay is longer in the second beat of the Abrupt transient alterations in autonomic tone
Wenckebach group; meanwhile, prolonged con- can also cause sudden block of one or more P
duction time decreases progressively in the sub- waves without altering the PR interval of the con-
sequent beats. The consequence is that the ducted P wave before or after the block. Such
interval between the successive beats progres- condition is a typical normal finding at night dur-
sively decreases, although the conduction time ing sleep. Thus, apparent type II AV block would
increases. The RR interval produced by the block be produced at the AV node. Increased vagal
is shorter than the double of the RR interval that tone, producing an AV block, usually lengthens
precedes the block impulse (the shortest inter- the PP interval at the same time [4].
val). The RR interval that follows the noncon- Vagal stimulation generally increases and
ducted beat (the first beat of the new Wenckebach vagolytic agents decrease the extent of type I AV

ERRNVPHGLFRVRUJ
23 Atrioventricular Blocks 269

block. Atropine can minimally improve conduc- lar rate is less than 40 beats/min. A hereditary
tion in the AV node and markedly faster sinus form caused by degeneration of the His bundle
rate, which may result in higher AV block degree. and bundle branches has been linked to the
Counterwise, carotid sinus massage generally SCN5A gene [2].
improves and atropine worsens AV conduction in Paroxysmal AV block can be caused by a too
patients with a conduction block within or below strong response of the AV node to vagotonic
the His bundle. reflexes. Surgery, electrolyte disturbances, myo-
Similarly, exercise or isoproterenol by increas- endocarditis, tumors, Chagas disease, rheumatoid
ing the sinus rate and improving AV conduction nodules, calcific aortic stenosis, myxedema, poly-
may resolve the supra-Hisian block and impair myositis, infiltrative processes (e.g., amyloidosis,
sub-Hisian one [2]. sarcoidosis, scleroderma), and many other com-
First-degree and type I second-degree AV mon and unusual conditions can produce AV
block can occur in normal healthy children; a block.
Wenckebach AV block can be also normal in In children, the most common cause of AV
well-trained athletes, probably related to a higher block is congenital. Children are most often
vagal tone at rest. asymptomatic, but in some cases symptoms
develop and pacemaker implantation becomes
necessary. Mortality from congenital AV block is
Third-Degree AV Block highest in the neonatal period, decreases during
childhood and adolescence, and increases slowly
In the total AV block, no atrial activity is con- later. Syncope can occur in patients with congen-
ducted to the ventricles; the atria and ventricles ital heart block at any age. It is difficult to predict
are controlled by independent pacemakers the prognosis. A persistent heart rate at rest of
(atrioventricular dissociation) [4]. The atrial 50 beats/min or less correlates with the incidence
pacemaker can be sinus or ectopic (tachycardia, of syncope in children with congenital complete
flutter, or fibrillation) (Fig. 23.4). AV block [2].
Complete AV block may arise at AV node
level (usually congenital), within the bundle of
His, or distally below the His (usually acquired). The association of a long PR plus a right
The ventricular focus that arise below the bundle branch block and left anterior hemi-
region of the block. Sites of ventricular pacemaker block is called bifascicular block.
activity that are in or closer to the His bundle are
more stable and can produce a faster escape rate
than those located distally in the ventricular con-
duction system. The ventricular escape rate in General Management
acquired total heart block is less than 40 beats/
min but can be faster in congenital complete AV First- or second-degree AV blocks are usually
block [5]. asymptomatic. Sometimes clinical manifesta-
Blocks proximal to the His bundle generally tions of first- and second-degree AV block consist
exhibit normal QRS complexes and rates of of palpitations or subjective feelings of the hearts
4060 beats/min. missing a beat. Persistent 2:1 AV block can
In patients with AV nodal block, atropine usu- produce symptoms of chronic bradycardia.
ally speeds both atrial and ventricular rates. Complete AV block can be accompanied by signs
Exercise can reduce the extent of AV nodal block. and symptoms of reduced cardiac output, syn-
An acquired complete AV block is more often cope or presyncope, angina, or palpitations from
distal to the bundle of His because of trifascicular ventricular tachyarrhythmias [2].
conduction disturbance. The QRS complex is Death in patients with untreated atrioventricu-
abnormal (wide QRS complex), and the ventricu- lar (AV) block is due not only to heart failure

ERRNVPHGLFRVRUJ
270 A. Romandini and M. Morelli

Fig. 23.4 The ventricular rate arise below the region of the block. The atrial pacemaker can be (a) ectopic (tachycardia,
flutter, or fibrillation) or (b) sinus

(HF) secondary to low cardiac output but mainly During early in-hospital phase, when conduc-
to sudden cardiac death caused by prolonged tion blocks are likely to be evanescent, vagolytic
asystole or bradycardia-triggered ventricular agents (such as atropine) are useful for patients
tachyarrhythmia [3]. who have AV nodal disturbances, whereas cate-

ERRNVPHGLFRVRUJ
23 Atrioventricular Blocks 271

cholamines (such as isoproterenol) can be used The diagnosis of AV block is usually made
transiently to treat patients who have heart block from standard ECG when persistent. Prolonged
at any site. Isoproterenol should be used with electrocardiogram monitoring strategy [standard
extreme caution in patients who have acute myo- 24 h Holter ECG or implantable loop recorder
cardial infarction. The use of transcutaneous or (ILR)] may be needed to diagnose an intermittent
temporary transvenous pacing is preferable [2]. AV block (Table 23.1). When an AV block is
For symptomatic AV block or high-grade AV suspected but not documented, provocative
block (e.g., infra-Hisian, type II AV block, third- testing or an electrophysiological study (EPS)
degree heart block not caused by congenital AV may also be needed (Table 23.2). Since there is
block), permanent pacemaker is the treatment of no defined heart rate below which treatment is
choice. indicated, correlation between symptoms and
bradyarrhythmia is essential to give the indica-
tion for permanent cardiac pacing.
23.3 Guidelines Tilt table testing and carotid sinus massage are
indicated when reflex syncope is suspected in the
Diagnosis setting of an atypical presentation. Exercise
testing is indicated in patients who experience
Early identification of a potentially reversible syncope during or shortly after exertion.
cause (drug effects, acute myocardial infarction,
intoxication, electrolyte disorders) is the first step
toward treatment.
As long as the stroke volume increases in
Table 23.1 Prolonged electrocardiogram monitoring strat-
order to compensate for the decrease in heart egy (standard 24 h Holter ECG or implantable loop recorder)
rate, patients with bradycardia can remain com- may be needed to diagnose an intermittent AV block
pletely asymptomatic. First-degree AV block and Prolonged
type I second-degree AV block with marked PR electrocardiogram
prolongation (0.3 s) can lead to symptoms, monitoring strategy Provocative test strategy
because atrial contraction occurs very early in Holter Carotid sinus massage
diastole overlapping with early diastolic filling External loop recorder Tilt table test
and diastolic mitral regurgitation may occur Remote at-home Electrophysiological study
telemetry
between the end of atrial filling and the onset of
Implantable loop Exercise test
ventricular contraction. recorder
While the permanent forms of AV blocks are
caused by an intrinsic disease of the AV conduc-
tion system, the etiology of intermittent block Table 23.2 When an AV block is suspected but not docu-
can be difficult to determine. The diagnosis of mented, provocative testing or an electrophysiological
intermittent AV block is often only presumed but study (EPS) may also be needed
not documented by ECG [6]. Frequency of Suggested ECG monitoring
AV block can be asymptomatic in young, symptoms technique
healthy individuals or during sleep, but patients Daily 24 h Holter, in-hospital
telemetric monitoring
with sustained or frequent bradyarrhythmia are
Every 23 days 4872 h Holter, in-hospital
often symptomatic (e.g., easy fatigability,
telemetric monitoring
reduced exercise capacity, symptoms of HF, irri- Every week 7day Holter or external loop
tability, lassitude, and inability to concentrate). recorder
Dizziness, presyncope, and syncope, due to a Every month 1430-day external loop
sudden decrease in cerebral blood flow, are com- recorder
mon symptoms with intermittent severe forms of Less than once per Implantable loop recorder
AV block. month

ERRNVPHGLFRVRUJ
272 A. Romandini and M. Morelli

Indications for Pacing in Patients leading to inadequate cerebral perfusion. In


with Persistent Acquired patients with syncope and documented third- or
Atrioventricular Block second-degree AV block due to intrinsic disease
of the AV conduction system, there is general
consensus that pacing prevents recurrence of syn-
Pacing is not indicated in patients with cope and may improve survival regardless of
AV block which is due to reversible documented symptom ECG correlation [1].
causes (class I, level C). Other types of intermittent AV block include
Pacing should be considered in patients vagal (extrinsic) and idiopathic forms.
with second-degree type 1 AV block In the setting of reflex syncope when ECG
which causes symptoms or is found to symptom correlation is established, there is evi-
be located at intra- or infra-His levels at dence that dual-chamber pacing can at least
EPS (class IIa, level C). partially prevent recurrences, reducing the syn-
Pacing is indicated in patients with cope burden in patients 40 years [6].
third- or second-degree type 2 AV In patients with a clinical diagnosis of reflex
block irrespective of symptom (class I, syncope and asymptomatic pause >6 s, there is
level C). weak evidence that cardiac pacing may be effec-
tive for the reduction of syncopal recurrences.
In patients with reflex syncope, cardiac pacing
should be the last choice in selected patients (e.g.,
In patients with third- or second-degree type 2 elderly; history of recurrent syncope and frequent
AV block, pacing prevents recurrence of syncope injuries, probably due to lack of prodromal
and improves survival in adults [3]. symptoms).
As a general rule, for intermittent bradycardia,
pacing may be required only for short periods of
Intermittent AV Block time; therefore, permanent ventricular stimula-
tion should be avoided through proper device
Persistent AV block clearly indicates an intrinsic setting.
conduction system disease. On the other hand,
the meaning of intermittent block is less clear, Indications for Pacing in Intermittent
resulting from variable contributions of intrinsic Documented AV Block
and extrinsic mechanisms.
Intermittent/paroxysmal AV block that
occurs in patients with underlying heart disease Intermittent/paroxysmal AV block (includ-
and/or BBB is usually considered as a manifes- ing AF with slow ventricular
tation of intrinsic disease of the AV conduction conduction)
system. The diagnosis of intrinsic AV block is Pacing is indicated in patients with
supported by an infra-Hisian block documented intermittent/paroxysmal intrinsic
at the EPS and, if the block onset is located after third- or second-degree AV block
atrial or ventricular premature beats, during (I; C).
increased heart rate (tachy-dependent AV block) Reflex asystolic syncope
or decreased heart rate (brady-dependent AV Pacing should be considered in
block). patients >40 years with recurrent,
During the intermittent block, the cardiac unpredictable, reflex syncopes and
rhythm may become dependent on ectopic, unre- documented symptomatic pause/s
liable, pacemaker sites that often have slow rates due to sinus arrest or AV block or the
(2540 bpm). Syncope can occur due to long combination of the two (IIa; B).
delay before ectopic pacemaker sites take over,

ERRNVPHGLFRVRUJ
23 Atrioventricular Blocks 273

during ILR observation. Decision to implant a


Asymptomatic pauses (AV block) PM in this setting is determined by an individual
Pacing should be considered in riskbenefit evaluation. There are some
patients with history of syncope and subgroups of patients who could benefit from
documentation of asymptomatic this strategy: old patients with unpredictable (no
pauses >6 s due to sinus arrest, sinus- or very short prodromes) and recurrent syncope
atrial block, or AV block (IIa; C). that expose them to high risk of traumatic injury.
Pacing is not indicated in reversible Obviously, for asymptomatic patient with
causes of AB block (III). BBB, permanent PM implantation is not
indicated.

Suspected AV Block in Patient Indication for Cardiac Pacing


with Syncope and Baseline Bundle in Patients with BBB
Branch Block (BBB)

Electrophysiological study (EPS) should mea- BBB, unexplained syncope, and abnor-
sure the Hisventricular (HV) interval at baseline mal EPS: Pacing is indicated in patients
and during stress by incremental atrial pacing. with syncope, BBB, and positive EPS
The progression rate to AV block at 4 years was defined as interval of 70 ms, or sec-
4 % in patients with HV interval <70 ms, 12 % ond- or third-degree HisPurkinje block
in those with HV interval of 70100 ms, and demonstrated during incremental atrial
24 % in those with HV interval >100 ms. The pacing or with pharmacological stress
development of intra- or infra-His block at incre- (I; B).
mental atrial pacing is highly predictive of Alternating BBB: Pacing is indicated in
impending AV block but is rarely observed [3]. patients with alternating BBB with or
In patients with unexplained syncope and without symptoms (I; C).
bifascicular block, EPS is highly sensitive in BBB, unexplained syncope, and nondi-
identifying patients with intermittent or impend- agnostic investigations: Pacing may be
ing high-degree AV block, though a negative considered in selected patients with
electrophysiological investigation cannot rule out unexplained syncope and BBB [old
intermittent/paroxysmal AV block as the cause of patients with unpredictable (no or very
syncope. Indeed, in patients with negative elec- short prodromes) and recurrent syncope
trophysiological studies, intermittent or stable that expose them to high risk of trau-
AV block was still documented by ILR in about matic injury] (IIb; B).
50 % of cases [3]. Asymptomatic BBB: Pacing is not indi-
The alternating bundle branch block is defined cated for BBB in asymptomatic patients
as block in all three fascicles detected on succes- (III).
sive ECGs: right bundle branch block (RBBB)
and left bundle branch block (LBBB) or RBBB
with associated left anterior fascicular block on
one ECG and associated left posterior fascicular Choice of Pacing Mode
block on another ECG]. These patients progress
quickly toward complete AV block. Therefore, a Dual-chamber pacing is superior over ventricular
PM is usually implanted as soon as possible, even pacing as regards symptom improvement, while
in the absence of a history of syncope [3]. there is evidence of non-superiority about sur-
Only half of patients with unexplained syn- vival and morbidity (hospitalization, HF). On the
cope, basal bundle branch block, and nondiag- other hand, bicameral pacing increases complica-
nostic investigations had a documented AV block tion risk and costs [3].

ERRNVPHGLFRVRUJ
274 A. Romandini and M. Morelli

References
Acquired AV block: In patients with sinus
rhythm, dual-chamber PM should be 1. Lee S, Wellens HJ, Josephson ME (2009) Paroxysmal
preferred over single-chamber ventricular atrioventricular block. Heart Rhythm 6(8):12291234
2. Bonow RO, Mann DL (2012) Braunwalds Heart
pacing for avoiding PM syndrome and Disease: a textbook of cardiovascular medicine. 9th
improving quality of life [IIa; A]. edition. Philadelphia: Saunders; pp 818823
3. Brignole M, Auricchio A, Baron-Esquivias G,
Bordachar P, Boriani G, Breithardt OA, Cleland J,
Deharo JC, Delgado V, Elliott PM, Gorenek B, Israel
CRT should be considered if clinical symp- CW, Leclercq C, Linde C, Mont L, Padeletti L, Sutton
toms of HF (NYHA IIIII), a wide QRS complex R, Vardas PE (2013) ESC guidelines on cardiac pacing
(>120 ms with appearance left BBB like), and a and cardiac resynchronization therapy: the task force
on cardiac pacing and resynchronization therapy of
severely reduced LVEF (<35 %) are present [3]. the European Society of Cardiology (ESC). Developed
It is advisable that mode-switch algorithm be in collaboration with the European Heart Rhythm
activated; episodes of AF during follow-up Association (EHRA) 15(8):10701118
should be assessed (to anticoagulant therapy). 4. Pietro D (2008) Aritmie. Fisiopatologia e diagnosi.
Roma: Cesi editor: 6567
In patient with AF associated with AV block, 5. Rolands DJ (2004) Interpretazione dellelettro-
rate-responsive pacing is associated with better cardiogramma. Treviso: Pro.med editor: 502547
exercise performance, improved daily activities, 6. El-Sherif N, Jalife J (2009) Paroxysmal atrioventricu-
decreased dyspnea, reduced chest pain and palpi- lar block: are phase 3 and phase 4 block mechanisms
or misnomers? Heart Rhythm 6(10):15141521
tations, and improved quality of life, compared
with fixed-rate pacing [3].

Permanent AF and AV block: Ventricular


pacing with rate-response function is
recommended (I; C).

ERRNVPHGLFRVRUJ
Part VIII
Channelopathies

ERRNVPHGLFRVRUJ
Brugada Syndrome
24
Alessandro Barbarossa and Giulia Pongetti

24.1 Case Report The patient had a previous (5 years ago) syn-
copal episode also at rest, without prodromes,
that was not deeply evaluated.
A 35-year-old man was referred to the
emergency room (ER) for acute syncope, at
rest, during a febrile state. Its onset was Allergies
sudden and without any prodromic symp-
tom or convulsion. There was a rapid None
recovery of consciousness without any
neurological impairment and loss of urine.
The patient was admitted to our hospital for Medications
further investigations.
None

Medical History and Cardiovascular Vital Signs


Risk Factors
Temperature: 39 C
Smoker Heart rate: 105 bpm
No personal history for cardiovascular dis- Blood pressure: 120/80 mmHg
ease. Previous routine ECGs recorded for Respiratory rate: 18 bpm
professional sport level resulted normal. Oxygen saturation while breathing ambient
Positive familiar history for sudden cardiac air: 98 %
death (SCD): One 40-year-old cousin sud- Glasgow coma scale (GCS): 15
denly died during sleep.

Physical Examination
A. Barbarossa (*) G. Pongetti
Clinica di Cardiologia e Aritmologia, Universit General: No acute distress, alert, awake, and well
Politecnica delle Marche, oriented. Normally developed and nourished
Via Conca, 71, Ancona 60126, Italy Head, eyes, ears, nose, and throat: Normo-
e-mail: alessandro.barbarossa@hotmail.it; cephalic, atraumatic, mucous membranes moist,
giuliapongetti@gmail.com

Springer International Publishing Switzerland 2015 277


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_24

ERRNVPHGLFRVRUJ
278 A. Barbarossa and G. Pongetti

extraocular muscles intact, pupils equally round tion) and the patient was at rest and did not
and reactive to light and accommodation complain of any prodromic symptom (like
bilaterally, bilateral tympanic membrane intact, dizziness, headache, sweating, pallor, and nau-
bilateral sclera anicteric, and no conjunctival sea). Syncope due to orthostatic hypotension
injection seems also unlikely because he was sitting when
Neck: Supple, no jugular venous distention, no the episode occurred; moreover, he was not tak-
lymphadenopathy, and no carotid bruit ing any drug therapy.
Cardiovascular: Regular rate and rhythm; S1 A neurogenic cause of the loss of conscious-
and S2 are normal; no murmurs, rubs, or gal- ness (like epilepsy) could be a possible hypothe-
lops; point of maximal intensity non-displaced sis if we consider the young age and the sudden
and non-sustained; no hepatojugular reflux; onset, but the absence of other signs (tongue bite,
and capillary refill less than 2 s tonic-clonic seizures, typical aura, urinary incon-
Lungs: No rales, rhonchus, or wheezes; no tinence, rapid recover without neurological
egophony; no alterations in tactile fremitus; impairment) made it less likely.
and normal percussion According to these clinical features, cardiac
Abdomen: Mild overweight, no pulsatile masses, syncope was the most likely diagnosis. ECG and
normal bowel sounds in all four quadrants, no echocardiogram were performed during hospital-
high-pitched or tinkling sounds, resonant to per- ization together with routine laboratory tests.
cussion, soft, non-distended/non-tender, no
rebound or guarding, no costovertebral angle
tenderness, and no hepatosplenomegaly ECG (Fig. 24.1)
Extremities: No cyanosis or clubbing and mild
peripheral edema Sinus rhythm, heart rate 95 bpm, normal atrio-
Neurological: Cranial nerve II through XII ventricular conduction (PQ 150 ms), cardiac
intact and no focal deficit electrical axis +60, and coved-type ST-segment
Psychiatric: Normal affect, no hallucinations, elevation about 4 mm, followed by a negative T
normal speech, and no dysarthria wave in V1 and an ST-segment elevation with a
Skin: Intact, warm, no rashes, and no lesions saddleback appearance in V2 with a biphasic T
wave. QTc 450 ms

What Are the Possible Causes


of Syncope? [1] Echocardiography

Normal dimension of both atria (LA diameter


1. Cardiac syncope: arrhythmias (brady- or M-mode = 30 mm, LA area 4 c = 16 cm2, RA area
tachycardia) and structural heart disease 4 c = 12 cm2). Normal size and wall thickness of
2. Reflex (neurally mediated): vasovagal, the left ventricle (iLVEDV 61 ml/m2) which
situational, carotid sinus syncope shows a preserved systolic function (ejection
3. Syncope due to orthostatic hypotension: fraction with Simpsons method 0.61). Normal
primary and secondary autonomic fail- size and global function of the right ventricle
ure, drug induced, and volume depletion (TAPSE 24 mm)
4. Neurogenic syncope Normal morphology of the cardiac valves;
mild tricuspid regurgitation and normal systolic
pressure gradient (PASP = 20 mmHg)
The inferior vena cava has a normal size
Reflex syncope seems unlikely because there (16 mm) and physiological collapsing during
were no triggering situations (like emotion, gas- inspiration.
trointestinal stimulation, or postprandial situa- Absence of pericardial effusion

ERRNVPHGLFRVRUJ
24 Brugada Syndrome 279

Fig. 24.1 ECG performed at hospital admission

Normal diastolic pattern without increased Inflammation index: VES 30 mm/h (n.v
filling pressure (E/A 0.8, E/E 3.7, E dec time <27 mm/h) and CRP 0.9 mg/dl (n.v <0.6 ng/ml)
193 m/s).
What Are the Possible Causes
Conclusion There are normal echocardiography of Cardiac Syncope?
findings for a 35-year-old man.
Arrhythmia

Routine Laboratory Tests


Bradycardia
Complete blood count: mild leukocytosis Sinus node dysfunction (including
(13.3 10^3/mmc) bradycardia/tachycardia syndrome)
Cholesterol (total, HDL, LDL) and TG: Atrioventricular conduction system
normal disease (AV blocks)
Hepatic function (GOT, GPT, -GT, ALP, Implanted device malfunction
total bilirubin, direct and indirect): normal Tachycardia
Thyroid function (TSH, FT3, FT4): normal Supraventricular arrhythmias
Renal function (creatinine, BUN): normal Ventricular arrhythmias (idiopathic,
Electrolytes (Na+, K+, Ca++, Mg++, Cl): secondary to structural heart disease
normal or to channelopathies)
Fasting blood glucose: 94 m/dl Drug-induced bradycardia and
(5.22 mmol/l) tachyarrhythmias
Troponin I-hs: 0.006 ng/ml (n.v <0.055 ng/ml)

ERRNVPHGLFRVRUJ
280 A. Barbarossa and G. Pongetti

Structural Disease the Brugada pattern, because the previous ECGs


have been described to be normal. It is well
known that the typical Brugada pattern may vary
Cardiac according to body temperature, autonomic tone,
Cardiac valvular disease and also drug intake. This ECG pattern is
Acute myocardial infarction/ described to be associated with risks of ventricu-
ischemia lar tachyarrhythmias (torsades de pointes or
Idiopathic dilated cardiomyopathy ventricular fibrillation mainly), as possible cause
Hypertrophic cardiomyopathy of syncope or sudden cardiac death.
Arrhythmogenic right ventricular
dysplasia (ARVD) Final diagnosis Brugada syndrome (syncope in
Cardiac masses young man with Brugada type 1 pattern at the
Pericardial disease/tamponade ECG)
Congenital anomalies of coronary An electrophysiological study was specifi-
arteries cally not indicated in this particular situation
Prosthetic valves dysfunction because the patient was symptomatic for unex-
Others plained syncope and there was a familiar exam-
Pulmonary embolism ple of sudden death at young age.
Acute aortic dissection Therefore, we implanted a subcutaneous car-
Pulmonary hypertension dioverter defibrillator (S-ICD) in primary preven-
tion. We chose a subcutaneous device instead of
an intravenous one because sudden death in
Brugada syndrome is due to polymorphic ven-
A precise and careful medical history is the tricular tachycardia (e.g., torsades de pointes) or
key to achieve a correct etiologic diagnosis of ventricular fibrillation (VF) that did not required
syncope. Thanks to this we have already removed anti-tachycardial pacing (ATP) but only defibril-
some possible causes of syncope (orthostatic lation. Moreover, our patient was young and did
hypotension, reflex, and neurological syncope) not require cardiac pacing. Finally, the subcuta-
and epilepsy (we executed an electroencephalog- neous device implantation is less invasive (device
raphy which did not show any abnormality). So lead is extrathoracic), and an eventual explant
we focused on the most likely cardiac causes. (e.g., for infection) is easier compared to an intra-
The patient did not show any metabolic disor- venous device.
der at the laboratory tests or physical signs of Quinidine has been reported as a possible
intoxication. He did not assume any drug as pos- pharmacological aid, but the existing data are not
sible cause of arrhythmias. so numerically sufficient to be considered a life-
Physical exam together with ECG and echo- saving therapy.
cardiography did exclude a structural heart dis-
ease together with pulmonary embolism and
acute aortic dissection. 24.2 Brugada Syndrome
A possible arrhythmic origin seemed to us to
be the most likely cause of this syncope. During Denition
hospitalization, the patient never showed brady-
cardia or paroxystic atrioventricular block of dif- Brugada syndrome is an autosomal dominant
ferent grades. At ECG, the repolarization pattern genetic disorder, first described in 1992, with
did show a typical Brugada type 1 pattern with a variable expression characterized by typical pat-
coved ST-segment elevation 2 mm and a nega- terns on the surface electrocardiogram (ECG)
tive T wave in V1. The febrile state during which and an increased risk of ventricular tachyarrhyth-
the ECG has been recorded could have unmasked mias and sudden cardiac death [2, 3].

ERRNVPHGLFRVRUJ
24 Brugada Syndrome 281

Pattern Versus Syndrome which the elevated ST segment descends


toward the baseline and then rises again to an
Brugada pattern (BrP) and Brugada syndrome upright or biphasic T wave.
(BrS) differ each other for the presence or
absence of symptoms: Moving the right precordial chest leads (V1
V2) up to the second or third intercostal space may
increase the sensitivity of detecting these abnor-
Brugada pattern: patients with typical malities [6]. This may be crucial because those
ECG features who are asymptomatic with a type 1 Brugada pattern only in high chest
and have no other clinical criteria leads seem to have a similar rate of cardiac events
Brugada syndrome: patients with typical during >1 year of follow-up as those with type 1
ECG features who have experienced Brugada ECG in standard position [7]. With that
sudden cardiac arrest (SCA), sustained single ECG recording, it is likely to diagnose a
ventricular tachyarrhythmia, or unex- Brugada type 1 without referring to a drug chal-
plained syncope lenge (see below) in a high percentage of patients.

Epidemiology
ECG Patterns
Prevalence of type 1 Brugada ECG pattern ranges
Initial classification considered three main ECG from 0.012 to 1.0 % in different population regis-
patterns [4], but in the last consensus, this classi- tries [810]. A meta-analysis reports an event
fication was changed unifying types 2 and 3 into rate of 10 % at 2.5 years in patients with type 1
a unique pattern (Fig. 24.2) [5]: ECG pattern [11].

Type 1 (classic Brugada type 1 ECG): ST-segment


elevation (2 mm) descends with an upward Pathogenesis
convexity to an inverted T wave in at least one
right precordial chest lead (V1V2). This is A variety of factors may contribute to the clinical
referred to as the coved type. manifestations of Brugada syndrome including
Type 2 (combined from the original designation cardiac sodium channel (SCN) gene mutation,
of type 2 and 3 patterns): ST segment has a right ventricular abnormalities, autonomic tone,
saddleback STT wave configuration, in and fever.

Fig. 24.2 Current electrocardiographic classification of Brugada pattern

ERRNVPHGLFRVRUJ
282 A. Barbarossa and G. Pongetti

Genetic Brugada syndrome demonstrates the syndrome are sudden cardiac arrest or syncope
autosomal dominant inheritance with variable (due to a tachyarrhythmia). Palpitations are rare
expression of mutation in the cardiac sodium and usually are caused by an episode of atrial fibril-
channel genes (mainly SCN5A) [12]. The lation that is common in Brugada syndrome [18].
ST-segment elevation and T wave inversions seen
in the right precordial leads seem to be due to a Sudden cardiac arrest (SCA) and syn-
different length of the action potentials in the epi- cope One-third of patients have SCA or syncope
cardial compared to the endocardial cells of the as the first manifestation of Brugada syndrome.
right ventricle [3, 13]. Usually these manifestations are more common
during the night and during sleep but not during
Arrhythmias The cells with impaired sodium physical exercise [19].
channel function may fail to propagate the action
potential, resulting in localized conduction Atrial fibrillation Patients with Brugada have an
blocks. Due to the abbreviation of phase two, increased incidence of atrial fibrillation (1020 %).
these cells have a much shorter refractory period Its presence has been associated also with an
and recovery of excitability compared to the sur- increased risk of ventricular fibrillation (VF) [20].
rounding cells. This combination (short refrac-
tory periods and localized conduction blocks) Nocturnal agonal respiration As abovemen-
provides the substrate for localized reentry (phase tioned, ventricular arrhythmias are frequent dur-
2 reentry) between epicardium and endocardium ing sleep; the clinical manifestation of these
as arrhythmia substrate [14]. arrhythmias may be nocturnal agonal respiration
associated with a gasping breath.
Structural abnormalities Historically, Brugada
syndrome is not associated with structural heart
disease. However, new evidence suggests the pres- Diagnosis and Risk Stratication
ence of microscopic area of fibrosis in the right
ventricular output tract (RVOT). Nevertheless, Drug Challenge
further investigations in this filed are necessary. In patients with type 2 Brugada pattern, a type 1
pattern may be unmasked by sodium channel
Fever Data from a retrospective review of blocker drugs (e.g., flecainide, procainamide,
patients with Brugada syndrome suggest that ajmaline). The importance of unmasking the type
fever is a main trigger for ECG changes and also 1 Brugada ECG pattern relates to its relevance in
possibly cardiac arrest [15]. confirming the diagnosis of Brugada syndrome,
particularly in patients without symptoms [4]. The
Autonomic tone An imbalance between sympa- test may be useful in patients with type 2 pattern,
thetic and parasympathetic activation seems to be asymptomatic, and with familiar history for SCD.
also present in the Brugada syndrome, as sug-
gested by frequent nocturnal occurrence of the Flecainide: 2 mg/kg over 10 min
tachyarrhythmias (torsades de pointes) and ST intravenously
elevation occurrence during the recovery phase Procainamide: 10 mg/kg over 10 min
of exercise tests (vagal rebound) [16, 17]. intravenously
Ajmaline: 1 mg/kg over 10 min intravenously

Clinical Features Electrophysiological Study (EPS)


EPS is not indicated in patients with Brugada pat-
Patients with a Brugada pattern may remain asymp- terns and high risks features (previous SCA or
tomatic and the typical pattern may be a casual syncope and positive familiar history). EPS in
finding. Usually the first clinical manifestations of asymptomatic patients with type 1 pattern remains

ERRNVPHGLFRVRUJ
24 Brugada Syndrome 283

an area of investigation and debate in which there Class IIb: ICD implantation may be considered
is discordant evidence [21, 22]. Despite this, 2005 in patients with a diagnosis of Brugada syn-
guidelines recommended EPS test in asymptom- drome who develop VF during programmed
atic patients with type 1 pattern [4]. A recent electrical stimulation (inducible patients).
study founded a short refractory period recovery Class III: ICD implantation is not indicated in
(<200 ms) as an adjunctive parameter to possibly asymptomatic patients with a drug-induced
indicate an ICD implantation [21]. type 1 ECG and on the basis of a family his-
tory of SCD alone.
Genetic Testing [12]
Comprehensive or BrS1 (SCN5A) targeted Pharmacological therapy plays a marginal role
BrS genetic testing can be useful particularly in Brugada syndrome; quinidine has been shown
for the patients in which there is a clinical to prevent induction of VF and suppress spontane-
index of suspicion for BrS based on examina- ous ventricular arrhythmias in a clinical setting.
tion of the patients clinical history, family However, quinidine is recommended only as [23]:
history, and expressed electrocardiogram
(resting 12-lead ECGs and/or provocative Class IIa: patients with ICD and multiple shocks
drug challenge testing). Class IIa: patients with contraindication to ICD
Genetic testing is not indicated in the setting
of an isolated type 2 Brugada ECG pattern. Conclusions
Brugada syndrome is one possible cause of
Prognostic Factors SCD in the young population with an incidence
The major risk factor for patients with the Brugada that is probably underestimated. ECG is funda-
ECG pattern is a history of ventricular tachyar- mental for diagnosis; however, it is important to
rhythmias leading to SCA or syncope; these remember that the typical pattern may not
patients should be considered at high risk [4, 22]. always be founded in all the consecutive ECGs
Atrial fibrillation, positive familiar history for in the same subject, because it can vary in rela-
SCD, and male gender are less powerful predic- tion to different environmental conditions.
tors of future events. About risk stratification, medical history plays
a central role because history of syncope or
SCA is so far the only major risk criteria.
Treatment ICD implantation is the only effective
therapy for these patients at the moment.
To date, the only proven effective therapeutic S-ICD could be chosen because of the exter-
strategy for the prevention of SCD in BrS patients nal lead in a relative young patient not needing
is the implantable cardioverter defibrillator ATP therapy or cardiac pacing.
(ICD), and this therapy is principally guided by
the clinical history.
Recommendation [23] are as follows: References

Class I: ICD implantation is recommended in 1. Moya A, Sutton R, Ammirati F, Blanc J-J, Brignole M,
patients with a diagnosis of Brugada syndrome Dahm JB, Deharo J-C, Gajek J, Gjesdal K, Krahn A,
Massin M, Pepi M, Pezawas T, Ruiz Granell R, Sarasin
who are survivors of a cardiac arrest and/or have F, Ungar A, van Dijk JG, Walma EP, Wieling W (2009)
documented spontaneous sustained ventricular Guidelines for the diagnosis and management of syn-
tachycardia (VT) with or without syncope. cope (version 2009). Eur Heart J [Internet]. 30:2631
Class IIa: ICD implantation can be useful in 2671. [Cited 2012 Jul 12]. Available from: http://www.
pubmedcentral.nih.gov/articlerender.fcgi?artid=32955
patients with a spontaneous diagnostic type 1 36&tool=pmcentrez&rendertype=abstract
ECG who have a history of syncope judged to 2. Brugada P, Brugada J (1992) Right bundle branch
be likely caused by ventricular arrhythmias. block, persistent ST segment elevation and sudden

ERRNVPHGLFRVRUJ
284 A. Barbarossa and G. Pongetti

cardiac death: a distinct clinical and electrocardio- an urban population in the United States. Pacing Clin
graphic syndrome. A multicenter report. J Am Coll Electrophysiol [Internet]. 32:704708. [Cited 2014
Cardiol [Internet]. 20:13911396. [Cited 2014 Dec Dec 17]. Available from: http://www.ncbi.nlm.nih.
10]. Available from: http://www.ncbi.nlm.nih.gov/ gov/pubmed/19545330
pubmed/1309182 11. Gehi AK, Duong TD, Metz LD, Gomes JA, Mehta D
3. Alings M, Wilde A (1999) Brugada syndrome: clin- (2006) Risk stratification of individuals with the
ical data and suggested pathophysiological mecha- Brugada electrocardiogram: a meta-analysis. J
nism. Circulation [Internet]. 99:666673. [Cited 2014 Cardiovasc Electrophysiol [Internet]. 17:577583.
Dec 17]. Available from: http://www.ncbi.nlm.nih. [Cited 2014 Dec 17]. Available from: http://www.
gov/pubmed/9950665 ncbi.nlm.nih.gov/pubmed/16836701
4. Antzelevitch C, Brugada P, Borggrefe M, Brugada J, 12. Ackerman MJ, Priori SG, Willems S, Berul C,
Brugada R, Corrado D, Gussak I, LeMarec H, Brugada R, Calkins H, Camma J, Ellinor PT, Gollob
Nademanee K, Perez Riera AR, Shimizu W, Schulze- M, Hamilton R, Hershberger RE, Judge DP, Le Marec
Bahr E, Tan H, Wilde A (2005) Brugada syndrome: H, McKenna WJ, Schulze-Bahr E, Semsarian C,
report of the second consensus conference: endorsed Towbin J a, Watkins H, Wilde A, Wolpert C, Zipes DP
by the Heart Rhythm Society and the European Heart (2011) HRS/EHRA expert consensus statement on the
Rhythm Association. Circulation [Internet]. 111:659 state of genetic testing for the channelopathies and
670. [Cited 2012 Nov 3]. Available from: http://www. cardiomyopathies this document was developed as a
ncbi.nlm.nih.gov/pubmed/15655131 partnership between the Heart Rhythm Society (HRS)
5. Bays de Luna A, Brugada J, Baranchuk A, Borggrefe and the European Heart Rhythm Association (EHRA).
M, Breithardt G, Goldwasser D, Lambiase P, Riera Heart Rhythm [Internet]. 8:13081339. [Cited 2012
AP, Garcia-Niebla J, Pastore C, Oreto G, McKenna Jul 18]. Available from: http://www.ncbi.nlm.nih.gov/
W, Zareba W, Brugada R, Brugada P (2012) Current pubmed/21787999
electrocardiographic criteria for diagnosis of Brugada 13. Gussak I, Antzelevitch C, Bjerregaard P, Towbin JA,
pattern: a consensus report. J Electrocardiol [Internet]. Chaitman BR (1999) The Brugada syndrome: clini-
45:433442. [Cited 2014 Dec 10]. Available from: cal, electrophysiologic and genetic aspects. J Am Coll
http://www.ncbi.nlm.nih.gov/pubmed/22920782 Cardiol [Internet]. 33:515. [Cited 2014 Dec 17].
6. Sangwatanaroj S, Prechawat S, Sunsaneewitayakul B, Available from: http://www.ncbi.nlm.nih.gov/
Sitthisook S, Tosukhowong P, Tungsanga K (2001) pubmed/9935001
New electrocardiographic leads and the procainamide 14. Aiba T, Shimizu W, Hidaka I, Uemura K, Noda T,
test for the detection of the Brugada sign in sudden Zheng C, Kamiya A, Inagaki M, Sugimachi M,
unexplained death syndrome survivors and their rela- Sunagawa K (2006) Cellular basis for trigger and
tives. Eur Heart J [Internet]. 22:22902296. [Cited maintenance of ventricular fibrillation in the Brugada
2014 Dec 9]. Available from: http://www.ncbi.nlm. syndrome model: high-resolution optical mapping
nih.gov/pubmed/11728150 study. J Am Coll Cardiol [Internet]. 47:20742085.
7. Miyamoto K, Yokokawa M, Tanaka K, Nagai T, [Cited 2014 Dec 17]. Available from: http://www.
Okamura H, Noda T, Satomi K, Suyama K, Kurita T, ncbi.nlm.nih.gov/pubmed/16697328
Aihara N, Kamakura S, Shimizu W (2007) Diagnostic 15. Singh I, Ramteke VK (1996) The role of omental
and prognostic value of a type 1 Brugada electrocar- transfer in Buergers disease: New Delhis experience.
diogram at higher (third or second) V1 to V2 record- Aust N Z J Surg [Internet]. 66:372376. [Cited 2014
ing in men with Brugada syndrome. Am J Cardiol Dec 17]. Available from: http://www.ncbi.nlm.nih.
[Internet]. 99:5357. [Cited 2014 Dec 17]. Available gov/pubmed/8678856
from: http://www.ncbi.nlm.nih.gov/ 16. Matsuo K, Kurita T, Inagaki M, Kakishita M, Aihara
pubmed/17196462 N, Shimizu W, Taguchi A, Suyama K, Kamakura S,
8. Matsuo K, Akahoshi M, Nakashima E, Seto S, Yano Shimomura K (1999) The circadian pattern of the
K (2004) Clinical characteristics of subjects with the development of ventricular fibrillation in patients with
Brugada-type electrocardiogram. J Cardiovasc Brugada syndrome. Eur Heart J [Internet]. 20:465
Electrophysiol [Internet]. 15:653657. [Cited 2014 470. [Cited 2014 Dec 15]. Available from: http://
Dec 17]. Available from: http://www.ncbi.nlm.nih. www.ncbi.nlm.nih.gov/pubmed/213350
gov/pubmed/15175059 17. Miyazaki T, Mitamura H, Miyoshi S, Soejima K,
9. Miyasaka Y, Tsuji H, Yamada K, Tokunaga S, Saito Aizawa Y, Ogawa S (1996) Autonomic and antiar-
D, Imuro Y, Matsumoto N, Iwasaka T (2001) rhythmic drug modulation of ST segment elevation in
Prevalence and mortality of the Brugada-type electro- patients with Brugada syndrome. J Am Coll Cardiol
cardiogram in one city in Japan. J Am Coll Cardiol [Internet]. 27:10611070. [Cited 2014 Dec 17].
[Internet]. 38:771774. [Cited 2014 Dec 17]. Available from: http://www.ncbi.nlm.nih.gov/
Available from: http://www.ncbi.nlm.nih.gov/ pubmed/8609322
pubmed/11527631 18. Rodrguez-Maero M, Namdar M, Sarkozy A,
10. Patel SS, Anees S, Anees SS, Ferrick KJ (2009) Casado-Arroyo R, Ricciardi D, de Asmundis C,
Prevalence of a Brugada pattern electrocardiogram in Chierchia G-B, Wauters K, Rao JY, Bayrak F, Van

ERRNVPHGLFRVRUJ
24 Brugada Syndrome 285

Malderen S, Brugada P (2013) Prevalence, clinical 21. Priori SG, Gasparini M, Napolitano C, Della Bella P,
characteristics and management of atrial fibrillation in Ottonelli AG, Sassone B, Giordano U, Pappone C,
patients with Brugada syndrome. Am J Cardiol Mascioli G, Rossetti G, De Nardis R, Colombo M
[Internet]. 111:362367. [Cited 2014 Dec 17]. (2012) Risk stratification in Brugada syndrome:
Available from: http://www.ncbi.nlm.nih.gov/ results of the PRELUDE (PRogrammed ELectrical
pubmed/23206922 stimUlation preDictive valuE) registry. J Am Coll
19. Atarashi H, Ogawa S, Harumi K, Hayakawa H, Cardiol [Internet]. 59:3745. [Cited 2014 Dec 17].
Sugimoto T, Okada R, Murayama M, Toyama J Available from: http://www.ncbi.nlm.nih.gov/
(1996) Characteristics of patients with right bundle pubmed/22192666
branch block and ST-segment elevation in right pre- 22. Brugada J, Brugada R, Brugada P (2003) Determinants
cordial leads. Idiopathic Ventricular Fibrillation of sudden cardiac death in individuals with the elec-
Investigators. Am J Cardiol [Internet]. 78:581583. trocardiographic pattern of Brugada syndrome and no
[Cited 2014 Dec 17]. Available from: http://www. previous cardiac arrest. Circulation [Internet].
ncbi.nlm.nih.gov/pubmed/8806350 108:30923096. [Cited 2014 Dec 17]. Available from:
20. Kusano KF, Taniyama M, Nakamura K, Miura D, http://www.ncbi.nlm.nih.gov/pubmed/14623800
Banba K, Nagase S, Morita H, Nishii N, Watanabe A, 23. Priori SG, Wilde AA, Horie M, Cho Y, Behr ER,
Tada T, Murakami M, Miyaji K, Hiramatsu S, Berul C, Blom N, Brugada J, Chiang C-E, Huikuri H,
Nakagawa K, Tanaka M, Miura A, Kimura H, Fuke S, Kannankeril P, Krahn A, Leenhardt A, Moss A,
Sumita W, Sakuragi S, Urakawa S, Iwasaki J, Ohe T Schwartz PJ, Shimizu W, Tomaselli G, Tracy C (2013)
(2008) Atrial fibrillation in patients with Brugada syn- Executive summary: HRS/EHRA/APHRS expert
drome relationships of gene mutation, electrophysiol- consensus statement on the diagnosis and manage-
ogy, and clinical backgrounds. J Am Coll Cardiol ment of patients with inherited primary arrhythmia
[Internet]. 51:11691175. [Cited 2014 Dec 17]. syndromes. Heart Rhythm [Internet]. 10:e85e108.
Available from: http://www.ncbi.nlm.nih.gov/ Available from: http://www.ncbi.nlm.nih.gov/pub
pubmed/18355654 med/23916535

ERRNVPHGLFRVRUJ
Long QT Syndrome
25
Maria Vittoria Matassini and Alessandro Maolo

25.1 Case Report


Routine blood tests including electro-
lytes were all within the normal ranges.
The 24-h Holter ECG recording revealed
A 33-year-old woman was referred to our during syncope an episode of polymor-
cardiology department after an aborted phic ventricular tachycardia that was
sudden cardiac death. very likely to be a torsade de pointes
Few days before cardiologic visit, an acci- with later spontaneous sinus rhythm
dental diagnosis of long QT interval was restoration, despite the presence of
done. The cardiologist noticed a QT many artifacts (Fig. 25.1).
interval of 560 ms with a cardiac rate of The patient was then transferred to our
58 bpm. Therefore, a 24-h ECG Holter arrhythmology and cardiology clinic for
recording was booked. On the day of the further evaluation and treatment.
recording, the patient collapsed while
standing at the red traffic light. She had a
spontaneous recovery of consciousness
in a few seconds with no memory left. Medical History and Cardiovascular
The patient was then transferred to the ER Risk Factors
at the nearest hospital. She fully recov-
ered without neurological sequelae, and No cardiovascular risks factor.
the ECG showed sinus rhythm with a No family history of sudden cardiac death or
prolonged QT interval (580 ms, 55 bpm). other structural heart diseases.
No history of syncope.
2009: the patient underwent cardiologic visit
for nocturnal episodes of palpitations with
sudden awakening from sleep and sweating.
ECG showed a long QT interval (QTc 560 ms).
M.V. Matassini A. Maolo (*)
Clinica di Cardiologia e Aritmologia Ospedali
Riuniti di Ancona, Via Conca 71, 60126,
Ancona, Italy Allergies
e-mail: mavimatassini@yahoo.it;
alessandro.maolo@alice.it Dust, cat hair, pollen

Springer International Publishing Switzerland 2015 287


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_25

ERRNVPHGLFRVRUJ
288 M.V. Matassini and A. Maolo

Fig. 25.1 Holter ECG recording showing a torsade de pointes. The smaller trace below shows the spontaneous sinus
rhythm recovering

Medications Extremities: no cyanosis or clubbing and no


peripheral edema
None

Routine Laboratory Tests Performed


Vital Signs
Complete blood count: normal
Temperature: 36 C Cholesterol (total, HDL, LDL) and TG:
Heart rate: 55 bpm normal
Arterial blood pressure: 100/60 mmHg Hepatic function (GOT, GPT, -GT, ALP,
Respiratory rate: 18 breaths/min total bilirubin, direct, and indirect): normal
Oxygen saturation: 100 % Thyroid function (TSH, FT3, FT4): normal
Renal function (creatinine, BUN): normal
Serum electrolytes: potassium 4.4 mEq/l,
Physical Examination sodium 138 mEq/l, and magnesium 1.8 mg/dl

General: no fatigue, no acute distress; alert,


awake, and oriented. Well developed and well Instrumental Examination
nourished
Neck: supple, no jugular venous distention, no The recorded ECG (Fig. 25.2a, b) revealed sinus
lymphadenopathy, and no carotid bruit rhythm with 58 bpm heart rate, normal atrioven-
Cardiovascular: regular rate and rhythm, S1 tricular and intraventricular conduction, and nor-
and S2 normal, no murmurs, rubs or gallops, mal ventricular repolarization with a corrected
point of maximal intensity non-displaced and QT interval 600 ms long.
non-sustained, no hepatojugular reflux, and A complete echocardiographic examination
capillary refill less than 2 s was performed and showed normal dimensions
Lungs: no rales, no rhonchi or wheezes, no and function of cardiac chambers, a mild pro-
egophony, no alterations in tactile fremitus, lapse of the posterior mitral leaflet with trace of
and normal percussion mitral regurgitation, and a mild tricuspid regurgi-
Abdomen: no pulsatile masses, normal bowel tation with normal pulmonary artery systolic
sounds in all four quadrants, no high-pitched pressure.
or tinkling sounds, resonant to percussion, No reversible cause of QT interval prolonga-
soft, non-distended/non-tender, no rebound or tion was found.
guarding, no costovertebral angle tenderness, All the family members underwent ECG
and no hepatosplenomegaly evaluation, and either the father or brother

ERRNVPHGLFRVRUJ
25 Long QT Syndrome 289

a b

Fig. 25.2 (a, b) Standard rest 12-lead ECG showing a prolonged QT interval (600 ms)

showed a prolonged QT interval. They both were ECG with or without T wave [1]. These abnormal-
not taking any medications which could alter the ities of ventricular repolarization can be present in
QT interval, and laboratory findings did not show other family members of the affected subject.
electrolyte abnormalities. The natural history of LQTS is characterized
by the development of life-threatening ventricu-
lar arrhythmias such as torsade de pointes (TdP).
Final Diagnosis This arrhythmia can be self-limiting and asymp-
tomatic or they can manifest with syncope or
These findings altogether were suggestive for sudden cardiac death (SCD) [2].
hereditary long QT syndrome. Genetic assessments The prevalence of this genetic disorder is esti-
were therefore performed and it is still underway. mated to be at least 1:5,000 subjects. Considering
that the affected subject can be asymptomatic and
there are cases of sudden cardiac death with no
25.2 Long QT Syndrome defined etiology, the real prevalence of LQTS
may be considerably higher [1, 2].
Epidemiology Moreover, 1040 % of affected patients pres-
ent a nondiagnostic QT interval at rest because of
Hereditary long QT syndrome (LQTS) is a con- an intermittent nature of the ECG abnormalities
genital disorder characterized by a prolongation of or a borderline QT value that make the diagnosis
the QT interval detectable on the standard 12-lead challenging [3].

ERRNVPHGLFRVRUJ
290 M.V. Matassini and A. Maolo

Long QT syndrome more often manifests feet, phenotype known as Timothy syndrome
before puberty in males and after puberty in (LQT8)
females, and the probability of a cardiac event Cavelolin-3 gene, with increase in late sodium
depends on the degree of QT prolongation. current (LQT9)
Among untreated symptomatic patients, mor- SCN4B gene, with increase in late sodium
tality is high, with 20 % of deaths in the first year current (LQT10)
after the first cardiac event and approximately
50 % within 10 years [2]. In addition, 1520 % of affected patients pres-
ent a negative genetic assessment.
The majority of LQTS is inherited as an autoso-
Pathophysiology mal dominant trait: the Romano-Ward syndrome
affects 1 per 5,000 persons. Sporadic (or de novo)
Since the discovery of the first LQTS-responsible alterations occur 510 % of the time. The autosomal
genes in 1990s, a lot more genes have been recessive form of LQTS, also known as Jervell and
related to the syndrome and more than 500 muta- Lange-Nielsen syndrome, was firstly described in
tions are described nowadays, resulting in 10 dif- 1957. It probably affects less than 1 per million peo-
ferent phenotypes (LQTS 110). ple and results from complete loss of Kv7.1 channel
In about 75 % of patients, mutations involve function, which precipitates sensorineural deafness
three main causative genes: in addition to LQT1 or LQT5 mutation [2, 4].

KCNQ1-encoded Kv7.1 channel subunit (IKs


potassium channel alpha subunit; LQT1) Genotype-Phenotype Correlation
KCNH2-encoded Kv11.1 subunit or hERG
(IKr potassium channel alpha subunit; LQT2) The three main genotypes (LQT1, LQT2, and
SCN5A-encoded Nav1.5 (INa sodium chan- LQT3) present distinctive clinical phenotypes
nel alpha subunit, LQT3) with important implications in diagnosis, treat-
ment, and long-term clinical course. Indeed, the
Nearly 70 % of all LQTS are secondary to recognition of gene-specific features can be used
loss-of-function mutations involving IKs (30 to choose the most suitable therapy and to give
35 %) or IKr (2540 %) potassium channels, the correct recommendations to probands and
while approximately 5 % result in a gain-of- their family. The gene-specific phenotypes
function mutation involving sodium channel. include different triggers of ventricular arrhyth-
Less frequently, patients with congenital LQTS mias, different ages of symptom onset, and spe-
present mutation involving: cific ECG morphologies [2, 4].
Patients with LQT1 and less part of LQT2
Ankyrin-B gene: a cytoskeletal membrane patients usually present the greater risk of cardiac
adapter which acts as an anchoring protein events after triggers associated with adrenergic
that binds proteins involved in cardiac electro- stimulation [5]. Life-threatening arrhythmias
physiology and cellular calcium homeostasis have been shown to occur under specific circum-
(LQT4) stances: while diving and swimming for LQT1
The auxiliary subunits to KCNQ1 (mink; patients or acoustic stimuli (such as alarm clock
LQT5) or KCNH2 (MiRP1, LQT6) ringing) or sudden awakening for LQT2 patients
KCNJ2-encoded Kir2.1 potassium channel [4, 68].
with consequent reduction in Kir2.1 current On the other hand, LQT3 is characterized by
(LQTS7), known as Andersen-Tawil the higher risk of events at rest or during sleep.
syndrome (ATS), with the phenotype domi- Patients with LQT3 have fewer events during
nated by skeletal abnormalities exercise or stress because they significantly
CACNA1C-encoded L-type calcium channel shorten their QTc at high frequencies and
subunit with increase in Cav1.2 current and therefore they become less susceptible to
associated with syndactyly in both hands and catecholamine-induced arrhythmias [9]. These
ERRNVPHGLFRVRUJ
25 Long QT Syndrome 291

LQT3 LQT2 LQT1

II

aVF

V5

Fig. 25.3 Distinctive ECG patterns of the three main LQTS genotypes. (With permission from Prof. Arthur Moss with
the consens of the original publisher (Circulation))

Table 25.1 Characteristics of different genotypes of LQTS


QT shortening Age of first
Genotype Higher risk of events ECG during exercise presentation
LQT1 Diving/swimming T waves broad-based with high amplitude No; may lengthen Lower
LQT2 Alarm clock Low-amplitude ST-T in the limb leads, Variable
biphasic in the precordial leads
LQT3 Rest/sleeping Narrow T waves, late-onset peak or Yes
biphasic, long isoelectric ST-T segment

findings have consistent clinical implications and Moreover, every genotype has a different
explain why these patients have less or no benefit response to exercise. LQT1 patients fail to appro-
at all from beta-blocker therapy. priately shorten their QT interval during exercise
Age of first manifestations varies from one and QTc may further lengthen. This effect is due
genotype to another. In fact, generally, the age of to IKs that in normal subjects is responsible for
first events is lower in LQT1 than in LQT2 and the abbreviation of the action potential at higher
LQT3 [4]. heart rates. On the contrary, patients with LQT3
Some differences can be noticed on the stan- show shortening of QT interval during exercise.
dard 12-lead ECG too. Each one of the three In LQT2, QTc duration behavior is variable [4].
major genotypes (LQT1 to LQT3) seems to have Finally, LQT1 patients have a peculiar response
a distinctive T-wave repolarization pattern on the to epinephrine. Epinephrine infusion has an excel-
ECG, as shown in Fig. 25.3. lent performance in the detection of LQTS1
because it provokes a significant and stable pro-
longation of the QTc in these patients [10, 12].
LQT1: T waves are broad-based with a Table 25.1 shows the characteristics of differ-
high amplitude. ent genotypes of LQTS.
LQT2: ST-T segment has a low ampli-
tude in the limb leads and a biphasic
pattern in the precordial leads. Clinical Presentation
LQT3: T waves are narrow, with late-onset
peak or biphasic and preceded by a long The long QT syndromes main manifestations are
isoelectric ST-T segment [1, 2, 4]. electrocardiographic anomalies, syncope, cardiac
arrest, and sudden cardiac death.
ERRNVPHGLFRVRUJ
292 M.V. Matassini and A. Maolo

Table 25.2 Scoring system to evaluate the probability of Some useful tests have been proposed to
LQTS
unmask the concealed forms of LQTS, and ongo-
ECG QTc 480 ms 3 ing clinical trials are evaluating and validating
findings QTc = 460470 ms 2 them. These tests are:
QTc = 450 ms (in males) 1
Torsade de pointes 2 Exercise test: patients with LQT1 mutation
T wave alternans 1 seem to have a more marked QT prolongation
Notched T waves (three leads) 1 during exercise (burst and gradual exercise
Rest heart rate below the second 0.5 showed the same results), while those with
percentile for age
LQT2 mutation have an exceeding hysteresis
Clinical Syncope with stress 2
history of the QT interval (difference between QT
Syncope without stress 1
interval measured during exercise and 2 min in
Congenital deafness 0.5
the recovery phase at similar heart rates)
Family Family members with LQTS 1
history (score 4) [1719].
Unexplained sudden cardiac death 0.5 Changing position: sudden changes from
below age 30 among immediate supine to standing position can provoke a pro-
family members longation of the QT interval mostly in LQT1
and LQT2 patients. This can be explained by
The ECG anomalies are very different and they the complex mechanism involving sympa-
depend in part on the type of QT syndrome. The thetic and vagal systems and their recruitment
main alteration is the prolongation of the QT inter- of IKs and IKr currents for an adequate inotropy
val, but is not always present at rest. In a certain and chronotropy response [17, 18, 20, 21].
percentage of patients, the QT interval can even be Epinephrine test: epinephrine infusion showed
normal (10 % in LQT3 and 37 % in LQT1) [12]. excellent results in unmasking concealed
In these cases, the QT intervals prolongation can forms of LQTS, but the clinical trials available
be evident during exercise test or infusion of epi- by now have few patients, and this test cannot
nephrine [4, 911]. Furthermore, repolarization be used as a routine exam so far.
can have strange aspects. For example, notches on
the T wave are typical of LQT2. According to the latest consensus paper, the
Syncope, cardiac arrest, and sudden death are presence of a pathogenic mutation in LQTS
the result of ventricular arrhythmias due to the genes per se can be a diagnostic criterion [22].
arrhythmogenic condition strictly linked to the A scoring system was created to evaluate the
genetic and electrical disorders of the disease. probability of LQTS when QT interval is normal
These arrhythmias are most of all torsade de or the genetic mutation is not unequivocal [23].
pointes VT that become clinically manifest This scoring system is still used and is shown in
depending on their duration. Sudden death occurs Table 25.2.
when the arrhythmia is not self-limiting and Thats why according to the latest HRS/
degenerates into ventricular fibrillation. EHRA/HPRS consensus document, diagnosis of
LQTS can be surely done if:

Diagnosis There is an LQTS risk score of 3.5 (exclud-


ing secondary causes of QT prolongation).
In many cases, the diagnosis of LQTS can easily There is a pathogenic mutation in LQTS
be done with a standard 12-lead ECG at rest, genes.
measuring the QT interval and correcting it with There is a QTc 500 ms in repeated ECG
Bazetts formula (QTc = QT (ms)/RR(s)). (excluding secondary causes of QT
However, there are hidden cases of LQTS prolongation).
in which the QT interval can be normal or Moreover, LQTS can be diagnosed if there
borderline at rest (about 2025 %) [1216]. is a QTc 499 ms and >480 ms in repeated

ERRNVPHGLFRVRUJ
25 Long QT Syndrome 293

ECG (excluding secondary causes of QT


prolongation). Lifestyle changes: in particular avoid-
ance of hard physical exercise (espe-
cially swimming) in LQT1 subjects,
Management exposure of sudden loud noises (i.e.,
alarm clock or phone ringing) in LQT2
Before deciding the appropriate therapy for a subjects, and any drug prolonging QT
patient affected by LQTS, it is important to make interval in all LQTvS patients. Prompt
a careful risk stratification. Even if in some cases correction of electrolyte impairment is
identifying high-risk patients could be easy (i.e., important too.
some specific genetic mutation such as those Beta-blockers: are the first-line therapy,
found in Jervell and Lange-Nielsen syndrome or and they are indicated with full dosing
in Timothy syndrome [see below]), more fre- for age and weight in all LQTS patients
quently risk stratification becomes a hard matter, (regardless of QT prolongation) in the
and a wrong conclusion about the arrhythmic risk absence of contraindication such as
could be made. asthma. Nadolol and sustained-release
According to the latest consensus document propranolol should be preferred consid-
[22] could be considered as high-risk ering their long-acting activity and their
patients with: demonstrated better efficacy compared
to metoprolol [28].
Jervell and Lange-Nielsen syndrome (1.6 Implantable cardioverter defibrillator
6:1,000,000): particular variant of LQTS (ICD): is recommended in secondary
associated with severe and bilateral sensori- prevention (patients who survived car-
neural deafness, caused in 90 % of the cases diac arrest) and in primary prevention
by mutations in KCNQ1 (the remaining 10 % only for patient at very high risk for
is related to KCNE1 mutations) [24, 25] arrhythmia (see above). It could be con-
Timothy syndrome (20 cases worldwide): rare sidered in patients that experience
congenital defect (autosomal dominant) recurrent syncope while receiving beta-
caused by mutations of CACNA1C (gene blocker therapy. It is not indicated in
encoding the Cav1.2 subunit) and character- asymptomatic patients with LQTS
ized by physical malformation (syndactyly), (excluding those at very high risk) espe-
neurological and developmental defects cially if not receiving beta-blockers.
(autism), and QT prolongation with high Left cardiac sympathetic denervation
arrhythmic risk (frequent early childhood sud- (LCSD): is a surgical procedure aimed
den death) [26, 27] at the dissection of the main sympa-
Documented mutations of LQT1 affecting in thetic trunk in the upper thoracic region.
particular the cytoplasmic loops and those The procedure is done usually through a
responsible for dominant-negative ion current left supraclavicular incision and seems
effects to be effective in preventing the recur-
QTc >500 ms (in particular if affected by two rence of arrhythmic events in high-risk
unequivocally variants by genetic testing) and subjects [29]. In particular, LCSD is
very high risk if >600 ms recommended for patients refusing ICD
Syncope or cardiac arrest before age 7 and therapy or having contraindications to it
very high risk if it occurs in the first year of life and in those patients experiencing recur-
Recurrence of arrhythmic events during rence of syncope or arrhythmias during
optimal medical therapy beta-blocker therapy, when these drugs
are not tolerated, contraindicated, or
After adequate risk stratifications, the refused by the patient.
management of patients with LQTS includes:

ERRNVPHGLFRVRUJ
294 M.V. Matassini and A. Maolo

Other channel-specific therapies such as mex- frequent episodes of atrial fibrillation requiring
iletine, flecainide, and ranolazine (all sodium electrical cardioversion. Moreover, they
channel blockers) have been used in high-risk described the clinical case of a 37-year-old man
patients with recurrent events despite optimal with history of malignant ventricular arrhythmias
dosed beta-blockers, ICD, and LCSD. Their and a short QT interval on the ECG who died
effectiveness is encouraging, but they are still not suddenly at home [30].
considered as a routine therapy.

Genotypes-Phenotypes
25.3 Clinical Course
and Therapeutic The SQTS was associated with mutations of
Management some genes encoding the potassium channels
of the Clinical Case (KCNH2, KCNQ1, KCNJ2). These genes are
involved also in the pathogenesis of LQTS (in
For the specific clinical case discussed above, we particular LQT1 and LQT2), but on the contrary,
finally decided implanting one-chamber endo- in SQTS, the mutations are responsible for a gain
cavitary ICD. This choice was due to torsade de of function of the potassium channels. In few
pointes occurrence unrelated to secondary rea- patients with SQTS, mutations in the genes
sons. Furthermore, the classical ICD was pre- CACNA1C and CACNB2 (encoding the alpha-
ferred to S-ICD because we thought that the and beta-subunits of the L-type calcium channel
anti-tachycardia and anti-bradycardia pacing of the myocardium) responsible for a loss of
could have been useful in this patient. function of the channel were identified. These
Nadolol 40 mg once daily was also adminis- genes are usually involved also in the pathogen-
tered as starting dose. When employing a beta- esis of Brugada syndrome (in particular mani-
blocker, the choice is usually between two drugs: festing with type 1 ECG pattern).
nadolol and sustained-release propranolol. As The different genotypes correspond to dif-
explained above, these two drugs were demon- ferent phenotypes and different channels func-
strated to be superior to metoprolol in preventing tion disturbances (loss or gain of function), and
arrhythmic events in patients with LQTS. Nadolol they manifest with different ECG patterns
40 mg once a day is the starting dose, but the dos- [3135].
age could be increased up to 160 mg/day. The classification of the different forms of
A follow-up visit was performed 3 months short QT syndrome according to the genes muta-
later. The patients were completely asymptom- tions, the channel involved, and the ECG expres-
atic. The physical examination was normal. At sion is summarized in Table 25.3.
the ICD interrogation, any arrhythmia was
recorded.
Clinical Presentation

25.4 Short QT Syndrome The clinical onset of the SQTS is unpredictable


and extremely variable.
Short QT syndrome (SQTS) is a relatively new Relying on the data available up to now and
disease and one of the rarest channelopathies. taking into account the largest series of cases (29
The first hypothesis of a possible correlation subjects) published by Giustetto in 2006 [23],
between an idiopathic short QT interval and sud- cardiac arrest seems to be the most frequent
den cardiac death dates back to 2000, when manifestation (34 %) and is often the first clinical
Gussak, P. Brugada, and J. Brugada described the presentation (28 %). In that series, syncope was
finding of a persistent short QT interval in an present in 24 % of patients and palpitations in
entire family, including a 17-year-old girl having 31 % of patients with frequent documentation of

ERRNVPHGLFRVRUJ
25 Long QT Syndrome 295

Table 25.3 Summary table of genes and channels involved, channels function modifications and ECG patterns in
SQTS
Gene Channel Function disturbance ECG
SQTS1 KCNH2 IKr Gain

SQTS2 KCNQ1 IKs Gain

SQTS3 KCNJ2 IK1 Gain

SQTS4 CACNA1C ICaL Loss

SQTS5 CACNB2B ICaL Loss

atrial fibrillation or flutter also in young patients. Therapy


For this reason, it is important to consider SQTS
as a differential diagnosis when a lone atrial Because of the small series and few data avail-
fibrillation is found in young patients and in chil- able, there is a lack of evidence regarding the
dren. However, in that series, the median age of clinical management of patients with SQTS. The
presentation was 30 years with a wide range ICD implantation in patients who survive cardiac
between 4 months and 62 years [23]. arrest or experimented sustained ventricular
tachycardia is obvious considering the high
lethality of this syndrome, but the issue is more
Diagnosis questionable when talking about primary preven-
tion. The hardest point is doing a correct risk
Considering the rarity of the syndrome, a univo- stratification without strong risk factors con-
cal definition of the diagnostic criteria does not firmed up to now.
exist. According to the latest HRS/EHRA/HPRS Quinidine and sotalol are the two drugs used
consensus document [36], a diagnosis of SQTS up to now exploiting their ability in prolonging
can be made: QT interval. However, quinidine has proven to be
effective in prolonging QT interval only in
If a QTc interval 330 ms is present, mea- patients with SQTS, while sotalol could be effec-
sured avoiding bradycardia and tachycardia tive in the other subtypes [3739].
and using Bazetts formula For these reasons, the authors of the latest HRS/
If a QTc interval <360 ms is present, associ- EHRA/HPRS consensus document [36] conclude
ated with at least one among pathogenic muta- that the most reasonable management is:
tion, family history of SQTS, history of
familiar sudden cardiac death at age 40 years, ICD implantation (recommended, Class I)
and VT/VF in absence of structural heart in symptomatic patients with a diagnosis of
disease SQTS who survived cardiac arrest and/or

ERRNVPHGLFRVRUJ
296 M.V. Matassini and A. Maolo

have documented spontaneous sustained VT 10. Shimizu W, Noda T, Takaki H et al (2004) Diagnostic
value of epinephrine test for genotyping LQT1,
with or without syncope
LQT2, and LQT3 forms of congenital long QT syn-
ICD implantation (considered, Class IIb) in drome. Heart Rhythm 1:276283
asymptomatic patients with a diagnosis of 11. Vyas H, Hejilik J, Ackerman MJ (2006) Epinephrine
SQTS and a family history of SCD QT stress testing in the evaluation of congenital long
QT syndrome: diagnostic accuracy of the paradoxical
Quinidine (considered, Class IIb) in asymp-
QT response. Circulation 113(11):13851392
tomatic patients with a diagnosis of SQTS and 12. Zipes DP, Camm AJ, Borggrefe M et al (2006) ACC/
a family history of SCD AHA/ESC 2006 guidelines for management of
Sotalol (considered, Class IIb) in asymptom- patients with ventricular arrhythmias and the
prevention of sudden cardiac death A report of the
atic patients with a diagnosis of SQTS and a
American College of Cardiology/American Heart
family history of SCD Association Task Force and the European Society of
Cardiology Committee for Practice Guidelines (Writing
Finally, it is important to remember the pri- Committee to Develop Guidelines for Management of
Patients With Ventricular Arrhythmias and the
mary importance of an appropriate ICD program-
Prevention of Sudden Cardiac Death) Developed in col-
ming to avoid inappropriate shocks due to laboration with the European Heart Rhythm Association
oversensing of tall T waves present in the ECG and the Heart Rhythm Society. Europace 8:746837
pattern of SQTS. 13. Priori SG, Schwartz PJ, Napolitano C et al (2003)
Risk stratification in the long-QT syndrome. N Engl J
Med 348(19):18661874
14. Moss AJ, Zareba W, Benhorin J et al (1995) ECG
T-wave patterns in genetically distinct forms of the
References hereditary long QT syndrome. Circulation 92(10):
29292934
1. Goldemberg I, Moss AJ (2008) Long QT syndrome. J 15. Schwartz PJ, Malliani A (1975) Electrical alternation
Am Coll Cardiol 51:22912300 of the T-wave: clinical and experimental evidence of
2. Ackerman MJ, Priori SG, Willems S et al (2011) its relationship with the sympathetic nervous system
HRS/EHRA expert consensus statement on the state and with the long Q-T syndrome. Am Heart J
of genetic testing for the channelopathies and cardio- 89(1):4550
myopathies. Europace 13:10771109 16. Malfatto G, Beria G, Sala S et al (1994) Quantitative
3. Tester DJ, Ackerman MJ (2007) Postmortem long QT analysis of T wave abnormalities and their prognostic
syndrome genetic testing for sudden unexplained implications in the idiopathic long QT syndrome. J
death in the young. J Am Coll Cardiol 49:240246 Am Coll Cardiol 23(2):296301
4. Schwartz PJ, Priori SG, Spazzolini C et al (2001) 17. Schwartz PJ, Crotti L (2011) QTc behavior during
Genotype-phenotype correlation in the long-QT syn- exercise and genetic testing for the long-QT syn-
drome: gene-specific triggers for life-threatening drome. Circulation 124(20):21812184
arrhythmias. Circulation 103:89 18. Wong JA et al (2010) Utility of treadmill testing in
5. Moss AJ, Robinson JL, Gessman L et al (1999) identification and genotype prediction in long-QT
Comparison of clinical and genetic variables of car- syndrome. Circulation 3:120125
diac events associated with loud noise versus swim- 19. Sy RW, van der Werf C, Chattha IS et al (2011)
ming among subjects with the long QT syndrome. Am Derivation and validation of a simple exercise-based
J Cardiol 84:876 algorithm for prediction of genetic testing in relatives
6. Ali RH, Zareba W, Moss AJ et al (2000) Clinical and of LQTS probands. Circulation 124(20):21872194
genetic variables associated with acute arousal and 20. Horner JM, Horner MM, Ackerman MJ (2011) The
nonarousal-related cardiac events among subjects diagnostic utility of recovery phase QTc during tread-
with long QT syndrome. Am J Cardiol 85:457 mill exercise stress testing in the evaluation of long
7. Ackerman MJ, Tester DJ, Porter CJ (1999) Swimming, QT syndrome. Heart Rhythm 8(11):16981704
a gene-specific arrhythmogenic trigger for inherited 21. Viskin S, Postema PG, Bhuiyan ZA et al (2010) The
long QT syndrome. Mayo Clin Proc 74:1088 response of the QT interval to the brief tachycardia pro-
8. Batra AS, Silka MJ (2002) Mechanism of sudden car- voked by standing: a bedside test for diagnosing long
diac arrest while swimming in a child with the pro- QT syndrome. J Am Coll Cardiol 55(18):19551961
longed QT syndrome. J Pediatr 141:283 22. Walker BD, Krahn AD, Klein GJ, Skanes AC, Yee R,
9. Ackerman MJ, Khositseth A, Tester DJ et al (2002) Wang J, Hegele RA (2005) Effect of change in pos-
Epinephrine-induced QT interval prolongation: a ture and exercise on repolarization in patients with
gene-specific paradoxical response in congenital long long QT syndrome with HERG channel mutations.
QT syndrome. Mayo Clin Proc 77:413421 Can J Cardiol 21:3338

ERRNVPHGLFRVRUJ
25 Long QT Syndrome 297

23. Priori SG, Wilde AA, Horie M, Cho Y et al (2013) 31. Gussak I, Brugada P, Brugada J et al (2000) Idiopathic
HRS/EHRA/HPRS expert consensus statement on the short QT interval: a new clinical syndrome?
diagnosis and management of patients with inherited Cardiology 94:99102
primary arrhythmia syndromes. Heart Rhythm 32. Merino JL, Reviriego SM (2010) Short QT syndrome.
10(12):e85e108, EP Europace and Journal of E-Journal of the ESC Council for Cardiology Practice.
Arrhythmias http://www.escardio.org/Guidelines-&-Education/
24. Schwartz PJ, Moss AJ, Vincent GM, Crampton RS Journals-and-publications/ESC-journals-family/
(1993) Diagnostic criteria for the long-QT syndrome. E-journal-of-Cardiology-Practice/Volume-9/Short-
An Update. Circulation 88:782784 QT-Syndrome
25. Jervell F (1957) Lange-Nielsen. Congenital deaf- 33. Brugada R, Hong K, Dumaine R et al (2004) Sudden
mutism, functional heart disease with prolongation of death associated with short QT syndrome linked to
the QT interval and sudden death. Am Heart J mutations in HERG. Circulation 109:3035
54(1):5968 34. Bellocq C, van Ginneken AC, Bezzina CR et al (2004)
26. Tranebjaerg L, Samson RA, Green GE (2002) Jervell Mutation in the KCNQ1 gene leading to the short
and Lange-Nielsen syndrome. Gene Rev http://www. QT-interval syndrome. Circulation 109:23942397
ncbi.nlm.nih.gov/books/NBK1405/ 35. Priori SG, Pandit SV, Rivolta I et al (2005) A novel
27. Marks M, Whisler S, Clericuzio C, Keating M (1995) form of short QT syndrome (SQT3) is caused by a
A new form of long QT syndrome associated with mutation in the KCNJ2 gene. Circ Res 96:800807
syndactyly. J Am Coll Cardiol 25(1):5964 36. Antzelevitch C, Pollevick GD, Cordeiro JM et al
28. Splawski K, Timothy N, Decher P, Kumar F, Sachse (2007) Loss-of-function mutations in the cardiac cal-
A, Beggs M, Sanguinetti M, Keating M (2005) Severe cium channel underlie a new clinical entity character-
arrhythmia disorder caused by cardiac L-type calcium ized by ST- segment elevation, short QT intervals, and
channel mutations. Proc Natl Acad Sci USA sudden cardiac death. Circulation 115:442449
102(23):80898096 37. Giustetto C, Di Monte F, Wolpert C et al (2006) Short
29. Chockalingam P, Crotti L, Girardengo G et al (2012) QT syndrome: clinical findings and diagnosticthera-
Not all beta-blockers are equal in the management of peutic implications. Eur Heart J 27:24402447
long QT syndrome types 1 and 2: higher recurrence of 38. Giustetto C, Schimpf A (2011) Lon term follow-up of
events under metoprolol. J Am Coll Cardiol 60(20): patients with short QT syndrome. J Am Coll Cardiol
20922099 58(6):587595
30. Schwartz PJ, Priori SG, Cerrone M et al (2004) Left 39. Gaita F, Giustetto C, Bianchi F et al (2004) Short QT
cardiac sympathetic denervation in the management syndrome: pharmacological treatment. J Am Coll
of high-risk patients affected by the long-QT syn- Cardiol 43(8):14941499
drome. Circulation 109(15):18261833

ERRNVPHGLFRVRUJ
Catecholaminergic Polymorphic
Ventricular Tachycardia: 26
A Challenging Case of Epilepsy

Laura Cipolletta

26.1 Introduction
had a period of incoherent speech, followed
Cardiac channelopathies are potentially malig- by sleepiness lasting for several hours. He
nant conditions caused by mutations in the chan- suffered from urinary incontinence during
nels that alter ion transit across the cardiac episodes. These events occurred approxi-
myocyte cell membrane. The clinical presenta- mately every month, although they had
tion of these mutations is variable, ranging from increased in frequency over the past
isolated abnormalities on electrocardiogram 2 months.
(ECG) to sudden cardiac death. The episodes were triggered by anxiety
and never occurred during physical efforts,
although he had no active lifestyle.
26.2 Case Report Several anticonvulsant medications
were not effective, so inpatient EEG moni-
toring was performed. EEG demonstrated
frequent, independent, temporal intermit-
A 17-year-old boy was referred to the car- tent rhythmic delta activity, indicating an
diology clinic by a neurologist for evalua- increased risk for focal seizures; however,
tion of potential arrhythmia. He was treated the typical episode was not recorded during
for resistant focal epilepsy for 4 years. admission. A second EEG recorder cap-
During a typical event, he fell off, with tured a typical event. It showed a high-
generalized shaking, lasting several min- amplitude diffuse slowing waves
utes. Then he gasped and in few seconds (suggestive of extracerebral artifact), fol-
later he appeared to stop breathing. He also lowed by an abrupt, and diffuse, attenua-
tion lasting 60 s, and finally a slow recovery
to diffuse polymorphic delta frequencies.
There was no ECG recording during the
episode. The EEG pattern suggested cere-
bral hypoperfusion, so the patient was
L. Cipolletta referred to a cardiologist. At admission to
Clinica di Cardiologia e Aritmologia, Universit
Politecnica delle Marche, the cardiology department, he became anx-
Via Conca, 71, Ancona 60126, Italy ious and lost consciousness.
e-mail: lauracipo2006@libero.it

Springer International Publishing Switzerland 2015 299


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_26

ERRNVPHGLFRVRUJ
300 L. Cipolletta

Medical History and Cardiovascular potassium 3.9 mEq/l, sodium 140 mEq/l,
Risk Factors magnesium 1.8 mg/dl).

No cardiovascular risk factor; his family


history of structural heart disease, syncope, or Instrumental Examination
sudden cardiac death was unremarkable.
2010: diagnosis of focal epilepsy unrespon- Basal ECG was normal with several PVC.
sive to antiepileptic agents. The loss of consciousness corresponds to the
onset of polymorphic VT on telemetry ECG
monitoring (Figs. 26.1 and 26.2) and spontane-
Allergies ously the VT converted to sinus rhythm with
frequent premature ventricular complexes
None (PVCs) after a brief period of cardiopulmonary
resuscitation.

Medications
Clinical Course and Therapeutic
Valproic acid Management

This event was similar to his typical episodes.


Vital Signs During his hospital stay, he was treated with a
beta-blocker (propranolol 3 mg/kg/die) with no
Temperature: 36 C further episodes.
Heart rate: 90 bpm Genetic testing revealed a mutation in the
Arterial blood pressure: 100/60 mmHg cardiac ryanodine receptor gene (RyR2), consis-
Respiratory rate: 16 breaths/min tent with CPVT.
Oxygen saturation: 100 %

26.3 Catecholaminergic
Physical Examination Polymorphic Ventricular
Tachycardia (CPVT)
General appearance: Well developed, well
nourished, alert, and cooperative Epidemiology
Lungs: Clear to auscultation and percussion
without rales, rhonchi, wheezing, or dimin- The prevalence of CPVT is about 1:10,000. The
ished breath sounds mean age of onset is between 7 and 9 years,
Cardiovascular: Normal S1 and S2. No S3, although later onset has been reported.
S4, or murmurs. Regular rhythm. No periph- Approximately 30 % of patients have a family
eral edema, cyanosis, or pallor. Warm and history of stress-related syncope, seizure, or SCD
well-perfused extremities before age 40 years. There is a high level of pen-
Abdomen: Positive bowel sounds. Soft and non- etrance of the disease (7580 %) [1].
distended. No guarding or rebound. No masses

Clinical Presentation
Routine Laboratory Test
Syncope triggered by exercise or emotion is the
Tests were normal (hemoglobin 13.4 g/dl, white typical clinical presentation of CPVT patients
blood cells 6,290/mmc, creatinine 0.7 mg/dl, and sometimes a reproducible, stress-related,

ERRNVPHGLFRVRUJ
26 Catecholaminergic Polymorphic Ventricular Tachycardia: A Challenging Case of Epilepsy 301

I
II
III
aVR
aVL
aVF
V1
V2
V3
V4
V5
V6
1000ms

Fig. 26.1 Onset of polymorphic VT symptomatic for syncope, converted to sinus rhythm after cardiopulmonary
resuscitation

Fig. 26.2 Restoration of sinus rhythm with frequent monomorphic PVCs

bidirectional VT in the absence of a structural patients with CPVT. Ventricular arrhythmia


heart disease or QT interval prolongation could distinguishing CPVT is characterized by alternat-
be documented. CPVT appears as one of the ing QRS axis with 180 rotation on a beat-to-beat
most malignant forms of ventricular arrhythmia. basis (bidirectional VT). However, this typical
By age 40, almost 80 % of untreated CPVT arrhythmia is not observed in all patients. With
patients develop symptoms (syncope, VT, or increasing exercise workload, a progressive
VF), and overall mortality is 3050 % [2]. The worsening of arrhythmias is a characteristic fea-
first manifestation of the disease can be SCD in a ture of CPVT. A heart rate of 110130 beats/min
significant number of cases [3]. is the cutoff to the appearance of ventricular
arrhythmias during exercise stress testing, ini-
tially as polymorphic PVCs. Then, the complex-
Electrocardiographic Features ity and frequency of arrhythmias progressively
worsen as workload increases: ventricular bigem-
Brugada-like ST elevation, QT interval abnor- iny, run of polymorphic PVCs, and polymorphic
malities, and atrioventricular and intraventricular or bidirectional VT. Bidirectional VT may degen-
conduction defects are absent in resting ECG of erate into polymorphic VT and VF, if exercise is

ERRNVPHGLFRVRUJ
302 L. Cipolletta

not quickly stopped. On the recovery phase, VT reasonable [1, 4]. Invasive EP testing is of no
rate and VT duration progressively diminish until value in the diagnosis or risk stratification in
arrhythmias disappear. During exercise, at a patients with CPVT. Programmed electrical
range of heart rates similar to or slower than that stimulation is not useful to arrhythmia induction
of ventricular arrhythmias, isolated premature [1, 3]. Genetic testing should be performed in all
atrial complexes, nonsustained supraventricular definitive CPVT probands (see Fig. 26.3).
tachycardia, and short runs of AF usually are
observed [3, 4].
Genetics of CPVT

Diagnosis of CPVT Mutations in genes that encode for Ca2+ regula-


tory proteins are the principal cause of CPVT. The
Resting ECG, echocardiography, and EP testing genetic variants of CPVT that have been
frequently are completely normal. CPVT diagno- described are the following: an autosomal domi-
sis is clinically based on symptoms (syncope or nant trait (CPVT1, most common) caused by
aborted SCD), family history, and response to mutations in the RyR2 gene and a recessive form
exercise or isoproterenol infusion. Holter moni- (CPVT2, rare) associated with homozygous
toring, exercise, and drug provocation demon- mutations in the CASQ2 gene [1].
strated ventricular arrhythmias in more than 80 % RyR2 is the major calcium release channel of
of patients. Not infrequently, syncopal episodes the sarcoplasmic reticulum, mediating excita-
are considered as vasovagal in origin, and no fur- tioncontraction coupling. RyR2 mutations are
ther workup is performed. If the loss of con- present in approximately 5070 % of patients
sciousness is associated with convulsions, as it with CPVT. Typical CPVT mutation of RyR2s
happened in our case, it may be misdiagnosed as shows gain-of-function defects following chan-
epileptic seizures if a prolonged circulatory arrest nel activation by PKA phosphorylation (in
resulted in brain ischemia [1]. response to beta-adrenergic stimulation), leading
When a syncopal episode induced by exercise to an uncontrolled Ca2+ release from the sarco-
or emotion occurs in a child or in a young patient plasmic reticulum during electrical diastole that
with a normal resting ECG and no structural heart increases the diastolic membrane depolarizations
disease, CPVT should be hypothesized [1, 3]. (DADs) and triggers arrhythmias [2].
The most important test for diagnosis of Another sarcoplasmic reticulum Ca2+ buffer-
CPVT is the exercise stress test that induces ven- ing protein associated with RyR2 is CASQ2 that
tricular arrhythmias in at least 80 % of CPVT plays an important role in the control of Ca2+
patients. When the sinus rate exceeds an individ- release from the sarcoplasmic reticulum to the
ual threshold rate (usually at 110130 beats/min), cytosol. The hypothesis is that some of these
VT typically appears. A diagnostic and highly mutations compromise CASQ2 synthesis and
reproducible marker of CPVT is the progressive reduce expression or provoke complete absence
worsening of arrhythmias during exercise. of CASQ2 in the heart and others cause expres-
Intravenous infusion of catecholamines (e.g., iso- sion of defective CASQ2 proteins with abnormal
proterenol or epinephrine) can also provoke pro- regulation of cellular Ca2+ homeostasis [2, 3].
gressive ventricular arrhythmias [2, 4]. After a
first syncopal episode, if a high suspicion of
CPVT exists, repeated exercise stress test is fun- Mechanism of Ventricular
damental to avoid delay in the diagnosis [4]. Arrhythmias in CPVT
Also, when a maximal exercise stress test is dif-
ficult, continuous ambulatory monitoring may The central pathogenic abnormality in CPVT
reveal arrhythmias typical for CPVT. In some results from abnormalities in the control of sarco-
cases, an implantation of a loop recorder could be plasmic reticulum Ca2+ release. The L-type Ca2+

ERRNVPHGLFRVRUJ
26 Catecholaminergic Polymorphic Ventricular Tachycardia: A Challenging Case of Epilepsy 303

Fig. 26.3 Diagnostic


protocol of catecholaminergic Diagnostic protocol
polymorphic ventricular Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT)
tachycardia

Syncope without prodromes


- During exercise or emotions
- Starting from infancy
Resuscitated sudden cardiac death
Family history of sudden cardiac death
Normal EKG

Suspect of catecholaminergic ventricular


tachycardia

Clinical history, physical examination


Genealogical tree
Treadmill test
Epinephrine test
24-h Holter ECG
Echocardiogram

Echocardiogram:
Exclusion of structural heart disease
Treadmill test/ epinephrine test:
Ventricular arrhythmia that increases in frequency and
complexity while increases the exercise threshold/ epinephrine
infusion and decreases until it disappears during recopevy
With/without:
Holter:
Ventricular arrhythmia related to exercise/emotion

CPVT diagnosis

channels in the cell membrane permit Ca2+ influx calcium adenosine triphosphatase (SERCA)
during the action potential plateau that triggers resequesters most of the surplus Ca2+ in the cytosol
more Ca2+ release (Ca2+ transients) from the sarco- into the sarcoplasmic reticulum, controlled by the
plasmic reticulum into the cytosol via activation of phosphoprotein phospholamban. Additionally, to
Ca2+ release channels (RyR2). This amplifying balance the Ca2+ that enters with the Ca2+ current,
process, known as Ca2+-induced Ca2+ release, Na+Ca2+ exchanger extrudes from the cell some
causes a quick raise in cytosolic Ca2+ concentration of the Ca2+.
to an optimal level required for binding of Ca2+ to RyR2 mutations alter the physiological prop-
troponin C and induction of contraction [5]. During erties and function of RyR2 using some molecular
diastole, the sarcoplasmic/endoplasmic reticulum mechanisms not fully understood. It has been

ERRNVPHGLFRVRUJ
304 L. Cipolletta

suggested that the binding affinity of RyR2 for the arrhythmias related with intracellular Ca2+
regulatory protein calstabin-2 is reduced by overload and the DADs documented during digi-
CPVT mutations in RyR2. The closed conforma- talis toxicity. Mutations in RyR2 or CASQ2 lead
tional state of the RyR2 channel is stabilized by to the activation of the Na+Ca2+ exchanger
calstabin-2, which enables the channel to close enhanced by cytosolic Ca2+ overload. This mech-
completely during diastole (at low intracellular anism generates a net inward current (the so-
Ca2+ concentrations) and prevents aberrant Ca2+ called transient inward current) that provokes
leakage from the sarcoplasmic reticulum, ensur- DADs and if they reach the threshold for Na+
ing muscle relaxation. The binding affinity of cal- channel activation can trigger abnormal beats [1,
stabin-2 is worsened by PKA phosphorylation 3]. Low-amplitude DADs usually are not clini-
(induced by beta-adrenergic stimulation) of the cally significant. A decrease in the initiating
mutant RyR2 channels, increasing the probability cycle length probably is the most important fac-
of an open state at diastolic Ca2+ concentrations. tor that leads subthreshold DADs to reach thresh-
So, diastolic Ca2+ leak from the sarcoplasmic old; indeed fast heart rates increase both the
reticulum during stress or exercise is the result of amplitude and rate of the DADs. Moreover, cat-
the incomplete closure of the mutant RyR2 chan- echolamines can facilitate the development of
nel during diastole [1, 6]. Other groups have sug- DADs because the stimulation of beta-adrenergic
gested that in CPVT the mutated RyR2 channel is receptors and subsequently cAMP increases the
more sensitive to luminal (sarcoplasmic reticu- L-type Ca2+ current and leads to an increase in
lum) Ca2+. So under baseline conditions, where transsarcolemmal Ca2+ influx and intracellular
sarcoplasmic reticulum load is normal, there is no Ca2+ overload; also the enhancement of the activ-
Ca2+ leak, but under beta-adrenergic (sympa- ity of the Na+Ca2+ exchanger increases the like-
thetic) stimulation, sarcoplasmic reticulum Ca2+ lihood of DAD-mediated triggered activity;
concentration raises above the reduced threshold, finally, the enhancement of the uptake of Ca2+ by
causing Ca2+ to leak out of the sarcoplasmic retic- the sarcoplasmic reticulum leads to a greater
ulum. A third hypothesis for RyR2-related CPVT amount of Ca2+ stored in the sarcoplasmic reticu-
is that the intermolecular interactions between lum and the subsequent major release of Ca2+
discrete RyR2 domains necessary for proper fold- from the sarcoplasmic reticulum during contrac-
ing of the channel and self-regulation of channel tion. The increased susceptibility to ventricular
gating are impaired by the mutations in RyR2 [6]. arrhythmias in CPVT patients during exercise
The most important Ca2+ storage protein in the and emotional stress is associated with increased
sarcoplasmic reticulum is calsequestrin (CASQ2) sympathetic stimulation and higher heart rates.
and forms a part of a quaternary complex with Sometimes in CPVT, spontaneous Ca2+ release
RyR2, triadin, and junction, which play a major and DADs can happen without Ca2+ overload. For
role in regulating intracellular Ca2+. Calsequestrin, example, mutations in RyR2 or CASQ2 provoke
as a high-capacity, low-affinity Ca2+-binding pro- defective Ca2+ signaling that lowers the sarcoplas-
tein, is able to bind luminal Ca2+ during diastole, mic reticulum Ca2+ threshold for spontaneous
buffering Ca2+ within the sarcoplasmic reticulum Ca2+ release below the normal baseline level caus-
and preventing diastolic Ca2+ release via RyR2 to ing the so-named perceived Ca2+ overload [6].
the cytosol. For effective termination of sarco-
plasmic reticulum Ca2+ release and prevention of
spontaneous Ca2+ release during diastole, the Mechanism of the Bidirectional
control of RyR2s by luminal Ca2+ is required. Morphology of Ventricular
CASQ2 mutations lead to diminished Ca2+ sig- Tachycardia
naling refractoriness and generation of arrhyth-
mogenic spontaneous Ca2+ releases [6]. The main The EP mechanisms leading to the characteristic
arrhythmogenic mechanism in CPVT is DADs bidirectional morphology of the VT are not
and triggered activity because the bidirectional fully understood. Some of the proposed mecha-
ECG pattern of this VT corresponds to the nisms are the following: changes in conduction

ERRNVPHGLFRVRUJ
26 Catecholaminergic Polymorphic Ventricular Tachycardia: A Challenging Case of Epilepsy 305

direction from a single ventricular focus, one VT twisting of the points of the QRS complexes),
focus that triggers another focus, and firing from frequent in LQTS patients [13].
double ventricular foci (i.e., the right and left api- Bidirectional VT can also occur in patients
cal portions of the heart) [2, 6]. with type 7 LQTS (LQT7, AndersenTawil syn-
Others showed that left posterior inferior drome) linked to mutations in the KCNJ2 gene,
origin accounts for the majority of cases. which may be considered a CPVT phenocopy,
Additionally, the site of origin of bidirectional particularly in patients with AndersenTawil
VT has been recognized in the Purkinje network, syndrome having borderline QT interval prolon-
with alternating firing from the right and left gation. SCD is rare among AndersenTawil syn-
branches of the Purkinje fibers [3]. drome and KCNJ2 mutation carriers [8].
A recent experimental model identified a Furthermore, typical of AndersenTawil syn-
ping-pong mechanism as etiology of ventricu- drome are the extracardiac features such as peri-
lar arrhythmias in CPVT, so in different regions odic paralysis and facial dysmorphism [13].
of the HisPurkinje system (HPS) or ventricles, Ankyrin-B syndrome can also manifest with
DAD-induced triggered activity develops at dif- catecholamine-mediated ventricular arrhythmias.
ferent heart rate thresholds. First, if the heart rate Cardiac ankyrin-B is a structural membrane
rises above a certain threshold, ventricular adapter protein encoded by ANK2 gene. Loss-of-
bigeminy appears from a single site in the HPS or function mutations result in increased intracellular
ventricular myocardium. Ventricular bigeminy concentration of Ca2+ and a greater risk of fatal
shortens the RR cycle length that induces DAD- arrhythmia. This syndrome has been categorized
triggered beats from a second focus within the under LQTS (LQT4), but the QT interval prolon-
HPS, with the latter reciprocally activating PVCs gation is inconsistent and the degrees of cardiac
from the first focus; this process is repeated back dysfunction and arrhythmias observed (including
and forth, in a ping-pong pattern. The bidirec- bradycardia, sinus arrhythmia, idiopathic VF,
tional VT characteristic of CPVT is produced by adrenergically mediated VT, and SCD) are vari-
the reciprocating bigeminy from the two sites. able, so ankyrin-B syndrome is considered as a
When three or more sites concurrently develop different clinical entity compared to classic LQTS.
bigeminy, the result is the development of poly- The typical ECG pattern and the structural
morphic VT, whereas when repetitive DADs gen- abnormalities of the RV distinguish ARVD from
erate a run of triggered activity from a single site, CPVT despite the development of exercise-
the result is the development of monomorphic provoked arrhythmias. The typical arrhythmia in
VT [7]. ARVD is monomorphic VT with a left bundle
branch block (LBBB) pattern that is totally different
from the polymorphic PVCs or VT in CPVT [4].
Differential Diagnosis In contrast to CPVT, arrhythmias in Brugada
syndrome appear usually at rest or during sleep so
The first step is the exclusion of other inherited patients with Brugada syndrome do not manifest
arrhythmogenic cardiac disorders that can cause polymorphic PVCs on physical effort. Moreover,
malignant ventricular tachyarrhythmias. Measure Brugada syndrome shows the characteristic ST
of QT interval excludes the diagnosis of SQTS (if segment elevation in the precordial ECG leads at
QTc interval less than 320 ms) or LQTS (if QTc baseline and after provocation testing with Na+
interval more than 450 ms). The first syncope in channel blockers, absent in CPVT [4].
CPVT patients more often occurs during child-
hood, whereas clinical symptoms of LQTS typi-
cally start around puberty. Risk Stratication
Furthermore, the typical arrhythmia in CPVT
patients is bidirectional VT with a beat-to-beat Because of the relatively small number of
180 rotation of the QRS complex, in contrast to patients, reported risk stratification for SCD in
the torsades de pointes (characterized by the CPVT is difficult. CPVT patients with prior

ERRNVPHGLFRVRUJ
306 L. Cipolletta

cardiac arrest and those in whom pharmacologi- Table 26.1 Treatment of catecholaminergic polymor-
phic ventricular tachycardia (CPVT)
cal therapy does not suppress are considered at
high risk for SCD. Programmed electrical stimu- Catecholaminergic polymorphic ventricular tachycardia
lation typically fails in inducing ventricular (CPVT) treatment
arrhythmias and is of no value for risk stratifica- Genotype, if available
Beta-blockers (BB) at maximum tolerated doses:
tion. Furthermore, the predictive value of
In patients with CPVT clinical diagnosis (Class I
inducibility of ventricular arrhythmias by cate-
indication, level of evidence: C) [11]
cholamine infusion or exercise for risk stratifica- In patients without clinical symptoms of CPVT but
tion has not been confirmed. with genetic diagnosis of CPVT during childhood
(Class IIa indication, level of evidence: C) [11]
In patients with CPVT and genetic diagnosis in adult
Pharmacological Therapy age without any symptoms of tachycardia (Class IIb
indication, level of evidence: C) [11]
Nadolol 40 and 80 mg:
The first line of treatment for CPVT is beta-
Kids: 12.5 mg/kg/die, q.d.
blockers and should be promptly initiated to pre- Adults: maximal doses 160 mg/die, q.d.
vent the occurrence of ventricular Propranolol:
tachyarrhythmias. Untreated CPVT has a poor Kids: 2.53.5 mg/kg/die, t.i.d. (every 8 h)
prognosis, so drug therapy is indicated for all Adults: 160 mg/die (q.d.)
clinically diagnosed patients and also for all Treadmill test could help titrate beta-blocker doses
silent carriers of a CPVT mutation. The most (target heart rate of <85 % of maximum heart rate,
significant reduction of ventricular arrhythmia burden
widely used beta-blockers are nadolol (12.5 mg/
during exercise test)
kg/day) and propranolol (2.53.5 mg/kg/day).
Left thoracic sympathectomy [24]
Intravenous propranolol is the treatment of choice Kids with syncope during beta-blocker therapy,
for acute management of CPVT (Table 26.1). to delay/avoid ICD implantation
Exercise stress testing and Holter monitoring ICD patients with frequent ICD therapy (ATP/
can help determine the adequate beta-blocker shock/electrical storm)
dosage for arrhythmia control, but the absence of Flecainide [9]:
exercise-provoked arrhythmias does not com- On top of maximum tolerated doses of beta-
blockers and ICD [patients with high burden of
pletely exclude the risk of arrhythmia recurrence, complex ventricular arrhythmias in the exercise test
and the maximum tolerated dose of beta-blockers during follow-up or frequent ICD therapy (ATP/
should be prescribed to optimize control of shock/electrical storm)]
arrhythmias with the aim of avoiding the achieve- Doses: 150200 mg/die; range: 100300 mg/die
ment of the threshold rate for CPVT [3, 4]. ICD: CPVT + resuscitated sudden cardiac death (Class
I indication, level of evidence: C) [11]
Recent studies demonstrate that the addition
ICD + BB in CPVT patients with syncope and/or
of flecainide to beta-blocker therapy can effec- sustained VT during treatment with BB (Class IIa
tively reduce exercise-induced ventricular indication, level of evidence: C) [11]
arrhythmias in CPVT patients not controlled by Absolute contraindication to competitive sports [13]
beta-blocker therapy alone [9]. Direct blockade Relative contraindication to recreational sports [13]
of RyR2 channels and reduction of Ca2+ spark Avoid other arrhythmia triggers: loud noises,
amplitude rather than Na+ channel blockade emotional stress
mediate the flecainide effects [10]. If the patient is implanted with ICD: periodic
follow-up to monitor recorded arrhythmias or ICD
Limited data suggest that verapamil (an inhibi- therapy
tor of RyR2) can be an alternative option for the Urgent medical consultation in case of syncope or
treatment of CPVT. Also, verapamil used in com- nocturnal agonal respiration
bination with beta-blockers can provide additional Evaluation recommended to first-degree family
protection. However, there is no conclusive members

ERRNVPHGLFRVRUJ
26 Catecholaminergic Polymorphic Ventricular Tachycardia: A Challenging Case of Epilepsy 307

evidence for recommending verapamil alone or in high mortality of CPVT, it is important to study
combination with beta-blockers because of the both first- and second-degree relatives to find
small number of patients and the limited follow-up potential CPVT patients [14]. Exercise testing
and also its impact on prognosis is not known [4]. and Holter monitoring are used for screening
family members. Moreover, some CPVT patients
Implantable Cardioverter Debrillator may not have arrhythmias in the exercise stress
ICD therapy is recommended for CPVT patients test during early childhood, but later in life, a
who have survived a cardiac arrest or those who change in the phenotype may occur. Therefore,
continue to have symptoms (syncope or sustained follow-up with repeated exercise stress tests is
or hemodynamically unstable VT) despite ade- indicated. When a gene mutation has been identi-
quate beta-blocker therapy [11]. fied in the proband, genetic testing is recom-
Approximately half of ICD recipients mended to screen family members [3].
experience an appropriate shock to terminate VT
during 2 years of follow-up. It is important to
maintain the maximum tolerated dose of
References
beta-blockers in ICD patients to help reduce the
risk of arrhythmic storms and ICD shocks [1, 2]. 1. Liu N, Ruan Y, Priori SG (2008) Catecholaminergic
polymorphic ventricular tachycardia. Prog Cardiovasc
Catheter Ablation Dis 51:2330
When ventricular arrhythmias are triggered by 2. Mohamed U, Napolitano C, Priori SG (2007)
Molecular and electrophysiological bases of catechol-
monomorphic PVCs, catheter ablation of the aminergic polymorphic ventricular tachycardia. J
focus of PVCs may be attempted to reduce Cardiovasc Electrophysiol 18:791797
the burden of arrhythmias and ICD shocks. The 3. Napolitano C, Priori SG (2007) Diagnosis and treat-
initiating beat of VT usually shows an LBBB ment of catecholaminergic polymorphic ventricular
tachycardia. Heart Rhythm 4:675678
inferior axis pattern, suggestive of a ventricular 4. Ylanen K, Poutanen T, Hiippala A et al (2010)
outflow tract origin [12]. Catecholaminergic polymorphic ventricular tachycar-
dia. Eur J Pediatr 169:535542
Sympathetic Denervation 5. Wehrens XH (2007) The molecular basis of catechol-
aminergic polymorphic ventricular tachycardia: what
Small case series suggest the effectiveness of left are the different hypotheses regarding mechanisms?
cardiac sympathetic denervation in patients with Heart Rhythm 4:794797
recurrent symptoms despite beta-blocker therapy 6. Gyorke S (2009) Molecular basis of catecholaminer-
or those experiencing intractable arrhythmic gic polymorphic ventricular tachycardia. Heart
Rhythm 6:123129
storms or frequent ICD shocks [2]. 7. Baher AA, Uy M, Xie F et al (2011) Bidirectional
ventricular tachycardia: ping pong in the His-Purkinje
Participation in Sports system. Heart Rhythm 8:599605
All CPVT patients with documented exercise- or 8. Tsuboi M, Antzelevitch C (2006) Cellular basis for
electrocardiographic and arrhythmic manifestations
isoproterenol-induced VT should avoid all competi- of Andersen-Tawil syndrome (LQT7). Heart Rhythm
tive sports. A less restrictive approach may be pos- 3:328335
sible for the genotype-positive/phenotype-negative 9. Van der Werf C, Kannankeril PJ, Sacher F et al (2011)
(asymptomatic, no inducible VT) athlete [13]. Flecainide therapy reduces exercise-induced ventricu-
lar arrhythmias in patients with catecholaminergic
polymorphic ventricular tachycardia. J Am Coll
Cardiol 57(22):22442254
Family Screening 10. Hwang HS, Hasdemir C, Laver D et al (2011)
Inhibition of cardiac Ca2+ release channels (RyR2)
determines efficacy of class I antiarrhythmic drugs in
After a diagnosis of CPVT in a family member catecholaminergic polymorphic ventricular tachycar-
because of the severe clinical manifestations and dia. Circ Arrhythm Electrophysiol 4:128135

ERRNVPHGLFRVRUJ
308 L. Cipolletta

11. Zipes DP, Camm AJ, Borggrefe M et al (2006) ACC/ 12. Sy RW, Gollob MH, Klein GJ et al (2011) Arrhythmia
AHA/ESC 2006 guidelines for management of patients characterization and long-term outcomes in catechol-
with ventricular arrhythmias and the prevention of sud- aminergic polymorphic ventricular tachycardia. Heart
den cardiac death: a report of the American College of Rhythm 8:864871
Cardiology/American Heart Association Task Force and 13. Zipes DP, Ackerman MJ, Estes NA III et al (2005)
the European Society of Cardiology Committee for Task Force 7: arrhythmias. J Am Coll Cardiol
Practice Guidelines (writing committee to develop 45:13541363
Guidelines for Management of Patients With Ventricular 14. Ackerman MJ, Priori SG, Willems S et al (2011)
Arrhythmias and the Prevention of Sudden Cardiac HRS/EHRA expert consensus statement on the State
Death): developed in collaboration with the European of Genetic Testing for the channelopathies and cardio-
Heart Rhythm Association and the Heart Rhythm myopathies. Heart Rhythm 8:13081339
Society. Circulation 114(10):e385e484

ERRNVPHGLFRVRUJ
Part IX
Miscellanea

ERRNVPHGLFRVRUJ
Sudden Cardiac Death
27
Laura Cipolletta

27.1 Introduction usually no premonitory symptoms. If symptoms


are present, they are nonspecific such as weak-
Sudden cardiac arrest (SCA) and sudden cardiac ness, chest discomfort, shortness of breath, and
death (SCD) are defined as the sudden cessation palpitations [2].
of cardiac activity with hemodynamic collapse,
usually due to sustained ventricular tachycardia/
ventricular fibrillation (VT/VF). Patients with 27.2 Case Report
structural heart disease especially coronary heart
disease are defined at greater risk.
An intervention (e.g., defibrillation) or a sponta- A 17-year-old female patient was admitted
neous reversion to sinus rhythm is defined as SCA to hospital after an episode of SCA that
(or aborted SCD), and if the patient dies, the event ensued during physical activity. The elec-
is called SCD [1]. However, typically the term trocardiogram (ECG) recorded during the
SCD is used to describe both fatal and nonfatal car- episode showed a pulseless VT (Fig. 27.1).
diac arrest occurring 1 h from symptom onset.
The specific causes of SCA vary with patient
age and different kinds of population studied. Most
commonly ventricular fibrillation (VF) in the set- Medical History and Cardiovascular
ting of structural heart disease causes hemody- Risk Factors
namic collapse and subsequently SCA.
The underlying cardiac disease and the rapid- Ebstein anomaly and severe pulmonary steno-
ity of resuscitation maneuvers (defibrillation) are sis; her family history of structural heart
the determinant factors to establish the outcome disease, syncope, or sudden cardiac death was
following SCA. The insufficient cerebral blood unremarkable.
flow due to SCA mostly results in loss of con- 2014: She had undergone surgical tricuspid
sciousness within seconds to minutes. There are valve replacement with a biological prosthetic
valve and cavopulmonary anastomosis.

L. Cipolletta
Clinica di Cardiologia e Aritmologia, Universit Allergies
Politecnica delle Marche,
Via Conca, 71, Ancona 60126, Italy None
e-mail: lauracipo2006@libero.it

Springer International Publishing Switzerland 2015 311


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_27

ERRNVPHGLFRVRUJ
312 L. Cipolletta

Fig. 27.1 Onset of polymorphic VT symptomatic for syncope, converted to sinus rhythm after cardiopulmonary
resuscitation

Medications Lungs: Clear to auscultation and percussion


without rales, rhonchi, wheezing, or
None diminished breath sounds
Cardiovascular: Normal S1 and S2. No S3 and
S4. Systolic murmur 2/6 at Erbs point.
Vital Signs Regular rhythm. No peripheral edema, cyano-
sis, or pallor. Warm and well-perfused
Temperature: 36.8 C extremities
Heart rate: 80 bpm Abdomen: Positive bowel sounds. Soft, non-
Arterial blood pressure: 110/70 mmHg distended. No guarding or rebound. No masses
Respiratory rate: 18 breaths/min
Oxygen saturation: 92 %
Routine Laboratory Test

Physical Examination Tests were normal (hemoglobin 12.8 g/dl; white


blood cells 9680/mmc; creatinine 0.82 mg/dl;
General appearance: Well developed, well potassium 3.7 mEq/l; sodium 138 mEq/l; magne-
nourished, alert, and cooperative sium 1.7 mg/dl).

ERRNVPHGLFRVRUJ
27 Sudden Cardiac Death 313

Instrumental Examination Clinical Course and Therapeutic


Management
Basal ECG after DC shock was normal with
incomplete right bundle branch block and spe- During hospital stay, she was treated with a beta-
cific abnormalities of repolarization. blocker (propranolol 3 mg/kg/day) with no fur-
The loss of consciousness was caused by a ther episodes. Genetical analysis and exercise
monomorphic pulseless VT (Fig. 27.1), con- test ruled out polymorphic ventricular tachycar-
verted to sinus rhythm by external DC shock dia as the etiology of the VT.
after a brief period of cardiopulmonary The VT episode was treated with an external
resuscitation. direct current cardioversion, and the patient was
Figure 27.2 shows the magnetic resonance of then referred to our center to attempt a VT abla-
the patient. The right ventricle was severely tion. Unfortunately, the fact that the right ventri-
enlarged, with a papyraceous right ventricular cle (RV) was papyraceous and the free ventricular
wall. wall was thin would have exposed the patient to a
high risk of catheter perforation.
Because transvenous implantable cardio-
verter defibrillator (ICD) implantation had the
same risk of cardiac perforation and access to
the right cardiac chambers had been made
unfeasible by cavopulmonary anastomosis,
subcutaneous (S)-ICD implantation was con-
sidered. In assessing the patients adequacy for
S-ICD implantation, the morphology of the
QRS was screened to avoid T-wave oversens-
ing. Because of the bizarre morphology of the
QRS attributable to severe dilatation of the RV,
QRS screening was performed with the left
arm electrode placed to the right side of the
xiphoid process. Under local anesthesia and
Fig. 27.2 Cardiac magnetic resonance conscious sedation, the catheter was inserted

Fig. 27.3 Chest X-ray of S-ICD implant

ERRNVPHGLFRVRUJ
314 L. Cipolletta

subcutaneously from the pocket in the left pacing. Sudden cardiac death should not be used to
describe events that are not fatal [1].
midaxillary region to the right side of the
xiphoid process. The tip was then advanced up
to the manubriosternal junction, 1 cm to the
right of the midsternal line (Fig. 27.3). At the Epidemiology
end of the procedure, a defibrillation test was
performed: Clinical VT was induced, correctly SCD accounts for approximately 15 % of the
sensed, and treated after 12.5 s with a 65 J total mortality in the industrialized countries [4].
shock (conventional polarity configuration). At The incidence of SCD increases as a function
2-month follow-up examination, the patient of advancing age. The prognosis of patients expe-
did not have any complications, and no signifi- riencing SCA varies significantly according to the
cant events were recorded. initial rhythm, although the outcome remains poor,
despite advances in the treatment of heart disease.
The incidence of experiencing SCA is
27.3 Sudden Cardiac Death increased by age, sex, and underlying cardiac dis-
ease [5]. Indeed, men are two to three times more
Denitions likely to experience SCA than women. In the
Womens Health Initiative, 161,808 postmeno-
Various criteria have been used to define SCA pausal women were followed for an average of
and SCD in the medical literature [3]. It is diffi- 10.8 years, and the incidence rate of SCD was 2.4
cult to find a specific definition for several main per 10,000 women/year, and nearly one-half of
reasons: First of all, only two-thirds of events are women who experienced SCD did not have prior
witnessed, making the diagnosis difficult to clinically recognized coronary heart disease [6].
establish; secondly, the cardiac rhythm at clinical The presence of clinically recognized heart
presentation is unknown in many cases, so it is disease and of coronary heart disease (CHD) risk
not possible to restrict the definition of SCA to factors, respectively, increases six- to tenfold and
documented cases of VF; finally, the suddenness two- to fourfold the risk of SCA [7].
of death is generally defined by the duration of
symptoms prior to SCA, but in approximately
one-third of cases, the duration of symptoms is Etiology
not clearly defined.
For these reasons, the criteria proposed do not SCA is more frequent in male with structural
rely upon the cardiac rhythm at the time of the heart disease, especially CHD. The most frequent
event. The criteria focus on the sudden pulseless cause of SCA is CHD that amounts up to 70 % of
event occurred out of hospital and on the absence SCAs. It is possible that SCA occur during an
of a noncardiac condition (e.g., pulmonary embo- acute coronary syndrome (ACS) such as in
lism, intracranial hemorrhage, central airway patients with a chronic, stable CHD [8]. Usually
obstruction) as the cause of cardiac arrest. in this setting, prior myocardial damage and scar
The following definitions of SCA and SCD act as a substrate for SCA. On the contrary, SCD
were presented by the 2006 American College of occurs more commonly in the absence of an iden-
Cardiology/American Heart Association/Heart tifiable acute cardiac event.
Rhythm Society (ACC/AHA/HRS): Ten percent of cases of out-of-hospital SCA is
Sudden cardiac arrest is the sudden cessation of represented by other forms of structural heart dis-
cardiac activity so that the victim becomes unre- ease, both acquired and hereditary. Examples of
sponsive, with no normal breathing and no signs of such disorders include the following:
circulation. If corrective measures are not taken
rapidly, this condition progresses to sudden death.
Cardiac arrest should be used to signify an event as 1. SCD is responsible for approximately one-
described above, that is reversed, usually by CPR third of deaths in heart failure and
and/or defibrillation or cardioversion, or cardiac cardiomyopathy.

ERRNVPHGLFRVRUJ
27 Sudden Cardiac Death 315

2. Left ventricular hypertrophy due to hyperten- hypomagnesemia), and the proarrhythmic effect
sion or other causes. of some antiarrhythmic drugs. The mortality is
3. Myocarditis. highest in the first month after acute myocardial
4. Hypertrophic cardiomyopathy. infarction (MI) in patients with ejection fraction
5. Arrhythmogenic right ventricular (EF) of less than 30 % [1].
cardiomyopathy. In addition, direct trauma over precordium
6. Congenital coronary artery anomalies. precipitated SCA that results from commotio
7. Mitral valve prolapse. cordis.
The cumulative risk of SCD has been esti-
Data from different reports documented mated at 1520 % of adults with aortic stenosis,
approximately 1012 % of cases of SCA among with the risk being higher in symptomatic patients
subjects without structural heart disease, under and equal to or less than 5 % in asymptomatic
age 45, while a lower value of about 5 % has patients. Mitral valve prolapse is usually non-
been described when older patients are included life-threatening, and its link with SCD has never
[9, 10]. been definitely demonstrated. Reported SCD
Hereditary factors that contribute to coro- rates in patients with Wolff-Parkinson-White
nary heart disease risk have been thought to (WPW) syndrome have been 0.15 %, due in most
operate nonspecifically for the SCD syndrome. to the development of atrial fibrillation (AF) with
However, several studies have identified muta- a rapid ventricular response that degenerates to
tions and relevant polymorphisms along the VF. Genetic influences modulate the risk of SCD
cascade from atherogenesis to plaque destabili- in the setting of CHD. The Paris Prospective
zation, thrombosis, and arrhythmogenesis, each Study I, analyzing more than 7,000 men followed
of which is associated with a risk of a coronary for an average of 23 years, found that a parental
event. history of SCD increased the relative risk of SCD
for offspring to 1.8, without elevating the risk for
MI. When both parents had SCD, the relative risk
for SCD in offspring was 9.4. Genetic influences
SCA in the absence of apparent structural
may act through multiple mechanisms, such as
heart disease can occur in different
modulation of the substrate, atherothrombosis,
settings:
electrogenesis impulse propagation, neural con-
trol, and regulation [1].
1. Brugada syndrome
Some individuals can have inherited abnor-
2. Idiopathic VF
malities that are not manifest until triggered by
3. Congenital or acquired long QT
an external event. For example, autonomic mod-
syndrome
ulation associated with certain types of activity,
4. Arrhythmogenic right ventricular cardio-
as well as drugs that affect cardiac repolariza-
myopathy
tion, can convert a subclinical genetic abnor-
5. Catecholaminergic polymorphic VT
mality to SCD. Among the genetic factors, the
6. Familial SCD of uncertain cause
most common are DNA variants called poly-
7. Wolff-Parkinson-White syndrome
morphisms that may be present in a large pro-
portion of the population and create susceptibility
for SCD. Single nucleotide polymorphisms
A major role in the pathogenesis of SCA, in (SNPs) are DNA variants that can be associated
addition to the presence of the above underlying with a functional consequence. For example, a
disorders, is played by acute superimposed trig- polymorphism identified in the alpha 2b adren-
gers. These include ischemia, autonomic nervous ergic receptor is associated with an increased
system activation, psychosocial factors, electro- risk of MI and SCD [1]. Nevertheless, to create
lyte disturbances (particularly hypokalemia and a risk for SCD, a combination of polymorphisms

ERRNVPHGLFRVRUJ
316 L. Cipolletta

in different genes may be required, interacting Anyway, regular exercise should be encouraged
with a specific trigger or substrate, because mil- for the primary prevention of coronary heart dis-
lions of SNPs are present in the DNA of each ease and consequently SCA. Despite a small
individual [1]. transient increase in risk during and shortly after
strenuous exercise, rate of SCD is lower among
exercisers compared with sedentary men [17]. A
Risk Factors reduced risk of SCD is associated with a lower
resting heart rate and increased heart rate vari-
An increased risk of SCA is associated with a ability, characteristics which developed after
number of clinical characteristics and other regular exercise.
factors among patients without prior clinically Patients should be advised to pay attention to
recognized heart disease [11, 12]. Coronary heart possible symptoms of coronary heart disease,
disease and SCA have a lot of risk factors in even if they are trained exercisers.
common. These include family history of myo- Hypertrophic cardiomyopathy, anomalous
cardial infarction, hypertension, dyslipidemia, coronary artery of wrong sinus origin, myocardi-
diabetes mellitus, obesity, cigarette smoking, and tis, and arrhythmogenic right ventricular cardio-
physical inactivity [13, 14]. myopathy [18, 19] are an exception to the lower
The risk of SCA in patients with coronary overall risk associated with intensive exercise
heart disease is strongly related to current ciga- that occurs in patients with certain, often unrec-
rette smoking and the number of cigarettes ognized underlying heart diseases.
smoked per day. In the Nurses Health Study, A family history of SCA, which could be
101,018 women were followed for 30 years, and associated with myocardial infarction, is related
current smokers had a significantly greater risk of to a 1.5- to 1.8-fold increased risk of SCA [20].
SCD than women who had never smoked Traditional risk factors that tend to aggregate in
(adjusted hazard ratio 2.44, 95 % CI 1.803.31), families, such as hypercholesterolemia, hyper-
and the risk was increased even among women tension, diabetes mellitus, and obesity, do not
who smoked 1 to 14 cigarettes per day (adjusted explain the increase in risk. However, it is likely
hazard ratio 1.84, 95 % CI 1.162.92) [15]. In that interactions of mutations or polymorphisms
this study, the risk of SCD declined over time in in specific genes and environmental factors influ-
a linear fashion after quitting smoking, reaching ence this risk.
the same risk of SCD as never smokers 20 years Elevated serum C-reactive protein (CRP) is
after quitting [15]. also associated with an increased risk of SCA
The risk of SCA is particularly high among [21]. Higher serum concentrations of CRP could
current smokers and declines rapidly after stop- be an expression of chronic inflammation that has
ping smoking, to reduce the risk of SCA and a been implicated as a risk factor for cardiovascu-
multitude of other complications. lar diseases (including acute coronary syndromes
During and up to 30 min after strenuous exer- and stroke).
cise, the risk of SCA is transiently increased com- Heavy alcohol consumption (six or more
pared to other times [16]. In fact, the actual risk drinks per day) or binge drinking increases the
during any one episode of vigorous exercise is risk for SCD [22, 23]. In fact, moderate alcohol
very low (1 per 1.51 million episodes of exercise) intake (e.g., one to two drinks per day) is related
[17]. Also, the magnitude of the transient increase to a reduction of the risk of SCD [22].
in risk during acute exercise is lower for people Clinical observations have suggested that
who perform regular exercise compared with peo- acutely stressful situations could have a possible
ple for whom exercise is unusual [16, 17]. relation with a higher risk of SCA. For example,
Data from long-term exercise intervention tri- major disasters, such as earthquakes and war,
als among apparently healthy persons that focus result in a rapid transient increase in the rate of
upon major disease end points are absent. SCA in populations [15]. Social support from

ERRNVPHGLFRVRUJ
27 Sudden Cardiac Death 317

others may reduce the risk associated with stress- Class I and III antiarrhythmic agents were not
ful life events. able to reduce total and SCD mortality in
Excessive caffeine intake has been investi- patients at risk for SCD [30]. In fact, both
gated as a potential risk factor for SCA [24], but classes do not direct their actions on cardiac
no significant association with SCA has been muscle or specialized conducting tissues that
found. have been shown effective for prevention of
After a myocardial infarction, elevated plasma SCD. Beta-blockers, ACE inhibitors, angioten-
nonesterified fatty acid (free fatty acid) concen- sin receptor-blocking agents, lipid-lowering
trations were associated with ventricular arrhyth- agents, spironolactone, and fibrinolytic and
mias and SCD [25]. However, in the antithrombotic agents were judged likely effec-
Cardiovascular Health Study, in a population- tive especially in primary prevention. Since
based cohort of older adults [26], nonesterified SCD is for the most part the result of a ventricu-
fatty acids were not associated with SCD. lar tachyarrhythmia, these drugs act on the bio-
Moreover, in a population-based case-control chemical, ischemic, and fibrotic processes that
study among people without prior clinically underlie the onset or maintenance of life-threat-
recognized heart disease, cases of SCA had ening ventricular arrhythmias.
higher concentrations of trans isomers of linoleic The mechanisms of cardiac arrest are repre-
acid in red blood cell membranes [27]. sented by electromechanical dissociation, asys-
In contrast, in several studies, a higher dietary tole and heart block, and VF. More permanent
intake and higher plasma levels of long-chain n-3 changes could also occur, such as plaque rup-
polyunsaturated fatty acids (eicosapentaenoic ture, but it is likely that transient factors inter-
acid and docosahexaenoic acid) are associated act with a fixed substrate to precipitate the
with a lower risk of SCD [28]. arrhythmia. The possibilities include physical
activity, transient ischemia, hypoxia, stretch,
ion channel abnormalities, pH and electrolyte
Mechanisms of Sudden Cardiac Death changes, inflammation, neuroendocrine actions,
and drugs.
The rhythm most often recorded at the time of
sudden cardiac arrest is VF. Previous studies
suggest that up to 80 % occur via this mecha-
Management
nism and up to 20 % are attributed to bradyar-
rhythmias, including advanced atrioventricular
(AV) block and asystole [1]. Bayes de Luna
et al. [31] reported that in 157 ambulatory The acute management of survivors of
patients who had SCD while undergoing Holter SCA includes the following:
recording, 62.4 % had VF, 16.5 % had bradyar-
rhythmias, 12.7 % had torsades de pointes, and 1. Identification and treatment of acute
8.3 % had primary VT. The true incidence of reversible causes
bradyarrhythmias is not easy to understand 2. Evaluation for structural heart disease
since a rhythm beginning as VF may appear to 3. Evaluation for primary electrical dis-
be asystole during the first ECG recording. eases in patients without identifiable
Advanced AV block or significant bradycardia arrhythmic triggers or cardiac structural
can provoke VF. It is difficult to identify clearly abnormalities
the electrophysiological mechanism responsible 4. Neurologic and psychologic assessment
for SCD. Mechanisms may be multifactorial 5. Evaluation of family members in
and are different depending on the specific car- selected patients with a suspected or
diac abnormality, and a rhythm can begin via confirmed heritable syndrome
one mechanism and proceed via a different one.

ERRNVPHGLFRVRUJ
318 L. Cipolletta

Noninvasive Management Other Noninvasive ECG Techniques


Also other noninvasive ECG techniques such as
ECG signal-averaged ECG (SAECG), heart rate vari-
A noninvasive evaluation is recommended with ability (HRV), baroreflex sensitivity, and heart
resting 12-lead ECG in all patients who are eval- rate turbulence may be useful to improve the
uated for ventricular arrhythmias (class I, level of diagnosis and risk stratification of patients with
evidence: A). It allows to identify various con- ventricular arrhythmias or who are at risk of
genital abnormalities associated with ventricular developing life-threatening ventricular arrhyth-
arrhythmias and SCD (e.g., LQTS, SQTS, mias (class IIb, level of evidence: B) [1].
Brugada syndrome, ARVC) but also other ECG
modifications, such as those due to electrolyte Echocardiography
disturbances, or evidence suggesting underlying Echocardiography is recommended for the
structural disease, such as bundle branch block; subset of patients at high risk for the develop-
Q waves indicative of ischemic heart disease, ment of SCD, such as those with dilated,
ventricular hypertrophy, or infiltrative cardiomy- hypertrophic, or RV cardiomyopathies, AMI
opathy; and AV block. QRS duration and repolar- survivors, or relatives of patients with inher-
ization abnormalities are both independent ited disorders associated with SCD (class I,
predictors of SCD. A QRS duration greater than level of evidence: B).
130 ms has been associated with increased mor-
tality in patients with a reduced LVEF (equal to Invasive Tests
or less than 30 %). Prospective studies have also
reported an association between ST-segment Coronary Angiography
depression or T-wave abnormalities and increased Coronary angiography can be useful in evaluat-
risk of cardiovascular death and SCD in particu- ing the presence of significant obstructive CHD
lar [1]. QTc greater than 440 ms has been demon- in patients with life-threatening ventricular
strated to predict cardiovascular death with arrhythmias or in survivors of SCD, who have an
adjusted relative risk of 2.1 [31]. Some data sug- intermediate or greater probability of having
gest that the correlation between QTc and sur- CHD by age, symptoms, and gender (class IIa,
vival could be J shaped. So, also relatively level of evidence: C).
short QTc intervals have also been associated
with increased risk. The definition of short Electrophysiological Study (EPS)
QT syndrome as a QTc less than 300 ms has been Invasive tests such as EPS were required in the
proven to be an independent predictor of primary prevention of SCD in patients with the
SCD [32]. same characteristic of patients enrolled in
MADIT, MUSTT, and BEST-ICD trials [1].
Exercise Testing Inducibility of VT in patients with NSVT on
Among noninvasive evaluation, exercise testing is Holter monitoring identified a population at
useful in patients with known or suspected exercise- high risk for VT/VF and ICD use in the MADIT
induced ventricular arrhythmias, including cate- trial [33]. Lower heart rate, lower EF, and a
cholaminergic VT (class I, level of evidence: B). longer interval between MI and an EPS corre-
late with higher inducibility. In patients with
T-Wave Alternans (TWA) ischemic heart disease, asymptomatic NSVT,
It is reasonable to use TWA to improve the diag- and an EF less than 40 %, inducibility of sus-
nosis and risk stratification of patients with ven- tained VT identifies patients at high risk of
tricular arrhythmias or who are at risk for subsequent VT. Persistent inducibility while
developing life-threatening ventricular arrhyth- receiving antiarrhythmic drugs predicts a
mias (class IIa, level of evidence: A). worse prognosis. Patients in whom amiodarone

ERRNVPHGLFRVRUJ
27 Sudden Cardiac Death 319

suppressed VT inducibility or slowed VT to a when immediate defibrillation is available, with


mean cycle length of greater than 400 ms had response time of less than 30 s, such as in moni-
30 % higher mortality compared with patients tored intensive care units and EP laboratories,
who did not respond to amiodarone and had an survival from VF is greater than 90 % [34].
ICD [1]. Synchronized cardioversion is used for unsta-
In dilated cardiomyopathy (DCM), EPS has a ble monomorphic VT, while high-energy unsyn-
low predictive value and low inducibility [1]. chronized shocks at defibrillation doses treat
EPS is not useful in long QT syndrome unstable polymorphic VT and VF. Monomorphic-
(LQTS) and hypertrophic cardiomyopathy. His tolerated VT generally is converted to sinus
role is not yet clear in Brugada syndrome because rhythm by monophasic waveform cardioversion
of the lack of large randomized studies and a uni- synchronized shocks at initial energies of 100 J
form protocol [1]. or higher.
In arrhythmogenic right ventricular cardio- The general aims of advanced life support are
myopathy (ARVC), EPS is useful to guide abla- to establish hemodynamically effective cardiac
tion [1]. rhythm, to optimize ventilation, and to maintain
and support the restored circulation.
Epinephrine, 1 mg intravenously, is adminis-
Management of Cardiac Arrest tered and followed by repeated defibrillation
attempts at 360 J. Epinephrine may be repeated at
Cardiac arrest is described as an abrupt loss of 35-min intervals with defibrillator shocks in-
effective blood flow that causes immediate loss between doses, but high-dose epinephrine does
of consciousness and leads immediately to death not provide added benefit [35].
if untreated. Bradyarrhythmic events or pulseless electrical
The most common electrical mechanisms for activity could be precipitated by metabolic and
cardiac arrest are VF and pulseless VT but also transient factors that must be controlled.
could be a severe bradyarrhythmias, asystole, or Simultaneously, the rescuer should correct the
pulseless electrical activity. Survival probabili- chemistry of the blood focusing on ventilation
ties are better for VT/VF at first presentation than (i.e., improved oxygenation, reversal of acidosis,
for bradyarrhythmias or asystole. and improvement of the underlying electrophysi-
A rapid response time is the major determi- ological condition). Metabolic acidosis of cardiac
nant of survival. So cardiopulmonary resuscita- arrest is corrected by adequate oxygenation of the
tion (CPR) should be started immediately after blood, but sometimes additional correction can be
contacting a response team (class I, level of evi- achieved by intravenous administration of sodium
dence: A). bicarbonate. Excessive quantities can be deleteri-
In an out-of-hospital setting, if an automatic ous. Possible reversible causes, particularly for
external defibrillator (AED) is available, shock bradyarrhythmia and asystole, should be consid-
therapy should be administered according to the ered and/or treated promptly, such as pulmonary
guidelines on CPR (class I, level of evidence: C). embolus, acute MI, hypovolemia, hypoxia, cardiac
Reversible causes and factors contributing to tamponade, tension pneumothorax, acidosis, drug
cardiac arrest should be managed during overdose, hypothermia, and hyperkalemia [1].
advanced life support, such as management of
mechanical factors, hypoxia, electrolyte distur-
bances, and volume depletion (class I, level of Primary Prevention
evidence: C).
If no CPR is initiated, the decrease in cardiac Primary prevention of SCA is tailored according
arrest survival is about 710 % per minute and to the different categories of patients, such as
34 % per minute with CPR. On the contrary, general population, patients surviving an acute

ERRNVPHGLFRVRUJ
320 L. Cipolletta

myocardial infarction, patients with heart failure evaluation, such as with an electrocardiogram or
and cardiomyopathy, and patients with one of the echocardiogram.
congenital disorders associated with an increased
risk of SCA (e.g., Brugada syndrome, congenital Risk factor reduction: As mentioned above,
long QT syndrome, WPW). the majority of the traditional risk factors
associated with coronary heart disease are
General Population also associated with SCA.
Two approaches could be applied to reduce the
risk of SCA in the general population: screening
and risk stratification to identify individuals who
Thus, reduction of these risk factors may
may benefit from specific interventions (e.g.,
affect the incidence of SCA in the gen-
stress testing, screening ECGs) and behavior that
eral population. Such interventions
target the underlying disorders that predispose to
include:
SCA and reduce the risk in any individual (e.g.,
smoking cessation or other lifestyle Effective treatment of hypercholestero-
modifications). lemia
Effective treatment of hypertension
Screening and risk stratification: In patients Heart-healthy diet
with known elevated risk of SCA (e.g., Regular exercise
patients with a prior myocardial infarction), Smoking cessation
further risk stratification can identify sub- Moderation of alcohol consumption
groups that benefit from specific therapies, Effective treatment of diabetes
such as an ICD. On the contrary, in the gen-
eral population without known cardiovascu-
lar disease, routine screening with 12-lead
electrocardiography, exercise stress testing, There is no definitive evidence that risk fac-
or Holter monitoring hardly or ineffectively tor reduction in the general population lowers
identifies populations at an increased risk the rate of SCA. On the contrary, several studies
of SCA. have demonstrated that treatment of risk factors
can lower total cardiovascular and coronary
mortality. Reduction of cardiovascular risk fac-
Key point in risk stratification of the gen- tors may reduce SCA rates as well because the
eral population could be the following: majority of CHD mortality is due to SCD.
In observational studies of populations at low
Identifying risk factors for cardiovascu- cardiovascular risk, greater dietary fatty fish
lar disease according to standard intake was associated with lower cardiac mortal-
guidelines ity [36]. This benefit is partially due to a reduced
Screening for coronary heart disease in risk of SCD. From these results, subsequent ran-
selected patients domized trials evaluated the benefit of fish oil
supplements in high-risk populations [37].
Routine additional testing for the SCA risk The benefit of pharmacologic doses of n-3
stratification is not recommended. polyunsaturated fatty acids is little compared to
the intake of one to two servings of fatty fish per
week. The pharmacologic use of fish oil supple-
A complex topic matter of debate is the pre- ments should be restricted to patients with refrac-
participation evaluation of athletes. There are tory hypertriglyceridemia in whom the periodic
conflicting opinions regarding the appropriate- monitoring of apolipoprotein B levels is recom-
ness of screening and, if so, the optimal screening mended [37].

ERRNVPHGLFRVRUJ
27 Sudden Cardiac Death 321

Post-myocardial Infarction status for more than 1 year (class I, level of evi-
The risk of SCA is increased in patients who have dence: B) [1].
had a myocardial infarction (MI), and this risk var- Ventricular arrhythmias and SCD are common
ies significantly according to a number of factors. in patients with symptomatic acute and chronic
The strategies to the prevention of SCA in HF and LV systolic dysfunction. In the setting of
such patients include the following: acute HF, ventricular arrhythmias may be poorly
tolerated and early cardioversion should be per-
Standard medical therapies: Both beta- formed, rather than attempting pharmacological
blockers and ACE inhibitors (or angiotensin termination of arrhythmia.
II receptor blockers) reduce overall mortality In addition, standard medical therapies for HF
after an MI and are routinely administered. (beta-blockers, ACE inhibitors or angiotensin II
These agents also lower the incidence of receptor blockers, and aldosterone inhibitors
SCD. such as spironolactone or eplerenone) may lower
Risk stratification: To identify those patients the risk of SCA [1].
at the highest risk of SCA.
ICD implantation in selected patients: ICD
therapy is recommended for primary preven- Secondary Prevention
tion to reduce SCD in patients with LV dys-
function due to prior MI who are at least 40 ICD Therapy
days post-MI, have an LVEF less than or equal An implantable cardioverter defibrillator (ICD)
to 3040 %, are NYHA functional class II or is the preferred therapeutic modality in survi-
III, are receiving chronic optimal medical vors of SCA. The ICD does not prevent the
therapy, and have a reasonable expectation of recurrence of malignant ventricular arrhyth-
survival with a good functional status for more mias, but it effectively terminates these arrhyth-
than 1 year (class I, level of evidence: A) [1]. mia recurrences.
ICD patients who have frequent arrhythmia
In patients with LV dysfunction due to prior recurrences and device discharges may benefit
MI who are at least 40 days post-MI, have an from adjunctive therapies, such as antiarrhythmic
LVEF of less than or equal to 3035 %, are drugs or catheter ablation.
NYHA functional class I on chronic optimal Coronary revascularization reduces the risk
medical therapy, and have a reasonable expecta- of SCD in patients with VF when evidence of
tion of survival with a good functional status for acute myocardial ischemia is documented to pre-
more than 1 year, implantation of an ICD is rea- cede the onset of VF (class I, level of evidence:
sonable (class IIa, level of evidence: B) [1]. B) [1]. With evidence of prior MI and significant
LV dysfunction, if coronary revascularization
Heart Failure and Cardiomyopathy cannot be realized, the primary therapy of
Patients with heart failure and left ventricular sys- patients resuscitated from VF should be the ICD
tolic dysfunction, regardless of the etiology, are at in patients who are receiving chronic optimal
an increased risk of SCA. Primary prevention with medical therapy and those who have a reason-
an ICD is recommended in selected patients with able expectation of survival with a good func-
either ischemic or nonischemic cardiomyopathy tional status for more than 1 year (class I, level
(NICM). ICD therapy is recommended for primary of evidence: A) [1].
prevention to reduce total mortality by a reduction In patients with LV dysfunction due to prior
in SCD in patients with nonischemic heart disease MI with hemodynamically unstable sustained
who have an LVEF less than or equal to 3035 %, VT, receiving chronic optimal medical therapy,
are NYHA functional class II or III, are receiving with a reasonable expectation of survival for
chronic optimal medical therapy, and have a reason- more than 1 year, ICD is the effective therapy to
able expectation of survival with a good functional reduce SCD (class I, level of evidence: A) [1].

ERRNVPHGLFRVRUJ
322 L. Cipolletta

ICD therapy is recommended for secondary causes. ICD implantation is indicated in patients
prevention of SCD in patients who survived VF who are receiving chronic optimal medical ther-
or hemodynamically unstable VT or VT with apy and who have a reasonable expectation of
syncope and who have an LVEF less than or survival with a good functional status for more
equal to 40 %, who are receiving chronic optimal than 1 year (class I, level of evidence: B) [1].
medical therapy, and who have a reasonable Congenital heart disease represents a spectrum
expectation of survival with a good functional of anatomical and physiological defects with an
status for more than 1 year (class I, level of evi- intrinsic risk of arrhythmias and late SCD. Overall,
dence: A) [1]. congenital heart defects are the principal cause of
Amiodarone is a reasonable therapy to reduce infant mortality (less than 1 year of age) in the
symptoms due to recurrent hemodynamically sta- industrialized world. About all defects can now be
ble VT for patients with LV dysfunction due to repaired or palliated, with greater than 1 million
prior MI who cannot have or refuse to have an ICD worldwide long-term survivors after surgery for
implanted (class IIa, level of evidence: C) [1]. congenital heart disease [38].
ICD implantation is a reasonable treatment of Due to the low incidence of late SCD in post-
recurrent ventricular tachycardia in patients post- operative congenital heart disease patients, pro-
MI with normal or near-normal ventricular func- spective randomized clinical trials do not exist to
tion who are receiving chronic optimal medical identify either risk factors for SCD or the role of
therapy and who have a reasonable expectation of primary prevention therapies.
survival with a good functional status for more During infancy and childhood, greater than
than 1 year (class IIa, level of evidence: C) [1]. 75 % of deaths in patients with congenital heart
disease are in-hospital events, during the periop-
Antiarrhythmic Drugs erative period [39].
Antiarrhythmic drugs are less effective than an The remaining deaths often occur in patients
ICD for secondary prevention of SCD. Thus, their with other congenital anomalies in an out-of-
use is limited to the adjunctive role in frequent ven- hospital setting.
tricular arrhythmia recurrences or in patients who Beyond 20 years of age, a progressive increase
do not want or are not candidates for an ICD (e.g., in the incidence of sudden and total cardiac mor-
due to marked comorbidities or end-stage heart tality occurs in postoperative congenital heart
failure with a life expectancy less than 1 year). disease patients [40].
Amiodarone, sotalol, and/or other beta- Congenital heart defects associated with the
blockers are recommended pharmacological greatest risks of late SCD are tetralogy of Fallot,
adjuncts to ICD therapy to suppress symptomatic D- and L-transposition of the great arteries, aortic
ventricular tachyarrhythmias in optimally treated stenosis, and functional single ventricle [41].
patients with HF (class I, level of evidence: C) [1]. The additional factors most likely associated
Curative catheter ablation or amiodarone may with an increased risk of SCD due to ventricular
be considered in place of ICD therapy to improve arrhythmias appear to be volume overload due to
symptoms in patients with LV dysfunction due to pulmonary insufficiency and QRS duration
prior MI and recurrent hemodynamically stable greater than 160 ms [42]. EPS for risk stratifica-
VT whose LVEF is greater than 40 % (class IIb, tion in these patients is not useful, in part due to
level of evidence: B) [1]. variable study protocols and response to such
testing [1].
The mechanism of SCD appears to be atrial flut-
Congenital Heart Disease ter with 1:1 AV conduction, followed by myocar-
dial ischemia resulting in polymorphic VT or VF or
ICD implantation is indicated in patients with progressive ventricular dysfunction that results in
congenital heart disease who are survivors of car- ventricular arrhythmias [42]. Moreover, a positive
diac arrest after exclusion of any reversible response to EPS, independent of the clinical indica-

ERRNVPHGLFRVRUJ
27 Sudden Cardiac Death 323

tion, may identify patients with a high risk of late that is not manageable by reprogramming or
SCD [42]. Also, there are non-arrhythmic causes of changing drug therapy or who do not wish long-
late sudden death in postoperative patients, includ- term drug therapy (class I, level of evidence: C)
ing cerebral or pulmonary embolism, endocarditis, [1]. Finally, ablation is indicated in patients with
and aneurysm rupture [40]. WPW syndrome resuscitated from sudden car-
Coronary artery abnormalities are another diac arrest due to rapidly conducted atrial fibrilla-
class of congenital anomalies that may result in tion causing VF (class I, level of evidence: B) [1].
SCD. The most common congenital coronary Less evidence exists regarding ablation of
artery anomaly leading to SCD in the young is Purkinje fiber potentials that may be considered
anomalous origin of the left coronary artery from in patients with ventricular arrhythmia storm
the right sinus of Valsalva. The mechanism of consistently provoked by PVCs of similar mor-
SCD is that acute angulation of the coronary phology (class IIb, level of evidence: C) [1].
ostium or compression of the left coronary artery Also ablation of asymptomatic PVCs may be
on the region between the aortic wall and RVOT considered when the PVCs are very frequent to
results in acute myocardial ischemia and the sub- avoid or treat tachycardia-induced cardiomyopa-
sequent VT or VF. The risk of SCD is greatest thy and when PVCs are trigger of untolerated
during the first three decades of life. Diagnosis VT/VF [1] (class IIb, level of evidence: C).
may be difficult because only one-third of
patients have exertional syncope or angina.
Coronary angiography is needed to definitive References
diagnosis and the treatment is surgical revascu-
larization. Similar risks for SCD have also been 1. Zipes DP, Camm AJ, Borggrefe M et al (2006)
American College of Cardiology; American Heart
defined for anomalous origin of the right coro-
Association Task Force; European Society of
nary artery from the left sinus of Valsalva [41]. Cardiology Committee for Practice Guidelines;
Anomalous origin of the left coronary artery European Heart Rhythm Association; Heart Rhythm
from the pulmonary artery generally is mani- Society. Europace 8:746837
2. Demirovic J, Myerburg RJ (1994) Epidemiology of
fested during the first month of life. Myocardial
sudden coronary death: an overview. Prog Cardiovasc
ischemia and dysfunction develop with the nor- Dis 37:39
mal decline in pulmonary vascular resistance 3. Zheng ZJ, Croft JB, Giles WH, Mensah GA (2001)
when coronary perfusion is shunted to the pul- Sudden cardiac death in the United States, 1989 to
1998. Circulation 104:2158
monary circulation. The diagnosis is echocardio-
4. Chugh SS, Jui J, Gunson K et al (2004) Current burden
graphic during infancy, and the definitive of sudden cardiac death: multiple source surveillance
treatment is surgical reimplantation of the left versus retrospective death certificate-based review in a
coronary ostium [41]. large U.S. community. J Am Coll Cardiol 44:1268
5. Kuller LH (1980) Sudden deathdefinition and epide-
miologic considerations. Prog Cardiovasc Dis 23:1
6. Drory Y, Turetz Y, Hiss Y et al (1991) Sudden unex-
Ablation pected death in persons less than 40 years of age. Am
J Cardiol 68:1388
7. Topaz O, Edwards JE (1985) Pathologic features of
First of all, ablation is indicated in patients who
sudden death in children, adolescents, and young
are at low risk for SCD, who are drug intolerant, adults. Chest 87:476
or who do not wish long-term drug therapy and 8. Chugh SS, Kelly KL, Titus JL (2000) Sudden cardiac
have sustained predominantly monomorphic VT death with apparently normal heart. Circulation
102:649
that is drug resistant (class I, level of evidence: C)
9. Survivors of out-of-hospital cardiac arrest with appar-
[1]. Secondly, it is indicated in patients with bun- ently normal heart. Need for definition and standard-
dle branch reentrant VT (class I, level of evi- ized clinical evaluation. Consensus Statement of the
dence: C) [1]. Thirdly, it is indicated as adjunctive Joint Steering Committees of the Unexplained
Cardiac Arrest Registry of Europe and of the
therapy in patients with an ICD who are receiv-
Idiopathic Ventricular Fibrillation Registry of the
ing multiple shocks as a result of sustained VT United States (1997) Circulation 95:265

ERRNVPHGLFRVRUJ
324 L. Cipolletta

10. Siscovick DS, Weiss NS, Hallstrom AP et al (1982) 28. Harper CR, Jacobson TA (2001) The fats of life: the
Physical activity and primary cardiac arrest. JAMA role of omega-3 fatty acids in the prevention of coro-
248:3113 nary heart disease. Arch Intern Med 161:2185
11. Friedlander Y, Siscovick DS, Weinmann S et al (1998) 29. Albert CM, Campos H, Stampfer MJ et al (2002)
Family history as a risk factor for primary cardiac Blood levels of long-chain n-3 fatty acids and the risk
arrest. Circulation 97:155 of sudden death. N Engl J Med 346:1113
12. Siscovick DS, Weiss NS, Fletcher RH, Lasky T 30. Marchioli R, Barzi F, Bomba E et al (2002) Early pro-
(1984) The incidence of primary cardiac arrest during tection against sudden death by n-3 polyunsaturated
vigorous exercise. N Engl J Med 311:874 fatty acids after myocardial infarction: time-course
13. Kark JD, Goldman S, Epstein L (1995) Iraqi missile analysis of the results of the Gruppo Italiano per lo
attacks on Israel. The association of mortality with a Studio della Sopravvivenza nellInfarto Miocardico
life-threatening stressor. JAMA 273:1208 (GISSI)-Prevenzione. Circulation 105:1897
14. Siscovick DS, Raghunathan TE, King I et al (1995) 31. Bayes de Luna A, Coumel P, Leclercq JF (1989)
Dietary intake and cell membrane levels of long-chain Ambulatory sudden cardiac death: mechanisms of
n-3 polyunsaturated fatty acids and the risk of primary production of fatal arrhythmia on the basis of data
cardiac arrest. JAMA 274:1363 from 157 cases. Am Heart J 117:151159
15. Trichopoulos D, Katsouyanni K, Zavitsanos X et al 32. Alberte C, Zipes DP (2003) Use of nonantiarrhythmic
(1983) Psychological stress and fatal heart attack: the drugs for prevention of sudden cardiac death. J
Athens (1981) earthquake natural experiment. Lancet Cardiovasc Electrophysiol 14:S87S95
1:441 33. Schouten EG, Dekker JM, Meppelink P et al (1991)
16. Kromhout D, Bosschieter EB, De Lezenne Coulander QT interval prolongation predicts cardiovascular mor-
C (1985) The inverse relation between fish consump- tality in an apparently healthy population. Circulation
tion and 20-year mortality from coronary heart dis- 84:15161523
ease. N Engl J Med 312:1205 34. Capucci A, Aschieri D, Piepoli MF et al (2002) Tripling
17. Albert CM, Mittleman MA, Chae CU et al (2000) survival from sudden cardiac arrest via early defibrilla-
Triggering of sudden death from cardiac causes by tion without traditional education in cardiopulmonary
vigorous exertion. N Engl J Med 343:1355 resuscitation. Circulation 106(9):10651070
18. Maron BJ, Carney KP, Lever HM et al (2003) 35. Gussak I, Brugada P, Brugada J et al (2000) Idiopathic
Relationship of race to sudden cardiac death in com- short QT interval: a new clinical syndrome?
petitive athletes with hypertrophic cardiomyopathy. J Cardiology 94:99102
Am Coll Cardiol 41:974 36. Moss AJ, Hall WJ, Cannom DS et al (1996) Improved
19. Corrado D, Basso C, Schiavon M, Thiene G (1998) survival with an implanted defibrillator in patients
Screening for hypertrophic cardiomyopathy in young with coronary disease at high risk for ventricular
athletes. N Engl J Med 339:364 arrhythmia. Multicenter Automatic Defibrillator
20. Jouven X, Desnos M, Guerot C, Ducimetire P (1999) Implantation Trial Investigators. N Engl J Med 335:
Predicting sudden death in the population: the Paris 19331940
Prospective Study I. Circulation 99:1978 37. Gueugniaud PY, Mols P, Goldstein P et al (1998) A
21. Albert CM, Ma J, Rifai N et al (2002) Prospective comparison of repeated high doses and repeated stan-
study of C-reactive protein, homocysteine, and plasma dard doses of epinephrine for cardiac arrest outside
lipid levels as predictors of sudden cardiac death. the hospital. European Epinephrine Study Group. N
Circulation 105:2595 Engl J Med 339:15951601
22. Albert CM, Manson JE, Cook NR et al (1999) Moderate 38. Perloff JK, Warnes CA (2001) Challenges posed by
alcohol consumption and the risk of sudden cardiac adults with repaired congenital heart disease.
death among US male physicians. Circulation 100:944 Circulation 103:26372643
23. Wannamethee G, Shaper AG (1992) Alcohol and sud- 39. Centers for Disease Control and Prevention (CDC)
den cardiac death. Br Heart J 68:443 (2002) State-specific mortality from sudden cardiac
24. Weinmann S, Siscovick DS, Raghunathan TE et al deathUnited States, 1999. MMWR Morb Mortal
(1997) Caffeine intake in relation to the risk of pri- Wkly Rep 51:123126
mary cardiac arrest. Epidemiology 8:505 40. Gatzoulis MA, Balaji S, Webber SA et al (2000) Risk
25. Jouven X, Charles MA, Desnos M, Ducimetire P factors for arrhythmia and sudden cardiac death late
(2001) Circulating nonesterified fatty acid level as a after repair of tetralogy of Fallot: a multicentre study.
predictive risk factor for sudden death in the popula- Lancet 356:975981
tion. Circulation 104:756 41. Alexander ME, Walsh EP, Saul JP et al (1999) Value
26. Djouss L, Biggs ML, Ix JH et al (2012) Nonesterified of programmed ventricular stimulation in patients
fatty acids and risk of sudden cardiac death in older with congenital heart disease. J Cardiovasc
adults. Circ Arrhythm Electrophysiol 5:273 Electrophysiol 10:10331044
27. Lemaitre RN, King IB, Raghunathan TE et al (2002) 42. Liberthson RR (1996) Sudden death from cardiac
Cell membrane trans-fatty acids and the risk of pri- causes in children and young adults. N Engl J Med
mary cardiac arrest. Circulation 105:697 334:10391044

ERRNVPHGLFRVRUJ
The Role of Home Monitoring
28
Laura Cipolletta and Jenny Ricciotti

28.1 Case Report


At the time of implantation, ventricu-
lar sensing with a stable intrinsic ampli-
tude of 11 mV without T-wave sensing
A 42-year-old Caucasian man suffering was achieved. The ICD was programmed
from type 1 Brugada syndrome was with a VT detection zone at 179 bpm with
referred to our department in 2007 after an only monitoring and no therapy and a
episode of palpitations followed by syn- second detection zone at 214 bpm, with
cope while driving his car. The electrocar- antitachycardia pacing (ATP) during
diogram (EKG) showed a type 1 Brugada charging and shock at 36/40/40 J for three
syndrome pattern, and further history times.
revealed two syncopal episodes 2 years Subsequent follow-up was organized
later during fever. When asked about sud- with outpatient visits every 6 months; in
den cardiac death in his family, he reported addition, starting from February 2011,
that a maternal uncle had died at age of 40 when the ICD generator was replaced at the
for unknown reasons. end of battery life, the patient was equipped
A transvenous single-chamber implant- with a remote monitoring system (St. Jude
able cardioverter defibrillator (ICD), St. Medical Merlin.net) that can record data
Jude Fortify VR, equipped with a ventricu- relative to arrhythmic events and technical
lar electrocatheter system was implanted, details of the device and leads and transmit
and the patient was subsequently dis- regularly to the cardiology center (on aver-
charged in stable condition. age, 1 transmission/month, plus any addi-
tional transmission in case of detected
alarm episodes).
The patient had never received ICD
therapy over 6 years follow-up; all func-
tional and lead integrity parameters were
normal during all the follow-up visits.
L. Cipolletta (*) J. Ricciotti
Clinica di Cardiologia e Aritmologia, Universit
Politecnica delle Marche,
Via Conca, 71, Ancona 60126, Italy
e-mail: lauracipo2006@libero.it;
jen.ricciotti@tiscali.it

Springer International Publishing Switzerland 2015 325


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_28

ERRNVPHGLFRVRUJ
326 L. Cipolletta and J. Ricciotti

Figure 28.1 shows an episode interpreted as


In the summer of 2013, we received a ventricular tachycardia (VT)/ventricular fibrilla-
nonprogrammed remote transmission with an tion (VF) with delivery of ATP during charging
alert of many episodes of tachyarrhythmia, of the capacitors; indeed the beats marked as VF
marked as supraventricular and ventricular, correspond to deflections of high amplitude, not
both sustained and nonsustained, with con- constantly present, identifiable as noise in the
sequent ICD therapy deliveries. By exam- ventricular channel.
ining all the transmissions in detail, we can Noise and artifacts can lead to oversensing
see how the delivered therapies resulted and cause inappropriate ICD therapies; they can
inappropriately and recognition of arrhyth- be classified into different types:
mias was incorrect.
External noise (e.g., magnetic interference)
usually present on all sensing channels, con-
What Are the Possible Causes stant and regular, with high frequency
for Inappropriate ICD Therapy? (~5060 Hz)
Myopotential noise with high frequency and
low and constant amplitude, which can vary
Supraventricular arrhythmias with rapid with respiration
ventricular conduction Internal noise, due to problem in the lead con-
Abnormal sensing (and esp. oversensing ductors with high-frequency and highly vari-
with double T-wave counting) able amplitude, which occurs intermittently,
Lead malfunction separated by periods of isoelectric baseline
Electromagnetic interference and typically on one lead/channel

Fig. 28.1 Episode marked as VT/VF. The far-field electrogram (A) is present on the top, the near-field ventricular
sense channel (V) is present in the middle tracing, and the marker channel is on the bottom

ERRNVPHGLFRVRUJ
28 The Role of Home Monitoring 327

Fig. 28.2 VF episode secondary to T-wave oversensing (top). Capture test (bottom)

The episodes marked as VF are likely to be an Figure 28.2 shows another episode labeled as
artifact with the characteristics of high amplitude VF, inappropriately treated with ATP, and capaci-
and low frequency, not constant but intermittent, tors charging subsequently aborted; in reality it is
which suggest a problem in the integrity of ven- not a real VF but an oversensing phenomenon
tricular lead. from double T-wave counting.

ERRNVPHGLFRVRUJ
328 L. Cipolletta and J. Ricciotti

The patient was called for an ambulatory visit. Physical Examination


During ICD interrogation, the anomalies already
showed with remote monitoring were confirmed, Weight 70 kg, height 167 cm, and estimated
the usual tests were performed, and the parame- body mass index (BMI) of 25.
ters of sensing, pacing, and ventricular lead General: alert, awake, and oriented.
impedance (sensing 11.7 mV, impedance pacing Skin: normal in appearance, texture, and tem-
460 , shock impedance 50 ) were evaluated. It perature; no rashes; no lesions; no erythema.
was noticed that each paced ventricular beat was Head, eyes, ears, nose, and throat: normal.
followed by artifacts recorded on ventricular Neck: no abnormal adenopathy in the cervical
channel; that confirmed the malfunction of the or supraclavicular areas. Thyroid gland is nor-
device (Fig. 28.2). Furthermore the noise could mal without masses. No carotid bruit and no
be reproduced by isometric maneuvers and with jugular venous distention.
pocket manipulation. Cardiovascular: Regular rate and rhythm; S1
After confirmation of ICD system malfunc- and S2 normal; no murmurs, rubs, or gallops
tion, the patient was hospitalized in the cardiol- heard; point of maximal intensity nondis-
ogy clinic. placed and nonsustained; and no hepatojugu-
lar reflux.
Lungs: Lungs are clear to auscultation and
Medical History and Cardiovascular percussion bilaterally and no alterations in
Risk Factors tactile fremitus.
Abdomen: The abdomen is symmetrical with-
No cardiovascular risk factor. Hiatal hernia. out distention; bowel sounds are normal in
Thalassemia trait quality and intensity in all areas. No masses or
splenomegaly noted; liver span is 6 cm by
percussion.
Allergies Extremities: No cyanosis, clubbing, or edema
noted. Peripheral are normal in all areas.
No allergy is referred by the patient. Neurologic: Cranial nerves IIXII are normal.
Motor and sensory examination of the upper and
lower extremities is normal. Reflexes are normal
Social History and symmetrical bilaterally in both extremities.

The patient works as an employer. Does not


smoke or drink alcohol. He never used illicit Routine EKG at Rest (Fig. 28.3)
drugs.
Conclusions: sinus rhythm, normal atrioventricu-
lar conduction, and type 1 Brugada syndrome
Medications pattern saddleback in V2.
All the routine laboratory tests were performed,
He was not on any medication. including complete blood count, liver and renal
functions, electrolytes, thyroid function, and lipid
concentrationsall resulted normal.
Vital Signs

Temperature 36.4 C, heart rate 75 bpm, blood Echocardiography


pressure 120/80 mmHg, respiratory rate 16
breaths per minute, and oxygen saturation in Tricuspid Aortic Valve with Normal Valve
ambient air 97 % Opening. Normal size of aortic bulb, ascending

ERRNVPHGLFRVRUJ
28 The Role of Home Monitoring 329

Fig. 28.3 EKG. Conclusions: sinus rhythm, normal atrioventricular conduction, type 1 Brugada syndrome pattern
saddleback in V2

aorta, aortic arch, and abdominal aorta. Left exclude a macroscopic failure of the lead, a flu-
atrium of normal size (LA diameter oroscopy was made, which showed no images
M-mode = 30 mm, area 14 cm2). Right atrium was related to fractures or insulation defects of the
normal (area 4c = 14 cm2). Prolapse of the anterior conductors.
mitral leaflet. The tricuspid valve was normal. Given the sensing defect of the ventricular
Normal Dimensions and Thicknesses of the lead, we have reprogrammed sensitivity parame-
Left Ventricle. Normal contractile function (ejec- ters, in order to reduce the oversensing phenom-
tion fraction 60 % with Simpsons biplane enon; when sensitivity parameters of an ICD are
method) and normal wall motion. Right ventricle changed, it is always advisable to perform a defi-
of normal size and with normal contractile func- brillation threshold test (DFT).
tion (TAPSE = 29 mm). Inferior vena cava of nor- During the DFT (Fig. 28.4) after the first
mal size (13 mm), with normal collapse during shock at 2 J to induce VF, some noise entered the
inspiration. The pericardium is normal. Color ventricular channel hiding the real VF; when the
Doppler: mild mitral regurgitation. Normal dia- noise has stopped, the VF signal was very low
stolic function. Minimal tricuspid regurgitation. and unclassified beats were recorded. The shock
PAPs about 20 mmHg. was effectively delivered late by the device, and
There are a lot of factors that should be during the pacing post-shock, every paced beat
weighed in the management of a failed ICD was followed by noise and ICD began to recharge
lead. already the capacitors. To avoid that other
First, the suspected mechanism of lead fail- inappropriate shocks were delivered, ICDs were
ure should be evaluated; for this reason to turned off.

ERRNVPHGLFRVRUJ
330 L. Cipolletta and J. Ricciotti

Fig. 28.4 Defibrillation threshold test

What Are the Treatment Options


in Case of Malfunction of an ICD 2. Lead extraction. The 2009 Heart
Ventricular Lead? Rhythm Society consensus recommen-
dations for the extraction of a nonfunc-
tional, noninfected ICD lead [1] are
1. Add a new pace/sense lead alone, leav- reported in Fig. 28.5. The extraction
ing in place the old lead, if the shock procedure has a high morbidity with
circuit is working correctly. This may be possible fatal complications (cardiac or
the appropriate strategy in older patients, vascular avulsion, cardiac tamponade,
patients with multiple comorbidities, or pulmonary and systemic embolism,
patients for whom lead extraction is retained ICD lead fragments, lead dis-
prohibitively high risk. The disadvan- lodgement, or damage to other existing
tages of adding new leads without leads). Passive fixation leads and
removal of the failed leads include mul- dual-coil leads may be more difficult to
tiple leads crossing the tricuspid valve, extract because of increased adhesions.
lead-to-lead interaction, and an
increased risk of future lead-related
problems. If lead extraction became Management in every case needs to be
necessary in the future, the complexity individualized.
and risks would be higher with more In our case, given the young age of the patient,
leads. with no comorbidities, with a long life expec-
tancy, and with an average time from the

ERRNVPHGLFRVRUJ
28 The Role of Home Monitoring 331

Fig. 28.5 Indications to transvenous lead extraction in patients with nonfunctional leads [1]

implantation of the lead, we decided to proceed implanted subcutaneously without using


with the extraction. The procedure was per- intracardiac and intravascular leads.
formed under local anesthesia by an interven- The indications for implant of an S-ICD are
tional cardiology in the electrophysiology lab the same as a transvenous ICD, excluding the
with a cardiac surgeon on standby. After lead iso- need for pacing for bradycardia and recurring
lation with an excimer laser sheath system, its ventricular tachycardia (VT) that is reliably ter-
removal was performed without complications. minated with antitachycardia pacing (ATP).
Few days later, the patient underwent implan- For these reasons, it is therefore particu-
tation of a subcutaneous ICD (S-ICD, Cameron larly indicated for young patients, with an
Health). This new type of cardioverter defibrilla- active lifestyle and a long life expectancy,
tor was approved in 2012 by the FDA; it is at risk of sudden cardiac death due to ven-

Fig. 28.6 Chest X-ray postimplantation of the S-ICD (posteroanterior and lateral views)

ERRNVPHGLFRVRUJ
332 L. Cipolletta and J. Ricciotti

tricular tachyarrhythmias, as in the genetic parameters can be individualized. This function of


arrhythmogenic syndromes (Brugada syn- the system can be summarized in three steps:
drome, long and short QT syndrome, and other
channelopathies). 1. The first step is the communication between
After the implantation, a chest X-ray was per- the implanted generator and the patient device,
formed that confirmed the correct placement of named CardioMessenger (CM). On the sched-
the S-ICD system (Fig. 28.6). uled time, the patient must be from 20 cm to
The patient was discharged in stable condition. 2 m far from the CM to ensure successful
transmission.
Conclusion 2. The second step is the delivery of the CM
message to the service central, via GSM/
Inappropriate therapies delivered from the ICD, GPRS cellular. A specific website shows the
due to the malfunction of ventricular lead, treated device periodical report of the Biotronik
with transvenous lead extraction and implant of Home Monitoring system that includes the
the new S-ICD. Home monitoring was crucial for intracavitary EKG. At the top, the mark chan-
early defect detection. nel is visualized, and at the bottom, the
electrocardiograms are observed. The system
automatically performs the following trans-
28.2 Home Monitoring missions: (1) Every 24 h, at a physician-
scheduled time, the device sends a transmission
Introduction containing the data recorded along the last
24 h that is forwarded by CM to the service
Routine device follow-up assessment at regular center. (2) A message is immediately sent
intervals is required after implant of an elec- when a serious cardiac event or change hap-
tronic device (PMK or ICD) [2]. The purpose of pens in the device parameters.
these assessments is to evaluate the patient and 3. In the third step, the physician receives a
the device control. Traditionally, device follow- report issued from the service center, named
up clinic is designated to realize these assess- CardioReport, that is possible to consult in an
ments. In the last years, a new technology such Internet-based secure webpage. The physi-
as the remote monitoring (RM) is developed cian, from any computer, will have access to
and permits surveillance and device assessment whole information related to the event and to
from any location of the patient. RM is impor- the device and leads, which will have the fol-
tant to facilitate early diagnosis of arrhythmia lowing characteristics:
and evaluate earlier than in-office visit problems
with the device or leads. Now, in the interna- Home page: that allows the physician to rap-
tional guidelines, RM of implantable devices idly visualize patients who need more
has been indicated as a new standard for patient attention
follow-up, and it has been defined as an alterna- Consolidated report (CardioReport), continu-
tive to the large part of scheduled follow-up vis- ously updated
its [3]. Control and recognition of changed events
and the possibility to recognize alert changes
Fields to add comments and remarks, which
Types of Remote Monitoring Systems relate to both the patient and his/her clinical
history
Home Monitoring (Biotronik) Time line: history of detailed events on a time
It was introduced first in 2001. This technology line
automatically permits the transmission of data on a
daily basis at fixed intervals and soon after a clini- CareLink Network
cally relevant event has occurred. It is wireless and With this system, data from the device, via a
does not require patient action. The surveillance common telephonic connection, are sent to a ser-
ERRNVPHGLFRVRUJ
28 The Role of Home Monitoring 333

vice center. The physician enters a personal low-up, the use of remote pacemaker interrogation
username and password in a specific website to detected actionable events that are potentially
access to data sent. A transmission is made by the important more quickly and more frequently than
patient connecting the RM device to the tele- transtelephonic rhythm strip recordings [7].
phone plug to establish the first connection with Several other clinical studies showed the ben-
the cardiac implantable electronic device (CIED) efits related to the use of RM such as the reduc-
and pressing the button to start the data transfer. tion of outpatient clinic workload, better patient
The CareLink website immediately allows the quality of life, improved implanted system sur-
consultation of exactly the same actualized data veillance, and continuous patient monitoring to
observed in the office. It is always possible to early detect harmful clinical events and to
consult every transmission from the file archive improve patient outcome [812].
on the website. All more recent Medtronic CIED
are compatible with CareLink. The parameters Latitude (Boston Scientic)
showed in the website are the following: This system also utilizes wireless transmission
from the device to a communicator and then to a
Complete program of the CIED central repository. From there, the data is made
Initial report available on an Internet platform. As with the
21 s of EGM recorded at the moment of send- other systems, the Internet site is the same as the
ing data respective manufacturers programmer in order
Battery voltage and estimated remaining life to make the system easier. With this system
of CIED scheduled appointments for remote controlling
Percentage of pacing can be programmed. Additionally, a red light on
Atrioventricular conduction the communicator prompts the patient to initiate
Atrial and ventricular impedance on-demand transmission if an alert has been
Atrial and ventricular automatic threshold detected. Furthermore, the Latitude System
High ventricular rate episodes optionally allows for connection of a blood pres-
High atrial rate episodes sure cuff and a weighing scale for ambulatory
Transthoracic impedance monitoring of heart failure patients.

It is possible to program manually automatic Merlin.net (St. Jude Medical)


transmissions at intervals not shorter than 21 days. The implant system is provided through wireless
The OptiVol sensor incorporated in the telemetry, and follow-up is provided through the
CareLink network measures the drop of intratho- RF Merlin@home transmitter, which receives
racic impedance upon intrapulmonary fluid accu- the DirectCall messages. The monitoring sys-
mulation. Yu et al. demonstrated that the impedance tem sends daily data (DirectAlerts) that are
drop preceded the onset of clinical heart failure exported to a database. A patient can start the
symptoms by a mean of 15 days [6]. This feature is interrogation pressing the start button of the
a powerful tool to prevent heart failure hospitaliza- Merlin@home system that has status lights for
tions in CRT and ICD patients if medical therapy is both the transmitter and the transmission in one
adjusted quickly on an outpatient basis. icon and connects to a telephonic line. The status
One of the first randomized studies that con- of the implanted device is checked on a daily
firmed the clinical value of CareLink network basis to send notification to the physician through
is the PREFER study that enrolled 980 patients e-mail, text message, phone call, or the Internet.
randomized 2:1. The primary endpoint was to
evaluate remote pacemaker interrogation for the SMARTVIEW (Sorin Group)
earlier diagnosis of serious events compared with It is the most recent RM system, approved by FDA
traditional transtelephonic monitoring and rou- in 2013. The device communicates with an at-
tine in-person evaluation. After 12 months fol- home unit that transmits stored data about device

ERRNVPHGLFRVRUJ
334 L. Cipolletta and J. Ricciotti

function system to the patient and appropriate cli- ity of impedance testing early after implant. The
nicians. Using this information, a cardiologist can sensitivity to predict HF events was at best 42 %
monitor disease progression and determine with a positive predictive valve of 38 %. The
whether the implant needs to be reprogrammed. DOT-HF trial [18] studied the clinical utility of
Installation of the SMARTVIEW remote monitor- the OptiVol to track changes in intrathoracic
ing system at the patients home is assisted by a impedance. All-cause mortality and rehospitaliza-
dedicated help desk provided by Sorin. tions did not change from control to
impedance-monitored therapy. The OptiVol is
being used to provide physicians with wireless
Clinical Utility alerts of threshold deviations for worsening car-
diac status in the OptiLink-HF trial [19]. To
Early AF Episodes Detection increase the predictive ability of a diagnostic
Rhythm disturbances can readily be identified in all algorithm, Whellan et al. in the PARTNERS-HF
of these devices allowing early detection of the trial [20] combined more device variables.
developmentofatrialfibrillation.Arrhythmia-related Utilizing data from an independent dataset, the
severe adverse events, caused by long-lasting unde- authors identified a fluid index >100 days or
tected atrial fibrillation, may be avoided by the any two of the following as criterion: (1) long
early detection of atrial fibrillation occurrence as atrial fibrillation duration, (2) rapid ventricular
soon as arrhythmia has been stored in the device rate during atrial fibrillation, (3) a high (60) fluid
memory because of the prompt clinical reaction of index, (4) decreased patient activity, (5) high
the physician. That is particularly meaningful in night heart rate, (6) low heart rate variability, (7)
asymptomatic patients, because this permits timely low CRT pacing, or (8) ICD shocks. Patients with
decisions concerning antithrombotic or anticoagu- positive device diagnostics had a hazard ratio of
lant therapy [22]. Remote monitoring has been also 5.5 (95 % CI 3.48.8; P<0.0001) of HF hospital-
associated with a reduced hospitalization rate for ization with pulmonary congestion within a
atrial fibrillation and a potential lower risk for month. More recently, Landolina et al. [10] in the
stroke [12]. EVOLVO study randomized 200 patients with a
Medtronic wireless ICD/CRT-D along with the
Heart Failure Monitoring CareLink network; half had remote transmission
The majority of these patients suffer from con- on, while the rest had remote transmission off.
comitant heart disease, reduced left ventricular In the control group, audible alerts were turned on
ejection fraction, and symptomatic heart failure. with no transmission of data, while in the inter-
Thoracic impedance can be measured with these vention group, all alerts were transmitted with no
implanted devices and has been considered as a audible alerts. The primary endpoint was emer-
surrogate marker of pulmonary congestion. When gency department or urgent in-office visits, and
a current is passed between the intracardiac lead the secondary endpoint was the number of visits
and the generator, the impedance to the conduc- related to worsening HF and arrhythmias or ICD-
tion of this current is inversely related to the fluid related events. Patients in the remote monitoring
content of the lungs. In a validation study, Yu et al. arm had significantly less events (0.59 vs. 0.93
[6] found that the impedance started to decrease events per year; incident rate ratio 0.65; 95 % CI
15 days prior to worsening heart failure symp- 0.490.88; P = 0.005). As expected for the directed
toms. In a more recent study of HF patients, intervention, the median time from the alert to
Abraham et al. showed that the sensitivity of ICD data review was 24.8 days for the standard
intrathoracic impedance monitoring was far supe- care arm compared with 1.4 days in the remote
rior to daily weight monitoring for predicting monitoring arm. The audible alerts from the ICD/
worsening HF events [16]. On the contrary, the CRT-D devices did not make much of a differ-
SENSE-HF trial failed to show utility of the same ence. In fact, also prior studies have indicated that
technology [17] and also revealed the unreliabil- many alerts go unnoticed by patients [21].

ERRNVPHGLFRVRUJ
28 The Role of Home Monitoring 335

PVC, NSVT, and Slow VT Episode the management of the huge workload that
Monitoring manufacturers advisory causes. The findings
Early detection of an high burden of PVC or in remote monitoring systems can be used to
NSVT/slow VT in CRT patients with heart fail- avoid clinical problems, but in their absence,
ure allows for early treatment, pharmacological nothing can be done [6, 7].
or ablative, of these conditions, and there is evi-
dence that hospitalization rates can be reduced, Early Identication of Device
as the biventricular pacing reduction and worsen- Troubleshooting
ing heart failure could be prevented. Oversensing, electromagnetic interferences, loss
of capture, undersensing, pacemaker-mediated
VT/VF Therapy tachycardia, and prompt starting of therapeutic
Remote monitoring permits to verify earlier measures are used for the early identification of
device therapies and decide a strategy device troubleshooting [23].
personalized for each patient. If the patient had
only an appropriate shock, he will be reassured Improving of Quality of Life
and hospital admission is avoided. In case of 2 While ICDs are highly effective in lowering the
shocks, the RM is useful to guarantee ER admis- rate of arrhythmic deaths in high-risk patients,
sion. RM has been also associated with less the delivery of shocks, especially when repeti-
appropriate and inappropriate ICD shocks [12] tive, remains a major cause of discomfort, anxi-
explained by the early suppression of inappropri- ety, depression, and poor quality of life. RM
ate therapies prompted by recurrent supraven- makes possible the very early detection of causes
tricular tachyarrhythmias or by oversensing and of appropriate and inappropriate ICD interven-
prevention of appropriate therapies by the early tions and rapid implementation of preventive
introduction of antiarrhythmic measures. measures. This may confer important advantages
for the quality of life of patient and device lon-
Monitoring of Lead Performance gevity [24].
Five-year failure rates have been reported to be
between 2 and 15 % [13], and lately problems
with high-profile ICD lead performance Infrastructure and Organization
(Medtronic Sprint Fidelis and St. Jude Medical of Remote Monitoring
Riata) have concerned the medical world [14].
For that purpose, some remote monitoring sys- Two-thirds of the centers in Europe do not have a
tems can report daily lead integrity measure- specific unit for RM, and the RM program is a
ments of more than 90 % of the time in most of part of their traditional device follow-up clinics.
cases [15]. However, lead alert system and The organization for RM in Europe is based
CareLink helped to identify the major defibril- primarily on nursing with easy communication
lator lead problems experienced with with the responsible physician for medical deci-
Medtronic Sprint Fidelis lead [3]. sions. Many centers, however, conduct only spo-
Unfortunately, contrary to other reports, not radic reviews or have no specific protocol, so the
always finding a lead failure with RM is pos- risk exists that some events may be undetected
sible. Several case reports warn that some- and that the opportunity to adjust therapy may
times the Riata leads can fail to deliver therapy not be undertaken [25].
although normal electrical parameters have
been recorded during in-office visits [4].
However, in the report of Hauser et al. [5], References
87 % of the leads analyzed, in a late stage of
disintegration, had shown electrical abnormal- 1. Wilkoff BL et al (2009) Transvenous lead extraction:
Heart Rhythm Society expert consensus on facilities,
ities, which may have been detected by remote
training, indications, and patient management. Heart
monitoring. Remote monitoring offers help for Rhythm 6(7):10851104

ERRNVPHGLFRVRUJ
336 L. Cipolletta and J. Ricciotti

2. Dubner S, Auricchio A, Steinberg JS, Vardas P, Stone P, able cardioverter defibrillator patients in clinical prac-
Brugada J et al (2012) ISHNE/EHRA expert consensus tice: impact on medical management and health-care
on remote monitoring of cardiovascular implantable resource utilization. Europace 10(2):164170
electronic devices (CIEDs). Europace 14:278293 16. Abraham WT, Compton S, Haas G, Foreman B,
3. Wilkoff BL, Auricchio A, Brugada J, Cowie M, Canby RC, Rishel R et al (2011) Intrathoracic imped-
Ellenbogen KA, Gillis AM et al (2008) HRS/EHRA ance vs daily weight monitoring for predicting wors-
expert consensus on the monitoring of cardiovascular ening heart failure events: results of the Fluid
implantable electronic devices (CIEDs): description Accumulation Status Trial (FAST). Congest Heart
of techniques, indications, personnel, frequency and Fail 17(2):5155
ethical considerations. Heart Rhythm 5:907925 17. Conraads VM, Tavazzi L, Santini M, Oliva F, Gerritse
4. Krebsbach A, Alhumaid F, Henrikson CA, Calkins H, B, Yu CM et al (2011) Sensitivity and positive predic-
Berger RD, Cheng A (2011) Premature failure of a tive value of implantable intrathoracic impedance
Riata defibrillator lead without impedance change or monitoring as a predictor of heart failure hospitaliza-
inappropriate sensing: a case report and review of the tions: the SENSE-HF trial. Eur Heart J 32(18):
literature. J Cardiovasc Electrophysiol 22:10701072 22662273
5. Hauser RG, McGriff D, Retel LK (2012) Riata 18. van Veldhuisen DJ, Braunschweig F, Conraads V,
implantable cardioverter-defibrillator lead failure: Ford I, Cowie MR, Jondeau G et al.; DOT-HF
analysis of explanted leads with a unique insulation Investigators (2011) Intrathoracic impedance moni-
defect. Heart Rhythm 9:742749 toring, audible patient alerts, and outcome in patients
6. Yu CM, Wang L, Chau E, Chan RH, Kong SL, Tang with heart failure. Circulation 124(16):17191726
MO et al (2005) Intrathoracic impedance monitoring 19. Brachmann J, Bhm M, Rybak K, Klein G, Butter C,
in patients with heart failure: correlation with fluid Klemm H et al (2011) Fluid status monitoring with a
status and feasibility of early warning preceding hos- wireless network to reduce cardiovascular-related hos-
pitalization. Circulation 112(6):841848 pitalizations and mortality in heart failure: rationale
7. Crossley GH, Chen J, Choucair W et al (2009) Clinical and design of the OptiLink HF Study (Optimization of
benefits of remote versus transtelephonic monitor- Heart Failure Management using OptiVol Fluid Status
ing of implanted pacemakers. J Am Coll Cardiol Monitoring and CareLink). Eur J Heart Fail
54(22):20122019. doi:10.1016/j.jacc.2009.10.001 13(7):796804
8. Varma N, Epstein A, Irimpen A, Schweikert R, Shah 20. Whellan DJ, Ousdigian KT, Al-Khatib SM, Pu W,
J, Love CJ, TRUST Investigators (2010) Efficacy and Sarkar S, Porter CB et al.; PARTNERS Study
safety of automatic remote monitoring for ICD fol- Investigators (2010) Combined heart failure device
low-up: the TRUST trial. Circulation 122:325332 diagnostics identify patients at higher risk of subse-
9. Crossley GH, Boyle A, Vitense H, Chang Y, Mead quent heart failure hospitalizations: results from
RH, CONNECT Investigators (2011) Clinical PARTNERS HF (Program to Access and Review
Evaluation of Remote Notification to Reduce Time to Trending Information and Evaluate Correlation to
Clinical Decision (CONNECT) Trial: the value of Symptoms in Patients With Heart Failure) study. J Am
wireless remote monitoring with automatic clinician Coll Cardiol 55(17):18031810
alerts. J Am Coll Cardiol 57:11811189 21. Simons EC, Feigenblum DY, Nemirovsky D, Simons
10. Landolina M, Perego GB, Lunati M, Curnis A, GR (2009) Alert tones are frequently inaudible among
Guenzati G, Vicentini A et al (2012) Remote monitor- patients with implantable cardioverter-defibrillators.
ing reduces healthcare utilization and improves quality Pacing Clin Electrophysiol 32(10):12721275
of care in heart failure patients with implantable defi- 22. Healey JS, Connolly SJ, Gold MR, IsraelCW, Van
brillators: the EVOLVO (Evolution of Management Gelder IC Capucci A et al.; for the ASSERT
Strategies of Heart Failure Patients with Implantable Investigators (2012) Subclinical atrial fibrillation and
Defibrillators) Study. Circulation 125:29852992 the risk of stroke. N Engl J Med 366:120129
11. Ricci RP, Morichelli L, Santini M (2009) Remote con- 23. Guedon-Moreau L, Lacroix D, Sadoul N, Clementy J,
trol of implanted devices through Home Monitoring Kouakam C, Hermida JS et al (2013) A randomized study
technology improves detection and clinical manage- of remote follow-up of implantable cardioverter defibril-
ment of atrial fibrillation. Europace 11:5461 lators: safety and efficacy report of the ECOST trial. Eur
12. Mabo P, Victor F, Bazin P, Ahres S, Babuty D, Da Heart J 34(8):605614. doi:10.1093/eurheartj/ehs425
Costa A et al (2012) A randomized trial of long-term 24. Schron EB, Exner DV, Yao Q, Yao Q, Jenkins LS,
remote monitoring of pacemaker recipients (The Steinberg JS, Cook JR, Kutalek SP, Friedman PL,
COMPAS trial). Eur Heart J 33:11051111 Bubien RS, Page RL, Powell J (2002) Quality of life
13. ECkstein J, Koller MT, Zabel M, Kalusche D, Schaer in the antiarrhythmics versus implantable defibrilla-
BA, Osswald S et al (2008) Necessity for surgical revi- tors trial: impact of therapy and influence of adverse
sion of defibrillator leads implanted long-term: causes symptoms and defibrillator shocks. Circulation
and management. Circulation 117(21):27272733 105:589594
14. Maisel WH, Kramer DB (2008) Implantable 25. Hernndez-Madrid A, Lewalter T, Proclemer A, Pison
cardioverter-defibrillator lead performance. Circulation L, Lip GY, Blomstrom-Lundqvist C (2014) Remote
117(21):27212723 monitoring of cardiac implantable electronic devices
15. Ricci RP, Morichelli L, Santini M (2008) Home in Europe: results of the European Heart Rhythm
Monitoring remote control of pacemaker and implant- Association survey. Europace 16(1):129132

ERRNVPHGLFRVRUJ
Drug-Related Adverse Events
29
Simone Maffei and Jenny Ricciotti

29.1 Case Report Vital Signs and Physical Examination

Temperature: 36.5 C
A 58-year-old woman presented to the Heart rate: 36 beats per minute
emergency department accompanied by Blood pressure: 150/90 mmHg
her family members for complaints of agi- Respiratory rate: 22 breaths/min; oxygen satu-
tation and dyspnea. She had a history of ration while breathing in ambient air, 97 %
bipolar disorder that required lithium, and General: awake but agitated and disoriented
there were no history of suicidal ideation, with aimless movements
recent medication changes, and improper Head, eyes, ears, nose, and throat: normoce-
medication administration and no family phalic, atraumatic, mucous membranes dehy-
history of sudden cardiac death. There drated, extraocular muscles intact, and pupils
were unknown cardiovascular risk factors equally round and reactive to light and accom-
or allergies. Home medication: only lithium modation bilaterally
(but the patient didnt remember the dosage Cardiovascular: regular bradycardia rhythm,
of the drug). S1 and S2 normal, and no pathological heart
According to the relatives, the patient murmurs
had been suffering from gastroenteritis Lungs: no rales, rhonchi, or wheezes; no alter-
with significant diarrhea few days before. ations in tactile fremitus; normal percussion
Abdomen: overweight, no pulsatile masses,
soft, nondistended/nontender, no hepatospleno-
megaly, slightly sore upon deep palpation in the
lower quadrants, and lively peristalsis
Extremities: no cyanosis or clubbing, no
edema, and dehydrated skin

Routine Laboratory Tests


S. Maffei (*) J. Ricciotti
Clinica di Cardiologia e Aritmologia, Universit Complete blood count: mild neutrophilic leu-
Politecnica delle Marche, kocytosis (WBC 12.160/mmc); hemoglobin
Via Conca, 71, Ancona 60126, Italy 12.4 g/dl
e-mail: maffo-85@hotmail.it; jen.ricciotti@tiscali.it

Springer International Publishing Switzerland 2015 337


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_29

ERRNVPHGLFRVRUJ
338 S. Maffei and J. Ricciotti

Hepatic function (GOT, GPT, -GT, ALP, and In this case, gastroenteritis has led to a state of
total, direct, and indirect bilirubin): normal dehydration which resulted in the deterioration of
Impaired renal function (creatinine 2.10 mg/ dl) renal function and an increase of lithium serum
Electrolytes: Na+ 133 mEq/l and K+ 3.4 mEq/l levels.
Myocardial necrosis markers: CK-MB 2.3 ng/ The patient was monitored and a fluid infusion
ml and TnI 0.30 ng/ml was set up with saline to restore an adequate
Lithium level: 3.7 seq/l blood volume and hydration state.
After a few hours, there was a gradual
A routine 12-lead resting ECG was performed reduction in blood creatinine and lithium levels
(Fig. 29.1ad). with the regression of the electrocardiographic
ECG showed marked sinus bradycardia (heart changes shown above (Fig. 29.2ad).
rate was 36 b/min), normal atrioventricular and The patient was discharged completely
intraventricular conduction, and diffuse altera- asymptomatic the next day, with a normal elec-
tions of ventricular repolarization with negative trocardiogram and with good renal function. We
T waves from V1 to V5; QTc = 435 msec. advised a further psychiatric check for a possible
optimization of the specific medical therapy.

What Are the Possible Causes


of Agitation and Dyspnea in This Lithium
Patient? Why Are There Sinus
Bradycardia and Diffuse Alterations Lithium salts are still used in the prophylaxis and
of Ventricular Repolarization? treatment of bipolar syndrome (manicdepres-
sive) with a dose ranging from 1 to 1.5 g per day
in the attack phase to 300400 mg daily as a
Acute coronary syndrome (ACS) maintenance dose. Lithium is still considered the
Electrolyte disorders (hypokalemia, drug of choice in the treatment of patients with
hyponatremia) affectivity bipolar, that is, for people who expe-
Drug intoxication rience alternate phases and periods of sadness
Exacerbation of renal failure and hopelessness (depression) with periods of
excitement and euphoria (mania).
Lithium salts have a narrow therapeutic index,
ACS is very unlikely because the patient and their use should always be subordinate to the
didnt present chest pain, there was a little move- possibility of periodic blood tests.
ment of cardiovascular enzymes (probably sec- The daily dose of lithium should be individu-
ondary to worsening of renal function), and an alized on the basis of the drug plasma levels. It is
echocardiogram showed no alterations in the important to determine the plasma concentration
kinetics. Blood tests showed slight alterations of of lithium (lithium levels) once per week for the
sodium and potassium that did not cause any first 2 months, once a month for the next 68
electrocardiogram abnormalities. months, and once every 23 months during the
Instead, there was a marked alteration of lith- maintenance phase of therapy. Lithium levels
ium levels (normal values, 0.61.2 mEq/L for should be checked after each change of the dose.
chronic bipolar disorder; 1.01.5 mEq/L in the Clinical signs of toxicity manifest themselves
case of acute manic episode) that could explain at concentrations of lithium levels equal to or
both agitation state and ECG changes. higher than 1.5 mEq/l. The majority of patients
Retention of lithium is favored by all those reach levels of toxicity when intercurrent diseases
conditions that may be associated with intravas- (diarrhea, vomiting, heart failure, renal failure) or
cular volume depletion. drug interactions (NSAIDs, ACE-I) are present.

ERRNVPHGLFRVRUJ
29 Drug-Related Adverse Events 339

Side Effects function abnormalities to underlie these side


effects have been postulated. However, most pre-
Lithium can cause a number of side effects in vious cases of conduction abnormalities occurred
various districts of the organism (see Table 29.1). in the setting of chronic lithium therapy or in
It is also known that lithium can cause various patients receiving toxic levels of lithium [1].
cardiac disorders, including conduction abnor- Although most side effects occur at the supra-
malities. The mechanisms underlying these con- therapeutic levels, there are several case reports
duction defects are not clearly understood. A of conduction abnormalities induced by lithium
wide range of lithium toxicities have been in patients at levels within the therapeutic range.
observed, from severe bradycardia to arrhyth- This suggests a wide range of individual respon-
mias. Direct effects on the sinus node and thyroid siveness to lithium [1].

Fig. 29.1 (ad) Image 1 ECG: sinus bradycardia, heart rate of 36 b/min, and negative T waves from V1 to V5

ERRNVPHGLFRVRUJ
340 S. Maffei and J. Ricciotti

Fig. 29.1 (continued)

ERRNVPHGLFRVRUJ
29 Drug-Related Adverse Events 341

a b

Fig. 29.2 (ad) Image 2 ECG: reduction of previous electrocardiographic changes

ERRNVPHGLFRVRUJ
342 S. Maffei and J. Ricciotti

Fig. 29.2 (continued)

Table 29.1 Lithium side effects


The major cardiovascular side effects of Side effects
lithium include: Nervous system disorders
Unmasking of Brugada syndrome Absences, seizures, drowsiness, dizziness, fatigue,
lethargy, psychomotor delays, confusion, restlessness,
Sinus node dysfunction
stupor, coma, tremors (twitching, clonic movements
Atrioventricular (AV) block of the legs), ataxia, dry mouth
Various arrhythmias (torsades de pointes, Nephrology
ventricular tachycardia, ventricular Decreased ability to concentrate urine, renal failure
fibrillation) (rarely), diabetes insipidus
Asymptomatic electrocardiographic Endocrinological
changes (changes in T waves character- Hypothyroidism, hyperparathyroidism, weight gain
ized by flattening, isoelectrical changes, (insulin-like effect)
or inversion) Sense organs
Prolongation of the QT interval Taste changes, alteration of thirst
Teratogenic
Likely association with Ebsteins anomaly (avoid use
during pregnancy)

ERRNVPHGLFRVRUJ
29 Drug-Related Adverse Events 343

29.2 Drug-Related ECG Adverse Table 29.2 Sodium channel-blocking drugs


Events Cardiovascular drugs: class Ia antiarrhythmic drugs,
class Ib antiarrhythmic drugs, class Ic antiarrhythmic
Several drugs and toxins can act on the cardio- drugs, verapamil, diltiazem, propanolol
vascular system, either at therapeutic level or sec- Psychiatric drugs: carbamazepine, cyclic
antidepressants, antipsychotics (loxapine),
ondary to overdose and poisoning. They may neuroleptics (thioridazine, mesoridazine),
cause ECG abnormalities, heart rhythm distur- citalopram
bances, depression of myocardial contractility, Antihistamines (diphenhydramine)
and vasodilation. Illicit drugs: cocaine
There are two main mechanisms underlying Other drugs: amantadine, chloroquine,
the adverse cardiovascular effects of pharmaco- hydroxychloroquine, orphenadrine, propoxyphene
logical and toxin agents: Toxins: quinine, saxitoxin, tetrodotoxin

A direct action on ion channels: sodium


(Na+) channel blockers, outward potassium Class Ia antiarrhythmic drugs are quinidine,
(K+) channel blockers, calcium channel procainamide, and disopyramide. They are potent
blockers (CCB), sodiumpotassium adenos- inhibitors of sodium channels and additionally
ine triphosphatase (Na+/K+ ATPase) blockers determine a block of potassium channels.
Effects on the autonomic nervous system: At high plasma concentrations, conduction
vagal and sympathetic action and beta- through the AV junction is slowed and the auto-
adrenergic block maticity and refractoriness of HisPurkinje sys-
tem are reduced; QT prolongation is the first sign
Drugs cause ECG changes also indirectly, in ECG and is related to the increased duration of
determining electrolyte and acidbase imbal- the action potential in ventricular muscle fibers;
ance. It must be remembered that many drugs with increasing doses, there is a progressive flar-
produce effects resulting from the combination ing of the QRS complex.
of several mechanisms. Sinus node dysfunction, varying degrees of
AV block, ventricular arrhythmias, and torsades
de pointes (TdP) can also occur; also during the
Drugs and Toxins Acting on Ion first administrations, quinidine can cause TdP, in
Channels a dose-independent manner.
Quinidine and disopyramide at high doses
Sodium Channel-Blocking Agents have a potent negative inotropic effect, resulting
(Table 29.2) in hypotension and shock. Toxic levels of quini-
During phase 0 of the action potential, the open- dine are responsible for a syndrome known as
ing of fast sodium channels causes Na+ influx and cinchonism, with tinnitus, blurred vision, photo-
depolarization of cardiac cell membrane; this led phobia, confusion, delirium, and abdominal pain.
to contraction of ventricular myocytes, which is Procainamide toxicity is similar to that of
expressed by QRS complex in ECG. quinidine but with less negative inotropic effects
Inhibitors of fast Na+ channels slow the rise of [35].
the depolarization in phase 0; in case of overdose, Class Ib antiarrhythmic drugs have a high
there is a progressive widening of the QRS com- affinity for sodium channels that are in the inacti-
plex, with the risk of heart block and ventricular vated state, with a rapid binding kinetic onoff.
arrhythmias. The depression of myocardial con- They reduce the duration of the ventricular action
tractility is responsible for hypotension induced potential but do not act on potassium channels
by these agents [2]. and therefore do not affect the QT interval [6].
Class I antiarrhythmic drugs belong to this Since lidocaine easily crosses the bloodbrain
category. barrier, neurological effects are the first to appear

ERRNVPHGLFRVRUJ
344 S. Maffei and J. Ricciotti

in the case of toxicity, with light-headedness, Table 29.3 Calcium channel blockers
seizures, confusion, hallucinations, and comatose Dihydropyridines 1st generation:
state. At cardiac level, there is a slowing of con- nicardipine, nifedipine
duction and reduction in myocardial contractility, 2nd generation:
felodipine, isradipine,
and with very high doses, asystole, advanced nimodipine
heart block, and refractory hypotension may 3rd generation:
occur [5]. amlodipine, nitrendipine
Class Ic antiarrhythmic drugs (flecainide, 4th generation:
lercanidipine, lacidipine
propafenone, encainide) bind to sodium channels
Phenylalkylamine Verapamil
in the activated state. At high plasma concentra-
Benzothiazepine Diltiazem
tions, these agents have a potent negative inotro- Diarylaminopropylamine Bepridil
pic effect; the PR interval and QRS duration are ether
prolonged, and QT interval generally is not very
affected.
Propafenone unlike the other drugs in this
class also possesses subclinical beta-blocker generation of action potential in the pacemaker
properties [5]. cells and for the contraction of smooth muscle
Cyclic antidepressants block sodium channels cells of the peripheral vessel walls.
in a manner similar to quinidine, slowing the rise Verapamil and diltiazem have a major affin-
of phase 0 of the action potential; in case of toxic- ity for cardiac calcium channels and in toxic
ity, an increase in duration of QRS and QT inter- states may induce sinus node and AV node dys-
val can occur, with a depression of myocardial function and depression of myocardial contrac-
contractility [6]. tility. Bradycardia, various degrees of AV
Block of the reuptake of catecholamines in the blocks, and accelerated junctional rhythms are
presynaptic nerve terminal and the antagonist common, and in most severe cases, asystole is a
effect on alpha-adrenergic receptors can cause possible complication; enlargement of QRS
hypotension. A study [7] suggested that an R complex may be an expression of a ventricular
wave in aVR 3 mm and a rightward terminal escape rhythm or a consequence of sodium
vector are predictive factors of cardiotoxicity channel block with a delay in the depolarization
from cyclic antidepressants and correlated with phase 0.
the risk of rhythm disturbances and seizures. At the systemic level, common manifestations
The initial management of all drug intoxica- are hyperglycemia and metabolic acidosis, nau-
tion is based on circulatory and airway support. sea and vomiting, lethargy, confusion, dizziness,
The mainstay treatment of sodium channel and slurred speech.
blocker toxicity is the intravenous administration Dihydropyridines have a major affinity for
of sodium bicarbonate that increases the extracel- vascular smooth muscle cells and, in overdoses,
lular sodium concentration and blood pH. may produce vasodilation, with severe hypoten-
Lidocaine is indicated in case of refractory sion and reflex tachycardia with palpitations and
ventricular arrhythmias. Gastric lavage and acti- flushing [5, 8].
vated charcoal may be effective in limiting Specific treatment of CCB intoxication
absorption if administrated early after drug inges- involves intravenous administration of calcium
tion (12 h). salts (calcium gluconate), the use of glucagon
that promotes calcium entry into cells, and the
Calcium Channel Blockers (CCB) use of vasopressors and inotropes in case of
CCBs block (see Table 29.3) the transport of cal- hypotension and shock; also insulin therapy may
cium ions into the cell from the extracellular be indicated, with a hyperinsulinemia/euglycemia
space, inhibiting voltage-sensitive L-type Ca2+ protocol because it can improve inotropy and
channels; these channels are responsible for the increase peripheral vascular resistance.

ERRNVPHGLFRVRUJ
29 Drug-Related Adverse Events 345

Potassium Channel Blockers phenomena with the risk of polymorphic ventric-


Important antiarrhythmic drugs, as amiodarone, ular tachycardia and TdP [10, 11].
dofetilide, sotalol, dronedarone, etc. (see In a patient with suspected QT prolongation
Table 29.4), belong to this class. Drugs of this induced by drugs, first, the possible cause must
group block the outward flow of potassium from be withdrawn, and, second, any associated condi-
the cell into the extracellular space. The block in tions such as the presence of electrolyte distur-
the output current of potassium slows the repolar- bances should be treated.
ization phase of the myocardium and prolongs The intravenous administration of magnesium
the duration of action potential [9]. sulfate is able to reduce the occurrence of arrhyth-
The most common ECG manifestation is the mias in the case of long QT; in the presence of
QT interval prolongation (QTc interval greater episodes of TdP in patients who do not respond
than 450 msec in men and 470 msec in women). to such treatment, an overdrive pacing may be
The delay of repolarization resulted in a lower indicated [5].
potential difference at the two sides of the cardiac
cell membrane; this can induce early after- SodiumPotassium ATPase Blockers
depolarization, triggered activity, and reentry Cardiac glycosides inhibit the sodiumpotas-
sium ATPase pump on the cell membrane; as a
result, there is an increase in intracellular sodium
and a secondary activation of the Na+/Ca++
Table 29.4 Potassium channel blocking drugs exchanger. This led to increased levels of intra-
Cardiovascular drugs: cellular calcium, with stimulation of contractile
Class IA antidysrhythmics: disopyramide, quinidine, fibers in myocytes and a positive inotropic effect;
procainamide they also increase vagal tone and slow AV con-
Class IC antidysrhythmics: encainide, flecainide, duction [12].
moricizine, propafenone
Class III antidysrhythmics: amiodarone, dofetilide, At therapeutic doses, digitalis induces a series
ibutilide, sotalol of ECG signs known as digitalis effect: flat or
Antianginal/vasodilators: bepridil, prenylamine, inverted T waves, ST segment depression, PR
terodiline prolongation, and shortening of the QT interval
Psychiatric drugs: with increased U-wave amplitude [13, 14].
Antipsychotics: chlorpromazine, droperidol,
haloperidol, mesoridazine, pimozide, quetiapine, In cardiac glycoside toxicity, electrocardio-
risperidone, thioridazine, ziprasidone graphic abnormalities are the result of increased
Cyclic antidepressants: amitriptyline, amoxapine, automaticity and slow conduction in the AV
desipramine, doxepin, imipramine, nortriptyline, node.
maprotiline
Other antidepressants: citalopram, venlafaxine Different arrhythmias can be produced; the
Phenothiazines most frequent manifestations are atrial fibrilla-
Antimicrobials: tion with slow ventricular response, paroxysmal
Fluoroquinolones: ciprofloxacin, gatifloxacin, atrial tachycardia with block, accelerated junc-
levofloxacin, moxifloxacin, sparfloxacin tional rhythm, ventricular ectopic complexes
Macrolides: clarithromycin, erythromycin
Others: pentamidine, chloroquine, halofantrine, (bigeminal and trigeminal), and bidirectional
hydroxychloroquine ventricular tachycardia.
Antihistamines: astemizole, diphenhydramine, The treatment of severe digoxin toxicity
loratadine, terfenadine, hydroxyzine consists in intravenous administration of
Serotonin 5-HT4 receptor agonist: cisapride digoxin-specific Fab fragments; they are able
Other drugs: to restore the activity of the sodiumpotassium
Arsenic trioxide
Probucol
ATPase pump, by displacing digoxin away
Synthetic opioids: levomethadyl from its receptors, leading to a rapid disap-
Opium alkaloids: papaverine pearance of signs and symptoms of intoxica-
Toxins: quinine, organophosphates tion [5].

ERRNVPHGLFRVRUJ
346 S. Maffei and J. Ricciotti

Drugs and Toxins Acting Overdose with agents with intrinsic sympa-
on the Autonomic Nervous System thomimetic activity can be expressed with hypo-
tension and tachycardia.
Beta-Blocker Antagonists Other manifestations of beta-blocker poison-
Pharmacodynamic properties of beta-blockers ing are bronchospasm, lethargy, coma, and
differ in relation to their affinity for beta- hypoglycemia.
adrenergic receptor (see Table 29.5): block of 1 In the management of patients with beta-
receptors has negative chronotropic and inotropic blocker intoxication, glucagon via IV infusion is
effects and reduces renin secretion, while inhibi- used that induces an increase in cAMP levels and
tion of 2 receptors induces relaxation of smooth an influx of calcium in the intracellular space,
muscle cells in the vessels, bronchial apparatus, secondary to the stimulation of a membrane
and gastrointestinal tract and influences glucose receptor in a different site of action compared to
metabolism (inhibition of glycogenolysis and that of beta-blockers. Other therapeutic interven-
gluconeogenesis) [15]. tions are the infusion of atropine, calcium, and
The most common manifestations of beta- catecholamines [5].
blocker toxicity include cardiovascular system
symptoms, with disorders of the conduction sys- Other Drugs Acting on the Sympathetic
tem (sinus node dysfunction, various degrees of Nervous System (Table 29.6)
AV blocks, intraventricular conduction delay) Drugs acting on alpha-adrenergic receptors are
and hypotension, secondary to altered myocar- classified as central and peripheral, with agonist
dial contractility and block of peripheral and antagonist properties.
receptors. Alpha-2 central agonists (clonidine,
Sotalol is the only beta-blocker with class III -methyldopa) and peripheral alpha-1 blockers
antiarrhythmic properties, via blockade of potas- (doxazosin, prazosin, terazosin) are principally
sium channels, causing an increase in the dura- used as antihypertensive drugs; overdose states
tion of repolarization phase, with QTc can be associated with rhythm disturbances
prolongation on ECG and risk of ventricular (sinus, atrial, junctional, and ventricular bradyar-
tachyarrhythmias and TdP. Also propanolol and rhythmias; first-degree AV block; ventricular
acebutolol can induce prolongation of the QTc tachyarrhythmias), severe hypotension, and car-
interval [16, 17]. diac failure.

Table 29.5 Selectivity of beta blockers


Drug 1-selectivity ISA -Adrenergic blockade Half-life (h)
Propanolol 35
Nadolol 1020
Timolol 35
Pindolol ++ 34
Labetalol + + 46
Carvedilol + 710
Metoprolol ++ 34
Atenolol ++ 58
Esmolol ++ 0.13
Acebutolol + + 812
Bisoprolol ++ 912
Nebivolol ++ 1030
Abbreviations: ISA intrinsic sympathomimetic activity

ERRNVPHGLFRVRUJ
29 Drug-Related Adverse Events 347

Table 29.6 Sympathomimetic drugs and toxins At high dosage, they commonly induce sinus
Drugs Beta-adrenergic agonists (albuterol, tachycardia, but in case of poisoning, we should
dobutamine, epinephrine, isoproterenol, assess for serious supraventricular and ventricu-
norepinephrine, ritodrine, terbutaline) lar arrhythmias.
Theophylline
Ergot alkaloids Clinical manifestations of anticholinergic
Phenylpropanolamine and other over-the- toxic syndrome are flushing, dry skin and mucous
counter sympathomimetics (decongestants membranes, mydriasis, altered mental status,
containing phenylephrine, sinus tachycardia with hypertension, and urinary
pseudoephedrine, ephedrine)
Monoamine oxidase inhibitors retention [19].
Caffeine
Chloral hydrate (sedative and hypnotic Cholinomimetic Toxicity
drug) The most common cause of toxicity from drugs
Illicit Cocaine
and toxins with cholinomimetic properties is
drugs Amphetamines
Phencyclidine the intoxication with organophosphate and
Delta-tetrahydrocannabinol (cannabis) carbamate pesticides. Organophosphorus pesti-
Psilocybin and other hallucinogens cides inhibit esterase enzymes (acetylcho-
Lysergic acid diethylamide
linesterase) with consequent accumulation of
Toxins Ethanol
acetylcholine and stimulation of acetylcholine
Hydrocarbon solvents (e.g., toluene,
benzene, chloroform, etc.) receptors in synapses of the autonomic nervous
Freon (and other fluorocarbon aerosols) system, central nervous system, and neuromus-
cular junctions.
In the early phase, tachycardia is present sec-
A lot of drugs and toxins can cause signs of ondary to the stimulation of nicotinic receptors,
hyperactivity of the sympathetic nervous system followed by the development of bradycardia by
both centrally and peripherally, with positive ino- the stimulation of muscarinic receptors; subse-
tropic and chronotropic stimulation (Table 29.6). quently, even after a few days, in cases of severe
At ECG, sinus tachycardia is very common; at intoxication, AV blocks and asystole may appear
high dosage, supraventricular and ventricular [5], with QT prolongation, ventricular tachycar-
arrhythmias may also occur. Cocaine can induce dia, and TdP.
vasospasm and platelet activation, causing myo- Cases of myocardial infarction have been
cardial ischemia and infarction also in young reported, secondary to parasympathetic hyperac-
subjects with no other cardiovascular risk factors tivity with coronary vasospasm and direct dam-
[18, 19]. age of pesticides on the myocardium [19].
Peripheral alpha-1 agonist drugs (imidazoline
derivatives) are used as nasal decongestants; at
References
high doses by topical administration, they can
induce systemic toxicity, hypertension, and 1. Sabharwal MS, Annapureddy N, Agarwal SK,
tachycardia. Ammakkanavar N, Kanakadandi V, Nadkarni GN
(2013) Severe bradycardia caused by a single dose of
Anticholinergic Toxicity lithium. Intern Med 52:767769
2. Roden DM (2001) Antiarrhythmic drugs. In: Hardman
A lot of drugs, toxins, and other natural sub- JG, Limbird LE (eds) The pharmacologic basis of
stances have anticholinergic properties: antihista- therapeutics, 10th edn. McGraw-Hill, New York,
mines, tricyclic antidepressants, antipsychotics pp 933970
(e.g., phenothiazines, clozapine, olanzapine), 3. Kim SY, Benowitz NL (1990) Poisoning due to class
Ia antiarrhythmic drugs. Drug Saf 5:393420
atropine, scopolamine, toxic plants from 4. Mathis AS, Gandhi AJ (2002) Serum quinidine con-
Solanaceae family containing belladonna alka- centration and effect on QT dispersion and interval.
loids, and toxic mushrooms (Amanita muscaria). Ann Pharmacother 36:11561161

ERRNVPHGLFRVRUJ
348 S. Maffei and J. Ricciotti

5. Jeremias A et al (2010) Cardiac intensive care, 2nd 14. Ma G, Brady WJ, Pollack M et al (2001) Electro-
edn. Elsevier Saunders, Philadelphia cardiographic manifestations: digitalis toxicity. J Emerg
6. Lewin NA, Nelson LS (2006) Goldfranks toxicologic Med 20(2):145152
emergencies, 8th edn. McGraw-Hill, New York 15. Bird SB (2007) Beta-adrenergic antagonists. In:
7. Liebelt EL, Francis PD, Woolf AD (1995) ECG lead Shannon MW et al (eds) Haddad and Winchesters
aVR versus QRS interval in predicting seizures and clinical management of poisoning and drug overdose,
arrhythmias in acute tricyclic antidepressant toxicity. 4th edn. Saunders Elsevier, Philadelphia, pp 975982.
Ann Emerg Med 26(2):195201, ISSN 0196-0644 ISBN 978-0-7216-0693-4
8. Pearigen PD, Benowitz NL (1991) Poisoning due to 16. Anderson AC (2008) Management of beta-adrenergic
calcium antagonists: experience with verapamil, blocker poisoning. Clin Pediatr Emerg Med 9:416
diltiazem and nifedipine. Drug Saf 6:408430 17. Delk C, Holstege CP, Brady WJ (2007) Electro-
9. Yap YG, Camm AJ (2003) Drug induced QT prolonga- cardiographic abnormalities associated with poison-
tion and torsades de pointes. Heart 89(11):13631372 ing. Am J Emerg Med 25(6):672687, ISSN
10. Sides GD (2002) QT interval prolongation as a bio- 0735-6757. Emerg Med 9(1), (March, 2008):416,
marker for torsades de pointes and sudden death in ISSN 1522-8401
drug development. Dis Markers 18(2):5762 18. Vincent GM, Anderson JL, Marshall HW (1983)
11. Nelson LS (2002) Toxicologic myocardial sensitiza- Coronary spasm producing coronary thrombosis and
tion. J Toxicol Clin Toxicol 40(7):867879 myocardial infarction. N Engl J Med 309:220239
12. Holstege CP, Eldridge DL, Rowden AK (2006) ECG 19. Murphy NG, Benowitz NL, Goldschlager N (2007)
manifestations: the poisoned patient. Emerg Med Clin Cardiovascular toxicology. In: Shannon MW et al (eds)
North Am 24(1):159177, ISSN 0733-8627 Haddad and Winchesters clinical management of poi-
13. Irwin J (2003) Intensive care medicine. In: Kirk M, soning and drug overdose, 4th edn. Saunders Elsevier,
Judge B (eds) Digitalis poisoning, 5th edn. Lippincott Philadelphia, pp 133165. ISBN 978-0-7216-0693-4
Williams & Wilkins, Baltimore

ERRNVPHGLFRVRUJ
Difcult Interpretation of ECG:
Small Clues May Make 30
the Difference. The Role
of the P Wave

Alessandro Maolo and Daniele Contadini

30.1 Case Report reduction of intensity at the apex. Mild sys-


tolic crescendo-decrescendo murmur at the
second right intercostal space with no radia-
A 24-year-old woman suspected of sclero- tion, early low-pitched diastolic decrescendo
derma was referred to the clinic of internal murmur, and no pansystolic plateau murmur
medicine for clinical evaluation. attributable to ventricular septal defect. Point
The patient presented with asthenia and of maximal intensity displaced to the center of
mild dyspnea for moderate physical activ- the chest. No hepatojugular reflux and capil-
ity. She reported sleeping with just one pil- lary refill less than 2 s.
low, and she had no episodes of nocturnal Lungs: rales at auscultation at the bases bilat-
dyspnea. She had no syncope or presynco- erally, no rhonchi or wheezes, no egophony,
pal symptoms. no alterations in tactile fremitus, and normal
percussion sounds
Abdomen: mild overweight, no pulsatile
masses, normal bowel sounds in all four quad-
Physical Examination rants, no high-pitched or tinkling sounds, res-
onant to percussion, soft, nondistended/
General: no fatigue, no acute distress, alert, nontender, no rebound or guarding, no costo-
awake, and oriented; well developed and well vertebral angle tenderness, and no
nourished hepatosplenomegaly
Neck: supple, no jugular venous distention, no Extremities: no cyanosis or clubbing and no
lymphadenopathy, and no carotid bruit peripheral edema
Cardiovascular: regular rate and rhythm, S1
normal, paradoxical splitting of the S2, and
Electrocardiogram
A. Maolo D. Contadini (*)
Clinica di Cardiologia e Aritmologia, Universit A routine ECG was performed and is described
Politecnica delle Marche,
Via Conca 71, Ancona 60126, Italy
as follows (Fig. 30.1a,b):
e-mail: alessandro.maolo@alice.it; daniele. Regular RR interval with a heart rate of
contadini@gmail.com 75 bpm.

Springer International Publishing Switzerland 2015 349


A. Capucci (ed.), Clinical Cases in Cardiology: A Guide to Learning and Practice,
DOI 10.1007/978-3-319-19926-9_30

ERRNVPHGLFRVRUJ
350 A. Maolo and D. Contadini

a b

Fig. 30.1 (a, b) Standard 12-lead ECG at rest. See the chapter for the description

P wave is apparently increased in amplitude part is high and narrow and its followed by a sec-
and duration; however, watching carefully, we ond part that is smaller and larger than the first
can notice that the amplitude of the P wave one. This is a likely sign of biatrial enlargement.
doesnt reach a pathologic value. In fact the max- P wave axis is 90, being negative in aVR and
imum amplitude is 0.20 mV in II lead. Also the aVL leads, biphasic in I lead, and positive in II,
duration is mildly prolonged, with the maximum II, and aVF leads. A negative P wave in aVL is
P wave duration of 120 milliseconds (ms) reached something to think about.
in II lead. P wave morphology can be considered PQ interval is normal (160 ms).
pathological: if we pay attention on the P wave in QRS complex is clearly enlarged (160 ms)
II, III, and aVF leads, we can notice that the first with a normal amplitude. However, it decreases

ERRNVPHGLFRVRUJ
30 Difficult Interpretation of ECG: Small Clues May Make the Difference. The Role of the P Wave 351

progressively from V1 to V6. Looking at precor- underwent a surgical correction when she was a
dial leads, the morphology is typical for right child.
bundle branch block. In the limb leads, the QRS Taking a new look to the previous ECG, we
complex has a strange morphology. A deep S can now relate some findings with the anatomic
wave, typical for RBBB, in I and aVL leads is abnormalities.
missing, and the intraventricular and interven- First of all, the axis of the P wave is not nor-
tricular conduction disturbance seems to be more mal. The notch on the apex of the P wave is not a
complex than in a simple RBBB. Also the rS sign of an atrial enlargement. In fact in young
morphology in aVR lead and the rSR in aVL people, the P wave duration can be normal up to
leads are not typical for RBBB. Axis is 130 ms. The notch is a sign of interatrial conduc-
indeterminable. tion disturbance compatible also with the previ-
Repolarization is not abnormal because this is ous surgical intervention.
secondary to the conduction disturbance. The QT The previously diagnosed RBBB is a left bun-
interval measures 400 ms and the QTc 447 ms. dle branch block. In dextrocardia, the morpho-
logical left bundle branch is placed on the right,
Conclusions and its conduction disturbance could be due to
ECG shows sinus rhythm, normal heart rate, right the interventricular septal defect that is always
and left atrial enlargement, normal atrioventricu- present in tetralogy of Fallot. The other intraven-
lar conduction, right bundle branch block, and tricular and interventricular conduction distur-
secondary repolarizations abnormalities. bances could be linked to the previous surgical
Looking at a previous chest X-ray, something intervention.
unexpected was found (Fig. 30.2). The atypical morphology of QRS complex in
The patient had a situs viscerum inversus with aVR and aVL leads can be related to dextrocardia
dextrocardia (please note the cardiac apex and too. In fact, in RBBB, rSR and qR morphol-
the gastric bubble placed on the right side). ogies in aVR lead and a deep or large S wave in
Moreover, the patient had tetralogy of Fallot and aVL lead are common findings, and in this case,
the morphologies are inverted between the two
leads.
By comparing the previous ECG with the fol-
lowing, it is visible that in the typical RBBB
(ECG from another patient below), the QRS mor-
phologies in aVR and aVL are switched
(Fig. 30.3a,b).
A sign that could suggest the presence of dex-
trocardia is the decreasing amplitude of the QRS
complex from V1 to V6 together with the nega-
tive P wave in aVL. This happens because the
electrodes are placed on the left part on the chest
and the electric signal from the heart is progres-
sively going farther from the precordial leads, in
particular from the left ones.

30.2 The Normal P Wave

P wave comes from the electrocardiogram sign of


Fig. 30.2 Chest X-ray. Please notice the cardiac apex atrial depolarization. The impulse starts from the
and the gastric bubble placed to the right side sinoatrial (SA) node and spreads to the right

ERRNVPHGLFRVRUJ
352 A. Maolo and D. Contadini

a b

Fig. 30.3 (a, b) Standard 12-lead ECG at rest from another patient having a situs solitus and an RBBB. Please compare
with the ECG in Fig. 30.1

atrium first and then through the interatrial sep- cordial leads from V3 to V6, the P wave is gener-
tum to the left atrium. ally positive. Conversely in V1 and V2, normal P
Its normal duration varies from 70 to 140 ms wave is often diphasic and sometimes even
even in normal subjects [1]. A P wave lasting negative.
more than 110 ms can be considered abnormal. Talking about the normal amplitude of the P
The P wave axis is the result of the electrical wave, we have to say that its affected by many
activation of the right atrium (directed anteriorly factors such as the distance of electrodes from the
first and then posteriorly when involving the infe- heart and the level of atrial fibrosis. In the limb
rior wall) and of the left atrium (directed posteri- leads, a P wave inferior or equal to 0.25 mV or
orly). Therefore, on the limb leads, the normal P 25 % of the following R wave is considered as
wave range is 0 to +75. Thats why the normal normal amplitude. In the precordial leads, the
P wave is positive in I and II leads and always normal amplitude is a bit lower and doesnt
negative in aVR lead. Moreover, also in the pre- exceed 0.15 mV [1].

ERRNVPHGLFRVRUJ
30 Difficult Interpretation of ECG: Small Clues May Make the Difference. The Role of the P Wave 353

30.3 Interatrial Conduction Delay ectopic focus can cause P waves of different mor-
phology related to the presence of intra-atrial or
Atrial depolarization begins in the sinus node; interatrial conduction disturbances. Likewise,
from here, the electrical impulse goes through the ectopic P waves originating from different foci
internodal fascicles (fibers that connect sinus can have similar or the same morphology [1, 3].
with the AV node). There are three bundles: the Despite this wide variability, some ectopic
Bachman fascicle (anterior), the Thorel fascicle foci are identifiable with fairly good precision.
(in an intermediate position), and the Wenckebach For example, if the ectopic impulse originates
fascicle (posterior). The last one conducts the from a focus close to the AV node, a negative P
impulse from the right to the left atrium, too. So, wave can be visible in II, III, and aVF leads,
the atria activation begins from the right atrium while it is positive in aVL and aVR leads. On the
and then reaches the left one. The normal dura- contrary, if the ectopic focus is located in the left
tion of P wave is from 50 to 80 ms. A P wave atrium, the P wave is negative in aVL and posi-
duration superior to 110 ms is pathological and tive in aVR (similarly to the P wave in dextrocar-
related to interatrial conduction delay. This con- dia), while in the inferior leads, it can be positive
dition is caused by a complete or partial interrup- or negative depending on the part of the left
tion of the Bachman fascicle or other connecting atrium in which the focus is placed.
atria fibers. Usually the interatrial conduction
delay is associated with a modification of the axis
of the terminal part of P wave. It is more negative 30.5 Right Atrial Enlargement
(from 30 to 60) with a caudal-cranial activa-
tion of the left atrium. This pattern of activation is Right atrial enlargement (RAE) is a common con-
visible as a biphasic P wave (positive-negative, sequence of various cardiopulmonary diseases. As
with terminal negative component evident in we already said, the atrial depolarization firstly
inferior leads) lasting more than or equal to involves the right atrium, so it is easy to understand
100 ms. The interatrial conduction delay is highly that RAE alterations interest the initial portion of P
specific of left atrium enlargement [1, 2]. wave with voltage increase but without modifica-
tion in terms of P wave duration (Fig. 30.4a, b).
Some typical P wave modifications can help to rec-
30.4 P Wave Other Than That ognize RAE: P wave (or initial positive P wave
from the Sinus Node deflection) > 0.15 mV (more specific 0.20 mV)
in V1 and V2 leads; an initial positive part > 40 ms
The P wave is defined not to be sinusal when it in V1 when P wave is biphasic in this lead; high
originates from an ectopic focus that can be and sharp P wave in inferior leads (II, III,
located in variable parts of the atria. A non-sinus aVF) 0.25 mV; and P wave axis +75 (it can
P wave can be present as a premature atrial reach +90 too). The last modification is very
impulse (isolated or in pairs), as an ectopic atrial strictly associated with COPD. Sometimes the
tachycardia (heart rate 100 bpm), or as an ecto- RAE correlated P wave modifications can be asso-
pic atrial rhythm (heart rate < 100 bpm). ciated with QRS modification as increased volt-
In all these cases, the P wave has a different ages of right ventricle electrical vectors or delayed
morphology and axis compared with the sinusal right ventricle depolarization [1, 2].
P wave. Analyzing its characteristics on the sur-
face ECG can help us to identify the location of
the ectopic focus, but it isnt always accurate. 30.6 P Pulmonale
This variability depends on the possible presence
of ectopic focus close to the sinus node and there- A P wave is defined pulmonale when it
fore originating P wave very similar to that pres- appears as a tall and peaked P wave in the
ent in normal sinus rhythm. Moreover, a single inferior limb leads (II, III, and aVF) and

ERRNVPHGLFRVRUJ
354 A. Maolo and D. Contadini

Fig. 30.4 Atrial activation


and P wave morphology in
normal atria (a), in right (b), a
left (c) and biatrial enlarge-
ment (d)

Atrial activation P wave

Right atrial activation


Left atrial activation

consequently having an axis superior to 70 [1]. severe COPD is responsible for alterations of
This finding on the ECG has different correla- the P wave axis more than its morphology. This
tions, but its usually associated with pulmonary axis deviation over 70 is due to the hyperinfla-
hypertension. The P pulmonale is most fre- tion of lungs, always present in these kinds of
quently associated with congenital heart disease patients.
such as tetralogy of Fallot, pulmonic stenosis, Despite all, the P pulmonale pattern isnt
tricuspid atresia, or interatrial septum defects strictly correlated with the increase of right atrial
and interventricular septal defects in the pres- volume, and it can be present even in healthy sub-
ence of Eisenmenger syndrome. Furthermore, in jects. In these cases, usually a sympathetic stimu-
patients with chronic cor pulmonale, this ECG lation, resulting in a stronger and synchronous
pattern has an incidence of about 20 %. [1] As atrial contraction, or the vertical position of the
expected, the P pulmonale is a marker of chronic heart can explain abnormalities in P wave axis
lung disease, but it is mostly associated with and morphology similar to those present in the P
COPD and then pulmonary fibrosis. Moreover, pulmonale pattern [1].

ERRNVPHGLFRVRUJ
30 Difficult Interpretation of ECG: Small Clues May Make the Difference. The Role of the P Wave 355

30.7 Left Atrial Enlargement P Wave Duration

Left atrial enlargement (LAE) is a common con- A prolonged P wave can be related to echocardio-
sequence of left ventricular or mitral valve dis- graphic findings of left or biatrial enlargement
eases. The left atrial depolarization is the last and consequently help us to identify patients with
part of atrial activation, so the LAE modifica- a high risk of developing atrial fibrillation. In a
tions involve the final component of P wave with clinical trial of 660 patients, who underwent
durations increase but without significant volt- dual-chamber pacemaker implantation, P wave
age alterations (Fig. 30.4a, c). Some typical P duration was measured using the standard 12-lead
wave modifications can help to recognize LAE: ECG (50 mm/s velocity). A P wave duration
P wave duration more than or equal to 120 ms in superior to 100 ms was identified in a group of
limb leads; left deviation of P wave axis more patients with a higher risk for developing persis-
negative than +30; and biphasic P wave in V1 tent atrial fibrillation and atrial fibrillation-related
with negative terminal component more than or hospitalization [4].
equal to 40 ms and with a minimum of 1 mV The P wave duration analysis can be more
bifid P wave (usually in dII, dI, aVL, V3, V4, V5, accurate using signal-averaged ECG (SAECG).
or V6), with an interval between the two peaks SAECG is a particular ECG technique using
longer than 40 ms. A more peak-to-peak prolon- cardiac electric signals from many surface elec-
gation could be the sign of a mitral disease, often trodes. The values measured with all the leads
a severe stenosis. This pattern is also defined as are averaged to minimize interference and to see
P mitrale [1, 2]. even the smallest alterations. A prolonged
signal-averaged P wave duration compared to
the standard 12-lead ECG was found to be a
30.8 Biatrial Enlargement more precise marker for the development of
atrial fibrillation [5].
The P wave abnormality during biatrial enlarge- Other important findings detectable using
ment can be simply considered as the summa of SAECG are atrial late potentials. Late potentials
the RAE and LAE alterations (Fig. 30.4a, d). are very-low-amplitude electric signals not visi-
ECG findings could be a P wave duration more ble with the standard 12-lead ECG. The QRS
than or equal to 120 ms associated with an ampli- late potentials originally were studied to esti-
tude more than or equal to 2.5 mV; a diphasic P mate the ventricular arrhythmia risk. Similarly
wave (positive-negative) in V1 with the positive the P wave (or atrial) late potential can be useful
component of at least 1.5 mV and a negative to stratify the risk for paroxysmal atrial fibrilla-
component longer than 40 ms with an amplitude tion. Budeus et al. hypothesized that atrial late
of 1 mV or more; and the presence of notched P potentials found on P wave SAECG could have a
wave in left precordial leads and high P wave in role in the development of paroxysmal atrial
right precordial leads [1, 2]. fibrillation [6]. However, the predictive value of
atrial late potentials has not been demonstrated
univocally yet.
30.9 P Wave and Arrhythmias

The role of P wave in differential diagnosis of P Wave Dispersion


tachycardias has already been described in previ-
ous chapters. However, some P wave abnormali- P wave dispersion is defined as the difference
ties during sinus rhythm can be useful as atrial between the maximum and minimum durations of
fibrillation predictor. That concept will be dis- P wave measured on the standard 12-lead ECG. A
cussed below. significant high value of P wave dispersion could

ERRNVPHGLFRVRUJ
356 A. Maolo and D. Contadini

be considered as the sign of abnormal atrial elec- affect accuracy and reliability of P wave
trical impulse propagation. In the literature, the dispersion as a noninvasive predictor of paroxys-
most used cutoff value for high P wave dispersion mal atrial fibrillation.
is considered to be 40 ms [7]. This value has been
demonstrated to have a sensitivity of 83 % and a
specificity of 85 % with a positive predictive value References
of 89 % in detecting subjects who had history of
1. Surawicz B, Knilans TK (2008) Chous electrocardi-
paroxysmal lone atrial fibrillation [8]. Calculating
ography in clinical practice, 6th edn. Saunders
the P wave dispersion on the standard 12-lead Elsevier, Philadelphia
ECG in 100 patients, it was found to be higher in 2. Oreto G (2008) Lelettrocardiogramma: un mosaico a
patients who had a history of paroxysmal atrial 12 tessere. Edi-ermes, Milano
3. Wu D, Denes P, Amat-y-Leon F et al (1975)
fibrillation [8]. Moreover, other studies demon-
Limitations of the surface electrocardiogram in diag-
strated that the P wave dispersion calculated on nosis of atrial arrhythmias. Am J Cardiol 336:91
the standard surface ECG could be an effective 4. Padelletti L, Capucci A et al (2007) Duration of
predictor of atrial fibrillation development in P-wave is associated with atrial fibrillation hospitaliza-
tions in patients with atrial fibrillation and paced for
postoperative period (after CABG) and in patients
bradycardia. Pacing Clin Electrophysiol 3:961969
with hypertension [9, 10]. 5. Steinberg SA, Guidera JS (1993) The signal-averaged
For a more accurate evaluation of P wave P wave duration: a rapid and noninvasive marker of risk
dispersion, SAECG was also used and some of atrial fibrillation. J Am Coll Cardiol 21:16451651
6. Budeus M et al (2003) Detection of atrial late poten-
more precise markers were studied. In particu-
tials with P wave signal-averaged electrocardiogram
lar, P wave dispersion index (P wave duration among patients with paroxysmal atrial fibrillation. Z
standard deviation/P wave duration mean value Kardiol 92(5):362369
100) calculated in 40 subjects was demon- 7. Dilaveris PE, Jalavos JE (2001) P wave dispersion: a
novel predictor of paroxysmal atrial fibrillation. Ann
strated to have an 83 % sensitivity and 81 %
Noninvasive Electrocardiol 6(2):159165
specificity values in the identification of 8. Dilaveris PE, Gialafos EJ, Sideris SK et al (1998)
patients with a history of paroxysmal atrial Simple electrocardiographic markers for the predic-
fibrillation [11]. tion of paroxysmal idiopathic atrial fibrillation. Am
Heart J 135:733738
The rationale for using P wave dispersion as a
9. Kloter Weber U, Osswald S, Huber M et al (1998)
predictor of atrial fibrillation is that a high dis- Selective versus nonselective antiarrhythmic approach
persion could be considered the electric sign of for prevention of atrial fibrillation after coronary sur-
nonhomogeneous atrial conduction and conse- gery: is there a need for preoperative risk stratifica-
tion? A prospective placebo-controlled study using
quently may reflect an electrical instability of the
low-dose sotalol. Eur Heart J 19:794800
atria. 10. Ciaroni S, Cuenoud L, Bloch A (2000) Clinical study
Nevertheless, the techniques used for ECG to investigate the predictive parameters for the onset
recording and P wave duration and dispersion of atrial fibrillation in patients with essential hyper-
tension. Am Heart J 139(5):814819
measurement were not standardized in the differ-
11. Villani GQ, Piepoli M, Rosi A, Capucci A (1996)
ent trials. Finally, we have to consider an interob- P-wave dispersion index: a marker of patients with par-
server and intraobserver variability that may oxysmal atrial fibrillation. Int J Cardiol 55(2):169175

ERRNVPHGLFRVRUJ

Вам также может понравиться