Você está na página 1de 465

10090-01_CH01.

qxd 8/24/06 5:40 PM Page 1

C H A P T E R

Basic Cardiac Structure


and Function
Vivek Iyer
1
Elazer R. Edelman
Leonard S. Lilly

CARDIAC ANATOMY AND HISTOLOGY Action Potential


Pericardium Refractory Periods
Surface Anatomy of the Heart Impulse Conduction
Internal Structure of the Heart Normal Sequence of Cardiac Depolarization
Impulse-Conducting System EXCITATION-CONTRACTION COUPLING
Cardiac Innervation Contractile Proteins in the Myocyte
Cardiac Vessels Calcium-Induced Calcium Release and the
Histology of Ventricular Myocardial Cells Contractile Cycle
BASIC ELECTROPHYSIOLOGY β-Adrenergic and Cholinergic Signaling
Ion Movement and Channels
Resting Potential

A knowledge of normal cardiac structure essential an intimate knowledge of the spa-


and function is crucial to understanding dis- tial relationships of cardiac structures. Such
eases that afflict the heart. This chapter re- information also proves helpful in under-
views basic cardiac anatomy and electro- standing the pathophysiology of heart dis-
physiology as well as the events that lead to ease. This section emphasizes the aspects of
cardiac contraction. cardiac anatomy that are important to the
clinician—that is, the “functional” anatomy.
CARDIAC ANATOMY
AND HISTOLOGY Pericardium
Although the study of cardiac anatomy dates The heart and roots of the great vessels are
back to ancient times, interest in this field enclosed by a fibroserous sac called the peri-
has recently gained momentum. The devel- cardium (Fig. 1.1). This structure consists of Fig. 1
opment of sophisticated cardiac-imaging two layers: a strong outer fibrous layer and
procedures such as coronary angiography, an inner serosal layer. The inner serosal layer
echocardiography, computed tomography, adheres to the external wall of the heart
and magnetic resonance imaging has made and is called the visceral pericardium. The

1
10090-01_CH01.qxd 8/24/06 5:40 PM Page 2

2 Chapter One

Superior Aorta heart is formed by the atria, mainly the left,


vena cava and lies between the lung hila. The anterior
Pulmonary surface of the heart is shaped by the right
artery atrium and ventricle. Because the left atrium
and ventricle lie more posteriorly, they form
only a small strip of this anterior surface.
Heart within
pericardium The inferior surface of the heart is formed by
both ventricles, primarily the left. This sur-
Inferior face of the heart lies along the diaphragm;
vena cava Diaphragm hence, it is also referred to as the diaphrag-
matic surface.
Observing the chest from an anteroposte-
Figure 1.1. The position of the heart in the chest.
rior view (as on a chest radiograph; see Chap-
The superior vena cava, aorta, and pulmonary artery ter 3), four recognized borders of the heart are
exit superiorly, whereas the inferior vena cava projects apparent. The right border is established by
inferiorly.
the right atrium and is almost in line with
the superior and inferior vena cavae. The
inferior border is nearly horizontal and is
visceral pericardium reflects back on itself formed mainly by the right ventricle, with a
and lines the outer fibrous layer, forming slight contribution from the left ventricle
the parietal pericardium. The space be- near the apex. The left ventricle and a por-
tween the visceral and parietal layers con- tion of the left atrium make up the left bor-
tains a thin film of pericardial fluid that al- der of the heart, whereas the superior border
lows the heart to beat in a minimal-friction is shaped by both atria. From this description
environment. of the surface of the heart emerge two basic
The pericardium is attached to the ster- “rules” of normal cardiac anatomy: (1) right-
num and the mediastinal portions of the sided structures lie mostly anterior to their
right and left pleurae. Its many connections left-sided counterparts, and (2) atrial cham-
to surrounding structures keep the pericar- bers are located mostly to the right of their
dial sac firmly anchored within the thorax corresponding ventricles.
and therefore help to maintain the heart in
its normal position.
Internal Structure of the Heart
Emanating from the pericardium in a su-
perior direction are the aorta, the pulmonary Four major valves in the normal heart direct
artery, and the superior vena cava (see Fig. blood flow in a forward direction and pre-
1.1). The inferior vena cava projects through vent backward leakage. The atrioventricular
the pericardium inferiorly. valves (tricuspid and mitral) separate the
atria and ventricles, whereas the semilunar
valves (pulmonic and aortic) separate the
Surface Anatomy of the Heart
ventricles from the great arteries. All four
The heart is shaped roughly like a cone and heart valves are attached to the fibrous car-
consists of four muscular chambers. The diac skeleton (Fig. 1.3), which is composed Fig. 3
right and left ventricles are the main pump- of dense connective tissue. The cardiac skele-
ing chambers. The less muscular right and ton also serves as a site of attachment for the
left atria deliver blood to their respective ventricular and atrial muscles.
ventricles. The surface of the heart valves and the
Several terms are used to describe the interior surface of the chambers are lined by
Fig. 2 heart’s surfaces and borders (Fig. 1.2). The a single layer of endothelial cells, termed
apex is formed by the tip of the left ventricle, the endocardium. The subendocardial tis-
which points inferiorly, anteriorly, and to sue contains fibroblasts, elastic and col-
the left. The base or posterior surface of the lagenous fibers, veins, nerves, and branches
10090-01_CH01.qxd 8/24/06 5:40 PM Page 3

Basic Cardiac Structure and Function 3

A
Brachiocephalic a. Left common carotid a.

Left subclavian a.

Aortic arch
Superior vena cava

Left pulmonary a.

Right pulmonary a.
Left pulmonary vv.
Right pulmonary vv.
Left atrial appendage
Right atrium

Left ventricle
Right heart border

Left heart border

Right ventricle Anterior interventricular


groove
Inferior vena cava

Inferior heart border Apex

B
Left common carotid a. Brachiocephalic a.
Left subclavian a.
Azygous v.

Aortic arch
Superior vena cava
Lig. arteriosum
Left pulmonary a.
Right pulmonary a.
Left pulmonary vv.
Right pulmonary vv.
Left
atrium

Right atrium

Left ventricle

Inferior vena cava

Inferior heart border

Figure 1.2. The heart and great vessels. A. The anterior view. B. The posterior aspect (or base), as
viewed from the back. a, artery; lig, ligamentum; vv, veins.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 4

4 Chapter One

Anterior

Pulmonic
valve

Aortic
valve

Tricuspid
valve
Mitral
valve
Annulus
fibrosus
Annulus
fibrosus

Posterior
Figure 1.3. The four heart valves viewed from above with atria removed.
The figure depicts the period of ventricular filling (diastole) during which the tri-
cuspid and mitral valves are open and the semilunar valves (pulmonic and aortic)
are closed. Each annulus fibrosus surrounding the mitral and tricuspid valves is
thicker than those surrounding the pulmonic and aortic valves; all four contribute
to the heart’s fibrous skeleton, which is composed of dense connective tissue.

of the conducting system and is continuous pid valve is located in the floor of the atrium
with the connective tissue of the heart muscle and opens into the right ventricle.
layer, the myocardium. The myocardium is The right ventricle (see Fig. 1.4) is roughly
the thickest layer of the heart and consists of triangular in shape, and its superior aspect
bundles of cardiac muscle cells, the histol- forms a cone-shaped outflow tract, which
ogy of which is described later in the chap- leads to the pulmonary artery. Although the
ter. External to the myocardium is a layer inner wall of the outflow tract is smooth,
of connective tissue and adipose tissue the rest of the ventricle is covered by a num-
through which pass the larger blood vessels ber of irregular bridges (termed trabeculae
and nerves that supply the heart muscle. carneae) that give the right ventricular wall
The epicardium is the outermost layer of a spongelike appearance. A large trabecula
the heart and is identical to, and just an- that crosses the ventricular cavity is called
other term for, the visceral pericardium pre- the moderator band. It carries a component
viously described. of the right bundle branch of the conducting
system to the ventricular muscle.
The right ventricle contains three papil-
Right Atrium and Ventricle
lary muscles, which project into the cham-
Opening into the right atrium are the su- ber and via their thin, stringlike chordae
perior and inferior vena cavae and the coro- tendineae attach to the edges of the tricus-
Fig. 4 nary sinus (Fig. 1.4). The vena cavae return pid valve leaflets. The leaflets, in turn, are at-
deoxygenated blood from the systemic veins tached to the fibrous ring that supports the
into the right atrium, whereas the coronary valve between the right atrium and ventri-
sinus carries venous return from the coronary cle. Contraction of the papillary muscles
arteries. The interatrial septum forms the prior to other regions of the ventricle tight-
posteromedial wall of the right atrium and ens the chordae tendineae, helping to align
separates it from the left atrium. The tricus- and restrain the leaflets of the tricuspid
10090-01_CH01.qxd 8/24/06 5:40 PM Page 5

Basic Cardiac Structure and Function 5

Pulmonary artery
Aorta
Pulmonic valve

Superior vena cava


Interventricular septum

Right atrium
Moderator
band

Trabeculae
carneae
Inferior vena cava

Coronary sinus

Tricuspid valve Right ventricle Papillary muscles


Figure 1.4. Interior structures of the right atrium and right ventricle. (Modified from Goss CM.
Gray’s Anatomy. 29th Ed. Philadelphia: Lea & Febiger, 1973:547.)

valve as they are forced closed. This action aortic valve. Inferior to this region, most of
prevents blood from regurgitating into the the ventricle is covered by trabeculae carneae,
right atrium during ventricular contraction. which are finer and more numerous than
At the apex of the right ventricular outflow those in the right ventricle.
tract is the pulmonic valve, which leads to The left ventricular chamber (see Fig. 1.5B)
the pulmonary artery. This valve consists of contains two large papillary muscles. These
three cusps attached to a fibrous ring. During are larger than their counterparts in the
relaxation of the ventricle, elastic recoil of the right ventricle, and their chordae tendineae
pulmonary arteries forces blood back toward are thicker but less numerous. The chordae
the heart, distending the valve cusps toward tendineae of each papillary muscle distrib-
one another. This action closes the pulmonic ute to both leaflets of the mitral valve. Sim-
valve and prevents regurgitation of blood back ilar to the case in the right ventricle, tensing
into the right ventricle. of the chordae tendineae during left ven-
tricular contraction helps restrain and align
the mitral leaflets, enabling them to close
Left Atrium and Ventricle
properly and preventing the backward leak-
Entering the posterior half of the left atrium age of blood.
Fig. 5 are the four pulmonary veins (Fig. 1.5). The The aortic valve separates the left ventri-
wall of the left atrium is about 2 mm thick, cle from the aorta. Surrounding the aortic
being slightly greater than that of the right valve opening is a fibrous ring to which is at-
atrium. The mitral valve opens into the left tached the three cusps of the valve. Just
ventricle through the inferior wall of the left above the right and left aortic valve cusps in
atrium. the aortic wall are the origins of the right
The cavity of the left ventricle is approx- and left coronary arteries (see Fig. 1.5B).
imately cone shaped and longer than that of
the right ventricle. In a healthy adult heart,
Interventricular Septum
the wall thickness is 9 to 11 mm, roughly 3
times that of the right ventricle. The aortic The interventricular septum is the thick wall
vestibule is a smooth-walled part of the left between the left and right ventricles. It is
ventricular cavity located just inferior to the composed of a muscular and a membranous
10090-01_CH01.qxd 8/24/06 5:40 PM Page 6

6 Chapter One

A
Right pulmonary
Aorta veins

Pulmonary
artery Left atrium

Aortic valve
Posterior leaflet
of mitral valve
Interventricular
septum

Right ventricle Papillary muscle

Left ventricle

B
Pulmonary artery
AORTA Posterior cusp
of aortic valve
Origin of right
coronary artery Origin of left
coronary artery
Interventricular
septum, Anterior cusp
membranous of mitral valve
part
Chordae
Interventricular tendineae
septum, muscular
part Anterior
papillary
Posterior muscle
R
IG

papillary
H

muscle
T
VE
N
TR

Trabeculae
IC

carneae
LE

Figure 1.5. Interior structures of the left atrium and left ventricle. A. The left
atrium and left ventricular (LV) inflow and outflow regions. B. Interior structures of the LV
cavity. (Modified from Agur AMR, Lee MJ. Grant’s Atlas of Anatomy. 9th Ed. Baltimore:
Williams & Wilkins, 1991:59.)

part (see Fig. 1.5B). The margins of this sep- small, oval-shaped membranous part of the
tum can be traced on the surface of the heart septum is thin and located just inferior to
by following the anterior and posterior in- the cusps of the aortic valve.
terventricular grooves. Owing to the greater To summarize the functional anatomic
hydrostatic pressure within the left ventri- points presented in this section, the follow-
cle, the large muscular portion of the sep- ing is a review of the path of blood flow
tum bulges toward the right ventricle. The through the heart: Deoxygenated blood is
10090-01_CH01.qxd 8/24/06 5:40 PM Page 7

Basic Cardiac Structure and Function 7

delivered to the heart through the inferior rior vena cava entrance and normally initi-
and superior vena cavae, which enter into ates the electrical impulse for contraction.
the right atrium. Flow continues through The atrioventricular (AV) node lies be-
the tricuspid valve orifice into the right ven- neath the endocardium in the inferoposte-
tricle. Contraction of the right ventricle pro- rior part of the interatrial septum.
pels the blood across the pulmonic valve to Distal to the AV node is the bundle of
the pulmonary artery and lungs, where car- His, which perforates the interventricular
bon dioxide is released and oxygen is ab- septum posteriorly. Within the septum, the
sorbed. The oxygen-rich blood returns to bundle of His bifurcates into a broad sheet of
the heart through the pulmonary veins to fibers that continues over the left side of the
the left atrium and then passes across the septum, known as the left bundle branch,
mitral valve into the left ventricle. Contrac- and a compact, cablelike structure on the
tion of the left ventricle pumps the oxy- right side, the right bundle branch.
genated blood across the aortic valve into The right bundle branch is thick and
the aorta, from which it is distributed to all deeply buried in the muscle of the inter-
other tissues of the body. ventricular septum and continues toward
the apex. Near the junction of the interven-
tricular septum and the anterior wall of the
Impulse-Conducting System
right ventricle, the right bundle branch be-
Fig. 6 The impulse-conducting system (Fig. 1.6) comes subendocardial and bifurcates. One
consists of specialized cells that initiate the branch travels across the right ventricular
heartbeat and electrically coordinate con- cavity in the moderator band, whereas the
tractions of the heart chambers. The sino- other continues toward the tip of the ven-
atrial (SA) node is a small mass of specialized tricle. These branches eventually arborize
cardiac muscle fibers in the wall of the right into a finely divided anastomosing plexus
atrium. It is located to the right of the supe- that travels throughout the right ventricle.

Sinoatrial node
Mitral valve

Membranous part of
Coronary sinus IV septum

Bifurcation of bundle
of His
Atrioventricular node Muscular part of
IV septum
Bundle of His
Left bundle branch

Right bundle branch Purkinje fibers under


endocardium of papillary
muscle
Moderator band

Figure 1.6. Main components of the cardiac conduction system. This system includes
the sinoatrial node, atrioventricular node, bundle of His, right and left bundle branches, and
the Purkinje fibers. The moderator band carries a large portion of the right bundle. IV, intra-
ventricular.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 8

8 Chapter One

Functionally, the left bundle branch is di- Coronary Arteries


vided into an anterior and a posterior fascicle
The heart muscle is supplied with oxygen and
and a small branch to the septum. The ante-
nutrients by the right and left coronary arter-
rior fascicle runs anteriorly toward the apex,
ies, which arise from the root of the aorta just
forming a subendocardial plexus in the area
above the aortic valve cusps (Fig. 1.7; see also Fig. 7
of the anterior papillary muscle. The poste-
Fig. 1.5B). After their origin, these vessels
rior fascicle travels to the area of the posterior
pass anteriorly, one on each side of the pul-
papillary muscle; it then divides into a suben-
monary artery (see Fig. 1.7).
docardial plexus and spreads to the rest of the
The large left main coronary artery passes
left ventricle.
between the left atrium and the pulmonary
The subendocardial plexuses of both ven-
trunk to reach the AV groove. There it di-
tricles send distributing Purkinje fibers to
vides into the left anterior descending
the ventricular muscle. Impulses within the
His-Purkinje system are transmitted first to (LAD) coronary artery and the circumflex
the papillary muscles and then throughout artery. The LAD travels within the anterior
the walls of the ventricles, allowing papil- interventricular groove toward the cardiac
lary muscle contraction to precede that of apex. During its descent on the anterior sur-
the ventricles. This coordination prevents face, the LAD gives off septal branches that
regurgitation of blood flow through the AV supply the anterior two thirds of the inter-
valves, as discussed earlier. ventricular septum and the apical portion of
the anterior papillary muscle. The LAD also
gives off diagonal branches that supply the
Cardiac Innervation anterior surface of the left ventricle. The cir-
The heart is innervated by both parasympa- cumflex artery continues within the left AV
thetic and sympathetic afferent and efferent groove and passes around the left border of
nerves. Preganglionic sympathetic neurons lo- the heart to reach the posterior surface. It
cated within the upper five to six thoracic gives off large obtuse marginal branches
levels of the spinal cord synapse with second- that supply the lateral and posterior wall of
order neurons in the cervical sympathetic the left ventricle.
ganglia. Traveling within the cardiac nerves, The right coronary artery (RCA) travels
these fibers terminate in the heart and great in the right AV groove, passing posteriorly
vessels. Preganglionic parasympathetic fibers between the right atrium and ventricle. It
originate in the dorsal motor nucleus of the supplies blood to the right ventricle via acute
medulla and pass as branches of the vagus marginal branches. In most people, the distal
nerve to the heart and great vessels. Here the RCA gives rise to a large branch, the poste-
fibers synapse with second-order neurons lo- rior descending artery (see Fig. 1.7C). This
cated in ganglia within these structures. A vessel travels from the inferoposterior aspect
rich supply of vagal afferents from the infe- of the heart to the apex and supplies blood to
rior and posterior aspects of the ventricles the inferior and posterior walls of the ventri-
mediates important cardiac reflexes, whereas cles and the posterior one third of the inter-
the abundant vagal efferent fibers to the SA ventricular septum. Just before giving off the
and AV nodes are active in modulating elec- posterior descending branch, the RCA usu-
trical impulse initiation and conduction. ally gives off the AV nodal artery.
The posterior descending and AV nodal
arteries arise from the RCA in 85% of the
Cardiac Vessels
population, and in such people, the coro-
The cardiac vessels consist of the coronary nary circulation is termed right dominant. In
arteries and veins and the lymphatics. The approximately 8%, the posterior descending
largest components of these structures lie artery arises from the circumflex artery in-
within the loose connective tissue in the stead, resulting in a left-dominant circula-
epicardial fat. tion. In the remaining population, the heart’s
10090-01_CH01.qxd 8/24/06 5:40 PM Page 9

Basic Cardiac Structure and Function 9

A Pulmonary artery

Left main
coronary artery
Aorta
Left circumflex
coronary artery

Left anterior
Right coronary descending
artery coronary artery

B C

Left circumflex Left circumflex


coronary artery coronary artery
Left anterior
descending
coronary artery
Right Obtuse
coronary Diagonal marginal
artery branch branches Right
coronary
artery

Acute
Posterior descending
marginal branch
coronary artery
Figure 1.7. Coronary artery anatomy. A. Schematic representation of the right and left coronary arteries demon-
strates their orientation to one another. The left main artery bifurcates into the circumflex artery, which perfuses the lat-
eral and posterior regions of the left ventricle (LV), and the anterior descending artery, which perfuses the LV anterior
wall, the anterior portion of the intraventricular septum, and a portion of the anterior right ventricular (RV) wall. The
right coronary artery (RCA) perfuses the right ventricle and variable portions of the posterior left ventricle through its
terminal branches. The posterior descending artery most often arises from the RCA. B. Anterior view of the heart demon-
strating the coronary arteries and their major branches. C. Posterior view of the heart demonstrating the terminal por-
tions of the right and circumflex coronary arteries and their branches.

posterior blood supply is contributed to From their epicardial locations, the coro-
from branches of both the RCA and the cir- nary arteries send perforating branches into
cumflex, forming a codominant circulation. the ventricular muscle, which form a richly
The blood supply to the SA node is also branching and anastomosing vasculature in
most often (70% of the time) derived from the walls of all the cardiac chambers. From
the RCA. However, in 25% of normal hearts, this plexus arise a massive number of cap-
the SA nodal artery arises from the circum- illaries that form an elaborate network
flex artery, and in 5% of cases, both the RCA surrounding each cardiac muscle fiber. The
and the circumflex artery contribute to this muscle fibers located just beneath the endo-
vessel. cardium, particularly those of the papillary
10090-01_CH01.qxd 8/24/06 5:40 PM Page 10

10 Chapter One

muscles and the thick left ventricle, are sup- that of skeletal muscle. However, unlike the
plied either by the terminal branches of the multinucleated skeletal myofibers, myocar-
coronary arteries or directly from the ven- dial cells contain only one or two centrally
tricular cavity through tiny vascular chan- located nuclei. Surrounding each myocar-
nels, known as thebesian veins. dial cell is connective tissue with a rich cap-
Collateral connections, usually <200 µm illary network.
in diameter, exist at the subarteriolar level Each myocardial cell contains numerous
between the coronary arteries. In the nor- myofibrils, which are long chains of indi-
mal heart, few of these collateral vessels are vidual sarcomeres, the fundamental con-
visible. However, they may become larger tractile units of the cell (Fig. 1.8). Each Fig. 8
and functional when atherosclerotic disease sarcomere is made up of two groups of
obstructs a coronary artery, thereby provid- overlapping filaments of contractile pro-
ing blood flow to distal portions of the ves- teins. Biochemical and biophysical interac-
sel from a nonobstructed neighbor. tions occurring between these myofila-
ments produce muscle contraction. Their
structure and function are described later in
Coronary Veins
the chapter.
The coronary veins follow a distribution Within each myocardial cell, the neigh-
similar to that of the major coronary arter- boring sarcomeres are all in register, produc-
ies. These vessels return blood from the myo- ing the characteristic cross-striated banding
cardial capillaries to the right atrium pre- pattern seen by light microscopy. The rela-
dominantly via the coronary sinus. The tive densities of the cross bands identify the
major veins lie in the epicardial fat, usually location of the contractile proteins. Under
superficial to their arterial counterparts. The physiologic conditions, the overall sarco-
thebesian veins, described earlier, provide mere length (Z-to-Z distance) varies between
an additional potential route for a small 2.2 and 1.5 µm during the cardiac cycle.
amount of direct blood return to the cardiac The larger dimension reflects the fiber
chambers. stretch during ventricular filling, whereas
the smaller dimension represents the extent
of fiber shortening during contraction.
Lymphatic Vessels
The myocardial cell membrane is termed
The heart lymph is drained by an exten- the sarcolemma. A specialized region of the
sive plexus of valved vessels located in the membrane is the intercalated disk, a dis-
subendocardial connective tissue of all four tinct characteristic of cardiac muscle tissue.
chambers. This lymph drains into an epi- Intercalated disks are seen on light micro-
cardial plexus from which are derived sev- scopic study as darkly staining transverse
eral larger lymphatic vessels that follow the lines that cross chains of cardiac cells at
distribution of the coronary arteries and irregular intervals. They represent the gap
veins. Each of these larger vessels then com- junction complexes at the interface of adja-
bines in the AV groove to form a single lym- cent cardiac fibers and establish structural
phatic conduit, which exits the heart to and electrical continuity between the my-
reach the mediastinal lymphatic plexus and ocardial cells.
ultimately the thoracic duct. Another functional feature of the cell
membrane is the transverse tubular system
(or T tubules). This complex system is char-
Histology of Ventricular
acterized by deep, fingerlike invaginations
Myocardial Cells
of the sarcolemma (Fig. 1.9; see also Fig. 1.8). Fig. 9
The mature myocardial cell (also termed the Similar to the intercalated disks, transverse
myocyte) measures up to 25 µm in diame- tubular membranes establish pathways for
ter and 100 µm in length. The cell shows rapid transmission of the excitatory electri-
a cross-striated banding pattern similar to cal impulses that initiate contraction. The
10090-01_CH01.qxd 8/24/06 5:40 PM Page 11

Basic Cardiac Structure and Function 11

Myofibril Sarcolemma T tubule

Mitochondrion

Sarcoplasmic
Reticulum
Mitochondrion

ACTIN MYOSIN

SARCOMERE
Figure 1.8. Myocardial cell. Top. Schematic representation of the ultrastructure of
the myocardial cell. The cell consists of multiple parallel myofibrils surrounded by mi-
tochondria. The T tubules are invaginations of the cell membrane (the sarcolemma)
that increase the surface area for ion transport and transmission of electrical impulses.
The intracellular sarcoplasmic reticulum houses most of the intracellular calcium and
abuts the T tubules. (Modified from Katz AM. Physiology of the Heart. 2nd Ed. New
York: Raven Press, 1992:21.) Bottom. Expanded view of a sarcomere, the basic unit
of contraction. Each myofibril consists of serially connected sarcomeres that extend
from one Z line to the next. The sarcomere is composed of alternating thin (actin) and
thick (myosin) myofilaments.

T tubule

Sarcolemma

Ca++ Ca++
Ca++
Ca++ Ca++
Ca++

Sarcoplasmic
reticulum
ATPase
Ca++
Ca++ Ca++ Ca++
Ca++

Terminal cisternae
Figure 1.9. Schematic view of the tubular systems of the myocardial cell. The T
tubules, invaginations of the sarcolemma, abut the sarcoplasmic reticulum at right angles
at the terminal cisternae sacs. This relationship is important in linking membrane excita-
tion with intracellular release of calcium from the sarcoplasmic reticulum.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 12

12 Chapter One

T tubule system increases the surface area 1. Pacemaker cells (e.g., SA node, AV node)
of the sarcolemma in contact with the extra- 2. Specialized rapidly conducting tissues
cellular environment, allowing the trans- (e.g., Purkinje fibers)
membrane ion transport accompanying ex- 3. Ventricular and atrial muscle cells
citation and relaxation to occur quickly and
synchronously. The sarcolemma of each of these cardiac
The sarcoplasmic reticulum (SR) is an cell types is a phospholipid bilayer that is
extensive intracellular tubular membrane largely impermeable to ions. There are spe-
network that complements the T tubule sys- cialized proteins interspersed throughout
tem both structurally and functionally. The the membrane that serve as ion channels,
SR abuts the T tubules at right angles in lat- cotransporters, and active transporters (Fig. Fig. 10
eral sacs, called the terminal cisternae (see 1.10). These help to maintain ionic concen-
Fig. 1.9). These sacs house most of intracel- tration gradients and charge differentials be-
lular calcium stores; the release of these tween the inside and the outside of the car-
stores is important in linking membrane ex- diac cells. Note that normally, the Na+ and
citation with activation of the contractile Ca++ concentrations are much higher out-
apparatus. Lateral sacs also abut the interca- side the cell and the K+ concentration is
lated disks and the sarcolemma, providing much higher inside.
each with a complete system for excitation-
contraction coupling. Ion Movement and Channels
To serve the tremendous metabolic de-
mand placed on the heart and the need for The movement of specific ions across the cell
a constant supply of high-energy phos- membrane serves as the basis of the action
phates, the myocardial cell has an abundant potential. Ion transport depends on two
concentration of mitochondria. These or- major factors: (1) the energetic favorability
ganelles are located between the individual and (2) the permeability of the membrane
myofibrils and constitute approximately 35% for the ion.
of cell volume (see Fig. 1.8).
Energetics
BASIC ELECTROPHYSIOLOGY
The two major forces that drive the energetics
Rhythmic contraction of the heart relies on of ion transport are the concentration gradient
the organized propagation of electrical im- and the transmembrane potential (voltage).
pulses along its conduction pathway. The Molecules diffuse from areas of high concen-
marker of electrical stimulation, the action tration to areas of lower concentration—the
potential, is created by a sequence of ion gradient between these values determines the
fluxes through specific channels in the rate of ion flow. For example, the extracellu-
sarcolemma. To provide a basis for under- lar Na+ concentration is normally 145 mM,
standing how electrical impulses lead to while the concentration inside the myocyte is
cardiac contraction, the process of cellular 15 mM. As a result, a strong force tends to
depolarization and repolarization is re- drive Na+ into the cell, down its concentra-
viewed here. This material serves as an im- tion gradient.
portant foundation for topics addressed The transmembrane potential of cells
later in the book, including electrocardiog- exerts an electrical force on ions (i.e., like
raphy (see Chapter 4) and cardiac arrhyth- charges repel one another, and opposite
mias (see Chapters 11 and 12). charges attract). The transmembrane po-
Cardiac cells capable of electrical excita- tential of a myocyte at rest is about −90 mV
tion are of three electrophysiologic types, (the inside of the cell is negative relative to
the properties of which have been studied the outside). Extracellular Na+, a positively
by intracellular microelectrode and patch- charged ion, is therefore attracted to the rel-
clamp recordings: atively negatively charged interior of the
10090-01_CH01.qxd 8/24/06 5:40 PM Page 13

Basic Cardiac Structure and Function 13

Na+ Ca++

A B C
K+

GAP Ca++
JUNCTION G Internal
ATP [Na+] 15 mM
Ca++ [K+] 150 mM
[Cl–] 5 mM
[Ca+ ] 10–7 M

Na+ Ca++

ATP ATP
D E F

Na+ Ca++ Exte


l rna
[Na+] 145 mM
K+
[K+] 5 mM
[Cl–] 120 mM
[Ca+ ] 2 mM

Figure 1.10. Ion channels, cotransporters, and active transporters of the myocyte. A. Sodium entry through the
fast sodium channel is responsible for the rapid upstroke (phase 0) of the action potential (AP) in nonpacemaker cells.
B. Calcium enters the cell through the slow calcium channel during phase 2 of the Purkinje fiber and muscle cell AP, and
is the main channel responsible for depolarization of pacemaker cells. C. Potassium exits through a potassium channel
to repolarize the cell during phase 3 of the AP, and open potassium channels help maintain the resting potential (phase
4) of nonpacemaker cells. D. Sodium-calcium exchanger helps maintain the low intracellular calcium concentration.
E. Sodium-potassium ATPase pump maintains concentration gradients for these ions. F, G. Active calcium transporters
aid removal of calcium to the external environment and sarcoplasmic reticulum, respectively.

cell. Thus, there is a strong tendency for Na+ Most types of ion channels share similar
to enter the cell because of both the steep protein sequences and structures, consisting
concentration gradient and the electrical of repeating transmembrane domains (Fig.
attraction. 1.11). Each of these domains contains six Fig. 11
membrane-spanning segments. The fourth
segment (see S4 in Fig. 1.11) includes a se-
Permeability
quence of positively charged amino acids
If there is such a strong force driving Na+ (lysine and arginine) that reacts to the mem-
into the cell, what keeps this ion from actu- brane potential, and therefore that segment
ally moving inside? The membrane of the is thought to confer voltage-sensitivity to
cell at its resting potential is not permeable the channel, as described next.
to sodium. The phospholipid bilayer of the The several types of cardiac ion chan-
cell membrane is composed of a hydropho- nels vary by two functional properties:
bic core that does not allow simple passage selectivity and gating. Each type of chan-
of charged, hydrophilic particles. Instead, nel is normally selective for a specific ion,
permeability of the membrane is depen- which is a manifestation of the size and
dent on the opening of specific ion chan- structure of its pore. For example, in cardiac
nels, specialized proteins that span the cell cells, some channels permit the passage of
membrane and contain hydrophilic pores sodium ions, some are specific for potas-
through which certain charged atoms can sium, and others allow only calcium to pass
pass under specific circumstances. through.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 14

14 Chapter One

A DOMAIN DOMAIN DOMAIN DOMAIN


I II III IV

SODIUM
CHANNEL
C
INACTIVATION GATE
N

CALCIUM
CHANNEL
C
N

POTASSIUM
CHANNEL

N
C N N
C C N C

B
SELECTIVITY
Extracellular FILTER

+ +
S1 S2 S3 S4 S5 S6
+ +
+
Intracellular

N C
Figure 1.11. Structure of ion channels. A. Ion channels consist of glycosylated pro-
teins arranged as repeating transmembrane domains. Each domain consists of six
membrane-spanning segments. The potassium channel has four separate domains in a
tetrameric structure, while the sodium and calcium channels contain four domains
covalently linked as a single unit. In the case of the sodium channel, the loop connect-
ing domains III and IV is believed to serve as the channel’s inactivation gate.
B. Enlarged view of a single domain of the sodium channel showing the six membrane-
spanning segments. The S4 segment of each domain contains a sequence of positively
charged amino acids, which confers the channel’s voltage sensitivity. The peptide loops
connecting segments 5 and 6 in each domain form the selectivity filter for the chan-
nel’s pore, which allows sodium, but not other ions, to pass through. (Parts A and B
are reproduced in part from Katz AM. Physiology of the Heart. 2nd Ed. New York:
Raven Press, 1992:427, 429, with permission.) (Continued)

An ion can pass through its specific chan- therefore, the greater the transmembrane
nel only at certain times. That is, the ion current.
channel is gated—at any given moment, the Cells contain a population of each type of
channel is either open or closed. The more ion channel, and each individual channel
time a channel is in its open state, the larger may be in the open or closed state; it is the
the number of ions that pass through it and voltage across the membrane that deter-
10090-01_CH01.qxd 8/24/06 5:40 PM Page 15

Basic Cardiac Structure and Function 15

C III
Voltage
4 sensor
2 3
1
5 6 Selectivity
filter (pore)

IV II

Inactivation I Inactivation
gate (closed) gate (open)
Figure 1.11. (Continued ) Structure of ion channels. C. A 3-dimensional schematic
of the sodium channel, showing how the four domains wrap around the channel’s
pore. The selectivity filter formed by the loops connecting segments 5 and 6 is shown
near the extracellular opening of the channel, while the inactivation gate (the loop
between domains III and IV) is displayed on the cytosolic side. (Reproduced from
Nelson CL, Cox MM. Lehninger’s Principles of Biochemistry. 3rd Ed. New York: Worth,
AQ4 2000:428, with permission.)

mines what fraction of these channels is through. In this resting state, the channels
open at a given time. Therefore, the gating are available for conversion to the open
of channels is said to be voltage sensitive. configuration.
As the membrane voltage changes during A rapid wave of depolarization causes the
depolarization and repolarization of the membrane potential to become less nega-
cell, specific channels open and close, with tive and activates the resting channels to
corresponding alterations in the ion fluxes the open state (see Fig. 1.12B). Na+ ions read-
across the sarcolemma. ily permeate through the open channels,
An example of voltage-sensitive gating and an inward Na+ current ensues. How-
is apparent in the cardiac channel known ever, the activated channels remain open
as the fast sodium channel. The trans- for only a brief time, a few thousandths of
membrane protein that forms this channel a second, and then spontaneously close
assumes various conformations depend- to create an inactive state (see Fig. 1.12C).
Fig. 12 ing on the cell’s membrane potential (Fig. Channels in the inactivated conformation
1.12). At a voltage of −90 mV (the typical cannot be directly converted back to the
resting voltage of a ventricular muscle cell), open state.
the channels are primarily in a closed, rest- The inactivated state persists until the
ing state, such that Na+ ions cannot pass membrane voltage has repolarized nearly
10090-01_CH01.qxd 8/24/06 5:40 PM Page 16

16 Chapter One

A B
CHANNEL CLOSED CHANNEL OPEN
Rapid depolarization
(RESTING)
Activation

gate III Na+

IV II
Outside ++++ ––––
++++ ––––

e
Cll
membrane

–––– –––– ++++ ++++


Insid
e
Inactivation

Na+
R
e
p
lo

s
u
a

o
ir

e
z

n
a

a
it

tn
o
n

o
C
p
S
CHANNEL CLOSED
(INACTIVE)
Na+

–––– ––––

++++ ++++

Figure 1.12. Schematic representation of gating of fast sodium channels. A. The four covalently linked trans-
membrane domains (I, II, III, IV) form the sodium channel, which is guarded by activation and inactivation gates. (Here,
domain I is cut away to show the transmembrane pore.) In the resting membrane, most channels are in a closed state.
Even though the inactivation gate is open, Na+ ions cannot easily pass through because the activation gate is closed.
B. A rapid depolarization changes the cell membrane voltage and forces the activation gate to open, presumably
mediated by translocation of the charged portions of segment 4 in each domain. With the channel in this con-
formation (in which both the activation and inactivation gates are open), Na+ ions permeate into the cell.
C. As the inactivation gate spontaneously and quickly closes, the sodium current ceases. The inactivation gating func-
tion is thought to be achieved by the peptide loop that connects domains III and IV and swings into the intracellular
opening of the channel pore (black arrow). The channel cannot reopen directly from this closed, inactive state. Cellu-
lar repolarization returns the channel to the resting condition (A). During repolarization, as high negative membrane
voltages are reachieved, the activation gate closes and the inactivation gate reopens.

back to its original resting level. Until it does rent again until the cell membrane has
so, the inactivated channel prevents any flow nearly fully repolarized and the channels re-
of sodium ions. Thus, during normal cellular cover from the inactivated to the closed rest-
depolarization, the voltage-dependent fast ing state.
sodium channels conduct for a short period Another important attribute of cardiac
and then inactivate, unable to conduct cur- fast sodium channels should be noted. If the
10090-01_CH01.qxd 8/24/06 5:40 PM Page 17

Basic Cardiac Structure and Function 17

transmembrane voltage of a cardiac cell is Calcium and potassium channels in cardiac


slowly depolarized and maintained chroni- cells also act in voltage-dependent fashions,
cally at levels less negative than the usual but they behave differently than the sodium
resting potential, inactivation of channels channels, as described in the next section.
occurs without initial opening and current
flow. Furthermore, as long as this partial de-
Resting Potential
polarization exists, the closed, inactive chan-
nels cannot recover to the resting state. In cardiac cells at rest, prior to excitation,
Thus, the fast sodium channels in such a cell the electrical charge differential between
are persistently unable to conduct Na+ ions. the inside and outside of a cell is known
This is the typical case in cardiac pacemaker as the resting potential. The magnitude of
cells (e.g., the SA and AV nodes) in which the the resting potential of a cell depends on two
membrane voltage is generally less negative main properties: (1) the concentration gra-
than −70 mV throughout the cardiac cycle. dients for all the different ions between the
As a result, the fast sodium channels in pace- inside and outside of the cell, and (2) which
maker cells are persistently inactivated and ion channels are open at rest.
do not play a role in the generation of the ac- As in other tissues such as nerve cells and
Box 1.1 tion potential in these cells (see Box 1.1). skeletal muscle, the potassium concentra-

Box 1.1 Mechanism of Fast Sodium Channels

A key characteristic of fast sodium channels is their ability to activate and then inactivate
rapidly when the cell is depolarized. The mechanism by which this occurs has been inves-
tigated for many decades. In the mid-1900s, Hodgkin and Huxley studied the action po-
tential in giant squid axons (J Physiol [Lond] 1952; 117:500–544). They found that ion
channels act as if they contain a series of “gates” that open and close in a specific pattern
when the membrane potential is altered. In the case of the sodium channel, the re-
searchers postulated the presence of m gates closed in the resting state and an h gate
open in the resting state. Depolarization of the membrane causes the m gates to open
quickly, which allows Na+ ions to pass through the channel (equivalent to the open chan-
nel in Fig. 1.12B). However, that same depolarization of the cell also causes the h gate to
close, which blocks the passage of sodium ions (the closed, inactive state in Fig. 1.12C).
Na+ can flow through the channel only when both sets of gates are open. Since the m
gates open faster than the h gate closes, there is a brief period (about 1 millisecond) dur-
ing which Na+ can pass through. After the membrane repolarizes to voltages more nega-
tive than about −60 mV, the m gates shut, the h gate reopens, and the channel returns
to the closed, resting state (see Fig. 1.12A), available for activation once again.
More recent research has demonstrated that ion channel activity is actually more com-
plex than suggested by this model, but there are important correlates with current mo-
lecular concepts. For example, the cluster of positively charged amino acids on segment
4 (S4) of the ion channel domain (see Fig. 1.11) is believed to be the voltage sensor for the
AQ1 m gates that cause the channel to open during depolarization. In the resting state, the
strong positive charge on S4 causes it to be pulled inward toward the negative membrane
potential. During depolarization, as the membrane charge becomes less, S4 can move out-
ward, resulting in a conformational change in the protein that results in channel opening.
Inactivation (the h gate) is thought to be achieved by the peptide loop that connects do-
mains III and IV of the sodium channel (see Figs. 1.11 and 1.12) and swings into and oc-
cludes the channel during depolarization.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 18

18 Chapter One

tion is much greater inside cardiac cells ward the cellular interior, an effect that
compared with outside the cells. This is at- slows their net exit from the cell. Thus, the
tributed to cell membrane transporters, the two opposing forces directing the flux of
most important of which is Na+K+-ATPase. potassium ions across their open channels
This protein “pump” couples the energy in the resting state are (1) the concentra-
of ATP hydrolysis to export three Na+ ions tion gradient, which favors outward pas-
out of the cell in exchange for the inward sage of potassium, and (2) the electrostatic
movement of two K+ ions. This acts to main- force, which attracts potassium back into
tain intracellular Na+ at low levels and intra- the cell (Fig. 1.13). At steady state, the Fig. 13
cellular K+ at high levels. equilibrium between these chemical and
Cardiac myocytes contain potassium electrical forces determines the resting
channels that are open in the resting state, potential, which is approximately −90 mV
at a time when other ionic channels (i.e., in ventricular muscle cells. This is pre-
sodium and calcium) are closed. Therefore, dicted by the equilibrium potential (Nernst
the resting cell membrane is much more potential) for potassium, as shown in Fig-
permeable to potassium than to other ions. ure 1.13.
As a result of its open channels at rest, K+ The permeability of the cardiac myocyte
fluxes in an outward direction down its cellular membrane for sodium is minimal
concentration gradient, removing positive in the resting state because the channels
charges from the cell. The predominant that conduct that ion are essentially closed.
counter ions for potassium within the cell However, there is a slight leak of sodium
are large negatively charged proteins that ions into the cell at rest. This tiny inward
are unable to diffuse outward along with K+. current of positively charged ions explains
Thus, as potassium ions exit the cell, the an- why the actual resting potential is slightly
ions that are left behind cause the interior of less negative than would be predicted if the
the cell to become electrically negative with cell membrane were truly only permeable to
respect to the outside. potassium. The sodium ions that slowly leak
However, as the interior of the cell be- into the myocyte at rest (and the much
comes more and more negatively charged larger amount that enters during the action
by the outward flux of potassium, the pos- potential) are continuously removed from
itively charged K+ ions are attracted back to- the cell and returned to the extracellular en-

Open
potassium
channels
Inside cell

CONCENTRATION [K+] out [K+] in


GRADIENT (5 mM) (150 mM)
+ –
+ –
ELECTRICAL K+
FORCE + –
+ –

Equilibrium (Nernst) potential = –26.7 ln ([K+] in/[K+] out) = –91mV


Figure 1.13. The resting potential of a cardiac muscle cell is deter-
mined by the balance between the concentration gradient and
electrostatic forces for potassium, because only potassium chan-
nels are open at rest. The concentration gradient favors outward move-
ment of K+, whereas the electrical force attracts the positively charged K+
ions inward. The equilibrium (resting) potential can be approximated by
the Nernst equation for potassium, as shown here.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 19

Basic Cardiac Structure and Function 19

vironment by Na+K+-ATPase, as previously


1 Ca++ influx
described.

Membrane potential (mV)


(and K+ efflux)
0
2

Action Potential
0 3
When the cell membrane voltage is altered, K+
–50
efflux
its permeability to specific ions changes be- Na+
influx
cause of the voltage-gating characteristics of
4
the ion channels. Each type of channel has
a characteristic pattern of activation and in- –100
activation that determines the progression
of the electrical signal. This discussion be- Inward
gins by following the development of the sodium
action potential in a typical cardiac muscle current
Fig. 14 cell (Fig. 1.14). The unique characteristics of
0
action potentials in cardiac pacemaker cells
are described next.
Inward
calcium
Cardiac Muscle Cell current

Until stimulated, the resting potential of a


0
cardiac muscle cell remains stable, at ap-
proximately −90 mV. This resting state be-
fore depolarization is known as phase 4 of Outward
the action potential. Following phase 4, four potassium
additional phases characterize depolariza- current
tion and repolarization of the cell (see Fig.
0
1.14). Time
Figure 1.14. Schematic representation of a my-
Phase 0 ocyte action potential (AP) and relative net ion
currents for Na1, Ca11, and K1. The resting po-
At the resting membrane voltage, sodium tential is represented by phase 4 of the AP. Follow-
ing depolarization, Na+ influx results in the rapid up-
and calcium channels are closed. Any pro- stroke of phase 0; a transient outward potassium
cess that makes the membrane potential less current is responsible for partial repolarization dur-
negative than the resting value causes some ing phase 1; slow Ca++ influx (and relatively low K+
efflux) results in the plateau of phase 2; and final
sodium channels to open. As these channels rapid repolarization largely results from K+ efflux
open, sodium ions rapidly enter the cell, during phase 3.
flowing down their concentration gradient,
and toward the negatively charged cellular
interior. The entry of Na+ ions into the cell
causes the transmembrane potential to be- membrane potential to zero and transiently
come progressively less negative, which in into the positive range.
turn causes more sodium channels to open The prominent influx of sodium ions is
and promotes further sodium entry into responsible for the rapid upstroke, or phase
the cell. When the membrane voltage ap- 0, of the action potential. However, the Na+
proaches the threshold potential (approxi- channels remain open for only a few thou-
mately −70 mV in cardiac muscle cells), sandths of a second and are then quickly in-
enough of these fast Na+ channels have activated, preventing further influx (see Fig.
opened to generate a self-sustaining inward 1.14). Thus, while activation of these fast
Na+ current. The magnitude of entry of pos- Na+ channels causes the rapid early depolar-
itively charged Na+ ions neutralizes the ization of the cell, the rapid inactivation
10090-01_CH01.qxd 8/24/06 5:40 PM Page 20

20 Chapter One

makes their major contribution to the ac- of rapid repolarization. Phase 3 completes
tion potential short lived. the action potential cycle, with a return to
resting phase 4, preparing the cell for the
next stimulus for depolarization.
Phase 1
To preserve normal transmembrane ionic
Following rapid phase 0 depolarization into concentration gradients, sodium and cal-
the positive voltage range, a brief current of cium ions that enter the cell during depo-
repolarization returns the membrane poten- larization must be returned to the extra-
tial to approximately 0 mV. The responsible cellular environment, and potassium ions
current is carried by the outward flow of K+ must return to the cell interior. As shown in
ions through a type of transiently activated Figure 1.10, Ca++ ions are removed by the
potassium channel. sarcolemmal Na+-Ca++ exchanger and to a
lesser extent by the ATP-consuming calcium
pump (sarcolemmal Ca++-ATPase). The cor-
Phase 2
rective exchange of Na+ and K+ across the
This relatively long phase of the action po- cell membrane is mediated by Na+K+-AT-
tential is mediated by the balance of an out- Pase, as described earlier.
ward K+ current in competition with an
inward Ca++ current, which flows through
Specialized Conduction System
specific L-type calcium channels. The latter
channels begin to open during phase 0, The process described in the previous sec-
when the membrane voltage reaches ap- tions applies to the action potential of car-
proximately −40 mV, allowing Ca++ to flow diac muscle cells. The cells of the specialized
down their concentration gradient into the conduction system (e.g., Purkinje fibers) be-
cell. Ca++ entry proceeds in a more gradual have similarly, although the resting poten-
fashion than the initial influx of sodium, be- tial is slightly more negative and the up-
cause with calcium channels, activation is stroke of phase 0 is even more rapid.
slower and the channels remain open much
longer compared with sodium channels (see
Pacemaker Cells
Fig. 1.14). During this phase, the Ca++ influx,
and the relatively low permeability to K+ ef- The upstroke of the action potential of car-
flux, maintains a voltage of approximately diac muscle cells does not normally occur
0 mV for a prolonged period known as the spontaneously. Rather, when a wave of
plateau. Calcium ions that enter the cell depolarization reaches the myocyte from
during this phase play a critical role in trig- neighboring cells, its membrane potential
gering additional internal calcium release becomes less negative and an action poten-
from the sarcoplasmic reticulum, which is tial is triggered.
important in initiating myocyte contrac- Certain heart cells do not require external
tion, as discussed later in the chapter. As the provocation to initiate their action poten-
Ca++ channels gradually inactivate and the tial. Rather, they are capable of self-initiated
efflux of K+ begins to exceed the influx of depolarization in a rhythmic fashion and
calcium, phase 3 begins. are known as pacemaker cells. They are en-
dowed with the property of automaticity,
by which the cells undergo spontaneous de-
Phase 3
polarization during phase 4. When the
This is the final phase of repolarization that threshold voltage is reached in such cells,
returns the transmembrane voltage back to the action potential upstroke is triggered
the resting potential of approximately −90 (Fig. 1.15). Fig. 15
mV. A continued outward potassium cur- Cells that display pacemaker behavior in-
rent and low membrane permeability for clude the SA node (the “natural pacemaker”
other cations are responsible for this period of the heart) and the AV node. Although
10090-01_CH01.qxd 8/24/06 5:40 PM Page 21

Basic Cardiac Structure and Function 21

of the action potential. Rather, this


0
pacemaker channel opens during repo-
Membrane potential (mV) Ca2+ larization of the cell, as the membrane
influx
K+
potential approaches its most negative
0 efflux values. The inward flow of positively
charged Na+ ions through the pace-
–40
4
maker channel causes the membrane
lf potential to become progressively less
negative during phase 4, ultimately
depolarizing the cell to its threshold
–80
Time voltage (see Fig 1.15.)
Figure 1.15. Action potential of a pace- 3. The phase 0 upstroke of the pacemaker
maker cell. Phase 4 is characterized by cell action potential is less rapid and
gradual, spontaneous depolarization owing reaches a lower amplitude than that of a
to the pacemaker current (If). When the
threshold potential is reached, at about cardiac muscle cell. These characteristics
−40 mV, the upstroke of the action poten- result from the fast sodium channels of
tial follows. The upstroke of phase 0 is less the pacemaker cells being inactivated
rapid than in nonpacemaker cells because
the current represents Ca++ influx through and the upstroke of the action potential
the relatively slow calcium channels. relying solely on Ca++ influx through the
relatively slow calcium channels.
Repolarization of pacemaker cells occurs
in a fashion similar to that of ventricular
atrial and ventricular muscle cells do not nor- muscle cells and relies on inactivation of the
mally display automaticity, they may do so calcium channels and increased activation
under disease conditions such as ischemia. of potassium channels with enhanced K+
The shape of the action potential of a pace- efflux from the cell.
maker cell is different from that of a ventric-
ular muscle cell in three ways:
1. The maximum negative voltage of pace- Refractory Periods
maker cells is approximately −60 mV, Compared with electrical impulses in nerves
substantially less negative than the rest- and skeletal muscle, the cardiac action po-
ing potential of ventricular muscle cells tential is much longer in duration. This re-
(−90 mV). The persistently less negative sults in a prolonged refractory period during
membrane voltage of pacemaker cells which the muscle cannot be restimulated.
causes the fast sodium channels within Such a long period is physiologically neces-
these cells to remain inactivated. sary because it allows the ventricles suffi-
2. Unlike that of cardiac muscle cells, cient time to empty their contents and refill
phase 4 of the pacemaker cell action before the next contraction.
potential is not flat but has an upward There are different levels of refractoriness
slope, representing spontaneous grad- during the action potential, as illustrated in
ual depolarization. This spontaneous Figure 1.16. The degree of refractoriness pri- Fig. 16
depolarization is the result of an ionic marily reflects the number of fast Na+ chan-
flux known as the pacemaker current nels that have recovered from their inactive
(also called the “funny current” and de- state and are capable of reopening. As phase
noted If). Current evidence indicates 3 of the action potential progresses, an in-
that the pacemaker current is carried creasing number of Na+ channels recover and
predominantly by Na+ ions. The ion can respond to the next depolarization. This,
channel through which the pacemaker in turn, corresponds to an increasing proba-
current passes is different from the fast bility that a stimulus will trigger an action
sodium channel responsible for phase 0 potential and result in a propagated impulse.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 22

22 Chapter One

Membrane potential (mV)


0 1

2
–50
Absolute
RP
3

–100

Supranormal
period
Effective Relative
RP RP
Figure 1.16. Refractory periods (RPs) of the myocyte. During the absolute re-
fractory period (ARP), the cell is unexcitable to stimulation. The effective refractory
period includes a brief time beyond the ARP during which stimulation produces a
localized depolarization that does not propagate (curve 1). During the relative re-
fractory period, stimulation produces a weak action potential (AP) that propagates,
but more slowly than usual (curve 2). During the supranormal period, a weaker-
than-normal stimulus can trigger an AP (curve 3).

The absolute refractory period refers to from cell to cell, because each myocyte is
the time during which the cell is completely connected to its neighbors through low-
unexcitable to a new stimulation. The effec- resistance gap junctions. The speed of tissue
tive refractory period includes the absolute depolarization (phase 0) and the conduction
refractory period but extends beyond it to velocity along the cell depend on the num-
include a short interval of phase 3, during ber of sodium channels and on the magni-
which stimulation produces a localized ac- tude of the resting potential. Tissues with a
tion potential that is not strong enough to high concentration of Na+ channels, such as
propagate further. The relative refractory pe- Purkinje fibers, have a large, fast inward cur-
riod is the interval during which stimula- rent, which spreads rapidly within and be-
tion triggers an action potential that is con- tween cells to support rapid conduction. In
ducted, but because the cell is stimulated contrast, the less negative the resting poten-
from a voltage less negative than the resting tial, the greater the number of inactivated
potential, its upstroke is less steep and of fast sodium channels, and therefore the less
lower amplitude and its conduction velocity rapid the upstroke velocity (Fig. 1.17). Thus, Fig. 17
slower than normal (as described in the next alterations in the resting potential greatly af-
section). Following the relative refractory fect the upstroke and conduction velocity of
period, a short “supranormal” period is pres- the action potential.
ent in which a less-than-normal stimulus
can trigger an action potential. The refrac-
Normal Sequence of
tory period of atrial cells is shorter than that
Cardiac Depolarization
of ventricular muscle cells, such that atrial
rates can generally exceed ventricular rates Electrical activation of the heartbeat is nor-
during rapid arrhythmias (see Chapter 11). mally initiated at the SA node (see Fig. 1.6).
The impulse spreads to the surrounding
atrial muscle through intercellular gap junc-
Impulse Conduction
tions that provide electrical continuity be-
During depolarization, the electrical impulse tween the cells. Ordinary atrial muscle fibers
spreads along each cardiac cell, and rapidly participate in the propagation of the im-
10090-01_CH01.qxd 8/24/06 5:40 PM Page 23

Basic Cardiac Structure and Function 23

Membrane potential (mV)


0
Phase
0

–50
B
A
–100
Figure 1.17. Dependence of speed of depolarization on resting poten-
tial. A. Normal resting potential (RP) and rapid rise of phase 0. B. Less negative
RP results in slower rise of phase 0 and lower maximum amplitude of the ac-
tion potential.

pulse from the SA to the AV node, although EXCITATION-CONTRACTION


in certain regions the fibers are more densely COUPLING
arranged, facilitating conduction.
Fibrous tissue surrounds the tricuspid This section reviews how the electrical
and mitral valves, such that there is no action potential leads to physical contrac-
direct electrical connection between the tion of cardiac muscle cells, a process known
atrial and ventricular chambers other than as excitation-contraction coupling. During
through the AV node. As the electrical this process, chemical energy in the form of
impulse reaches the AV node, a delay in high-energy phosphate compounds is trans-
conduction (approximately 0.1 sec) is en- lated into the mechanical energy of myo-
countered. This delay occurs because the cyte contraction.
small-diameter fibers in this region conduct
slowly, and the action potential is of the
Contractile Proteins in
“slow” pacemaker type (recall that the fast
the Myocyte
sodium channels are permanently inacti-
vated in pacemaker tissues, such that the Several distinct proteins are responsible for
upstroke velocity relies on the slower cal- cardiac muscle cell contraction (Fig. 1.18). Fig. 18
cium channels). The pause in conduction at Two of the proteins, actin and myosin, are
the AV node is actually beneficial because it the chief contractile elements. Two other
allows the atria time to contract and fully proteins, tropomyosin and troponin, serve
empty their contents before ventricular regulatory functions.
stimulation. In addition, the delay allows Myosin is arranged in thick filaments,
the AV node to serve as a “gatekeeper” of each composed of lengthwise stacks of ap-
conduction from atria to ventricles, which proximately 300 molecules. The myosin
is critical for limiting the rate of ventricular filament exhibits globular heads that are
stimulation during abnormally rapid atrial evenly spaced along its length and contain
rhythms. myosin ATPase, an enzyme that is necessary
After traversing the AV node, the cardiac for contraction to occur. Actin, a smaller
action potential spreads into the rapidly molecule, is arranged in thin filaments as an
conducting bundle of His and Purkinje α-helix consisting of two strands that inter-
fibers, which distribute the electrical im- digitate between the thick myosin filaments
pulses to the bulk of the ventricular muscle (see Fig. 1.8). Titin (also termed connectin)
cells. This allows for precisely timed stimu- is a protein that helps tether myosin to the
lation and contraction of the ventricular Z line of the sarcomere and provides elastic-
myocytes. ity to the contractile process.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 24

24 Chapter One

Tn-C
Tn-I
Tn-T

Actin

Myosin
head Tropomyosin

Myosin thick filament

Figure 1.18. Schematic diagram of the main contractile proteins of the


myocyte, actin and myosin. Tropomyosin and troponin (components TnI,
TnC, and TnT) are regulatory proteins.

Tropomyosin is a double helix that lies as ryanodine receptors (Fig. 1.19). When Fig. 19
in the grooves between the actin filaments calcium enters the cell and binds to the
and, in the resting state, inhibits the inter- ryanodine receptor, the receptor undergoes
action between myosin heads and actin, a conformational change, which results in
thus preventing contraction. Troponin sits a much greater release of Ca++ into the
at regular intervals along the actin strands cytosol from the abundant stores in the
and is composed of three subunits. The tro- terminal cisternae of the SR. Thus, the ini-
AQ2 ponin T (TnT) subunit links the troponin tial L-type Ca++ current signal is amplified
complex to the actin and tropomyosin mol- by this mechanism, known as calcium-
ecules. The troponin I (TnI) subunit inhibits induced calcium release (CICR), and the
the ATPase activity of the actin-myosin in- cytosolic calcium concentration dramati-
teraction. The troponin C (TnC) subunit is cally increases.
responsible for binding calcium ions that As calcium ions bind to TnC, the activity
regulate the contractile process. of TnI is inhibited, which induces a confor-
mational change in tropomyosin. The latter
event exposes the active site between actin
Calcium-Induced Calcium Release
and myosin, enabling contraction to proceed.
and the Contractile Cycle
Contraction ensues as myosin heads
The sensitivity of troponin C to calcium es- bind to actin filaments and “flex,” thus
tablishes a crucial role for intracellular Ca++ causing the interdigitating thick and thin
ions in cellular contraction. The cycling of filaments to move past each other in an
calcium in and out of the cytosol during ATP-dependent reaction (Fig. 1.20). The Fig. 20
each action potential effectively couples elec- first step in this process is activation of
trical excitation to physical contraction. the myosin head by hydrolysis of ATP, fol-
Recall that during phase 2 of the action lowing which the myosin head binds to
potential, activation of L-type Ca++ chan- actin and forms a cross bridge. The inter-
nels results in an influx of Ca++ ions into the action between the myosin head and actin
myocyte. The small amount of calcium that results in a conformational change in the
enters the cell in this fashion is not suffi- head, causing it to pull the actin filament
cient to cause contraction of the myofibrils, inward.
but it triggers a much greater Ca++ release Next, while the myosin head and actin
from the sarcoplasmic reticulum (SR), as are still attached, ADP is released, and a new
follows: The T tubule invaginations of the molecule of ATP then binds to the myosin
sarcolemmal membrane bring the L-type head, causing it to release the actin filament.
channels into close apposition with special- The cycle can then repeat. Progressive cou-
ized Ca++ release receptors in the SR, known pling and uncoupling of actin and myosin
10090-01_CH01.qxd 8/24/06 5:40 PM Page 25

Basic Cardiac Structure and Function 25

Na+
Ca++
Outside cell
ATP
Inside cell
Ca++ Ca++
Ryanodine
Ca++ receptor
Binds to +
Tn-C

Contraction Ca++

PL

ATP Sarcoplasmic
Ca++ SERCA reticulum

Figure 1.19. Calcium ion movements during excitation and contraction in


cardiac muscle cells. Ca++ enters the cell through calcium channels during
phase 2 of the action potential, triggering a much larger calcium release from
the sarcoplasmic reticulum (SR) via the ryanodine receptor complex. The binding
of cytosolic Ca++ to troponin C (TnC) allows contraction to ensue. Relaxation
occurs as Ca++ is returned to the SR by sarco(endo)plasmic reticulum calcium
ATPase (SERCA). Phospholamban (PL) is a major regulator of this pump, inhibit-
ing Ca++ uptake in its dephosphorylated state. Excess intracellular calcium is re-
turned to the extracellular environment by sodium-calcium exchange and to a
smaller degree by the sarcolemmal Ca++-ATPase.

causes the muscle fiber to shorten by in- by the ATP-consuming calcium pump, Ca++- AQ3
creasing the overlap between the myofila- ATPase (see Fig. 1.10).
ments within each sarcomere. In the pres- As cytosolic Ca++ concentrations fall and
ence of ATP, this process continues for as calcium ions dissociate from troponin C,
long as the cytosolic calcium concentration tropomyosin once again inhibits the actin-
remains sufficiently high to inhibit the myosin interaction, leading to relaxation
troponin-tropomyosin blocking action. of the contracted cell. The contraction-
Myocyte relaxation, like contraction, is relaxation cycle can then repeat with the
synchronized with the electrical activity of next action potential.
the cell. Toward the end of phase 2 of the ac-
tion potential, L-type channels inactivate, ar-
b-Adrenergic and
resting the influx of Ca++ into the cell and
Cholinergic Signaling
abolishing the trigger for CICR. Concur-
rently, calcium is pumped back into the SR There is substantial evidence that the con-
and out of the cell. Calcium is sequestered centration of Ca++ within the cytosol is the
back into the SR primarily by sarco(endo)- major determinant of the force of cardiac
plasmic reticulum Ca11 ATPase (SERCA), as contraction with each heartbeat. Mecha-
shown in Figure 1.19. The small amount of nisms that raise intracellular Ca++ concentra-
Ca++ that entered the cell through L-type cal- tion enhance force development, whereas
cium channels is removed via the sarcolem- factors that lower Ca++ concentration reduce
mal Na+-Ca++ exchanger and to a lesser extent the contractile force.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 26

26 Chapter One

Actin

ATP
-ATP
ADP Myosin

A. Activation of myosin head


by ATP hydrolysis
D. ADP release, ATP binding,
actin filament release

-ADP -ADP-Pi

C. Phosphate release
and power stroke B. Cross bridge formation
between myosin head
and actin filament

Pi
-ADP-Pi

Figure 1.20. The contractile process. A. Myosin head is activated by hydrolysis of ATP.
B. During cellular depolarization, cytoplasmic calcium concentration increases and removes the
troponin-tropomyosin inhibition, such that a cross bridge is formed between actin and myosin.
C. Inorganic phosphate (Pi), is released and a conformational change in the myosin head draws
the actin filament inward. D. ADP is released and replaced by ATP, causing the myosin head to
dissociate from the actin filament. As the process repeats, the muscle fiber shortens. The cycle
continues until cytosolic calcium concentration decreases at the end of phase 2 of the action
potential.

β-Adrenergic stimulation is one mechanism β-Adrenergic stimulation of the myocyte


that enhances calcium fluxes in the myocyte also enhances myocyte relaxation. The re-
and thereby strengthens the force of ventric- turn of Ca++ from the cytosol to the sar-
Fig. 21 ular contraction (Fig. 1.21). Catecholamines coplasmic reticulum (SR) is regulated by
(e.g., norepinephrine) bind to the myocyte phospholamban (PL), a low molecular
β1-adrenergic receptor, which is coupled to weight protein in the SR membrane. In its
and activates the G protein system (Gs) at- dephosphorylated state, PL inhibits Ca++
tached to the inner surface of the cell mem- uptake by SERCA (see Fig. 1.19). However,
brane. Gs in turn stimulates membrane- β-adrenergic activation of protein kinases
bound adenylate cyclase to produce cyclic (see Fig. 1.21) causes PL to become phos-
AMP (cAMP) from ATP. cAMP then activates phorylated, an action that blunts PL’s in-
intracellular protein kinases, which phos- hibitory effect. The subsequently greater
phorylate cellular proteins, including the uptake of calcium ions by the SR hastens
L-type calcium channels within the cell Ca++ removal from the cytosol, promoting
membrane. Phosphorylation of the calcium myocyte relaxation. The increased cAMP
channel augments Ca++ influx, which trig- activity also results in phosphorylation of
gers a corresponding increase in Ca++ release TnI, an action that inhibits actin-myosin
from the sarcoplasmic reticulum, thereby interactions and therefore further enhances
enhancing the force of contraction. relaxation of the cell.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 27

Basic Cardiac Structure and Function 27

Norepinephrine Acetylcholine

β1-andrenergic Muscarinic
receptor receptor
Adenylate Ca++
cyclase
+ –

+
Gs protein G1 protein
ATP
cAMP

Inactive
protein Active Ca++
kinases protein
kinases PL

+ Sarcoplasmic
Ca++ ATP reticulum

Figure 1.21. Effects of b-adrenergic and cholinergic stimulation on cardiac cellular


signaling and calcium ion movement. The binding of a ligand (e.g., norepinephrine) to
the β1-adrenergic receptor induces G protein–mediated stimulation of adenylate cyclase
and formation of cyclic AMP (cAMP). The latter activates protein kinases, which phosphor-
ylate cellular proteins, including ion channels. Phosphorylation of the slow Ca++ channel
enhances calcium movement into the cell and therefore strengthens the force of contrac-
tion. Protein kinases also phosphorylate phospholamban (PL), reducing the latter’s inhibi-
tion of Ca++ uptake by the sarcoplasmic reticulum. The enhanced removal of Ca++ from the
cytosol facilitates relaxation of the myocyte. Cholinergic signaling, triggered by acetyl-
choline binding to the muscarinic receptor, activates inhibitory G proteins that reduce
adenylate cyclase activity and cAMP production, thus antagonizing the effects of
β-adrenergic stimulation.

Cholinergic signaling via parasympathetic Thus, physiologic or pharmacologic cate-


inputs (mainly from the vagus nerve) op- cholamine stimulation of the myocyte β1-
poses the effects of β-adrenergic stimulation adrenergic receptor enhances contraction of
(see Fig. 1.21). Acetylcholine released from the cell, while cholinergic stimulation op-
parasympathetic nerve terminals binds to the poses that enhancement. These important
muscarinic M2 receptor on cardiac cells. This properties will be referred to in later chapters.
receptor also activates G proteins, but in dis-
tinction to the β-adrenergic receptor, it is
SUMMARY
coupled to Gi, an inhibitory G protein system.
Gi associated with cholinergic stimulation in- The anatomic structure, cellular composi-
hibits adenylate cyclase activity and reduces tion, and conduction pathways of the heart
cAMP formation. Gi also activates specific K+ form an efficient system for repetitive, or-
channels in the plasma membrane, which ganized contractions. As a result, the heart
hyperpolarizes the cell. At the sinus node, is capable of purposeful stimulation bil-
these actions of cholinergic stimulation serve lions of times during the lifespan of a nor-
to reduce heart rate. In the myocardium, the mal person. With each contraction cycle,
effect is to counteract the force of contraction the heart receives and propagates blood
induced by β-adrenergic stimulation. It through the circulation to provide nutri-
should be noted that ventricular cells are ents to and remove waste products from
much less sensitive to this cholinergic effect the body’s tissues.
than atrial cells, likely reflecting different de- The following chapters explore what can
grees of G protein coupling. go wrong with this remarkable system.
10090-01_CH01.qxd 8/24/06 5:40 PM Page 28

28 Chapter One

Acknowledgments Lohse MJ, Engelhardt S, Eschenhagen T. What is the


role of beta-adrenergic signaling in heart failure?
Contributors to the previous editions of this chap- Circ Res 2003;93:896–906.
ter were Kirsten Greineder, MD; Stephanie Harper, Opie LH. Heart Physiology, from Cell to Circulation.
MD; Scott Hyver, MD; Paul Kim, MD; Rajeev 4th Ed. Philadelphia: Lippincott Williams &
Malhotra, MD; Laurence Rhines, MD; James D. Wilkins, 2004.
Marsh, MD; Gary R. Strichartz, MD; and Leonard S. Rockman HA, Koch WJ, Lefkowitz RJ. Seven-
Lilly, MD. transmembrane-spanning receptors and heart
function. Nature 2002;415:206.
Additional Reading Wilcox BR, Cook AC, Anderson RH. Surgical Anatomy
of the Heart. Cambridge, MA: Cambridge Univer-
Bers DM. Cardiac excitation-contraction coupling. sity Press, 2005.
Nature 2002;415:198–205. Zipes DP, Jalife J, eds. Cardiac Electrophysiology:
Katz AM. Physiology of the Heart. 4th Ed. Philadel- From Cell to Bedside. 4th Ed. Philadelphia: Saun-
phia: Lippincott Williams & Wilkins, 2006. ders, 2004.
10090-01_CH01.qxd 8/24/06 5:40 PM Page a

Chapter 1—Author Queries


2. AU: Abbreviations changed per several Internet sources and Chapter 7.
3. AU: As on p. 22. Correct?
4. AU: ED: Permission still pending?

Chapter 1(Box)—Author Query


1. AU: Plural correct, as above?
10090-02_CH02.qxd 8/24/06 5:41 PM Page 29

C H A P T E R

The Cardiac Cycle:


Mechanisms of Heart
Sounds and Murmurs
2
Nicole Martin
Leonard S. Lilly

CARDIAC CYCLE MURMURS


Systolic Murmurs
HEART SOUNDS
Diastolic Murmurs
First Heart Sound (S1)
Continuous Murmurs
Second Heart Sound (S2)
Extra Systolic Heart Sounds
Extra Diastolic Heart Sounds

Cardiac diseases often cause abnormal CARDIAC CYCLE


findings on physical examination, includ-
ing pathologic heart sounds and murmurs. The cardiac cycle consists of precisely timed
These findings are clues to the underlying electrical and mechanical events that are re-
pathophysiology, and proper interpreta- sponsible for rhythmic atrial and ventricu-
tion is essential for successful diagnosis lar contractions. Figure 2.1 displays the pres- Fig. 1
and disease management. This chapter de- sure relationships between the left-sided
scribes heart sounds in the context of the cardiac chambers during the normal cardiac
normal cardiac cycle and then focuses on cycle and serves as a platform for describing
the origins of pathologic heart sounds and key events. Mechanical systole refers to ven-
murmurs. tricular contraction, and diastole to ven-
Many cardiac diseases are mentioned tricular relaxation and filling. Throughout
briefly in this chapter as examples of abnor- the cardiac cycle, the right and left atria
mal heart sounds and murmurs. Because accept blood returning to the heart from
each of these conditions is described in the systemic veins and from the pulmonary
greater detail later in the book, it is not nec- veins, respectively. During diastole, blood
essary to memorize the examples presented passes from the atria into the ventricles
here. Rather, it is preferable to understand across the open tricuspid and mitral valves,
the mechanisms by which the abnormal causing a gradual increase in ventricular di-
sounds are produced, so that their descrip- astolic pressures. In late diastole, atrial con-
tions will make sense in later chapters. traction propels a final bolus of blood into

29
10090-02_CH02.qxd 8/24/06 5:41 PM Page 30

30 Chapter Two

ECG
the mitral component slightly precedes that
of the tricuspid valve because of the earlier
electrical stimulation of left ventricular con-
AV closes

AV opens
traction (see Chapter 4).
As the right and left ventricular pressures
100
Pressure (mm Hg)

Aorta rapidly rise further, they soon exceed the


diastolic pressures within the pulmonary
artery and aorta, forcing the pulmonic and
LV aortic valves to open, and blood is ejected
into the pulmonary and systemic circula-
50
tions. The ventricular pressures continue to
MV opens increase during contraction, and because the
MV closes
pulmonic and aortic valves are open, the aor-
tic and pulmonary artery pressures rise, par-
LA c v
a allel to those of the corresponding ventricles.
Time At the conclusion of ventricular ejection,
the ventricular pressures fall below those of
the pulmonary artery and aorta (the pul-
monary artery and aorta are elastic struc-
S1 S2 tures that maintain their pressures longer),
DIASTOLE SYSTOLE DIASTOLE such that the pulmonic and aortic valves are
forced to close, producing the second heart
Figure 2.1. The normal cardiac cycle, showing pres-
sure relationships between the left-sided heart
sound, S2. Like the first heart sound (S1), this
chambers. During diastole, the mitral valve (MV) is sound consists of two parts: the aortic (A2)
open, so that the left atrial (LA) and left ventricular (LV) component normally precedes the pulmonic
pressures are equal. In late diastole, LA contraction
causes a small rise in pressure in both the LA and LV (the
(P2) because the diastolic pressure gradient
a wave). During systolic contraction, the LV pressure between the aorta and left ventricle is
rises; when it exceeds the LA pressure, the MV closes, greater than that between the pulmonary
contributing to the first heart sound (S1). As LV pressure
rises above the aortic pressure, the aortic valve (AV)
artery and right ventricle, forcing the aortic
opens, which is a silent event. As the ventricle begins to valve to shut more readily. The ventricular
relax and its pressure falls below that of the aorta, the pressures fall rapidly during the subsequent
AV closes, contributing to the second heart sound (S2).
As LV pressure falls further, below that of the LA, the
relaxation phase. As they drop below the
MV opens, which is silent in the normal heart. In addi- pressures in the right and left atria, the tri-
tion to the a wave, the LA pressure curve displays two cuspid and mitral valves open, followed by
positive deflections: the c wave represents a small rise in
LA pressure as the MV closes and bulges toward the
diastolic ventricular filling and repetition of
atrium, and the v wave is the result of passive filling of this cycle.
the LA from the pulmonary veins during systole, when Notice in Figure 2.1 that in addition to
the MV is closed.
the a wave, the atrial pressure curve displays
two other positive deflections during the
cardiac cycle: The c wave represents a small
each ventricle, an action that produces a rise in atrial pressure as the tricuspid and mi-
brief further rise in atrial and ventricle pres- tral valves close and bulge into their respec-
sures, termed the a wave (see Fig. 2.1). tive atria. The v wave is the result of passive
Contraction of the ventricles follows, sig- filling of the atria from the systemic and
naling the onset of mechanical systole. As pulmonary veins during systole, a period
the ventricles start to contract, the pressures during which blood accumulates in the atria
within them rapidly exceed atrial pressures. because the tricuspid and mitral valves are
This results in the forced closure of the tri- closed.
cuspid and mitral valves, which produces At the bedside, systole can be approxi-
the first heart sound, termed S1. This sound mated by the period from S1 to S2, and dias-
has two nearly superimposed components: tole from S2 to the next S1. Although the du-
10090-02_CH02.qxd 8/24/06 5:41 PM Page 31

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 31

ration of systole remains constant from beat the left side of the heart. Equivalent events
to beat, the length of diastole varies with the occur simultaneously in the right side of the
heart rate: the faster the heart rate, the shorter heart in the right atrium, right ventricle,
the diastolic phase. The main sounds, S1 and and pulmonary artery. At the bedside, clues
S2, provide a framework from which all other to right-heart function can be ascertained by
heart sounds and murmurs can be timed. examining the jugular venous pulse, which
The pressure relationships and events de- is representative of the right atrial pressure
picted in Figure 2.1 are those that occur in (see Box 2.1). Box 1

Box 2.1 Jugular Venous Pulsations and Assessment of Right-Heart Function

Bedside observation of jugular venous pulsations in the neck is a vital part of the car-
diovascular examination. With no structures impeding blood flow between the internal
jugular (IJ) veins and the superior vena cava and right atrium (RA), the height of the IJ
venous column (termed the jugular venous pressure, or JVP) is an accurate representa-
tion of the RA pressure. Thus, the JVP provides an easily obtainable measure of right-
heart function.
Typical fluctuations in the jugular ve- a
nous pulse during the cardiac cycle, man-
ifested by oscillations in the overlying v
skin, are shown in the figure (notice the
similarity to the left atrial pressure tracing y
x
in Fig. 2.1). There are two major upward
components, the a and v waves, fol-
lowed by two descents, termed x and y.
The x descent, which represents the pressure decline following the a wave, may be inter-
rupted by a small upward deflection (the c wave, denoted in the figure by the arrow) at
the time of tricuspid valve closure, but that is usually not distinguishable in the JVP. The a
wave represents transient venous distension caused by back pressure from RA contrac-
tion. The v wave corresponds to passive filling of the RA from the systemic veins during
systole, when the tricuspid valve is closed. Opening of the tricuspid valve in early diastole
allows blood to rapidly empty from the RA into the right ventricle; that fall in RA pressure
corresponds to the y descent.
Conditions that abnormally raise right-sided cardiac pressures (e.g., heart failure, tri-
cuspid valve disease, pulmonic stenosis, pericardial diseases) elevate the JVP, while re-
duced intravascular volume (e.g., dehydration) decreases it. In addition, specific disease
states can influence the individual components of the JVP, examples of which are listed
here for reference and explained in subsequent chapters:
Prominent a: right ventricular hypertrophy, tricuspid stenosis
Prominent v: tricuspid regurgitation
Prominent y: constrictive pericarditis

Technique of Measurement
The JVP is measured as the maximum vertical height of the internal jugular vein (in cm)
above the center of the right atrium, and in a normal person is ≤9 cm. Because the ster-
nal angle is located approximately 5 cm above the center of the RA, the JVP is calculated
10090-02_CH02.qxd 8/24/06 5:41 PM Page 32

32 Chapter Two

at the bedside by adding 5 cm to the vertical height of the top of the IJ venous column
above the sternal angle.
The right IJ vein is usually the easiest to evaluate because it extends directly upward
from the RA and superior vena cava. First, observe the pulsations in the skin overlying the
IJ with the patient supine and the head of the bed at about a 45° angle. Shining a light
obliquely across the neck helps to visualize the pulsations. Be sure to examine the IJ, not
the external jugular vein. The former is medial to, or behind, the sternocleidomastoid mus-
cle, while the external jugular is usually more lateral. Although the external jugular is typ-
ically easier to see, it does not accurately reflect RA pressure because it contains valves that
interfere with venous return to the heart.
If the top of the IJ column is not visible at 45°, the column of blood is either too low
(below the clavicle) or too high (above the jaw) to be measured in that position. In such
situations, the head of the bed must be lowered or raised, respectively, so that the top of
the column becomes visible. As long as the top can be ascertained, the vertical height
of the JVP above the sternal angle will accurately reflect RA pressure, no matter the angle
of the head of the bed.
Sometimes it can be difficult to distinguish the jugular venous pulsations from the
neighboring carotid artery. Unlike the carotid, the JVP is usually not palpable, it has a dou-
ble rather than a single upstroke, and it declines in most patients by assuming the seated
position or during inspiration.

HEART SOUNDS ates only a single sound. An exception oc-


curs in patients with right bundle branch
Typical stethoscopes contain two chest pieces block (see Chapter 4), in whom these com-
for auscultation of the heart. The concave ponents may be audibly split because of de-
“bell” chest piece, meant to be applied lightly layed closure of the tricuspid valve.
to the skin, accentuates low-frequency sounds. Three factors determine the intensity of S1:
Conversely, the flat “diaphragm” chest piece (1) the distance separating the leaflets of the
should be pressed firmly against the skin to open valves at the onset of ventricular con-
eliminate low frequencies and therefore ac- traction; (2) the mobility of the leaflets (nor-
centuate high-frequency sounds and mur- mal, or rigid because of stenosis); and (3) the
murs. Some modern stethoscopes incorpo- rate of rise of ventricular pressure (Table 2.1). Tab. 1
rate both the bell and diaphragm functions
into a single chest piece; in these models,
placing the piece lightly on the skin brings
out the low-frequency sounds, while firm
pressure accentuates the high-frequency
ones.

First Heart Sound (S1)


S1 is produced by closure of the mitral and
tricuspid valves in early systole and is loud-
Fig. 2 est near the apex of the heart (Fig. 2.2). It is
a high-frequency sound, best heard with the
diaphragm of the stethoscope. Although
mitral closure usually precedes tricuspid
closure, they are separated by only about Figure 2.2. Standard positions of stethoscope place-
0.01 sec, such that the human ear appreci- ment for cardiac auscultation.
10090-02_CH02.qxd 8/24/06 5:41 PM Page 33

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 33

a diminished S1 results from an abnormally


TABLE 2.1. Causes of Altered Intensity of First prolonged PR interval, which delays the
Heart Sound (S1)
onset of ventricular contraction. Following
Accentuated S1 atrial contraction, the mitral and tricuspid
valves have additional time to float back to-
1. Shortened PR interval
2. Mild mitral stenosis
gether so that the leaflets are forced closed
3. High cardiac output states or tachycardia from only a small distance apart.
(e.g., exercise or anemia) In patients with mitral regurgitation (see
Diminished S1 Chapter 8), S1 is often diminished in inten-
sity because the mitral leaflets may not come
1. Lengthened PR interval: first-degree AV nodal into full contact with one another as they
block close. In severe mitral stenosis, the leaflets are
2. Mitral regurgitation
3. Severe mitral stenosis
nearly fixed in position throughout the car-
4. “Stiff” left ventricle (e.g., systemic hypertension) diac cycle, and that reduced movement
lessens the intensity of S1.
AV, atrioventricular.
In patients with a “stiffened” left ventri-
cle (e.g., a hypertrophied chamber), atrial
The distance between the open valve leaf- contraction results in a higher-than-normal
lets at the onset of ventricular contraction pressure at the end of diastole. This greater
relates to the electrocardiographic PR inter- pressure causes the mitral leaflets to drift to-
val (see Chapter 4), the period between the gether more rapidly, forcing them closed
onset of atrial and ventricular activation. from a smaller-than-normal distance when
Atrial contraction at the end of diastole ventricular contraction begins and thus re-
forces the tricuspid and mitral valve leaflets ducing the intensity of S1.
apart. As they start to drift back together,
ventricular contraction forces them shut, Second Heart Sound (S2)
from whatever position they are at, as soon
as the ventricular pressure exceeds that in The second heart sound results from the clo-
the atrium. An accentuated S1 results when sure of the aortic and pulmonic valves and
the PR interval is shorter than normal be- therefore has aortic (A2) and pulmonic (P2)
cause the valve leaflets do not have suffi- components. Unlike S1, which is usually
cient time to drift back together and are heard as a single sound, the components of
therefore forced shut from a relatively wide S2 vary with the respiratory cycle: they are
distance. normally fused as one sound during expira-
Similarly, in mild mitral stenosis (see tion but become audibly separated during
Chapter 8) a prolonged diastolic pressure inspiration, a situation termed normal or
gradient exists between the left atrium and physiologic splitting (Fig. 2.3). Fig. 3
ventricle, which keeps the mobile portions One explanation for normal splitting of
of the mitral leaflets farther apart than nor- S2 is as follows. Expansion of the chest dur-
mal during diastole. Because the leaflets are ing inspiration causes the intrathoracic pres-
relatively wide apart at the onset of systole, sure to become more negative. The negative
they are forced shut loudly when the left pressure transiently increases the capaci-
ventricle contracts. tance (and reduces the impedance) of the in-
S1 also may be accentuated when the trathoracic pulmonary vessels. As a result,
heart rate is more rapid than normal (i.e., there is a temporary delay in the diastolic
tachycardia) because diastole is shortened “back pressure” of the pulmonary artery re-
and the leaflets have insufficient time to drift sponsible for closure of the pulmonic valve.
back together before the ventricles contract. Thus, P2 is delayed; that is, it occurs later
Conditions that reduce the intensity of S1 during inspiration than during expiration.
are also listed in Table 2.1. In first-degree Inspiration has the opposite effect on A2.
atrioventricular (AV) block (see Chapter 12), Because the capacity of the intrathoracic
10090-02_CH02.qxd 8/24/06 5:41 PM Page 34

34 Chapter Two

A. Physiologic (normal) P2
splitting

B. Widened splitting P2

P2
C. Fixed splitting

P2
D. Paradoxical splitting
(Note reversed position of
A2 and P2)

Figure 2.3. Splitting patterns of the second heart sound (S2). A2, aortic component; P2, pulmonic component
of S2; S1, first heart sound.
10090-02_CH02.qxd 8/24/06 5:41 PM Page 35

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 35

pulmonary veins is increased by the nega- that condition, chronic volume overload of
tive pressure generated by inspiration, the the right-sided circulation results in a high-
venous return to the left atrium and ventri- capacitance, low-resistance pulmonary vas-
cle temporarily decreases. Reduced filling of cular system. This alteration in pulmonary
the LV causes a reduced stroke volume dur- artery hemodynamics delays the back pres-
ing the next systolic contraction and there- sure responsible for closure of the pulmonic
fore shortens the time required for LV emp- valve. Thus, P2 occurs later than normal,
tying. Therefore, aortic valve closure (A2) even during expiration, such that there is
occurs slightly earlier in inspiration than wider than normal separation of A2 and P2.
during expiration. The combination of an The pattern of splitting does not change (i.e.,
earlier A2 and delayed P2 during inspiration it is fixed) during the respiratory cycle be-
causes audible separation of the two com- cause (1) inspiration does not substantially
ponents. Since these components are high- increase further the already elevated pul-
frequency sounds, they are best heard with monary vascular capacitance, and (2) aug-
the diaphragm of the stethoscope, and split- mented filling of the right atrium from the
ting of S2 is usually most easily appreciated systemic veins during inspiration is counter-
near the second left intercostal space next to balanced by a reciprocal decrease in the left-
the sternum (the pulmonic area). to-right transatrial shunt, eliminating respi-
Abnormalities of S2 include alterations in ratory variations in right ventricular filling.
its intensity and changes in the pattern of Paradoxical splitting (or reversed split-
splitting. The intensity of S2 depends on the ting) refers to audible separation of A2 and P2
velocity of blood coursing back toward the during expiration that disappears on inspira-
valves from the aorta and pulmonary artery tion, the opposite of the normal situation. It
after the completion of ventricular contrac- reflects an abnormal delay in the closure of
tion, and the suddenness with which that the aortic valve such that P2 precedes A2. In
motion is arrested by the closing valves. In adults, the most common cause is left bun-
systemic hypertension or pulmonary arte- dle branch block (LBBB). In LBBB, the spread
rial hypertension, the diastolic pressure in of electrical activity through the left ventri-
the respective great artery is higher than cle is impaired, resulting in delayed ventric-
normal, such that the velocity of the blood ular contraction and late closure of the aor-
surging toward the valve is elevated and S2 is tic valve such that it follows P2. During
accentuated. Conversely, in severe aortic or inspiration, as in the normal case, the pul-
pulmonic valve stenosis, the valve commis- monic valve closure sound is delayed and
sures are nearly fixed in position, such that the aortic valve closure sound moves earlier.
the contribution of the stenotic valve to S2 is This results in narrowing and often superim-
diminished. position of the two sounds; thus, there is no
Widened splitting of S2 refers to an in- apparent split at the height of inspiration
crease in the time interval between A2 and P2, (see Fig. 2.3). In addition to LBBB, paradoxi-
such that the two components are audibly cal splitting may be observed under circum-
separated even during expiration and become stances in which left ventricular ejection is
more widely separated in inspiration (see Fig. greatly prolonged, such as aortic stenosis.
2.3). This pattern is usually the result of de-
layed closure of the pulmonic valve, which
Extra Systolic Heart Sounds
occurs in right bundle branch block and pul-
monic valve stenosis. Extra systolic heart sounds may occur in
Fixed splitting of S2 is an abnormally early, mid-, or late systole.
widened interval between A2 and P2 that per-
sists unchanged through the respiratory
Early Extra Systolic Heart Sounds
cycle (see Fig. 2.3). The most common ab-
normality that causes fixed splitting of S2 is Abnormal early systolic sounds, or ejection
an atrial septal defect (see Chapter 16). In clicks, occur shortly after S1 and coincide with
10090-02_CH02.qxd 8/24/06 5:41 PM Page 36

36 Chapter Two

the opening of the aortic or pulmonic valves the aortic or pulmonic root with the onset
Fig. 4 (Fig. 2.4). These sounds have a sharp, high- of blood flow into the vessel. The aortic ejec-
pitched quality, so they are heard best with tion click is heard at both the base and the
the diaphragm of the stethoscope placed apex of the heart and does not vary with res-
over the aortic and pulmonic areas. Ejection piration. In contrast, the pulmonic ejection
clicks indicate the presence of aortic or pul- click is heard only at the base and its inten-
monic valve stenosis or dilatation of the pul- sity diminishes during inspiration (see Chap-
monary artery or aorta. In stenosis of the aor- ter 16).
tic or pulmonic valve, the sound occurs as
the valve leaflets reach their maximal level of
Mid- or Late Extra Systolic Heart Sounds
ascent into the great artery, just prior to
blood ejection. At that moment, the rapidly Clicks occurring in mid- or late systole are
ascending valve reaches its elastic limit and usually the result of systolic prolapse of
decelerates abruptly, an action thought to re- the mitral or tricuspid valves, in which
sult in the sound generation. In dilatation of the leaflets bulge abnormally from the ven-
the root of the aorta or pulmonary artery, the tricular side of the atrioventricular junction
sound is associated with sudden tensing of into the atrium during ventricular contrac-
tion, often accompanied by valvular regur-
gitation. They are loudest over the mitral or
tricuspid auscultatory regions, respectively.
ECG

Extra Diastolic Heart Sounds


Extra heart sounds in diastole include the
Aorta opening snap (OS), the third heart sound
(S3), the fourth heart sound (S4), and the
LV pericardial knock.

MV opens Opening Snap


Opening of the mitral and tricuspid valves is
LA normally silent, but mitral or tricuspid valvu-
lar stenosis (usually the result of rheumatic
heart disease; see Chapter 8) produces a
sound, termed a snap, when the affected valve
opens. It is a sharp, high-pitched sound, and
S4 S1 S2 S3 its timing does not vary significantly with
Ejection OS
respiration. In mitral stenosis (which is much
click
more common than tricuspid valve steno-
Figure 2.4. Timing of extra systolic sis), the OS is heard best between the apex
and diastolic heart sounds. S4 is pro-
duced by atrial contraction into a “stiff” and the left sternal border, just after the aor-
left ventricle (LV). An ejection click fol- tic closure sound (A2), when the left ventric-
lows the opening of the aortic or pul- ular pressure falls below that of the left
monic valve in cases of valve stenosis or
dilatation of the corresponding great atrium (see Fig. 2.4).
artery. S3 occurs during the period of Because of its proximity to A2, the A2–OS
rapid ventricular filling; it is normal in sequence can be confused with a widely
young people, but its presence in adults
implies LV contractile dysfunction. The split second heart sound. However, careful
timing of an opening snap (OS) is placed auscultation at the pulmonic area during
for comparison, but it is not likely that all inspiration reveals three sounds occurring
of these sounds would appear in the
same person. LA, left atrium; MV, mitral in rapid succession (Fig. 2.5), which corre- Fig. 5
valve. spond to aortic closure (A2), pulmonic clo-
10090-02_CH02.qxd 8/24/06 5:41 PM Page 37

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 37

mitral stenosis, the opening snap is widely


separated from A2, whereas in more severe
stenosis, the A2–OS interval is narrower.

Expiration Third Heart Sound (S3)


OS When present, an S3 occurs in early diastole,
following the opening of the atrioventricu-
lar valves, during the ventricular rapid fill-
ing phase (see Fig. 2.4). It is a dull, low-
pitched sound best heard with the bell of
S1 S2
the stethoscope. A left-sided S3 is typically
loudest over the cardiac apex while the pa-
tient lies in the left lateral decubitus posi-
tion. A right-sided S3 is better appreciated
at the lower-left sternal border. Production
of the S3 appears to result from tensing of
the chordae tendineae during rapid filling
and expansion of the ventricle.
A third heart sound is a normal finding in
Inspiration children and young adults. In these groups,
OS
an S3 implies the presence of a supple ven-
tricle capable of normal rapid expansion in
early diastole. Conversely, when heard in
middle-aged or older adults, an S3 is often a
sign of disease, indicating volume overload
A2P2 owing to congestive heart failure, or the in-
Figure 2.5. Timing of the opening snap (OS) in mitral creased transvalvular flow that accompanies
stenosis does not change with respiration. On inspi- advanced mitral or tricuspid regurgitation.
ration, normal splitting of the second heart sound (S2) is
observed so that three sounds are heard. A2, aortic com-
A pathologic S3 is sometimes referred to as a
ponent; P2, pulmonic component of S2; S1, first heart ventricular gallop.
sound.

Fourth Heart Sound (S4)


sure (P2), and then the opening snap (OS). When an S4 is present, it occurs in late di-
The three sounds become two on expiration astole and coincides with contraction of
because A2 and P2 normally fuse. the atria (see Fig. 2.4). This sound is gener-
The severity of stenosis can be approxi- ated by the left (or right) atrium vigorously
mated by the time interval between A2 and contracting against a stiffened ventricle.
the opening snap: the more advanced the Thus, an S4 usually indicates the presence
stenosis, the shorter the interval. This oc- of cardiac disease—specifically, a decrease
curs because the degree of left atrial pressure in ventricular compliance typically result-
elevation corresponds to the severity of mi- ing from ventricular hypertrophy or myo-
tral stenosis. When the ventricle relaxes in cardial ischemia. Like an S3, the S4 is a dull,
diastole, the greater the left atrial pressure, low-pitched sound and is best heard with
the earlier the mitral valve opens. Com- the bell of the stethoscope. In the case
pared with severe stenosis, mild disease is of the more common left-sided S4, the
marked by a less elevated left atrial pressure sound is loudest at the apex, with the pa-
is less elevated, lengthening the time it tient lying in the left lateral decubitus
takes for the left ventricular pressure to fall position. S4 is sometimes referred to as an
below that of the atrium. Therefore, in mild atrial gallop.
10090-02_CH02.qxd 8/24/06 5:41 PM Page 38

38 Chapter Two

Quadruple Rhythm or Summation Gallop Murmurs are described by their timing,


intensity, pitch, shape, location, radiation,
In a patient with both an S3 and S4, those
and response to maneuvers. Timing refers to
sounds, in conjunction with S1 and S2, pro-
whether the murmur occurs during systole
duce a quadruple beat. If a patient with a
or diastole or is continuous (i.e., begins in
quadruple rhythm develops tachycardia, di-
systole and continues into diastole). The in-
astole becomes shorter in duration, the S3
tensity of the murmur is typically quantified
and S4 coalesce, and a summation gallop
by a grading system. In the case of systolic
results. The summation of S3 and S4 is heard
murmurs:
as a long middiastolic, low-pitched sound,
often louder than S1 and S2.

Grade 1/6 (or I/VI): Barely audible (i.e., med-


Pericardial Knock ical students may not
hear it!)
A pericardial knock is an uncommon, high- Grade 2/6 (or II/VI): Faint but immediately
pitched sound that occurs in patients with audible
severe constrictive pericarditis (see Chapter Grade 3/6 (or III/VI): Easily heard
14). It appears early in diastole soon after Grade 4/6 (or IV/VI): Easily heard and asso-
S2 and can be confused with an opening ciated with a palpable
snap or an S3. However, the knock appears thrill
slightly later in diastole than the timing of Grade 5/6 (or V/VI): Very loud; heard with
an opening snap and is louder and occurs stethoscope lightly on
earlier than the ventricular gallop. It results chest
Grade 6/6 (or VI/VI): Audible without the
from the abrupt cessation of ventricular fill-
stethoscope directly on
ing in early diastole, which is the hallmark
the chest wall
of constrictive pericarditis.

MURMURS And in the case of diastolic murmurs:


A murmur is the sound generated by turbu-
lent blood flow. Under normal conditions,
the movement of blood through the vascular Grade 1/4 (or I/IV): Barely audible
bed is laminar, smooth and silent. However, Grade 2/4 (or II/IV): Faint but immediately
as a result of hemodynamic and/or structural audible
changes, laminar flow can become disturbed Grade 3/4 (or III/IV): Easily heard
Grade 4/4 (or IV/IV): Very loud
and produce an audible noise. Murmurs re-
sult from any of the following mechanisms:
1. Flow across a partial obstruction
(e.g., aortic stenosis) Pitch refers to the frequency of the murmur,
ranging from high to low. High-frequency
2. Increased flow through normal structures
murmurs are caused by large pressure gra-
(e.g., aortic systolic murmur associated
dients between chambers (e.g., aortic steno-
with a high-output state, such as anemia)
sis) and are best appreciated using the di-
3. Ejection into a dilated chamber (e.g., aphragm chest piece of the stethoscope.
aortic systolic murmur associated with Low-frequency murmurs imply less of a pres-
aneurysmal dilatation of the aorta) sure gradient between chambers (e.g., mitral
4. Regurgitant flow across an incompetent stenosis) and are best heard using the stetho-
valve (e.g., mitral regurgitation) scope’s bell piece.
5. Abnormal shunting of blood from one Shape describes how the murmur changes
vascular chamber to a lower-pressure in intensity from its onset to its comple-
chamber (e.g., ventricular septal defect) tion. For example, a crescendo–decrescendo
10090-02_CH02.qxd 8/24/06 5:41 PM Page 39

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 39

(or “diamond-shaped”) murmur first rises right, Valsalva (forceful expiration against a
and then falls off in intensity. Other shapes closed airway), or clenching of the fists, each
include decrescendo (i.e., the murmur begins of which alters the heart’s loading condi-
at its maximum intensity and grows softer) tions and can affect the intensity of many
and uniform (the intensity of the murmur murmurs. Examples of the effects of maneu-
does not change). vers on specific murmurs are presented in
Location refers to the murmur’s region of Chapter 8.
maximum intensity and is usually described When reporting a murmur, some or all of
in terms of specific auscultatory areas (see these descriptors are mentioned. For exam-
Fig. 2.2): ple, you might describe a particular patient’s
murmur of aortic stenosis as “A grade III/
VI high-pitched, crescendo–decrescendo sys-
tolic murmur, heard best at the upper-right
Aortic area: Second to third right inter-
sternal border, radiating toward the neck.”
costal spaces, next to sternum
Pulmonic area: Second to third left intercostal
spaces, next to sternum Systolic Murmurs
Tricuspid area: Lower-left sternal border
Mitral area: Cardiac apex Systolic murmurs are subdivided into sys-
tolic ejection murmurs, pansystolic mur-
murs, and late systolic murmurs (Fig. 2.6). A Fig. 6
systolic ejection murmur is typical of aor-
From their primary locations, murmurs are tic or pulmonic valve stenosis. It begins after
often heard to radiate to other areas of the the first heart sound and terminates before
chest, and such patterns of transmission re- or during S2, depending on its severity and
late to the direction of the turbulent flow. whether the obstruction is of the aortic or
Finally, similar types of murmurs can be pulmonic valve. The shape of the murmur is
distinguished from one another by simple of the crescendo–decrescendo type (i.e., its
bedside maneuvers, such as standing up- intensity rises and then falls).

Click

Figure 2.6. Classification of systolic murmurs. Ejection murmurs are


crescendo–decrescendo in configuration, whereas pansystolic murmurs are
uniform throughout systole. A late systolic murmur often follows a midsys-
tolic click and suggests mitral (or tricuspid) valve prolapse.
10090-02_CH02.qxd 8/24/06 5:41 PM Page 40

40 Chapter Two

The ejection murmur of aortic stenosis be-


gins in systole after S1, from which it is sep-
Fig. 7 arated by a short audible gap (Fig. 2.7). This
gap corresponds to the period of isovolu-
metric contraction of the left ventricle (the
period after the mitral valve has closed but
before the aortic valve has opened). The
S1 A 2 P2
murmur becomes more intense as flow in-
creases across the aortic valve during the
rise in left ventricular pressure (crescendo).
Then, as the ventricle relaxes, forward flow
decreases, and the murmur lessens in in-
tensity (decrescendo) and finally ends prior
to the aortic component of S2. The murmur
may be immediately preceded by an ejec-
tion click, especially in mild forms of aortic
stenosis.
S1 A2 P2
Although the intensity of the murmur
does not correlate well with the severity of
aortic stenosis, other features do. For exam-
ple, the more severe the stenosis, the longer it
takes to force blood across the valve, and the
Fig. 8 later the murmur peaks in systole (Fig. 2.8).
Also, as shown in Figure 2.8, as the severity of

S1 P2
Figure 2.8. The severity of aortic stenosis affects
the shape of the systolic murmur and the heart
sounds. A. In mild stenosis, an ejection click (EJ) is often
present, followed by an early peaking crescendo–
decrescendo murmur and a normal aortic component of
S2 (A2). B. As stenosis becomes more severe, the peak of
the murmur becomes more delayed in systole and the
intensity of A2 lessens. The prolonged ventricular ejec-
tion time delays A2 so that it merges with or occurs after
the pulmonic component of S2 (P2); the ejection click
may not be heard. C. In severe stenosis, the murmur
peaks very late in systole, and A2 is usually absent be-
cause of immobility of the valve leaflets. S1, first heart
sound; S2, second heart sound.

stenosis increases, the aortic component of S2


softens because the leaflets become more
rigidly fixed in place.
Aortic stenosis causes a high-frequency
S1 S2 murmur, reflecting the sizable pressure gra-
dient across the valve. It is best heard in the
“aortic area” at the second and third right
Figure 2.7. Systolic ejection murmur of aortic steno- intercostal spaces close to the sternum. The
sis. There is a short delay between the first heart sound
(S1) and the onset of the murmur. LV, left ventricle; S2, murmur typically radiates toward the neck
second heart sound. (the direction of turbulent blood flow) but
10090-02_CH02.qxd 8/24/06 5:41 PM Page 41

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 41

often can be heard in a wide distribution, in- Tricuspid valve regurgitation is best heard
cluding the cardiac apex. along the left lower sternal border. It gener-
The murmur of pulmonic stenosis also be- ally radiates to the right of the sternum and
gins after S1, it may be preceded by an ejec- is high pitched and blowing in quality. The
tion click, but unlike aortic stenosis, it may intensity of the murmur increases with
extend beyond A2. That is, if the stenosis is inspiration because the negative intratho-
severe, it will result in a very prolonged right racic pressure induced during inspiration
ventricular ejection time, elongating the enhances venous return to the heart. The
murmur, which will continue beyond A2 latter augments right ventricular stroke vol-
and end just before closure of the pulmonic ume, thereby increasing the amount of re-
valve (P2). Pulmonic stenosis is usually loud- gurgitated blood.
est at the second to third left intercostal The murmur of a ventricular septal defect is
spaces close to the sternum. It does not ra- heard best at the fourth to sixth left inter-
diate as widely as aortic stenosis, but some- costal spaces, is high pitched, and may be as-
times it is transmitted to the neck or left sociated with a palpable thrill. The intensity
shoulder. of the murmur does not increase with in-
Young adults often have benign systolic spiration, nor does it radiate to the axilla,
ejection murmurs owing to increased systolic which helps distinguish it from tricuspid
flow across normal aortic and pulmonic and mitral regurgitation, respectively. Of
valves. This type of murmur often becomes note, the smaller the VSD, the greater the
softer or disappears when the patient sits turbulence of blood flow between the left
upright. and right ventricles and the louder the mur-
Pansystolic (also termed holosystolic) mur. Some of the loudest murmurs ever
murmurs are caused by regurgitation of heard are those associated with small VSDs.
blood across an incompetent mitral or tri- Late systolic murmurs begin in mid-to-
cuspid valve or through a ventricular septal late systole and continue to the end of sys-
defect (VSD; see Fig. 2.6). These murmurs tole. The most common example is mitral
are characterized by a uniform intensity regurgitation caused by mitral valve prolapse—
throughout systole. In mitral and tricuspid bowing of abnormally redundant and elon-
valve regurgitation, as soon as ventricular gated valve leaflets into the left atrium dur-
pressure exceeds atrial pressure (i.e., when S1 ing ventricular contraction (see Fig. 2.6).
occurs), there is immediate retrograde flow This murmur is usually preceded by a
across the regurgitant valve. Thus, there is midsystolic click and is described further in
no gap between S1 and the onset of these Chapter 8.
pansystolic murmurs, in contrast to the
systolic ejection murmurs discussed earlier.
Diastolic Murmurs
Similarly, there is no significant gap be-
tween S1 and the onset of the systolic Diastolic murmurs are divided into early de-
murmur of a VSD, because left ventricular crescendo murmurs and mid-to-late rum-
systolic pressure exceeds right ventricular bling murmurs (Fig. 2.9). Early diastolic Fig. 9
systolic pressure (and flow occurs) quickly murmurs result from regurgitant flow
after the onset of contraction. through either the aortic or pulmonic valve,
The pansystolic murmur of advanced mi- with the former being much more common
tral regurgitation continues through the aor- in adults. If produced by aortic valve regurgi-
tic closure sound because left ventricular tation, the murmur begins at A2, has a de-
pressure remains greater than that in the left crescendo shape, and terminates before the
atrium at the time of aortic closure. The next S1. Because diastolic relaxation of the
murmur is heard best at the apex, is high left ventricle is rapid, a pressure gradient de-
pitched and “blowing” in quality, and often velops immediately between the aorta and
radiates toward the left axilla; its intensity lower-pressured left ventricle in aortic re-
does not change with respiration. gurgitation, and the murmur therefore dis-
10090-02_CH02.qxd 8/24/06 5:41 PM Page 42

42 Chapter Two

• Aortic regurgitation
• Pulmonic regurgitation

S1 S2 S1

• Mild mitral or tricuspid stenosis

S1 S2 S1

• Severe mitral or tricuspid stenosis

S1 S2 S1

Figure 2.9. Classification of diastolic murmurs. A. An early diastolic decrescendo murmur is typi-
cal of aortic or pulmonic valve regurgitation. B. Mid-to-late low-frequency rumbling murmurs are usu-
ally the result of mitral or tricuspid valve stenosis, which follows a sharp opening snap (OS). Presystolic
accentuation of the murmur occurs in patients in normal sinus rhythm because of the transient rise in
atrial pressure during atrial contraction. C. In more severe mitral or tricuspid valve stenosis, the open-
ing snap and diastolic murmur occur earlier and the murmur is prolonged. S1, first heart sound; S2, sec-
ond heart sound.

plays its maximum intensity at its onset. begins after S2 and is preceded by an open-
Thereafter in diastole, as the aortic pressure ing snap. The shape of this murmur is
falls and the LV pressure increases (as blood unique. Following the opening snap, the
fills the ventricle), the gradient between the murmur is at its loudest because the pres-
two chambers diminishes and the murmur sure gradient between the atrium and ven-
decreases in intensity. Aortic regurgitation is tricle is at its maximum. The murmur then
a high-pitched murmur, best heard using decrescendos or disappears totally during
the diaphragm of the stethoscope along the diastole as the transvalvular gradient de-
left sternal border with the patient sitting, creases. The degree to which the murmur
leaning forward, and exhaling. fades depends on the severity of the steno-
Pulmonic regurgitation in adults is usually sis. If the stenosis is severe, the murmur
owing to the presence of pulmonary arterial is prolonged; if the stenosis is mild, the
hypertension. It has an early diastolic de- murmur disappears in mid-to-late diastole.
crescendo murmur profile similar to that of Whether the stenosis is mild or severe, the
aortic regurgitation, but it is best heard in murmur intensifies at the end of diastole in
the pulmonic area and its intensity may in- patients in normal sinus rhythm, when
crease with inspiration. atrial contraction augments flow across the
Mid-to-late diastolic murmurs result valve (see Fig. 2.9). The murmur of mitral
from either turbulent flow across a stenotic stenosis is low pitched and is heard best
mitral or tricuspid valve or less commonly with the bell of the stethoscope at the apex,
from abnormally increased flow across a while the patient lies in the left lateral de-
normal mitral or tricuspid valve (see Fig. cubitus position. The much less common
2.9). If resulting from stenosis, the murmur murmur of tricuspid stenosis is better aus- 1 LINE SHORT
10090-02_CH02.qxd 8/24/06 5:41 PM Page 43

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 43

cultated at the lower sternum, near the xi- begins in early systole, crescendos to its
phoid process. maximum at S2, then decrescendos until the
Hyperdynamic states such as fever, ane- next S1 (Fig. 2.10). Fig. 10
mia, hyperthyroidism, and exercise cause The “to-and-fro” combined murmur in a
increased flow across the normal tricuspid patient with both aortic stenosis and aortic
and mitral valves and can therefore result in regurgitation could be mistaken for a con-
a diastolic murmur. In patients with ad- tinuous murmur (see Fig. 2.10). During sys-
vanced mitral regurgitation, the expected tole, there is a diamond-shaped ejection
systolic murmur can be accompanied by an murmur, and during diastole a decrescendo
additional diastolic murmur owing to the murmur. However, in the case of a to-and-
increased volume of blood that must return fro murmur, the sound does not extend
across the valve to the left ventricle in dias- through S2 because it has discrete systolic
tole. Similarly, patients with either tricuspid and diastolic components.
regurgitation or an atrial septal defect (see
Chapter 16) may display a diastolic flow
murmur across the tricuspid valve. SUMMARY
Abnormal heart sounds and murmurs are
Continuous Murmurs common in acquired and congenital heart
disease and can be predicted by the underly-
Continuous murmurs are heard throughout ing pathology. Although it may seem diffi-
the cardiac cycle without an audible hiatus cult to remember even the basic features pre-
between systole and diastole. Such murmurs sented here, it will become easier as you learn
result from conditions in which there is a more about the pathophysiology of these
persistent pressure gradient between two conditions, and as your experience in physi-
structures during systole and diastole. An cal diagnoses grows. For now, just remember
example is the murmur of patent ductus ar- that the information is here, and refer to it as
teriosus, in which there is an abnormal com- needed. Tables 2.2 and 2.3 and Figure 2.11 Tab. 2,
munication between the aorta and pul- summarize features of the heart sounds and Tab. 3,
monary artery (see Chapter 16). During murmurs described in this chapter. Fig. 11
systole, blood flows from the high-pressure
ascending aorta through the ductus into the
lower-pressure pulmonary artery. During di- Acknowledgments
astole, the aortic pressure remains greater Contributors to the previous editions of this chapter
than that in the pulmonary artery and flow were Oscar Benavidez, MD; Bradley S. Marino, MD;
continues across the ductus. This murmur Allan Goldblatt, MD; and Leonard S. Lilly, MD.

• Patent ductus arteriosus

S1 S2 S1

• Aortic stenosis and regurgitation


• Pulmonic stenosis and regurgitation

S1 S2 S1
Figure 2.10. A continuous murmur peaks at, and extends through, the second heart sound (S2).
A to-and-fro murmur is not continuous; rather, there is a systolic component and a distinct diastolic com-
1 LINE SHORT ponent, separated by S2. S1, first heart sound.
10090-02_CH02.qxd 8/24/06 5:41 PM Page 44

44 Chapter Two

TABLE 2.2. Common Heart Sounds

Sound Location Pitch Significance

S1 Apex High Normal closure of mitral and tricuspid


valves
S2 Base High Normal closure of aortic (A2) and
pulmonic (P2) valves
Extra systolic sounds
Ejection clicks Aortic: apex and base High Aortic or pulmonic stenosis, or dilata-
Pulmonic: base High tion of aortic root or pulmonary artery
Mid-to-late click Mitral: apex High Mitral or tricuspid valve prolapse
Tricuspid: LLSB High
Extra diastolic sounds
Opening snap Apex High Mitral stenosis
S3 Left-sided: apex Low Normal in children
Abnormal in adults: indicates heart
failure or volume overload state
S4 Left-sided: apex Low Reduced ventricular compliance

LLSB, lower left sternal border.

TABLE 2.3. Common Murmurs

Murmur Type Example Location and Radiation

Systolic ejection Aortic stenosis 2nd right intercostal space →


neck (but may radiate
widely)
Pulmonic stenosis 2nd–3rd left intercostal spaces

Pansystolic Mitral regurgitation Apex → axilla


Tricuspid regurgitation Left lower sternal border →
right lower sternal border

Late systolic Mitral valve prolapse Apex → axilla

Early diastolic Aortic regurgitation Along left side of sternum


Pulmonic regurgitation Upper left side of sternum

Mid- or late diastolic Mitral stenosis Apex


10090-02_CH02.qxd 8/24/06 5:41 PM Page 45

The Cardiac Cycle: Mechanisms of Heart Sounds and Murmurs 45

Aortic area Pulmonic area


Ejection-type murmur Ejection-type murmur
• Aortic stenosis • Pulmonic stenosis
• Flow murmur • Flow murmur

Left sternal border


Early diastolic murmur
• Aortic regurgitation
• Pulmonic regurgitation

Mitral area
Tricuspid area Pansystolic murmur
Pansystolic murmur • Mitral regurgitation
• Tricuspid regurgitation Mid-to-late diastolic murmur
• Ventricular septal defect • Mitral stenosis

Mid-to-late diastolic
murmur
• Tricuspid stenosis
• Atrial septal defect
Figure 2.11. Locations of maximum intensity of common murmurs.

Additional Reading LeBlond RF, DeGowin RL, Brown DD. DeGowin’s


Diagnostic Examination. 7th Ed. New York:
Bickley LS. Bates’ Guide to Physical Examination McGraw-Hill, 2004.
and History Taking. 8th Ed. Philadelphia: Lippin- Orient JM, Sapira JD. Sapira’s Art and Science of Bed-
cott Williams & Wilkins, 2003. side Diagnosis. 3rd Ed. Philadelphia: Lippincott
Constant J. Essentials of Bedside Cardiology. 2nd Ed. Williams & Wilkins, 2005.
Totowa, NJ: Humana Press, 2003.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 46

C H A P T E R

Diagnostic Imaging and


Cardiac Catheterization
Nicole Martin
3
Patricia Challender Come

CARDIAC RADIOGRAPHY Measurement of Blood Flow


Cardiac Silhouette Calculation of Vascular Resistance
Pulmonary Manifestations of Heart Disease Contrast Angiography
ECHOCARDIOGRAPHY NUCLEAR IMAGING
Ventricular Assessment Assessment of Myocardial Perfusion
Valvular Lesions Radionuclide Ventriculography
Coronary Artery Disease Assessment of Myocardial Metabolism
Cardiomyopathy COMPUTED TOMOGRAPHY
Pericardial Disease
MAGNETIC RESONANCE IMAGING
CARDIAC CATHETERIZATION
Measurement of Pressure

Imaging plays a central role in the assess- memorize the details. This chapter is meant as
ment of cardiac function and pathology. a reference to be consulted as needed.
Traditional imaging modalities such as chest
radiography, echocardiography (echo), car-
CARDIAC RADIOGRAPHY
diac catheterization with cineangiography,
and nuclear imaging are fundamental in the The extent of penetration of x-rays through
diagnosis and management of cardiovascu- the body is inversely proportional to tissue
lar diseases. These techniques are being in- density. Air-filled tissues, such as the lung,
creasingly supplemented by newer modali- absorb few x-rays and expose the underly-
ties, including computed tomography and ing film, causing it to appear black. In con-
magnetic resonance imaging. trast, dense materials, such as bone, absorb
This chapter presents an overview of imag- more radiation and appear white, or radio-
ing techniques as they are used to assess the paque. For a boundary to show between two
cardiovascular disorders described in the structures, they must differ in density. Myo-
book. It would be beneficial to familiarize cardium, valves, and other intracardiac
yourself with the information now, but not to structures have densities similar to that of

46
10090-03_CH03.qxd 8/31/06 5:24 PM Page 47

Diagnostic Imaging and Cardiac Catheterization 47

adjacent blood; consequently, radiography terior chest. This positioning places the heart
cannot delineate these structures unless they close to the x-ray film so that its image is only
happen to be calcified. Conversely, heart bor- minimally distorted, allowing for an accurate
ders adjacent to a lung are depicted clearly be- assessment of size. In the standard lateral
cause the heart and an air-filled lung have dif- view, the patient’s left side is placed against
ferent densities. If the lung adjacent to the the film plate and the x-rays pass through
heart is diseased, however (as in pulmonary the body from right to left. The frontal ra-
edema, consolidation, or collapse), the lung diograph is particularly useful for assessing
density will match that of the heart and the the size of the left ventricle, left atrial ap-
cardiac border will be poorly defined. pendage, pulmonary artery, aorta, and supe-
Frontal and lateral radiographs are rou- rior vena cava; the lateral view evaluates right
tinely used to assess the heart and lungs (Fig. ventricular size, posterior borders of the left
Fig. 1 3.1). The frontal view is usually a posterior- atrium and ventricle, and the anteroposterior
anterior image in which the x-rays are trans- diameter of the thorax. In some cases, opti-
mitted from behind (i.e., posterior to) the mal evaluation of the heart requires right and
patient, travel through the body, and then left anterior oblique views as well.
expose a sheet of film placed against the an-
Cardiac Silhouette
Chest radiographs are used to evaluate the
size of heart chambers and the pulmonary
consequences of cardiac disease. Alterations
in chamber size are reflected by changes in
the cardiac silhouette. In the frontal view of
adults, the heart shadow should occupy 50%
or less of the maximal width of the thorax,
measured between the inner margins of the
ribs (although in children, normal cardiac
diameter may be up to 60% of the thoracic
width). Thus, the cardio:thoracic ratio is used
instead of absolute measurements to account
for differences in body habitus.
In several situations, the cardiac silhou-
ette inaccurately reflects heart size. An ele-
vated diaphragm or narrow chest anteropos-
terior diameter, for example, may cause the
heart to appear to spread out transversely.
Consequently, the silhouette on a posterior-
anterior chest film may be greater than 50%
of the thorax even though the actual heart
size is normal. Therefore, the chest antero-
posterior diameter should be assessed on the
lateral view before the frontal image is deter-
Figure 3.1. Posteroanterior (A and B) and lateral (C
and D) chest radiographs of a person without car- mined to truly represent an enlarged heart.
diopulmonary disease, illustrating cardiac chambers The presence of a pericardial effusion around
and valves. AO, aorta; AV, azygos vein; IVC, inferior the heart can also enlarge the cardiac silhou-
vena cava; LA, left atrium; LAA, left atrial appendage;
LPA, left pulmonary artery; LV, left ventricle; MPA, main ette, because fluid and myocardium affect
pulmonary artery; MV, mitral valve; RA, right atrium; RPA, x-ray penetration similarly.
right pulmonary artery; RV, right ventricle; SVC, superior Radiographs can depict dilatation of the
vena cava; TV, tricuspid valve. (Reprinted with permission
from Come PC, ed. Diagnostic Cardiology: Noninvasive cardiac chambers and great vessels. Hyper-
Imaging Techniques. Philadelphia: JB Lippincott, 1985.) trophy alone may not result in radiographic
10090-03_CH03.qxd 8/31/06 5:24 PM Page 48

48 Chapter Three

abnormalities, because it generally occurs at cle, accompanied by signs of pulmonary hy-


the expense of the cavity’s internal volume pertension, suggests mitral stenosis (Fig. 3.2). Fig. 2
and produces little or no change in overall In contrast, dilatation of the pulmonary
cardiac size. Hypertrophy is more readily artery and right heart chambers, but without
suspected from abnormalities on the elec- enlargement of the left-sided heart dimen-
trocardiogram or by directly measuring wall sions, suggests pulmonary vascular obstruc-
thickness by echocardiography (as discussed tion or increased pulmonary artery blood
later in the chapter). Major causes of chamber flow (e.g., owing to an atrial septal defect;
and great vessel dilatation include heart fail- Fig. 3.3). Fig. 3
ure, valvular lesions, abnormal intracardiac The shape of the dilated chamber may also
and extracardiac communications (shunts), provide etiologic clues. For instance, in left
and certain pulmonary disorders. Because ventricular volume overload owing to valvu-
dilatation takes time to develop, recent le- lar insufficiency, the ventricle tends to en-
sions, such as acute mitral valve insuffi- large primarily in its long axis, displacing the
ciency, may present without apparent car- apex downward and to the left. In contrast,
diac enlargement. when left ventricular dilation results from pri-
The pattern of chamber enlargement may mary myocardial dysfunction, left ventricu-
suggest specific disease entities. For example, lar length and width are generally both in-
dilatation of the left atrium and right ventri- creased, causing the heart to appear globular.

Figure 3.2. Posteroanterior chest radiograph of a patient with severe


mitral stenosis and secondary pulmonary vascular congestion. The ra-
diograph shows a prominent left atrial appendage (arrowheads) with con-
sequent straightening of the left-heart border and suggestion of a double-
density right cardiac border (arrows) produced by the enlarged left atrium.
The aortic silhouette is small, which suggests chronic low cardiac output. Ra-
diographic signs of pulmonary vascular congestion include increased caliber
of upper-zone pulmonary vessel markings and decreased caliber of lower-
zone vessels.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 49

Diagnostic Imaging and Cardiac Catheterization 49

Figure 3.3. Posteroanterior chest radiograph of a patient with pul-


monary hypertension secondary to an atrial septal defect. Radiographic
signs of pulmonary hypertension include pulmonary artery dilatation (black
arrows; compare with the appearance of left atrial appendage dilatation in
Fig. 3.2) and large central pulmonary arteries (white arrows) associated with
small peripheral vessels (a pattern known as peripheral pruning).

Chest radiographs can also detect dila- increased vascular markings, redistribution
tation of the aorta and pulmonary artery. of blood flow from the bases to the apices of
Causes of aortic dilatation include aneurysm, the lungs (termed cephalization of vessels),
Fig. 4 dissection, and aortic valve disease (Fig. 3.4). pulmonary edema, the presence of abnor-
Normal aging and atherosclerosis may also mal septal lines (termed Kerley lines), and
cause the aorta to become dilated and tor- pleural effusions (Fig. 3.5). Blood flow redis- Fig. 5
tuous. The pulmonary artery may be en- tribution appears as an increase in the num-
larged in patients with left-to-right shunts, ber or width of vascular markings at the
which cause increased pulmonary blood apex. Interstitial and alveolar pulmonary
flow, and in those with pulmonary hyper- edemas produce opacity radiating from the
tension of diverse causes (see Fig. 3.3). Iso- hilar region bilaterally (known as a “butter-
lated enlargement of the proximal left pul- fly” or “bat-wing” pattern) and air broncho-
monary artery is seen in some patients with grams, respectively. Kerley B lines, which
pulmonic stenosis. depict fluid in interlobular spaces at the pe-
riphery of the lung, result from interstitial
edema. Pleural effusions cause blunting of
Pulmonary Manifestations of
the costodiaphragmatic angles.
Heart Disease
Changes in pulmonary blood flow may
The appearance of the pulmonary vascula- also alter the appearance of the pulmonary
ture reflects abnormalities of pulmonary ar- vessels. Focal oligemia (decreased flow) may
terial and venous pressures and pulmonary result from pulmonary embolism or replace-
blood flow. Increased pulmonary venous ment of functioning lung tissue by emphy-
pressure, as occurs in left-heart failure, causes sematous bullae. The finding of enlarged
10090-03_CH03.qxd 8/31/06 5:24 PM Page 50

50 Chapter Three

Figure 3.4. Posteroanterior chest radiograph of a patient with


aortic stenosis and insufficiency secondary to a bicuspid aortic
valve. In addition to poststenotic dilatation of the ascending aorta
(black arrows), the transverse aorta (white arrow) is prominent, sug-
gesting aortic insufficiency in addition to stenosis.

central pulmonary arteries, but small periph- cause mechanical deformation of the piezo-
eral vessels (termed peripheral pruning), sug- electric element. The machine measures the
gests pulmonary hypertension (see Fig. 3.3). time elapsed between the initiation and re-
Tab. 1 Table 3.1 summarizes the major radio- ception of the sound waves, thus enabling it
graphic findings in common forms of car- to calculate the distance between the trans-
diac disease. ducer and each anatomic reflecting surface.
Images are then constructed from these cal-
culations.
ECHOCARDIOGRAPHY
Three types of echocardiographic modali-
Echocardiography plays an essential role in ties are generally performed: M-mode, two-
the diagnosis and serial evaluation of many dimensional (2-D), and Doppler imaging.
cardiac disorders. It is safe, noninvasive, rel- Each type of imaging can be performed from
atively inexpensive, and capable of accu- various locations. Most commonly, transtho-
rately depicting a wide array of heart dis- racic studies are performed, in which images
eases. High-frequency (ultrasonic) waves, are obtained by placing the transducer on the
generated by a piezoelectric element, travel surface of the chest. When greater structural
through body tissue and are reflected at in- detail is required, transesophageal imaging is
terfaces where there are differences in the performed, as described later in this section.
acoustic impedance of adjacent tissues. The M-mode echocardiography was the first
reflected waves return to the transducer and cardiac application of ultrasonography. It is
10090-03_CH03.qxd 8/31/06 5:24 PM Page 51

Diagnostic Imaging and Cardiac Catheterization 51

Figure 3.5. Radiographs of patients


with congestive heart failure. These
are anteroposterior views (which may ex-
aggerate the size of the heart because it
is further from the x-ray film), taken with
portable x-ray machines at the bedside.
A. Mild congestive heart failure. Pul-
monary congestion is indicated by vas-
cular redistribution from the bases to the
apices of the lungs. The white spots la-
beled “L” are electrocardiographic leads
on the patient’s chest. B. Severe conges-
tive heart failure. Increased pulmonary
vascular markings are present through-
out the lung fields, along with peribron-
chiolar cuffing (black arrow) and pleural
effusion, which is indicated by blunting
of the costodiaphragmatic angle and
tracking up the right lateral hemithorax
(black arrowheads). The presence of in-
terstitial and alveolar edema produces
perihilar haziness and air bronchograms
(open arrows), which occur when the ra-
diolucent bronchial tree is contrasted B
with opaque edematous lung tissue.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 52

52 Chapter Three

TABLE 3.1. Chest Radiography of Common Cardiac Disorders

Disorder Finding

Congestive heart failure • Vascular redistribution from bases to apices of the lungs
• Interstitial and alveolar edema
• Perihilar haziness
• Peribronchiolar cuffing
• Air bronchograms
• Pleural effusions
Pulmonic valve stenosis • Poststenotic dilatation of pulmonary artery
• Normal cardiac chamber sizes
• Clear lung fields
Aortic valve stenosis • Poststenotic dilatation of ascending aorta
• Normal cardiac chamber sizes (until heart fails)
• Normal pulmonary vasculature
Aortic regurgitation • Left ventricular enlargement
• Dilated aorta
Mitral stenosis • Enlarged left atrium
• Small aorta (if chronic low cardiac output)
• Signs of pulmonary venous congestion
Mitral regurgitation • Left atrial dilatation
• Left ventricular dilatation
• If severe:
• Right ventricular dilatation
• Signs of congestive heart failure

now used rarely by itself because it provides patient’s skin, these include the standard
limited data from one narrow ultrasonic parasternal long axis, parasternal short axis,
beam, and only structures along that single apical four-chamber, apical two-chamber,
line are displayed. M-mode techniques apical three-chamber (also known as apical
continue to be valuable for measurement long axis), and subcostal views. The paraster-
of wall thicknesses and chamber diameters nal long axis view is recorded with the trans-
and for accurate timing of valve movements ducer in the third or fourth intercostal space
Fig. 6 (Fig. 3.6). to the left of the sternum. This view is par-
In 2-D echocardiography, multiple ul- ticularly useful for evaluation of the left
trasonic beams are transmitted through a atrium, mitral valve, left ventricle, and left
wide arc. The returning signals are inte- ventricular outflow tract, which includes
grated to produce two-dimensional images the aortic valve and adjacent interventricu-
of the heart on a video monitor. As a result, lar septum. To obtain parasternal short axis
this technique depicts anatomic relation- views, the transducer is rotated 90° from its
ships and defines the movement of cardiac position for the parasternal long axis view.
structures relative to one another. The wide The short axis images depict transverse
fields of view enhance the ability of 2-D planes of the heart. Several different levels
echocardiograms to detect and display wall are imaged to assess the aortic valve, mitral
and valve motion and intracardiac masses valve, and left ventricular wall motion.
such as vegetations, thrombi, and tumors. Apical TTE views are produced when the
Fig. 7 Each two-dimensional plane (Fig. 3.7) de- transducer is placed at the point of maximal
lineates only part of a given cardiac struc- cardiac impulse. The apical four-chamber view
ture. Optimal evaluation of the entire heart evaluates the mitral and tricuspid valves as
is achieved by using combinations of views. well as the atrial and ventricular chambers,
In transthoracic echocardiography (TTE), in including the motion of the lateral, septal,
which the transducer is placed against the and apical left ventricular walls. The apical
10090-03_CH03.qxd 8/31/06 5:24 PM Page 53

Diagnostic Imaging and Cardiac Catheterization 53

the rib cage, may provide a better ultrasonic


window, allowing visualization of all four
cardiac chambers.
Doppler imaging evaluates blood flow
direction and velocity and turbulence. Addi-
tionally, it permits estimation of pressure
gradients within the heart and great vessels.
1 Doppler studies are based on the physical
principle that waves reflected from a moving
object undergo a frequency shift according
to the moving object’s velocity relative to
2 the source of the waves. Color flow mapping
4
3 converts the Doppler signals to an arbitrarily
chosen scale of colors that represent direc-
tion, velocity, and turbulence of blood flow
in a semiquantitative way. The colors are su-
perimposed on 2-D images and show the lo-
cation of stenotic and regurgitant valvular
lesions and of abnormal communications
within the heart and great vessels. For exam-
ple, Doppler echocardiography in a patient
with mitral regurgitation shows a jet of ret-
rograde flow into the left atrium during sys-
1 2 3 4 tole (Fig. 3.8). Fig. 8
Figure 3.6. Schematic diagram of the heart in the
Sound frequency shifts are converted into
parasternal long axis view (upper drawing). Below it blood flow velocity measurements (auto-
is the electrocardiogram and M-mode echocardiogram matically calculated by the echo machine),
generated by changes in the direction of the transducer
from position 1 to position 4. Chamber sizes and wall
by the relationship
thicknesses can be measured along the vertical axis. Note fs • c
the sequential thickening and thinning of the ventricular v=
walls, corresponding to systolic contraction and diastolic 2 f O ( cosθ )
relaxation respectively, as well as movement of the aortic
and mitral valves. AMV, anterior mitral valve leaflet; AO, in which v equals the blood flow velocity
aortic root; AV, aortic valve; CW, chest wall; LA, left (m/sec); fs, the Doppler frequency shift (kHz);
atrium; LV, left ventricle; LVW, left ventricular wall; PMV,
AQ2 pulmonic valve; PPM, posterior papillary muscle region; c, the velocity of sound in body tissue (m/sec);
RV, right ventricle; RVW, right ventricular wall; S, ster- fo, the frequency of the sound pulse emitted
num; T, echocardiographic transducer. (Reprinted with from the transducer (MHz); and θ, the angle
permission from Come PC. Echocardiography in diagno-
sis and management of cardiovascular disease. Compr
between the transmitted sound pulse and the
Ther 1980;6:7–17. By permission of International Pub- mean axis of blood flow.
AQ3 lishing Group, Cleveland, OH.) Transesophageal echocardiography
(TEE) uses a miniaturized transducer
mounted at the end of a modified endoscope
two-chamber view shows only the left side of to transmit and receive ultrasound waves
the heart, and it depicts movement of the from within the esophagus, thus producing
anterior, inferior, and apical walls. very clear images of the neighboring cardiac
In some patients, such as those with ob- structures (Fig. 3.9) and much of the tho- Fig. 9
structive airways disease, the parasternal and racic aorta. Modern probes permit multipla-
apical views do not provide adequate depic- nar imaging and Doppler interrogation as
tion of cardiac structures because of signal well. TEE is particularly helpful in the as-
attenuation caused by excessive underlying sessment of aortic and atrial abnormalities,
air. In such patients, the subcostal view, in conditions that are less well visualized by
which the transducer is placed inferior to conventional transthoracic echo imaging. For
10090-03_CH03.qxd 8/31/06 5:24 PM Page 54

54 Chapter Three

Interventricular septum
RV

Ao
Aortic
LV valve
LA
Mitral
valve
A
LV posterior wall

RV
LV

RV LV
Mitral
Tricuspid valve
valve
RA LA
C

Figure 3.7. Transthoracic two-dimensional echocardiographic views.


A. Parasternal long axis view. B. Parasternal short axis view. Notice that the left ven-
tricle appears circular in this view, while the right ventricle is crescent shaped.
C. Apical four-chamber view. Ao, aorta; LA, left atrium; LV, left ventricle; RA, right
atrium; RV, right ventricle. (Modified from Sahn DJ, Anderson F. Two-Dimensional
Anatomy of the Heart. New York: John Wiley & Sons, 1982.)

example, TEE is more sensitive than transtho- mechanical valves reflect a large portion of ul-
racic echo for the detection of thrombus with- trasound waves, thus interfering with visual-
Fig. 10 in the left atrial appendage (Fig. 3.10), which ization of more-posterior structures (termed
is of great importance in patients who re- acoustic shadowing). TEE aids visualization of
quire electrical cardioversion of atrial fibril- the posterior chambers in such patients and
lation without prior anticoagulation (see is therefore the most sensitive noninvasive
Chapter 12). The proximity of the esophagus technique for evaluating perivalvular leaks.
to the heart makes TEE imaging particularly TEE is commonly used to evaluate pa-
advantageous in patients for whom trans- tients with cerebral ischemia of unexplained
thoracic echo images are unsatisfactory (e.g., etiology, because it can identify cardiovascu-
those with chronic obstructive lung disease). lar causes of emboli with a high sensitivity.
TEE is also advantageous in the evaluation These etiologies include intracardiac thrombi
of patients with prosthetic heart valves. Dur- or tumors, atherosclerotic debris within the
ing standard transthoracic imaging, artificial aorta, and valvular vegetations. It is also
10090-03_CH03.qxd 8/31/06 5:24 PM Page 55

Diagnostic Imaging and Cardiac Catheterization 55

right-to-left heart blood flow, or in the pres-


ence of an intrapulmonary shunt, bubbles
of contrast will appear in the left-sided
chambers as well. Newer perfluorocarbon-
based contrast agents have been developed
with sufficiently small particle size to inten-
tionally pass through the pulmonary circula-
tion. These agents are sometimes used to
opacify the left ventricular cavity and, via
the coronary arteries, the myocardium, en-
abling superior assessment of LV contraction
and myocardial perfusion.
Echocardiographic techniques can identify
valvular lesions, complications of coronary
artery disease, septal defects, intracardiac
masses, cardiomyopathy, ventricular hyper-
trophy, pericardial disease, aortic disease,
and congenital heart disease. Evaluation in-
cludes assessment of cardiac chamber sizes,
Figure 3.8. Doppler color flow mapping (reproduced wall thicknesses, wall motion, valvular func-
in gray tones) of mitral regurgitation (MR). The tion, blood flow, and intracardiac hemody-
Doppler image, recorded in systole, is superimposed on namics. A few of these topics are highlighted
an apical view of the left ventricle (LV), left atrium (LA),
and mitral valve (short arrow). The retrograde flow of MR in the sections that follow.
into the LA is indicated by the long arrow.
Ventricular Assessment
Two-dimensional echocardiography assesses
highly sensitive and specific for the detec- left ventricular systolic function by comput-
tion of aortic dissection. ing fractional changes between end-diastolic
In the operating room, TEE permits im- and end-systolic measurements. Left ven-
mediate evaluation after surgical repair of tricular width, area, length, and volume in
congenital and valvular lesions. In addition, diastole and systole are used to assess con-
imaging of ventricular wall motion can iden- tractile function and calculate the ventri-
tify periods of myocardial ischemia during cular ejection fraction. Beyond assessing
high-risk surgery. overall left ventricular systolic function,
Contrast echocardiography is frequently two-dimensional echocardiography depicts
used in the evaluation of congenital heart regional ventricular wall motion abnormal-
disease because it is highly sensitive for the ities, a common sign of coronary artery dis-
detection of abnormal intracardiac shunts. ease. Right ventricular systolic function is
In this technique, often called a “bubble generally assessed qualitatively because the
study,” an echocardiographic contrast agent right ventricle does not lend itself as easily
(e.g., agitated saline) is rapidly injected into to geometric modeling as does the left ven-
a peripheral (usually a brachial) vein. Using tricle. Echocardiography is useful in evalu-
standard echocardiographic imaging, the ating ventricular wall thickness and mass,
contrast can be visualized passing through which are important in patients with hyper-
the cardiac chambers. Normally, there is tension, aortic stenosis, and hypertrophic
rapid opacification of the right-sided cham- cardiomyopathy (Fig. 3.11). Fig. 11
bers, but because the contrast is filtered out Diastolic dysfunction (e.g., caused by is-
(harmlessly) in the lungs, it does not reach chemic disease, ventricular hypertrophy, or
the left-sided chambers. However, in the pres- restrictive cardiomyopathy; see Chapter 9)
ence of an intracardiac shunt with abnormal can also be evaluated by Doppler techniques.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 56

56 Chapter Three

LV
Basal Short Axis
25–30 cm from incisors

Four Chambers RV
30 cm
Transgastric
Transgastric
35–40 cm
Basal Short Axis
LA

RA

RV

Four Chambers

IVC LA

RA

LV
RV

Figure 3.9. Transesophageal echocardiographic views. IVC, inferior vena cava; LA,
left atrium; LV, left ventricle; RA, right atrium; RV, right ventricle. (Courtesy of Jane Freed-
man, MD, Boston University Medical Center, MA.)

LA

LAA

Thrombus

A B
Figure 3.10. Echocardiographic imaging of an intracardiac thrombus. A. Transesophageal
echocardiographic image demonstrates thrombus within the left atrial appendage. (Courtesy of Scott
Streckenbach, MD, Massachusetts General Hospital, Boston.) B. Schematic drawing of same image.
LA, left atrium; LAA, left atrial appendage.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 57

Diagnostic Imaging and Cardiac Catheterization 57

Figure 3.11. Left ventricular outflow tract (LVOT) obstruction in hypertrophic cardiomyopathy. No-
tice that the interventricular septum (S) is thicker and more echogenic than the posterior wall (P). A. Before
ventricular contraction, the LVOT is only slightly narrowed. B. During contraction, the rapidly flowing blood
through the LVOT incites a Venturi effect and abnormally draws the mitral valve apparatus anteriorly toward
the hypertrophied septum (arrow), creating a functional obstruction. LA, left atrium. LV, left ventricle.

For example, Doppler tissue imaging is a then the calculated peak pressure gradient
modality that can readily record the maxi- across the valve = 4 × 42 = 64 mm Hg.
mum velocity at which the mitral valve an- Other calculations permit noninvasive
nulus springs back from the left ventricle in determination of the cross-sectional area of
early diastole, as a measure of the chamber’s stenotic valves. For instance, the continuity
ability to relax. Standard Doppler measure- equation is often used to calculate aortic valve
ments of flow velocity across the mitral valve area. This equation assumes that blood flow
in early, compared with late, diastole also (F, expressed in cc/sec) is the same at the aor-
provide important information about ven- tic valve orifice (AV) as at a neighboring po-
tricular diastolic function. sition along the flow stream (e.g., in the left
ventricular outflow tract [LVOT]).
Valvular Lesions FLVOT = FAV

Echocardiography can accurately determine As shown in Figure 3.12, blood flow at any Fig. 12
underlying causes of valvular abnormalities, position along a flow stream can also be ex-
and Doppler imaging permits quantitation pressed as the product of the Doppler veloc-
of the degree of valvular stenosis and regurgi- ity (V, in cm/sec) and cross-sectional area (A,
tation. The pressure gradient across a stenotic in cm2) at that level. If location 1 in Figure
valve can be calculated from the maximum 3.12 represents a position in the LVOT and
blood flow velocity (v) measured distal to the location 2 represents the aortic valve, then
valve, using the simplified Bernoulli equation:
ALVOT × VLVOT = AAV × VAV
Pressure gradient = 4 × v 2
The cross-sectional area of the LVOT (ALVOT)
As an example, if the peak velocity recorded is calculated simply as π(d/2)2, in which d
distal to a stenotic aortic valve is 4 m/sec, represents the LVOT diameter, measured by
10090-03_CH03.qxd 8/31/06 5:24 PM Page 58

58 Chapter Three

1 2 dium. The location and degree of abnormal


systolic contraction and decreased systolic
wall thickening indicate the extent of an in-
farction and implicate the responsible coro-
nary artery. Infarct size measured by 2-D
V1 V2 echocardiography correlates well with other
methods of quantification, such as radioiso-
tope scanning and positron emission to-
A1 A2
mography (described later in the chapter).
A1 x V1 = A2 x V2 Echocardiography is also used to detect com-
Figure 3.12. The continuity equation. Within a closed plications of acute myocardial infarction,
flow stream, the volume rate of flow at any point (calcu- including intraventricular thrombus for-
lated as the cross-sectional area at that site multiplied by
the maximum flow velocity at the same location) is equal mation, papillary muscle rupture, valvular
to the volume rate of flow at sequential points. Thus, dysfunction, ventricular septal rupture, and
cross-sectional area and velocity at any location are in- aneurysm formation.
versely proportional to one another. Here, location 2 is
narrower than location 1. Therefore the velocity at loca- Although echocardiography can depict
tion 2 must be greater for the same volume to pass per those consequences of coronary artery dis-
unit time. ease, transthoracic echo resolution is usually
insufficient to directly image the coronary
arteries themselves. In a few patients, the
echocardiography, usually from the para- most proximal portions of the coronary ar-
sternal long axis view. The velocities (VLVOT teries can be delineated.
and VAV) are measured by Doppler interro- Stress echocardiography can be used to
gation, from the apical four-chamber view. diagnose the presence of coronary artery
The equation can then be solved for the aor- disease. This technique assesses the devel-
tic valve area (AAV): opment of left ventricular regional wall mo-
( ALVOT × VLVOT ) tion abnormalities induced by exercise or
AAV = after the infusion of specific pharmacologic
VAV
agents, such as dobutamine, dipyridamole,
Color Doppler analysis provides a quali- or adenosine (see Chapter 6). Reversible myo-
tative assessment of the severity of regurgi- cardial ischemia is recognized by a stress-
tant valve lesions. In mitral regurgitation induced wall motion abnormality.
(see Fig. 3.8), for example, the ratio of the re-
gurgitant jet color Doppler area to the entire
left atrial area has traditionally been used to
Cardiomyopathy
classify the regurgitation as mild, moderate, Cardiomyopathies are heart muscle disorders
or severe. More quantitative evaluation of that occur in three forms: dilated, hypertro-
mitral regurgitation can now be performed phic, and restrictive (see Chapter 10). Echo-
by what is known as the proximal isoveloc- cardiography can often distinguish among
ity surface area (PISA) method. This tech- these and permits assessment of the severity
nique uses advanced color Doppler tech- of systolic and diastolic dysfunction. For ex-
niques to calculate the regurgitant volume ample, Figure 3.11 depicts the asymmetri-
and effective regurgitant orifice area, two cally thickened ventricular walls found in
values that predict clinical outcomes in pa- classic hypertrophic cardiomyopathy.
tients with chronic mitral regurgitation.
Pericardial Disease
Coronary Artery Disease
Two-dimensional echocardiography can
Echocardiography demonstrates ventricular identify abnormalities in the pericardial cav-
wall motion abnormalities associated with ity (e.g., excessive pericardial fluid, fibrinous
infarcted or transiently ischemic myocar- material, tumor, and clot). Tamponade and
10090-03_CH03.qxd 8/31/06 5:24 PM Page 59

Diagnostic Imaging and Cardiac Catheterization 59

constrictive pericarditis, the main functional results in diastolic “collapse” of the right
consequences of pericardial disease (see atrium, right ventricle, and sometimes (in
Chapter 14), are associated with particular more extreme cases) the left-sided chambers
echocardiographic abnormalities. In tam- (Fig. 3.13). Constrictive pericarditis is associ- Fig. 13
ponade, the increased intrapericardial pres- ated with increased thickness or reflective-
sure compresses the cardiac chambers and ness of the pericardial echo, abnormal pat-

ECG
ECG

Figure 3.13. Echocardiographic studies of a patient with a peri-


cardial effusion causing cardiac tamponade. The apical four-
chamber two-dimensional image (upper panel) shows a large peri-
cardial effusion (PE) and inward collapse (white arrowheads) of the
right atrium (RA) and left atrium (LA). M-mode tracings (lower pan-
els) indicate early diastolic collapse of the right ventricular wall (black
arrows). AoV, aortic valve; ECG, electrocardiogram; LV, left ventricle;
MV, mitral valve; RV, right ventricle; RVOT, right ventricular outflow
tract. (Reprinted with permission from Cunningham MJ, Safian RD,
Come PC, et al. Absence of pulsus paradoxus in a patient with cardiac
tamponade and coexisting pulmonary artery obstruction. Am J Med
1987;83:973–976.)
10090-03_CH03.qxd 8/31/06 5:24 PM Page 60

60 Chapter Three

terns of diastolic left ventricular wall mo- blood flow. In 1929, Werner Forssmann per-
tion, alterations in pulmonary venous flow formed the first cardiac catheterization, on
patterns, and exaggerated changes in mitral himself, thus ushering in the era of invasive
and tricuspid valve inflow velocities during cardiology. Much of what is known about the
respiration. pathophysiology of valvular heart disease and
Tab. 2 Table 3.2 summarizes the salient echocar- congestive heart failure comes from decades
diographic features of common cardiac dis- of subsequent hemodynamic research in the
eases. cardiac catheterization laboratory.

CARDIAC CATHETERIZATION Measurement of Pressure


In the diagnosis of many cardiovascular ab- Before catheterization of an artery or vein,
normalities, intravascular catheters are in- the patient is mildly sedated, and a local
serted to measure pressures in the heart anesthetic is used to numb the skin site of
chambers, to determine cardiac output and catheter entry. The catheter, attached to a
vascular resistances, and to inject radiopaque pressure transducer outside the body, is then
material to examine heart structures and introduced into the appropriate blood ves-

TABLE 3.2. Echocardiography in Common Cardiac Disorders

Disorder Finding

Valvular lesions
Mitral stenosis • Enlarged left atrium
• Thickened mitral valve leaflets
• Decreased movement and separation of mitral valve leaflets
• Decreased mitral valve orifice
Mitral regurgitation • Enlarged left atrium (if chronic)
• Enlarged left ventricle (if chronic)
• Systolic flow from left ventricle into left atrium (by Doppler)
Aortic stenosis • Thickened aortic valve cusps
• Decreased valve orifice
• Increased left ventricular wall thickness
Aortic regurgitation • Enlarged left ventricle
• Abnormalities of aortic valve or aortic root
Left ventricular function
Myocardial infarction and • Hypokinetic, dyskinetic, or akinetic ventricular wall motion
complications • Decreased ejection fraction
• Thrombus within left ventricle
• Aneurysm of ventricular wall
• Septal rupture (abnormal Doppler flow)
• Papillary muscle rupture
• Pericardial effusion
Cardiomyopathies
Dilated • Enlarged ventricular chamber sizes
• Normal ventricular wall thicknesses
• Decreased systolic contraction
Hypertrophic • Normal or decreased ventricular chamber sizes
• Increased ventricular wall thickness
• Diastolic dysfunction (assessed by Doppler)
Restrictive • Normal or decreased ventricular chamber sizes
• Enlarged atria
• Increased ventricular wall thickness
• Ventricular contractile function often normal
• Diastolic dysfunction (assessed by Doppler)
10090-03_CH03.qxd 8/31/06 5:24 PM Page 61

Diagnostic Imaging and Cardiac Catheterization 61

sel. To measure pressures in the right atrium, suitable size (e.g., the inferior or superior
right ventricle, and pulmonary artery, a vena cava), the balloon at the catheter tip is
catheter is usually inserted into a femoral, manually inflated so that venous return of
brachial, or jugular vein. Pressures in the blood helps direct the catheter into the right-
aorta and left ventricle are measured via sided heart chambers and into the pulmo-
catheters inserted into a brachial or femoral nary artery. As it travels through the right
artery. Once in the blood vessel, the catheter side of the heart, recorded pressure measure-
is guided by fluoroscopy (x-ray images) to ments identify the catheter tip’s position (see
the area of study, where pressure measure- Box 3.1). Box 1
ments are made. Figure 3.14 depicts normal The normal right atrial (RA) pressure de-
Fig. 14 intracardiac and intravascular pressures. monstrates three positive deflections (see
The measurement of right-heart pressures Fig. 2.1): the a wave reflects right atrial con-
is performed with a specialized balloon- traction at the end of diastole, the c wave re-
tipped catheter (a common version of which sults from bulging of the tricuspid valve to-
is known as the Swan-Ganz catheter) that ward the right atrium as it closes in early
is advanced through the right side of the systole, and the v wave represents passive
heart with the aid of normal blood flow. filling of the right atrium from the systemic
The catheter is typically inserted percuta- veins during systole, when the tricuspid
neously into a peripheral vein (e.g., femoral, valve is closed. The negative deflection that
brachial, or internal jugular) and advanced follows the c wave is known as the x descent,
toward the chest. When it reaches a vein of and the negative deflection after the v wave

Aorta
PCW
100–140 2–10
60–90

PA LA
2–10
15–30
RA 4–12
LV
2–8
100–140
RV 3–12
15–30
2–8

Lungs Aorta
RA RV PA LA LV
15–30 15–30 PCW 100–140 100–140
2–8 2–10
2–8 4–12 2–10 3–12 60–90

Figure 3.14. Diagrams indicating normal pressures in the cardiac chambers


and great vessels. The top figure shows the normal anatomic relationship of the
cardiac chambers and great vessels, whereas the figure on the bottom shows a sim-
plified schematic to clarify the pressure relationships. Numbers indicate pressures in
mm Hg. LA, left atrial mean pressure; LV, left ventricular pressure; PA, pulmonary
artery pressure; PCW, pulmonary capillary wedge mean pressure; RA, right atrial
mean pressure; RV, right ventricular pressure.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 62

62 Chapter Three

Box 3.1 Intracardiac Pressure Tracings

When a catheter is inserted into a systemic vein and advanced into the right side of the
heart, each cardiac chamber produces a characteristic pressure tracing. It is important to
distinguish these tracings from one another to localize the position of the catheter tip and
to derive appropriate physiologic information.

ECG

Right ventricle Pulmonary artery


Pressure (mm Hg)

20
Pulmonary capillary wedge
a v
10
Right atrium
acv
x y

Time

is called the y descent. Often the a and c the pulmonary capillary wedge (PCW) tracing,
waves merge so that only two major positive which reflects the left atrial pressure (see Fig.
deflections are seen. In patients with atrial 3.14). Its characteristic shape is similar to the
fibrillation (see Chapter 12), the a wave is RA tracing, but the pressure values are usually
absent because there is no organized left higher and the tracing is often less clear (with
atrial contraction at the end of diastole. the c wave not observed) because of damped
As the catheter is advanced into the right transmission through the capillary vessels.
ventricle (RV), a dramatic increase in systolic
pressure is seen. The RV systolic waveform is
Right Atrial Pressure
characterized by a rapid upstroke and down-
stroke. In diastole, there is a gradual contin- Right atrial pressure is equal to the central
uous increase in RV pressure as the chamber venous pressure (estimated by the jugular
fills with blood. venous pressure on physical examina-
As the catheter is moved forward into the tion) because no obstructing valves impede
pulmonary artery (PA), the systolic pressure re- blood return from the veins into the right
mains the same as that in the RV (as long as atrium. Similarly, right atrial pressure nor-
there is no obstruction to RV outflow, such mally equals right ventricular pressure in
as pulmonic valve stenosis). However, three diastole because the right heart functions
characteristics of the tracing indicate entry as a “common chamber” when the tricus-
into the pulmonary artery: (1) the PA dias- pid valve is open. The mean right atrial
tolic pressure is higher than that of the RV; pressure is reduced when there is intra-
(2) the descending systolic portion of the PA vascular volume depletion. It is elevated
tracing inscribes a dicrotic wave, a small in right ventricular failure, right-sided val-
transient pressure increase that occurs after vular disease, and cardiac tamponade (in
the systolic peak and is related to pulmonic which the cardiac chambers are surrounded
valve closure; and (3) the diastolic portion of by high-pressure pericardial fluid; see Chap-
the PA tracing is downsloping compared ter 14).
with the upsloping RV diastolic pressure. Certain abnormalities cause characteris-
Further advancement of the catheter into tic changes in individual components of the
a branch of the pulmonary artery results in right atrial (and therefore jugular venous)
10090-03_CH03.qxd 8/31/06 5:24 PM Page 63

Diagnostic Imaging and Cardiac Catheterization 63

Tab. 3 pressure (Table 3.3). For example, a promi- sure increases when the right ventricle is sub-
nent a wave is seen in tricuspid stenosis and jected to pressure or volume overload and
right ventricular hypertrophy. In these con- may be a sign of right-heart failure.
ditions, the right atrium contracts vigor-
ously against the obstructing tricuspid valve
Pulmonary Artery Pressure
or stiffened right ventricle, respectively, gen-
erating a prominent pressure wave. Simi- Elevation of systolic and diastolic pulmonary
larly, amplified “cannon” a waves may be artery pressures occurs in three conditions:
produced by conditions of atrioventricular (1) left-sided heart failure; (2) parenchymal
dissociation (see Chapter 12), when the right lung disease (e.g., chronic bronchitis or end-
atrium contracts against a closed tricuspid stage emphysema); and (3) pulmonary vas-
valve. A prominent v wave is observed in tri- cular disease (e.g., pulmonary embolism,
cuspid regurgitation because normal right primary pulmonary hypertension, or acute
atrial filling is augmented by the regurgitated respiratory distress syndrome). Normally, the
blood in systole. pulmonary artery diastolic pressure is equiv-
alent to the left atrial pressure because of the
low resistance of the pulmonary vasculature
Right Ventricular Pressure
that separates them. If the left atrial pressure
Right ventricular systolic pressure is increased rises because of left-sided heart failure, both
by pulmonic valve stenosis or pulmonary hy- systolic and diastolic pulmonary artery pres-
pertension. Right ventricular diastolic pres- sures increase in an obligatory manner to

TABLE 3.3. Causes of Increased Intracardiac Pressures

Chamber and Measurement Causes

Right atrial pressure • Right ventricular failure


• Cardiac tamponade
a wave • Tricuspid stenosis
• Right ventricular hypertrophy
• Atrioventricular dissociation
v wave • Tricuspid regurgitation
• Right ventricular failure
Right ventricular pressure
Systolic • Pulmonic stenosis
• Right ventricular failure
• Pulmonary hypertension
Diastolic • Right ventricular failure
• Cardiac tamponade
• Right ventricular hypertrophy
Pulmonary artery pressure
Systolic and diastolic • Pulmonary hypertension
• Left-sided CHF
• Chronic lung disease
• Pulmonary vascular disease
Systolic only • Increased flow (L ∅ R shunt)
Pulmonary artery wedge pressure • Left-sided CHF
• Mitral stenosis or regurgitation
• Cardiac tamponade
a wave • Left ventricular hypertrophy
v wave • Mitral regurgitation
• Ventricular septal defect

CHF, congestive heart failure.


10090-03_CH03.qxd 8/31/06 5:24 PM Page 64

64 Chapter Three

maintain forward flow through the lungs. bed, left atrium, and left ventricle normally
This situation leads to “passive” pulmonary share the same pressures. Thus, the PCW can
hypertension. be used to estimate the left ventricular dias-
In certain conditions, however, pulmo- tolic pressure, a measurement of ventricular
nary vascular resistance becomes abnormally preload (see Chapter 9). As a result of this im-
high, causing pulmonary artery diastolic portant feature, monitoring of PCW is often
pressure to be elevated compared with left useful in managing critically ill patients in
atrial pressure. For example, pulmonary vas- the intensive care unit.
cular obstructive disease may develop as a Elevation of the mean PCW is seen in
complication of a chronic left-to-right car- left-sided heart failure and in mitral steno-
diac shunt, such as an atrial or ventricular sis or regurgitation. The individual compo-
septal defect (see Chapter 16). nents of the PCW tracing can also become
abnormally high. The a wave may be in-
creased in conditions of decreased left ven-
Pulmonary Artery Wedge Pressure
tricular compliance, such as left ventricular
If a catheter is advanced into the right or hypertrophy or acute myocardial ischemia.
left pulmonary artery, its tip will ultimately The v wave is greater than normal when
reach one of the small pulmonary artery there is increased left atrial filling during
branches and temporarily occlude forward ventricular contraction, as in mitral regur-
blood flow beyond it. During that time, a gitation.
column of stagnant blood stands between
the catheter tip and the portions of the pul-
Measurement of Blood Flow
monary capillary and pulmonary venous
Fig. 15 segments distal to it (Fig. 3.15). That col- Cardiac output is generally measured by
umn of blood acts as an “extension” of the either the thermodilution method or the Fick
catheter, and the pressure recorded through technique. In the thermodilution method,
the catheter reflects that of the downstream saline of a known temperature is injected
chamber—namely, the left atrium. Such a rapidly into the right heart via a catheter
pressure measurement is termed the pul- side hole located a specific distance proxi-
monary artery wedge pressure or pulmonary mal to the tip of the catheter. At the tip, a
capillary wedge pressure (PCW) and closely thermistor registers the surrounding tem-
matches the left atrial pressure in most peo- perature in the pulmonary artery, which is
ple. Furthermore, while the mitral valve is transiently altered by the injected saline.
open during diastole, the pulmonary venous The cardiac output is electronically calcu-

PA
LA

Pulmonary
artery catheter A pulmonary
vein

Catheter tip Shaded area represents


occludes branch “column of blood” between
of pulmonary artery catheter tip and LA
Pulmonary
capillaries
Figure 3.15. Diagram of a pulmonary artery catheter inserted into a branch of the pul-
monary artery (PA). Flow is occluded in the arterial, arteriolar, and capillary vessels beyond the
catheter; thus, these vessels act as a conduit that transmits the left atrial (LA) pressure to the
catheter tip.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 65

Diagnostic Imaging and Cardiac Catheterization 65

lated from the slope of the decay of the tem- Because the normal range of cardiac out-
perature change. put varies with a patient’s size, it is common
The Fick method is derived from the prin- to report the cardiac index, which is equal
ciple that consumption of oxygen by tissues to the cardiac output divided by the patient’s
is related to the O2 content removed from body surface area (normal range of cardiac
blood as it flows through the capillary bed. index = 2.6 − 4.2 L/min per square meter).

O2 consumption = O2 content removed


× Flow Calculation of
Vascular Resistance
⎛ mL O2 ⎞ ⎛ mL O2 ⎞ ⎛ mL blood ⎞
⎜⎝ ⎟=⎜ ⎟ ×⎜ ⎟
min ⎠ ⎝ mL blood ⎠ ⎝ min ⎠ Once pressures and cardiac output have been
determined, pulmonary and systemic vascu-
Or, in other terms: lar resistances can be calculated from the
following formulas:
O2 consumption = AVO2 difference
× Cardiac output MPAP − LAP
PVR = × 80
CO
in which the arteriovenous O2 (AVO2) dif- PVR, pulmonary vascular resistance (dynes-
ference equals the difference in oxygen con- sec-cm−5)
tent between the arterial and venous com- MPAP, mean pulmonary artery pressure
partments. Total body oxygen consumption (mm Hg)
can be determined by analyzing expired air LAP, mean left atrial pressure (mm Hg)
from the lungs, and arterial and venous O2 CO, cardiac output (L/min)
content is measured in blood samples. By re-
MAP − RAP
arranging the terms, the cardiac output can SVR = × 80
be calculated. CO
SVR, systemic vascular resistance (dynes-
O2 consumption
Cardiac output = sec-cm−5)
AVO2 difference MAP, mean arterial pressure (mm Hg)
RAP, mean right atrial pressure (mm Hg)
For example, if the arterial blood in a nor-
CO, cardiac output (L/min)
mal adult contains 190 mL of O2 per liter
and the venous blood contains 150 mL of The normal PVR ranges from 20 to 130 dynes-
O2 per liter, the arteriovenous difference is sec-cm−5. The normal SVR is 700 to 1,600
40 mL of O2 per liter. If this patient has a dynes-sec-cm−5.
measured O2 consumption of 200 mL/min,
the cardiac output is 5 L/min.
In many forms of heart disease, the car- Contrast Angiography
diac output is lower than normal. In that sit- This technique, performed at the time of
uation, the total body oxygen consumption catheterization, uses radiopaque contrast ma-
does not change significantly; however, a terial to visualize regions of the cardiovascu-
greater percentage of O2 is extracted per vol- lar system. A catheter is introduced into an
ume of circulating blood by the metaboliz- appropriate vessel and guided under fluo-
ing tissues. The result is a lower-than- roscopy to the site where the contrast will
normal venous O2 content and therefore an be injected. Following administration of
increased arteriovenous O2 difference. In the the contrast agent, x-rays are transmitted
example, if the patient’s venous blood O2 through the area of interest. A single expo-
content fell to 100 mL/L, the arteriovenous sure produces one film image, whereas a se-
O2 difference would be increased to 90 mL/L ries of x-ray exposures is recorded to produce
and the calculated cardiac output would be a “motion picture,” termed a cineangiogram
reduced to 2.2 L/min. (often simply called a cine).
10090-03_CH03.qxd 8/31/06 5:24 PM Page 66

66 Chapter Three

A specialized type of contrast angiogra- to examine the location and severity of coro-
phy, termed digital subtraction angiogra- nary atherosclerotic lesions. To maximize the
phy (DSA), was developed to provide a clear test’s sensitivity and reproducibility, each
image using less contrast material. In this patient is imaged in several standard views. If
technique, a computer processes the digital- a significant stenosis is detected, balloon
ized x-ray images and subtracts the back- angioplasty and stent placement can be per-
ground of soft tissue and bone, thus enhanc- formed, reopening the vessel (Figs. 3.17 and Fig. 17-18
ing the image of the blood vessel or chamber 3.18; see Chapter 6).
into which contrast material was injected. Some risk is associated with catheterization
DSA has advantages over conventional an- and contrast angiography. Complications
giography: smaller catheters may be used, are uncommon but include myocardial per-
the amount of contrast agent required may foration by the catheter, precipitation of ar-
be lower, and better image quality is usually rhythmias and conduction blocks, damage to
achieved. vessel walls, hemorrhage, dislodgement of
Selective injection of contrast material atherosclerotic plaques, and infection. Com-
into the heart chambers is used to identify plications resulting from the contrast medium
valvular insufficiency, abnormal wall thick- itself include anaphylaxis and renal toxicity.
ening, intracardiac shunts, thrombi within Table 3.4 summarizes the catheterization Tab. 4
the heart, and congenital malformations, findings in common cardiac abnormalities.
and also to measure ventricular contractile Therapeutic interventional catheterization
function. To image the right heart cham- techniques are described in Chapter 6.
bers, injection is made through a catheter
inserted into the inferior or superior vena
NUCLEAR IMAGING
cava, the right atrium, or the right ventricle.
The left side of the heart can be imaged by Heart function can be evaluated using in-
contrast injection through a catheter ad- jected, radioactively labeled tracers and
vanced into the left ventricle (Fig. 3.16). γ-camera detectors. The resulting images re-
A widespread application of contrast in- flect the distribution of the tracers within
jection is coronary artery angiography, used the cardiovascular system. Nuclear tech-

Figure 3.16. Left ventriculogram, in diastole (A) and systole (B) in the right anterior oblique projec-
tion, from a patient with normal ventricular contractility. A catheter (arrow) is used to inject contrast into
the left ventricle (LV). The catheter can also be seen in the descending aorta (arrowhead). AO, aortic root.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 67

Diagnostic Imaging and Cardiac Catheterization 67

LCX

LM
LM
8

Diagonal
LAD branch
LAD

Septal
perforators

A B

Figure 3.17. Cardiac catheterization and stenting of a proximal left anterior descending artery (LAD)
stenosis, shown in an anteroposterior cranial projection. A. When contrast agent is injected into the left
main coronary artery (LM), the left circumflex artery (LCX) fills normally but the LAD is almost completely occluded
at its origin (white arrow). B. After the stenosis is successfully stented, the LAD and its branches fill robustly. (Images
courtesy of Jeffrey Popma, MD, Brigham and Women’s Hospital, Boston, MA.)

A B

Figure 3.18. Cardiac catheterization and stenting of right coronary artery (RCA) stenoses. Both images are
obtained in the left anterior oblique (LAO) projection. A. The stenotic segment is located between the white arrows.
B. After stenting, the caliber of the vessel and flow have improved. (Courtesy of Jeffrey Popma, MD, Brigham and
Women’s Hospital, Boston, MA.)
10090-03_CH03.qxd 8/31/06 5:24 PM Page 68

68 Chapter Three

TABLE 3.4. Cardiac Catheterization and Angiography in Cardiac Disorders

Disorder Finding

Coronary artery disease • Identification of atherosclerotic lesions


Mitral regurgitation • Large systolic v wave in left atrial pressure tracing
Mitral stenosis • Abnormally high pressure gradient between left atrium and left ventricle
in diastole
Tricuspid insufficiency • Large systolic v wave in the right atrial pressure tracing
Aortic stenosis • Systolic pressure gradient between left ventricle and aorta
Congestive heart failure • Estimation of cardiac output
• Calculation of systemic and pulmonary vascular resistances

niques are used to assess myocardial perfu- mogenous distribution of thallium in the
sion, to image blood passing through the myocardial tissue. Conversely, myocardial
heart and great vessels, to localize and quan- regions that are scarred (by previous infarc-
tify myocardial ischemia and infarction, tion) or have reduced perfusion during ex-
and to assess myocardial metabolism. ercise (i.e., transient myocardial ischemia)
do not accumulate as much thallium as nor-
mal heart muscle. Consequently, these areas
Assessment of
will appear on the thallium scan as light or
Myocardial Perfusion
“cold” spots.
Ischemia and infarction resulting from coro- When evaluating for myocardial ischemia,
nary artery disease can be detected by myo- an initial set of images is taken right after ex-
cardial perfusion imaging using various ra- ercise and 201Tl injection. Well-perfused myo-
dioisotopes, including compounds labeled cardium will take up more tracer than is-
thallium-201 (201Tl) and technetium-99m chemic or infarcted myocardium at this
(99mTc). Currently, 99mTc-sestamibi and 99mTc- time. Delayed images are acquired several
tetrofosmin are widely used. Both 201Tl- and hours later, because 201Tl accumulation does
99mTc-labeled compounds are sensitive for
not remain fixed in myocytes. Rather, con-
the detection of ischemic or scarred myo- tinuous redistribution of the isotope occurs
cardium, but each compound has certain across the cell membrane. After 3 to 4 hours
advantages. For example, the 99mTc-labeled of redistribution, when additional images
agents provide better image quality and are are obtained, all viable myocytes will have
superior for detailed single photon emis- equal concentrations of 201Tl. Consequently,
sion computed tomography (SPECT, as in any uptake abnormalities on the initial pos-
Fig. 3.19). Conversely, superior detection of texercise scan that were caused by myocar-
myocardial cellular viability has been demon- dial ischemia will have resolved (“filled in”)
strated with 201Tl imaging. on the delayed scan (and are therefore termed
In the case of 201Tl imaging, the radioiso- “reversible” defects), and those representing
tope is injected intravenously while the pa- infarcted or scarred myocardium will persist as
tient is exercising on a treadmill or stationary cold spots.
bicycle. Because thallium is a potassium ana- Of note, some myocardial segments that
logue, it enters normal myocytes, a process demonstrate persistent 201Tl defects on both
thought to be partially governed by the stress and redistribution imaging are falsely
sodium-potassium ATPase pump. The intra- characterized as nonviable, scarred tissue.
cellular concentration of thallium, estimated Sometimes these areas represent ischemic,
by the density of the image, depends on vas- noncontractile, but metabolically active
cular supply (perfusion) and membrane areas that have the potential to regain func-
function (tissue viability). In the normal tion if an adequate blood supply is restored.
heart, the radionuclide scan shows a ho- For example, such areas may represent hi-
10090-03_CH03.qxd 8/31/06 5:24 PM Page 69

Diagnostic Imaging and Cardiac Catheterization 69

SHORT AXIS
B Anterior

STRESS Sep Lat

A Inferior

REST

HORIZONTAL
LONG AXIS
C Apex

STRESS Sep Lat

REST

Figure 3.19. Stress and rest myocardial perfusion single photon emission computed tomography images
(using 99mTc-tetrofosmin) of a patient with a high-grade stenosis within the proximal left anterior de-
scending coronary artery. A. Miniaturized reproduction of the complete scan showing tomographic images in
each of three views. The first, third, and fifth rows demonstrate images during stress, and the second, fourth, and
sixth rows are matching images acquired at rest. B and C. Enlarged selected panels from part A showing stress and
rest images in the short axis and horizontal long axis views. The arrows indicate regions of decreased perfusion dur-
ing stress but normal perfusion on the matching resting scans, consistent with inducible ischemia. Lat, lateral wall
of the LV; Sep, septal wall. (Courtesy of Marcelo Di Carli, MD, Brigham and Women’s Hospital, Boston, MA.)

bernating myocardium, segments that de- The uptake mechanism differs in that the
monstrate diminished contractile function compound crosses the myocyte membrane
owing to chronic reduction of coronary passively, driven by the negative membrane
blood flow (see Chapter 6). This viable potential. Once inside the cell, it further ac-
state (in which the affected cells can be cumulates in mitochondria, driven by that
predicted to regain function following organelle’s even more negative membrane
coronary revascularization) can often be potential. The myocardial distribution of
differentiated from irreversibly scarred myo- MIBI reflects perfusion at the moment of
cardium by repeat imaging at rest after the injection, and in contrast to thallium, it re-
injection of additional 201Tl to enhance up- mains fixed intracellularly; that is, it does not
take by viable cells. redistribute over time. Consequently, per-
99mTc-sestamibi (commonly referred to as
forming a MIBI procedure is more flexible, as
MIBI) is the most widely used 99mTc-labeled images can be obtained up to 4 to 6 hours
compound. This agent is a large lipophilic after injection and repeated as necessary. A
molecule that, like thallium, is taken up in MIBI study is usually performed as a 1-day
the myocardium in proportion to blood flow. protocol in which an initial injection, using
10090-03_CH03.qxd 8/31/06 5:24 PM Page 70

70 Chapter Three

a small tracer dose, and imaging are per- therapy, for risk stratification, and to predict
formed at rest. Then, a few hours later, the which patients would benefit from early me-
patient exercises and repeat imaging is per- chanical revascularization.
formed after injection of a larger tracer
dose. Radionuclide Ventriculography
Stress nuclear imaging studies with either
201Tl- or 99mTc-labeled compounds have Radionuclide ventriculography (RVG, also
greater sensitivity and specificity than stan- known as blood pool imaging) is used to an-
dard exercise electrocardiography for the de- alyze right and left ventricular function. A
tection of ischemia but are more expensive radioisotope (usually 99mTc) is bound to red
and should be ordered judiciously. Nuclear blood cells or to human serum albumin and
imaging is particularly appropriate for pa- then injected as a bolus. Nuclear images are
tients with certain baseline electrocardio- obtained at fixed time intervals as the la-
gram (ECG) abnormalities that preclude ac- beled material passes through the heart
curate interpretation of a standard exercise and great vessels. Multiple images are dis-
test. Examples include patients with elec- played sequentially to produce a dynamic
tronic pacemaker rhythms, those with left picture of blood flow. Calculations, such as
bundle branch block, and those who take determination of the ejection fraction, are
certain medications that alter the ST seg- based on the difference between radioactive
ment, such as digoxin. Nuclear scans also counts present in the ventricle at end dias-
provide more accurate anatomic localiza- tole and at end systole. Therefore, measure-
tion of the ischemic segment(s) and quan- ments are largely independent of any as-
tification of the extent of ischemia com- sumptions of ventricular geometry and are
pared with standard exercise testing. In highly reproducible. Studies suggest that
addition, electronic synchronizing (gating) RVG and echocardiography provide similar
of nuclear images to the ECG cycle permits left ventricular ejection fraction values, but
wall motion analysis. unlike echocardiography, RVG can also cal-
Patients with orthopedic or neurologic culate an accurate right ventricular ejection
conditions, as well as those with severe phys- fraction.
ical deconditioning or chronic lung disease, Radionuclide ventriculography is com-
may be unable to perform an adequate exer- monly used to assess baseline cardiac func-
cise test on a treadmill or bicycle. In such pa- tion in patients scheduled to undergo po-
tients, stress images can be obtained instead tentially cardiotoxic chemotherapy (e.g.,
by administering pharmacologic agents, such doxorubicin) and to follow cardiac function
as adenosine or dipyridamole. These agents over time in such patients. In addition, first-
induce diffuse coronary vasodilation, aug- pass imaging and scans gated to the ECG
menting blood flow to myocardium per- permit recognition of abnormal cardiac and
fused by healthy coronary arteries. Since vascular shunts.
ischemic regions are already maximally di-
lated (because of local metabolite accumula- Assessment of
tion), the drug-induced vasodilation causes a Myocardial Metabolism
“steal” phenomenon, reducing isotope up-
take in regions distal to significant coronary Positron emission tomography (PET) is a
stenoses (see Chapter 6). Alternatively, dobu- specialized nuclear imaging technique used
tamine (see Chapter 17) can be infused intra- to assess myocardial perfusion and viability.
venously to increase myocardial oxygen de- PET imaging employs positron-emitting iso-
mand to test for ischemia. topes (e.g., oxygen-15, carbon-11, rubidium-
In addition to its role in the diagnosis of 82, nitrogen-13, and fluorine-18) attached
myocardial ischemia, nuclear imaging can to metabolic or flow tracers. Sensitive detec-
be useful after acute myocardial infarction tors measure positron emission from the tra-
to assess the effectiveness of thrombolytic cer molecules.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 71

Diagnostic Imaging and Cardiac Catheterization 71

Myocardial perfusion is commonly as- tractile dysfunction owing to decreased flow


sessed using nitrogen-13–labeled ammonia represent scar tissue, or whether the region
or rubidium-82. These flow tracers are taken is still viable (e.g., hibernating myocar-
up by myocytes in proportion to blood dium). In scar tissue, both blood flow to the
flow. Myocardial viability can be determined affected area and 18FDG uptake are decreased.
by PET by studying glucose utilization in Because the myocytes in this region are per-
myocardial tissue. In normal myocardium manently damaged, such tissue is not likely
under fasting conditions, glucose is used to benefit from a revascularization proce-
for approximately 20% of energy produc- dure. Hibernating myocardium, in contrast,
tion, with free fatty acids providing the shows decreased blood flow but normal or
remaining 80%. In ischemic conditions, elevated 18FDG uptake. Such tissue may ben-
however, metabolism shifts toward glucose efit from mechanical revascularization, as
use, and the more ischemic the myocardial described in Chapter 6.
tissue, the stronger the reliance on glucose. Table 3.5 summarizes the radionuclide- Tab. 5
Fluoro-18 deoxyglucose (18FDG), created imaging abnormalities associated with com-
by substituting fluorine-18 for hydrogen in mon cardiac conditions.
2-deoxyglucose, is used to study glucose
uptake. This substance competes with
COMPUTED TOMOGRAPHY
glucose both for transport into myocytes
and for subsequent phosphorylation. Un- Computed tomography (CT) uses thin
like glucose, however, 18FDG is not metab- x-ray beams to obtain axial plane images.
olized and becomes trapped within the An x-ray tube is programmed to rotate
myocyte. around the body, and the generated beams
Combined evaluation of perfusion and are partially absorbed by body tissues. The
18FDG metabolism allows assessment of remaining beams emerge and are captured
both regional blood flow and glucose up- by electronic detectors, which relay infor-
take, respectively. PET scanning thus helps mation to a computer for image composi-
determine whether areas of ventricular con- tion. CT scanning typically requires admin-

TABLE 3.5. Nuclear Imaging in Cardiac Disorders

Disorder Finding

Myocardial ischemia
Stress-delayed reinjection 201Tl • Low uptake during stress with complete or partial fill-in with
delayed or reinjection images
Rest-stress 99mTc-labeled compounds • Normal uptake at rest with decreased uptake during stress
PET (N-13 ammonia/18FDG) • Decreased flow with normal or increased 18FDG uptake during
stress
Myocardial infarction
Stress-delayed reinjection 201Tl • Low uptake during stress and low uptake after reinjection
Rest-stress 99mTc-labeled compounds • Low uptake in rest and stress images
PET (N-13 ammonia/18FDG) • Decreased flow and decreased 18FDG uptake at rest
Hibernating myocardium
Rest-delayed 201Tl • Complete or partial fill-in of defects after reinjection
PET (N-13 ammonia/18FDG) • Decreased flow and increased 18FDG uptake at rest
Assessment of ventricular unction
99m
Tc RBC gated radionuclide • Assessment of global left and right ventricular function at rest
or during exercise
AQ4 Radionuclide ventriculography • Regional wall motion

18
FDG, fluoro-18 deoxyglucose; N-13, nitrogen-13; PET, positron emission tomography; RBC, red blood cell; 99m Tc, tech-
netium-99m; 201Tl, thallium-201.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 72

72 Chapter Three

istration of an intravenous contrast agent to images. A limitation of conventional CT


distinguish intravascular contents (i.e., techniques is the artifact generated by pa-
blood) from neighboring soft tissue struc- tient motion (i.e., breathing) during image
tures (e.g., myocardium). acquisition. Modern spiral CT imaging al-
Applications of CT in cardiac imaging in- lows more rapid image acquisition, often
clude assessment of the great vessels, peri- during a single breath-hold, at relatively
cardium, myocardial structures, and coro- lower radiation doses than conventional CT.
nary arteries. CT can be used to accurately Spiral CT is particularly important in the di-
diagnose aortic dissections and aneurysms agnosis of pulmonary embolism. When an
Fig. 20 (Fig. 3.20) and to monitor patients who intravenous iodine-based contrast agent is
have undergone surgical repair of these con- administered, emboli create the appearance
ditions. CT clearly delineates pericardial ef- of “filling defects” in otherwise contrast-
fusions as well as pericardial thickening and enhanced pulmonary vessels (Fig. 3.21). Fig. 21
calcification. Myocardial abnormalities, Electron beam computed tomography
such as regional hypertrophy or ventricular (EBCT), a technology developed particu-
aneurysms, and intracardiac thrombus for- larly for cardiac imaging, uses a direct elec-
mation can be distinctly visualized on CT tron beam to acquire images in a matter of

C D
AA
PA

AA AA

B RK

RK
Liver LK
LK

RCI LCI
LCI

LEI

Figure 3.20. Computed tomography (CT) imaging of aortic dissection. A and B. Axial images demonstrate an in-
timal flap (colored arrowheads) separating the true and false lumens. C. CT angiography (CTA) with three-dimensional
reconstructions. In this left anterior oblique view, the origin of the dissection (colored arrowhead) is apparent in the dis-
tal portion of the aortic arch. The dissection continues to the level of the renal arteries (white arrowhead) and beyond.
D. In this CTA left posterior oblique view, the dissection extends to the infrarenal aorta (white arrowhead) and involves
the left common and external iliac arteries (colored arrowhead). AA, ascending aorta; LCI, left common iliac artery; LEI,
left external iliac artery; LK, left kidney; PA, main pulmonary artery; RCI, right common iliac artery; RK, right kidney.
(Courtesy of Suhny Abbara, MD, Massachusetts General Hospital, Boston.) one line sh
10090-03_CH03.qxd 8/31/06 5:24 PM Page 73

Diagnostic Imaging and Cardiac Catheterization 73

Figure 3.21. Spiral computed tomography image demonstrating a massive pul-


monary embolism. The white arrows point to a large thrombus within the right pul-
monary artery. It appears as a filling defect within the otherwise contrast-enhanced pul-
monary vasculature. AA, ascending aorta; DA, descending aorta; LPA, left pulmonary
artery; PA, main pulmonary artery; RPA, right pulmonary artery; SVC, superior vena cava.

milliseconds. Rapid succession of images de- tector row CT scanners acquire as many as
picts cardiac structures at multiple times 64 anatomic sections with each rotation,
during a single cardiac cycle. Displaying providing excellent spatial resolution. Ad-
these images in a cine (“motion picture”) ministration of intravenous contrast and
format can provide estimates of left ventric- computer reformatting allows visualiza-
ular volumes, including stroke volume, and tion of the arterial lumen and regions of
ejection fraction, expanding the application coronary narrowings (Fig. 3.22). Because Fig. 22
of CT to include not only heart structure but image acquisition is timed with the cardiac
also function. Capable of detecting coronary cycle, a relatively low heart rate is desir-
artery calcification, EBCT has been used pri- able, such that a β-blocker is often given
marily to screen for coronary artery disease. prior to scanning.
Because calcified coronary artery plaques CT is not as sensitive as conventional
have a radiodensity similar to that of bone, angiography for the detection of coronary
they appear attenuated (white) on CT. The lesions, and it cannot adequately evaluate
Agatson score, a measure of total coronary stenosis within coronary artery stents. In
artery calcium, correlates well with athero- addition, this technique results in signifi-
sclerotic plaque burden, and predicts the cant radiation exposure. However, CT is
risk of nonfatal myocardial infarction and rapid, relatively inexpensive, and signifi-
cardiac death, independently of other cardiac cantly less invasive than conventional an-
risk factors. giography. Its role in assessing patients
Beyond assessing the coronary calcium with symptoms suggestive of coronary artery
score, newer CT technology can character- disease and for following the progression of
ize individual atherosclerotic plaques and known coronary disease is currently under
line short stenoses in great detail. Current multide- evaluation.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 74

Ao
RCA Ao
RA
LA LCX
LM LM LA
RCA RV
PA
LAD LCX
LAD
RV
LV LV

A B

Ao

PA

LAD

Figure 3.22. Computed tomography (CT) coronary angiography.


After a patient is imaged in a high-resolution axial CT scanner, three-di-
mensional reconstructions (termed volume renderings) are generated by
a computer. A. Volume rendering of a normal CT angiogram. B. Volume
rendering of a CT angiogram that demonstrates diffuse coronary artery
disease. Notice that the caliber of each vessel is irregular along its length.
C. This curved reformat of the left anterior descending artery (LAD) de-
picts the entire course of the vessel in a single, flat image, making it eas-
ier to detect stenoses. None are present here. Ao, aorta; LA, left atrium;
LCX, left circumflex artery; LM, left main coronary artery; LV, left ventri-
cle; PA, pulmonary artery; RA, right atrium; RCA, right coronary artery;
C RV, right ventricle. (Courtesy of Suhny Abbara, MD, Massachusetts Gen-
eral Hospital, Boston.)

MAGNETIC RESONANCE IMAGING gen nuclei, which align themselves with an


applied magnetic field. Radiofrequency ex-
Magnetic resonance imaging (MRI) uses a citation causes the nuclei to move out of
powerful magnetic field to obtain detailed alignment momentarily. As they return to
images of internal structures. This technique their resting states, the nuclei emit radio
is based on the magnetic polarity of hydro- waves, which are translated into computer-
10090-03_CH03.qxd 8/31/06 5:24 PM Page 75

Diagnostic Imaging and Cardiac Catheterization 75

generated images. Therefore, MRI requires mit evaluation of valvular and ventricular
no ionized radiation. Among all the imaging function.
modalities, MRI is best at differentiating tis- Two applications of cardiac MRI deserve
sue contrasts (blood, fluid, fat, and my- special mention. Coronary magnetic reso-
ocardium) and can often do so even without nance angiography (coronary MRA) is
the use of contrast agents. The addition of a noninvasive, contrast-free angiographic-
gadolinium-based (noniodinated) contrast imaging modality. Laminar blood flow ap-
agents allows further characterization of car- pears as bright signal intensity, whereas
diac structures and tissues using the latest turbulent blood flow, at the site of stenosis,
MRI techniques. results in less bright or absent signal inten-
The detail of soft tissue structures is often sity. This technique has shown high sensi-
exquisitely demonstrated in magnetic reso- tivity and accuracy for the detection of im-
Fig. 23 nance images (Fig. 3.23). Cardiac MRI has portant coronary artery disease in the left
an established role in assessing congenital main coronary artery and in the proximal
anomalies, such as shunts, and diseases of and midportions of the three major coro-
the aorta, including aneurysm and dissec- nary vessels. Coronary MRA is also useful
tion. It is also used to assess left and right in delineating coronary artery congenital
ventricular mass and volume, intravascular anomalies.
thrombus, cardiomyopathies, and neoplas- In contrast-enhanced MRI, a gadolinium-
Fig. 24 tic disease (Fig. 3.24). ECG-gated and cine based agent is administered intravenously
MRI techniques capture images at discrete to differentiate between impaired (but vi-
times in the cardiac cycle, and therefore per- able) myocardial segments and truly in-

Figure 3.23. Cardiac magnetic resonance images of a normal person. A. Three-chamber


long axis view of the heart in diastole and systole showing the left ventricle (LV), right ventricle
(RV), and left atrium (LA). The mitral valve (MV), aortic valve (AV), ascending aorta (AAO), and
descending aorta (DA) are also imaged. B. Midventricular short axis view demonstrating the LV,
RV, and left ventricular papillary muscles (PMs). PW, posterior wall; S, septum. (Courtesy of
Raymond Y. Kwong, MD, Brigham and Women’s Hospital, Boston, MA.)
10090-03_CH03.qxd 8/31/06 5:24 PM Page 76

76 Chapter Three

LV LV

RV RV

RA RA

LA LA

A B

Figure 3.24. Magnetic resonance imaging of an intracardiac mass. Both images are apical four-chamber views.
A. Before a gadolinium-based contrast agent is administered, an abnormal left atrial mass (indicated by the oval)
demonstrates diminished signal relative to the surrounding tissue. In this respect, it resembles a nonvascular thrombus.
B. After contrast injection, the mass enhances similar to the surrounding tissue, indicating that it is vascularized. Biopsy
revealed a spindle-cell carcinoma. LA, left atrium; LV, left ventricle; RA, right atrium; RV, right ventricle. (Courtesy of
Raymond Kwong, MD, Brigham and Women’s Hospital, Boston, MA.)

fracted tissue. This technique is based on rently available to assess cardiac structure
findings that gadolinium is excluded from and function. Many of these tools are ex-
viable cells with intact cell membranes but pensive and yield similar information. For
can permeate and concentrate in infarcted example, estimates of ventricular contractile
(i.e., irreversibly damaged) areas. Nonviable function can be made by echocardiogra-
myocardial segments therefore appear “hy- phy, nuclear imaging, contrast angiogra-
perenhanced” relative to viable, reversibly phy, gated CT, or MRI. Myocardial viability
Fig. 25 impaired myocardium (Fig. 3.25). In this can be assessed using nuclear-imaging stud-
way, gadolinium-enhanced MRI can help ies, gadolinium MRI, or dobutamine echo-
select patients who are likely to benefit from cardiography.
Determining the single best test for any
revascularization procedures.
given patient depends on a number of fac-
tors. One is the ease by which images may be
obtained. In a critically ill patient, bedside
SUMMARY echocardiography provides an easily ob-
This chapter has presented an overview of tained measure of left ventricular systolic
imaging and catheterization techniques cur- function. Obtaining similar information
10090-03_CH03.qxd 8/31/06 5:24 PM Page 77

Diagnostic Imaging and Cardiac Catheterization 77

LV LV
RV RV

A B

Figure 3.25. Gadolinium-enhanced magnetic resonance images


demonstrating a region of nonviable myocardium. Both images are
short axis views. A. Imaging before administration of gadolinium demon-
strates thinning of the anterior and anteroseptal myocardium (colored
arrow) suggestive of infarcted tissue. B. After contrast injection, the ante-
rior and anteroseptal segments of the left ventricle selectively enhance
(white arrows), indicating that scar tissue is present. Because more than
half the thickness of the ventricular wall is scarred, coronary revascular-
ization would have a low likelihood of improving contractile function of
these myocardial segments. LV, left ventricle; RV, right ventricle. (Courtesy
of Raymond Kwong, MD, Brigham and Women’s Hospital, Boston, MA.)

from a nuclear study would require radio- choice of an imaging approach. When used
isotope administration and a trip to the appropriately, each imaging tool can pro-
nuclear scanner. Another factor to consider vide important information to guide the di-
is the degree of invasiveness of a given agnosis and management of cardiovascular
imaging technique. Expense, available disorders.
equipment, and institutional preference and Table 3.6 summarizes the uses of imaging Tab. 6
expertise also play roles in determining the techniques described in this chapter.

TABLE 3.6. Summary of Cardiac Imaging Techniques

Imaging Technique Finding Examples of Clinical Uses

Chest radiography • Cardiac and mediastinal contours • Detect chamber dilatation


• Pulmonary vascular markings • Identify consequences of stenotic
and regurgitant valve lesions and
intracardiac shunts
Transthoracic • Wall thickness, chamber dimensions • Assess global and segmental ventric-
echocardiography ular contraction
(TTE) • Anatomic relationships and motion • Identify valvular abnormalities and
of cardiac structures vegetations
• Flow direction, turbulence, and • Diagnose consequences of myocar-
velocity measurements dial infarction (e.g., ventricular
aneurysm, papillary muscle rupture,
intraventricular thrombus)
• Echo contrast studies • Identify myocardial, pericardial, and
congenital abnormalities
• Stress echocardiography
Transesophageal • Similar to TTE but higher resolution • Visualize intracardiac thrombus
echocardiography
(TEE)

(Continued)
10090-03_CH03.qxd 8/31/06 5:24 PM Page 78

78 Chapter Three

TABLE 3.6. (Continued)

Imaging Technique Finding Examples of Clinical Uses

Cardiac catheterization • Pressure measurement • Evaluate intracardiac pressures


(e.g., in valvular disease, heart fail-
ure, pericardial disease)
• Contrast angiography • Visualize ventricular contractile
function, regurgitant valve lesions
Nuclear SPECT imaging • Regional myocardial perfusion • Detect, quantify, and localize myo-
(using 99mTc-labeled cardial ischemia
compounds or 201Tl)
• Myocardial viability • Perform stress testing in patients
with baseline ECG abnormalities
Radionuclide • Ventricular contractile function • Calculate ventricular ejection fraction
ventriculography and quantitate intracardiac shunts
Positron emission • Myocardial perfusion and meta- • Evaluate contractile function
tomography (PET) bolism
Computed tomo- • Anatomy and structural relation- • Diagnose disease of the great vessels
graphy (CT) ships (aortic dissection, pulmonary
embolism)
Magnetic resonance • Detailed soft tissue anatomy • Assess and stenoses (multidetector
imaging (MRI) CT) myocardial structure and func-
tion (e.g., ventricular mass and vol-
ume, neoplastic disease, intracardiac
thrombus, cardiomyopathies)

ECG, electrocardiogram; SPECT, single photon emission computed tomography; 99mTc, technetium-99m; 201Tl, thallium-201.

Acknowledgment Nuclear Cardiology endorsed by the American


Heart Association. J Am Coll Cardiol 2005;46:
AQ1 The authors are grateful to Suhny Abbara, MD; 1587–1605.
Sharmila Dorbala, MD; Raymond Kwong, MD: and Clouse ME, Chen J, Krumholz HM. How useful is
Jeffrey Popma, MD, for their helpful suggestions. computed tomography for screening for coronary
Contributors to the previous editions of this chapter artery disease? Circulation 2006;113:125–146.
were Deborah Bucino, MD; Sharon Horesh, MD;
DiMario C, Sutaria N. Coronary angiography in the
Shona Pendse, MD; Albert S. Tu, MD; Patrick Yachim-
angioplasty era: projections with a meaning.
ski, MD, and Patricia C. Come, MD.
Heart 2005;91:968–976.
Edelman RR. Contrast-enhanced MR imaging of the
heart: overview of the literature. Radiology 2004;
Additional Reading 232:653–668.

Armstrong WF, Zoghbi WA. Stress echocardiogra- Enriquez-Sarano M, Avierinos J-F, Messika-Zeitoun D,
phy: current methodology and clinical applica- et al. Quantitative determinants of the outcome of
tions. J Am Coll Cardiol 2005;45:1739–1747. asymptomatic mitral regurgitation. N Engl J Med
2005;352:875–883.
Aviles RJ, Messerli AW, Askari AT, et al. Introductory
Guide to Cardiac Catheterization. Philadelphia: Feigenbaum H. Feigenbaum’s Echocardiography.
Lippincott Williams & Wilkins, 2005. 6th Ed. Philadelphia: Lippincott Williams &
Wilkins, 2004.
Baim D, Grossman W. Grossman’s Cardiac Catheter-
ization, Angiography and Intervention. 7th Ed. Ghesani M, DePuey G, Rozanski A. Role of F-18 FDG
Philadelphia: Lippincott Williams & Wilkins, 2005. positron emission tomography (PET) in the as-
sessment of myocardial viability. Echocardiogra-
Brindis RG, Douglas PS, Hendel RC, et al. ACCF/ASNC
phy 2005;22:165–177.
appropriateness criteria for single-photon emis-
sion computed tomography myocardial perfusion Hillis GS, Bloomfield P. Basic transthoracic echocar-
imaging (SPECT MPI): a report of the American diography. BMJ 2005;330:1432–1436.
College of Cardiology Foundation Quality Strate- Hoffmann MH, Shi H, Schmitz BL, et al. Noninva-
gic Directions Committee Appropriateness Crite- sive coronary angiography with multislice com-
ria Working Group and the American Society of puted tomography. JAMA 2005;293:2471–2478.
10090-03_CH03.qxd 8/31/06 5:24 PM Page 79

Diagnostic Imaging and Cardiac Catheterization 79

Otto CM. The Practice of Clinical Echocardiography. cardiology. Arch Intern Med 2005;165:2345–
3rd Ed. Philadelphia: Elsevier Saunders, 2004. 2353.
Quiroz R, Kucher N, Zou KH, et al. Clinical validity Schoenhagen P, Halliburton SS, Stillman AE, et al.
of a negative computed tomography scan in pa- Noninvasive imaging of coronary arteries: current
tients with suspected pulmonary embolism: a sys- and future role of multi-detector row CT. Radiol-
tematic review. JAMA 2005;293:2012–2017. ogy 2004;232:7–17.
Raggi P. Role of electron-beam computed tomogra- Schuijf JD, Shaw LJ, Wijns W, et al. Cardiac imaging
phy and nuclear stress testing in cardiovascular in coronary artery disease: differing modalities.
risk assessment. Am J Cardiol 2005;96:20–27. Heart 2005;91:1110–1117.
Raggi P, Taylor A, Fayad Z, et al. Atherosclerotic Sengupta PP, Khandheria BK. Transesophageal echo-
plaque imaging: contemporary role in preventive cardiography. Heart 2005;91:541–547.
10090-03_CH03.qxd 8/31/06 5:24 PM Page a

Chapter 3—Author Queries


1. AU: Correct that these folks are MDs?
2. AU: PMV is a label on the figure that needs defining here. Is the one added here correct?
3. ED: Are both w
Ò ith permission ” and “by permission” necessary?
4. AU: Added text correct?
10090-04_CH04.qxd 8/31/06 5:25 PM Page 80

C H A P T E R

The Electrocardiogram
Lilit Garibyan
Leonard S. Lilly
4
ELECTRICAL MEASUREMENT— Calibration
SINGLE-CELL MODEL Heart Rhythm
ELECTROCARDIOGRAM LEAD Heart Rate
REFERENCE SYSTEM Intervals (PR, QRS, QT)
Mean QRS Axis
SEQUENCE OF NORMAL CARDIAC ACTIVATION Abnormalities of the P Wave
INTERPRETATION OF THE Abnormalities of the QRS Complex
ELECTROCARDIOGRAM ST Segment and T Wave Abnormalities

Cardiac contraction relies on the organized On the right side of the diagram, a voltmeter
flow of electrical impulses through the heart. records the electrical potential across the
The electrocardiogram (ECG) is an easily ob- cell on graph paper. In the resting state, the
tained recording of that activity, and it pro- cell is polarized; that is, the entire outside of
vides a wealth of information about cardiac the cell is electrically positive with respect to
structure and function. This chapter presents the inside, because of the ionic distribution
the electrical basis of the ECG in health and across the cell membrane, as described in
disease and leads the reader through the ba- Chapter 1. In this resting state, the volt-
sics of interpretation. To become fully adept meter electrodes, which are placed on op-
at this technique and to practice the princi- posite outside surfaces of the cell, do not de-
ples described here, you may wish to consult tect any electrical activity, because there is
one of the complete electrocardiographic no electrical potential difference between
textbooks listed at the end of the chapter. them (the myocyte surface is homogeneously
charged).
This equilibrium is disturbed, however, by
ELECTRICAL MEASUREMENT—
stimulation of the cell (see Fig. 4.1B). During
SINGLE-CELL MODEL
the action potential, as cations rush across
This section begins by observing the propa- the sarcolemma into the cell, the polarity
gation of an electrical impulse within a single at the stimulated region transiently reverses,
Fig. 1 cardiac muscle cell, illustrated in Figure 4.1. such that the outside becomes negatively

80
10090-04_CH04.qxd 8/31/06 5:25 PM Page 81

The Electrocardiogram 81

E
Figure 4.1. Depolarization of a single cardiac muscle cell. A. In
the resting state, the surface of the cell is positively charged relative to
the inside. Because the surface is homogeneously charged, the volt-
meter electrodes outside the cell do not record any electrical potential
difference (“flat line” recording). B. Stimulation of the cell initiates de-
polarization (shaded area); the outside of the depolarized region be-
comes negatively charged relative to the inside. Because the current
of depolarization is directed toward the (+) electrode of the voltmeter,
an upward deflection is recorded. C. Depolarization spreads, creating
a greater upward deflection by the recording electrode. D. The cell has
become fully depolarized. The surface of the cell is now completely
negatively charged compared with the inside. Because the surface is
again homogeneously charged, a flat line is recorded by the voltmeter.
E. Notice that if the position of the voltmeter electrodes had been re-
versed, the wave of depolarization would have traveled away from the
(+) electrode, causing the deflection to be downward.

charged with respect to the inside; that is, By convention, the direction of electrical
the region depolarizes. At that moment, an current is said to flow from areas that are
electrical potential is created on the cell negatively charged to those that are posi-
surface between the depolarized area (nega- tively charged. When a depolarization cur-
tively charged surface) and the still-polarized rent is directed toward the (+) electrode of the
(positively charged surface) portions of the voltmeter, an upward deflection is recorded.
cell. As a result, an electrical current begins Conversely, if it is directed away from the (+)
flowing between these two regions. electrode, a downward deflection is recorded.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 82

82 Chapter Four

Because the depolarization current in this


example proceeds from left to right—that is,
toward the (+) electrode—an upward deflec-
tion is recorded by the voltmeter. As the A
wave of depolarization spreads along the
cell, additional electrical forces directed to-
ward the (+) electrode record an even greater
upward deflection (see Fig. 4.1C). Once
the cell has become fully depolarized (see
Fig. 4.1D), its outside is completely nega-
tively charged with respect to the inside, the
opposite of the initial resting condition. B
However, because the surface charge is homo-
geneous once again, the external electrodes
measure a potential difference of zero and
the voltmeter records a neutral “flat line”
during this period.
C
Note that in Figure 4.1E, if the electrode
wires of the voltmeter had been reversed so
that the (+) pole was placed to the left of
the cell, then as the wave of depolarization
proceeded toward the right, it would have
D
headed away from the (+) electrode and the
recorded deflection would have been down- Figure 4.2. Sequence of repolarization of a single
ward. This relationship should be kept in cardiac muscle cell. A. As repolarization commences,
positive charges reemerge on the surface of the cell, and
mind when the polarity of ECG leads is de- a current flows from the still-negatively charged surface
scribed in the next section. areas to the repolarized region. Because the current is di-
Depolarization of the cell initiates cardiac rected away from the (+) electrode of the voltmeter, a
downward deflection is recorded. B. Repolarization pro-
muscle contraction and is then followed by gresses. C. Repolarization has been completed, and the
repolarization, the process by which the outside surface of the cell is once again homogeneously
cellular charges return to the resting state charged, so that no further electrical potential is detected
Fig. 2 (Fig. 4.2). As the left side of the cell begins to (flat line once again). D. In the human heart, repolariza-
tion proceeds in a direction opposite that of depolarization
repolarize in this example, its surface charge (would be from right to left in this example, and the wave
becomes positive once again. A current is of repolarization would be upright). Therefore, the de-
therefore generated from the still negatively flections of depolarization and repolarization of the nor-
mal intact heart are in the same direction, as shown here.
charged surface toward the positively charged Notice that the wave of repolarization is of lower ampli-
area. Because this current is directed away tude and more prolonged than that of depolarization.
from the voltmeter’s (+) electrode, a down-
ward deflection is recorded, opposite to that
which was observed during the process of
depolarization. that of depolarization. This occurs because
Repolarization is a slower process than de- myocardial action potential durations are
polarization, so that the inscribed deflection more prolonged in cells near the inner en-
of repolarization is wider and of lower mag- docardium (the first cells stimulated by
nitude. Once the cell has returned to the Purkinje fibers) than in myocytes near the
resting state, the surface charges are once outer epicardium (the last cells to depolar-
again homogeneous and no further electrical ize). Thus, the endocardium is the first re-
potential is detected, resulting in a flat line gion to depolarize but the last region to re-
on the voltmeter recording (see Fig. 4.2C). polarize. As a result, the recorded pattern of
It is important to note that in the intact repolarization on an ECG is usually the in-
human heart, the sequence of repolarization verse of what was presented in this example;
actually proceeds in the direction opposite that is, the current of repolarization (nega-
10090-04_CH04.qxd 8/31/06 5:25 PM Page 83

The Electrocardiogram 83

tive-to-positive flow) in Figure 4.2 is nor-


mally directed toward the (+) electrode and TABLE 4.1. Proper Positions of ECG
Chest Electrodes
inscribes an upright deflection on the record-
ing. Therefore, in a normal heart, the forces V1 4th ICS, 2 cm to the right of sternum
of depolarization and repolarization are usu- V2 4th ICS, 2 cm to the left of sternum
ally oriented in the same direction on the V3 Midway between V2 and V4
V4 5th ICS, left midclavicular line
ECG recording (see Fig. 4.2D).
V5 5th ICS, left anterior axillary line
The depolarization and repolarization of V6 5th ICS, left midaxillary line
a single cardiac muscle cell have been con-
sidered here. As the wave of depolarization ICS, intercostal space

spreads rapidly through the heart, each cell


generates electrical forces, and it is the sum
of these forces, measured at the skin’s sur- A complete ECG tracing is produced by
face, recorded by the ECG machine. The di- recording the electrical activity between
rection and magnitude of the deflections on these standard electrodes in specific patterns.
the ECG recording depend on how the elec- Figure 4.4 demonstrates the orientation of Fig. 4
trical forces are aligned to a set of specific the six standard reference axes (termed limb
reference axes, known as ECG leads. leads) in the body’s frontal plane, which are
electronically constructed as described in the
following paragraphs.
ELECTROCARDIOGRAM LEAD
The ECG machine records lead aVR by
REFERENCE SYSTEM
selecting the right-arm electrode as the (+)
When the electrocardiogram was first in- pole with respect to the other electrodes.
vented, the recording was made by dunking This is known as a unipolar lead, because
the patient’s arms and legs into large buck- there is no single (−) pole; rather, all the other
ets filled with electrolyte solution and wired electrodes are averaged to create a composite
to the machine. That process naturally was (−) reference. When the instantaneous elec-
fairly messy and fortunately is no longer trical activity of the heart points in the di-
necessary. Instead, wire electrodes are placed rection of the right arm, an upward deflec-
directly on the skin, held in place by adhe- tion is recorded in lead aVR. However, when
sive tabs, in the standard arrangement shown the electrical forces are heading away from
Fig. 3 in Figure 4.3. The right-leg electrode is not the right arm, the ECG inscribes a down-
used for measurement but serves as an elec- ward deflection in aVR.
Tab. 1 trical ground. Table 4.1 lists the standard lo- Similarly, lead aVF is recorded by setting
cations of the chest electrodes. the left leg as the (+) pole, such that a positive

A B

Right arm Left arm


electrode electrode
Chest V1 V6
electrodes
V2 V5
V3 V4
Right leg Left leg
electrode electrode

Figure 4.3. Placement of electrocardiogram (ECG) electrodes. A. Standard


positions. B. Close-up view of chest electrode placement.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 84

84 Chapter Four

Unipolar Limb Leads

(+)

aVR aVL
aVF

(+)

Bipolar Limb Leads

(–) (+) (–) (–)


I

II III

(+) (+)

Figure 4.4. The six limb leads are formed from the electrodes placed on the arms and left
leg. Each unipolar lead has a (+) designated electrode; for the unipolar leads, the (−) pole is an
average of the other electrodes. Each bipolar lead has specific (−) and (+) designated electrodes.

deflection is recorded when forces are di- By overlaying the six limb leads together,
rected toward the feet. Lead aVL is selected a reference system is established (Fig. 4.5). In Fig. 5
when the left-arm electrode is made the (+) the figure, each lead is presented with its (+)
pole and records an upward deflection when pole designated by an arrowhead and the (−)
electrical activity is aimed in that direction. aspect by dashed lines. Note that each 30°
In addition to these three unipolar limb sector of the circle falls along the (+) or (−)
leads, three bipolar leads are part of the stan- pole of one of the standard six ECG leads.
dard ECG recording (see Fig. 4.4). Bipolar in- Also note that the (+) pole of lead I points to
dicates that one limb electrode is the (+) pole
and another single electrode provides the (−)
reference. In this case, the ECG machine in-
scribes an upward deflection if electrical TABLE 4.2. Limb Leads
forces are heading toward the (+) electrode Lead () Electrode () Electrode
and records a downward deflection if the
forces are heading toward the (−) electrode. A Bipolar leads
I LA RA
simple mnemonic to remember the place-
II LL RA
ment of the bipolar leads is that the lead III LL LA
name indicates the number of l’s in the place- Unipolar leads
ment sites. For example, lead III connects the aVR RA *
left arm to the left leg, lead II connects the aVL LA *
aVF LL *
right arm to the left leg, and lead I connects
Tab. 2 the left arm to the right arm. Table 4.2 lists *(−) Electrode constructed by combining all other electrodes.
how the six limb leads are derived. LA, left arm; LL, left leg; RA, right arm.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 85

The Electrocardiogram 85

0° and that, by convention, measurement of


the angles proceeds clockwise as +30°, +60°,
and so forth. The complete ECG recording
provides a simultaneous “snapshot” of the
heart’s electrical activity, taken from the per-
spective of each of these lead reference lines.
Figure 4.6 demonstrates how the magni- Fig. 6
tude and direction of electrical activity are
represented by the ECG recording in each
lead. This figure should be studied until the
following four points are clear:
1. An electrical force directed toward the (+)
pole of a lead results in an upward deflec-
tion on the ECG recording of that lead.
2. Forces that head away from the (+) elec-
trode result in a downward deflection in
Figure 4.5. The axial reference system is created by that lead.
combining the leads shown in Figure 4.4. Each lead
has a (+) region indicated by the arrowhead and a (−) re- 3. The magnitude of the deflection, either
gion indicated by the dashed line. upward or downward, reflects how paral-

Figure 4.6. Relationship of the magnitude and direction of electrical activity to the ECG lead.
A. The electrical vector is oriented parallel to lead I and aimed toward the (+) electrode; therefore, a tall
upward deflection is recorded by the lead. B. The vector is still oriented toward the (+) region of lead I
but not parallel to the lead, so that only a component of the force is recorded. The recorded deflection
is still upward but less tall compared with that shown in A. C. The electrical vector is perpendicular to
lead I. so that no deflection is generated. D. The vector is directed toward the (−) region of lead I, caus-
ing the ECG to record a downward deflection.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 86

86 Chapter Four

lel the electrical force is to the axis of the the junction of the right atrium and the su-
lead being examined. The more parallel perior vena cava (Fig. 4.8). The wave of de- Fig. 8
the electrical force is to the lead, the polarization rapidly spreads through the
greater the magnitude of the deflection. right and left atria and then reaches the atri-
4. An electrical force directed perpendicular oventricular (AV) node, where it encounters
to an electrocardiographic lead does not an expected delay. The impulse then travels
register any activity by that lead (a flat rapidly through the bundle of His and into
line on the recording). the right and left bundle branches. These di-
vide into the Purkinje fibers, which radiate
The six standard limb leads examine the elec-
toward the myocardial fibers, stimulating
trical forces in the frontal plane of the body.
them to contract.
However, because electrical activity travels in Each heartbeat is represented on the
three dimensions, recordings from a perpen- ECG by three major deflections that record
Fig. 7 dicular plane are also essential (Fig 4.7A).
the sequence of electrical propagation (see
This is accomplished by the use of six elec- Fig. 4.8B). The P wave represents depolar-
trodes placed on the anterior and left lateral ization of the atria. Following the P wave,
aspect of the chest (see Fig. 4.3B), creating the the tracing returns to the flat baseline as a
chest (or precordial) leads. The orientation result of the conduction delay at the AV
of these leads around the heart is shown in node. The second deflection of the ECG, the
Figure 4.7B. These are unipolar leads and, as QRS complex, represents depolarization of
with the unipolar limb leads, electrical forces the ventricular muscle cells. After the QRS
directed toward these individual (+) electrodes complex, the tracing returns to baseline
result in an upward deflection on the record- once again, and after a brief delay, repolar-
ing of that lead and forces heading away ization of the ventricular cells is signaled by
record a downward deflection. the T wave. Occasionally, an additional
The standard complete electrocardiogram small deflection follows the T wave (the U
prints samples from each of the six limb leads wave), which is believed to represent late
and each of the six chest leads, examples of phases of ventricular repolarization.
which are presented later in the chapter. The QRS complex may take one of several
shapes but can always be subdivided into in-
dividual components (Fig. 4.9). If the first Fig. 9
SEQUENCE OF NORMAL
deflection of a QRS complex is downward, it
CARDIAC ACTIVATION
is known as a Q wave. However, if the initial
Conduction of electrical impulses through deflection is upward, then that particular
the heart is an orderly process. The normal complex does not have a Q wave. The R wave
beat begins at the sinoatrial node, located at is defined as the first upward deflection,

Figure 4.7. The chest (precordial) leads. A. The cross-sectional plane of


the chest. B. Arrangement of the six chest electrodes shown in the cross-
sectional plane.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 87

The Electrocardiogram 87

B
Figure 4.8. Cardiac conduction pathway. A. The electrical impulse be-
gins at the sinoatrial (SA) node (1) then traverses the atria (2). After a
delay at the AV node (3), conduction continues through the bundle of
His and into the right and left bundle branches (4). The latter divide into
Purkinje fibers, which stimulate contraction of the myocardial cells.
B. Corresponding waveforms on the ECG recording: (1) the SA node dis-
charges (too small to generate any deflection on ECG), (2) P wave in-
scribed by depolarization of the atria, (3) delay at the AV node, and
(4) depolarization of the ventricles (QRS complex). The T wave represents
ventricular repolarization.

Figure 4.9. Examples of QRS complexes. A. The first deflection is down-


ward (Q wave), followed by an upward deflection (R wave), and then another
downward wave (S wave). B. Because the first deflection is upward, this com-
plex does not have a Q wave; rather, the downward deflection after the
R wave is an S wave. C. A QRS complex without downward deflections lacks
Q and S waves. D. QRS composed of only a downward deflection; this is a
Q wave but is often referred to as a QS complex. E. A second upward de-
flection (seen in bundle branch blocks) is labeled R′.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 88

88 Chapter Four

whether or not a Q wave is present. Any no further net electrical force is generated,
downward deflection following the R wave is and the ECG voltage recording returns to
known as an S wave. Figure 4.9 demonstrates baseline in both leads. Thus, in this example
several variations of the QRS complex. In cer- of depolarization in a normal heart, lead aVL
tain pathologic states, such as bundle branch inscribes an initial small Q wave followed by
blocks, additional deflections may be in- a tall R wave. Conversely, in lead aVF, there
scribed, as shown in the figure. Study Figure is an initial upward deflection (R wave) fol-
4.9 to gain confidence in differentiating a Q lowed by a downward S wave.
from an S wave. The sequence of depolarization in the
Fig. 10 Figure 4.10 illustrates the course of nor- horizontal plane of the body is also evident
mal ventricular depolarization as it is on examining the six chest leads (Fig. 4.11). Fig. 11
recorded by two of the ECG leads, aVF and Once again, recall that the first region to de-
aVL. The recording in aVF represents electri- polarize is the left ventricular aspect of the
cal activity from the perspective of the infe- interventricular septum. The sequence of
rior (i.e., underside) aspect of the heart, and depolarization proceeds from the midven-
aVL records from the perspective of the left tricular septum toward the right ventricle
lateral side. Recall that in the resting state, (which is anterior to the left ventricle), to-
the surfaces of myocardial cells are posi- ward the cardiac apex, and then around to
tively charged compared with the inside, the lateral walls of both ventricles. Because
but these external ECG leads record zero the initial forces are directed anteriorly—
voltage, as the sum of electrical forces in that is, toward the (+) pole of V1—the initial
each region are canceled by equal and op- deflection recorded by lead V1 is upward.
posite forces. These same initial forces are heading away
The initial portion of ventricular myo- from V6 (which overlies the lateral wall of
cardium that is stimulated to depolarize is the left ventricle), so an initial downward
the midportion of the interventricular sep- deflection is recorded there. As the wave of
tum, on the left side. Because depolariza- depolarization spreads, the forces of the left
tion reverses the cellular charge, the surface ventricle outweigh those of the right, and
of that region becomes negative with re- the vector swings posteriorly toward the
spect to the inside, and an electrical current bulk of the left ventricular muscle. As the
is generated (see Fig. 4.10B, arrow). This forces swing away from lead V1, the deflec-
initial force heads away from the left ven- tion there becomes downward, whereas it
tricle, toward the right ventricle and inferi- becomes more upright in lead V6. Leads V2
orly. Because the force is heading away from through V5 record intermediate steps in this
the (+) pole region of lead aVL, an initial process, such that the R wave becomes pro-
downward deflection is recorded in that gressively taller from lead V1 through lead
lead. At the same time, forces are heading V6 (see Fig. 4.11E). Typically, the height of
in the direction of the (+) pole region of the R wave becomes greater than the depth
lead aVF, causing an initial upward deflec- of the S wave in lead V3 or V4; the lead in
tion to be recorded there. As the wave of which this occurs is termed the “transition”
depolarization spreads through the myo- lead.
cardium, the sequence of net electrical charge A normal complete 12-lead ECG is pre-
occurs as depicted by the series of arrows in sented later in the chapter (see Fig. 4.28).
Figure 4.10. The ECG is recorded on a special grid
As the lateral walls of the ventricles are de- divided into lines spaced 1 mm apart in
polarized, the forces of the thicker left side both the horizontal and vertical directions.
outweigh those of the right. Therefore, the Each fifth line is made heavier to facilitate
arrow swings further and further toward the measurement. On the vertical axis, voltage
left ventricle (leftward and posteriorly). At is measured in millivolts (mV), and in the
the completion of depolarization, the myo- standard case, each 1-mm line separation
cytes again become homogeneously charged, represents 0.1 mV. The horizontal axis rep-
10090-04_CH04.qxd 8/31/06 5:25 PM Page 89

The Electrocardiogram 89

Figure 4.10. Normal ventricular depolarization as recorded by leads aVL and aVF. A. In the resting state, the
surface is homogeneously charged so that the leads do not record any electrical potential. B. The first area to depo-
larize is the left side of the ventricular septum. This results in forces heading away from aVL (downward deflection on
aVL recording) but toward the (+) region of aVF, such that an upward deflection is recorded by that lead. C and D.
Depolarization continues; the forces from the thicker-walled left ventricle outweigh those of the right, such that the
electrical vector swings leftward and posteriorly toward aVL (upward deflection) and away from aVF. E. At the com-
pletion of depolarization, the surface is again homogeneously charged, and no further electrical voltage is recorded.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 90

90 Chapter Four

Figure 4.11. Sequence of depolarization recorded by the chest (precordial) leads. A–D. Depolar-
ization begins at the left side of the septum, and then the forces progress posteriorly toward the left ven-
tricle. Thus, V1, which is an anterior lead, records an initial upward deflection followed by a downward
wave, whereas V6, a posterior lead, inscribes the opposite. E. In the normal pattern of the QRS from V1 to
V6, the R wave becomes progressively taller and the S wave less deep.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 91

The Electrocardiogram 91

resents time. Because the standard recording 6. Abnormalities of the P wave


speed is 25 mm/sec, each 1 mm division rep- 7. Abnormalities of the QRS (hypertrophy,
resents 0.04 sec and each heavy line (5 mm) bundle branch block, infarction)
Fig. 12 represents 0.2 sec (Fig. 4.12). 8. ST segment and T wave abnormalities

INTERPRETATION OF THE Calibration


ELECTROCARDIOGRAM
ECG machines routinely inscribe a 1.0-mV
Many cardiac disorders alter the ECG re- vertical signal at the beginning or end of
cording in a diagnostically useful way. It is each 12-lead tracing to document the volt-
important to interpret each tracing in a age calibration of the machine. In the nor-
standard fashion to avoid missing subtle ab- mal case, each 1-mm vertical box on the
normalities. Here is a commonly followed ECG paper represents 0.1 mV, so that the cal-
sequence of analysis: ibration signal records a 10-mm deflection
1. Check voltage calibration (e.g., see Fig. 4.28). However, in patients with
markedly increased voltage of the QRS com-
2. Heart rhythm
plex (e.g., some patients with left ventricular
3. Heart rate hypertrophy or bundle branch blocks), the
4. Intervals (PR, QRS, QT) very large deflections would not fit on the
5. Mean QRS axis ECG tracing. To facilitate interpretation in

Paper Speed: 25 mm/sec

PR QT

5 mm = 0.5 mV
(1 mm = 0.1 mV)

QRS

5 mm = 0.2 sec
(1 mm = 1 small box = 0.04 sec)
Figure 4.12. Enlarged view of an ECG strip. The paper travels through the machine at
25 mm/sec, so that each 1 mm on the horizontal axis represents 0.04 sec. Each 1 mm on the verti-
cal axis represents 0.1 mV. Interval measurements in this example are as follows: PR interval (from
the beginning of the P wave to the beginning of the QRS) = small boxes = 0.16 sec; QRS duration (from
the beginning to the end of the QRS complex) = 1.75 small boxes = 0.07 sec; and QT interval (from the
beginning of the QRS to the end of the T wave) = 8 small boxes = 0.32 sec. The corrected QT
QT
interval = . Because the R–R interval = 15 small boxes (0.6 sec), the corrected QT interval =
R −R
0.32
= 0.41 sec.
0.6
10090-04_CH04.qxd 8/31/06 5:25 PM Page 92

92 Chapter Four

such a case, the recording is purposely made which corresponds to the heart rate in beats
at half the standard voltage (i.e., each 1-mm per minute, as illustrated in Figure 4.13. Fig. 13
box = 0.2 mV), and this is indicated on the When the rhythm is irregular, the heart
ECG tracing by a change in the height of the rate may be approximated by taking advan-
1.0-mV calibration signal (at half the stan- tage of the time markers, spaced 3 seconds
dard voltage, the signal would be 5 mm tall). apart, often printed at the top or bottom of
It is important to check the height of the cal- the ECG tracing (see Fig. 4.13, method 3).
ibration signal on each ECG to ensure that
the voltage criteria used to define specific ab-
Intervals (PR, QRS, QT)
normalities is applicable.
The PR interval, QRS interval, and QT inter-
val (see Fig. 4.12) are measured in the limb
Heart Rhythm
lead recordings. For each of these, it is ap-
The normal cardiac rhythm, initiated by de- propriate to take the measurement in the
polarization of the sinus node, is known as limb lead in which the interval is the longest
sinus rhythm and is present if (1) every in duration (the intervals can vary a bit in
P wave is followed by a QRS; (2) every each lead). The PR interval is measured
QRS is preceded by a P wave; (3) the P wave is from the onset of the P wave to the onset of
upright in leads I, II, and III; and (4) the PR the QRS. The QRS interval is measured from
interval is greater than 0.12 sec (three small the beginning to the end of the QRS com-
boxes). If the heart rate in sinus rhythm is be- plex. The QT interval is measured from the
tween 60 and 100 bpm, then normal sinus beginning of the QRS to the end of the
rhythm is present. If less than 60 bpm, the T wave. The normal ranges of the intervals
rhythm is sinus bradycardia; if greater than are listed in Table 4.3, along with conditions Tab. 3
100 bpm, the rhythm is sinus tachycardia. associated with abnormal values.
Other abnormal rhythms (termed arrhyth- Because the QT interval varies with heart
mias or dysrhythmias) are described in Chap- rate (the faster the heart rate, the shorter the
ters 11 and 12. QT), the corrected QT interval is determined
by dividing the measured QT by the square
root of the R–R interval (see Fig. 4.12). When
Heart Rate
the heart rate is in the normal range (60 to
The standard ECG paper speed is 25 mm/sec. 100 bpm), a rapid rule can be applied: if the
Therefore, QT interval is less than half the interval be-
tween two consecutive QRS complexes, then
25 mm/sec × 60 sec/min
Heart rate (bpm) = the QT interval is within the normal range.
Number of mm
between beats
Mean QRS Axis
or more simply,
The mean QRS axis represents the average of
1,500
Heart rate (bpm) = the instantaneous electrical forces generated
Number of small during the sequence of ventricular depolar-
boxes between ization. The normal value is between −30°
2 consecutive beats and +90° (Fig. 4.14). A mean axis that is more Fig. 14
It is rarely necessary, however, to determine negative than −30° implies left axis devia-
the exact heart rate, and a more rapid deter- tion, whereas an axis greater than +90° rep-
mination can be made with just a bit of resents right axis deviation. The axis can be
memorization. Simply “count off” the num- accurately determined by plotting the QRS
ber of large boxes between two consecutive complexes of different leads on the axial ref-
QRS complexes, using the sequence erence diagram (see Fig. 4.5), but this is te-
dious and rarely necessary. It is generally
300—150—100—75—60—50 sufficient to note whether the axis is nor-
10090-04_CH04.qxd 8/31/06 5:25 PM Page 93

The Electrocardiogram 93

Method 1
First, count the number of small boxes (1 mm each) between two adjacent
QRS complexes (i.e., between 2 “beats”). Then, since the standard paper
speed is 25 mm/sec:
Heart Rate (25 mm/sec  60 sec/min) 1,500
(beats/min)  Number of mm between beats  number of mm between beats
In this example, there are 23 mm between the first 2 beats.
23 mm between beats

Therefore, the heart rate  1,500  65 bpm


23

Method 1 is particularly helpful for measuring fast heart rates (100 bpm)

Method 2
The “count-off” method requires memorizing the sequence:
300 - 150 - 100 - 75 - 60 - 50
Then use this sequence to count the number of large boxes between two
consecutive beats:
300 100 60
Start 150 75 50
here

The second QRS falls between the 75 and 60 bpm; therefore, the
heart rate is approximately midway between them  67 bpm. Knowing
that the heart rate is approximately 60–70 bpm is certainly close enough.
Figure 4.13. Methods to calculate heart rate.

mal, deviated to the left, or deviated to the flection greater than downward deflection),
right. If a more precise measurement is then the mean vector falls within the normal
needed, the simplified approach described range (Fig. 4.15).a If the QRS in either lead I or Ftn. a,
next can be employed. II is not primarily upward, then the axis is Fig. 15
Recall from Figure 4.5 that each ECG lead abnormal, and the approximate axis should
has a (+) region and a (−) region. Electrical then be determined by the rapid method de-
activity directed toward the (+) half results scribed in the following paragraphs.
in an upward deflection, whereas activity to-
ward the (−) half results in a downward de-
a
flection on the ECG recording of that lead. Note that some textbooks recommend (and some prac-
titioners prefer) examining leads I and aVF, rather than
To determine whether the axis is normal
leads I and II, to determine if the mean axis is normal.
or abnormal, examine the QRS complexes in This is acceptable, but be aware that a mean axis that
limb leads I and II. If the QRS is primarily falls between 0° and −30° would be erroneously classi-
positive in both of these leads (upward de- fied as “abnormal” by that method.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 94

94 Chapter Four

Method 3

ECG recording paper often indicates 3-sec time markers at the top or
bottom of the tracing:
marker marker
3 sec

To calculate the heart rate, count the number of QRS complexes between
the 3-sec markers ( 6 beats in this example) and multiply by 20. Thus,
the heart rate here is approximately 120 bpm.

It’s even easier (and more accurate) to count the number of complexes
between the first and third markers on the strip (representing 6 sec
of the recording) and then multiply by 10 to determine the heart rate.

Method 3 is particularly helpful for measuring irregular heart rates.


Figure 4.13. (Continued) Methods to calculate heart rate.

First, consider the special example in Fig- After arrow c, the electrical vector swings
Fig. 16 ure 4.16. A sequence of ventricular depolar- into the positive region of lead I, so that up-
ization is represented in this figure by arrows ward deflections are recorded.
a through e. The initial deflection (repre- In this special example, in which electri-
senting left septal depolarization) points to cal forces begin exactly opposite lead I’s (+)
the patient’s right side. Because it is directed electrode and terminate when pointed di-
completely away from the (+) pole of lead I, rectly at that electrode, note that the mean
a strong downward deflection is recorded by electrical vector points straight downward
the lead. As depolarization continues, the (in the direction of arrow c), perpendicular to
arrow swings downward and to the left, re- the lead I axis. Also notice the configuration
sulting in less negative deflections in lead I. of the inscribed QRS complex in lead I.

TABLE 4.3. Electrocardiographic Intervals

Interval Normal Decreased In Increased In

PR 0.12–0.20 sec • Preexcitation syndrome • First-degree AV block


(3–5 small boxes) • Junctional rhythm
QRS ≤0.10 sec • Bundle branch blocks
(≤2.5 small boxes) • Ventricular ectopic beat
• Toxic drug effect (e.g., quinidine)
• Severe hyperkalemia
QT Corrected QTa ≤0.44 sec • Hypercalcemia • Hypocalcemia
• Tachycardia • Hypokalemia (↑ QT interval owing to AQ1,
↑ T wave) AQ2
• Hypomagnesemia
• Myocardial ischemia
• Congenital prolongation of QT
• Toxic drug effect (e.g., quinidine)
a
Corrected QT is negative. AQ3
10090-04_CH04.qxd 8/31/06 5:25 PM Page 95

The Electrocardiogram 95

Left a
xis
d ev
iat
i

on
90°
120° 60° Left axis deviation
150° 30° • Inferior wall myocardial infarction
aVR aVL • Left anterior fascicular block
• Left ventricular hypertrophy
(sometimes)

180° I 0°

Right axis deviation


• Right ventricular hypertrophy
150° 30° • Acute right heart strain
(e.g., massive pulmonary embolism)
III • Left posterior fascicular block
II
Ri 120° aVF 60°
gh 90°
ta
xis
dev
iatio n

Figure 4.14. A normal mean QRS axis falls within the shaded area (between 30º and
+90°°). A mean axis more negative than −30° is termed left axis deviation, whereas an axis more
positive than +90° is right axis deviation. The figure shows common conditions that result in axis
deviation.

There is a downward deflection, followed the (+) pole of that lead. If it is predomi-
by an upward deflection of equal magni- nantly negative, then it points away from
tude (when the upward and downward de- the lead’s (+) pole. In the example, the iso-
flections of a QRS are of equal magnitude, electric complex appears in lead I; therefore,
it is termed an isoelectric complex). Thus, the next step is to inspect the perpendicular
when an ECG limb lead inscribes an isoelectric lead, which is aVF (see Fig. 4.5 if this rela-
QRS complex, it indicates that the mean elec- tionship is not clear). Because the QRS com-
trical axis of the ventricles is perpendicular to plex in aVF is primarily upward, the mean
that lead. axis points toward its (+) pole, which is in
Therefore, an easy way to determine the fact located at +90°.
mean QRS axis is to glance at the six limb To summarize, the mean QRS axis is cal-
lead recordings and observe which one has culated as follows:
the most isoelectric-appearing complex: the
1. Inspect limb leads I and II. If the QRS is
mean axis is simply perpendicular to it. One
primarily upward in both, then the axis
step remains. When the mean axis is per-
is normal and you are done. If not, then
pendicular to a lead, it could be perpendicu-
proceed to the next step.
lar in either a clockwise or a counterclock-
wise direction. In the example of Figure 2. Inspect the six limb leads and determine
4.16, the isoelectric complex appears in lead which one contains the QRS that is most
I, such that the mean vector could be at +90° isoelectric. The mean axis is perpendicu-
or it could be at −90°, because both are per- lar to that lead.
pendicular. Determining which of these it is 3. Inspect the lead that is perpendicular to
requires inspecting the recording of the ECG the lead containing the isoelectric com-
lead that is perpendicular to the one inscrib- plex. If the QRS in that perpendicular
ing the isoelectric complex (and is therefore lead is primarily upward, then the mean
parallel to the mean axis). If the QRS is pre- axis points to the (+) pole of that lead. If
dominantly upright in that perpendicular primarily negative, then the mean QRS
lead, then the mean vector points toward points to the (−) pole of that lead.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 96

96 Chapter Four

If the QRS complex is mainly upward in limb lead I, Similarly, if the QRS is predominantly upward in limb
then the mean axis falls within the “+” region of that lead II, then the mean axis falls within the “+” half of
lead, shown as the shaded half of the circle below. lead II, shown as the shaded half here:

–90°

–30°

+150°

+90° +60°

If the QRS is predominantly upright in both


leads I and II, then the mean axis must fall
within their common “+” regions: between
–30° and +90°.

–30°

Normal
Axis

+90°

Figure 4.15. The mean axis is within the normal range if the QRS complex is predominantly upright in limb
leads I and II.

Conditions that result in left or right axis or back from the chest, as it may be in pa-
deviation are listed in Figure 4.14. In addi- tients with chronic obstructive lung disease;
tion, the vertical position of the heart in in such a case, the mean axis is said to be
many normal children and adolescents may indeterminate.
result in a slightly rightward mean axis
(>+90°).
Abnormalities of the P Wave
In some patients, isoelectric complexes
are inscribed in all the limb leads. That situ- The P wave represents depolarization of the
ation arises when the heart is tilted, so that right atrium followed quickly by depolariza-
the mean QRS is pointing straight forward tion of the left atrium; the two components
10090-04_CH04.qxd 8/31/06 5:25 PM Page 97

The Electrocardiogram 97

e component of the P wave is larger than nor-


d mal (taller than 2.5 mm in lead II).
a e Lead I
c
Left atrial enlargement is best observed in
b d b lead V1. Normally, V1 inscribes a P wave with
c a
an initial positive deflection reflecting right
atrial depolarization (directed anteriorly),
followed by a negative deflection, owing to
Mean the left atrial forces oriented posteriorly (see
axis Fig. 1.2 for anatomic relationships). Left atrial
enlargement is therefore manifested by a
greater-than-normal negative deflection (at
least 1 mm wide and 1 mm deep) in lead V1
Lead aVF
(see Fig. 4.17).
Figure 4.16. Sequence of ventricular depolarization
when the mean axis is +90°°. Because the mean axis is
perpendicular to limb lead I, an isoelectric QRS complex Abnormalities of the
(height of upward deflection = height of downward de- QRS Complex
flection) is recorded by that lead (see text for details).
Ventricular Hypertrophy
Hypertrophy of the left or right ventricle
are nearly superimposed on one another causes the affected chamber to generate
Fig. 17 (Fig. 4.17). The P wave is usually best visual- greater-than-normal electrical activity. Or-
ized in lead II, the lead that runs most par- dinarily, the forces produced by the thicker-
allel to the flow of electrical current through walled left ventricle are more prominent
the atria from the sinoatrial to the AV node. than those of the right. However, in right
When the right atrium is enlarged, the initial ventricular hypertrophy, the augmented

Figure 4.17. The P wave represents superimposition of right atrial (RA) and left atrial
(LA) depolarization. RA depolarization occurs slightly earlier than LA depolarization. In RA
enlargement, the initial component of the P wave is prominent (>2.5 mm tall) in lead II. In LA
enlargement, there is a large terminal downward deflection in lead V1 (>1 mm wide and
>1 mm deep).
10090-04_CH04.qxd 8/31/06 5:25 PM Page 98

98 Chapter Four

right-sided forces may outweigh those of the leads I and aVL) show taller-than-normal R
left. Therefore, chest leads V1 and V2, which waves. Leads on the other side of the heart (V1
overlie the right ventricle, record greater- and V2) demonstrate the opposite: deeper-
than-normal upward deflections: the R wave than-normal S waves. Many criteria have
becomes taller than the S wave in those been proposed for the diagnosis of left ven-
leads, the opposite of the normal situation tricular hypertrophy by ECG. Three of the
Fig. 18 (Fig. 4.18). In addition, the increased right most helpful criteria are listed in Figure 4.18.
ventricular mass shifts the mean axis of the
heart, resulting in right axis deviation
Bundle Branch Blocks
(greater than +90°).
In left ventricular hypertrophy, greater- Interruption of conduction through the right
than-normal forces are generated by that or left bundle branches may develop from is-
chamber, which simply exaggerates the nor- chemic or degenerative damage. As a result,
mal situation. Leads that directly overlie the the affected ventricle does not depolarize in
left ventricle (chest leads V5 and V6 and limb the normal sequence. Rather than rapid uni-

A V6

RIGHT VENTRICULAR HYPERTROPHY


RV LV

• R > S in lead V1
1
• Right axis deviation
4 2

V1

B
V6

3 LEFT VENTRICULAR HYPERTROPHY

• S in V1 plus
R in V5 or V6 ≥ 35 mm or
1 • R in aVL > 11 mm or
2 • R in Lead I > 15 mm

V1

Figure 4.18. Ventricular hypertrophy. The arrows indicate the sequence of average electrical forces
during ventricular depolarization. A. Right ventricular (RV) hypertrophy. The RV forces outweigh those of
the left, resulting in tall R waves in leads V1 and V2 and a deep S wave in lead V6. B. Left ventricular (LV)
hypertrophy exaggerates the normal pattern of depolarization, with greater-than-normal forces directed
toward the LV, resulting in a tall R wave in V6 and a deep S wave in lead V1.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 99

The Electrocardiogram 99

form stimulation by the Purkinje fibers, the boxes), an incomplete bundle branch block is
cells of that ventricle must rely on relatively present. If the QRS duration is greater than
slow myocyte-to-myocyte spread of electrical 0.12 sec (3.0 small boxes), complete bundle
activity traveling from the unaffected ventri- branch block is identified.
cle. This delayed process prolongs depolar- In right bundle branch block (Fig. 4.19A; Fig. 19
ization and widens the QRS complex. When see also Fig. 4.29), normal depolarization of
a bundle branch block widens the QRS dura- the right ventricle does not occur. In this
tion to 0.10 to 0.12 sec (2.5 to 3.0 small case, initial depolarization of the ventricular

Figure 4.19. Bundle branch blocks. Interruption of conduction through the right
(RBBB) or left (LBBB) bundles results in delayed, slowed activation of the respective
ventricle and widening of the QRS complex. A. In RBBB, normal initial activation of
the septum (1) is followed by depolarization of the left ventricle (2). Slow cell-to-
cell spread activates the right ventricle (RV) after the left ventricle (LV) has nearly
fully depolarized, so that the late forces generated by the RV are unopposed.
Therefore, V1 records an abnormal terminal upward deflection (R′), and V6 records
an abnormal, terminal deep S wave (3). B. In LBBB, the initial septal depolarization
is blocked, such that initial forces are oriented from right to left. Thus, the normal
initial R wave in V1 and Q wave in V6 are absent (1). After the RV depolarizes, late,
slow activation of the LV results in a terminal upward deflection in V6 and down-
ward deflection in V1 (3).
10090-04_CH04.qxd 8/31/06 5:25 PM Page 100

100 Chapter Four

septum (which is stimulated by a branch of posterior fascicular blocks (also termed hemi-
the left bundle) is unaffected so that the blocks). The main significance of fascicular
normal small R wave in lead V1 and small Q blocks in ECG interpretation is that they can
wave in lead V6 are recorded. As the wave of markedly alter the mean ECG axis.
depolarization spreads down the septum Anatomically, the anterior fascicle of the
and into the left ventricular free wall, the se- left bundle runs along the front of the left
quence of depolarization is indistinguish- ventricle toward the anterior papillary mus-
able from normal, because left ventricular cle (which is located in the anterior and
forces normally outweigh those of the right. superior portion of the chamber), whereas
However, by the time the left ventricle has the posterior fascicle travels to the posterior
almost fully depolarized, slow cell-to-cell papillary muscle (which is located in the
spread has finally reached the “blocked” right posterior, inferior, and medial aspect of the
ventricle and depolarization of that cham- left ventricle). Under normal conditions,
ber begins, unopposed by left ventricular conduction via the left anterior and left
activity (because that chamber has nearly posterior fascicles proceeds simultaneously,
fully depolarized). This prolonged depolar- such that electrical activation of the left
ization process widens the QRS complex, ventricle is uniform, spreading from the
and since the terminal portion of the QRS bases of the two papillary muscles. However,
complex represents right ventricular forces if conduction is blocked in one of the two
acting alone, there is a terminal upward de- divisions, then initial LV depolarization
flection (known as R′ wave) over the right arises exclusively from the unaffected fasci-
ventricle in lead V1, and a downward de- cle (Fig. 4.20). Fig. 20
flection (S wave) in V6 on the opposite side of In the case of left anterior fascicular
the heart. block (LAFB), left ventricular activation be-
Left bundle branch block produces even gins via the left posterior fascicle alone, at
greater QRS abnormalities. In this situation, the posterior papillary muscle, and then
normal initial depolarization of the left sep- spreads to the rest of the ventricle. Because
tum does not occur; rather, the right side the left posterior fascicle first activates the
of the ventricular septum is first to depolar- posterior, inferior, medial region of the left
ize, through branches of the right bundle. ventricle, the initial impulses are directed
Thus, the initial forces of depolarization are downward (i.e., toward the feet) and toward
directed toward the left ventricle instead of the patient’s right side (see Fig. 4.20). This
the right (see Fig. 4.19B; see also Fig. 4.30). results in a positive deflection (initial small
Therefore, an initial downward deflection is R wave) in the inferior leads (leads II, III, and
recorded in V1 and the normal small Q wave aVF) and a negative deflection (small Q wave)
in V6 is absent. Only after depolarization in the left lateral leads, I and aVL. As depo-
of the right ventricle does slow cell-to- larization then spreads upward and to the
cell spread reach the left ventricular myo- left, toward the “blocked” anterior, superior,
cytes. These slowly conducted forces inscribe and lateral regions of the left ventricle, a
a widened QRS complex with terminally positive deflection (R wave) is inscribed in
upward deflections in the leads overlying leads I and aVL, while a negative deflection
the left ventricle (V5 and V6), as shown in (S wave) develops in the inferior leads. The
Figure 4.19B. predominance of these leftward forces, re-
Recall from Chapter 1 that the left bundle sulting from the abnormal activation of the
branch subdivides into two main divisions, anterior superior left ventricular wall results
termed fascicles: the left anterior fascicle and in left axis deviation (generally more nega-
left posterior fascicle. Although a left bundle tive than −45 degrees). A complete 12-lead
branch block implies that conduction is ECG demonstrating LAFB is shown later
blocked in the entire left bundle branch, im- (see Fig. 4.34).
pairment can also occur in just one of the Left posterior fascicular block (LPFB) is
two fascicles, resulting in left anterior or left less common than LAFB. In LPFB, ventricu-
10090-04_CH04.qxd 8/31/06 5:25 PM Page 101

MV A

LV
P
L AFB LPFB

aVL aVL

X
1
X

aVF aVF

aVL aVL

aVF aVF

aVL aVL

aVF aVF

• Left axis deviation • Right axis deviation


• Small Q in leads aVL and I • Small R in leads aVL and I
• Small R in inferior leads (II, III, aVF) • Small Q in inferior leads (II, III, aVF)
Figure 4.20. Left anterior and left posterior fascicular blocks. The schematic at the top of the figure shows the left
ventricle (LV) in the frontal plane. The mitral valve (MV) chordae tendineae insert into the anterior (A) and posterior (P)
papillary muscles, which are important landmarks: the anterior fascicle of the left bundle branch courses toward the an-
terior papillary muscle, whereas the posterior fascicle travels to the posterior papillary (the fascicles are not shown). No-
tice that the anterior papillary muscle is actually superior to the posterior papillary muscle. Left side of figure: In left
anterior fascicular block (LAFB), activation begins solely in the region of the posterior papillary muscle (panel 1) because
initial conduction to the anterior papillary muscle is blocked (denoted by the X). As a result, the initial forces of depo-
larization are directed downward towards the feet, producing an initial positive R wave in lead aVF and a negative Q wave
in lead aVL. As the wave of depolarization spreads toward the left side and superiorly, aVF begins to register a negative
deflection and aVL starts to record a positive deflection (panel 2). Panel 3 shows the complete QRS complexes at the end
of depolarization. Right side of figure: In left posterior fascicular block (LPFB; denoted by the X), LV activation begins
solely in the region of the anterior papillary muscle (panel 1). Thus, the initial forces are directed upward and toward the
patient’s left side, producing an initial R wave in aVL and a Q wave in aVF. Panels 2 and 3 show how the spread of de-
polarization travels in the direction opposite that of LAFB.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 102

102 Chapter Four

lar activation begins via the left anterior fas-


cicle at the base of the anterior papillary
muscle (see Fig. 4.20). As that anterosupe-
rior left ventricular region depolarizes, the
initial forces are directed upward and to the
patient’s left (creating a positive R wave in
leads I and aVL and a negative Q wave in
the inferior leads). As the impulse then
spreads downward and to the right toward
the initially blocked region, an S wave is in-
scribed in leads I and aVL, while an R wave
is recorded in leads II, III, and aVF. Because Figure 4.21. Compared with small Q waves gener-
the bulk of these delayed forces head to- ated during normal depolarization, pathologic
Q waves are more prominent with a width ≥1 mm
ward the patient’s right side, right axis devi- (1 small box) and depth >25% of the height of the
ation of the QRS mean axis is expected (see QRS complex.
Fig. 4.36).
LAFB and LPFB do not result in signifi-
cant widening of the QRS (in contrast to appear in some leads. For example, initial
right or left bundle branch blocks) because septal depolarization routinely inscribes a
rapidly conducting Purkinje fibers bridge small Q wave in leads V6 and aVL. Normal
the territories served by the anterior and Q waves are of short duration (≤0.04 sec, or
posterior fascicles. Therefore, although the 1 small box) and of low magnitude (<25% of
sequence of conduction is altered, the total the QRS total height). A pathologic Q wave
time required for depolarization is usually is more prominent (Fig. 4.21), having a width Fig. 21
only slightly prolonged. Also note that al- ≥1 small box in duration and a depth >25%
though left and right bundle branch blocks of the total height of the QRS. The ECG
are most easily recognized by analyzing the leads in which pathologic Q waves appear
patterns of depolarization in the precordial reflect the anatomic site of an infarction
leads, in the case of LAFB and LPFB, it is the (Table 4.4; see Fig. 4.22). Tab. 4,
recordings in the limb leads (as in Fig. 4.20) Pathologic Q waves develop in the leads Fig. 22
that are most helpful. overlying infarcted tissue because necrotic
muscle does not generate electrical forces.
Rather, the ECG electrode over that region
Myocardial Infarction
detects electrical currents from the healthy
The hallmark of transmural myocardial in- tissue on opposite regions of the ventricle,
farction (MI) is the pathologic Q wave. Re- which are directed away from the infarct and
call that it is normal for an initial Q wave to the recording electrode, thus inscribing the

TABLE 4.4. Localization of Myocardial Infarction

Anatomic Site Leads with Abnormal ECG Complexesa Coronary Artery Most Often Responsible

Inferior II, III, aVF RCA


Anteroseptal V1–V2 LAD
Anteroapical V3–V4 LAD (distal)
Anterolateral V5–V6, I, aVL CFX
Posterior V1–V2 (tall R wave, not Q wave) RCA
a
Pathologic Q waves in all of leads V1–V6 implies an “extensive anterior MI” usually associated with a proximal left coronary
artery occlusion.
CFX, left circumflex coronary artery; LAD, left anterior descending coronary artery; RCA, right coronary artery, TSP1;
10090-04_CH04.qxd 8/31/06 5:25 PM Page 103

The Electrocardiogram 103

downward deflection (Fig. 4.23). Q waves are


permanent evidence of a myocardial infarc- Fig. 23
tion; only rarely do they disappear over time.
Notice in Table 4.4 that in the case of a
posterior wall myocardial infarction (see Fig.
4.22), it is not pathologic Q waves that are
evident on the ECG. Because standard elec-
trodes are not placed on the patient’s back
overlying the posterior wall, other leads must
be relied on to indirectly identify the pres-
ence of such an infarction. Chest leads V1
and V2, which are directly opposite the pos-
terior wall, record the inverse of what leads
placed on the back would demonstrate.
Therefore, taller-than-normal R waves in leads
V1 and V2 are the equivalent of a pathologic
Q wave in the diagnosis of a posterior wall
MI. It may be recalled that right ventricular
hypertrophy also produces tall R waves in
leads V1 and V2, but unlike right ventricular
hypertrophy, right axis deviation is not usu-
ally a feature of posterior wall MI.
It is important to note that if a Q wave ap-
pears in only a single ECG lead, it is not di-
agnostic of an infarction. True pathologic
Qs should appear in the groupings listed in
Table 4.4 and Figure 4.22. For example, if a
pathologic Q wave is present in lead III but
not in II or aVF, it likely does not indicate an
infarction. Also, Q waves are disregarded in
lead aVR because electrical forces are normally
directed away from the right arm. Finally, in
the presence of left bundle branch block, Q
waves are usually not helpful in the diagno-
sis of MI because of the markedly abnormal
pattern of depolarization in that condition.
In infarctions in which Q waves develop,
appropriately termed Q-wave infarctions,
the entire thickness of a myocardial segment
is usually involved. Therefore, this type of
MI is often called a transmural infarct. As de-
scribed in Chapter 7, infarctions are not al-
ways transmural but may involve only the
Figure 4.22. Relationship between ECG leads and
cardiac anatomic regions. A. The leads listed in paren- subendocardial layers of the myocardium. In
theses are those that reflect infarction of these regions. this case, pathologic Q waves do not usually
B. Miniaturized schematic drawings of a 12-lead ECG develop, because the remaining viable cells
showing the standard orientation of printed samples
from each lead. The major anatomic groupings are indi- are able to generate some electrical activity;
cated. Note that while the presence of pathologic Q waves such MIs are therefore called non–Q-wave
in leads V1 and V2 are indicative of anteroseptal infarc- infarctions. In either case, certain ST seg-
tion, tall initial R waves in those leads are seen in poste-
rior wall infarction. ment and T wave abnormalities evolve dur-
ing acute Q-wave and non–Q-wave infarc-
10090-04_CH04.qxd 8/31/06 5:25 PM Page 104

104 Chapter Four

A
Anterior Anterolateral
(V1–V6) (V5–V6, I, aVL)

Anteroseptal Posterior
(V1–V2) (Tall R in
V1–V2)

Inferior
Anteroapical (II, III, aVF)
(V3–V4)

B Anteroseptal Anteroapical Anterolateral

I aVR V1 V4 I aVR V1 V4

II aVL V2 V5 II aVL V2 V5

III aVF V3 V6 III aVF V3 V6

Inferior
Figure 4.23. Sequence of depolarization recorded by lead aVL, overlying a lateral wall infarction
(black region). A pathologic Q wave is recorded because the necrotic muscle does not generate electrical
forces; rather, at the time when the lateral wall should be depolarizing (panel 3), the activation of the healthy
muscle on the opposite side of the heart is unopposed, such that forces head away from aVL. The terminal
R wave recorded by aVL reflects depolarization of the remaining viable myocardium beyond the infarct.

tions, as discussed in the next section. The polarization is very sensitive to myocardial
electrocardiographic differences between perfusion, patients with coronary artery dis-
these types of MI are summarized as follows: ease often demonstrate reversible deviations
of the ST segments and T waves during myo-
cardial ischemia.
Acute ST As described in the previous section, pa-
Type of Pathologic Segment thologic Q waves are indicative of an MI but
Infarction Q Waves Deviation
do not differentiate between an acute event
Q-wave MI Yes ST elevation and an MI that occurred weeks or years ear-
Non–Q-wave No ST depression lier. However, acute MI does result in a se-
MI (and/or T wave quence of ST and T wave abnormalities that
inversion) permit this distinction (Fig. 4.24). The initial Fig. 24
abnormality during an acute Q-wave MI is
elevation of the ST segment, often with a
ST Segment and T Wave peaked appearance of the T wave. At this
Abnormalities early stage, myocardial cells are still viable
Among the most common important ab- and Q waves have not yet developed. Within
normalities of the ST segments and T waves several hours, however, myocyte death leads
are those that represent myocardial ische- to loss of the amplitude of the R wave, and
mia and infarction. Because ventricular re- pathologic Q waves begin to be inscribed by
10090-04_CH04.qxd 8/31/06 5:25 PM Page 105

The Electrocardiogram 105

Figure 4.24. ECG evolution during acute Q-wave myocardial infarction (also termed acute ST-elevation MI).

the ECG leads positioned over the infarct ter- charged compared with the normally fully
ritory. During the first 1 to 2 days following repolarized areas, an electrical current is gen-
infarction, the ST segments remain elevated, erated between the two regions. This current
the T wave inverts, and the Q wave deepens. is directed away from the more negatively
Several days later, the ST segment elevation charged ischemic area, causing the baseline
returns to baseline, but the T waves remain of the ECG leads overlying that region to shift
inverted. Weeks or months following the in- downward. Because the ECG machine Record-
farct, the ST segment and T waves have often ing only relative position, rather than ab-
returned to normal, but the pathologic Q solute voltages, ECG machines do not make
waves persist, a permanent marker of the MI. the downward deviation of the baseline no-
If the ST segment remains elevated several ticeable. Following ventricular depolariza-
tion (indicated by the QRS complex), after all
weeks later, it is likely that a bulging fibrotic
the myocardial cells have fully depolarized
scar (ventricular aneurysm) has developed at
(including those of the injured zone), the net
the site of infarction.
electrical potential surrounding the heart is
These evolutionary changes of the QRS,
true zero. However, compared with the ab-
ST, and T waves are recorded by the leads
normally displaced downward baseline, there
overlying the zone of infarction (see Table is the appearance of ST segment elevation (see
4.4). Typically, reciprocal changes are seen Fig. 4.25). As the myocytes then repolarize,
in leads opposite that site. For example, in the injured cells return to the abnormal state
acute anteroseptal MI, ST segment elevation of diastolic potassium ion leak, and the ECG
is expected in chest leads V1 and V2; simulta- again inscribes the abnormally depressed
neously, however, reciprocal changes (ST de- baseline. Thus, ST elevation in acute MI may
pression) may be inscribed by the leads over- in part reflect an abnormal shift of the record-
lying the opposite (inferior) region, namely ing baseline.
in leads II, III, and aVF. In non–Q-wave myocardial infarctions, it
The mechanism by which ST segment de- is ST segment depression, rather than eleva-
viations develop during acute MI has not tion, that often develops in the leads over-
been established with certainty. It is believed, lying the infarct (see Chapter 7). In this situ-
however, that the abnormality results from ation, the diastolic potassium leak of injured
injured myocardial cells immediately adja- cells adjacent to the infarct generates elec-
cent to the infarct zone producing abnormal trical forces heading from the inner endo-
systolic or diastolic currents. One explana- cardium to the outer epicardium and there-
tion, the diastolic current theory, contends that fore toward the overlying ECG electrode.
these cells are capable of depolarization but Thus, the baseline of the ECG is shifted up-
are abnormally “leaky” for potassium ions, ward (see Fig. 4.25). Following full cardiac de-
preventing them from ever fully repolarizing polarization, the electrical potential of the
Fig. 25 (Fig. 4.25). Because the surface of such par- heart returns to true zero but, relative to the
tially depolarized cells in the resting (dias- abnormal baseline, gives the appearance of
tolic) state would be relatively negatively ST segment depression.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 106

106 Chapter Four

Transmural MI

Normal baseline Heart fully depolarized

Recording
Injured segment is electrode Baseline shifted downward
partially depolarized
prior to stimulation

Nontransmural MI

Baseline
shifted
upward
Heart fully depolarized

Injured segment is Recording


partially depolarized electrode Normal baseline
prior to stimulation
Figure 4.25. ST deviations in acute MI: diastolic injury current. Top. Ionic leak re-
sults in partial depolarization of injured myocardium in diastole, prior to electrical stimu-
lation, which produces forces heading away from that site and shifts the ECG baseline
downward. This is not noticeable on the ECG because only relative, not absolute, volt-
ages are recorded. Following stimulation, when the entire myocardium has fully depo-
larized, the voltage is true zero but gives the appearance of ST elevation compared with
the abnormally depressed baseline. Bottom. In nontransmural MI, the process is similar,
but the ionic leak arises from the subendocardial tissue so that the partial depolarization
before stimulation is directed toward the recording electrode; hence, the baseline is
shifted upward. When fully depolarized, the voltage is true zero, but the ST segment ap-
pears depressed compared with the shifted baseline.

The systolic current theory of ST segment between Q-wave and non–Q-wave myocar-
shifts contends that in addition to reducing dial infarctions, because the critical thera-
the resting membrane potential, ischemic peutic approaches are different. Decisions
injury shortens the action potential dura- about therapy must be made within min-
tion of affected cells. As a result, the is- utes of evaluating the patient, usually while
chemic cells repolarize faster than neigh- acute ST and T wave deviations are present
boring normal myocytes; therefore, a voltage on the ECG but before Q waves would be ex-
gradient develops between the two zones, pected to have formed. Thus, for the pur-
creating an electrical current directed toward pose of such decision making, an evolving
the ischemic area. This gradient occurs dur- Q-wave MI is referred to as an acute ST-eleva-
ing the ST interval of the ECG, resulting in tion MI. Similarly, a non–Q-wave MI is fre-
ST elevation in the leads overlying the is- quently labeled a non–ST-elevation MI.
Fig. 26 chemic region (Fig. 4.26). Other common causes of ST segment and
As discussed in Chapter 7, when evaluat- T wave abnormalities caused by alterations
ing a patient with acute chest pain, it is very in myocyte repolarization are illustrated in
important to identify and rapidly distinguish Figure 4.27. Fig. 27
10090-04_CH04.qxd 8/31/06 5:25 PM Page 107

The Electrocardiogram 107

ST elevation
Ischemic +
Normal +
–90 mV +
Ischemic cells
repolarize more Recording
idl th l l t d
Figure 4.26. ST deviation in acute MI: systolic injury current. A. Compared with normal myocytes (solid
line), ischemic myocytes (dashed line) display a reduced resting membrane potential and repolarize more rapidly.
B. More rapid repolarization causes the surface of the ischemic zone to be relatively positively charged at the
time the ST segment is inscribed. The associated electrical current (arrows) is directed toward the recording elec-
trode overlying that site, so that the ST segment is abnormally elevated.

Figure 4.27. Conditions that alter repolarization of myocytes and therefore result in ST seg-
ment and T wave abnormalities.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 108

108 Chapter Four

SUMMARY ple normal and abnormal ECGs, with their


interpretations, are shown in Figures 4.28
The electrocardiogram provides a wealth of through 4.36. Fig. 28-36
information about the structure and integrity Disturbances of the cardiac rhythm iden-
of the heart and is one of the most impor- tified by ECGs are discussed in Chapters 11
tant diagnostic tools in cardiology. With the and 12.
knowledge of this chapter in hand, the reader
should be well prepared to practice analyz-
ing electrocardiograms in any of the com- Acknowledgments
plete ECG texts listed under “Additional Contributors to the previous editions of this chapter
Tab. 5 Reading.” Table 4.5 summarizes the sug- were Price Kerfoot, MD; Kyle Low, MD; and Leonard
gested sequence of ECG interpretation. Sam- S. Lilly, MD.

TABLE 4.5. Summary of Sequence of ECG Interpretation

1. Calibration
• Check 1.0 mV vertical box inscription (normal standard = 10 mm)
2. Rhythm
• Sinus rhythm is present if
• Each P wave is followed by a QRS complex
• Each QRS is preceded by a P wave
• P wave is upright in leads I, II, and III
• PR interval is >0.12 sec (3 small boxes)
• If these criteria are not met, determine type of arrhythmia (see Chapter 12)
3. Heart rate
• Use one of three methods:
• 1,500/(number of mm between beats)
• Count-off method: 300—150—100—75—60—50
• Number of beats in 6 seconds ∞ 10
• Normal rate = 60–100 bpm (bradycardia <60, tachycardia >100)
4. Intervals
• Normal PR = 0.12–0.20 sec (3–5 small boxes)
• Normal QRS ≤ 0.10 sec (≤2.5 small boxes)
• Normal QT ≤ half the R–R interval, if heart rate normal
5. Mean QRS axis
• Normal if QRS is primarily upright in leads I and II (+90° to −30°)
• Otherwise, determine axis by isoelectric or perpendicular method
6. P wave abnormalities
• Inspect P in leads II and V1 for left and right atrial enlargement
7. QRS wave abnormalities
• Inspect for left and right ventricular hypertrophy
• Inspect for bundle branch blocks
• Inspect for pathologic Q waves: What anatomic distribution?
8. ST segment or T wave abnormalities
• Inspect for ST elevations:
• Transmural infarct pattern
• Pericarditis (see Chapter 14)
• Inspect for ST depressions or T wave inversions:
• Subendocardial ischemia or infarct
• Commonly accompany ventricular hypertrophy or bundle branch blocks
• Metabolic or chemical abnormalities (see Fig. 4.27)
9. Compare with patient’s previous ECGs
6
10090-04_CH04.qxd
8/31/06

I aVR V1 V4
5:25 PM

II aVL
Page 109

V2 V5

III aVF V3 V6

Figure 4.28. 12-lead ECG (normal). The rectangular upward deflection at the beginning of each line is the voltage calibration signal (1 mV). Rhythm: normal sinus. Rate: 70 bpm.
Intervals: PR, 0.17; QRS, 0.06; QT, 0.40 sec. Axis: 0° (QRS is isoelectric in lead aVF). The P wave, QRS complex, ST segment, and T waves are normal. Notice the gradual increase in R
wave height between leads V1 through V6.

109
110
10090-04_CH04.qxd

I aVR V1 V4
8/31/06
5:25 PM

II aVL V2 V5
Page 110

III aVF V3 V6

Figure 4.29. 12-lead ECG (abnormal). Rhythm: normal sinus. Rate: 75 bpm. Intervals: PR, 0.16; QRS, 0.15; QT, 0.42 sec. Axis: indeterminate (isoelectric in all limb leads). P wave: left
atrial enlargement (1 mm wide and 1 mm deep in lead V1). QRS: widened with RSR′ in lead V1 consistent with right bundle branch block (RBBB). Also, pathologic Q waves are in
leads II, III, and aVF, consistent with inferior wall myocardial infarction (an old one, because the ST segments do not demonstrate an acute injury pattern).
10090-04_CH04.qxd
8/31/06
5:25 PM
Page 111

Figure 4.30. 12-lead ECG (abnormal). Rhythm: normal sinus. Rate: 68 bpm. Intervals: PR, 0.16; QRS, 0.16; QT, 0.40 sec. Axis: +15°. P wave: normal. QRS: widened with RR′
in leads V4–V6 consistent with left bundle branch block (LBBB). The ST segment and T wave abnormalities are secondary to LBBB.

111
112
10090-04_CH04.qxd
8/31/06
5:25 PM
Page 112

Figure 4.31. 12-lead ECG (abnormal). Rhythm: normal sinus. Rate: 66 bpm. Intervals: PR, 0.16; QRS, 0.08; QT, 0.40 sec. Axis: +10°. P wave: normal. QRS: pathologic Q waves in
leads V1–V4, consistent with anteroseptal and anteroapical myocardial infarction (MI). The ST segment and T waves do not demonstrate an acute injury pattern; thus, the MI is old.
10090-04_CH04.qxd
8/31/06
5:25 PM
Page 113

Figure 4.32. 12-lead ECG (abnormal). Rhythm: sinus bradycardia. Rate: 55 bpm. Intervals: PR, 0.20 (in aVF); QRS, 0.10; QT, 0.44 sec. Axis: normal (QRS is predominantly upright
in leads I and II). P wave: normal. QRS: voltage in chest leads is prominent but does not meet criteria for ventricular hypertrophy; pathologic Q waves are present in II, III, and aVF, in-
dicating inferior wall MI, and the tall R waves in V1 and V2 are consistent with posterior MI involvement as well. Marked ST segment elevation is apparent in II, III, and aVF, indicating
that this is an acute MI.

113
114
10090-04_CH04.qxd
8/31/06
5:25 PM
Page 114

Figure 4.33. 12-lead ECG (abnormal). Rhythm: sinus bradycardia. Rate: 55 bpm. Intervals: PR, 0.24 (first-degree AV block; see Chapter 12); QRS, 0.09; QT, 0.44 sec. Axis: 0°.
P wave: normal. QRS: left ventricular hypertrophy (LVH): S in V1 (14 mm) ++ R in V5 (22 mm) > 35 mm. Pathologic Q waves in leads III and aVF raise the possibility of an old inferior
MI. The ST segment depression and T wave inversion are secondary to the abnormal repolarization resulting from LVH.
10090-04_CH04.qxd
8/31/06
5:25 PM
Page 115

Figure 4.34. 12-lead ECG (abnormal). Rhythm: normal sinus. Rate: 68 bpm. Intervals: PR, 0.24 (first-degree AV block; see Chapter 12); QRS, 0.10; QT, 0.36 sec. Axis: −45° (left axis
deviation). P wave: left atrial enlargement (terminal deflection of P wave in V1 is 1 mm wide and 1 mm deep—just barely). QRS: pattern of left anterior fascicular block (LAFB; see
Fig. 4.20). The abnormally small R waves in leads V2–V4 are associated with LAFB resulting from the reduction of initial anterior forces. The ST segment and T waves are unremarkable.

115
116
10090-04_CH04.qxd
8/31/06
5:25 PM
Page 116

Figure 4.35. 12-lead ECG (abnormal). Rhythm: normal sinus. Rate: 95 bpm. Intervals: PR, 0.20; QRS, 0.10; QT, 0.34 sec. Axis: ++160° (right axis deviation [RAD]). P wave: right
atrial enlargement (P wave in lead II is > 2.5 mm tall). QRS: right ventricular hypertrophy (RVH): R > S in V1 with RAD. The T waves are inverted in the anterior leads, at least in part
reflecting abnormal repolarization owing to RVH.
10090-04_CH04.qxd 8/31/06 5:25 PM Page 117

The Electrocardiogram 117

I aVR V1 V4

II aVL V2 V5

III aVF V3 V6

Figure 4.36. 12-lead ECG (abnormal). Rhythm: normal sinus. Rate: 62 bpm. Intervals: PR, 0.14; QRS, 0.10; QT, 0.52
(corrected QT, 0.53, which is prolonged). Axis: +95° (right axis deviation [RAD]). QRS: pattern of left posterior fascic-
ular block, with small R wave in leads I and aVL, small Q wave in leads II, III, and aVF, and right axis deviation (RAD;
see Fig. 4.20). The prolonged QT interval in this patient is the result of antiarrhythmic medication.

Additional Reading Surawicz B, Knilans TK. Chou’s Electrocardiography


in Clinical Practice. 5th Ed. Philadelphia: Saun-
Dubin D. Rapid Interpretation of EKGs. 6th Ed. ders, 2001.
Tampa: Cover Publishing, 2000. Wagner GS. Marriott’s Practical Electrocardiography.
Goldberger AL, Goldberger E. Clinical Electrocardio- 10th Ed. Baltimore: Lippincott Williams & Wilkins,
graphy: A Simplified Approach. 6th Ed. St. Louis: 2001.
Mosby Year Book, 1999.
O’Keefe JH Jr, Hammill SC, Freed MS, et al. The Com-
plete Guide to ECGs. 2nd Ed. Royal Oak, Michi-
gan: Physicians’ Press, 2002.
10090-04_CH04.qxd 8/31/06 5:25 PM Page a

Chapter 4—Author Queries (tables)


1. AU: Correct?
2. AU: Correct?
3. AU: Edit correct?
10090-05_CH05.qxd 8/31/06 5:35 PM Page 118

C H A P T E R

Atherosclerosis
James L. Young
Peter Libby
5
VASCULAR BIOLOGY OF ATHEROSCLEROSIS ATHEROSCLEROSIS RISK FACTORS
Normal Arterial Wall Traditional Risk Factors
Atherosclerotic Arterial Wall Biomarkers
Complications of Atherosclerosis Management of Risk Factors

Atherosclerosis is the leading cause of mor- of the vascular wall, and immune cells. This
tality and morbidity in the developed world. process can smolder throughout adulthood,
Through its major manifestations of cardio- punctuated by acute cardiovascular events.
vascular disease and stroke, it is predicted to This chapter consists of two sections. The
be the leading global killer by the year 2020. first section describes the normal arterial wall,
Historically known as “hardening of the ar- the pathogenesis of atherosclerotic plaque
teries,” atherosclerosis derives its name from formation, and pathologic complications
the Greek roots athere-, meaning “gruel,” and that lead to clinical symptoms. The second
-skleros, meaning “hardness.” section relates findings from population stud-
Although the pathologic hallmarks of ath- ies to attributes that lead to this condition,
erosclerosis have been studied for more than thereby offering opportunities for preven-
a century, its pathogenesis has remained tion and treatment.
a topic of great debate. In the early 1900s,
Anitchkow and Chalatow demonstrated that VASCULAR BIOLOGY OF
feeding rabbits a high-cholesterol diet in- ATHEROSCLEROSIS
duces humanlike atherosclerotic lesions in
blood vessels, supporting the view of athero-
Normal Arterial Wall
sclerosis as a mere disorder of lipid accumu- The arterial wall consists of three layers Fig. 1
lation. This belief persisted through most of (Fig. 5.1): the intima, closest to the arterial
the twentieth century, but more recent evi- lumen and therefore most “intimate” with
dence has generated a different view of ath- the blood; the media, which is the middle
erosclerosis as a chronic inflammatory condi- layer; and the outer adventitia. The intima
tion, involving lipids, thrombosis, elements consists of a single layer of endothelial cells

118
10090-05_CH05.qxd 8/31/06 5:35 PM Page 119

Atherosclerosis 119

Lumen

Endothelial cells Intima


Internal elastic lamina

Smooth muscle cells Media

External elastic lamina


Adventitia
Figure 5.1. Schematic diagram of the arterial wall. The intima, the innermost layer, overlies the mus-
cular media demarcated by the internal elastic lamina. The external elastic lamina separates the media
from the outer layer, the adventitia.

that acts as a metabolically active barrier be- cellular matrix. Understanding the dysfunc-
tween circulating blood and the vessel wall. tion that leads to atherosclerosis requires
The media is the thickest layer of the normal knowledge of the normal function of these
artery. Boundaries of elastin, known as the components.
internal and external elastic laminae, sepa-
rate this middle layer from the intima and
Endothelial Cells
adventitia, respectively. The media is com-
posed of smooth muscle cells and extra- In a healthy artery, the endothelium serves
cellular matrix, and subserves the contrac- structural, metabolic, and signaling func-
tile and elastic functions of the vessel. The tions that maintain homeostasis of the ves-
elastic component, more prominent in sel wall. The tightly adjoined endothelial
large arteries (e.g., the aorta and its primary cells form a barrier that contains blood with-
branches), stretches during the high pres- in the lumen of the vessel and limits the pas-
sure of systole and then recoils during dias- sage of large molecules from the circulation
tole, propelling blood forward. The muscu- into the subendothelial space, which is also
lar component, more prominent in smaller known as the subintima.
arteries such as arterioles, constricts or re- As blood traverses the vascular tree, it en-
laxes to alter vessel resistance and therefore counters antithrombotic molecules produced
luminal blood flow (flow = pressure ÷ resis- by the endothelium that prevent it from
tance; see Chapter 6). The adventitia con- clotting. Some of these molecules reside on
tains the nerves, lymphatics, and blood ves- the endothelial surface (e.g., heparan sul-
sels (vasa vasorum) that nourish the cells of fate, thrombomodulin, and plasminogen
the arterial wall. activators; see Chapter 7), while other an-
Far from an inert conduit, the living arte- tithrombotic products of the endothelium
rial wall is a scene of dynamic interchange enter the circulation (e.g., prostacyclin and
between its cellular components, most im- nitric oxide [NO]; see Chapter 6). Although
portantly endothelial cells, vascular smooth a net anticoagulant state normally prevails,
muscle cells, and their surrounding extra- the endothelium can also produce prothrom-
10090-05_CH05.qxd 8/31/06 5:35 PM Page 120

120 Chapter Five

botic molecules when subjected to various molecules, which anchor mononuclear cells
stressors. to the endothelium and facilitate their mi-
Endothelial cells also secrete substances gration to the site of injury. As described
that modulate contraction of smooth mus- later, under the adverse influences present
cle cells in the underlying medial layer. These during atherogenesis, endothelial cells sim-
substances include vasodilators (e.g., NO, ilarly recruit leukocytes to the vessel wall.
prostacyclin) and vasoconstrictors (e.g., en- Thus, the normal endothelium provides a
dothelin) that alter the resistance of the ves- protective, nonthrombogenic surface with
sel and therefore luminal blood flow. In a homeostatic vasodilator and anti-inflamma-
normal artery, the predominance of va- tory properties (Fig. 5.2). Fig. 2
sodilator substances results in net smooth
muscle relaxation. Several of the aforemen-
Vascular Smooth Muscle Cells
tioned endothelial products (e.g., heparan
sulfate, NO) also function within the vessel Smooth muscle cells within the vessel wall
wall to inhibit proliferation of smooth mus- have both contractile and synthetic capabil-
cle cells into the intima, thus enforcing their ities. Various vasoactive substances modu-
normal residence within the media. late the contractile function, resulting in vaso-
Endothelial cells also play an important constriction or vasodilatation. Such agonists
role in regulating the immune response. include circulating molecules (e.g., angio-
In the absence of pathologic stimulation, tensin II), those released from local nerve
healthy arterial endothelial cells resist leu- terminals (e.g., acetylcholine), and others
kocyte adhesion and are therefore anti- originating from the overlying endothelium
inflammatory. However, endothelial cells in (e.g., endothelin, NO).
postcapillary venules respond to local injury Normal synthetic functions of smooth
or infection by secreting chemokines, chem- muscle cells include production of the col-
icals that attract white blood cells to the lagen, elastin, and proteoglycans that form
area. Such stimulation also causes endothe- the vascular extracellular matrix (see Fig 5.2).
lial cells to produce cell surface adhesion Smooth muscle cells also have the capability

NORMAL ACTIVATED

ENDOTHELIAL
CELLS
• Impermeable to large molecules Permeability
• Anti-inflammatory Inflammatory cytokines
• Resist leukocyte adhesion Leukocyte adhesion molecules
• Promote vasodilation Vasodilatory molecules
• Resist thrombosis Antithrombotic molecules

SMOOTH MUSCLE
CELLS

• Normal contractile function Inflammatory cytokines


• Maintain extracellular matrix Altered matrix synthesis
• Contained in medial layer Migration and proliferation
into subintima
Figure 5.2. Endothelial and smooth muscle cell activation by inflammation. Normal endothelial
and smooth muscle cells maintain the integrity and elasticity of the normal arterial wall while limiting
immune cell infiltration. Inflammatory activation of these vascular cells corrupts their normal functions
and favors proatherogenic mechanisms that drive plaque development.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 121

Atherosclerosis 121

to synthesize vasoactive and inflammatory inflammatory process, including (1) endo-


mediators, including interleukin 6 (IL-6) and thelial dysfunction, (2) accumulation of li-
tumor necrosis factor α (TNF-α), which pro- pids within the intima, (3) recruitment of
mote leukocyte proliferation and induce leukocytes and smooth muscle cells to the
endothelial expression of leukocyte adhe- vessel wall, (4) formation of foam cells, and
sion molecules. These synthetic functions (5) deposition of extracellular matrix, as
become more prominent at sites of athero- summarized in Figure 5.3 and described in Fig. 3
sclerotic plaque and may contribute to its the following sections. Rather than follow a
pathogenesis. sequential path, the cells of atherosclerotic
lesions continuously interact and compete
with each other, shaping the plaque over
Extracellular Matrix
decades into one of many possible profiles.
In healthy arteries, fibrillar collagen and This section categorizes these mechanisms
elastin make up most of the extracellular into three pathologic stages: (1) the fatty
matrix in the medial layer. Interstitial colla- streak, (2) plaque progression, and (3) plaque
gen fibrils, constructed from intertwining disruption (Fig. 5.4). Fig. 4
helical proteins, possess great biomechani-
cal strength, while elastin provides flexibil-
Fatty Streak
ity. Together these components maintain
the structural integrity of the vessel despite Fatty streaks represent the earliest visible
the high pressure within the lumen. Recent lesions of atherosclerosis. On gross inspec-
evidence suggests that the extracellular ma- tion, they appear as areas of yellow discol-
trix also regulates the growth of its resident oration on the artery’s inner surface; how-
cells. Native fibrillar collagen, in particular, ever, they neither protrude substantially
can inhibit smooth muscle cell proliferation into the arterial lumen nor impede blood
in vitro. Furthermore, the matrix influences flow. Surprisingly, fatty streaks exist in the
cellular responses to stimuli: matrix-bound aorta and coronary arteries of most people
cells respond in a specific manner to growth by age 20. They do not cause symptoms, and
factors and are less likely to undergo apop- in some locations in the vasculature, they
tosis (programmed cell death). may regress over time. Although the precise
initiation of fatty streak development is not
known, observations in animal models sug-
Atherosclerotic Arterial Wall
gest that various stressors cause early en-
The arterial wall is a dynamic and regulated dothelial dysfunction, as described in the
system. However, noxious elements can cor- next section. Such dysfunction allows entry
rupt normal homeostasis and pave the way and modification of lipids within the sub-
for atherogenesis. For example, as described intima, where they serve as proinflamma-
later, vascular endothelial and smooth mus- tory mediators that initiate recruitment of
cle cells react readily to inflammatory medi- leukocytes and foam cell formation—the
ators such as IL-1 and TNF-α. These inflam- pathological hallmarks of the fatty streak
matory agents can also activate vascular cells (Fig. 5.5). Fig. 5
to produce IL-1 and TNF-α, contrary to past
dogma that only cells of the immune system
Endothelial Dysfunction
synthesize such cytokines.
Realizing that immune cells were not the Injury to the arterial endothelium represents a pri-
AQ1 only source of proinflammatory agents, in- mary event in atherogenesis. Such injury can re-
vestigations into the role of “activated” en- sult from exposure to diverse agents, includ-
dothelial and smooth muscle cells in athe- ing physical forces and chemical irritants.
rogenesis burgeoned. This fundamental The role of physical stress is suggested by
research has identified several key compo- observations that atherosclerosis tends to
nents that contribute to the atherosclerotic form at arterial branch points (i.e., arterial
10090-05_CH05.qxd 8/31/06 5:35 PM Page 122

122 Chapter Five

Foam cell
8
1 Monocytes
Macrophage Cell apoptosis
LDL 4
Cell 7
Vascular adhesion Smooth muscle
endothelium molecule mitogens
Scavenger 6 Smooth muscle
Oxidized LDL IL-1 MCP-1
5 receptor proliferation
Internal elastic 2 Smooth muscle
lamina 3
migration

Figure 5.3. Schematic diagram of the evolution of atherosclerotic plaque. (1) Accumulation of lipoprotein parti-
cles in the intima. The darker color depicts modification of the lipoproteins (e.g., by oxidation or glycation). (2) Oxida-
tive stress, including constituents of modified LDL (mLDL), induces local cytokine elaboration. (3) These cytokines pro-
mote increased expression of adhesion molecules that bind leukocytes and of chemoattractant molecules (e.g., monocyte
chemoattractant protein 1 [MCP-1]) that direct leukocyte migration into the intima. (4) After entering the artery wall
in response to chemoattractants, blood monocytes encounter stimuli such as macrophage colony–stimulating factor
(M-CSF) that augment their expression of scavenger receptors. (5) Scavenger receptors mediate the uptake of modified
lipoprotein particles and promote the development of foam cells. Macrophage foam cells are a source of additional cy-
tokines and effector molecules such as superoxide anion (O2− ) and matrix metalloproteinases. (6) Smooth muscle cells
migrate into the intima from the media. Note the increasing intimal thickness. (7) Intimal smooth muscle cells divide and
elaborate extracellular matrix, promoting matrix accumulation in the growing atherosclerotic plaque. In this manner, the
fatty streak evolves into a fibrofatty lesion. (8) In later stages, calcification can occur (not depicted) and fibrosis contin-
ues, sometimes accompanied by smooth muscle cell death (including programmed cell death, or apoptosis), yielding a
relatively acellular fibrous capsule surrounding a lipid-rich core that may also contain dying or dead cells. IL-1, interleukin 1;
LDL, low-density lipoprotein. (Modified from Zipes D, Libby P, Bonow RO, et al., eds. Heart Disease: A Textbook of Cardio-
vascular Medicine. 7th Ed. Philadelphia: Elsevier Saunders, 2005:925.)

bifurcations). In straight sections of arteries, Moreover, laminar flow accentuates expres-


the normal laminar (i.e., smooth) shear sion of the antioxidant enzyme superoxide
forces favor the endothelial expression of dismutase, which protects against reactive
NO, which is beneficial as an endogenous oxygen species produced by chemical irri-
vasodilator, an inhibitor of platelet aggrega- tants or transient ischemia, as described in
tion, and an anti-inflammatory substance. Chapter 6. Conversely, disturbed flow is typ-

A B C

FATTY STREAK PLAQUE PROGRESSION PLAQUE DISRUPTION


• Endothelial dysfunction • Smooth muscle cell migration • Disrupted plaque integrity
• Lipoprotein entry and • Altered matrix synthesis and • Thrombus formation
modification degradation
• Leukocyte recruitment
• Foam cell formation
Figure 5.4. Stages of plaque development. A. The fatty streak develops as a result of endothelial dysfunction,
lipoprotein entry and modification, leukocyte recruitment, and foam cell formation. B. Plaque progression is char-
acterized by migration of smooth muscle cells into the intima, where they divide and elaborate extracellular ma-
trix. A lipid core is contained by the fibrous cap. C. Hemodynamic stresses and degradation of extracellular matrix
increase the susceptibility of the fibrous cap to rupture, allowing superimposed thrombus formation. (Modified
from Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation 2002;105:1136.)
10090-05_CH05.qxd 8/31/06 5:35 PM Page 123

Atherosclerosis 123

the result is an activated state manifested


Chemical irritants Hemodynamic stress by (1) impairment of the endothelium’s
role as a permeability barrier, (2) release
of inflammatory cytokines, (3) increased
Endothelial Dysfunction
production of cell surface adhesion mole-
cules that recruit leukocytes, (4) altered re-
lease of vasoactive substances (e.g., prosta-
Lipoprotein entry & Inflammatory
modification cytokines
cyclin and NO), and (5) interference with
normal antithrombotic properties. These
undesired effects of endothelial dysfunc-
tion lay the groundwork for subsequent
Chemokines Leukocyte adhesion events in the development of atherosclero-
molecules sis (see Fig 5.2).

Leukocyte recruitment Lipoprotein Entry and Modification


Unregulated update The activated dysfunctional endothelium
of modified LDL
no longer serves as an effective barrier to
the passage of circulating lipoproteins into
Foam Cell Formation
the arterial wall (see Box 5.1 for a review Box 1
Figure 5.5. Endothelial dysfunction is the primary
of the major lipoprotein pathways). In-
event in plaque initiation. Physical and chemical stres- creased endothelial permeability allows
sors alter the normal endothelium, allowing lipid entry the entry of low-density lipoprotein (LDL)
into the subintima and promoting inflammatory cytokine
release. This cytokine- and lipid-rich environment pro-
into the intima, a process facilitated by an
motes recruitment of leukocyte to the subintima, where elevated circulating LDL concentration.
they may transform into foam cells—a prominent inflam- Once within the intima, LDL accumulates
matory participant.
in the subendothelial space by binding to
components of the extracellular matrix
ical at arterial branch points, which impairs known as proteoglycans. This “trapping”
these locally atheroprotective endothelial increases the residence time of LDL within
functions. Accordingly, arteries with few the vessel wall, where the lipoprotein may
branches (e.g., the internal mammary artery) undergo chemical modifications that ap-
show relative resistance to atherosclerosis, pear critical to the development of athero-
whereas bifurcated vessels (e.g., the common sclerotic lesions. Hypertension, a major
carotid and left coronary arteries) are com- risk factor for atherosclerosis, may pro-
mon sites for atherosclerosis deposition. mote retention of lipoproteins in the in-
Endothelial dysfunction may also result tima by accentuating the production of
from exposure to a “toxic” chemical envi- LDL-binding proteoglycans by smooth mus-
ronment. For example, cigarette smoking, ab- cle cells.
normal circulating lipid levels, or diabetes— Oxidation is one type of modification that
all known risk factors for atherosclerosis—can befalls LDL trapped in the subendothelial
promote endothelial dysfunction. Each of space. It can result from the local action of
these states increases endothelial production reactive oxygen species and pro-oxidant en-
of reactive oxygen species—notably, super- zymes derived from activated endothelial or
oxide anion—which interact with other smooth muscle cells, or from macrophages
intracellular molecules to influence the meta- that penetrate the vessel wall. In diabetic
bolic and synthetic functions of the endothe- patients with sustained hyperglycemia, gly-
lium. Consequently, the cells are skewed to- cation of LDL can occur, a transformation that
ward proinflammatory processes. may ultimately render LDL antigenic and
When physical and chemical stressors proinflammatory. The biochemical modifi-
corrupt normal endothelial homeostasis, cations of LDL act early and contribute
10090-05_CH05.qxd 8/31/06 5:35 PM Page 124

124 Chapter Five

Box 5.1 The Lipoprotein Transport System

Lipoproteins ferry water-insoluble fats through the bloodstream. These particles consist of
a lipid core surrounded by more hydrophilic phospholipid, free cholesterol, and apolipopro-
teins (also called apoproteins, or apos). The apoproteins present on various classes of
lipoprotein molecules serve as the “conductors” of the system, directing the lipoproteins
to specific tissue receptors and mediating enzymatic reactions. Five major classes of
lipoproteins exist, distinguished by their densities, lipid constituents, and associated
apoproteins. In order of increasing density, they are chylomicrons, very low-density
lipoprotein (VLDL), intermediate-density lipoprotein (IDL), low-density lipopro-
teins (LDL), and high-density lipoprotein (HDL). The major steps in the lipoprotein
pathways are labeled in the accompanying figure and described as follows. The key
apoproteins (apos) at each stage are indicated in the figure in parentheses.

Exogenous Pathway Endogenous Pathway

Dietary fat
5
Bile acids and Non-hepatic
1
cholesterol cells
10
LDL
Liver (Apo B-100)
Intestine LPL
4 8 9
6 HL

Chylomicron
remnants VLDL IDL
Chylomicrons (Apo B-100, C, E) (Apo B-100, E)
(Apo B-48, A, C, E) (Apo B-48, E)

3 7
C, E
C, E

2 Lipoprotein lipase Lipoprotein lipase


P)
(CET
Apo
Apo

FFA FFA
HDL HDL

Muscle Adipose tissue Muscle Adipose tissue

Exogenous (Intestinal) Pathway


1. Dietary fats are absorbed by the small intestine and repackaged as chylo-
microns, accompanied by apo B-48. Chylomicrons are large particles, partic-
ularly rich in triglycerides, that enter the circulation via the lymphatic system.
2. Apo E and subtypes of apo C are transferred to chylomicrons from HDL par-
ticles in the bloodstream.
3. Apo C (subtype CII) enhances interactions of chylomicrons with lipoprotein
lipase (LPL) on the endothelial surface of adipose and muscle tissue. This re-
action hydrolyzes the triglycerides within chylomicrons into free fatty acids
(FFA), which are stored by adipose tissue or used for energy in cardiac and
skeletal muscle.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 125

Atherosclerosis 125

4. Chylomicron remnants are removed from the circulation by the liver, medi-
ated by apo E.
5. One fate of cholesterol in the liver is incorporation into bile acids, which are
exported to the intestine, completing the exogenous pathway cycle.

Endogenous (Hepatic) Pathway


Because dietary fat availability is not constant, the endogenous pathway provides a reli-
able supply of triglycerides for tissue energy needs:
6. The liver packages cholesterol and triglycerides into VLDL particles, accom-
panied by apo B-100 and phospholipid. The triglyceride content of VLDL is
much higher than that of cholesterol, but this is the main means by which the
liver releases cholesterol into the circulation.
7. VLDL is catabolized by LPL (similar to chylomicrons, as described in step 3),
releasing fatty acids to muscle and adipose tissue. During this process, VLDL
also interacts with HDL, exchanging some of its triglyceride for apo C sub-
types, apo E, and cholesteryl ester from HDL. The latter exchange (important
in reverse cholesterol transport, as described in the next section) is mediated
by cholesteryl ester transfer protein (CETP).
8. Approximately 50% of the VLDL remnants (IDL) are then cleared in the liver
by hepatic receptors that recognize apo E.
9. The remaining IDL is catabolized further by LPL and hepatic lipase (HL), which
remove additional triglyceride, apo E, and apo Cs, forming LDL particles.
10. Plasma clearance of LDL occurs primarily via LDL receptor–mediated endocy-
tosis in the liver and peripheral cells, directed by LDL’s apo B-100.

Cholesterol Homeostasis and Reverse Cholesterol Transport


Intracellular cholesterol content is tightly maintained by de novo synthesis, cellular uptake,
storage, and efflux from the cell. The enzyme HMG-CoA reductase is the rate-limiting
element of cholesterol biosynthesis, and cellular uptake of cholesterol is controlled by
receptor-mediated endocytosis of circulating LDL (see step 10). When intracellular cho-
lesterol levels are low, the transcription factor sterol regulatory element binding protein

apo A1
Internalized
cholesterol
ester
Free
Nascent
apo A1
HDL
LCAT Mature
HDL CETP

ABCA1 ABCG1 Transfer of


cholesterol
ester to
VLDL, IDL, LDL
(in exchange for TG)
Excess Peripheral
cholesterol cells
10090-05_CH05.qxd 8/31/06 5:35 PM Page 126

126 Chapter Five

(SREBP) is released from the endoplasmic reticulum. The active fragment of SREBP enters the
nucleus to increase transcription of HMG-CoA reductase and the LDL receptor, which,
through their subsequent actions, tend to normalize the intracellular cholesterol content. The
process is illustrated in the accompanying figure. AQ3
Under conditions of intracellular cholesterol excess, peripheral cells increase the tran-
scription of the ATP binding cassette A1 and G1 genes (ABCA1 and ABCG1, respectively).
The ABCA1 gene codes for a transmembrane protein transporter that initiates efflux of
cholesterol from the cell to lipid-poor circulating apo AI (which is synthesized by the liver
and intestine), thus forming nascent (immature) HDL particles. ABCG1 facilitates further
efflux of cholesterol to more-mature HDL particles. As free cholesterol is acquired by cir-
culating HDL, it is esterified by lecithin cholesterol acyltransferase, an enzyme activated by
apo AI. The hydrophobic cholesterol esters move into the particle’s core. Most cholesterol
esters in HDL can then be exchanged in the circulation (via the enzyme CETP) with any of
the apo B–containing lipoproteins (i.e., VLDL, IDL, LDL), which deliver the cholesterol back
to the liver. HDL can also transport cholesterol to the liver and hormone-producing tissues
via a scavenger receptor termed SR-B1.

to the inflammatory mechanisms initiated where they likely furnish an important


by endothelial dysfunction, and they may source of cytokines.
promote inflammation throughout the lifes- Modified LDL and proinflammatory cy-
pan of the plaque. In the fatty streak, and tokines (e.g., IL-1, TNF-α) can induce LAM
likely throughout plaque development, mod- and chemoattractant cytokine (chemokine)
ified LDL (mLDL) promotes leukocyte re- expression independently; however, mLDL
cruitment and foam cell formation. also potently stimulates endothelial and
smooth muscle cells to produce proinflam-
matory cytokines, thereby reinforcing the
Leukocyte Recruitment
direct action. This dual ability of mLDL
Recruitment of leukocytes (primarily mono- to promote leukocyte recruitment and in-
cytes and T lymphocytes) to the vessel wall flammation directly and indirectly persists
is a key step in atherogenesis. The process throughout atherogenesis.
depends on (1) expression of leukocyte ad-
hesion molecules (LAM) on the normally
Foam Cell Formation
nonadherent endothelial luminal surface,
and (2) chemoattractant signals (e.g., mono- After monocytes adhere to and penetrate
cyte chemotactic protein 1 [MCP-1], IL-8, the intima, they differentiate into phago-
AQ2 IP-10) that direct diapedesis (passage of cells cytic macrophages and imbibe lipoproteins
through the intact endothelium) into the to form foam cells. Importantly, foam cells
subintimal space. Two major subsets of LAM do not arise from uptake of LDL by the clas-
persist in the inflamed atherosclerotic plaque: sic cell surface LDL-receptor mechanism de-
(1) the immunoglobulin gene superfamily scribed in Box 5.1, because the high choles-
(particularly vascular cell adhesion molecule terol content within these cells actually
1 [VCAM-1] and intercellular adhesion mol- suppresses expression of the receptor. Fur-
ecule 1 [ICAM-1]), and (2) the selectins (par- thermore, the classic LDL receptor does not
ticularly E- and P-selectin). Despite the cen- recognize chemically modified LDL. Rather,
tral role of T lymphocytes in the immune macrophages rely on a family of “scavenger”
system, plaque LAMs and chemoattractant receptors that preferentially bind and in-
signals direct mainly monocytes to the form- ternalize mLDL. Unlike uptake via the clas-
ing lesion. Although outnumbered, T lym- sic LDL receptor, mLDL ingestion by scav-
phocytes localize within plaques at all stages enger receptors evades negative feedback
10090-05_CH05.qxd 8/31/06 5:35 PM Page 127

Atherosclerosis 127

inhibition and permits engorgement of the tion of smooth muscle cells from the arter-
macrophages by cholesterol-rich lipid, re- ial media into the intima, proliferation of
sulting in the typical appearance of foam the smooth muscle cells within the intima,
cells. Although such uptake initially may be and secretion of extracellular matrix macro-
beneficial (by sequestering proinflamma- molecules by the smooth muscle cells. Foam
tory mLDL particles), the impaired efflux of cells, activated platelets, and endothelial
these cells leads to local accumulation in the cells all elaborate substances that signal
plaque, mitigating their protective role and smooth muscle cell migration and prolifera-
Fig. 6
instead serving as a source of proinflamma- tion (Fig. 5.6).
tory cytokines that fuel atherosclerotic plaque Foam cells produce several factors that
progression. contribute to smooth muscle cell recruit-
ment. For example, they release platelet-
derived growth factor (PDGF), which likely
Plaque Progression
stimulates the migration of smooth muscle
Whereas endothelial cells play a central role cells across the internal elastic lamina
in formation of the fatty streak, smooth and into the subintimal space, where they
muscle cell migration into the intima dom- subsequently replicate. Foam cells also
inates early plaque progression. Spanning release cytokines and growth factors (e.g.,
decades of development, the typical ather- TNF-α, IL-1, fibroblast growth factor, and
osclerotic plaque acquires a distinct throm- transforming growth factor β [TGF-β]) that
bogenic lipid core that under-lies a protec- further stimulate smooth muscle cell
tive fibrous cap. Early plaque growth shows proliferation and synthesis of extracellular
a compensatory outward remodeling of the matrix proteins. Furthermore, these stimu-
plaque wall that preserves the diameter of latory cytokines induce smooth muscle
the arterial lumen and does not limit blood cell and leukocyte activation, promoting
flow. This stage can even evade detection by
angiography. Later plaque growth however
may significantly restrict the vessel lumen
Foam cells in fatty streak
and impede perfusion. Such flow-limiting
plaques can result in tissue ischemia, caus-
ing symptoms such as angina pectoris (see Tissue
Endothelial
Chapter 6) or intermittent claudication of dysfunction factor
the extremities (see Chapter 15).
NO
As described in Chapter 7, most acute PGI2
coronary syndromes (acute myocardial in- ↑ PDGF Thrombosis
farction or unstable angina pectoris) result and platelet
activation
when the fibrous cap of an atherosclerotic Heparinase
plaque ruptures and prothrombotic mole- Endothelial
cules within the lipid core are exposed, pre- heparan
sulfate
cipitating an acute thrombus that suddenly
occludes the arterial lumen. However, as de-
scribed later in the chapter, the extracellu- Smooth muscle cells migrate to intima,
proliferate, and produce extracellular matrix
lar matrix plays a pivotal role in fortifying
the fibrous cap, isolating the thrombogenic
Fibrous
plaque interior from coagulation substrates plaque
in the circulation.
Figure 5.6. Progression from the fatty streak involves
the migration and proliferation of smooth muscle
cells. Substances released from foam cells, dysfunctional
Smooth Muscle Cell Migration endothelial cells, and platelets contribute to this process.
IL-1, interleukin-1; NO, nitric oxide; PDGF, platelet-derived
The transition from fatty streak to fibrous growth factor; PGI2, prostacyclin; TGF-β, transforming
atheromatous plaque involves the migra- growth factor β; TNF-α, tumor necrosis factor α.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 128

128 Chapter Five

further cytokine release, thus reinforc- Extracellular Matrix Metabolism


ing and maintaining inflammation in the
As the predominant collagen-synthesizing
lesion.
cell type, smooth muscle cells should, via
It has been traditionally assumed that
their proliferation, favor fortification of the
plaque growth is gradual and continuous.
fibrous cap. However, net matrix deposition
However, current evidence suggests that
depends on the balance of synthesis by
this progression may be punctuated by
smooth muscle cells and degradation, medi-
subclinical events with bursts of smooth
ated in part by a class of proteolytic enzymes
muscle replication. For example, morpho-
known as matrix metalloproteinases (MMP).
logical evidence of resolved intraplaque
While PDGF and TGF-β stimulate smooth
hemorrhages indicates that small breaches
muscle cell production of interstitial colla-
in plaque integrity can occur without
gens, the T-lymphocyte–derived cytokine
clinical symptoms or signs. At the cellular
interferon γ (IFN-γ) inhibits smooth muscle
level, such breaches expose tissue factor
cell collagen synthesis. Furthermore, inflam-
from foam cells, which activates coagula-
matory cytokines stimulate local foam cells
tion and microthrombus formation. Acti-
to secrete collagen- and elastin-degrading
vated platelets within such microthrombi
MMP, thereby weakening the fibrous cap
release additional potent factors that can
and predisposing it to rupture (Fig. 5.7). Fig. 7
spur a local wave of smooth muscle cell mi-
gration and proliferation. These factors in-
clude PDGF and heparinase. The latter de- Plaque Disruption
grades heparan sulfate, a polysaccharide in
Plaque Integrity
the extracellular matrix that normally in-
hibits smooth muscle cell migration and Over decades, the tug-of-war between ma-
proliferation. trix synthesis and degradation continues,

Deg
ra dat
sis ion
the
S yn

Lumen

MMP
Collagen and
elastin Fibrous Cap

Smooth muscle
+ cell Foam +
– + cell
PDGF IFN-γ CD40L IL-1
TGF-β TNF-α
MCP-1
Lipid core
T lymphocyte
Figure 5.7. Matrix metabolism underlies fibrous cap integrity. The net deposition of extracellular ma-
trix is the result of competing synthesis and degradation reactions. Smooth muscle cells synthesize the bulk
of the fibrous cap constituents, such as collagen and elastin. However, foam cells also elaborate destructive
proteolytic enzymes, such as the collagen-degrading matrix metalloproteinases and the elastolytic cathepsins.
T-lymphocyte–derived factors favor destruction of the fibrous cap. However, all plaque residents contribute
to the cytokine milieu of the plaque, providing multiple activating and inhibitory stimuli as shown. IFN-γ,
interferon γ; IL-1, interleukin 1; MCP-1, monocyte chemoattractant protein 1; PDGF, platelet-derived growth
factor; TGF-β, transforming growth factor β; TNF-α, tumor necrosis factor α. (Modified from Libby P. The mo-
lecular bases of acute coronary syndromes. Circulation 1995;91:2844–2850; Young JL, Libby P, Schönbeck
U. Cytokines in the pathogenesis of atherosclerosis. Thromb Haemost 2002;88:554–567.) 1 LINE LO
10090-05_CH05.qxd 8/31/06 5:35 PM Page 129

Atherosclerosis 129

but not without consequences. Death of ruption of the fibrous cap leads to a tran-
smooth muscle and foam cells, either owing sient, nonobstructive mural thrombus or to
to excess inflammatory stimulation or by a completely occlusive clot, the latter being
contact activation of apoptosis pathways, implicated in >90% of acute clinical events.
liberates cellular contents, contributing im- The probability of a major thrombotic
bibed lipids and cellular debris to the grow- event reflects the balance between com-
ing lipid core. The size of the lipid core has peting processes of coagulation and fibri-
biomechanical implications for the stability nolysis. Inflammatory stimuli common in
of the plaque. With increasing size and pro- the plaque microenvironment (e.g., CD40L)
trusion into the arterial lumen, hemody- elicit tissue factor, the initiator of the extrin-
namic stress focuses on the plaque border sic coagulation pathway, from many plaque
abutting normal tissue, the so-called shoul- components, including smooth muscle cells,
der region. In addition to shouldering in- endothelial cells, and macrophage-derived
creased stress, local accumulation of foam foam cells. Beyond enhancing expression
cells and T lymphocytes at this site acceler- of the potent procoagulant tissue factor,
ates degradation of extracellular matrix, mak- inflammatory stimuli further support throm-
ing this region the most common site of bosis by favoring expression of antifibrino-
plaque rupture. lytics (e.g., plasminogen activator inhibitor-
The net deposition and distribution of 1) over expression of anticoagulants (e.g.,
the fibrous cap is an important determi- thrombomodulin, heparin-like molecules,
nant of overall plaque integrity. Whereas le- protein S) and profibrinolytic mediators (tis-
sions with thick fibrous caps may cause pro- sue plasminogen activator and urokinase-
nounced arterial narrowing, they have less type plasminogen activator; Fig. 5.9). More- Fig. 9
propensity to rupture. Conversely, plaques over, as described earlier, the activated
that have thinner caps (and often appear endothelium also promotes thrombin for-
less obstructive by angiography) tend to mation, coagulation, and fibrin deposition at
be fragile and more likely to rupture and in- the vascular wall.
cite thrombosis. Current terminology de- A person’s propensity toward coagulation
scribes the extreme spectrums of integrity as may be enhanced by genetics (e.g., the pres-
stable plaques (marked by a thick fibrous cap ence of a procoagulant prothrombin gene
and small lipid core) or vulnerable plaques mutation), coexisting conditions (e.g., dia-
(marked by a thin fibrous cap, rich lipid betes), and/or lifestyle factors (e.g., smoking,
core, extensive macrophage infiltrate, and a visceral obesity). Consequently, the concept
Fig. 8 paucity of smooth muscle cells; Fig. 5.8). De- of “vulnerable plaque” has expanded to
spite the common use of these terms, it is that of the “vulnerable patient” to acknow-
important to recognize that this distinction ledge other contributors to a person’s vas-
oversimplifies the heterogeneity of plaques cular risk.
and may overestimate the ability to foresee
a plaque’s “clinical future” based on struc-
Complications of Atherosclerosis
tural information.
Atherosclerotic plaques do not distribute
homogeneously throughout the vasculature.
Thrombogenic Potential
They usually develop first in the dorsal aspect
Rupture of atherosclerotic plaque does not of the abdominal aorta and proximal coro-
inevitably cause major clinical events such nary arteries, followed by the popliteal ar-
as myocardial infarction and stroke. As de- teries, descending thoracic aorta, internal
scribed in the previous section, small non- carotid arteries, and renal arteries. There-
occlusive thrombi may reabsorb into the fore, the regions perfused by these vessels
plaque, stimulating further smooth muscle most commonly suffer the consequences of
growth and fibrous deposition (see Fig 5.8). atherosclerosis.
It is in large part the thrombogenic potential Complications of atherosclerotic plaques—
NE LONG of the plaque that determines whether dis- including calcification, rupture, hemorrhage,
10090-05_CH05.qxd 8/31/06 5:35 PM Page 130

130 Chapter Five

Normal artery

Early atheroma

“Stable” plaque “Vulnerable” plaque


• Small lipid pool • Large lipid pool
• Thick fibrous cap • Thin fibrous cap
• Preserved lumen • Many inflammatory cells

Ruptured plaque
with thrombus formation

Healed rupture
• Narrowed lumen Acute
• Fibrous intima myocardial
infarction

Figure 5.8. Stable versus vulnerable plaques. Stable plaque is characterized by a small lipid pool and a thick fibrous
cap, whereas vulnerable plaque tends to have a large lipid pool and relatively thin fibrous cap. The latter is subject to
rupture, resulting in thrombosis. A resulting occlusive clot can cause an acute cardiac event, such as myocardial infarc-
tion. A lesser thrombus may resorb, but the wound-healing response stimulates smooth muscle cell proliferation and
collagen production, thereby thickening the fibrous cap and narrowing the vessel lumen further. (Modified from Libby
P. Inflammation in atherosclerosis. Nature 2002;420:868–874.)
10090-05_CH05.qxd 8/31/06 5:35 PM Page 131

Atherosclerosis 131

Favors Occlusive Thrombus Favors Thrombus Resorbtion


Pro-coagulant Anticoagulants
• Tissue factor • Thrombomodulin
• Protein S
• Heparin-like molecules
Antifibrinolytic Profibrinolytics
• PAI-1 • tPA, uPA

Figure 5.9. Competing factors in thrombosis. The clinical manifestations of


plaque disruption rely not only on the stability of the fibrous cap but also on the
thrombogenic potential of the plaque core. The balance of physiological media-
tors dictates the prominence of the thrombus, resulting in either luminal occlu-
sion or resorption into the plaque. PAI-1, plasminogen activator inhibitor 1; tPA,
tissue plasminogen activator; uPA, urokinase-type plasminogen activation.

and embolization—can have dire clinical quences in different organ systems (Fig. 5.10). Fig. 10
consequences owing to acute restriction of In the case of coronary plaque, lesions with
blood flow or alterations in vessel wall in- gradually progressive expansion and a thick
tegrity. These complications, which are dis- fibrous cap tend to narrow the vessel lumen
cussed in greater detail in later chapters, in- and result in intermittent chest discomfort
clude the following: on exertion (angina pectoris). In contrast,
plaque that does not significantly compro-
1. Calcification of atherosclerotic plaque,
mise the vessel lumen but has characteristics
which imparts a pipelike rigidity to the of vulnerability (thin fibrous cap and large
vessel wall and increases its fragility. lipid core) can rupture, leading to acute
2. Rupture or ulceration of atherosclerotic thrombosis and myocardial infarction (see
plaque, which exposes procoagulants Chapter 7). Such nonstenotic plaques are
within the plaque to circulating blood, often numerous and dispersed throughout
causing a thrombus to form at that site. the arterial tree, and because they do not sig-
Such thrombosis can occlude the vessel nificantly compromise the vessel lumen,
and result in infarction of the involved they do not produce symptoms and often
organ. Alternatively, the thrombus mate- evade detection by exercise testing or even
rial can incorporate into the lesion and angiography.
add to the bulk of the plaque. The description presented here of athero-
3. Hemorrhage into the plaque owing to genesis and its complications can explain
rupture of the fibrous cap or of the micro- the limitations of widely employed treat-
vessels that form within the lesion. The ments. For example, percutaneous interven-
resulting intramural hematoma may fur- tion (angioplasty and stent placement) of
ther narrow the vessel lumen. symptomatic coronary stenoses is an effec-
4. Embolization of fragments of disrupted tive strategy for relief of angina pectoris but
atheroma to distal vascular sites. has not been shown to prevent myocardial
5. Weakening of the vessel wall: the fibrous infarction or prolong life. This disparity
likely reflects the general nature of vulnera-
plaque subjects the neighboring medial
ble plaques as diffuse and angiographically
layer to increased pressure, which may
unimpressive, such that they tend not to at-
provoke atrophy and loss of elastic tissue
tract attention prior to rupture. It follows
with subsequent dilatation of the artery,
that lifestyle modifications and drug the-
forming an aneurysm.
rapies that curb the risk factors for plaque
The complications of atherosclerotic formation are a critical foundation for pre-
plaque may result in different clinical conse- venting progressive atherogenesis.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 132

132 Chapter Five

Stroke
• Embolic stroke 4
• Thrombotic stroke 2 3 Coronary artery disease
• Myocardial ischemia 1
• Unstable angina 2 3
• Myocardial infarction 2 3

Renal Artery Disease


• Atheroembolic renal disease 4
• Renal artery stenosis 1

Aneurysms 5

Peripheral artery disease


• Limb claudication 1

1 Narrowing of vessel by fibrous plaque


2 Plaque ulceration or rupture
3 Intraplaque hemorrhage
4 Peripheral emboli
5 Weakening of vessel wall

Figure 5.10. Clinical sequelae of atherosclerosis. Complications of atherosclerosis arise from the mechanisms
listed in the figure.

ATHEROSCLEROSIS RISK FACTORS them, have established the importance of


modifiable risk factors for atherosclerosis, in-
In the early portion of the twentieth cen- cluding (1) aberrant levels of circulating
tury, it was widely believed that atheroscle- lipids (dyslipidemia), (2) tobacco smoking,
rosis was an inevitable process of aging. How- (3) hypertension, (4) diabetes mellitus, and
ever, in 1948, the landmark Framingham (5) lack of physical activity and obesity
Heart Study began to examine the relation- (Table 5.1). The INTERHEART study suggests Tab. 1
ship between specific attributes and cardio- that these modifiable factors account for up
vascular disease, establishing the concept of to 90% of population-attributable cardiac
atherosclerotic risk factors. Among later risk. Major nonmodifiable risk factors include
studies, the Multiple Risk Factor Interven- advanced age, male gender, and heredity—
tion Trial screened more than 325,000 men that is, a history of coronary heart disease
to correlate risk factors with subsequent car- among first-degree relatives at a young age
diovascular disease and mortality. More re- (before age 55 for a male relative or before
cently, the INTERHEART study evaluated age 65 for a female relative).
such relationships in 29,000 survivors of In addition to these standard predictors,
myocardial infarction in 52 countries, rep- certain biological markers associated with
resenting varied ethnic and economic back- the development of cardiovascular events
grounds. These undertakings, and others like have been undergoing rigorous evaluation
10090-05_CH05.qxd 8/31/06 5:35 PM Page 133

Atherosclerosis 133

various critical functions in normal physiol-


TABLE 5.1. Common Cardiovascular ogy. All cells require cholesterol to form
Risk Factors
membranes and maintain fluidity of the
Modifiable Risk Factors phospholipid bilayer. Some cells use choles-
Dyslipidemia (elevated LDL, decreased HDL) terol to synthesize specialized products, such
Tobacco smoking as steroid hormones and bile salts.
Hypertension
Elevated levels of LDL particles correlate
Diabetes mellitus, metabolic syndrome
Lack of physical activity with an increased incidence of atherosclero-
Nonmodifiable risk factors sis and coronary artery disease. When pre-
Advanced age sent in excess, LDL can accumulate in the
Male gender subendothelial space and undergo the che-
Heredity
mical modifications that further damage the
HDL, high-density lipoprotein; LDL, low-density
lipoprotein. intima, as described earlier, initiating and
perpetuating the development of athero-
sclerotic lesions. Thus, LDL is commonly
known as “bad cholesterol.” Conversely, el-
as “novel” risk factors. These include ele- evated high-density lipoprotein (HDL) par-
vated circulating levels of (1) the amino ticles appear to protect against atheroscle-
acid metabolite homocysteine, (2) the spe- rosis, likely because of HDL’s ability to
cial lipoprotein particle Lp(a), and (3) cer- transport cholesterol away from the periph-
tain markers of inflammation, including eral tissues back to the liver for disposal
the acute-phase reactant C-reactive protein. (termed reverse cholesterol transport; see
The following sections address the tradi- Box 5.1) and because of its antioxidative
tional risk factors, followed by a description properties. Thus, HDL has earned the moni-
of the newer biomarkers. ker “good cholesterol.”
Elevated serum LDL may persist for many
Traditional Risk Factors reasons, including a high-fat diet or because
of abnormalities in the LDL-receptor clear-
Dyslipidemia ance mechanism. Patients with genetic de-
A large and consistent body of evidence es- fects in the LDL receptor, which leads to a
tablishes abnormal circulating lipid levels as condition known as familial hypercholes-
a major risk factor for the development of terolemia, cannot remove LDL from the cir-
atherosclerosis. Observational studies have culation efficiently. Heterozygotes with this
shown that the United States and other soci- condition have one normal and one defec-
eties in which consumption of saturated fat tive gene coding for the receptor. They dis-
and cholesterol levels are high, mortality play high plasma LDL levels and develop
rates from coronary disease are higher com- premature atherosclerosis. Homozygotes
pared with those in countries with tradition- who completely lack functional LDL recep-
ally low saturated fat intake and low serum tors may experience vascular events, such as
cholesterol levels (e.g., Japan and certain acute myocardial infarction, as early as the
Mediterranean nations). Similarly, data from first decade of life.
the Framingham Heart Study and other tri- Increasing evidence also implicates tri-
als have shown that the risk of ischemic glyceride-rich lipoproteins, such as very
heart disease increases with higher total se- low-density lipoprotein (VLDL) and inter-
rum cholesterol levels. The coronary risk is mediate density lipoprotein (IDL), in the
approximately twice as high for a person development of atherosclerosis. However,
with a total cholesterol level of 240 mg/dL it remains undetermined whether these
compared with a person whose cholesterol particles participate directly in atherogene-
level is 200 mg/dL. sis or simply keep company with low levels
However, not all lipoproteins bearing cho- of HDL cholesterol. Note that poorly con-
lesterol are harmful. In fact, cholesterol has trolled type 2 diabetes mellitus is often as-
10090-05_CH05.qxd 8/31/06 5:35 PM Page 134

134 Chapter Five

sociated with the combination of hyper- When lifestyle modifications fail to


triglyceridemia and low HDL levels. achieve target values, pharmacologic agents
are used to improve abnormal lipid levels.
The major groups of lipid-altering agents
Lipid-Altering Therapy
(see Chapter 17) include HMG-CoA reduc-
Strategies that improve abnormal lipid lev- tase inhibitors, niacin, fibric acid deriva-
els can limit the consequences of athero- tives, cholesterol intestinal absorption in-
sclerosis. Many large studies of patients with hibitors, and bile acid–binding agents. The
coronary disease show that dietary or phar- HMG-CoA reductase inhibitors (also known
macologic reduction of serum cholesterol as statins) have emerged as the most effec-
can slow the progression of atherosclerotic tive LDL-lowering drugs. They inhibit the
plaque. These trials form the basis of screen- rate-limiting enzyme responsible for cho-
ing guidelines, devised by the National Cho- lesterol biosynthesis. The resulting reduction
lesterol Education Program panel, which in intracellular cholesterol concentration
recommends a fasting lipid profile every promotes increased LDL-receptor expression
years for all adults. These guidelines iden- and thus augmented clearance of LDL parti-
tify an “optimal” LDL cholesterol level as cles from the bloodstream. Statins also
<100 mg/dL. Patients with established ath- lower the rate of VLDL synthesis by the
erosclerosis, or those who have equivalent liver (thus lowering circulating triglyceride
risk (e.g., diabetes) should receive treatment levels) and, by an unknown mechanism,
to attain this goal. An even lower goal of raise HDL.
<70 mg/dL is recommended for patients Major clinical trials evaluating statin ther-
with atherosclerotic disease at the highest apy have demonstrated consistently striking
risk of future vascular events: those who have reductions in ischemic cardiac events, the
recently sustained an acute coronary syn- occurrence of strokes, and (in many cases)
drome (see Chapter 7) and people with mul- mortality rates (Fig. 5.11). The benefits doc- Fig. 11
tiple risk factors, especially when diabetes, umented in these studies have extended to
the metabolic syndrome (described later in people with wide ranges of LDL, with or
the chapter), and/or smoking are present. without known preexisting atherosclerotic
Diet and exercise are two important com- disease.
ponents of the risk reduction arsenal. For ex- The effects of statins likely derive from
ample, the Lyon Diet Heart Study demon- several mechanisms. The combination of
strated that patients with coronary disease lowering LDL and raising HDL may reduce
who were randomized to a Mediterranean- the lipid content of atherosclerotic plaques
style diet decreased their risk of recurrent and thus favorably affect their biologic ac-
cardiac events. The diet implemented in this tivity. Other potentially beneficial actions
study included replacement of saturated include increased NO synthesis, enhanced
fats with polyunsaturated fats (particularly fibrinolytic activity, inhibition of smooth
α-linolenic acid, an ω-3 fatty acid). In vitro muscle proliferation and monocyte recruit-
evidence indicates that polyunsaturated ment, and reduction in macrophage pro-
fats may activate a transcription factor (per- duction of matrix-degrading enzymes. In
oxisome proliferator-activated receptor α vitro studies suggest that statins may also
[PPAR-α]), which increases expression of reduce inflammation by inhibiting the
the major HDL apoprotein (apo AI) and the macrophage cytokines TNF-α, IL-1, and IL-6
enzyme lipoprotein lipase, and inhibits or by augmenting PPAR-α, thereby reducing
cytokine-induced expression of leukocyte ad- endothelial expression of leukocyte adhe-
hesion molecules on endothelial cells. These sion molecules and macrophage tissue fac-
actions are potentially antiatherogenic. Phys- tor production. Clinical trials have supported
ical activity and loss of excessive weight can an anti-inflammatory action of statins, be-
also improve the lipid profile, notably by cause they reduce plasma levels of C-reac-
lowering triglycerides and raising HDL. tive protein, a marker of inflammation de-
10090-05_CH05.qxd 8/31/06 5:35 PM Page 135

Atherosclerosis 135

Included patients Excluded patients with


with history of MI or CHD known CHD
Baseline LDL
or total High Normal Normal Normal High Normal Normal
cholesterol to High to High (low HDL)
35

AFCAPS / TexCAPS
30
CHD events (%)
Reduction in
25

WOSCOPS
20

HPS
4S
15

ASCOT
LIPID
CARE
10
5
0
4S: Scandinavian Simvastatin Survival Study. Lancet 1994;344:1383–1389.
AFCAPS/TexCAPS: The Air Force/Texas Coronary Atherosclerosis Prevention Study.
JAMA 1998;279:1615–1622.
ASCOT: Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering Arm. Lancet
2003;361:1149–1158.
CARE: Cholesterol and recurrent events. N Engl J Med 1996;335:1001–1009.
HPS: Heart Protection Study. Lancet. 2004;363:757–767.
LIPID: The long-term intervention with Pravastatin in ischaemic disease. N Engl J
Med 1998;339:1349–1357.
WOSCOPS: West of Scotland Coronary Primary Prevention Study. N Engl J Med
1995;333:1301–1307.
Figure 5.11. Statins reduce cardiovascular risk. Several major clinical studies have sup-
ported the beneficial role of statin-induced lipid lowering in reducing coronary heart dis-
ease (CHD) events. Regardless of baseline serum cholesterol levels, the benefits of statins
extend to patients with an established history of coronary disease and to those with sig-
nificant risk factors without such a history. HDL, high-density lipoprotein; LDL, low-density
lipoprotein.

scribed later. It should be noted, however, Cigarette smoking could lead to athero-
that it is experimentally difficult to separate sclerotic disease in several ways, including
the LDL-lowering effect of statins from their enhanced oxidative modification of LDL,
anti-inflammatory mechanisms because of decreased circulating HDL levels, endothe-
the prominent role of oxidized LDL in initi- lial dysfunction owing to tissue hypoxia and
ating inflammatory cascades. Nonetheless, increased oxidant stress, increased platelet
accumulating clinical and experimental adhesiveness, increased expression of solu-
data do suggest that at least some measure of ble leukocyte adhesion molecules, inappro-
the statin benefit derives from mechanisms priate stimulation of the sympathetic ner-
apart from LDL lowering (so-called pleiotro- vous system by nicotine, and displacement
pic effects). of oxygen by carbon monoxide from hemo-
globin. Extrapolation from animal experi-
ments suggests that smoking not only accel-
Tobacco Smoking
erates atherogenesis but also increases the
Numerous studies have shown that cigarette propensity to thrombosis—both components
smoking increases the risk of atherosclerosis of the “vulnerable patient.”
and ischemic heart disease. Even minimal Fortunately, smoking cessation can reverse
smoking increases the risk, and the heaviest some of the adverse outcome. Epidemiologic
smokers are at the greatest danger of cardio- studies have shown that people who stop
vascular events. smoking greatly reduce their likelihood of
10090-05_CH05.qxd 8/31/06 5:35 PM Page 136

136 Chapter Five

coronary heart disease compared with those diastolic blood pressures. Regular exercise
who continue to smoke. In one study, after can also reduce resting blood pressure levels.
3 years of cessation, the risk of coronary artery Many medications can lower blood pres-
disease for former smokers became similar to sure, including diuretics, β-adrenergic recep-
subjects who never smoked. tor antagonists, drugs that interfere with the
renin-angiotensin system, calcium channel
blockers, and α-adrenergic inhibitors. The
Hypertension
benefits and limitations of these therapies
Elevated blood pressure (either systolic or di- are described in Chapters 13 and 17.
astolic) augments the risk of developing
atherosclerosis, coronary heart disease, and
Diabetes Mellitus and the
stroke (see Chapter 13). The association of
Metabolic Syndrome
elevated blood pressure with cardiovascular
disease does not appear to have a specific The global incidence of diabetes mellitus is
threshold. Rather, risk increases continu- estimated at 170 million people and pro-
ously with progressively higher pressure val- jected to grow 40% worldwide by 2030. In
ues. Particularly in older persons, systolic the United States alone, 18.2 million people
pressure predicts adverse outcomes more re- are diabetic, and projections suggest that
liably than does diastolic pressure. one of three children born in 2000 will
Hypertension may accelerate atheroscle- eventually develop the condition. With a
rosis in several ways. Animal studies have threefold to fivefold increased risk of acute
shown that elevated blood pressure injures coronary events, 80% of diabetic patients
vascular endothelium and may increase the succumb to atherosclerosis-related condi-
permeability of the vessel wall to lipopro- tions, including coronary heart disease,
teins. In addition to causing direct endothe- stroke, and peripheral artery disease. Accor-
lial damage, increased hemodynamic stress dingly, diabetes is considered an atheroscle-
can increase the number of scavenger recep- rotic risk equivalent, elevating it to the same
tors on macrophages, thus enhancing the risk category as people with a history of myo-
development of foam cells. Cyclic circum- cardial infarction.
ferential strain, increased in hypertensive The predisposition of diabetic patients to
arteries, can augment smooth muscle cell atherosclerosis may relate in part to the non-
production of proteoglycans that bind and enzymatic glycation of lipoproteins (which
retain LDL particles, promoting their accu- enhances uptake of cholesterol by scavenger
mulation in the intima and facilitating their macrophages, as described earlier) or to a pro-
oxidative modification. Angiotensin II, a thrombotic tendency and antifibrinolytic
mediator of hypertension, acts not only as a state that prevails in many patients with dia-
vasoconstrictor but also as a proinflamma- betes. Diabetic patients frequently have im-
tory cytokine. Thus, hypertension may also paired endothelial function, gauged by the
promote atherogenesis by contributing to reduced bioavailability of NO and increased
an inflammatory state. leukocyte adhesion. Tight control of serum
glucose levels in diabetic patients reduces the
risk of microvascular complications such as
Antihypertensive Therapy
retinopathy and nephropathy. At least one
Like dyslipidemias, treatment of hyperten- study has also demonstrated a reduction in
sion should start with lifestyle modifications macrovascular outcomes such as myocardial
but often requires pharmacologic interven- infarction and stroke in patients with type 1
tion. The Dietary Approaches to Stop Hyper- diabetes who followed an intense antidia-
tension studies demonstrate that a diet high betic regime. In addition, control of hyper-
in fruits and vegetables, with dairy products tension and dyslipidemia in diabetic patients
low in fat, and an overall reduced sodium convincingly reduces the risk of cardiac and
content significantly improves systolic and cerebrovascular complications.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 137

Atherosclerosis 137

The metabolic syndrome (previously known antioxidant and antiplatelet actions and im-
as the insulin resistance syndrome or syn- proves endothelium-dependent vasodilation.
drome X) is a descriptor for a clustering of risk Early observational studies suggested that
factors, including hypertension, hypertriglyc- hormone replacement therapy reduced the
eridemia, reduced HDL, cellular insulin resis- risk of coronary artery disease in postmeno-
tance (often leading to glucose intolerance), pausal women, prompting many physicians
and visceral obesity (excessive adipose tissue to prescribe such medications for cardiovas-
in the abdomen). This syndrome is associated cular prevention purposes. However, the
with a high risk for atherosclerosis in both Heart and Estrogen/progestin Replacement
diabetic and nondiabetic patients, and the Study actually demonstrated an association
National Health and Nutrition Examination between such hormone use and an early
Survey estimates that an astounding 44% of increased risk of vascular events in women
Americans have the metabolic syndrome with preexisting coronary disease. In addi-
based on current guidelines. The presence of tion, in 2003, randomized primary preven-
insulin resistance in this syndrome appears to tion studies from the Women’s Health Ini-
promote atherogenesis long before affected tiative were terminated prematurely because
persons develop overt diabetes. of the finding that estrogen-plus-progestin
treatment increased cardiovascular risk by
24% overall, with a striking 81% higher risk
Lack of Physical Activity
during the first year of therapy. Of note,
Exercise may mitigate atherogenesis in sev- these troubling outcomes did not appear
eral ways. In addition to beneficial effects on in the cohort of patients randomized to
the lipid profile and blood pressure, exercise the estrogen-only arm of the study. Further
enhances insulin sensitivity and the en- analyses will help determine if safe hormone
dothelial production of NO. Long-term replacement approaches are possible. Mean-
prospective studies of both men and women while, because currently available clinical
indicate that even modest activities, such trial data do not show that gonadal hor-
as brisk walking, can protect against cardio- mone therapy is cardioprotective and may
vascular mortality. Although no large ran- actually be harmful, such therapy should
domized primary prevention trials have ex- not be commenced at present for the sole
amined the effects of exercise on cardiac goal of reducing cardiovascular risk.
event rates, the proven benefits on the car-
diovascular risk profile should promote in-
Biomarkers
creased physical activity for anyone at risk
of developing atherosclerotic disease. Despite identification of the well-established
risk factors just described, one out of five car-
diovascular events occur in patients lacking
Estrogen Status
these attributes. In conjunction with the
Cardiovascular disease dominates over other growing knowledge about the pathogenesis
causes of mortality in women, including of atherosclerosis, several novel markers of
breast and other cancers. Before menopause, atherosclerotic risk have emerged.
women have a lower incidence of coronary
events than men. After menopause, however,
Homocysteine
men and women have similar rates. This ob-
servation suggests that estrogen (the levels of Some studies have shown a significant rela-
which decline after menopause) may have tionship between circulating levels of the
atheroprotective properties. Physiologic es- amino acid homocysteine and the incidence
trogen levels in premenopausal women raise of coronary, cerebral, and peripheral artery
HDL and lower LDL and lipoprotein (a), de- disease. The balance of current data suggest
scribed later in the chapter. Experimentally, an overall modest contribution of this factor
estrogen also exhibits potentially beneficial to cardiovascular risk in population studies.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 138

138 Chapter Five

The mechanism by which homocysteine ers of inflammation have undergone evalu-


might increase atherosclerotic risk remains ation as predictors of cardiac risk. Recall that
undetermined, but current evidence suggests the process of lipoprotein entry and modifi-
that abnormally high levels may promote cation in the vessel wall triggers the release
oxidative stress, vascular inflammation, and of cytokines, followed by leukocyte infiltra-
platelet adhesiveness. Hyperhomocysteine- tion, more cytokine release, and smooth
mia can result from genetic defects in me- muscle migration into, and proliferation
thionine metabolism or from insufficient di- within, the intima. Involved cytokines (e.g.,
etary intake of folic acid, a cofactor in the IL-6) mobilize to the liver and incite in-
methionine pathway. Although folic acid and creased production of acute-phase reactants,
other B-vitamin supplements reduce high including C-reactive protein (CRP), fibrino-
serum homocysteine levels, data thus far have gen, and serum amyloid A.
not proved that such therapy actually reduces Of these molecules, CRP has shown the
atherosclerotic disease or its complications. greatest promise as a marker of low-grade sys-
temic inflammation associated with athero-
sclerotic disease. Large studies of apparently
Lipoprotein (a)
healthy men and women indicate that those
Lipoprotein (a), referred to as Lp(a) and pro- with higher basal CRP levels have a greatly
nounced “L-P-little-a,” has also been identi- increased risk of adverse cardiovascular out-
fied as an independent risk factor for coro- comes, independent of serum cholesterol lev-
nary artery disease in some studies. Lp(a) is els. Recent prospective studies affirm high-
a special form of LDL whose major apo- sensitivity CRP as an independent predictor
lipoprotein (apo B-100) links by a disulfide of myocardial infarction, stroke, peripheral
bridge to another protein, known as apo(a). artery disease, and sudden cardiac death.
Apo(a) structurally resembles plasminogen, a In addition to its role as a marker of risk,
plasma protein important in the endogenous CRP may actually participate as a mediator of
lysis of fibrin clots (see Chapter 7). Thus, the atherogenesis. For example, CRP can acti-
detrimental effect attributed to Lp (a) may re- vate complement and thus contribute to
late to competition with normal plasminogen a sustained inflammatory state. The lipid-
activity. As with homocysteine, not all popu- lowering HMG-CoA reductase inhibitors de-
lation studies support a link between Lp(a) crease CRP levels, but current evidence does
and cardiovascular events, though people not yet prove that therapy should specifi-
with the highest Lp(a) levels do convincingly cally target CRP lowering.
appear to be at increased risk.
An person’s Lp(a) level is primarily deter-
Infection
mined by inheritance and has a skewed dis-
tribution, with higher levels among blacks. Several studies have identified infectious
Diet and exercise have little influence on agents (e.g., herpes viruses, Chlamydia pneu-
Lp(a), and drugs designed to specifically moniae) within some atherosclerotic lesions,
lower its level do not exist. However, nico- raising the question of their potential role in
tinic acid (niacin) is one agent that reduces atherogenesis. The studies have generated
Lp(a) as one of its multiple beneficial lipid substantial controversy, and definite proof
effects. Thus far, no evidence shows that of a causal role is lacking. Although it is un-
specifically targeting Lp(a) by drug therapy certain if certain infections truly play a role
improves cardiovascular outcomes. in atherogenesis, investigation of the possi-
bility is prompted by theoretical reasons. For
example, Chlamydia species produce heat
C-Reactive Protein and Other Markers
shock protein 60 (HSP-60), which activates
of Inflammation
macrophages and stimulates the production
Because the pathogenesis of atherosclerosis of matrix metalloproteinases that can impair
involves inflammation at every stage, mark- the stability of the atherosclerotic plaque’s fi-
10090-05_CH05.qxd 8/31/06 5:35 PM Page 139

Atherosclerosis 139

brous cap. Other possible atherogenic prop- 2. Early in atherogenesis, injurious and
erties of chlamydial HSP-60 and bacterial en- inflammatory stimuli activate endothe-
dotoxins include induction of foam cell for- lial and smooth muscle cells. The result-
mation, lipoprotein oxidation, and increased ing cascade of events recruits immune
procoagulant activity. Although the patho- cells to the vessel wall, fueling a per-
genic relationships remain unproven, some sistent inflammatory state believed to
researchers believe that infectious agents fur- underlie progression of the disease (see
nish an additional source of endothelial in- Fig. 5.2).
jury and inflammation that could initiate or 3. Mechanisms that contribute to athero-
exacerbate atherogenesis. To date, a number sclerosis shape the forming plaque over
of well-powered trials have not shown that decades (see Figs. 5.3 through 5.7).
antibiotic treatment directed against such Plaque diversity can be broadly catego-
putative infections reduces the risk of future rized as either stable or vulnerable (see
cardiac events in survivors of acute coronary Fig. 5.8).
syndromes. 4. Clinical expression of atherosclerosis
commonly results from narrowing of the
Management of Risk Factors vessel lumen, calcification or weakening
of the arterial wall, or plaque disruption
Despite accumulating knowledge of the with superimposed thrombus formation.
pathogenesis of atherosclerosis and its clini- Common manifestations include angina
cal sequelae, this age-old adversary to human pectoris, myocardial infarction, stroke,
life remains a major cause of death in the and peripheral arterial disease (see Fig.
modern world. Although improvements 5.10).
in cardiovascular care have reduced age- 5. Many of the risk factors that lead to ather-
adjusted mortality from this condition, it will osclerosis are modifiable (see Table 5.1),
continue to be a menace as our population providing the opportunity for physicians
ages and developing countries embrace the and other health care providers to educate
adverse dietary and activity habits of a West- patients to help prevent or reduce the pro-
ern lifestyle. Ongoing research of the biology gression of the disease.
of atherosclerosis, as well as advances in ther-
apeutic procedures and medications, will un-
doubtedly continue to further our abilities
Acknowledgments
to combat this condition. Yet we have not Contributors to the previous editions of this chapter
fully capitalized on what we already know: were Rushika Fernandopulle, MD; Gopa Bhat-
much cardiovascular risk is modifiable. Effec- tacharyya, MD; Mary Beth Gordon, MD; Joseph
Loscalzo, MD, PhD; and Peter Libby, MD.
tive control of the modifiable risk factors de-
scribed earlier remains a critical component
to tame this global scourge. It is here that the Additional Reading
patient-physician relationship and the role of AHA Science Advisory: Lyon Diet Heart Study. Bene-
medical professionals as community leaders fits of a Mediterranean-style, National Cholesterol
advocating healthy lifestyles remain of cardi- Education Program/American Heart Association
nal importance. Step I Dietary Pattern on cardiovascular disease.
Circulation 2001;103:1823–1825.
Ansell BJ, Watson KE, Fogelman AM, et al. High-
SUMMARY density lipoprotein function. J Am Coll Cardiol
2005;46:1792–1798.
1. The normal arterial wall is a three-layered Beckman JA, Creager MA, Libby P. Diabetes and ath-
erosclerosis. JAMA 2002;287:2570–2581.
structure, key aspects of which include a
Bhatt DL, Steg PG, Hirsch AT. International preva-
single endothelial layer in the intima, lence, recognition, and treatment of cardiovascu-
smooth muscle cells in the media, and lar risk factors in outpatients with atherothrom-
the outer adventitia (see Fig. 5.1). bosis. JAMA 2006;295:180–189.
10090-05_CH05.qxd 8/31/06 5:35 PM Page 140

140 Chapter Five

Executive summary of the Third Report of the Na- Libby P, Ridker PM, Maseri A. Inflammation and ath-
tional Cholesterol Education Project Expert Panel erosclerosis. Circulation 2002;105:1135–1143.
on Detection, Evaluation, and Treatment of High Nathan DM, Cleary PA, Backlund JC, et al. Intensive
Blood Cholesterol in Adults. JAMA 2001;285: diabetes treatment and cardiovascular disease in
2486–2496. patients with type 1 diabetes. N Eng J Med 2005;
Gotto AM. Evolving concepts of dyslipidemia, ath- 353:2643–2653.
erosclerosis, and cardiovascular disease. J Am Coll Ridker PM, Libby P. Risk factors for atherothrom-
Cardiol 2005;46:1219–1224. botic disease. In: Zipes D, Libby P, Bonow R,
Grundy SM, Cleeman JI, Daniels SR, et al. Diagnosis Braunwald E, ed. Braunwald’s Heart Disease: A
and management of the metabolic syndrome. An Textbook of Cardiovascular Medicine. 7th Ed.
American Heart Association/ National Heart, Lung, Philadelphia: Elsevier Saunders, 2005:939–958.
and Blood Institute scientific statement. Circula- Schönbeck U, Libby P. Inflammation, immunity,
tion 2005;112:2735–2752. and HMG-CoA reductase inhibitors: statins as
Hansson GK. Mechanisms of disease: Inflammation, antiinflammatory agents? Circulation 2004;109:
atherosclerosis, and coronary artery disease. N Engl II-18–II-26.
J Med 2005;352:1685–1695. Tsimikas S, Brilakis ES, Miller ER, et al. Oxidized
Libby P. Atherosclerosis: the new view. Scientific phospholipids, Lp(a) lipoprotein, and coronary
American 2002;286:46–55. artery disease. N Engl J Med 2005;353:46–57.
Libby P. The forgotten majority: unfinished business Yusuf S, Hawken S, Ounpuu S, et al. Effect of poten-
in cardiovascular risk reduction. J Am Coll Cardiol tially modifiable risk factors associated with myo-
2005;46:1225–1228. cardial infarction in 52 countries (the INTER-
HEART study). Lancet 2004;364:937–952.
10090-05_CH05.qxd 8/31/06 5:35 PM Page a

Chapter 5—Author Queries


1. AU: Edit fixes misplaced modifier, but does it retain intended meaning?
2. AU: Please provide a definition of IP-10 abbreviation.
3. AU: Correct to add this reference to the figure?
10090-06_CH06.qxd 8/31/06 5:22 PM Page 141

C H A P T E R

Ischemic Heart Disease


Haley Naik
Marc S. Sabatine
6
Leonard S. Lilly

DETERMINANTS OF MYOCARDIAL OXYGEN History


SUPPLY AND DEMAND Physical Examination
Myocardial Oxygen Supply Diagnostic Studies
Myocardial Oxygen Demand Natural History
PATHOPHYSIOLOGY OF ISCHEMIA TREATMENT
Fixed Vessel Narrowing Medical Treatment of an Acute Episode of Angina
Endothelial Cell Dysfunction Medical Treatment to Prevent Recurrent Ischemic
Other Causes of Myocardial Ischemia Episodes
Medical Treatment to Prevent Acute Cardiac Events
CONSEQUENCES OF ISCHEMIA
Revascularization
Ischemic Syndromes
Medical Versus Revascularization Therapy
CLINICAL FEATURES OF CHRONIC
STABLE ANGINA

In 1772, the British physician William Heber- the leading cause of death in industrialized
den reported a disorder in which patients nations.
developed an uncomfortable sensation in The clinical presentation of ischemic
the chest when walking. Labeling it angina heart disease can be highly variable and
pectoris, Heberden noted that this discom- forms a spectrum of syndromes (Table 6.1). Tab. 1
fort would disappear soon after the patient For example, ischemia may be accompa-
stood still but would recur with similar nied by the same exertional symptoms de-
activities. Although he did not know the scribed by Heberden. In other cases, it may
cause, it is likely that he was the first to occur without any clinical manifestations
describe the symptoms of ischemic heart at all, a condition termed silent ischemia.
disease, a condition of imbalance between This chapter describes the causes and con-
myocardial oxygen supply and demand sequences of chronic ischemic heart dis-
most often caused by atherosclerosis of the ease syndromes and provides a framework
coronary arteries. Ischemic heart disease for the diagnosis and treatment of affected
now afflicts millions of Americans and is patients.

141
10090-06_CH06.qxd 8/31/06 5:22 PM Page 142

142 Chapter Six

TABLE 6.1. Clinical Definitions

Syndrome Description

Ischemic heart disease Condition in which imbalance between myocardial oxygen supply and demand
results in myocardial hypoxia and accumulation of waste metabolites; most
often caused by atherosclerotic disease of the coronary arteries (coronary artery
disease)
Angina pectoris Uncomfortable sensation in the chest and neighboring anatomic structures
produced by myocardial ischemia
Stable angina Chronic pattern of transient angina pectoris, precipitated by physical activity or
emotional upset, relieved by rest within a few minutes; episodes often associ-
ated with temporary depression of the ST segment, but permanent myocardial
damage does not result
Variant angina Typical anginal discomfort, usually at rest, which develops because of coronary
artery spasm rather than an increase of myocardial oxygen demand; episodes
often associated with transient shifts of the ST segment (usually ST elevation)
Unstable angina Pattern of increased frequency and duration of angina episodes produced by less
exertion or at rest; high frequency of progression to myocardial infarction if
untreated
Silent ischemia Asymptomatic episodes of myocardial ischemia; can be detected by electro-
cardiogram and other laboratory techniques
Myocardial infarction Region of myocardial necrosis usually caused by prolonged cessation of blood sup-
ply; most often results from acute thrombus at site of coronary atherosclerotic
stenosis; may be first clinical manifestation of ischemic heart disease, or there
may be a history of angina pectoris

Angina pectoris remains the most com- Myocardial Oxygen Supply


mon manifestation of ischemic heart dis-
ease and literally means “strangling in the The supply of oxygen to the myocardium de-
chest.” Although other conditions may lead pends on the oxygen content of the blood
to similar discomfort, angina refers specifi- and the rate of coronary blood flow. The
cally to the uncomfortable sensation in the oxygen content is determined by the hemo-
chest and neighboring structures that arises globin concentration and the degree of sys-
from an imbalance between myocardial oxy- temic oxygenation. In the absence of ane-
gen supply and demand. mia or lung disease, oxygen content remains
fairly constant. In contrast, coronary blood
flow is much more dynamic, and regulation
DETERMINANTS OF MYOCARDIAL of that flow is responsible for matching the
OXYGEN SUPPLY AND DEMAND oxygen supply with metabolic requirements.
As in all blood vessels, coronary artery
In the normal heart, the oxygen require-
ments of the myocardium are continuously flow (Q) is directly proportional to the ves-
matched by the coronary arterial supply. sel’s perfusion pressure (P) and is inversely
Even during vigorous exercise, when the proportional to coronary vascular resistance
metabolic needs of the heart increase, so (R). That is,
does the delivery of oxygen to the myo- P
cardial cells so that the balance is main- Q=
R
tained. The following sections describe the AQ1
key determinants of myocardial oxygen However, unlike other arterial systems in
Fig. 1 supply and demand in a normal person (Fig. which the greatest blood flow occurs during
6.1) and how they are altered by the pres- systole, the predominance of coronary perfusion
ence of atherosclerotic coronary artery dis- takes place during diastole. The reason for this
ease (CAD). is that systolic flow is obstructed by com-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 143

Ischemic Heart Disease 143

Myocardial oxygen supply Myocardial oxygen demand

O2 content Wall stress


(P r/2h)
Coronary blood flow
• coronary perfusion pressure
• coronary vascular resistance Heart rate
• external compression
• intrinsic regulation
• local metabolites
• endothelial factors
Contractility
• neural innervation

Figure 6.1. Major determinants of myocardial oxygen supply and demand.


h, ventricular wall thickness; P, ventricular pressure; r, ventricular radius.

pression of the small coronary branches as Intrinsic Control of Coronary Tone


they course through the contracting myo-
Unlike most tissues, the heart cannot in-
cardium. Coronary flow is unimpaired in
crease oxygen extraction on demand be-
diastole because the relaxed myocardium
cause in its basal state it removes nearly as
does not compress the coronary vasculature.
much oxygen as possible from its blood sup-
Thus, in the case of the coronaries, perfu-
ply. Thus, any additional oxygen requirement
sion pressure can be approximated by the
must be met by an increase in blood flow, and
aortic diastolic pressure. Conditions that de-
autoregulation of coronary vascular resis-
crease aortic diastolic pressure (such as hy- tance is the most important mediator of this
potension or aortic valve regurgitation) de- process. Factors that participate in the regu-
crease coronary artery perfusion pressure lation of coronary vascular resistance in-
and may impair myocardial oxygen supply. clude the accumulation of local metabolites,
Coronary vascular resistance is the other endothelium-derived substances, and neural
major determinant of coronary blood flow. innervation.
In the normal artery, this resistance is dy-
namically modulated by (1) forces that ex-
ternally compress the coronary arteries and Metabolic Factors
(2) factors that alter intrinsic coronary tone. The accumulation of local metabolites signif-
icantly affects coronary vascular tone and acts
External Compression to modulate myocardial oxygen supply to
meet changing metabolic demands. During
External compression is exerted on the coro- states of hypoxemia, aerobic metabolism and
nary vessels during the cardiac cycle by con- oxidative phosphorylation in the mitochon-
traction of the surrounding myocardium. dria are inhibited. High-energy phosphates,
The degree of compression is directly related including adenosine triphosphate (ATP),
to intramyocardial pressure and is therefore cannot be regenerated. Consequently, adeno-
greatest during systole, as described in the sine diphosphate (ADP) and monophosphate
previous section. Moreover, when the myo- (AMP) accumulate and are subsequently de-
cardium contracts, the subendocardium, ad- graded to adenosine. Adenosine is a potent
jacent to the high intraventricular pressure, vasodilator and is thought to be the prime
is subjected to greater force than are the metabolic mediator of vascular tone. By bind-
outer muscle layers. This is one reason why ing to receptors on vascular smooth muscle,
the subendocardium is the region most vul- adenosine decreases calcium entry into cells,
nerable to ischemic damage. which leads to relaxation, vasodilatation,
10090-06_CH06.qxd 8/31/06 5:22 PM Page 144

144 Chapter Six

and increased coronary blood flow. Other tribute to the regulation of vascular tone.
metabolites that act locally as vasodilators Vasodilators produced by the endothelium
include lactate, acetate, hydrogen ions, and include nitric oxide (NO), prostacyclin, and
carbon dioxide. endothelium-derived hyperpolarizing factor
(EDHF). Endothelin 1 is an example of an
endothelium-derived vasoconstrictor.
Endothelial Factors
The identification and important actions
Endothelial cells of the arterial wall produce of endothelium-derived NO are described in
numerous vasoactive substances that con- Box 6.1. Box 1

Box 6.1 Endothelium-Derived Relaxing Factor, Nitric Oxide, and the Nobel Prize

Normal arterial endothelial cells synthesize potent vasodilator substances that contribute
to the modulation of vascular tone. Among the first of these to be identified were prosta-
cyclin (an arachidonic acid metabolite) and a substance termed endothelium-derived re-
laxing factor (EDRF).
EDRF was first studied in the 1970s. In experimental preparations, it was shown that
acetylcholine (ACh) has two opposite actions on blood vessels. Its direct effect on vascu-
lar smooth muscle cells is to cause vasoconstriction, but when an intact endothelial lining
overlies the smooth muscle cells, vasodilation occurs instead. Subsequent experiments
showed that ACh causes the endothelial cells to release the chemical mediator EDRF,
which quickly diffuses to the adjacent smooth muscle cells and results in their relaxation
with subsequent vasodilation of the vessel.
Subsequent research demonstrated that the mysterious EDRF is actually the nitric oxide
(NO) radical. When ACh (or another endothelial-dependent vasodilator such as serotonin
or histamine) binds to endothelial cells, intracellular free calcium increases, activating the
enzyme nitric oxide synthase (NOS). NOS catalyzes the formation of NO from the amino
acid L-arginine (see the accompanying figure). NO diffuses from the endothelium to the
adjacent vascular smooth muscle, where it activates guanylyl cyclase (G-cyclase). G-cyclase
in turn forms cyclic guanosine monophosphate (cGMP) from guanosine triphosphate. The
increased intracellular cGMP results in smooth muscle cell relaxation through mechanisms
that involve a reduction in cytosolic Ca++. The increase in cGMP is also associated with the
beneficial antimigratory effects of the smooth muscle cells.
AGONIST
(e.g., ACh, histamine, serotonin)

Nitric oxide L-Citruline


Endothelial synthase
cell L-Arginine
O2 Nitric oxide

Nitroprusside
or nitroglycerin Nitric oxide

Smooth G-cyclase
muscle cell GTP cGMP

RELAXATION
10090-06_CH06.qxd 8/31/06 5:22 PM Page 145

Ischemic Heart Disease 145

In contrast to the endothelial-dependent vasodilators, some agents cause smooth mus-


cle relaxation independent of the presence of endothelial cells. For example, the drugs
sodium nitroprusside and nitroglycerin result in vasodilation by providing an exogenous
source of NO to vascular smooth muscle cells, thereby activating G-cyclase and forming
cGMP without endothelial cell participation.
In the cardiac catheterization laboratory, the intracoronary administration of ACh in a
normal person causes vasodilation of the vessel, presumably through the release of NO.
However, in conditions of endothelial dysfunction, such as atherosclerosis, intracoronary
ACh administration results in paradoxical vasoconstriction instead. This likely reflects re-
duced production of NO by the dysfunctional endothelial cells, resulting in unopposed di-
rect vasoconstriction of the smooth muscle by ACh. Of particular interest is that the loss
of vasodilatory response to infused ACh is evident in persons with certain cardiac risk fac-
tors (e.g., elevated LDL cholesterol, hypertension, cigarette smoking) even before the phys-
ical appearance of atheromatous plaque. Thus, the impaired release of NO may be an early
and sensitive predictor for the later development of atherosclerotic lesions.
The significance of these discoveries was highlighted in 1998, when the Nobel Prize in
medicine was awarded to the scientists who discovered the critical role of NO as a cardio-
vascular signaling molecule.

Modified from Furchgott RF. The discovery of endothelium-derived relaxing factor and its importance in the
identification of nitric oxide. JAMA 1996;276:1186–1188.

NO regulates vascular tone by diffusing derived NO, it is a diffusible substance re-


into and then relaxing neighboring arterial leased by the endothelium that hyperpolar-
smooth muscle by a cyclic guanosine mono- izes (and therefore relaxes) neighboring vas-
phosphate (cGMP)–dependent mechanism. cular smooth muscle cells. EDHF is released
The production of NO by normal endothe- by many of the same factors that stimulate
lium occurs in the basal state and is addition- NO, including acetylcholine and normal
ally stimulated by many substances and con- pulsatile blood flow. In the coronary circu-
ditions. For example, its release is augmented lation, EDHF appears to be more important
when the endothelium is exposed to acetyl- in modulating relaxation in small arterioles
choline (ACh), thrombin, products of aggre- than in the large conduit arteries.
gating platelets (e.g., serotonin and ADP), or Endothelin 1 is a potent vasoconstrictor
even the shear stress of blood flow. Although produced by endothelial cells that partially
the direct effect of many of these substances counteracts the actions of the endothelial
on vascular smooth muscle is to cause vaso- vasodilators. Its expression is stimulated by
constriction, the induced release of NO from several factors, including thrombin, angio-
the normal endothelium results in vasodilata- tensin II, epinephrine, and the shear stress
Fig. 2 tion instead (Fig. 6.2). of blood flow.
Prostacyclin, an arachidonic acid metabo- Under normal circumstances, the healthy
lite, has vasodilator properties similar to those endothelium promotes vascular smooth mus-
of NO (see Fig. 6.2). It is released from en- cle relaxation (vasodilatation) through elabo-
dothelial cells in response to many stimuli, ration of substances such as NO and prosta-
including hypoxia, shear stress, acetylcho- cyclin, the influences of which predominate
line, and platelet products (e.g., serotonin). It over the endothelial vasoconstrictors (see
causes relaxation of vascular smooth muscle Fig. 6.2). However, as described later in
by a cyclic AMP–dependent mechanism. the chapter, dysfunctional endothelium
EDHF also appears to have important (e.g., in atherosclerotic vessels) secretes re-
vasodilatory properties. Like endothelial- duced amounts of vasodilators, causing
10090-06_CH06.qxd 8/31/06 5:22 PM Page 146

146 Chapter Six

Figure 6.2. Endothelium-derived vasoactive substances and their


regulators. Endothelium-derived vasodilators are shown on the left and
include nitric oxide (NO), prostacyclin, and endothelium-derived hyper-
polarizing factor (EDHF). Endothelin 1 is an endothelium-derived vaso-
constrictor. In the normal state, the vasodilator influence predominates
over that of vasoconstriction. ACh, acetylcholine.

the balance to shift toward vasoconstriction Myocardial Oxygen Demand


instead.
The three major determinants of myocardial
oxygen demand are (1) ventricular wall stress,
Neural Factors (2) heart rate, and (3) contractility (which is
The neural control of vascular resistance has also termed the inotropic state). Additionally,
both sympathetic and parasympathetic com- very small amounts of oxygen are consumed
ponents. Under normal circumstances, the in providing energy for basal cardiac metabo-
contribution of the parasympathetic ner- lism and electrical depolarization.
vous system appears minor, but sympathe- Ventricular wall stress (s) is the tangen-
tic receptors play an important role. Coro- tial force acting on the myocardial fibers,
nary vessels contain both α-adrenergic tending to pull them apart, and energy is ex-
and β2-adrenergic receptors. Stimulation of pended in opposing that force. Wall stress is
α-adrenergic receptors results in vasocon- related to intraventricular pressure (P), the
striction, whereas β2-receptors promote radius of the ventricle (r), and ventricular
vasodilatation. wall thickness (h) and is approximated by
It is the interplay among the metabolic, LaPlace’s relationship:
endothelial, and neural regulating factors P ×r
that determines the net impact on coronary s=
2h
vascular tone. For example, catecholamine
stimulation of the heart may initially cause Thus, wall stress is directly proportional to
coronary vasoconstriction via the α-adrenergic systolic ventricular pressure. Circumstances
receptor neural effect. However, catechola- that increase pressure development in the
mine stimulation also increases myocardial left ventricle, such as aortic stenosis or hy-
oxygen consumption through increased pertension, increase wall stress and myocar-
heart rate and contractility (β1-adrenergic ef- dial oxygen consumption. Conditions that
fect), and the resulting increased production decrease ventricular pressure, such as anti-
of local metabolites induces net coronary di- hypertensive therapy, reduce myocardial
latation instead. oxygen consumption.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 147

Ischemic Heart Disease 147

Because wall stress is also directly propor- the available blood supply and myocardial
tional to the radius of the left ventricle, con- metabolic demands.
ditions that augment left ventricular (LV)
filling (e.g., mitral or aortic regurgitation)
PATHOPHYSIOLOGY OF ISCHEMIA
raise wall stress and oxygen consumption.
Conversely, any physiologic or pharmaco- The traditional view has been that myocar-
logic maneuver that decreases LV filling and dial ischemia in CAD results from fixed ath-
size (e.g., nitrate therapy) reduces wall stress erosclerotic plaques that narrow the vessel’s
and myocardial oxygen consumption. lumen and limit myocardial blood supply.
Finally, wall stress is inversely proportional However, recent research has demonstrated
to ventricular wall thickness because the that the reduction of blood flow in this
force is spread over a greater muscle mass. A condition results from the combination of
hypertrophied heart has lower wall stress and fixed vessel narrowing and abnormal vascu-
oxygen consumption per gram of tissue than lar tone, contributed to by atherosclerosis-
a thinned-wall heart. Thus, when hypertro- induced endothelial cell dysfunction.
phy develops in conditions of chronic pres-
sure overload, such as aortic stenosis, it serves
Fixed Vessel Narrowing
a compensatory role in reducing oxygen con-
sumption. The hemodynamic significance of fixed ath-
The second major determinant of my- erosclerotic coronary artery stenoses relates
ocardial oxygen demand is heart rate. If the to both the fluid mechanics and the anatomy
heart rate accelerates—during physical exer- of the vascular supply.
tion, for example—the number of contrac-
tions and the amount of ATP consumed per
Fluid Mechanics
minute increases and oxygen requirements
rise. Conversely, slowing the heart rate (e.g., Poiseuille’s law states that for flow through
using a β-blocker drug) decreases ATP uti- a vessel,
lization and oxygen consumption.
ΔP πr 4
The third major determinant of oxygen Q=
demand is myocardial contractility, a mea- 8ηL
sure of the force of contraction (see Chapter in which Q is flow, ΔP is the pressure differ-
9). Circulating catecholamines, or the ad- ence between the points being measured, r is
ministration of positive inotropic drugs, the vessel radius, η is the fluid viscosity, and
directly increase the force of contraction, L is the vessel length. By analogy to Ohm’s
which augments oxygen utilization. Con- law, flow is also equal to the pressure differ-
versely, negative inotropic effectors, such as ence divided by the resistance (R) to flow:
β-adrenergic–blocking drugs, decrease myo-
cardial oxygen consumption. ΔP
Q= AQ2
In the normal state, autoregulatory me- R
chanisms adjust coronary tone to match
myocardial oxygen supply with oxygen re- By combining these two formulas, resistance
quirements. In the absence of obstructive to blood flow in a vessel can be expressed as:
coronary disease, these mechanisms main- 8ηL
tain a fairly constant rate of coronary flow, R=
πr 4
as long as the aortic perfusion pressure is ap-
proximately 60 mm Hg or greater. In the set- Thus, vascular resistance is governed, in part,
ting of advanced coronary atherosclerosis, by the geometric component L/r 4. That is,
however, the fall in perfusion pressure distal the hemodynamic significance of a stenotic
to the arterial stenosis, along with dysfunc- lesion depends on its length and, far more
tion of the endothelium of the involved seg- importantly, on the degree of vessel narrow-
ment, sets the stage for a mismatch between ing (i.e., the reduction of r) that it causes.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 148

148 Chapter Six

Anatomy tance vessels can dilate to provide adequate


blood flow. When a stenosis narrows the di-
The coronary arteries consist of large, prox-
ameter by more than approximately 70%,
imal epicardial segments and smaller, distal
resting blood flow is normal, but maximal
resistance vessels. The proximal vessels are
blood flow is reduced even with full dilata-
subject to overt atherosclerosis that results
tion of the resistance vessels. In this situa-
in stenotic plaques. The distal vessels are
tion, when oxygen demand increases (e.g.,
usually free of flow-limiting plaques and can
from the elevated heart rate and force of
adjust their vasomotor tone in response to
contraction during physical exertion), coro-
metabolic needs. These resistance vessels
nary flow reserve is inadequate, oxygen de-
serve as a reserve, increasing their diameter
mand exceeds supply, and myocardial is-
with exertion to meet increasing oxygen de-
chemia results. If the stenosis compromises
mand and dilating, even at rest, if a proxi-
the vessel lumen by more than approxi-
mal stenosis is sufficiently severe.
mately 90%, then even with maximal dilata-
The hemodynamic significance of a coro-
tion of the resistance vessels, blood flow may
nary artery narrowing depends on both the
be inadequate to meet basal requirements
degree of stenosis of the epicardial portion
and ischemia can develop at rest.
of the vessel and the amount of compen-
Although collateral channels (see Chap- AQ3
satory vasodilatation the distal resistance ves-
ter 1) may become apparent between unob-
Fig. 3 sels are able to achieve (Fig. 6.3). If a steno-
structed coronaries and sites distal to ather-
sis narrows the lumen diameter by less than
osclerotic stenoses and the flow can buffer
60%, the maximal potential blood flow
the fall in myocardial oxygen supply, it is
through the artery is not significantly al-
often not sufficient to prevent ischemia dur-
tered and, in response to exertion, the resis-
ing exertion in critically narrowed vessels.

Endothelial Cell Dysfunction


In addition to fixed vessel narrowing, the
other major contributor to reduced myocar-
dial oxygen supply in chronic CAD is en-
dothelial dysfunction. Abnormal endothelial
cell function can contribute to the patho-
physiology of ischemia in two ways: (1) by
inappropriate vasoconstriction of coronary
arteries and (2) through loss of normal an-
tithrombotic properties.

Inappropriate Vasoconstriction
In normal persons, physical activity or men-
tal stress results in measurable coronary
artery vasodilatation. This effect is thought
Figure 6.3. Resting and maximal coronary blood to be regulated by activation of the sympa-
flow are affected by the magnitude of proximal ar-
terial stenosis (percent lesion diameter). The dotted thetic nervous system, with increased blood
line indicates resting blood flow, and the solid line repre- flow and shear stress stimulating the release
sents maximal blood flow (i.e., when there is full dilata- of endothelial-derived vasodilators, such as
tion of the distal resistance vessels). Compromise of max-
imal blood flow is evident when the proximal stenosis NO. It is postulated that in normal people,
reduces the coronary lumen diameter by more than the relaxation effect of NO outweighs the di-
∼70%. Resting flow may be compromised if the stenosis rect α-adrenergic constrictor effect of cate-
exceeds ∼90%. (Modified from Gould KL, Lipscomb K. Ef-
fects of coronary stenoses on coronary flow reserve and cholamines on arterial smooth muscle, such
resistance. Am J Cardiol 1974;34:50.) that vasodilatation results. However, in pa-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 149

Ischemic Heart Disease 149

tients with dysfunctional endothelium (e.g., paired release of NO and prostacyclin allows
atherosclerosis), an impaired release of en- platelets to aggregate and to secrete their po-
dothelial vasodilators leaves the direct cate- tentially harmful procoagulants and vaso-
cholamine effect unopposed, such that rela- constrictors.
tive vasoconstriction occurs instead. The
resultant decrease in coronary blood flow
Other Causes of
contributes to ischemia. Even the vasodi-
latory effect of local metabolites (such as
Myocardial Ischemia
adenosine) is attenuated in patients with In addition to atherosclerotic CAD, other
dysfunctional endothelium, further uncou- conditions may result in an imbalance be-
pling the regulation of vascular tone from tween myocardial oxygen supply and de-
metabolic demands. mand and result in ischemia. Other causes of
In patients with risk factors for CAD, such decreased myocardial oxygen supply include
as hypercholesterolemia, diabetes mellitus, (1) decreased perfusion pressure owing to
hypertension, and cigarette smoking, im- hypotension (e.g., in a patient with hypo-
paired endothelial-dependent vasodilation volemia or septic shock) and (2) a severely
is noted even before visible atherosclerotic decreased blood oxygen content (e.g., marked
lesions have developed. This suggests that anemia or impaired oxygenation of blood
endothelial dysfunction occurs very early in by the lungs). For example, a patient with
the atherosclerotic process. massive bleeding from the gastrointestinal
Inappropriate vasoconstriction also ap- tract may develop myocardial ischemia
pears to be important in acute coronary syn- and angina pectoris, even in the absence of
dromes, such as unstable angina and myo- atherosclerotic coronary disease, because
cardial infarction (MI). As described in of reduced oxygen supply (i.e., the loss of
Chapter 7, the usual cause of acute coronary hemoglobin and hypotension).
syndromes is disruption of atherosclerotic On the other side of the balance, a pro-
plaque, with superimposed platelet aggrega- found increase in myocardial oxygen de-
tion and thrombus formation. Normally, mand can cause ischemia even in the ab-
the products of platelet aggregation in a de- sence of coronary atherosclerosis. This can
veloping clot (e.g., serotonin, ADP) result in occur, for example, with rapid tachycardias,
vasodilatation because they stimulate the profound acute hypertension, or severe aor-
endothelial release of NO. However, with tic stenosis.
dysfunctional endothelium, the direct vaso-
constricting actions of platelet products pre-
CONSEQUENCES OF ISCHEMIA
Fig. 4 dominate (Fig. 6.4), further compromising
flow through the arterial lumen. The consequences of ischemia reflect the in-
adequate myocardial oxygenation and local
accumulation of metabolic waste products.
Loss of Normal
For example, during ischemia, the myocytes
Antithrombotic Properties
convert from aerobic to anaerobic meta-
In addition to their vasodilatory actions, bolic pathways. The reduced generation of
factors released from endothelial cells (in- ATP impairs the interaction of the contrac-
cluding NO and prostacyclin) also exert an- tile proteins and results in a transient re-
tithrombotic properties by interfering with duction of both ventricular systolic contrac-
platelet aggregation (see Fig. 6.4). However, tion and diastolic relaxation (both of which
in states of endothelial cell dysfunction, re- are energy-dependent processes). The conse-
lease of these substances is reduced; there- quent elevation of LV diastolic pressure is
fore, the antithrombotic effect is attenu- transmitted (via the left atrium and pulmo-
ated. Thus, in syndromes characterized by nary veins) to the pulmonary capillaries and
thrombosis (i.e., the acute coronary syn- can precipitate pulmonary congestion and
dromes described in Chapter 7), the im- the symptom of dyspnea. In addition, meta-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 150

150 Chapter Six

Figure 6.4. The interaction between platelets and endothelial


cells. A. Normal endothelium. Aggregating platelets release thrombox-
ane (TXA2) and serotonin (5-HT), the direct vascular effects of which
cause contraction of vascular smooth muscle and vasoconstriction.
However, platelet products (e.g., adenosine diphosphate [ADP], 5-HT)
also stimulate the endothelial release of the potent vasodilators nitric
oxide (NO) and prostacyclin, such that the net effect is smooth muscle
relaxation instead. Endothelial production of NO and prostacyclin also
serves antithrombotic roles, which limit further platelet aggregation.
B. Dysfunctional endothelium demonstrates impaired release of the va-
sodilator substances, such that net smooth muscle contraction and
vasoconstriction supervene. The reduced endothelial release of NO and
prostacyclin diminishes their antiplatelet effect, such that thrombosis
proceeds unchecked.

bolic products such as lactate, serotonin, The ultimate fate of myocardium sub-
and adenosine accumulate locally. It is jected to ischemia depends on the severity
suspected that one or more of these com- and duration of the imbalance between
pounds activate peripheral pain receptors in oxygen supply and demand. It was previ-
the C7 through T4 distribution and may be ously thought that ischemic cardiac injury
the mechanism by which the discomfort of results in either irreversible myocardial
angina is produced. The accumulation of necrosis (i.e., myocardial infarction) or rapid
local metabolites and transient abnormalities and full recovery of myocyte function (e.g.,
of myocyte ion transport may also precipitate after a brief episode of typical angina). It is
dangerous arrhythmias (see Chapter 11). now known that in addition to those out-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 151

Ischemic Heart Disease 151

comes, ischemic insults can sometimes re- Ischemic Syndromes


sult in a period of prolonged contractile dys-
function without myocyte necrosis, and re- Depending on the underlying pathophysio-
covery of normal function may ultimately logic process, and the timing and severity of
follow. a myocardial ischemic insult, a spectrum of
For example, stunned myocardium refers distinct clinical syndromes may result, as il-
lustrated in Figure 6.5. Fig. 5
to tissue that, after suffering a period of se-
vere ischemia (but not necrosis), demon-
strates prolonged systolic dysfunction even Stable Angina
after the return of normal myocardial blood
flow. In this setting, the functional, bio- Chronic stable angina is generally caused by
chemical, and ultrastructural abnormalities fixed, obstructive atheromatous plaque in
following ischemia are reversible and con- one or more coronary arteries (see Fig. 6.5B).
tractile function gradually recovers. The The pattern of symptoms is usually related to
mechanism responsible for this delayed re- the degree of stenosis. As described in the ear-
covery of function involves myocyte cal- lier section on pathophysiology, when ather-
cium overload and the accumulation of oxy- osclerotic stenoses narrow a coronary artery
gen-derived free radicals during ischemia. In lumen diameter by more than approximately
general, the magnitude of stunning is pro- 70%, the reduced flow capacity may be suffi-
portional to the degree of the preceding is- cient to serve the low cardiac oxygen needs at
chemia, and this state is likely the patho- rest but is insufficient to compensate for any
physiologic response to an ischemic insult significant increase in oxygen demand (see
that just falls short of causing irreversible Fig. 6.3). During physical exertion, for exam-
necrosis. ple, activation of the sympathetic nervous
In contrast, hibernating myocardium system results in increased heart rate, blood
refers to tissue that manifests chronic ven- pressure, and contractility, all of which aug-
tricular contractile dysfunction in the face ment myocardial oxygen consumption. Dur-
of a persistently reduced blood supply, usu- ing the period that oxygen demand exceeds
ally because of multivessel CAD. In this sit- available supply, myocardial ischemia re-
uation, irreversible damage has not occurred sults, often accompanied by the chest dis-
and ventricular function can promptly im- comfort of angina pectoris. The ischemia and
prove if appropriate blood flow is restored symptoms persist until the increased de-
(e.g., by coronary angioplasty or bypass sur- mand is alleviated and oxygen balance is
gery). The importance of the difference be- restored. A pattern of chronic, predictable,
tween the two types of myocardium is sum- transient angina during exertion or emo-
Box 2 marized in Box 6.2. tional stress is termed stable angina.

Box 6.2 Distinguishing Stunned and Hibernating Myocardia

The concepts of stunned and hibernating myocardium are very important in the clinical
setting. Such regions of myocardia contract poorly when imaged (e.g., by echocardiogra-
phy or contrast angiography) and can appear indistinguishable from irreversibly infarcted
heart muscle. However, they can be differentiated from necrotic regions by special imag-
ing studies (e.g., dobutamine echocardiography, thallium-201 viability study, or positron
emission tomography; see Chapter 3). That distinction often influences the decision of
whether to undertake percutaneous angioplasty/stenting or coronary bypass procedures,
because stunned or hibernating myocardium would be expected to improve with me-
chanical revascularization, whereas truly infarcted myocardium would not.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 152

152 Chapter Six

Figure 6.5. Pathophysiologic findings in anginal syndromes. A. Normal coronary ar-


teries are widely patent, and the endothelium functions normally. B. In stable angina, ath-
erosclerotic plaque and inappropriate vasoconstriction (caused by dysfunctional endothe-
lium) reduce the vessel lumen’s size and coronary blood flow. C. In unstable angina,
disruption of the plaque triggers platelet aggregation, thrombus formation, and vasocon-
striction, all of which contribute to reduced coronary blood supply. D. In variant angina,
atherosclerotic plaques are absent; rather, ischemia is owing to intense vasospasm that re-
duces myocardial oxygen supply.

Potentially contributing to the inadequate not necessarily constant. Rather, it can vary
oxygen supply in stable angina is inappro- from moment to moment because of changes
priate coronary vasoconstriction caused, at in the superimposed coronary vascular tone.
least in part, by atherosclerosis-associated For some patients with stable angina, alter-
endothelial dysfunction. Recall that nor- ations in tone play a minimal role in the de-
mally, the high myocardial oxygen demand creased myocardial oxygen supply, and the
during exertion is balanced by an increased level of physical activity required to precipi-
supply of blood as the accumulation of local tate angina is fairly constant. These patients
metabolites induces vasodilatation. With have fixed-threshold angina. In other cases, the
endothelial cell dysfunction, however, va- degree of dynamic obstruction caused by
sodilatation is impaired and the vessels may vasoconstriction or vasospasm plays a more
paradoxically vasoconstrict instead, in re- prominent role, and such patients may have
sponse to exercise-induced catecholamine variable-threshold angina. For example, on a
stimulation of α-adrenergic receptors on the given day, a patient with variable-threshold
coronary artery smooth muscle cells. angina can experience exertion without chest
As a result, the extent of coronary artery discomfort, but on another day, the same de-
narrowing in patients with atherosclerosis is gree of myocardial oxygen demand does pro-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 153

Ischemic Heart Disease 153

duce symptoms. The difference reflects alter- pain, and such instances are referred to as
ations in vascular tone over the sites of fixed silent ischemia. These asymptomatic episodes
stenosis. Other clinical features of chronic sta- can occur in patients who on other occasions
ble angina are described in greater detail later experience typical symptomatic angina. Con-
in the chapter. versely, in some patients, silent ischemia
may be the only manifestation of CAD. It
may be difficult to diagnose silent ischemia,
Unstable Angina
but its presence can be detected by laboratory
A patient with chronic stable angina may ex- techniques such as continuous ambulatory
perience a sudden increase in the tempo and electrocardiography, or it can be elicited by
duration of ischemic episodes, occurring exercise stress testing, as described later in the
with lesser degrees of exertion and even at chapter. One study estimated that silent is-
rest. This acceleration of symptoms is known chemic episodes occur in 40% of patients
as unstable angina, which can be a precursor with stable symptomatic angina and in 2.5%
to an acute MI. Unstable angina and acute to 10% of asymptomatic middle-aged men.
MI are also known as acute coronary syn- When considering the importance of anginal
dromes and result from distinct pathophysi- discomfort as a physiologic warning signal,
ologic mechanisms, most commonly rup- the asymptomatic nature of silent ischemia
ture of an unstable atherosclerotic plaque becomes all the more alarming.
with subsequent platelet aggregation and The reason why some episodes of is-
thrombosis (see Fig. 6.5C). The acute coro- chemia are silent whereas others are symp-
nary syndromes are described in Chapter 7. tomatic has not been elucidated. The degree
of ischemia cannot fully explain the dispar-
Variant Angina ity, because even MI may present without
symptoms in some patients. However, silent
A small minority of patients manifest epi-
ischemia is particularly common among di-
sodes of focal coronary artery spasm in the
abetic patients, suggesting the possibility of
absence of overt atherosclerotic lesions,
impaired pain sensation resulting from pe-
and this syndrome is known as variant or
ripheral neuropathy.
Prinzmetal angina. In this case, intense va-
sospasm alone reduces coronary oxygen sup-
ply and results in angina (see Fig. 6.5D). The Syndrome X
mechanism by which such profound spasm
develops is not completely understood but The term syndrome X refers to patients with
may involve increased sympathetic activity typical symptoms of angina pectoris who
in combination with endothelial dysfunc- have no evidence of significant atheroscle-
tion. It is thought that many patients with rotic coronary stenoses on coronary an-
variant angina may actually have early ath- giograms. Some of these patients may show
erosclerosis manifested only by a dysfunc- definite laboratory signs of ischemia during
tional endothelium, because the response to exercise testing. The pathogenesis of is-
endothelium-dependent vasodilators (e.g., chemia in this situation may be related to
ACh and serotonin) is often abnormal. inadequate vasodilator reserve of the coro-
Variant angina often occurs at rest be- nary resistance vessels. It is thought that
cause ischemia in this case results from to the resistance vessels (which are too small
transient reduction of the coronary oxygen to be visualized by coronary angiography)
supply, rather than an increase in myocar- may not dilate appropriately during periods
dial oxygen demand. of increased myocardial oxygen demand.
Microvascular dysfunction, vasospasm, and
hypersensitive pain perception may also
Silent Ischemia
contribute to this syndrome. Patients with
Episodes of cardiac ischemia sometimes occur syndrome X have a better prognosis than
in the absence of perceptible discomfort or those with overt atherosclerotic disease.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 154

154 Chapter Six

CLINICAL FEATURES OF CHRONIC Accompanying Symptoms


STABLE ANGINA During the discomfort of an acute anginal
History attack, generalized sympathetic and para-
sympathetic stimulation may result in tachy-
The most important part of the clinical eval-
cardia, diaphoresis, and nausea. Ischemia also
uation of ischemic heart disease is the pa-
results in transient dysfunction of LV sys-
tient’s self-described history. Because chest
tolic contraction and diastolic relaxation.
pain is such a common complaint, it is im-
The resultant elevation of LV diastolic pres-
portant to focus on the characteristics that
sure is transmitted to the pulmonary vascu-
help distinguish myocardial ischemia from
lature and often causes shortness of breath
other causes of discomfort. From a diagnostic
(dyspnea) during the episode. Transient fa-
standpoint, it would be ideal to interview and
tigue and weakness are also common, partic-
examine a patient during an actual episode of
ularly in elderly patients.
angina, but most people are asymptomatic
during routine office or clinic examinations.
Therefore, a careful history probing several Precipitants
features of the discomfort should be elicited.
Angina, when not caused by pure vasospasm,
is precipitated by conditions that increase
Quality myocardial oxygen demand (e.g., increased
Angina is most often described as a “pres- heart rate, contractility, or wall stress). These
sure,” “discomfort,” “tightness,” “burning,” include physical exertion, anger, and other
or “heaviness” in the chest. It is rare that the emotional excitement. Additional factors
sensation is actually described as a “pain,” that increase myocardial oxygen demand
and often a patient will correct the physi- that can precipitate anginal discomfort in pa-
cian who refers to the anginal symptom as tients with CAD include a large meal or cold
such. Occasionally, a patient likens the sen- weather. The latter induces peripheral vaso-
sation to “an elephant sitting on my chest.” constriction, which in turn augments myo-
Anginal discomfort is neither sharp nor cardial wall stress as the left ventricle con-
stabbing, and it does not vary significantly tracts against the increased resistance.
with inspiration or movement of the chest Angina is generally relieved within min-
wall. It is a steady discomfort that lasts a few utes after the cessation of the activity that
minutes, yet rarely more than 5 to 10 min- precipitated it and even more quickly (within
utes. It always lasts more than a few seconds, 3 to 5 minutes) by sublingual nitroglycerin.
and this helps to differentiate it from sharper This response can help differentiate myocar-
and briefer musculoskeletal pains. dial ischemia from many of the other condi-
While describing angina, the patient may tions that produce chest discomfort.
place a clenched fist over his or her sternum, Patients who experience angina primarily
referred to as the Levine sign, as if defining owing to increased coronary artery tone or
the constricting discomfort by that tight grip. vasospasm often develop symptoms at rest,
independent of activities that increase myo-
cardial oxygen demand.
Location
Anginal discomfort is usually diffuse rather
Frequency
than localized to a single point. It is most
often located in the retrosternal area or in Although the level of exertion necessary to
the left precordium but may occur any- precipitate angina may remain fairly con-
where in the chest, back, arms, neck, lower stant, the frequency of episodes varies con-
face, or upper abdomen. It often radiates to siderably because patients quickly learn
the shoulders and inner aspect of the arms, which activities cause their discomfort and
especially on the left side. avoid them. It is thus important to inquire
10090-06_CH06.qxd 8/31/06 5:22 PM Page 155

Ischemic Heart Disease 155

about reductions in activities of daily living pain, spinal osteoarthritis, and cervical radi-
when taking the history. culitis. The history remains of paramount im-
portance in distinguishing myocardial is-
chemia from these disorders. In contrast to
Risk Factors
angina pectoris, gastrointestinal causes of re-
In addition to the description of chest dis- current chest pain are often precipitated by
comfort, a careful history should uncover risk certain foods and are unrelated to exertion.
factors that predispose to atherosclerosis and Musculoskeletal causes of chest discomfort
CAD, including cigarette smoking, hyper- tend to be more superficial or can be localized
cholesterolemia, hypertension, diabetes, and to a discrete spot (i.e., the patient can point to
a family history of premature coronary dis- the pain with one finger) and often vary with
ease (see Chapter 5). changes in position. Similarly, the presence of
pleuritic pain (sharp pain aggravated by res-
piratory movements) argues against angina as
Differential Diagnosis
the cause; this symptom is more likely a result
Several conditions can mimic angina pec- of pericarditis (or pulmonary conditions such
toris, including gastroesophageal reflux, eso- as pleuritis, pneumonia, or pulmonary em-
phageal spasm, biliary pain, pericarditis, and bolism). Useful differentiating features are
musculoskeletal conditions such as chest wall listed in Table 6.2. Tab. 2

TABLE 6.2. Causes of Recurrent Chest Pain

Condition Differentiating Features

Cardiac
Myocardial ischemia • Retrosternal tightness or pressure; typically radiates to neck, jaw, or left
shoulder and arm
• Lasts a few minutes (usually <10)
• Brought on by exertion, relieved by rest
• Relieved by nitroglycerin
• ECG: transient ST depressions or elevations, or flattened or inverted T waves
Pericarditis • Sharp, pleuritic pain that varies with position; friction rub on auscultation
• Can last for hours to days
• ECG: diffuse ST elevations and PR depression
Gastrointestinal
Gastroesophageal reflux • Retrosternal burning
• Precipitated by certain foods, worsened by supine position, unaffected by
exertion
• Relieved by antacids, not by nitroglycerin
Peptic ulcer disease • Epigastric ache or burning
• Occurs after meals, unaffected by exertion
• Relieved by antacids, not by nitroglycerin
Esophageal spasm • Retrosternal pain accompanied by dysphagia
• Precipitated by meals, unaffected by exertion
• May be relieved by nitroglycerin
Biliary colic • Constant, deep pain in right upper quadrant; can last hours
• Brought on by fatty foods, unaffected by exertion
• Not relieved by antacids or nitroglycerin
Musculoskeletal
Costochondral syndrome • Sternal pain worsened by chest movement
• Costochondral junctions tender to palpation
• Relieved by anti-inflammatory drugs, not by nitroglycerin
Cervical radiculitis • Constant ache or shooting pains, may be in a dermatomal distribution
• Worsened by neck motion

ECG, electrocardiogram.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 156

156 Chapter Six

Physical Examination myocardial ischemia as the cause. Because


many of these tests are costly, it is important
If it is possible to examine a patient during an
to choose the appropriate studies for each
anginal attack, several transient physical
patient.
Fig. 6 signs may be detected (Fig. 6.6). An increased
heart rate and blood pressure are common
because of the augmented sympathetic re- Electrocardiogram
sponse. Myocardial ischemia may lead to
One of the most useful tools is an electro-
papillary muscle dysfunction and therefore
cardiogram (ECG) obtained during an angi-
mitral regurgitation. Ischemia-induced re-
nal episode. Although this is easy to arrange
gional ventricular contractile abnormalities
when symptoms occur in hospitalized pa-
can sometimes be detected as an abnormal
bulging impulse on palpation of the left tients, it may not be possible to “catch”
chest. Ischemia decreases ventricular com- episodes in people seen on an outpatient
pliance, producing a stiffened ventricle and basis. During myocardial ischemia, ST seg-
therefore an S4 gallop on physical examina- ment and T wave changes often appear (Fig. Fig. 7
tion during atrial contraction (see Chapter 6.7). Acute ischemia usually results in tran-
2). However, if the patient is free of chest dis- sient horizontal or downsloping ST segment
comfort during the examination, there may depressions and T wave flattening or inver-
be no abnormal cardiac physical findings. sions. Occasionally, ST segment elevations
Physical examination should also assess are seen, suggesting more severe transmural
for signs of atherosclerotic disease in more ac- myocardial ischemia, and can be observed
cessible vascular beds. For example, carotid during the intense vasospasm of variant
bruits may indicate the presence of cere- angina. In contrast to the ECG of a patient
brovascular disease, whereas femoral artery with an acute MI, the ST deviations seen in
bruits or diminished pulses in the lower ex- patients with stable angina quickly normal-
tremities can be a clue to peripheral arterial ize with resolution of the patient’s symp-
disease (see Chapter 15). toms. In fact, ECGs obtained during periods
free of ischemia are completely normal in
approximately half of patients with stable
Diagnostic Studies
angina. In others, chronic “nondiagnostic”
Once angina is suspected, several diagnostic ST and T wave deviations may be present.
procedures may be helpful in confirming Evidence of a previous MI (e.g., pathologic

Figure 6.6. Pathophysiology of physical signs during acute myocardial ischemia.


10090-06_CH06.qxd 8/31/06 5:22 PM Page 157

Ischemic Heart Disease 157

Figure 6.7. Common transient ECG abnormalities during ischemia. Subendocardial ischemia
causes ST segment depressions and/or T wave flattening or inversions. Severe transient transmural
ischemia can result in ST segment elevations, similar to the early changes in acute myocardial infarc-
tion. When transient ischemia resolves, so do the electrocardiographic changes.

Q waves) on the ECG also points to the pres- with ischemia develop (i.e., >1 mm hori-
ence of underlying coronary disease. zontal or downsloping ST segment depres-
sions). Among patients who later undergo
diagnostic coronary angiography, standard
Stress Testing
exercise testing has a sensitivity of approxi-
Because ECGs obtained during or between mately 65% to 70% and specificity of 75% to
episodes of chest discomfort may be normal, 80% for the detection of anatomically sig-
such tracings do not rule out underlying nificant coronary artery disease.
ischemic heart disease. For this reason, pro- The stress test is considered markedly pos-
vocative exercise or pharmacologic stress itive if one or more of the following signs of
tests are valuable diagnostic and prognostic severe ischemic heart disease occur: (1) is-
aids. chemic ECG changes develop in the first
3 minutes of exercise or persist 5 minutes
after exercise has stopped; (2) the magnitude
Standard Exercise Testing
of the ST segment depressions is >2 mm;
For many patients suspected of having coro- (3) the systolic blood pressure decreases dur-
nary artery disease, a standard exercise test ing exercise (i.e., resulting from ischemia-
is performed. During this test, the patient induced impairment of contractile function);
exercises on a treadmill or a stationary bicy- (4) high-grade ventricular arrhythmias de-
cle to progressively higher workloads and is velop; or (5) the patient cannot exercise for
observed for the development of chest dis- at least 2 minutes because of cardiopul-
comfort or inordinate dyspnea. The heart monary limitations. Patients with markedly
rate and ECG are continuously monitored, positive tests are more likely to have severe
and blood pressure is checked at regular in- multivessel coronary disease.
tervals. The test is continued until angina The utility of a stress test may be affected
develops, signs of myocardial ischemia ap- by the patient’s medications. For example,
pear on the ECG, a target heart rate is β-blockers or certain calcium channel block-
achieved (85% of the maximal predicted ers may blunt the ability to achieve the tar-
heart rate, which is calculated as 220 minus get heart rate. In these situations, one must
the patient’s age), or the patient becomes consider the purpose of the stress test. If it is
too fatigued to continue. to determine whether ischemic heart disease
The test is considered positive if the pa- is present, then those medications are typi-
tient’s typical chest discomfort is repro- cally withheld for 24 to 48 hours before the
duced or if ECG abnormalities consistent test. On the other hand, if the patient has
10090-06_CH06.qxd 8/31/06 5:22 PM Page 158

158 Chapter Six

known ischemic heart disease and the pur- test results are discordant with the clinical
pose of the test is to assess the efficacy of the suspicion of coronary disease.
current medical regimen, testing should be As described in Chapter 3, positron emis-
performed while the patient takes his or her sion tomography is a specialized nuclear
usual antianginal medications. imaging technique used to assess myocar-
dial perfusion and cellular viability. It is not
universally available but is particularly ca-
Nuclear Imaging Studies
pable of distinguishing between regions of
Because the standard exercise test relies on ischemia, infarction, and hibernating myo-
ischemia-related changes on the ECG, the cardium.
test is less useful in patients with baseline
abnormalities of the ST segments (e.g., as
Exercise Echocardiography
seen in left bundle branch block or LV hy-
pertrophy). In addition, the standard exer- At many centers, exercise testing with echo-
cise stress test sometimes yields equivocal cardiographic imaging is another diagnostic
results in patients for whom the clinical sus- technique used to diagnose myocardial is-
picion of ischemic heart disease is high. In chemia in patients with baseline ST or T wave
these situations, radionuclide imaging can abnormalities or in those with equivocal
be combined with exercise testing to over- standard stress tests. In this procedure, LV
come these limitations and to increase the contractile function is assessed by echocar-
sensitivity and specificity of the study. diography at baseline and immediately after
AQ4 During myocardial perfusion imaging treadmill or bicycle exercise. The test indi-
(see Chapter 3), a radionuclide (commonly cates inducible myocardial ischemia if re-
either a technetium-99m–labeled compound gions of ventricular contractile dysfunction
or thallium-201) is injected intravenously at develop with exertion.
peak exercise, after which imaging is per-
formed. The radionuclide accumulates in
Pharmacologic Stress Tests
proportion to the degree of perfusion of vi-
able myocardial cells. Therefore, areas of For patients unable to exercise (e.g., those
poor perfusion (i.e., regions of ischemia) with hip or knee arthritis), pharmacologic
during exercise do not accumulate ra- stress testing can be performed using various
dionuclide and appear as “cold spots” on agents, including the inotrope dobutamine
the image. However, irreversibly infarcted (which increases myocardial oxygen demand
areas also do not take up the radionuclide, by stimulating the heart rate and force of
and they too will appear as cold spots. To contraction) or the vasodilators dipyridamole
differentiate between transient ischemia or adenosine. Dipyridamole blocks the cellu-
and infarcted tissue, repeat imaging is per- lar uptake and destruction of adenosine and
formed several hours later. If the cold spot thereby increases its circulating concentra-
fills in, a region of transient ischemia has tion. When adenosine binds to its vascular
been identified. If the cold spot remains receptors, coronary vasodilatation results.
unchanged, a region of irreversible infarc- As ischemic regions are already maximally
tion is likely. dilated (in compensation for the epicardial
Radionuclide exercise tests are 80% to coronary stenoses), the drug-induced vaso-
90% sensitive and 80% to 90% specific for dilatation increases flow to the myocardium
the presence of clinically significant CAD. perfused by healthy coronary arteries and
Because these techniques are expensive, thus “steals” blood away from the diseased
their use in screening for CAD should be re- segments. As a result, nuclear imaging (using
served for (1) patients in whom baseline thallium-201 or technetium-99m–labeled
ECG abnormalities preclude interpretation compounds) performed right after adeno-
of a standard exercise test or (2) improve- sine or dipyridamole administration displays
ment in test sensitivity when standard stress ischemic myocardium as regions of relatively
10090-06_CH06.qxd 8/31/06 5:22 PM Page 159

Ischemic Heart Disease 159

decreased perfusion (cold spots). Alterna- Although coronary angiography is con-


tively, pharmacologic stress testing can be sidered the “gold standard” for the diagno-
performed with echocardiography in place sis of CAD, it should be noted that it
of nuclear imaging. provides only anatomic information. The
clinical significance of lesions detected by
angiography depends not only on the de-
Coronary Angiography gree of narrowing but also on the patho-
The most direct means of identifying coro- physiologic consequences. Therefore, treat-
nary artery stenoses is by coronary angiog- ment decisions are made not only on the
raphy, in which atherosclerotic lesions are finding of such stenoses but even more so
visualized radiographically following the on their functional effects, manifested by
injection of radiopaque contrast material the patient’s symptoms, the viability of the
into the artery (Fig. 6.8; see Chapter 3). myocardium segment served by stenotic
Although generally safe, the procedure is as- vessels, and the degree of ventricular con-
sociated with a small risk of serious compli- tractile dysfunction. Furthermore, standard
cations directly related to its invasive na- arteriography does not reveal the composi-
ture. Therefore, coronary angiography is tion of coronary atherosclerotic plaque or its
typically reserved for patients whose angi- vulnerability to rupture (see Chapter 5).
Newer noninvasive techniques that visual-
nal symptoms do not respond adequately
ize coronary anatomy in great detail (e.g.,
to pharmacologic therapy, for those with
multidetector CT scanning; see Chapter 3)
an unstable presentation, or when the re-
are under study to determine their proper
sults of noninvasive testing are so abnormal
role in diagnosis and therapeutic planning
that severe CAD warranting revasculariza-
in patients with suspected CAD.
tion is likely.

Natural History
The patient with chronic angina may show
no change in a stable pattern of ischemia for
many years. In some patients, however, the
course may be punctuated at any time by
the occurrence of unstable angina, MI, or
sudden cardiac death. These complications
are often related to acute thrombosis at the
site of disrupted atherosclerotic plaque (see
Chapter 7). Why some patients, but not oth-
ers, sustain these complications remains a
subject of intense clinical and basic science
investigation and may relate to the vulnera-
bility of plaque to rupture.
Before the current era of sophisticated
pharmacotherapy, coronary angioplasty, and
surgical revascularization procedures, stud-
ies showed that the annual mortality rate
of patients with CAD corresponded to the
number of vessels containing significant
stenoses. For example, patients with ad-
vanced stenoses within a single coronary
vessel could expect an annual mortality rate
Figure 6.8. Example of coronary angiography. Injec-
tion of the right coronary artery demonstrates a stenosis of <4%. Those with two involved vessels had
in the midportion of the vessel, indicated by the arrow. an annual mortality rate of 7% to 10%, and
10090-06_CH06.qxd 8/31/06 5:22 PM Page 160

160 Chapter Six

those with advanced, three-vessel disease and demand, and (2) prevent acute coro-
showed a 10% to 12% mortality rate. If the nary syndromes and death in patients with
left main artery was significantly stenosed, chronic CAD.
the mortality rate was greatly increased
(15% to 25%). These outcomes were worse
Medical Treatment of an Acute
in corresponding patients with subnormal
Episode of Angina
ventricular contractile function.
More recent studies have confirmed that When experiencing angina, the patient
the location and extent of coronary stenoses should cease physical activity. Sublingual
are important but also that other critical nitroglycerin, an organic nitrate, is the drug
predictors of mortality include (1) the ex- of choice in this situation. Placed under the
tent of impaired LV contractile function, tongue, this medication produces a slight
(2) poor exercise capacity, and (3) the mag- burning sensation as it is absorbed through
nitude of clinical anginal symptoms. These the mucosa, and it begins to take effect in 1
predictors are taken into account when con- to 2 minutes. Nitrates relieve ischemia pri-
templating treatment decisions. marily through vascular smooth muscle re-
The mortality associated with CAD has laxation, particularly venodilatation. Venodi-
declined significantly in recent decades: the latation reduces venous return to the heart,
age-adjusted death rate has fallen by more with a subsequent decline in LV volume (a
than 50%. This is likely related to (1) athero- determinant of wall stress). The latter de-
sclerotic risk reduction through improved creases myocardial oxygen consumption,
lifestyle changes (e.g., less tobacco use, less thus helping to restore oxygen balance in
dietary fat consumption, and more exercise); the ischemic heart.
(2) improved therapeutic strategies and lon- A second action of nitrates is to dilate the
gevity following acute MI (see Chapter 7); coronary vasculature, with subsequent aug-
and (3) advances in the pharmacologic and mentation of coronary blood flow. This ef-
mechanical therapies for chronic CAD. fect may be of minimal value in patients
with angina in whom maximal coronary di-
latation has already resulted from the accu-
TREATMENT
mulation of local metabolites. However,
The goals of therapy in chronic ischemic when coronary vasospasm plays a role in the
heart disease are to decrease the frequency of development of ischemia, nitrate-induced
anginal attacks, to prevent acute coronary coronary vasodilatation may be particularly
syndromes such as myocardial infarction, beneficial.
and to prolong survival. A long-term crucial
step is to address the risk factors that led to
Medical Treatment to Prevent
the development of atherosclerotic coronary
Recurrent Ischemic Episodes
disease. Data convincingly demonstrate the
benefit of smoking cessation, cholesterol re- Pharmacologic agents are also the first line
duction, blood pressure control, and serum of defense in the prevention of anginal at-
glucose control in lowering the risk of coro- tacks. The goal of these agents is to decrease
nary disease events (see Chapter 5). Im- the cardiac workload (i.e., reduce myocar-
provements in other risk factors for CAD, in- dial oxygen demand) and to increase myo-
cluding obesity and physical inactivity, are cardial perfusion. The three classes of med-
also likely to reduce the risk of adverse out- ications commonly used are the organic
comes, although the benefits of these inter- nitrates, β-adrenergic blockers, and calcium
ventions are less well documented. channel blockers (Table 6.3). Tab. 3
The following sections describe medical Organic nitrates (e.g., nitroglycerin, iso-
and surgical strategies to (1) reduce is- sorbide dinitrate, isosorbide mononitrate), as
chemia and its symptoms by restoring the previously mentioned, relieve ischemia pri-
balance between myocardial oxygen supply marily through venodilatation (i.e., lower
10090-06_CH06.qxd 8/31/06 5:22 PM Page 161

Ischemic Heart Disease 161

TABLE 6.3. Pharmacologic Agents in the Treatment of Angina

Drug Class Mechanism of Action Adverse Effects

Organic nitrates ↓ Myocardial O2 demand • Headache


↓ Preload (venodilatation) • Hypotension
• Reflex tachycardia
↑ O2 supply
↑ Coronary perfusion
↓ Coronary vasospasm
β-blockers ↓ Myocardial O2 demand • Excessive bradycardia
↓ Contractility • ↓ LV contractile function
↓ Heart rate • Bronchoconstriction
• May worsen diabetic control
• Fatigue
Calcium channel blockers ↓ Myocardial O2 demand • Headache, flushing
(agent specific; see footnote)
↓ Preload (venodilatation) ↓ LV contraction (V, D)
↓ Wall stress (↓BP) • Marked bradycardia (V, D)
↓ Contractility (V, D) • Edema (especially N, D)
↓ Heart rate (V, D) • Constipation (especially V)
↑ O2 supply
↑ Coronary perfusion
↓ Coronary vasospasm
Aspirin ↓ Platelet aggregation • Gastrointestinal irritation or bleeding

BP, blood pressure; D, diltiazem; LV, left ventricular; N, nifedipine and other dihydropyridine calcium++ channel antagonists;
V, verapamil.

wall stress results from a smaller ventricular There is no evidence that nitrates improve
radius) and possibly through coronary va- survival or prevent infarctions in patients
sodilatation. The organic nitrates are the with chronic CAD, and they are used purely
oldest of the antianginal drugs and come in for symptomatic relief. Common side effects
several preparations. Sublingual nitroglyc- include headache, lightheadedness, and pal-
erin tablets or sprays are used in the treat- pitations induced by reflex sinus tachycar-
ment of acute attacks because of their rapid dia. The latter can be prevented by combin-
onset of action. In addition, when taken im- ing a β-blocker with the nitrate regimen.
mediately before a person engages in activi- -Blockers (see Chapter 17) exert their
ties known to provoke angina, these rapidly antianginal effect primarily by reducing myo-
acting nitrates are useful as prophylaxis against cardial oxygen demand. They are directed
anginal attacks. against β-receptors, of which there are two
Longer-acting anginal prevention can be classes: β1-adrenergic receptors are restricted
achieved through a variety of nitrate prepa- to the myocardium, whereas β2-adrenergic re-
rations, including oral tablets of isosorbide ceptors are located throughout blood vessels
dinitrate (or mononitrate) or a transdermal and the bronchial tree. The stimulation of
nitroglycerin patch, which is applied once a β1-receptors by endogenous catecholamines
day. A limitation to chronic nitrate therapy and exogenous sympathomimetic drugs in-
is the development of drug tolerance (i.e., creases heart rate and contractility. Conse-
decreased effectiveness of the drug during quently, β-adrenergic antagonists decrease
continued administration), which occurs to the force of ventricular contraction and heart
some degree in most patients. This unde- rate, thereby relieving ischemia by reducing
sired effect can be overcome by providing a myocardial oxygen demand. In addition,
nitrate-free interval for several hours each slowing the heart rate may benefit myo-
day, usually while the patient sleeps. cardial oxygen supply by augmenting the
10090-06_CH06.qxd 8/31/06 5:22 PM Page 162

162 Chapter Six

time spent in diastole, the phase when coro- ischemia by (1) decreasing oxygen demand
nary perfusion primarily occurs. (venodilatation reduces ventricular filling
In addition to suppressing angina, several and size, arterial dilatation reduces the resis-
studies have shown that β-blockers decrease tance against which the left ventricle con-
the rates of recurrent infarction and mortal- tracts, and both actions reduce wall stress);
ity following an acute MI (see Chapter 7). and (2) increasing myocardial oxygen sup-
Moreover, they have been shown to reduce ply via coronary dilatation. By the latter
the likelihood of an initial MI in patients mechanism, they are also potent agents for
with hypertension. Thus, β-blockers are the relief of coronary artery vasospasm.
first-line therapy in the treatment of CAD. Nondihydropyridine calcium channel
β-Blockers are generally well tolerated but blockers (verapamil and diltiazem) also act
have several potential side effects. For exam- as vasodilators but are not as potent in this
ple, they may precipitate bronchospasm in regard as the dihydropyridines. However,
patients with underlying asthma by antago- these agents have additional beneficial
nizing β2-receptors in the bronchial tree. Al- antianginal effects stemming from their
though β1-selective blockers are theoretically more potent cardiac depressant actions:
less likely to exacerbate bronchospasm in they reduce the force of ventricular con-
such patients, drug selectivity for the β1- traction (inotropy) and slow the heart rate.
receptor is not complete, and in general, Accordingly, verapamil and diltiazem also
all β-blockers should be avoided in patients decrease myocardial oxygen demand by
with significant obstructive airway disease. these mechanisms.
β-Blockers are also generally not used in Questions have been raised about the
patients with acutely decompensated LV dys- safety of short-acting calcium channel–block-
function because they could intensify heart ing drugs in the treatment of ischemic heart
failure symptoms by further reducing in- disease. In meta-analyses of randomized tri-
otropy. (However, as described in Chapter 9, als, these drugs have been associated with
β-blockers actually improve outcomes in pa- an increased incidence of MI and mortality.
tients with stable heart failure conditions.) β- The adverse effect may relate to the rapid
Blockers are also relatively contraindicated hemodynamic effects and blood pressure
in patients with marked bradycardia or cer- swings induced by the short-acting agents.
tain types of heart block to avoid additional Therefore, only long-acting calcium channel
impairment of electrical conduction. blockers are recommended in the treatment
β-Blockers sometimes cause fatigue and of chronic angina, generally as second-line
sexual dysfunction. They should be used with drugs if symptoms are not controlled by
caution in insulin-treated diabetic patients β-blockers and nitrates.
because they can mask tachycardia and other The three standard groups of antianginal
catecholamine-mediated responses that can drugs described in this section can be used
warn of hypoglycemia. One might also ex- alone or in combination. However, care
pect that β-blockers would decrease myocar- should be taken in combining a β-blocker
dial blood perfusion by blocking the vasodi- with a nondihydropyridine calcium chan-
lating β2-adrenergic receptors on the coronary nel blocker (verapamil or diltiazem) because
arteries. However, this effect is usually atten- the additive negative chronotropic effect
uated by autoregulation and vasodilation of can cause excessive bradycardia, and the
the coronary vessels owing to the accumula- combined negative inotropic effect could
tion of local metabolites. precipitate heart failure in patients with LV
Calcium channel blockers (see Chapter contractile dysfunction.
17) antagonize voltage-gated L-type calcium In 2006, the Food and Drug Administra-
channels, but the actions of the individual tion approved ranolazine, a fourth type of
drugs of this group vary. The dihydropy- anti-ischemic therapy. This medication has
ridines (e.g., nifedipine and amlodipine) are been shown to decrease the frequency of
potent vasodilators. They relieve myocardial anginal episodes and improve exercise ca-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 163

Ischemic Heart Disease 163

pacity in patients with chronic CAD but dif- allergy or gastric bleeding), aspirin should
fers from other anti-ischemic drugs in that it be continued indefinitely in all patients
does not affect the heart rate or blood pres- with CAD.
sure. Although its mechanism of action has Thienopyridines, including clopidogrel,
not been fully elucidated, it is believed to in- compose another group of antiplatelet
hibit the late phase of the action potential’s agents. As described in Chapter 17, they ir-
inward sodium current (INa+) in ventricular reversibly bind to the platelet ADP receptor
myocytes. That late phase tends to be abnor- P2Y12, thereby preventing platelet activation
mally enhanced in ischemic myocardium, and aggregation. When compared with as-
and the associated increased sodium influx pirin in clinical trials of patients with stable
results in higher-than-normal intracellular atherosclerotic disease, clopidogrel resulted
Ca++ (mediated by the transsarcolemmal in only a modest reduction of cardiovascu-
Na+-Ca++ exchanger; see Fig. 1.10). This cal- lar events. However, the combination of as-
cium overload is thought to result in im- pirin and clopidogrel is superior to aspirin
paired diastolic relaxation (i.e., diastolic dys- alone in reducing death and ischemic com-
function; see Chapter 9) and contractile plications in patients with acute coronary
inefficiency. Inhibition of the late INa+ by ra- syndromes and in those undergoing elective
nolazine counters these pathologic effects. percutaneous coronary stenting.
Pending further studies and experience, ra- Lipid-regulating therapy also reduces
nolazine is currently approved only for pa- cardiovascular clinical events. In particular,
tients who have not responded adequately HMG-CoA reductase inhibitors (statins; see
to the standard antianginal drugs described Chapter 17) lower MI and death rates in pa-
earlier. tients with established coronary disease and
Although useful in controlling symptoms in those at high risk of developing CAD. The
of angina, none of the antianginal drug benefits of statin therapy are believed to ex-
groups has been shown to slow or reverse tend beyond their lipid-altering effects, be-
the atherosclerotic process responsible for cause there is evidence that they decrease
the arterial lesions of chronic CAD. More- vascular inflammation and improve endo-
over, although β-blockers have demon- thelial cell dysfunction and thus may help
strated mortality benefits in patients after stabilize atherosclerotic plaques. All patients
MI, none of has been shown to improve with CAD should have their LDL cholesterol
longevity in patients with chronic stable maintained at <100 mg/dL. Moreover, some
angina and preserved LV function. trials of patients with established athero-
sclerotic disease have demonstrated that in-
tensive lipid lowering (with high-dose statin
Medical Treatment to Prevent
therapy) is superior to moderate lipid-lower-
Acute Cardiac Events
ing therapy in preventing ischemic events
Platelet aggregation and thrombosis are key and cardiovascular death. As a result, cur-
elements in the pathophysiology of acute rent national guidelines include an optional
MI and unstable angina (see Chapter 7). Anti- goal of LDL <70 mg/dL for patients with
platelet therapy reduces the risk of these known CAD, especially those at highest risk
acute coronary syndromes in patients with (e.g., following an acute coronary syndrome
chronic angina and should be a standard or those with multiple major risk factors, es-
part of the regime used to treat CAD. Aspirin pecially diabetes or continued smoking).
has antithrombotic actions through inhibi- Angiotensin-converting enzyme (ACE)
tion of platelet aggregation (and therefore inhibitors, known to be beneficial in the
reduces the release of platelet-derived pro- treatment of hypertension (see Chapter 13),
coagulants and vasoconstrictors) as well as heart failure (see Chapter 9), and following
anti-inflammatory properties that may be im- myocardial infarction (see Chapter 7), have
portant in stabilizing atheromatous plaque. been studied more recently as chronic ther-
Unless contraindications are present (e.g., apy for patients with stable CAD not com-
10090-06_CH06.qxd 8/31/06 5:22 PM Page 164

164 Chapter Six

plicated by heart failure. Some (but not all) rent symptoms within 6 months owing to
of these trials have shown reduced rates of restenosis of the dilated artery and require
death, myocardial infarction, and stroke. additional coronary interventions. In rigor-
Thus, many cardiologists recommend that ous angiographic studies, the incidence of
an ACE inhibitor be included in the medical restenosis after PTCA has been found to be
regimen of patients with chronic CAD. even greater—as high as 50%.
Fortunately, advances in percutaneous
techniques have included the development
Revascularization
of coronary stents that can be placed at the
Patients with angina that becomes asymp- time of PCI and significantly reduce the rate
tomatic during pharmacologic therapy are of restenosis. Coronary stents are slender,
usually followed by their physicians with cagelike stainless-steel support devices that in
continued emphasis on reducing cardiac their collapsed configuration can be threaded
risk factors. However, more aggressive coro- into the region of stenosis by a catheter. Once
nary revascularization is pursued if (1) the in position, the stent is expanded into its
patient’s symptoms of angina do not re- open position by inflating a high-pressure
spond adequately to antianginal drug ther- balloon in its interior (Fig. 6.9). The balloon Fig. 9
apy, (2) unacceptable side effects of medica- and attached catheter are then removed, but
tions occur, or (3) the patient is found to the stent is left permanently in place to serve
have high-risk coronary disease for which as a scaffold to maintain arterial patency. Be-
revascularization is known to improve sur- cause stents are thrombogenic, a combina-
vival (as described in the next section). The tion of oral antiplatelet agents (typically, as-
two techniques used to accomplish me-
chanical revascularization are percutaneous
coronary intervention and coronary artery
bypass graft surgery. A Artery wall Stenosis
Percutaneous coronary interventions
(PCIs) include percutaneous transluminal
coronary angioplasty (PTCA), a procedure
performed under fluoroscopy in which a
balloon-tipped catheter is inserted through a Balloon Stent in
peripheral artery (usually, femoral, brachial, catheter collapsed
or radial) and maneuvered into the stenotic configuration
segment of a coronary vessel. The balloon at
the end of the catheter is then inflated B
under high pressure to dilate the stenosis,
after which the balloon is deflated and the
catheter is removed from the body. The im-
provement in the size of the coronary lumen
increases coronary perfusion and myocar- Balloon inflation to expand stent
dial oxygen supply. Effective dilatation of
the stenosis results from compression of the C
atherosclerotic plaque and often by creating
a fracture within the lesion and stretching
the underlying media. Many types of coro-
nary stenoses are amenable to balloon dila-
tion, and complications are infrequent. The Figure 6.9. Placement of a coronary artery stent.
risk of MI during the procedure is less than A. A stent, in its original collapsed state, is advanced into
1.5%, and mortality is less than 1%. Unfor- the coronary stenosis on a balloon catheter. B. The bal-
loon is inflated to expand the stent. C. The balloon is de-
tunately, approximately one third of patients flated and the catheter is removed from the body, leaving
who undergo standard PTCA develop recur- the stent permanently in place. AQ5
10090-06_CH06.qxd 8/31/06 5:22 PM Page 165

Ischemic Heart Disease 165

pirin plus clopidogrel) is crucial after stent


implantation.
Internal
Compared with conventional PTCA, stent mammary
implantation results in larger luminal diam- artery
eters, decreased restenosis rates, and reduced
need for repeat angioplasty procedures. Al-
though restenosis resulting from vessel elas-
tic recoil is greatly diminished by standard
metal stent placement, neointimal prolifera- Saphenous
vein
tion (i.e., migration of smooth muscle cells graft
and production of extracellular matrix) re-
mains an important cause of in-stent resteno- LAD
sis and recurrent anginal symptoms.
To address the problem of restenosis after
PCI, drug-eluting stents have been devised.
These special stents are fabricated with a RCA
polymer coat that incorporates an antipro-
Figure 6.10. Coronary artery bypass surgery. A. The
liferative medication, such as sirolimus (an left internal mammary artery originates from the left sub-
immunosuppressive agent that inhibits T- clavian artery, and in this schematic, is anastomosed to
cell activation) or paclitaxel (which inter- the left anterior descending (LAD) coronary artery distal
to a tight stenosis (black segment). B. One end of a
feres with cellular microtubule function). saphenous vein graft is sutured to the proximal aorta and
The medication is released from the stent the other end to the right coronary artery (RCA) distal to
over a period of 2 to 4 weeks, and this ap- a stenotic segment.
proach has shown great effect at preventing
neointimal proliferation and clinical resteno-
sis. Prolonged courses of combination an- (IMA, a “superfluous” branch of each sub-
tiplatelet therapy (aspirin plus clopidogrel clavian artery)—that can be directly anasto-
for several months, followed by aspirin in- mosed distal to a stenotic coronary site.
definitely) are necessary for patients who re- Vein grafts have a patency rate of up to 80%
ceive drug-eluting stents, to prevent late at 12 months but are vulnerable to acceler-
thrombosis, a complication that has occurred ated atherosclerosis; 10 years after surgery,
on occasion in patients who stop taking an- more than 50% have occluded. In contrast,
tiplatelet medications. IMA grafts are more resistant to atheroscle-
Although percutaneous revascularization rosis with a patency rate of 90% at 10 years.
techniques are generally superior to stan- Therefore, IMA grafts are often used to per-
dard medical therapy for relief of angina, it fuse sites of critical flow such as the left an-
is important to note that, in the setting of terior descending artery. Recent evidence
stable coronary disease, they have not been supports the use of aggressive lipid-lowering
shown to reduce the risk of MI or death. drug therapy after CABG to improve the
Coronary artery bypass graft (CABG) long-term patency rates of bypass grafts.
surgery entails grafting portions of a pa- In recent years, less invasive surgical alter-
tient’s native blood vessels to bypass ob- natives to conventional CABG have been ex-
structed coronary arteries. Two types of sur- plored. These include “minimally invasive”
gical grafts are used (Fig. 6.10). The first operations with smaller incisions, the use of
employs native veins—typically, a section of transcutaneous ports with videoscopic ro-
the saphenous vein (a “superfluous” vessel botic assistance, and “off-pump” procedures,
removed from the leg) that is sutured from which avoid the use of cardiopulmonary by-
the base of the aorta to a coronary segment pass (heart-lung) machines. Experience with
downstream from the region of stenosis. The these techniques is growing as their advan-
second method uses arterial grafts—most tages and limitations are studied in compari-
commonly, an internal mammary artery son with standard CABG.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 166

166 Chapter Six

Medical Versus search advancements. New surgical tech-


Revascularization Therapy niques (increased use of various arterial
grafts, less invasive operations); novel ad-
Many patients with chronic, stable angina juncts to stenting (potent antithrombotic
can be successfully managed with pharma- drugs, advanced approaches to prevent
cologic therapy alone. However, if anginal in-stent restenosis); and progress in phar-
symptoms prove refractory to a good phar- macologic management (e.g., aggressive
macologic program, or if intolerable drug use of statins and antithrombotic drugs)
side effects develop, coronary angiography will likely further improve outcomes and
is usually recommended for further thera- better define the best therapeutic approaches
peutic planning. For patients whose angina for specific subsets of patients with chronic
is controlled by medications, it is standard CAD.
to perform noninvasive testing (e.g., exercise
testing, echocardiography) to identify those
with high-risk disease, because the long-term SUMMARY
prognosis for such patients can be improved 1. Cardiac ischemia results from an imbal-
by coronary revascularization.
ance between myocardial oxygen supply
Once coronary angiography is obtained,
and demand. Myocardial oxygen supply
the decision to proceed with percutaneous
is determined by the oxygen content of
intervention or bypass graft surgery depends
the blood and coronary blood flow. The
on several considerations, including those
latter is dependent on the coronary per-
Fig. 4 listed in Table 6.4. In general, patients with
fusion pressure and coronary vascular re-
persistent episodes of angina and signifi-
sistance. Key regulators of myocardial
cant stenoses in one to two coronary arter-
oxygen demand include myocardial wall
ies are good candidates for PCI, as are certain
stress, heart rate, and contractility.
lower-risk patients with three-vessel disease.
Conversely, patients who fare better over the 2. In the presence of atherosclerotic dis-
long term with CABG include those with ease, myocardial oxygen supply is com-
significant (>50%) stenosis of the left main promised. Atherosclerotic plaques cause
coronary artery and patients with multi- vascular lumen narrowing and reduce
vessel disease who also have reduced LV coronary blood flow. In addition, ather-
contractile function or diabetes. osclerosis-associated endothelial cell
Each of the previously described appro- dysfunction causes inappropriate vaso-
aches for the treatment of coronary disease constriction of the coronary resistance
is benefiting from rapidly developing re- vessels.

TABLE 6.4. Relative Advantages of Coronary Revascularization Procedures

Percutaneous Coronary Coronary Artery Bypass


Interventions (PCI) Graft Surgery (CABG)

Less invasive than CABG More effective for long-term relief of angina than PCI or pharma-
cologic therapy
Shorter hospital stay and easier Most complete revascularization
recuperation than CABG
Superior to pharmacologic therapy Improved survival in patients with
for relief of angina • >50% left main stenosis
• 3-vessel CAD, especially if LV contractile function is impaired
• 2-vessel disease with tight (>75%) LAD stenosis, especially if LV
contractile function is impaired
• Diabetes and multivessel disease

CAD, coronary artery disease; LV, left ventricle; LAD, left anterior descending coronary artery; MI, myocardial infarction.
10090-06_CH06.qxd 8/31/06 5:22 PM Page 167

Ischemic Heart Disease 167

3. Angina pectoris is the most frequent Chaitman BR. Ranolazine for the treatment of chronic
symptom of intermittent ischemia, and angina and potential use in other cardiovascular
conditions. Circulation 2006;113:2462–2472.
its diagnosis relies heavily on the patient’s
Eagle KA, Guyton RA, Davidoff R, et al. ACC/AHA
description of the discomfort. Angina may 2004 guideline update for coronary artery bypass
be accompanied by signs and symptoms of graft surgery: summary article: a report of the
adrenergic stimulation, pulmonary con- American College of Cardiology/American Heart
gestion, and transient LV systolic and di- Association Task Force on Practice Guidelines. Cir-
culation 2004;110:1168–1176.
astolic dysfunction.
Gibbons RJ, Abrams J, Chatterjee K, et al. ACC/AHA
4. Laboratory studies useful in the diagnosis 2002 Guideline Update for the Management of
of angina include the electrocardiogram Patients With Chronic Stable Angina-Summary
(ST segment and T wave abnormalities), Article: A Report of the American College of Car-
exercise (or pharmacologic) stress testing, diology/American Heart Association Task Force
on Practice Guidelines (Committee on the Man-
and coronary angiography. agement of Patients With Chronic Stable Angina).
5. Standard pharmacologic therapy for the Circulation 2003;107:149–158.
treatment of chronic angina includes Hannan EL, Racz MJ, Walford G, et al. Long-term
agents to prevent ischemia and relieve outcomes of coronary-artery bypass grafting ver-
sus stent implantation. N Engl J Med 2005;352:
symptoms (organic nitrates, β-blockers, 2174–2183.
and calcium channel antagonists, alone Henderson RA, Pocock SJ, Clayton TC, et al; Second
or in combination) as well as agents that Randomized Intervention Treatment of Angina
reduce the risk of acute coronary syn- (RITA-2) Trial Participants. Seven-year outcome in
dromes and death (e.g., aspirin, anticho- the RITA-2 trial: coronary angioplasty versus med-
ical therapy. J Am Coll Cardiol 2003;42:1161–1170.
lesterol therapy, and consideration of
Hill J, Timmis A. Exercise tolerance testing. BMJ
ACE inhibitors). Modifiable risk factors
2002;324:1084–1087.
for atherosclerosis should be corrected. Iakovou, I, Schmidt, T, Bonizzoni, E, et al. Incidence,
6. Revascularization with PCI or CABG predictors, and outcome of thrombosis after suc-
surgery may provide relief from ischemia cessful implantation of drug-eluting stents. JAMA
in patients with chronic angina who are 2005;293:2126–2130.
LaRosa JC, Grundy SM, Waters DD, et al.; Treating to
refractory to, or unable to tolerate, med-
New Targets (TNT) Investigators. Intensive lipid
ical therapy. CABG confers improved sur- lowering with atorvastatin in patients with stable
vival rates to certain high-risk groups. coronary disease. N Engl J Med 2005;352:1425–
1435.
Pearson TA, Mensah GA, Alexander RW, et al. Mark-
Acknowledgments ers of inflammation and cardiovascular disease:
application to clinical and public health practice:
Contributors to the previous editions of this chapter A statement for healthcare professionals from the
were Christopher P. Chiodo, MD; Carey Farquhar, Centers for Disease Control and Prevention and
MD; Anurag Gupta, MD; Rainu Kaushal, MD; the American Heart Association. Circulation 2003;
William Carlson, MD; Michael E. Mendelsohn, MD; 107:499–511.
Patrick T. O’Gara, MD; Marc S. Sabatine, MD; and Ridker PM, Cannon CP, Morrow DA, et al.; PROVE
Leonard S. Lilly, MD. IT-TIMI 22 Investigators. C-reactive protein levels
and outcomes after statin therapy. N Engl J Med
2005:352;20–28.
Additional Reading Serruys PW, Kutryk MJB, Ong ATL. Coronary-artery
stents. N Engl J Med 2006;354:483–495.
Abrams J. Chronic stable angina. N Eng J Med 2005; Smith SC Jr, Feldman TE, Hirshfeld JW, et al.
352:2524–2533. ACC/AHA/SCAI 2005 guideline update for percu-
Babapulle MN, Joseph L, Belisle P, et al. A hierarchi- taneous coronary intervention—summary article:
cal Bayesian meta-analysis of randomised clinical a report of the American College of Cardiology/
trials of drug-eluting stents. Lancet 2004;364: American Heart Association Task Force on Practice
583–591. Guidelines. Circulation 2006;113:156–175.
10090-06_CH06.qxd 8/31/06 5:22 PM Page a

Chapter 6—Author Queries

1. AU: Equal sign correct?


2. AU: Equation 6-1 did not include delta symbol. OK?
3. AU: Please confirm x-reference; the term is not used in text of chapter 1.
4. AU: The term “scintigraphy” is not used in ch. 3.
5. ED: Depending on how artist draws this from provided scrap, permission may be needed
10090-07_CH07.qxd 8/31/06 4:50 PM Page 168

C H A P T E R

Acute Coronary
Syndromes
Haley Naik
7
Marc S. Sabatine
Leonard S. Lilly

PATHOGENESIS OF ACUTE Acute Treatment of Unstable Angina and


CORONARY SYNDROMES Non–ST-Elevation Myocardial Infarction
Normal Hemostasis Acute Treatment of ST-Elevation Myocardial
Endogenous Antithrombotic Mechanisms Infarction
Pathogenesis of Coronary Thrombosis Adjunctive Therapies
Nonatherosclerotic Causes of Acute Coronary COMPLICATIONS
Syndromes Recurrent Ischemia
PATHOLOGY AND PATHOPHYSIOLOGY Arrhythmias
Early Changes in Infarction Myocardial Dysfunction
Late Changes in Infarction Right Ventricular Infarction
Functional Alterations Mechanical Complications
Pericarditis
CLINICAL FEATURES OF ACUTE
Thromboembolism
CORONARY SYNDROMES
Clinical Presentation RISK STRATIFICATION AND MANAGEMENT
Diagnosis of Acute Coronary Syndromes FOLLOWING MYOCARDIAL INFARCTION
TREATMENT OF ACUTE
CORONARY SYNDROMES

Acute coronary syndromes (ACS) are life- physiologic mechanism, as this chapter will
threatening conditions that can punctuate examine.
the course of patients with coronary artery The frequency of ACS is staggering: more
disease at any time. These syndromes en- than 1.6 million people are admitted to hos-
compass a continuum that ranges from an pitals in the United States each year with
unstable pattern of angina pectoris to the these conditions. Despite that daunting sta-
development of a large acute myocardial tistic, mortality associated with ACS has
infarction (MI), a condition of irreversible substantially and continuously declined in
Fig. 1 necrosis of heart muscle (Fig. 7.1). All acute recent decades as a result of major therapeu-
syndromes share a common initiating patho- tic and preventive advances. This chapter

168
10090-07_CH07.qxd 8/31/06 4:50 PM Page 169

Acute Coronary Syndromes 169

ACUTE CORONARY SYNDROMES

UNSTABLE ANGINA –
NO -EST LEV TI A NO MI -ETS LEV TI A MN O I

Figure 7.1. The continuum of acute coronary syndromes ranges from unstable angina, through
non–ST-elevation myocardial infarction (MI), to ST-elevation MI.

considers the events that lead to ACS, the thrombogenic connective tissue is exposed.
pathologic and functional changes that fol- Primary hemostasis is the first line of defense
low, and therapeutic approaches that ame- against bleeding. This process begins within
liorate the aberrant pathophysiology. seconds of vessel injury and is mediated by
circulating platelets, which adhere to colla-
gen in the vascular subendothelium and
PATHOGENESIS OF ACUTE
aggregate to form a “platelet plug.” While
CORONARY SYNDROMES
the primary hemostatic plug forms, the ex-
More than 90% of ACS result from disrup- posure of subendothelial tissue factor trig-
tion of an atherosclerotic plaque with sub- gers the plasma coagulation cascade, initiat-
sequent platelet aggregation and formation ing the process of secondary hemostasis. The
of an intracoronary thrombus. The throm- plasma coagulation proteins involved in se-
bus transforms a region of plaque narrowing condary hemostasis are sequentially activa-
to one of severe or complete occlusion, and ted at the site of injury and ultimately form
the impaired blood flow causes a marked a fibrin clot by the action of thrombin. The
imbalance between myocardial oxygen sup- resulting clot stabilizes and strengthens the
ply and demand. The form of ACS that re- platelet plug.
sults depends on the degree of coronary ob- The normal hemostatic system mini-
struction and associated ischemia (see Fig. mizes blood loss from injured vessels, but
7.1). A partially occlusive thrombus is the there is little difference between this physi-
typical cause of the closely related syn- ologic response and the pathologic process
dromes unstable angina (UA) and non–ST- of coronary thrombosis triggered by disrup-
elevation myocardial infarction (NSTEMI, tion of atherosclerotic plaques.
also referred to as non–Q-wave MI), with the
latter being distinguished from the former by
Endogenous Antithrombotic
the presence of myocardial necrosis. At the
Mechanisms
other end of the spectrum, if the thrombus
completely obstructs the coronary artery, the Normal blood vessels, including the coro-
results are more severe ischemia and a larger nary arteries, are replete with safeguards that
amount of necrosis, manifesting as an ST- prevent spontaneous thrombosis and occlu-
elevation myocardial infarction (STEMI, sion, some examples of which are shown in
also referred to as Q-wave MI). Figure 7.2. Fig. 2
The responsible thrombus in ACS appears
to be generated by interactions among the
Inactivation of Clotting Factors
atherosclerotic plaque, the coronary endo-
thelium, circulating platelets, and the dyna- Several natural inhibitors tightly regulate the
mic vasomotor tone of the vessel wall, which coagulation process to oppose clot forma-
overwhelm the natural antithrombotic me- tion and maintain blood fluidity. The most
chanisms described in the next section. important of these are antithrombin III,
proteins C and S, and tissue factor pathway
inhibitor.
Normal Hemostasis
Antithrombin III is a plasma protein that
When a normal blood vessel is injured, the irreversibly binds to thrombin and other
endothelial surface becomes disrupted and clotting factors, inactivating them and facili-
10090-07_CH07.qxd 8/31/06 4:50 PM Page 170

170 Chapter Seven

Tissue 4 tPA
factor
Plasminogen

VII Fibrin
clot
Xa 3
Plasmin
TFPI

Protein S Fibrin
Inactivated
split
Va,VIIIa
products
factors
Protein C*
TM 5
Thrombin 2
Prostacyclin
Protein C and
NO

Irreversible
Thrombin thrombin
inhibition
Antithrombin III
1
Heparan
sulfate

Figure 7.2. Endogenous protective mechanisms against thrombosis and


vessel occlusion. (1) Inactivation of thrombin by antithrombin III (AT), the effec-
tiveness of which is enhanced by binding of AT to heparan sulfate. (2) Inactivation
of clotting factors Va and VIIIa by activated protein C (protein C*), an action that
is enhanced by protein S. Protein C is activated by the thrombomodulin (TM)-
thrombin complex. (3) Inactivation of factor VII/tissue factor complex by tissue fac-
tor pathway inhibitor (TFPI). (4) Lysis of fibrin clots by tissue plasminogen activator
(tPA). (5) Inhibition of platelet activation by prostacyclin and NO.

tating their clearance from the circulation the thrombin-thrombomodulin complex ac-
(see mechanism 1 in Fig. 7.2). The effective- tivates protein C. Activated protein C de-
ness of antithrombin III is increased a thou- grades factors Va and VIIIa (see mechanism
sandfold by binding to heparan sulfate, a 2 in Fig. 7.2), thereby inhibiting coagula-
heparin-like molecule normally present on tion. The presence of protein S in the circu-
the luminal surface of endothelial cells. lation enhances the inhibitory function of
Protein C or protein S and thrombo- protein C.
modulin form a natural anticoagulant sys- Tissue factor pathway inhibitor (TFPI)
tem that inactivates the “acceleration” fac- is a plasma serine protease inhibitor that
tors of the coagulation pathway (i.e., factors is activated by coagulation factor Xa. The
Va and VIIIa). Protein C is synthesized in combined factor Xa-TFPI binds to and in-
the liver and circulates in an inactive form. activates the complex of tissue factor with
Thrombomodulin is a thrombin-binding factor VIIa that normally triggers the extrin-
receptor normally present on endothelial sic coagulation pathway (see mechanism 3
cells. Thrombin bound to thrombomod- in Fig. 7.2). Thus, TFPI serves as a negative
ulin cannot convert fibrinogen to fibrin (the feedback inhibitor that interferes with coag-
final reaction in clot formation). Instead, ulation.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 171

Acute Coronary Syndromes 171

Lysis of Fibrin Clots tors) and by reducing shear stress (an indu-
cer of platelet activation).
Tissue plasminogen activator (tPA) is a
Nitric oxide (NO) is similarly secreted by
protein secreted by endothelial cells in re-
endothelial cells, as described in Chapter 6.
sponse to many triggers of clot formation. It acts locally to inhibit platelet activation,
tPA cleaves the protein plasminogen to form and it too serves as a potent vasodilator.
active plasmin, which in turn enzymatically
degrades fibrin clots (see mechanism 4 in
Fig. 7.2). When tPA binds to fibrin in a form- Pathogenesis of
ing clot, its ability to convert plasminogen Coronary Thrombosis
to plasmin is greatly enhanced. Normally, the mechanisms shown in Figure
7.2 serve to prevent spontaneous intravas-
Endogenous Platelet Inhibition cular thrombus formation. However, abnor-
and Vasodilatation malities associated with atherosclerotic le-
sions may overwhelm these defenses and
Prostacyclin is synthesized and secreted by result in coronary thrombosis and vessel oc-
endothelial cells (see mechanism 5 in Fig. clusion (Fig. 7.3). Atherosclerosis contributes Fig. 3
7.2), as described in Chapter 6. Prostacyclin to thrombus formation by (1) plaque rup-
increases platelet levels of cyclic AMP and ture, which exposes the circulating blood
thereby strongly inhibits platelet activation elements to thrombogenic substances, and
and aggregation. It also indirectly inhibits (2) endothelial dysfunction with the loss of
coagulation via its potent vasodilating pro- normal protective antithrombotic and vaso-
perties. Vasodilatation helps guard against dilatory properties.
thrombosis by augmenting blood flow (which Atherosclerotic plaque rupture is consid-
minimizes contact between procoagulant fac- ered the major trigger of coronary thrombo-

Atherosclerosis

Figure 7.3. Mechanisms of coronary thrombus formation. Factors that contribute to this process include
plaque disruption (e.g., rupture), activation of platelets and the clotting cascade, and inappropriate vaso-
constriction and loss of normal antithrombotic defenses because of dysfunctional endothelium.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 172

172 Chapter Seven

sis. The underlying causes of plaque disrup- gen), activators of the coagulation cascade
tion are (1) chemical factors that destabi- (e.g., factor Va), and vasoconstrictors (e.g.,
lize atherosclerotic lesions and (2) physical thromboxane and serotonin). The devel-
stresses to which the lesions are subjected. oping intracoronary thrombus, intraplaque
As described in Chapter 5, atherosclerotic hemorrhage, and vasoconstriction all con-
plaques consist of a lipid-laden core sur- tribute to narrowing the vessel lumen,
rounded by a fibrous external cap. Sub- creating turbulent blood flow that contri-
stances released from inflammatory cells butes to shear stress and further platelet ac-
within the plaque can compromise the in- tivation.
tegrity of the fibrous cap. For example, T Dysfunctional endothelium, which is
lymphocytes elaborate γ-interferon, which apparent even in mild atherosclerotic coro-
inhibits collagen synthesis by smooth mus- nary disease, also increases the likelihood of
cle cells and thereby interferes with the thrombus formation. In the setting of en-
usual strength of the cap. Additionally, cells dothelial dysfunction, reduced amounts of
within atherosclerotic lesions produce en- vasodilators (e.g., NO and prostacyclin) are
zymes (e.g., metalloproteinases) that de- released and inhibition of platelet aggrega-
grade the interstitial matrix, further com- tion by these factors is impaired, resulting in
promising plaque stability. A weakened or the loss of a key defense against thrombosis.
thin-capped plaque is subject to rupture, Not only is dysfunctional endothelium
particularly in its “shoulder” region (the less equipped to prevent platelet aggrega-
border with the normal arterial wall that is tion, but also it is less able to counteract the
subjected to high circumferential stress) ei- vasoconstricting products of platelets. Dur-
ther spontaneously or by physical forces, ing thrombus formation, vasoconstriction
such as intraluminal blood pressure and tor- is promoted both by platelet products
sion from the beating myocardium. (thromboxane and serotonin) and by throm-
ACS sometimes occur in the setting of bin within the developing clot. The normal
certain triggers, such as strenuous physical platelet-associated vascular response is va-
activity or emotional upset. The activation sodilatation, because platelet products stim-
of the sympathetic nervous system in these ulate endothelial NO and prostacyclin re-
situations increases the blood pressure, lease, the influences of which predominate
heart rate, and force of ventricular contrac- over direct platelet-derived vasoconstrictors
tion—actions that may stress the athero- (see Fig. 6.4). However, reduced secretion of
sclerotic lesion, thereby causing the plaque endothelial vasodilators in atherosclerosis al-
to fissure or rupture. In addition, MI is most lows vasoconstriction to proceed unchecked.
likely to occur in the early morning hours. Similarly, thrombin in a forming clot is a po-
This observation may relate to the tendency tent vascular smooth muscle constrictor in
of key physiologic stressors (such as systolic the setting of dysfunctional endothelium.
blood pressure, blood viscosity, and plasma Vasoconstriction causes torsional stresses that
epinephrine levels) to be most elevated at can contribute to plaque rupture or can tran-
that time of day, and these factors subject siently occlude the stenotic vessel through
vulnerable plaques to rupture. heightened arterial tone. The reduction in
Following plaque rupture, thrombus for- coronary blood flow caused by vasocon-
mation is provoked via the mechanisms striction also reduces the washout of coagu-
shown in Figure 7.3. The exposure of tissue lation proteins, thereby enhancing throm-
factor from the atheromatous core triggers bogenicity.
the coagulation pathway, while the expo-
sure of subendothelial collagen activates
Significance of Coronary Thrombosis
platelets. Activated platelets release the
contents of their granules, which include The formation of an intracoronary throm-
facilitators of platelet aggregation (e.g., bus results in one of several potential out-
adenosine diphosphate [ADP] and fibrino- comes (Fig. 7.4). For example, plaque rup- Fig. 4
10090-07_CH07.qxd 8/31/06 4:50 PM Page 173

Acute Coronary Syndromes 173

Coronary thrombus

Small thrombus Partially occlusive Occlusive


(non–flow limiting) thrombus (Transient thrombus
ischemia) (Prolonged
ischemia)

No ECG ST segment
changes depression and/or ST elevation
T wave inversion (Q waves later)

Healing and
plaque enlargement

Unstable angina Non–ST-segment ST-segment


elevation MI elevation MI
Figure 7.4. Consequences of coronary thrombosis. A small thrombus formed on superficial plaque rup-
ture may not result in symptoms or electrocardiogram (ECG) abnormalities, but healing and fibrous organi-
zation may incorporate the thrombus into the plaque, causing the atherosclerotic lesion to enlarge. A par-
tially occlusive thrombus (with or without superimposed vasospasm) narrows the arterial lumen, restricts blood
flow, and can cause unstable angina or a non–ST-elevation MI, either of which may result in ST segment de-
pression and/or T wave inversion on the ECG. A totally occlusive thrombus with prolonged ischemia is the
most common cause of ST-elevation MI, in which the ECG initially shows ST segment elevation, followed by
Q wave development. An occlusive thrombus that recanalizes, or one that develops in a region served by ad-
equate collateral blood flow, may result in less prolonged ischemia and a non–ST-elevation MI instead. Mark-
ers of myocardial necrosis include cardiac-specific troponins and creatine kinase MB isoenzyme.

ture is sometimes superficial, minor, and self- cause of spontaneous recanalization or by


limited, such that only a small, nonocclusive relief of superimposed vasospasm), the se-
thrombus forms. In this case, the thrombus verity and duration of ischemia will be less,
may simply become incorporated into the and a smaller non–ST-elevation MI or UA
growing atheromatous lesion through fibro- are the more likely outcomes. The distinc-
tic organization, or it may be lysed by natural tion between a non–ST-elevation MI and
fibrinolytic mechanisms. Recurrent asymp- UA is based on the degree of the ischemia
tomatic plaque ruptures of this type may and whether the event is severe enough to
cause gradual progressive enlargement of the cause necrosis, indicated by the presence
coronary stenosis. of certain serum biomarkers (see Fig. 7.4).
However, deeper plaque rupture may re- Nonetheless, UA and NSTEMI act quite alike,
sult in greater exposure of subendothelial and the management of these entities is
collagen and tissue factor, with formation of similar.
a larger thrombus that more substantially Occasionally, a non–ST-elevation infarct
occludes the vessel’s lumen. Such obstruc- may result from total coronary occlusion. In
tion may cause prolonged severe ischemia this case, it is likely that a substantial col-
and the development of an acute coronary lateral blood supply (see Chapter 1) limits
syndrome. If the intraluminal thrombus at the extent of necrosis, such that a larger ST-
the site of plaque disruption totally occludes elevation MI is prevented.
the vessel, blood flow beyond the obstruc-
tion will cease, prolonged ischemia will
Nonatherosclerotic Causes of
occur, and an MI (usually an ST-elevation
Acute Coronary Syndromes
MI) will result. Conversely, if the thrombus
partially occludes the vessel (or if it totally Rarely, mechanisms other than acute throm-
occludes the vessel but only transiently be- bus formation can precipitate an acute coro-
10090-07_CH07.qxd 8/31/06 4:50 PM Page 174

174 Chapter Seven

TABLE 7.1. Causes of Acute Coronary Syndromes

• Atherosclerosis with superimposed thrombus


• Vasculitic syndromes (see Chapter 15)
• Coronary emboli (e.g., from endocarditis, artificial valves)
• Congenital anomalies of the coronary arteries
• Coronary trauma or aneurysm
• Severe coronary artery spasm (primary or cocaine-induced)
• Increased blood viscosity (e.g., polycythemia vera, thrombocytosis)
• Markedly increased myocardial oxygen demand (e.g., severe aortic stenosis)

Tab. 1 nary syndrome (Table 7.1). These should be span the entire thickness of the myocardium
suspected when an ACS occurs in a young and result from total, prolonged occlusion
patient or a person with no coronary risk of an epicardial coronary artery. Conversely,
factors. For example, coronary emboli from subendocardial infarcts exclusively involve
mechanical or infected cardiac valves can the innermost layers of the myocardium.
lodge in the coronary circulation, or inflam- The subendocardium is particularly suscep-
mation from acute vasculitis can initiate coro- tible to ischemia because it is the zone
nary occlusion. Occasionally, intense tran- subjected to the highest pressure from the
sient coronary spasm can sufficiently reduce ventricular chamber, has few collateral con-
myocardial blood supply to result in UA or nections that supply it, and is perfused by
infarction. vessels that must pass through layers of con-
Another cause of ACS is cocaine abuse. tracting myocardium.
Cocaine increases sympathetic tone by Infarction represents the culmination of
blocking the presynaptic reuptake of norep- a disastrous cascade of events, initiated by
inephrine and by enhancing the release of ischemia, that progresses from a potentially
adrenal catecholamines, which can lead to reversible phase to irreversible cell death.
vasospasm and therefore decreased myo- Myocardium that is supplied directly by an
cardial oxygen supply. An acute coronary occluded vessel may die quickly. The adja-
syndrome may ensue because of increased cent tissue may not necrose immediately be-
myocardial oxygen demand resulting from cause it may be sufficiently perfused by
cocaine-induced sympathetic myocardial nearby patent vessels. However, the neigh-
stimulation (increased heart rate and blood boring cells may become increasingly is-
pressure) in the face of the decreased oxygen chemic over time, as demand for oxygen
supply. continues in the face of decreased oxygen
supply. Thus, the region of infarction may
subsequently extend outward. The amount
PATHOLOGY AND
of tissue that ultimately succumbs to infarc-
PATHOPHYSIOLOGY
tion therefore relates to (1) the mass of my-
MI (either STEMI or NSTEMI) results when ocardium perfused by the occluded vessel,
myocardial ischemia is sufficiently severe to (2) the magnitude and duration of impaired
cause myocyte necrosis. Although by defin- coronary blood flow, (3) the oxygen demand
ition UA does not result in necrosis, MI may of the affected region, (4) the adequacy of
subsequently ensue if the underlying patho- collateral vessels that provide blood flow
physiology of the unstable pattern of angina from neighboring nonoccluded coronary ar-
is not promptly corrected. teries, and (5) the degree of tissue response
In addition to their clinical classifications, that modifies the ischemic process.
infarctions can be described pathologically The pathophysiologic alterations that
by the extent of necrosis they produce with- transpire during MI occur in two stages:
in the myocardial wall. Transmural infarcts early changes at the time of acute infarction
10090-07_CH07.qxd 8/31/06 4:50 PM Page 175

Acute Coronary Syndromes 175

and late changes during myocardial healing contributes to alterations in the transmem-
and remodeling. brane electrical potential, predisposing the
myocardium to lethal arrhythmias. Intracel-
lular calcium accumulates in the damaged
Early Changes in Infarction
myocytes and is thought to contribute to
Early changes include the histologic evolution the final common pathway of cell destruc-
of the infarct and the functional impact of tion through the activation of degradative
oxygen deprivation on myocardial contractil- lipases and proteases.
ity. These changes culminate in coagulative Collectively, these metabolic changes de-
necrosis of the myocardium in 2 to 4 days. crease myocardial function as early as 2 min-
utes following occlusive thrombosis. Without
intervention, irreversible cell injury ensues
Cellular Changes
in 20 minutes and is marked by the deve-
As oxygen levels fall in the myocardium lopment of membrane defects. Proteolytic
supplied by an abruptly occluded coronary enzymes leak across the myocyte’s altered
vessel, there is a rapid shift from aerobic to membrane, damaging adjacent myocardium,
Fig. 5 anaerobic metabolism (Fig. 7.5). Because mi- and the release of certain macromolecules
tochondria can no longer oxidize fats or pro- into the circulation serves as a clinical marker
ducts of glycolysis, high-energy phosphate of acute infarction.
production drops dramatically and anaero- Edema of the myocardium develops with-
bic glycolysis leads to the accumulation of in 4 to 12 hours, as vascular permeability in-
lactic acid. This results in a lowered pH. creases and interstitial oncotic pressure rises
Furthermore, the paucity of high-energy (because of the leak of intracellular pro-
phosphates such as adenosine triphosphate teins). The earliest histologic changes of
(ATP) interferes with the transmembrane irreversible injury are wavy myofibers,
NaK-ATPase, with resultant elevation in in- which appear as intercellular edema sepa-
tracellular Na and extracellular K. Rising Na rates the myocardial cells that are tugged
contributes to cellular edema. Membrane about by the surrounding, functional myo-
leak and rising extracellular K concentration cardium (Fig. 7.6). Contraction bands can Fig. 6

Myocardial hypoxia

ATP

Impaired Na-+, K+-ATPase Anaerobic metabolism

Extracellular Intracellular Intracellular Intracellular


K+ Na+ Ca++ H+

Altered Intracellular Chromatin clumping


membrane edema ATP Protein denaturation
potential Proteases
Lipases

Arrhythmias Cell death


Figure 7.5. Mechanisms of cell death in myocardial infarction. Acute ischemia rapidly de-
pletes the intracellular supply of adenosine triphosphate (ATP) as aerobic metabolism fails. Subse-
quent intracellular acidosis and impairment of ATP-dependent processes culminate in intracellular
calcium accumulation, edema, and cell death.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 176

176 Chapter Seven

Figure 7.6. Pathologic evolution in acute myocardial infarction. A. Early wavy myofibers and edema; viable
myocardium is at lower left. B. Coagulation necrosis and dense infiltration of neutrophils. C. Necrotic myocytes largely
removed by phagocytes (7 to 10 days); viable myocardium at lower left. D. Granulation tissue with early collagen de-
position; new capillaries have formed (arrows). E. Late fibrotic scarring. (Reprinted with permission from Schoen FJ.
Interventional and Surgical Cardiovascular Pathology—Clinical Correlations and Basic Principles. Philadelphia: Saun-
ders, 1989:67.)
10090-07_CH07.qxd 8/31/06 4:50 PM Page 177

Acute Coronary Syndromes 177

often be seen near the borders of the infarct: replaced by fibrous tissue. Macrophages in-
sarcomeres are contracted and consolidated vade the inflamed myocardium shortly after
and appear as bright eosinophilic belts. neutrophil infiltration and remove necrotic
An acute inflammatory response, with in- tissue. This period of tissue resorption is
filtration of neutrophils, begins after approx- termed yellow softening because connective
imately 4 hours and incites further tissue tissue elements are destroyed and removed
damage. Within 18 to 24 hours, coagulation along with dead myocardial cells. The phago-
necrosis is evident with pyknotic nuclei and cytic clearing, combined with thinning and
bland eosinophilic cytoplasm, seen by light dilation of the infarcted zone, results in
microscopy. These early changes are de- structural weakness of the ventricular wall
monstrated in Figure 7.6 and summarized in and the possibility of myocardial wall rup-
Tab. 2 Table 7.2. ture at this stage. Fibrosis subsequently en-
sues, and scarring is complete by 7 weeks
Gross Changes after infarction (see Fig. 7.6).

Gross morphologic changes do not appear


until 18 to 24 hours after coronary occlusion, Functional Alterations
although certain staining techniques (e.g.,
Impaired Contractility and Compliance
tetrazolium) permit the pathologist to iden-
tify regions of infarction earlier. Most often, The destruction of functional myocardial
ischemia and infarction begin in the suben- cells in infarction quickly leads to impaired
docardium and then extend laterally and ventricular contraction (systolic dysfunc-
outward toward the epicardium. tion). Cardiac output is further compro-
mised because synchronous contraction of
myocytes is lost. Specific terms are used to
Late Changes in Infarction
describe the types of wall motion abnormal-
Late pathologic changes in the course of ities that can result. A localized region of re-
acute MI (see Table 7.2) include (1) the clear- duced contraction is termed hypokinetic; a
ing of necrotic myocardium and (2) the de- segment that does not contract at all is called
position of collagen to form scar tissue. akinetic; and a dyskinetic region is one that
Irreversibly injured myocytes do not re- bulges outward during contraction of the re-
generate; rather, the cells are removed and maining functional portions of the ventricle.

TABLE 7.2. Pathologic Time Line in Transmural Infarction

Time Event

Early changes
1–2 min ATP levels fall; cessation of contractility
10 min 50% depletion of ATP; cellular edema, decreased membrane potential and suscepti-
bility to arrhythmias
20–24 min Irreversible cell injury
1–3 hours Wavy myofibers
4–12 hours Hemorrhage, edema, PMN infiltration begins
18–24 hours Coagulation necrosis (pyknotic nuclei with eosinophilic cytoplasm), edema
2–4 days Total coagulation necrosis (no nuclei or striations, rimmed by hyperemic tissue);
monocytes appear; PMN infiltration peaks
Late changes
5–7 days Yellow-softening from resorption of dead tissue by macrophages
7 days Ventricular remodeling
7 weeks Fibrosis and scarring complete

ATP, adenosine triphosphate; PMN, polymorphonuclear leukocyte.


10090-07_CH07.qxd 8/31/06 4:50 PM Page 178

178 Chapter Seven

During ACS, the left ventricle is also ad- In the early post-MI period, infarct ex-
versely compromised by diastolic dysfunc- pansion may occur, with affected ventricular
tion. Ischemia and/or infarction impair segment enlarging without additional my-
diastolic relaxation (an energy-dependent ocyte necrosis. Infarct expansion represents
process; see Chapter 1), which reduces ven- thinning and dilatation of the necrotic zone
tricular compliance and contributes to ele- of tissue, likely because of “slippage” be-
vated ventricular filling pressures. tween the muscle fibers, resulting in a de-
creased volume of myocytes in the region.
Infarct expansion can be detrimental because
Stunned Myocardium
it increases ventricular size, which (1) aug-
Sometimes transient myocardial ischemia ments wall stress, (2) impairs systolic con-
can result in a very prolonged, but gradu- tractile function, and (3) increases the likeli-
ally reversible, period of contractile dys- hood of aneurysm formation.
function. For example, as described in In addition to early expansion of the in-
Chapter 6, stunned myocardium is tissue farcted territory, remodeling of the ventricle
that demonstrates prolonged systolic dys- may also involve dilatation of the overworked
function after a discrete episode of severe noninfarcted segments, which are subjected to
ischemia, despite restoration of adequate increased wall stress. This dilatation begins in
blood flow, and gradually regains contrac- the early postinfarct period and continues
tile force days to weeks later. Stunning may over the ensuing weeks and months. Initially,
play an important role in patients with UA chamber dilatation serves a compensatory
or in myocardium adjacent to the region role because it increases cardiac output via the
of an acute infarction. In both instances, Frank-Starling mechanism (see Chapter 9),
prolonged contractile dysfunction of af- but progressive enlargement may ultimately
fected ventricular segments may be evi- lead to heart failure and predisposes to ven-
dent after the event, simulating infarcted tricular arrhythmias.
tissue. However, if the tissue is simply Adverse ventricular remodeling can be
stunned rather than necrotic, its function beneficially modified by certain interven-
will recover over time. tions. At the time of infarction, for example,
reperfusion therapies limit infarct size and
therefore decrease the likelihood of infarct
Ischemic Preconditioning
expansion. In addition, drugs that interfere
Brief ischemic insults to a region of myo- with the renin-angiotensin system have
cardium may render that tissue more resis- been shown to attenuate progressive remod-
tant to subsequent episodes, a phenomenon eling and to reduce short- and long-term
termed ischemic preconditioning. The clin- mortality after infarction (as discussed later
ical relevance is that patients who sustain an in the chapter).
MI in the context of recent angina experi-
ence less morbidity and mortality than those
CLINICAL FEATURES OF ACUTE
without preceding ischemic episodes. The
CORONARY SYNDROMES
mechanism of this phenomenon is unknown
but appears to involve ischemia-related acti- Because ACS represent disorders along a
vation of adenosine receptors. continuum, their clinical features overlap.
In general, the severity of symptoms and as-
sociated laboratory findings progress from
Ventricular Remodeling
UA on one side of the continuum, through
Following an MI, changes occur in the geom- NSTEMI, to STEMI on the other end of
etry of both the infarcted and noninfarcted the continuum (see Fig. 7.1). Distinguishing
ventricular muscle. Such alterations in cham- among these syndromes is based on the
ber size and wall thickness affect long-term clinical presentation, electrocardiographic
ventricular function and prognosis. findings, and serum biomarkers of myocar-
10090-07_CH07.qxd 8/31/06 4:50 PM Page 179

Acute Coronary Syndromes 179

dial damage. To institute appropriate imme- increase in the frequency, duration, and/or
diate therapy, the most important distinc- intensity of ischemic episodes; (2) episodes of
tion to make is between ACS that cause ST angina that occur at rest, without provoca-
segment elevation on the electrocardiogram tion; or (3) the new onset of anginal episodes,
(STEMI) and those acute syndromes that do described as severe, in a patient without pre-
not (UA and NSTEMI). vious symptoms of coronary artery disease.
Historically, MIs have been classified as These features are different from the pattern
Q-wave or non–Q-wave infarctions. Dogma of chronic stable angina, in which instances
held that transmural infarcts produce Q of chest discomfort are predictable, brief, and
waves (after initial ST elevation) on the elec- nonprogressive, occurring only during physi-
trocardiogram (ECG), whereas subendocar- cal exertion or emotional stress. Patients with
dial infarcts generate ST depressions without UA may progress further along the contin-
Q-wave development. However, it is now uum of ACS and develop evidence of necrosis
known that these ECG findings do not reli- (i.e., acute NSTEMI or STEMI) unless the con-
ably correlate with the pathologic findings dition is recognized and promptly treated.
and that much overlap exists among the
types of infarction. Moreover, the use of Q Acute Myocardial Infarction
waves to classify ACS is now less clinically
important, because Q waves, unlike ST The symptoms and physical findings of
changes, may take hours or longer to develop acute MI (both STEMI and NSTEMI) can be
and cannot be used to make early therapeu- predicted from the pathophysiology de-
tic decisions. Thus, for the remainder of this scribed earlier in this chapter and are sum-
chapter, the terms STEMI and NSTEMI will marized in Table 7.3. The discomfort expe- Tab. 3
be used instead of Q-wave and non–Q-wave rienced during an MI resembles angina
MI, respectively. pectoris qualitatively but is usually more se-
vere, lasts longer, and may radiate more
widely. Like angina, the sensation may result
Clinical Presentation from the release of mediators such as adeno-
sine and lactate from ischemic myocardial
Unstable Angina
cells onto local nerve endings. Because is-
UA presents as an acceleration of ischemic chemia in acute MI persists and proceeds to
symptoms in one of three ways: (1) a cres- necrosis, these provocative substances con-
cendo pattern in which a patient with tinue to accumulate and activate afferent
chronic stable angina experiences a sudden nerves for longer periods. The discomfort is

TABLE 7.3. Signs and Symptoms of Myocardial Infarction

1. Characteristic pain • Severe, persistent, typically substernal


2. Sympathetic effect • Diaphoresis
• Cool and clammy skin
3. Parasympathetic (vagal effect) • Nausea, vomiting
• Weakness
4. Inflammatory response • Mild fever
5. Cardiac findings • S4 (and S3 if CHF present) gallop
• Dyskinetic bulge (in anterior wall MI)
• Pericardial friction rub (if pericarditis present)
• Systolic murmur (if mitral regurgitation or VSD)
6. Other • Pulmonary rales (if CHF present)
• Jugular venous distention (if right ventricular MI)

CHF, congestive heart failure; MI, myocardial infarction; S3, third heart sound; S4, fourth heart sound; VSD, ventricular
septal defect.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 180

180 Chapter Seven

often referred to other regions of the C7 tive of volume overload in the presence of
through T4 dermatomes, including the neck, failing LV systolic function, may also be
shoulders, and arms. Initial symptoms are heard. A friction rub may be present if in-
usually rapid in onset and briskly crescendo flammation has extended to the pericardium.
to leave the victim with a profound “feeling A systolic murmur may appear if ischemia-
of doom.” Unlike a transient attack of angina, induced papillary muscle dysfunction causes
the pain does not wane with rest, and there mitral valvular insufficiency, or if the infarct
may be little response to the administration ruptures through the interventricular sep-
of sublingual nitroglycerin. tum to create a ventricular septal defect (as
The chest discomfort associated with an discussed later in the chapter).
acute MI is sometimes severe, but not always. Myocardial necrosis also activates systemic
In fact, up to 25% of patients who sustain an responses to inflammation. Cytokines such as
MI are asymptomatic during the acute event, interleukin 1 (IL-1) and tumor necrosis factor
and the diagnosis is made only in retro- (TNF) are released from macrophages and vas-
spect. This is particularly common among cular endothelium in response to tissue in-
diabetic patients who may not adequately jury. These mediators evoke an array of clini-
sense pain because of peripheral neuropa- cal responses, including low-grade fever.
thy. In addition, some patients who present Not all patients with severe chest pain are
with MI complicated by acute pericarditis in the midst of MI or UA. Table 7.4 lists other Tab. 4
may feel more of a sharp, pleuritic-type pain common causes of chest discomfort and clin-
(see Chapter 14), rather than the typical MI ical, laboratory, and radiographic features to
symptoms. differentiate them from an acute coronary
The combination of intense discomfort syndrome.
and baroreceptor unloading (if hypotension
is present) may trigger a dramatic sympa- Diagnosis of Acute
thetic nervous system response. Systemic Coronary Syndromes
signs of subsequent catecholamine release
include diaphoresis (sweating), tachycardia, The diagnosis of, and distinctions among,
and cool and clammy skin caused by vaso- the acute coronary syndromes is made on the
constriction. basis of (1) the patient’s presenting symp-
If the ischemia affects a sufficiently large toms, (2) acute ECG abnormalities, and
amount of myocardium, left ventricular (LV) (3) detection or absence of specific serum
contractility can be reduced (systolic dys- markers of myocardial necrosis (see Fig. 7.4
function) thereby decreasing the stroke vol- and Table 7.5). Specifically, UA is a clinical Tab. 5
ume and causing the diastolic volume and diagnosis supported by the patient’s symp-
pressure within the LV to rise. The increase in toms, transient ST abnormalities on the ECG
LV pressure, compounded by the ischemia- (usually ST depression and/or T wave inver-
induced stiffness of the chamber (diastolic sion), and the absence of serum biomarkers
dysfunction), is conveyed to the left atrium of myocardial necrosis. Non–ST segment el-
and pulmonary veins. The resultant pul- evation MI is distinguished from UA by the
monary congestion decreases lung compli- detection of serum markers of necrosis and
ance and stimulates juxtacapillary receptors. often more persistent ST or T wave abnor-
These J receptors effect a reflex that results in malities. The hallmark of ST-elevation MI is
rapid, shallow breathing and evokes the an appropriate clinical history coupled with
subjective feeling of dyspnea. Transudation ST elevations on the ECG plus detection of
of fluid into the alveoli exacerbates this serum markers of myocardial necrosis.
symptom.
Physical findings during an acute MI de-
ECG Abnormalities
pend on the location and extent of the in-
farct. The S4 sound, indicative of atrial con- ECG abnormalities, which reflect abnormal
traction into a noncompliant left ventricle, electrical currents during ACS, are usually
is frequently present. An S3 sound, indica- manifest in characteristic ways. In UA or
10090-07_CH07.qxd 8/31/06 4:50 PM Page 181

Acute Coronary Syndromes 181

TABLE 7.4. Conditions That May Be Confused With Acute Coronary Syndromes

Condition Differentiating Features

Cardiac
Acute coronary syndrome • Retrosternal pressure, radiating to neck, jaw, or left shoulder and arm; more
severe and lasts longer than previous anginal attacks
• ECG: localized ST elevations or depressions
Pericarditis • Sharp pleuritic pain (worsens with inspiration)
• Pain varies with position (relieved by sitting)
• Friction rub auscultated over precordium
• Electrocardiogram: diffuse ST elevations (see Chapter 14)
Aortic dissection • Tearing, ripping pain that migrates over time (chest and back)
• Asymmetry of arm blood pressures
• Widened mediastinum on chest radiograph
Pulmonary
Pulmonary embolism • Localized pleuritic pain, accompanied by dyspnea
• Pleural friction rub may be present
• Predisposing conditions for venous thrombosis
Pneumonia • Pleuritic chest pain
• Cough and sputum production
• Abnormal lung auscultation and percussion (i.e., consolidation)
• Infiltrate on chest radiograph
Pneumothorax • Sudden sharp, pleuritic unilateral chest pain
• Decreased breath sounds and hyperresonance of affected side
• Chest radiograph: increased lucency and absence of pulmonary markings
Gastrointestinal
Esophageal spasm • Retrosternal pain, worsened by swallowing
• History of dysphagia
Acute cholecystitis • Right upper quadrant abdominal tenderness
• Often accompanied by nausea
• History of fatty food intolerance

NSTEMI, ST segment depression and/or T of the T wave and Q-wave development (Fig.
Fig. 7 wave inversions are most common (Fig. 7.7). 7.8). Note that these characteristic patterns of Fig. 8
These abnormalities may be transient, occur- ECG abnormalities in ACS can be minimized
ring just during chest pain episodes in UA, or or aborted by early therapeutic interventions.
they may persist in patients with NSTEMI. In
contrast, as described in Chapter 4, STEMI
Serum Markers of Infarction
presents with a temporal sequence of abnor-
malities: initial ST segment elevation, fol- Necrosis of myocardial tissue causes disrup-
lowed over the next hours to day by inversion tion of the sarcolemma, so that intracellular

TABLE 7.5. Distinguishing Features of Acute Coronary Syndromes

Myocardial Infarction

Feature Unstable Angina NSTEMI STEMI

Typical symptoms Crescendo, rest, or new Prolonged “crushing” chest pain, more
onset severe angina severe and wider radiation than usual angina
Serum biomarkers No Yes Yes
Electrocardiogram ST depression and/or ST depression and/or ST elevation
initial findings T wave inversion T wave inversion (and Q waves later)

NSTEMI, non–ST-elevation myocardial infarction; STEMI, ST-elevation myocardial infarction.


10090-07_CH07.qxd 8/31/06 4:50 PM Page 182

182 Chapter Seven

Figure 7.7. ECG abnormalities in unstable angina and non–ST-


elevation myocardial infarction.

macromolecules leak into the cardiac inter- tween myosin and actin (see Chapter 1). It
stitium and ultimately into the bloodstream consists of three subunits: TnC, TnI, and
Fig. 9 (Fig. 7.9). Detection of such molecules in the TnT. Although these subunits are found
serum, particularly cardiac-specific troponins both in skeletal and cardiac muscle, the car-
and creatine kinase MB isoenzyme, serves diac forms of troponin I (cTnI) and troponin
important diagnostic and prognostic roles. T (cTnT) are structurally unique, and highly
In patients with STEMI or NSTEMI, these specific assays for their detection in the
markers rise above a threshold level in a de- serum have been developed. Because serum
fined temporal sequence. levels are virtually absent in healthy persons,
the presence of even minor elevations of
cTnI or cTnT serves as a sensitive and pow-
Cardiac-Specific Troponins
erful marker of myocyte damage.
Troponin (Tn) is a regulatory protein in Cardiac troponins begin to rise 3 to 4 hours
muscle cells that controls interactions be- after the onset of an MI, peak between 18 and

Figure 7.8. ECG evolution during ST-elevation myocardial infarction.


10090-07_CH07.qxd 8/31/06 4:50 PM Page 183

Acute Coronary Syndromes 183

50

Multiples of MI threshold
20

10 Troponins

5
CK-MB
2

1 MI Threshold

1 2 3 4 5 6 7 8 9 10
Days after onset of infarction
Figure 7.9. Evolution of serum biomarkers in acute myocardial infarction
(MI). Serum creatine kinase (and CK-MB isoenzyme) begins to rise 3 to 8 hours
after the onset of the acute infarct and peaks at 24 hours. Cardiac troponins
are highly sensitive for myocardial injury and remain detectable in the serum for
many days after the acute infarct. (Modified from Wu AH, Apple FS, Gibler WB,
et al. National Academy of Clinical Biochemistry Standards of Laboratory Prac-
tice: Recommendations for the use of cardiac markers in coronary artery dis-
eases. Clin Chem 1999;45:1104–1121.)

36 hours, and then decline slowly, allowing side the heart, including the uterus, prostate,
for detection for up to 10 to 14 days. Thus, gut, diaphragm, and tongue. CK-MB also
their measurement may be helpful for de- makes up 1% to 3% of the creatine kinase in
tection of MI for nearly 2 weeks after the skeletal muscle. In the absence of trauma to
event occurs. Given their high sensitivity these other organs and tissues, elevation of
and specificity, cardiac troponins are the pre- CK-MB is highly suggestive of myocardial
ferred serum biomarkers to detect myocardial injury. To facilitate the diagnosis of MI
necrosis. using this marker, it is common to calculate
the ratio of CK-MB to total CK. The ratio is
usually >2.5% in the setting of myocardial
Creatine Kinase injury and even less when CK-MB elevation
The enzyme creatine kinase (CK) reversibly is from another source.
transfers a phosphate group from creatine The serum level of CK-MB starts to rise
phosphate, the endogenous storage form 3 to 8 hours following infarction, peaks at
of high-energy phosphate bonds, to ADP, 24 hours, and returns to normal within 48
producing ATP. Because creatine kinase is to 72 hours (see Fig. 7.9). This temporal se-
found in the heart, skeletal muscle, brain, quence is important because other sources
and many other organs, serum concentra- of CK-MB (e.g., skeletal muscle injury) or
tions of the enzyme may become elevated other non-MI cardiac conditions that raise
following injury to any of these tissues. serum levels of the isoenzyme (e.g., myo-
There are, however, three isoenzymes of carditis) do not usually show this delayed
CK that improve diagnostic specificity of its peaking pattern.
origin: CK-MM (found mainly in skeletal The detection of cardiac troponins in the
muscle), CK-BB (located predominantly in serum is a much more sensitive marker for
the brain), and CK-MB (localized mainly in myocardial necrosis than CK-MB. As a re-
the heart). It should be noted that small sult, many patients with ACS are found to
amounts of CK-MB are found in tissues out- have small elevations of cardiac troponins
10090-07_CH07.qxd 8/31/06 4:50 PM Page 184

184 Chapter Seven

but unremarkable CK-MB test results. Before TREATMENT OF ACUTE


the advent of troponin assays, such occur- CORONARY SYNDROMES
rences would have been labeled as UA (be-
cause of the absence of the CK-MB biomarker) Successful management of ACS requires rapid
but are now more accurately classified as initiation of therapy to limit myocardial
NSTEMI. damage and minimize complications. Ther-
Because troponin and CK-MB levels do apy must address the intracoronary throm-
not become elevated in the serum until at bus that incited the syndrome and provide
least a few hours after the onset of MI symp- anti-ischemic measures to restore the bal-
toms, a single normal value drawn early in ance between myocardial oxygen supply
the course of evaluation (e.g., in the hospi- and demand. Although certain therapeutic
tal emergency department) does not rule aspects are common to all ACS, there is a
out an acute MI; thus, the diagnostic utility critical difference in the approach to patients
of these biomarkers is limited in that critical who present with ST segment elevation
period. As a result, early decision making (STEMI) compared with those without ST
in patients with acute coronary syndromes segment elevation (UA and NSTEMI). Pa-
often relies most heavily on the patient’s tients with STEMI typically have total occlu-
history and ECG findings. sion of a coronary artery and benefit from
Sometimes the early diagnosis of MI can immediate reperfusion therapies (pharmaco-
remain uncertain even after careful evalua- logic or mechanical), while patients without
tion of the patient’s history, ECG, and serum ST elevation do not (Fig. 7.10 and as discussed Fig. 10
biomarkers. In such a situation, an additional later in the chapter).
diagnostic study that may be useful is echo- General in-hospital measures for any pa-
cardiography, which typically reveals abnor- tient with ACS include admitting the pa-
malities of ventricular contraction in the re- tient to an intensive care setting where
gion of ischemia or infarction. continuous ECG monitoring for arrhyth-

Symptoms of Acute Coronary Syndrome

ST elevation ECG No ST elevation


(STEMI) (UA/ NSTEMI)

Reperfusion approach All patients Antithrombotic approach


1. Aspirin 1. Aspirin
2. Heparin (UFH or LMWH) 2. Heparin (UFH or LMWH)
3. Clopidogrel 3. Clopidogrel
4. Choose reperfusion method: 4. For high risk patients:
A. Fibrinolytic drug • GP IIb/IIIa inhibitor
B. Primary PCI • Proceed to cardiac cath
• with GP IIb/IIIa inhibitor

Figure 7.10. Management strategies in acute coronary syndromes (ACS). The interventions on the left side
should be considered in patients who present with ST-elevation myocardial infarction (STEMI), while those on the
right side are appropriate in unstable angina (UA) and non–ST-elevation MI (NSTEMI). All patients with ACS can ben-
efit from the measures listed in the center column. ACE, angiotensin-converting enzyme; ECG, electrocardiogram;
GP, glycoprotein; LMWH, low molecular weight heparin; PCI, percutaneous coronary intervention; UFH, unfraction-
ated heparin.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 185

Acute Coronary Syndromes 185

mias is undertaken. The patient is initially cardial oxygen demand by diminishing ve-
maintained at bedrest to minimize myo- nous return to the heart (reduced preload
cardial oxygen demand, while supplemen- and therefore less wall stress). Nitrates may
tal oxygen is administered (by face mask also improve coronary flow and prevent va-
or nasal cannula), if there is any degree of sospasm through coronary vasodilatation.
hypoxemia, to improve oxygen supply. In UA or NSTEMI, nitroglycerin is often ini-
Analgesics, such as morphine, are admin- tially administered by the sublingual route,
istered to reduce chest pain and anxiety followed by a continuous intravenous infu-
and thus further reduce myocardial oxygen sion. In addition to providing symptomatic
needs. relief of angina, intravenous nitroglycerin
is useful as a vasodilator in patients with
ACS accompanied by heart failure or severe
Acute Treatment of Unstable hypertension.
Angina and Non–ST-Elevation Nondihydropyridine calcium channel
Myocardial Infarction antagonists (i.e., verapamil and diltiazem)
exert anti-ischemic effects by decreasing
The management of UA and NSTEMI is es-
heart rate and contractility and through
sentially the same and is therefore discussed
their vasodilatory properties (see Chapter 6).
as one entity, whereas the approach to STEMI
These agents do not confer mortality benefit
is described later. The primary focus of treat-
to patients with ACS and are reserved for
ment for UA and NSTEMI consists of anti-
those in whom ischemia persists despite
ischemic medications to restore the balance
β-blocker and nitrate therapies, or for those
between myocardial oxygen supply and de-
with contraindications to β-blocker use.
mand and antithrombotic therapy aimed at
They should not be prescribed to patients
stabilizing the underlying partially occlusive
with LV systolic dysfunction, because clini-
coronary thrombus.
cal trials have shown adverse outcomes in
such cases.
Anti-ischemic Therapy
The same pharmacologic agents used to de- Antithrombotic Therapy
crease myocardial oxygen demand in chronic The purpose of antithrombotic therapy,
stable angina are appropriate in UA and including antiplatelet and anticoagulant
NSTEMI but are often administered more medications, is to prevent further propaga-
aggressively. b-Blockers decrease sympa- tion of the partially occlusive intracoronary
thetic drive to the myocardium, thus reduc- thrombus while facilitating its dissolution
ing oxygen demand, and contribute to elec- by endogenous mechanisms. Aspirin in-
trical stability. This group of drugs reduces hibits platelet synthesis of thromboxane A2,
the likelihood of progression from UA to MI a potent mediator of platelet activation, and
and lowers mortality rates in patients who is one of the most important interventions
present with infarction. In the absence of to reduce mortality in patients with all
contraindications (e.g., marked bradycardia, forms of ACS. It should be administered im-
bronchospasm, decompensated heart fail- mediately on presentation and continued
ure, or hypotension), a β-blocker is initially indefinitely in patients without contraindi-
administered intravenously and then con- cations to its use (e.g., allergy or underlying
verted to an oral regimen to achieve a target bleeding disorder).
heart rate of approximately 60 bpm. Such Because aspirin blocks only one pathway
therapy is usually continued indefinitely of platelet activation and aggregation, other
after hospitalization because of proven long- antithrombotic agents have also been stud-
term mortality benefits following an MI. ied. The thienopyridines (clopidogrel and
Nitrates help bring about anginal relief ticlopidine) inhibit ADP-mediated activa-
through venodilatation, which lowers myo- tion of platelets (see Chapter 17) and can be
10090-07_CH07.qxd 8/31/06 4:50 PM Page 186

186 Chapter Seven

used as a substitute in aspirin-allergic pa- fined to high-risk patients (e.g., those with
tients. In addition, the combination of as- elevated troponin levels or who experience
pirin plus clopidogrel is superior to aspirin recurrent episodes of chest pain).
alone in reducing cardiovascular mortality,
recurrent cardiac events, or stroke in pa-
Conservative Versus Early Invasive
tients with UA or NSTEMI.
Management of UA and NSTEMI
Intravenous unfractionated heparin
(UFH), an anticoagulant, is also standard Many patients with UA or NSTEMI stabilize
therapy for UA and NSTEMI. It binds to an- following institution of the therapies de-
tithrombin III, which greatly increases the scribed in previous section, while others
potency of the plasma protein in the inacti- progress to a more severe form of ACS. There
vation of clot-forming thrombin. UFH addi- is currently no definitive way to predict
tionally inhibits coagulation factor Xa, slow- which path a patient will take or to quickly
ing thrombin formation and thereby further determine which patients have such severe
impeding clot development. In patients with underlying CAD that revascularization is
UA or NSTEMI, UFH improves cardiovascu- warranted. These uncertainties have led to
lar outcomes and reduces the likelihood of two diagnostic strategies in UA/NSTEMI
progression from UA to MI. It is adminis- management: (1) an early invasive approach,
tered as a weight-based bolus, followed by in which urgent cardiac catheterization is
continuous infusion, with adjustment in its performed and coronary revascularization
dosage determined by measurements of the undertaken as indicated, or (2) a conservative
serum activated partial thromboplastin time approach, in which the patient is managed
(aPTT). with medications (as detailed in the previous
To overcome the shortcomings of UFH, section) and undergoes angiography only if
low molecular weight heparins (LMWHs) ischemic episodes spontaneously recur or if
have been developed. Like UFH, LMWHs in- the results of a subsequent stress test indicate
teract with antithrombin III but preferen- residual inducible ischemia. The conservative
tially inhibit coagulation factor Xa. LMWHs approach offers the advantage of avoiding
provide a more predictable pharmacologic costly and potentially risky invasive proce-
response than UFH. As a result, LMWHs are dures. On the other hand, an early invasive
easier to use, prescribed as one or two daily strategy allows rapid identification and de-
subcutaneous injections based on the pa- finitive treatment (i.e., revascularization) for
tient’s weight. Unlike UFH, repeated moni- those with critical coronary disease.
toring of blood tests and dosage adjust- Although early randomized trials com-
ments are not necessary. In clinical trials in paring these two approaches suggested that
patients with UA or NSTEMI, the LMWH each leads to comparable outcomes, the
enoxaparin (see Chapter 17) provides clinical weight of more recent data (in the era of
outcomes and safety comparable, if not su- coronary stents and platelet GP IIb/IIIa re-
perior, to UFH. ceptor antagonists) indicate that an early in- AQ1
The glycoprotein (GP) IIb/IIIa receptor vasive approach results in superior outcomes
antagonists (which include the mono- with lower rates of recurrent angina and re-
clonal antibody abciximab and the small- infarction. An early invasive approach ap-
molecule eptifibatide and tirofiban) are po- pears to be most beneficial in patients with
tent antiplatelet agents that block the final high-risk features, such as those with ST seg-
common pathway of platelet aggregation ment deviations at the time of presentation,
(see Chapter 17). These agents are very ef- elevated serum troponin levels, and the pres-
fective in reducing cardiac events in patients ence of multiple cardiac risk factors. As inva-
undergoing percutaneous coronary inter- sive techniques and pharmacologic thera-
vention (PCI). Their benefit in patients with pies improve over time, the optimal strategy
UA or NSTEMI who are managed pharma- in patients with UA or NSTEMI continues to
cologically is more modest, generally con- be an area of active investigation.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 187

Acute Coronary Syndromes 187

Acute Treatment of ST-Elevation thereby restoring blood flow and limiting


Myocardial Infarction myocardial damage. Please note that this
discussion does not pertain to patients with
In contrast to UA and NSTEMI, the culprit UA or NSTEMI, who do not benefit from fib-
artery in STEMI is typically completely oc- rinolytic therapy.
cluded. Thus, to limit myocardial damage, Currently used fibrinolytic agents include
the major focus of acute treatment for such the recombinant tissue-type plasminogen
patients is to achieve prompt reperfusion of activator alteplase (tPA), reteplase (rPA),
the jeopardized myocardium using either fib- and tenecteplase (TNK-tPA). Streptokinase,
rinolytic drugs or percutaneous coronary one of the earliest studied fibrinolytics, is no
revascularization. These approaches reduce longer in widespread use in the United
the extent of myocardial necrosis and greatly States. Each drug functions by stimulating
improve survival. To be effective, they must the natural fibrinolytic system, transforming
be undertaken as soon as possible; the earlier the inactive precursor plasminogen into the
the intervention occurs, the greater the active protease plasmin, which lyses fibrin
amount of myocardium that can be sal- clots. Although the intracoronary thrombus
vaged. Decisions about therapy must be is the target, plasmin has poor substrate
made within minutes of a patient’s arrival to specificity and can degrade other proteins,
the hospital, based on the history and elec- including fibrinogen. As a result, bleeding is
trocardiographic findings, often before serum the most common complication of these
markers of necrosis would be expected to rise. drugs. However, unlike the older fibrinolytic
In addition, as is the case in UA and streptokinase, the newer agents bind prefer-
NSTEMI, specific medications should be ini- entially to fibrin in a formed thrombus (i.e.,
tiated promptly to prevent further thrombo- the intracoronary clot), thereby generating
sis and to restore the balance between my- plasmin locally at that site, with less inter-
ocardial oxygen supply and demand. For ference of coagulation in the general circu-
example, antiplatelet therapy with aspirin lation (Fig. 7.11). Nonetheless, bleeding re- Fig. 11
also decreases mortality rates and rates of mains the most important risk with all
reinfarction after STEMI. It should be ad- fibrinolytic agents.
ministered immediately on presentation and rPA and TNK-tPA are derivatives of tPA
continued daily thereafter. Intravenous un- with longer half-lives. Their main advantage
fractionated heparin is typically infused to is that they can be administered as IV boluses,
help maintain patency of the coronary vessel which is more convenient and less prone to
and is an important adjunct to modern fibri- incorrect administration than the continuous
nolytic regimes. b-Blockers reduce myo- intravenous infusion necessary for tPA.
cardial oxygen demand and lower the risk of Administration of fibrinolytic agents in
recurrent ischemia, arrhythmias, and rein- the early hours of an acute STEMI restores
farction. In the absence of contraindications blood flow in most (70% to 80%) of coro-
(e.g., asthma, hypotension, or significant nary occlusions and significantly reduces
bradycardia), a β-blocker should be adminis- the extent of tissue damage. Improved artery
tered, usually intravenously at first and then patency translates into substantially in-
orally. Nitrate therapy, usually intravenous creased survival rates and fewer postinfarc-
nitroglycerin, is used to help control ischemic tion complications. The rapid initiation of
pain and also serves as a beneficial vasodilator fibrinolysis is crucial: patients who receive
in patients with heart failure or severe hyper- therapy within 2 hours of the onset of symp-
tension during acute infarction. toms of STEMI have half the mortality rate of
those who receive it after 6 hours.
Successful reperfusion is marked by the re-
Fibrinolytic Therapy
lief of chest pain, return of the ST segments to
Fibrinolytic drugs accelerate lysis of the oc- baseline, and earlier-than-usual peaking of
clusive intracoronary thrombus in STEMI, serum markers of necrosis, such as cardiac-
10090-07_CH07.qxd 8/31/06 4:50 PM Page 188

188 Chapter Seven

A
tPA P tPA P* Degrade clot without
systemic lytic state

Fibrin clot

B
P
SK
SK

P*

P P*

Systemic lytic
P* state
Fibrin clot

Figure 7.11. Examples of fibrinolytic agents used in ST-elevation myo-


cardial infarction. A. Tissue plasminogen activator (tPA) cleaves fibrin-bound
plasminogen (P) to form active plasmin (P*), which degrades the fibrin clot. The
selectivity of tPA for fibrin-bound P results in localized thrombolysis and mini-
mizes generalized systemic fibrinolysis. TNK-tPA and rPA (see text) act similarly
to tPA but can be administered as boluses, thus simplifying drug administra-
tion. B. The older fibrinolytic streptokinase (SK) combines with fibrin-bound
and circulating plasminogen to form an active complex, which in turn activates
additional plasminogen molecules. The lack of selectivity for fibrin-bound plas-
minogen results in more of a systemic lytic state.

specific troponins and CK-MB. During reper- postinfarction survival advantage of tPA com-
fusion, transient arrhythmias are common pared with streptokinase, at the expense of a
and do not usually require treatment. To pre- slightly increased risk of intracranial hemor-
vent immediate vessel reocclusion after suc- rhage with tPA. More recent trials compared
cessful thrombolysis, antithrombotic regimens tPA with the newer agents rPA and TNK-tPA
are administered, as described in the next and found similar clinical efficacies for all
section. three agents. The most important message
Because the major risk of thrombolysis is from these trials is that early and sustained
bleeding, contraindications to such therapy patency of the infarct-related coronary artery
include situations in which necessary fibrin improves survival. No matter which fibri-
clots within the circulation would be jeopar- nolytic is selected, it must be administered as
dized (e.g., patients with active peptic ulcer soon as possible, ideally within 30 minutes of
disease or an underlying bleeding disorder, the patient’s presentation to the hospital.
patients who have had a recent stroke or who
are recovering from recent surgery). Conse-
Adjunctive Antithrombotic Therapies
quently, approximately 30% of patients may
After Fibrinolysis
not be suitable candidates for thrombolysis.
Several large-scale comparisons of fibri- As previously indicated, aspirin is a main-
nolytic agents have been conducted. In 1993, stay of therapy in patients with ACS and is
the international GUSTO-1 trial found a small typically initiated on the patient’s presen-
10090-07_CH07.qxd 8/31/06 4:50 PM Page 189

Acute Coronary Syndromes 189

tation. Anticoagulants administered with symptom onset to hospital arrival) or are in


fibrinolytic therapy in STEMI enhance clot cardiogenic shock.
lysis and reduce reocclusion rates. Thus, In addition to aspirin and intravenous
for patients treated with tPA, rPA, or TNK- unfractionated heparin, patients undergo-
tPA, adjunctive IV unfractionated heparin ing primary PCI typically receive an intra-
should be administered for up to 48 hours. venous GP IIb/IIIa receptor antagonist in AQ3
Low molecular weight heparins have conjunction with the procedure to reduce
been tested as an alternative to unfraction- thrombotic complications. Also, for most
ated heparin and have been shown to re- patients who receive coronary stents during
duce ischemic complications but at an in- PCI, the oral thienopyridines (e.g., clopido-
creased risk of intracranial hemorrhage in grel) have been shown to reduce the risk of
older patients. ischemic complications and stent thrombo-
The antiplatelet agent clopidogrel, ad- sis. Clopidogrel is therefore typically begun
ministered in combination with aspirin, has in association with the procedure and then
been shown to further reduce mortality and continued for weeks to months, depending
major cardiovascular events in STEMI pa- on the type of stent placed.
tients who receive fibrinolytic drugs; its ad-
ministration should be considered in such pa- Adjunctive Therapies
tients. Conversely, the antiplatelet GP IIb/IIIa
receptor antagonists have not demonstrated Angiotensin-converting enzyme (ACE) in-
AQ2 hibitors limit adverse ventricular remodel-
a survival benefit in those treated with fibri-
ing and reduce the incidence of heart failure,
nolysis.
recurrent ischemic events, and mortality fol-
lowing an MI. Their benefit is additive to
Primary Percutaneous that of aspirin and β-blocker therapies, and
Coronary Intervention they have shown favorable improvements
especially in higher-risk patients—those with
An alternative to fibrinolytic therapy in pa-
anterior wall infarctions or LV systolic dys-
tients with acute STEMI is immediate car-
function.
diac catheterization and PCI of the lesion
Cholesterol-lowering statins (HMG-CoA
responsible for the infarction. This ap-
reductase inhibitors) reduce mortality rates
proach is termed primary PCI and involves
of patients with coronary artery disease (see
angioplasty, and usually stenting, of the Chapter 5). Clinical trials of patients with
culprit vessel. Primary PCI is a very effective ACS have demonstrated that it is safe to
method for reestablishing coronary perfu- begin statin therapy early during hospital-
sion and, in clinical trials performed at ization, and that an intensive lipid-lowering
highly experienced medical centers, has regimen, designed to achieve a low-density
achieved optimal flow in the infarct-related lipoprotein (LDL) level <70 mg/dl, provides
artery in more than 95% of patients. Com- greater protection against subsequent car-
pared with fibrinolytic therapy, primary diovascular events and death than “stan-
PCI leads to greater survival with lower rates dard” targets (i.e., achieving an LDL level
of reinfarction and bleeding. Therefore, pri- <100 mg/dL). The benefits of statin therapy
mary PCI is usually the preferred reperfusion may extend beyond lipid lowering, because
approach in acute STEMI, if the procedure this group of drugs has attributes that can
can be performed by an experienced opera- improve endothelial dysfunction, inhibit
tor in a rapid fashion (within 90 minutes of platelet aggregation, and impair thrombus
hospital presentation). In addition, primary formation.
PCI is preferred for patients who have con- In addition to the short-term use of he-
traindications to fibrinolytic therapy or are parin anticoagulation described earlier, a
unlikely to do well with fibrinolysis, includ- more prolonged course followed by oral an-
ing those who present late (>3 hours from ticoagulation (i.e., warfarin) is appropriate
10090-07_CH07.qxd 8/31/06 4:50 PM Page 190

190 Chapter Seven

for patients at high risk of thromboem- Recurrent Ischemia


bolism—for example, patients with docu-
Postinfarction angina has been reported to
mented intraventricular thrombus (typi-
occur in 20% to 30% of patients following
cally identified by echocardiography) or
an MI. This rate has not been reduced by the
atrial fibrillation and those who have suf-
use of thrombolytic therapy, but it is lower
fered a large acute anterior MI with akinesis
in those who have undergone percutaneous
of that territory (which is susceptible to
angioplasty or coronary stent implantation
thrombus formation because of the stagnant
as part of early MI management. Indicative
blood flow).
of inadequate residual coronary blood flow,
it is a poor omen and correlates with an in-
COMPLICATIONS creased risk for reinfarction. Such patients
usually require urgent cardiac catheteriza-
In unstable angina, the potential complica- tion, often followed by revascularization by
tions include death (5% to 10%) or progres- percutaneous techniques or coronary artery
sion to infarction (10% to 20%) over the en- bypass surgery.
suing days and weeks. Once infarction has
transpired, especially STEMI, complications
Arrhythmias
can result from the inflammatory, mechan-
ical, and electrical abnormalities induced Arrhythmias occur frequently during acute
by regions of necrosing myocardium (Fig. MI and are a major source of mortality prior
Fig. 12 7.12). Early complications result from my- to hospital arrival. Modern coronary care
ocardial necrosis itself. Those that develop units are highly attuned to the detection and
several days to weeks later reflect the in- treatment of rhythm disturbances; thus,
flammation and healing of necrotic tissue. once a patient is hospitalized, arrhythmia-

Myocardial Infarction

Ventricular Electrical Tissue Pericardial


Contractility
thrombus instability necrosis inflammation
Cardiogenic
shock
Embolism Arrhythmias Pericarditis
Ischemia Hypotension

Coronary
perfusion
pressure

Papillary Ventricular Ventricular


muscle septal rupture
infarction/ defect
ischemia
Cardiac
Mitral tamponade
regurgitation

Congestive heart failure

Figure 7.12. Complications of MI. Infarction results in decreased contractility, electrical instability, and tissue
necrosis, which lead to the indicated sequelae.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 191

Acute Coronary Syndromes 191

TABLE 7.6. Arrhythmias in Acute Myocardial Infarction

Rhythm Cause

Sinus bradycardia • ↑Vagal tone AQ4


• ↓SA nodal artery perfusion
Sinus tachycardia • Pain and anxiety
• CHF
• Volume depletion
• Pericarditis
• Chronotropic drugs (e.g., dopamine)
APBs, atrial fibrillation • CHF
• Atrial ischemia
VPBs, VT, VF • Ventricular ischemia
• CHF
AV block (1°, 2°, 3°) • IMI: ↑vagal tone and ↓AV nodal artery flow AQ5
• AMI: extensive destruction of conduction tissue

AMI, anterior myocardial infarction; APBs, atrial premature beats; AV, atrioventricular; CHF, congestive heart failure; IMI, in-
ferior myocardial infarction; SA, sinoatrial; VPBs, ventricular premature beats; VF, ventricular fibrillation; VT, ventricular
tachycardia.

associated deaths are uncommon. Mecha- 3. Autonomic stimulation (sympathetic and


nisms that contribute to arrhythmogenesis parasympathetic).
Tab. 6 after MI include the following (Table 7.6): 4. Administration of potentially arrhythmo-
1. Anatomic interruption of perfusion to genic drugs (e.g., dopamine).
structures of the conduction pathway
(e.g., sinoatrial node, atrioventricular Ventricular Fibrillation
node, bundle branches); the normal
Ventricular fibrillation (rapid, disorganized
perfusion of pertinent components of
electrical activity of the ventricles) is largely
the conduction system is reviewed in
Tab. 7 responsible for episodes of sudden cardiac
Table 7.7.
death during the course of acute MI. Most
2. Accumulation of toxic metabolic prod- fatal episodes occur before hospital arrival, a
ucts (e.g., cellular acidosis) and abnormal trend that will hopefully improve with in-
transcellular ion concentrations owing to creasing availability of automatic external
membrane leaks. defibrillators in public places. Episodes of
ventricular fibrillation that occur during the
first 48 hours of MI are often related to tran-
sient electrical instability, and the long-term
TABLE 7.7. Blood Supply of the
Conduction System prognosis of survivors of such events is not
affected. However, ventricular fibrillation
Conduction Pathway Primary Arterial Supply occurring later than 48 hours after the acute
SA node • RCA (70% of patients)
MI usually reflects severe LV dysfunction
AV node • RCA (85% of patients) and is associated with high subsequent mor-
Bundle of His • LAD (septal branches) tality rates.
RBB • Proximal portion by LAD Ventricular ectopic beats, ventricular ta-
• Distal portion by RCA chycardia, and ventricular fibrillation dur-
LBB
ing an acute MI arise from either reentrant
Left anterior fascicle • LAD
Left posterior fascicle • LAD and PDA circuits or enhanced automaticity of ven-
tricular cells (see Chapter 11). Ventricular
AV, atrioventricular; LAD, left anterior descending coronary ectopy is common but usually not treated
artery; LBB, left bundle branch; PDA, posterior descending
artery; RBB, right bundle branch; RCA, right coronary artery; unless the abnormal beats become conse-
SA, sinoatrial. cutive, multifocal, or frequent. Intravenous
10090-07_CH07.qxd 8/31/06 4:50 PM Page 192

192 Chapter Seven

lidocaine (a class IB antiarrhythmic drug de- tion, ventricular remodeling, arrhythmias,


scribed in Chapter 17) is effective in prevent- and acute mechanical complications of MI
ing ventricular fibrillation in the ischemic (described later in the chapter) may culmi-
setting but is not indicated prophylactically nate in heart failure. Signs and symptoms of
in the management of acute MI because of such decompensation include dyspnea, pul-
its potential side effects and because cardiac monary rales, and a third heart sound (S3).
care unit personnel are proficient at arrhy- Treatment consists of standard heart failure
thmia detection and treatment, should that therapy (see Chapter 9).
complication occur.
Cardiogenic Shock
Supraventricular Arrhythmias
Cardiogenic shock is a condition of severely
Supraventricular arrhythmias are also com- decreased cardiac output and hypotension
mon in acute MI. Sinus bradycardia results (systolic blood pressure <90 mm Hg) with in-
from either excessive vagal stimulation or adequate perfusion of peripheral tissues that
sinoatrial nodal ischemia, usually in the set- develops when more than 40% of the LV
ting of an inferior wall MI. Sinus tachycardia mass has infarcted. It may also follow certain
occurs frequently and may result from many severe mechanical complications of MI de-
causes, especially pain and anxiety, heart scribed later. Demise in cardiogenic shock is
failure, drug administration (e.g., dopamine), self-perpetuating because (1) hypotension
or intravascular volume depletion. Because leads to decreased coronary perfusion, which
sinus tachycardia increases myocardial oxy- exacerbates ischemic damage, and (2) de-
gen demand and could exacerbate ischemia, creased stroke volume increases LV size and
identifying and treating its cause are impor- therefore augments myocardial oxygen de-
tant. Atrial premature beats and atrial fibrilla- mand (see Fig. 7.12). Despite aggressive
tion (see Chapter 12) may result from atrial treatment, the mortality rate of patients in
ischemia or atrial distention secondary to cardiogenic shock is greater than 70%.
LV failure. Patients in cardiogenic shock require in-
travenous inotropic agents (e.g., dobuta-
Conduction Blocks mine) to increase cardiac output and arterial
vasodilators to reduce the resistance to LV
Conduction blocks (atrioventricular nodal contraction. These patients are often stabi-
block and bundle branch blocks) develop lized by the placement of an intra-aortic bal-
commonly in acute MI. They may result from loon pump. Inserted into the aorta through
ischemia or necrosis of conduction tracts (see a femoral artery, the pump consists of an in-
Table 7.7), or in the case of atrioventricular flatable, flexible chamber that expands dur-
blocks, may develop transiently because of in- ing diastole to increase intra-aortic pressure,
creased vagal tone. Vagal activity may be in- thus augmenting perfusion of the coronary
creased because of stimulation of afferent arteries and the peripheral tissues. During
fibers by the inflamed myocardium or as a re- systole, it deflates to create a “vacuum” that
sult of generalized autonomic activation in serves to reduce the afterload of the left ven-
association with the pain of an acute MI. tricle, thus aiding the ejection of blood into
the aorta. Early cardiac catheterization and
Myocardial Dysfunction revascularization (PCI or coronary artery by-
pass graft) has the potential to improve the
Congestive Heart Failure
prognosis of patients in cardiogenic shock.
Acute cardiac ischemia results in impaired
ventricular contractility (systolic dysfunc-
Right Ventricular Infarction
tion) and increased myocardial stiffness
(diastolic dysfunction), both of which may Approximately one third of patients with in-
lead to symptoms of heart failure. In addi- farction of the LV inferior wall also develop
10090-07_CH07.qxd 8/31/06 4:50 PM Page 193

Acute Coronary Syndromes 193

necrosis of portions of the right ventricle, be- On occasion, a pseudoaneurysm results


cause the same coronary artery (usually the if rupture of the free wall is incomplete and
right coronary) perfuses both regions in most held in check by thrombus formation that
patients. The resulting abnormal contraction “plugs” the hole in the myocardium. This
and decreased compliance of the right ven- situation is the cardiac equivalent of a time
tricle lead to signs of right-sided heart failure bomb, because subsequent complete rup-
(e.g., jugular venous distention) out of pro- ture into the pericardium and tamponade
portion to signs of left-sided failure. In addi- could follow. If detected (usually by echo-
tion, profound hypotension may result cardiography), surgical repair may prevent
when right ventricular dysfunction impairs an otherwise disastrous outcome.
blood flow through the lungs, leading to the
left ventricle becoming underfilled. In this
Ventricular Septal Rupture
setting, intravenous volume infusion often
serves to correct hypotension, guided by he- This complication is analogous to LV free
modynamic measurements via a transvenous wall rupture, but the abnormal flow of blood
pulmonary artery catheter (see Chapter 3). is not directed across the LV wall into the
pericardium. Rather, blood is shunted across
the ventricular septum from the left ventri-
Mechanical Complications
cle to the right ventricle, usually precipitat-
Mechanical complications following MI re- ing congestive heart failure because of sub-
sult from tissue ischemia and necrosis. sequent volume overload of the pulmonary
capillaries. A loud systolic murmur at the left
sternal border, representing transseptal flow,
Papillary Muscle Rupture
is common in this situation. Although each
Ischemic necrosis and rupture of an LV pap- results in a systolic murmur, ventricular sep-
illary muscle may be rapidly fatal because tal rupture can be differentiated from acute
of acute severe mitral regurgitation, which mitral regurgitation by the location of the
causes the valve leaflets to lose their anchor- murmur (see Fig. 2.11), by Doppler echocar-
ing attachments. Partial rupture, with more diography, or by measuring the O2 satura-
moderate regurgitation, is not immediately tion of blood in the right-sided heart cham-
lethal but may result in symptoms of heart bers through a transvenous catheter. The O2
failure or pulmonary edema. Because it has a content in the right ventricle is abnormally
more precarious blood supply, the postero- higher than that in the right atrium if there
medial LV papillary muscle is more suscepti- is shunting of oxygenated blood from the
ble to infarction than the anterolateral one; left ventricle across the septal defect.
therefore, this complication is more com-
mon following an inferoposterior MI.
True Ventricular Aneurysm
A late complication of MI, a true ventricular
Ventricular Free Wall Rupture
aneurysm occurs weeks to months after the
An infrequent but deadly complication, rup- acute event. It develops as the ventricular
ture of the LV free wall through a tear in the wall is weakened, but not perforated, by the
necrotic myocardium, may occur within the phagocytic clearance of necrotic tissue, and
first 2 weeks following MI. It is more com- it results in a localized outward bulge (dysk-
mon among women and patients with a his- inesia) when the residual viable heart mus-
tory of hypertension. Hemorrhage into the cle contracts. Unlike the pseudoaneurysm
pericardial space owing to LV free wall rup- described earlier, a true aneurysm does not
ture results in rapid cardiac tamponade, in involve communication between the LV
which blood fills the pericardial space and se- cavity and the pericardium, so that rupture
verely restricts ventricular filling (see Chapter and tamponade do not develop. Complica-
14). Survival is rare. tions of LV aneurysm include (1) thrombus
10090-07_CH07.qxd 8/31/06 4:50 PM Page 194

194 Chapter Seven

formation within this region of stagnant Thromboembolism


blood flow, serving as a potential source of
emboli to peripheral organs; (2) ventricular Stasis of blood flow in regions of impaired
arrhythmias associated with the stretched LV contraction after an MI may incite intra-
myofibers; and (3) heart failure resulting cavity thrombus formation, especially when
from reduced forward cardiac output, be- the infarction involves the LV apex, or
cause some of the LV stroke volume is when a true aneurysm has formed. Subse-
“wasted” by filling the aneurysm cavity dur- quent thromboemboli can result in devas-
ing systole. tating infarction of peripheral organs (e.g., a
Clues to the presence of an LV aneurysm cerebrovascular accident, or stroke, caused
include persistent ST segment elevations on by embolism to the brain).
the ECG weeks after the acute ST-elevation
MI and a bulge at the LV border on chest ra- RISK STRATIFICATION AND
diography. The abnormality can be con- MANAGEMENT FOLLOWING
firmed by echocardiography. MYOCARDIAL INFARCTION
The most important predictor of post-MI
Pericarditis outcome is the extent of LV dysfunction.
Acute pericarditis may occur in the early (in- Other features that portend adverse out-
hospital) post-MI period as necrosis and comes include early recurrence of ischemic
neutrophilic infiltrates extend from the my- symptoms, a large volume of residual myo-
ocardium to the adjacent pericardium (see cardium still at risk because of severe under-
Chapter 14). Sharp pain, fever, and a peri- lying coronary disease, and high-grade ven-
cardial friction rub are often manifest in this tricular arrhythmias.
situation and help distinguish pericarditis To identify patients at high risk for com-
from the discomfort of recurrent myocardial plications who may benefit from cardiac
ischemia. Anticoagulants are relatively con- catheterization and revascularization, exer-
traindicated in MI complicated by pericarditis cise treadmill testing is often performed
to avoid hemorrhage from the inflamed peri- (unless the patient has already undergone
cardial lining. The frequency of MI-associated catheterization and corrective percutaneous
pericarditis has declined since the introduc- revascularization for the presenting coro-
tion of acute reperfusion strategies, because nary syndrome). Patients with significantly
those approaches limit the extent of myocar- abnormal results, or those who demonstrate
dial damage and inflammation. an early spontaneous recurrence of angina,
are referred for cardiac catheterization to de-
fine their coronary anatomy.
Dressler Syndrome Standard postdischarge therapy include
Dressler syndrome is another uncommon aspirin, a β-blocker, and an HMG-CoA re-
form of pericarditis that can occur over the ductase inhibitor (statin) to achieve a long-
first several weeks following hospitaliza- term LDL value of <70 mg/dL. ACE in-
tion for MI. The cause is unclear, but an im- hibitors are prescribed to patients who have
mune process directed against damaged LV contractile dysfunction. Rigorous atten-
myocardial tissue is suspected to play a tion to other cardiac risk factors, such as
role. The syndrome is heralded by fever, smoking, hypertension, and diabetes, is also
malaise, and sharp, pleuritic chest pain typ- mandatory, and a formal exercise rehabili-
ically accompanied by leukocytosis, an tation program often speeds convalescence.
elevated erythrocyte sedimentation rate,
and a pericardial effusion. Similar to other
SUMMARY
forms of acute pericarditis, Dressler syn-
drome generally responds to high-dose as- 1. Acute coronary syndromes include UA,
pirin therapy. NSTEMI, and STEMI. Most ACS episodes
10090-07_CH07.qxd 8/31/06 4:50 PM Page 195

Acute Coronary Syndromes 195

are caused by the formation of intracoro- mechanical intervention, is beneficial in


nary thrombus at the site of atheroscle- high-risk patients.
rotic plaque. Plaque rupture is the usual 6. Acute treatment for STEMI includes early
trigger for thrombus formation through reperfusion strategies with thrombolytic
activation of platelets and the coagula- drugs or PCI. Other important measures
tion cascade. Atherosclerosis-induced include antiplatelet therapy (aspirin,
endothelium dysfunction contributes clopidogrel), a β-blocker, and frequently,
to the process by producing decreased nitrate and heparin therapies. Often a
amounts of vasodilators and antithrom- statin and an ACE inhibitor are also
botic mediators. appropriate.
2. Distinctions among types of ACS are 7. Potential complications of infarction
based on the severity of ischemia and include arrhythmias (e.g., ventricular
whether myocardial necrosis results. tachycardia and fibrillation), atrioven-
ST-elevation MI is associated with an oc- tricular blocks, bundle branch blocks,
clusive thrombus and severe ischemia and supraventricular arrhythmias. Car-
with necrosis. ACS without ST elevation diogenic shock or congestive heart failure
(non–ST-elevation MI and UA) usually may develop because of ventricular dys-
result from partially occlusive thrombi function or the development of mechan-
with less intense ischemia. Compared ical complications (e.g., acute mitral re-
with UA, the insult in NSTEMI is of suffi- gurgitation or ventricular septal defect).
cient magnitude to cause some degree of In addition, wall motion abnormalities
myocardial necrosis. of the affected segment may predispose
3. ACS result in biochemical and mechani- to thrombus formation.
cal changes that impair systolic contrac- 8. Standard pharmacologic therapy follow-
tion, decrease myocardial compliance ing discharge from the hospital includes
(diastolic dysfunction), and predispose measures to reduce the risks of throm-
to arrhythmias. Infarction initiates an bosis (aspirin, clopidogrel), recurrent is-
inflammatory response that clears nec- chemia (a β-blocker), progressive athero-
rotic tissue and leads to scar formation. sclerosis (cholesterol-lowering therapy,
Transient severe ischemia without infarc- usually a statin), and adverse ventricular
tion can cause “stunned” myocardium, a remodeling (an ACE inhibitor, especially
condition of contractile dysfunction that if LV dysfunction is present). Systemic
persists beyond the period of ischemia, anticoagulation with warfarin is indi-
with subsequent gradual recovery of cated if an intraventricular thrombus, a
function. large akinetic segment, or atrial fibrilla-
4. The diagnosis of specific ACS relies on tion are present.
the patient’s history, ECG abnormalities, 9. Post-ACS risk stratification can identify
and appearance of specific biomarkers patients at high risk of recurrent is-
in the serum (cardiac troponins and/or chemia, reinfarction, or death. Impaired
CK-MB). LV function, high-grade ventricular ar-
5. Acute treatment of UA and NSTEMI in- rhythmias, and ischemic changes during
cludes measures to restore balance be- exercise testing all portend unfavorable
tween myocardial oxygen supply and outcomes and warrant further investiga-
demand (β-blockers, nitrates) and stabi- tion and treatment.
lization of the intracoronary thrombus
(aspirin, unfractionated or low molecular Acknowledgment
weight heparin, and possibly additional
The authors thank Frederick Schoen, MD, for his
antiplatelet therapies [e.g., thienopy-
helpful suggestions. Contributors to the previous edi-
ridines, GP IIb/IIIa receptor antagonists]). tions of this chapter were J. G. Fletcher, MD; Anurag
Statin therapy is usually indicated. Early Gupta, MD; Marc S. Sabatine, MD; William Carlson,
coronary angiography, with subsequent MD; Patrick T. O’Gara, MD; and Leonard S. Lilly, MD.
10090-07_CH07.qxd 8/31/06 4:50 PM Page 196

196 Chapter Seven

Additional Reading Fox KA, Poole-Wilson P, Clayton TC, et al. 5-year


outcome of an interventional strategy in non-ST-
American Heart Association. Stabilization of the pa- elevation acute coronary syndrome: the British
tient with acute coronary syndromes. In: 2005 Heart Foundation RITA 3 randomised trial. Lancet
American Heart Association Guidelines for Car- 2005;366:914–920.
diopulmonary Resuscitation and Emergency Car- Gershlick AH, Stephens-Lloyd A, Hughes S, et al. Res-
diovascular Care. Circulation 2005;112:IV-89–IV- cue angioplasty after failed thrombolytic therapy
110. for acute myocardial infarction. N Engl J Med
Antman EM, Anbe DT, Armstrong PW, et al. 2005;353:2758–2768.
ACC/AHA guidelines for the management of pa- Giugliano RP. The year in non-ST-segment elevation
tients with ST-elevation myocardial infarction— acute coronary syndromes. J Am Coll Cardiol
Executive Summary: a report of the American Col- 2005;46:905–919.
lege of Cardiology/American Heart Association Harrington RA, Becker RC, Ezekowitz M, et al. Anti-
Task Force on Practice Guidelines. Circulation thrombotic therapy for coronary artery disease:
2004;110:588–636. the Seventh ACCP Conference on Antithrombotic
Boersma E, Harrington RA, Moliterno DJ, et al. Platelet and Thrombolytic Therapy. Chest 2004;126:513S–
glycoprotein IIb/IIIa inhibitors in acute coronary 548S.
syndromes: a meta-analysis of all major randomised Jaffe AS, Babuin L, Apple FS. Biomarkers in acute car-
clinical trials. Lancet 2002;359:189–198. diac disease: the present and the future. J Am Coll
Braunwald E. Application of current guidelines to the Cardiol 2006;48:1–11.
management of unstable angina and non-ST- Keeley EC, Boura JA, Grines CL. Primary angioplasty
elevation myocardial infarction. Circulation 2003; versus intravenous thrombolytic therapy for acute
108:III-28–III-37. myocardial infarction: a quantitative review of 23
Braunwald E, Antman EM, Beasley JW, et al. randomised trials. Lancet 2003;361:13–20.
ACC/AHA guidelines for management of patients Mehta SR, Cannon CP, Fox KAA, et al. Routine vs. se-
with unstable angina and non-ST-segment eleva- lective invasive strategies in patients with acute
tion myocardial infarction-2002: summary article. coronary syndromes: A collaborative meta-analysis
Circulation 2002;106:1893–1900. of randomized trials. JAMA 2005;293:2908–2917.
Bursi F, Enriquez-Sarano M, Jacobsen SJ, et al. Mitral Petersen JL, Mahaffey KW, Hasselblad V, et al. Efficacy
regurgitation after myocardial infarction: a re- and bleeding complications among patients ran-
view. Am J Med 2006;119:103–112. domized to enoxaparin or unfractionated heparin
Cannon CP, Braunwald E, McCabe CH, et al; Pravas- for antithrombin therapy in non-ST-segment ele-
tatin or Atorvastatin Evaluation and Infection Ther- vation acute coronary syndromes: a systematic
apy—Thrombolysis in Myocardial Infarction overview. JAMA 2004;292:89–96.
(PROVE-IT) 22 investigators. Intensive versus mod- Ray KK, Cannon CP, McCabe CH, et al. Early and
erate lipid lowering with statins after acute coronary late benefits of high-dose atorvastatin in patients
syndromes. N Engl J Med 2004;350:1495–1504. with acute coronary syndromes. J Am Coll Cardiol
Clopidogrel and Metoprolol in Myocardial Infarction 2005;46:1405–1410.
Trial (COMMIT) collaborative group. Addition Sabatine MS, Cannon CP, Gibson CM, et al. Clo-
of clopidogrel to aspirin in 45,852 patients with pidogrel as Adjunctive Reperfusion Therapy
acute myocardial infarction: randomized placebo- (CLARITY)–Thrombolysis in Myocardial Infarc-
controlled trial. Lancet 2005;366:1607–1621. tion (TIMI) 28 investigators. Addition of clopi-
de Winter RJ, Windhausen F, Cornel JH, et al. Inva- dogrel to aspirin and fibrinolytic therapy for
sive versus Conservative Treatment in Unstable myocardial infarction with ST-segment eleva-
Coronary Syndromes (ICTUS) investigators. Early tion. N Engl J Med 2005;352:1179–1189.
invasive versus selectively invasive management
for acute coronary syndromes. N Engl J Med
2005;353:1095–1104.
10090-07_CH07.qxd 8/31/06 4:50 PM Page a

Chapter 7—Author Queries

1. AU: Terminology used above. Correct here?


2. AU: Terminology used earlier. Correct here?
3. AU: Correct term?
4. AU: Please provide correct symbol
5. AU: Please provide correct symbol
10090-08_CH08.qxd 8/31/06 4:51 PM Page 197

C H A P T E R

Valvular Heart Disease


Mia M. Edwards
Patrick T. O’Gara
8
Leonard S. Lilly

RHEUMATIC FEVER PULMONIC VALVE DISEASE


Pulmonic Stenosis
MITRAL VALVE DISEASE
Pulmonic Regurgitation
Mitral Stenosis
Mitral Regurgitation PROSTHETIC VALVES
Mitral Valve Prolapse INFECTIVE ENDOCARDITIS
AORTIC VALVE DISEASE Pathogenesis
Aortic Stenosis Clinical Manifestations
Aortic Regurgitation
TRICUSPID VALVE DISEASE
Tricuspid Stenosis
Tricuspid Regurgitation

This chapter reviews the pathophysiologic usually identify the type of abnormalities
abnormalities in patients with valvular heart that are present. Assessment of the severity of
disease. Each of the common valvular con- the valve lesions can then be facilitated by in-
ditions is discussed separately because uni- formation from the electrocardiogram, chest
fying principles do not govern the behavior radiograph, echocardiogram, and in some
of all stenotic or regurgitant valves. Effective cases, cardiac magnetic resonance imaging
management of affected patients requires (see Chapter 3). In selected patients, further
accurate identification of the valvular le- investigation with exercise testing or cardiac
sion, a determination of its severity, and a catheterization may be necessary to define
clear understanding of the pathophysiologic fully the significance of the condition and
consequences and natural history of the guide therapy.
condition.
The evaluation of a patient with a sus-
RHEUMATIC FEVER
pected valvular lesion begins at the bedside
with a careful history and physical exami- Acute rheumatic fever (ARF) was once among
nation from which the trained clinician can the most common causes of valvular heart

197
10090-08_CH08.qxd 8/31/06 4:51 PM Page 198

198 Chapter Eight

disease, but its incidence has waned consider- Pathologically, rheumatic carditis (i.e., car-
ably in the past half-century in industrialized diac inflammation) may affect all three layers
societies. In the 1940s, the yearly incidence of the heart (pericardium, myocardium, and Fig. 1
exceeded 200,000 cases in the United States, endocardium). Histopathologic examination
whereas the disease is now rare. The decline often demonstrates the Aschoff body (Fig.
of this condition immediately preceded or co- 8.1), an area of focal fibrinoid necrosis sur-
incided with the introduction of penicillin, as rounded by inflammatory cells, including
well as with improvement of general health lymphocytes, plasma cells, and macro-
care and relief from overcrowding. Although phages, that later resolve to form fibrous
occasional local outbreaks occur, a major scar tissue. The most devastating sequelae
resurgence has not been seen in this country. result from inflammatory involvement of
Nevertheless, in developing parts of the the valvular endocardium, which leads to
world, ARF continues to be a scourge with ful- chronic rheumatic heart disease character-
minant consequences. ized by permanent deformity and impair-
ARF is an inflammatory condition that ment of one or more cardiac valves. Symp-
primarily involves the heart, skin, and con- toms of valvular dysfunction, however,
nective tissues. It is a complication of upper generally do not become manifest until 10 to
respiratory tract infections caused by group 30 years after ARF has subsided. This latency
A streptococci and mainly occurs in chil- period may be considerably shorter with the
dren and young adults. During epidemics, more aggressive disease observed in devel-
approximately 3% of patients with acute oping countries.
streptococcal pharyngitis develop ARF 2 to The most common presenting symptoms
3 weeks after the initial throat infection. Al- of ARF are chills, fever, fatigue, and migra-
though the pathogenesis remains unknown, tory arthralgias. The cardinal symptoms
it does not involve direct bacterial infection and clinical manifestations of the disease
of the heart. Some proposed mechanisms that establish the diagnosis are known as
include the elaboration of a toxin by the the Jones criteria (Table 8.1). During the Tab. 1
streptococci or autoimmune cross-reactivity acute episode, carditis may be associated
between bacterial and cardiac antigens. with tachycardia, decreased left ventricular

Figure 8.1. Histopathologic appearance of an Aschoff body in acute rheumatic


carditis. Mononuclear inflammatory cells surround a center of focal necrosis. (From
Schoen FJ. The heart. In: Kumar V, Abbas A, Fausto N, eds. Robbins and Cotran
Pathologic Basis of Disease. 7th Ed. Philadelphia: Elsevier Saunders, 2005:593. With
AQ1 permission requested.)
10090-08_CH08.qxd 8/31/06 4:51 PM Page 199

Valvular Heart Disease 199

MITRAL VALVE DISEASE


TABLE 8.1. Jones Criteria for Diagnosis of
Rheumatic Fevera Mitral Stenosis
Major criteria Etiology
Carditis
Polyarthritis The most common cause of mitral stenosis
Sydenham chorea (involuntary movements) (MS) is rheumatic fever. Approximately 50%
Erythema marginatum (skin rash with advancing of patients with symptomatic MS provide a
edge and clearing center) history of ARF occurring, on average, 20 years
Subcutaneous nodules
Minor criteria
before presentation. These patients display
Migratory arthralgias typical rheumatic deformity of the valve on
Fever echocardiographic and pathologic examina-
Increased acute phase reactants (ESR, leukocytosis) tions as described below. Other rare causes
Prolonged PR interval on electrocardiogram of MS (less than 1%) include congenital
Evidence of streptococcal infection
stenosis of the mitral valve leaflets, promi-
Antistreptolysin O antibodies
Positive throat culture for streptococci group A
nent calcification extending from the mitral
annulus onto the leaflets in elderly patients,
a
Diagnosis requires evidence of streptococcal infection or endocarditis with very large vegetations
and either: two major criteria, or one major plus two minor
criteria.
that obstruct the valve orifice.
ESR, erythrocyte sedimentation rate.

Pathology
Acute and recurrent inflammation produce
contractility, a pericardial friction rub, a the typical pathologic features of rheumatic
transient murmur of mitral or aortic regur- MS. These include fibrous thickening and cal-
gitation, or a middiastolic murmur at the cification of the valve leaflets, fusion of the
cardiac apex (termed the Carey-Coombs commissures (the borders where the leaflets
murmur). These transient murmurs likely meet), and thickening and shortening of the
reflect turbulent flow across inflamed valve chordae tendineae.
leaflets. Treatment of the acute episode in-
cludes the use of high-dose aspirin to re-
duce inflammation, penicillin to eliminate Pathophysiology
residual streptococcal infection, and ther- In early diastole in the normal heart, the mi-
apy for complications such as congestive tral valve opens and blood flows freely from
heart failure and pericarditis. the left atrium (LA) into the left ventricle
During the chronic phase of this condi- (LV), such that there is a negligible pressure
tion, stenosis or regurgitation of cardiac difference between the two chambers. In
valves is common, most often affecting the MS, however, there is obstruction to blood
mitral valve. Forty percent of patients with flow across the valve such that emptying of
rheumatic heart disease will develop mitral the LA is impeded and there is an abnormal
stenosis. An additional 25% will develop pressure gradient between the LA and LV
aortic stenosis or regurgitation in addition (Figs. 8.2 and 8.3). As a result, the left atrial Fig. 2-3
to the mitral abnormality. Infrequently, the pressure is higher than normal, a necessary
tricuspid valve is affected as well. feature for blood to be propelled forward
Recurrences of ARF in affected patients across the obstructed valve. The normal
can incite further cardiac damage. Therefore, cross-sectional area of the mitral valve ori-
individuals who have experienced ARF fice is 4 to 6 cm2. Hemodynamically signifi-
should receive low-dose penicillin prophy- cant MS becomes apparent when the valve
laxis at least until early adulthood, by which area is reduced to less than 2 cm2. Although
time exposure and susceptibility to strepto- left ventricular pressures are usually normal
coccal infections have diminished. in MS, impaired filling of the chamber across
10090-08_CH08.qxd 8/31/06 4:51 PM Page 200

200 Chapter Eight

Elevated pulmonary
and right-heart pressures

Aorta
Pressure
Volume
LA

LV

NORMAL MITRAL
(DIASTOLE) STENOSIS
Figure 8.2. Pathophysiology of mitral stenosis. In the normal
heart, blood flows freely from the left atrium (LA) into the left ventri-
cle (LV) during diastole. In mitral stenosis, there is obstruction to LA
emptying. Thus, LA pressure increases, which in turn elevates pul-
monary and right-heart pressures.

ECG

OS

Figure 8.3. Hemodynamic profile of mitral stenosis. The left atrial (LA) pressure is
elevated, and there is a pressure gradient (shaded area) between the LA and left ven-
tricle (LV) during diastole. Compare with schematic of normal tracing (see Fig. 2.1). Ab-
normal heart sounds are present: there is a diastolic opening snap (OS) that corre-
sponds to the opening of the mitral valve, followed by a decrescendo murmur. There
is accentuation of the murmur just before S1 owing to the increased pressure gradient
when the LA contracts (presystolic accentuation).
10090-08_CH08.qxd 8/31/06 4:51 PM Page 201

Valvular Heart Disease 201

the narrowed mitral valve may reduce LV Left atrial dilatation stretches the atrial con-
stroke volume and cardiac output. duction fibers and may disrupt the integrity
The high left atrial pressure in MS is pas- of the cardiac conduction system, resulting
sively transmitted to the pulmonary circula- in atrial fibrillation (a rapid irregular heart
tion, resulting in increased pulmonary ve- rhythm; see Chapter 12). Atrial fibrillation
nous and capillary pressures (see Fig. 8.2). causes the cardiac output to fall further in
This elevation of hydrostatic pressure in the MS because the increased heart rate shortens
pulmonary vasculature may cause transuda- diastole. This reduces the time available for
tion of plasma into the lung interstitium and blood to flow across the obstructed mitral
alveoli. The patient may therefore experience valve to fill the left ventricle and also results
dyspnea and other symptoms of congestive in markedly elevated left atrial pressure.
heart failure. In severe cases, significant ele- The relative stagnation of blood flow in
vation of pulmonary venous pressure leads to the dilated left atrium in MS, especially
the opening of collateral channels between when combined with the development of
the pulmonary and bronchial veins. Subse- atrial fibrillation, predisposes to intra-atrial
quently, the high pulmonary vascular pres- thrombus formation. Thromboemboli to pe-
sures may rupture a bronchial vein into the ripheral organs may follow, leading to dev-
lung parenchyma, resulting in coughing up astating complications such as cerebrovas-
of blood (hemoptysis). cular occlusion (stroke). The likelihood of
The elevation of left atrial pressure in MS developing systemic thromboembolic com-
can result in two distinct forms of pul- plications in a patient with MS correlates
monary hypertension: passive and reactive. with the patient’s age and the dimensions of
Most patients with MS exhibit passive pul- the left atrial appendage (a portion of the
monary hypertension, related to the back- left atrium); it correlates inversely with the
ward transmission of the elevated LA pres- patient’s cardiac output. Patients who de-
sure into the pulmonary vasculature. This velop atrial fibrillation are at very high risk
actually represents an “obligatory” increase of stroke and require chronic anticoagula-
in pulmonary artery pressure that develops tion therapy.
to preserve forward flow in the setting of in- The turbulent blood flow across the ob-
creased left atrial and pulmonary venous structed mitral valve in MS predisposes to in-
pressures. Additionally, approximately 40% fective endocarditis (discussed later); however,
of patients with MS demonstrate reactive pul- that complication occurs less frequently in
monary hypertension with medial hypertro- MS than in other forms of acquired valvular
phy and intimal fibrosis of the pulmonary disease.
arterioles. Reactive pulmonary hypertension
can be “beneficial” because the increased ar-
teriolar resistance impedes blood flow into Clinical Manifestations and Evaluation
the engorged pulmonary capillary bed and Presentation
thus reduces capillary hydrostatic pressure
(thereby “protecting” the pulmonary capil- The natural history of MS is variable. The 10-
laries from even higher pressures). However, year survival of untreated patients after onset
this benefit is at the cost of decreased blood of symptoms is 50% to 60%. Survival exceeds
flow through the pulmonary vasculature 80% in asymptomatic or minimally sympto-
with resultant elevation of the right-sided matic patients at 10 years. Longevity is much
heart pressures, as the right ventricle pumps more limited for patients with advanced
against the increased resistance. Chronic el- symptoms and is dismal for those who de-
evation of right ventricular pressure leads to velop significant pulmonary hypertension,
hypertrophy and dilatation of that chamber with a mean survival less than 3 years.
and ultimately to right-sided heart failure. The clinical presentation of MS depends
Chronic pressure overload of the left largely on the degree of reduction in valve
atrium in MS leads to left atrial enlargement. area. The more severe the stenosis, the greater
10090-08_CH08.qxd 8/31/06 4:51 PM Page 202

202 Chapter Eight

the symptoms related to elevation of left sition, causing the closure sound to be loud
atrial and pulmonary venous pressures. The (see Chapter 2). In late stages of the disease,
earliest manifestations are those of dyspnea the intensity of S1 may normalize or become
and reduced exercise capacity. In mild MS, reduced as the valve leaflets thicken, calcify,
dyspnea may be absent at rest; however, it and become immobile.
develops on exertion as LA pressure rises A main feature of auscultation in MS is a
with the exercise-induced increase in blood high-pitched “opening snap” (OS) that fol-
flow through the heart and faster heart rate lows S2. The OS is thought to result from the
(i.e., decreased diastolic filling time). Other sudden tensing of the chordae tendineae
conditions and activities that increase heart and stenotic leaflets on opening of the
rate and cardiac blood flow, and therefore valve. The interval between S2 and the OS re-
precipitate or exacerbate symptoms of MS, lates inversely to the severity of MS. The
include fever, anemia, hyperthyroidism, more severe the MS, the higher the LA pres-
pregnancy, rapid arrhythmias such as atrial sure and the earlier the valve is forced open
fibrillation, exercise, emotional stress, and in diastole. The OS is followed by a low-
sexual intercourse. frequency decrescendo murmur (termed a
With more severe MS (i.e., a smaller valve diastolic rumble) caused by turbulent flow
area) dyspnea occurs even at rest. Increasing across the stenotic valve during diastole (see
fatigue and more-severe signs of pulmonary Fig. 8.3). The duration, but not the inten-
congestion, such as orthopnea and paroxys- sity, of the diastolic murmur relates to the
mal nocturnal dyspnea, occur. With ad- severity of MS. The more severe the stenosis,
vanced MS and pulmonary hypertension, the longer it takes for the LA to empty and
signs of right-sided heart failure ensue, in- for the gradient between the LA and LV to
cluding jugular venous distention, hepato- dissipate. Near the end of diastole, contrac-
megaly, ascites, and peripheral edema. Com- tion of the LA causes the pressure gradient
pression of the recurrent laryngeal nerve by between the LA and LV to rise again (see Fig.
the enlarged pulmonary artery or left atrium 8.3); therefore, the murmur briefly becomes
may cause hoarseness. louder (termed presystolic accentuation). This
Less often, the diagnosis of MS is heralded final accentuation of the murmur does not
by one of its complications: atrial fibrilla- occur if atrial fibrillation has developed, be-
tion, thromboembolism, infective endocardi- cause there is no effective atrial contraction
tis, or hemoptysis, as described in the earlier in that situation.
section on pathophysiology. Murmurs caused by other valvular lesions
are often found concurrently in patients
with MS. For example, mitral regurgitation
Examination
(discussed later in the chapter) frequently
On examination, there are several typical coexists with MS. Additionally, right-sided
findings of MS. Palpation of the left anterior heart failure caused by severe MS may in-
chest may reveal a right ventricular “tap” in duce tricuspid regurgitation as a result of to
patients with increased right ventricular pres- right ventricular enlargement. A diastolic
sure. Auscultation discloses a loud S1 (the first decrescendo murmur along the left sternal
heart sound, which is associated with mitral border may be present owing to coexistent
valve closure) in the early stages of the dis- aortic regurgitation (because of rheumatic
ease. The increased S1 results from the high involvement of the aortic leaflets) or pul-
pressure gradient between the atrium and monic regurgitation (because of MS-induced
ventricle, which keeps the mobile portions of pulmonary hypertension).
the mitral valve leaflets widely separated The electrocardiogram in MS routinely
throughout diastole; at the onset of systole, shows left atrial enlargement and, if pul-
ventricular contraction abruptly slams the monary hypertension has developed, right
leaflets together from the relatively wide po- ventricular hypertrophy. Atrial fibrillation 1 LINE SHORT
10090-08_CH08.qxd 8/31/06 4:51 PM Page 203

Valvular Heart Disease 203

may be present. The chest radiograph reveals is recommended for patients with MS with
left atrial enlargement, pulmonary vascular atrial fibrillation or if previous embolic
redistribution, interstitial edema, and Kerley episodes have occurred.
B lines resulting from edema within the pul- If symptoms of MS persist despite diuretic
monary septae (see Chapter 3). With the therapy and control of rapid heart rates, me-
development of pulmonary hypertension, chanical correction of the stenosis is war-
right ventricular enlargement and promi- ranted. Percutaneous balloon mitral valvulo-
nence of the pulmonary arteries also appear. plasty was first introduced in 1985 and is
Echocardiography is of major diagnostic now the treatment of choice for MS in ap-
value in MS. It reveals thickened mitral propriately selected patients. During this
leaflets and abnormal fusion of their com- procedure, a balloon catheter is advanced
missures with restricted separation during from the femoral vein into the right atrium,
diastole. Left atrial enlargement can be as- across the atrial septum (by creating a small
sessed, and if present, intra-atrial throm- septal defect there), and advanced through
bus may be visualized. The mitral valve area the narrowed mitral valve orifice. The bal-
can be measured directly on cross-sectional loon is then rapidly inflated, thereby “crack-
views or calculated from Doppler-echocar- ing” open the fused commissures. The pro-
diographic velocity measurements. Patients cedure is safest and most effective in the
can be stratified into groups of disease sever- absence of complicating features, such as
ity based partly on the mitral valve area. A mitral regurgitation, extensive valve calcifi-
normal mitral valve orifice measures be- cation, or atrial thrombus. The results of this
tween 4 and 6 cm2. A reduced mitral valve procedure in randomized trials compare fa-
area of ≤2 cm2 correlates with mild MS, and vorably with those of surgical treatment.
a valve area of 1 to 1.5 cm2 correlates with Approximately 5% of patients undergoing
moderate MS. Severe MS is defined by a valve balloon mitral valvuloplasty are left with
area of ≤1 cm2. Although cardiac catheteri- a residual atrial septal defect. Less frequent
zation is not necessary to confirm the diag- complications include cerebral emboli at
nosis of MS, it is sometimes performed to the time of valvuloplasty, cardiac perfora-
calculate the valve area by direct hemody- tion, or the creation of mitral regurgitation
namic measurements and to clarify whether requiring subsequent surgical repair. The
significant mitral regurgitation, pulmonary estimated event-free survival 7 years after val-
hypertension, or coronary artery disease is vuloplasty is 67% to 76%.
present. Surgical options are undertaken for cor-
recting MS in individuals whose anatomy is
Treatment not ideal for balloon valvuloplasty. These
techniques include open mitral commissu-
Therapy of MS includes long-term penicillin
rotomy (an operation in which the stenotic
to prevent recurrent ARF in young people
commissures are separated under direct
and, in all patients, intermittent antibiotic
visualization), and in severe disease, mitral
prophylaxis against infective endocarditis
valve replacement. Open MV commissu-
(discussed later in the chapter). Diuretics are
rotomy is effective, and restenosis occurs
prescribed to treat symptoms of vascular
in <20% of patients over 10 to 20 years of
congestion. If atrial fibrillation has devel-
follow-up.
oped, digoxin may be useful to slow the
rapid ventricular rate and thereby improve di-
astolic LV filling (see Chapter 17). β-Blockers, Mitral Regurgitation
or the calcium channel antagonists vera-
Etiology
pamil or diltiazem, are even more effective
alternatives to slow the heart rate. Anticoag- Normal closure of the mitral valve during
LINE SHORT ulant therapy (to prevent thromboembolism) systole requires the coordinated action of
10090-08_CH08.qxd 8/31/06 4:51 PM Page 204

204 Chapter Eight

each component of the valve apparatus. leaflets, interfering with their excursion and
Therefore, mitral regurgitation (MR) may re- systolic closure.
sult from structural abnormalities of the mi- Primary (idiopathic) rupture of chordae
tral annulus, the valve leaflets, the chordae tendineae is associated with severe acute
Fig. 4 tendineae, or the papillary muscles (Fig. 8.4). valvular incompetence. Ischemic heart dis-
For example, myxomatous degeneration of ease may scar or cause transient dysfunction
the valve (the etiology of mitral valve pro- of a papillary muscle, interfering with valve
lapse) causes MR because enlarged, redun- closure. Marked left ventricular enlargement
dant leaflets bow excessively into the LA of any cause results in MR because of two
during systole rather than opposing each mechanisms that interfere with mitral leaflet
other normally. Infective endocarditis can closure: (1) the spatial separation between the
result in MR because of leaflet perforation or papillary muscles is augmented, and (2) the
rupture of infected chordae. Rheumatic mitral annulus is stretched to an increased
fever may lead to MS, as already discussed, diameter.
or primarily MR if excessive shortening of During the 1990s, the commonly used
the chordae tendineae and retraction of the weight-loss drug combination of fenflu-
leaflets occur. Hypertrophic obstructive car- ramine and phentermine was, in some pa-
diomyopathy (see Chapter 10) is associated tients, associated with the development of
with abnormal systolic anterior motion of thickened plaques on the cardiac valves.
the anterior mitral leaflet, which prevents These patients were prone to develop MR, as
normal valve closure and results in signifi- well as aortic regurgitation and tricuspid
cant MR in 50% of patients. Calcification of valve disease. Thus, that drug combination
the mitral annulus can occur with normal is no longer used.
aging but is more common among patients
with hypertension, diabetes, or end-stage
Pathophysiology
renal disease. Such calcification impairs the
normal movement of the annulus and im- In MR, a portion of the left ventricular stroke
mobilizes the basal portion of the valve volume is ejected backward into the low-

Mitral annulus
Left atrium • Annular calcification

Leaflets
• Myxomatous degeneration (“MVP”)
• Rheumatic disease
• Endocarditis
• SAM (hypertrophic cardiomyopathy)

Chordae tendineae
• Rupture (idiopathic)
• Endocarditis

Papillary muscles
• Dysfunction or rupture

Left ventricle
• Cavity dilatation

Figure 8.4. The mitral valve apparatus and associated common etiologies of
mitral regurgitation. MVP, mitral valve prolapse; SAM, systolic anterior motion.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 205

Valvular Heart Disease 205

Pulmonary
edema

Dilated LA
High LA with less
pressure elevated
pressure

Figure 8.5. Pathophysiology of mitral regurgitation. In the normal heart, left ventricular (LV) contraction
during systole forces blood exclusively through the aortic valve into the aorta; the closed mitral valve prevents
regurgitation into the left atrium (LA). In mitral regurgitation (MR), a portion of LV output is forced backward
into the LA, so that forward cardiac output into the aorta is reduced. In acute MR, the LA is of normal size and
is relatively noncompliant, such that the LA pressure rises significantly and pulmonary edema may result. In
chronic MR, the LA has enlarged and is more compliant, so that LA pressure is less elevated and pulmonary
congestive symptoms are less common. LV enlargement and eccentric hypertrophy result from the chronically
elevated volume load.

Fig. 5 pressure LA (Fig. 8.5) during systole. As a re- and LA, (3) the systemic vascular resistance
sult, the forward cardiac output (into the opposing forward LV blood flow, (4) the left
aorta) is less than the lLV’s total output (for- atrial compliance, and (5) the duration of re-
ward flow plus + backward leak). Therefore, gurgitation with each systolic contraction.
the direct consequences of MR include (1) The regurgitant fraction in MR is defined as
an elevation of the left atrial volume and follows:
pressure, (2) a reduction of forward cardiac
Volume of MR
output, and (3) a volume-related stress on
Total LV stroke volume
the LV because the regurgitated volume re-
turns to the LV in diastole along with the and this ratio rises whenever the resistance
normal pulmonary venous return. To meet to aortic outflow is increased (i.e., blood fol-
normal circulatory needs and to eject the lows the path of least resistance). For exam-
additional volume, LV stroke volume must ple, high systemic blood pressure or the
rise. This increase is accomplished by the presence of aortic stenosis will increase the
Frank-Starling mechanism (see Chapter 9), regurgitant fraction. The extent to which left
whereby the elevated LV diastolic volume atrial pressure rises in response to the regur-
augments myofiber stretch and stroke vol- gitant volume is determined by the left atrial
ume with each contraction. The subsequent compliance. Compliance is a measure of the
hemodynamic consequences of MR vary de- chamber’s pressure-volume relationship, re-
pending on the degree of regurgitation and flecting the ease or difficulty with which the
how long it has been present. chamber can be filled (see Table 9.1).
The severity of MR and the ratio of for- In acute MR (e.g., owing to sudden rup-
ward cardiac output to backward flow are ture of chordae tendineae), left atrial com-
dictated by five factors: (1) the size of the pliance undergoes little immediate change.
mitral orifice during regurgitation, (2) the Because the LA is a relatively stiff chamber,
systolic pressure gradient between the LV its pressure increases substantially when it
10090-08_CH08.qxd 8/31/06 4:51 PM Page 206

206 Chapter Eight

is suddenly exposed to the regurgitant vol- In acute MR, the LV accommodates the
ume (see Fig. 8.5). This elevated pressure increased volume load returning from the
serves to prevent further regurgitation; how- LA according to the Frank-Starling relation-
ever, the high pressure is also transmitted ship. The result is a compensatory increase
backward to the pulmonary circulation. in the LV stroke volume, such that at the
Therefore, acute MR can result in rapid pul- end of each systolic contraction, LV volume
monary congestion and edema, a medical remains normal in the nonfailing heart. Sys-
emergency. tolic emptying of the ventricle is facilitated
In acute MR, the LA pressure, or the pul- in MR by the reduced total impedance to
monary capillary wedge pressure (an indi- LV contraction (i.e., the afterload is lower
rect measurement of LA pressure; see Chap- than normal), which is caused by a portion
ter 3), demonstrates a prominent v wave of the LV output being directed into the rel-
(often referred to as a cv wave when it is so atively low-impedance left atrium, rather
prominent that it merges with the preced- than into the higher-pressure aorta as in
ing c wave), reflecting the increased LA normal outflow.
Fig. 6 filling during systole (Fig. 8.6). Additionally, In contrast to the acute situation, the more
as in MS, pulmonary artery and right-heart gradual development of chronic MR (e.g.,
pressures passively rise such that forward owing to rheumatic valve disease) permits
flow through the heart is maintained. the LA to undergo compensatory changes
that lessen the effects of regurgitation on
the pulmonary circulation (see Fig. 8.5). In
ECG particular, the LA dilates and its compliance
increases such that the chamber is able to
accommodate a larger volume without a
substantial increase in pressure. Left atrial
dilatation is therefore adaptive in that it pre-
vents significant increases in pulmonary
Tall v vascular pressures. However, this adaptation
wave
occurs at the cost of inadequate forward car-
diac output, because the compliant LA be-
comes a preferred low-pressure “sink” for
left ventricular ejection, compared with the
greater impedance of the aorta. Conse-
quently, as progressively larger fractions of
blood regurgitate into the LA, the main
symptoms of chronic MR become those of
low forward cardiac output (e.g., weakness
and fatigue). In addition, chronic left atrial
dilatation predisposes to the development
of atrial fibrillation.
In chronic MR, the left ventricle also un-
Figure 8.6. Hemodynamic profile of mitral dergoes gradual compensatory dilatation in
regurgitation (MR). A large systolic v wave is
noted in the left atrial (LA) pressure tracing. A response to the volume load (through eccen-
holosystolic murmur is present in chronic MR (as tric hypertrophy; see Chapter 9). Compared
shown here), beginning at the first heart sound with acute MR, the increased ventricular
(S1) and continuing through the second heart
sound (S2). In severe acute MR, the systolic mur- compliance accommodates the augmented
mur may actually have a decrescendo qual- filling volume with relatively normal dias-
ity, reflecting rapid equilibration of LV and LA tolic pressures. Forward output in chronic
pressures owing to the relatively reduced LA
compliance. ECG, electrocardiogram; LV, left MR is preserved to near-normal levels for
ventricle. an extended period by maintaining a high 1 LINE SHORT
10090-08_CH08.qxd 8/31/06 4:51 PM Page 207

Valvular Heart Disease 207

stroke volume via the Frank-Starling mech- best heard along the left sternal edge or in
anism. Over years, however, the chronic the aortic area (see Chapter 2) and could be
volume overload results in deterioration of confused with the murmur of aortic stenosis
systolic function, a decline in forward out- (AS). Fortunately, the distinction between
put, and symptoms of heart failure. the systolic murmur of MR and that of AS
In summary, the main differences be- can be made by simple bedside maneuvers.
tween acute and chronic MR relate to a great If the patient is instructed to clench the fists,
extent on left atrial size and compliance (see systemic vascular resistance will increase,
Fig. 8.5): and the severity of MR and its murmur will
intensify, whereas the murmur of AS will
Acute MR: Normal LA size and compliance
not. Even more helpful in this distinction is
→ High LA pressure → High pulmonary
to notice the effect of varying cardiac cycle
venous pressure → Pulmonary conges-
length (the time between consecutive heart
tion and edema
beats) on the intensity of the systolic mur-
Chronic MR: Increased LA size and com- mur. In a patient with atrial fibrillation or
pliance → More normal LA and pul- with frequent premature beats, the LV fills
monary venous pressures, but low for- to a degree that directly depends on the pre-
ward cardiac output ceding cycle length (i.e., a longer cycle
length permits greater left ventricular fill-
Clinical Manifestations and Evaluation ing). The systolic murmur of AS becomes
louder in the beat after a long cycle length
Presentation
because even small pressure gradients are
As should be clear from the pathophysiol- amplified as more blood is ejected across the
ogy discussion, patients with acute MR usu- reduced aortic orifice. In MR, however, the
ally present with symptoms of pulmonary intensity of the murmur does not vary sig-
edema (see Chapter 9). The symptoms of nificantly because the change in the LV-to-
chronic MR are predominantly owing to low LA pressure gradient is minimally affected
cardiac output, especially during exertion, by alterations in the cycle length.
and consist of fatigue and weakness. Pa- In patients with severe acute MR, the sys-
tients with severe MR or those who develop tolic murmur is often different, reflecting
LV contractile dysfunction often complain the underlying pathophysiology. In this case,
of dyspnea, orthopnea, and/or paroxysmal the murmur may have a decrescendo quality,
nocturnal dyspnea. In severe chronic MR, reflecting the rapid equilibration between
symptoms of right-heart failure (e.g., in- LV and LA pressures in systole caused by the
creased abdominal girth, peripheral edema) relatively reduced LA compliance.
may develop as well. In addition to the systolic murmur, a
common finding in chronic MR is the pres-
ence of an S3, which reflects increased vol-
Examination
ume returning to the LV in early diastole
The physical examination of a patient with (see Chapter 2). In chronic MR, the palpated
chronic MR reveals an apical holosystolic cardiac apical impulse is often laterally dis-
(also termed pansystolic) murmur that radi- placed toward the axilla because of LV en-
ates to the axilla (see Fig. 8.6). This descrip- largement.
tion, accurate for rheumatic MR, has some The chest radiograph may display pulmo-
exceptions. For example, when ischemic nary edema in acute MR but in chronic
papillary muscle dysfunction interferes with asymptomatic MR more likely demonstrates
normal mitral valve closure, the regurgitant left ventricular and atrial enlargement, with-
jet may be directed toward the anterior left out pulmonary congestion. Calcification of
atrial wall, immediately posterior to the the mitral annulus may be seen if that is the
LINE SHORT aorta. In this setting, the murmur may be cause of the MR. In chronic MR, the electro-
10090-08_CH08.qxd 8/31/06 4:51 PM Page 208

208 Chapter Eight

cardiogram typically demonstrates left atrial currently the preferred operative technique
enlargement and signs of left ventricular hy- for appropriately selected patients. In the
pertrophy. Echocardiography can often iden- past, the operative mortality and the draw-
tify the structural cause of MR and grade its backs associated with the use of prosthetic
severity by color Doppler analysis. Left ven- valves were motivations for delaying surgery
tricular size and function (usually vigorous as long as possible. Studies showed that sur-
in the “compensated” heart because of the vival after mitral valve replacement was not
increased stroke volume) can be observed. clearly better than the natural history of the
Cardiac catheterization is useful for identify- disease, even though symptomatic improve-
ing a coronary ischemic cause (i.e., papillary ment was the rule. Fortunately, mitral repair
muscle dysfunction) and for grading the preserves native valve tissue, is associated
severity of MR. The characteristic hemody- with less impairment of postoperative LV
namic abnormality is a large v wave on the function, and eliminates many of the prob-
pulmonary capillary wedge pressure (reflect- lems of artificial valves.
ing LA pressure) tracing. Mitral repair involves the reconstruction
of parts of the valve responsible for the re-
gurgitation. For example, a perforated leaflet
Natural History and Treatment
may be patched with transplanted autolo-
The natural history of chronic MR is related gous pericardium, or ruptured chordae may
to its underlying cause. For example, in be reattached to a papillary muscle. In pa-
rheumatic heart disease, the course is one of tients who undergo a repair operation, the
very slow progression with a 15-year sur- postoperative survival rate appears to be bet-
vival rate of 70%. On the other hand, abrupt ter than the natural history of MR and has
worsening of chronic MR of any cause provided impetus toward earlier surgical in-
can occur with superimposed complica- tervention.
tions, such as rupture of chordae tendineae The operative mortality rate is approxi-
or endocarditis, and can result in an imme- mately 2% to 4% for mitral valve repair and
diate life-threatening situation. 5% to 7% for mitral replacement. These rates
Medical therapy of MR involves aug- are higher if concurrent coronary artery by-
menting forward cardiac output while re- pass grafting is performed. In general, mitral
ducing regurgitation into the LA and reliev- valve repair is more often appropriate for
ing pulmonary congestion. In acute MR younger patients with myxomatous in-
with heart failure, treatment includes intra- volvement of the mitral valve, and mitral
venous diuretics to relieve pulmonary edema replacement is more often undertaken in
and vasodilators (e.g., intravenous sodium older patients with more extensive valve
nitroprusside) to reduce the resistance to pathology.
forward flow and augment forward cardiac
output. In chronic MR, vasodilators are less
Mitral Valve Prolapse
useful and indicated only for the treatment
of accompanying hypertension or LV sys- Mitral valve prolapse (MVP) is a common
tolic dysfunction, because these drugs have and usually asymptomatic billowing of the
not been shown to delay the need for surgi- mitral leaflets into the LA during ventricular
cal correction. systole, sometimes accompanied by MR
Because chronic MR produces continu- (Fig. 8.7). Other names for this condition in- Fig. 7
ous left ventricular volume overload, it can clude floppy mitral valve, myxomatous mi-
slowly result in left ventricular contractile tral valve, or Barlow syndrome. MVP may be
impairment and, ultimately, heart failure. inherited as a primary autosomal dominant
Mitral valve surgery should be performed disorder with variable penetrance or may
before this deterioration occurs. Mitral valve occur as a part of other connective tissue dis-
repair (reconstruction of the native valve) is eases, such as Marfan syndrome or Ehlers- 1 LINE SHORT
10090-08_CH08.qxd 8/31/06 4:51 PM Page 209

Valvular Heart Disease 209

AO
LA

RV

LV

Figure 8.7. Mitral valve prolapse. Long axis view of the left
ventricle (LV) demonstrates myxomatous, elongated appear-
ance of the mitral valve with prolapse of the posterior leaflet
(arrow) into the left atrium (LA). Ao, aorta; RV, right ventricle.
(From Schoen FJ. The heart. In: Kumar V, Abbas A, Fausto N,
eds. Robbins and Cotran Pathologic Basis of Disease. 7th Ed.
Philadelphia: Elsevier Saunders, 2005:592. With permission
AQ2 requested.)

Danlos syndrome. Pathologically, the valve chordae tendineae as the leaflet is forced
leaflets, particularly the posterior leaflet, are back toward the left atrium; the murmur
enlarged, and the normal dense collagen corresponds to regurgitant flow through the
and elastin matrix of the valvular fibrosa is incompetent valve. The click and murmur
fragmented and replaced with loose myxo- are characteristically altered during dy-
matous connective tissue. Additionally, in namic auscultation at the bedside: maneu-
more severe lesions, elongated or ruptured vers that increase the volume of the LV (e.g.,
chordae, annular enlargement, or thickened sudden squatting, which increases venous
leaflets may be present. A recent rigorous return) delay the occurrence of prolapse in
echocardiographic study indicated that MVP systole and cause the click and murmur to
occurs in about 2% of the population and occur later (i.e., further from S1). Con-
is more common among women, especially versely, if the volume of blood in the LV is
those with thin, lean bodies. decreased (e.g., on sudden standing), pro-
MVP is often asymptomatic, but affected lapse occurs more readily and the click
individuals may describe chest pain or pal- and murmur occur earlier in systole (closer
pitations because of associated arrhythmias. to S1).
Most often it is identified on routine physi- Confirmation of the diagnosis is obtained
cal examination by the presence of a mid- by echocardiography, which demonstrates pos-
systolic click and late systolic murmur heard terior displacement of one or both mitral
best at the cardiac apex. The midsystolic leaflets into the left atrium during systole.
click is thought to correspond to the sudden The electrocardiogram and chest radiograph are
LINE SHORT tensing of the involved mitral leaflet or usually normal unless chronic MR has re-
10090-08_CH08.qxd 8/31/06 4:51 PM Page 210

210 Chapter Eight

sulted in left atrial and left ventricular en- damage, causing calcification of an other-
largement. wise normal trileaflet valve. However, there
The clinical course of MVP is most often is also evolving evidence of a common eti-
benign. Treatment consists of reassurance ology with atherosclerotic vascular disease.
about the usually good prognosis and anti- Studies have shown that as in atherosclero-
biotic prophylaxis (to prevent endocarditis) sis, the valve tissue of patients with this
only if substantial valve thickening or MR are form of AS display cellular proliferation, in-
present. Of the potential complications, the flammation, lipid accumulation, and in-
most common is the development of gradu- creased margination of macrophages and T
ally progressive MR. Occasionally, rupture of lymphocytes.
myxomatous chordae can cause sudden, se- In the case of a congenitally deformed
vere regurgitation and pulmonary edema. valve, years of turbulent flow across the valve
Other rare complications include infective disrupt the endothelium and collagen ma-
endocarditis, peripheral emboli owing to mi- trix of the leaflets, resulting in gradual cal-
crothrombus formation behind the redun- cium deposition. In rheumatic AS, endocar-
dant valve tissue, and atrial or ventricular ar- dial inflammation leads to organization and
rhythmias. fibrosis of the valve, and ultimately fusion
of the commissures and the formation of
calcified masses within the aortic cusps.
AORTIC VALVE DISEASE
Aortic Stenosis Pathophysiology
Etiology In AS, blood flow across the aortic valve is
Aortic stenosis (AS) is often caused by age- impeded during systole (Fig. 8.8). When the Fig. 8
related degenerative calcific changes of the valve, valve orifice area is reduced by more than
formerly termed senile AS. Calcific changes 50% of its normal size, significant elevation
that progress to AS may also develop in pa- of left ventricular pressure is necessary to
tients with congenitally deformed aortic valves drive blood into the aorta (Fig. 8.9). In ad- Fig. 9
(about 1% to 2% of the population is born vanced AS, it is common to measure peak
with an abnormal bicuspid aortic valve).
Most patients who present with AS after the
age of 65 have the age-related form, whereas
younger patients usually have calcification
of a congenitally bicuspid valve. AS may also
result from chronic rheumatic valve disease,
although the prevalence of this condition
has decreased dramatically in recent decades
in the United States. Approximately 95% of
patients who are found to have rheumatic Pressure
AS have coexisting rheumatic involvement
of the mitral valve.

Pathology
Figure 8.8. Pathophysiol-
The pathologic appearance in AS is depen- ogy of aortic stenosis (AS).
dent on its etiology. In age-related degener- The impediment to left ven-
ative AS, the classic teaching is that cumula- tricular (LV) outflow in AS
results in elevated LV pres-
tive “wear and tear” of valve motion over sures and secondary ventricu-
many years leads to endothelial and fibrous lar hypertrophy. 1 LINE SHORT
10090-08_CH08.qxd 8/31/06 4:51 PM Page 211

Valvular Heart Disease 211

Three major manifestations occur in pa-


ECG
tients with advanced AS: (1) angina, (2) ex-
150 ertional syncope, and (3) congestive heart
failure, all of which can be explained on the
130 basis of the underlying pathophysiology.
Each manifestation, in order, heralds an in-
110
creasingly ominous prognosis (Table 8.2). Tab. 2
AS may result in angina because it creates
a substantial imbalance between myocar-
dial oxygen supply and demand. Myocar-
dial oxygen demand is increased in two
50
ways. First, the muscle mass of the hyper-
30 trophied LV is increased, requiring greater-
than-normal perfusion. Second, wall stress
10 is increased because of the elevated systolic
ventricular pressure. In addition, AS reduces
myocardial oxygen supply because the ele-
vated left ventricular diastolic pressure re-
duces the coronary perfusion pressure gradi-
ent between the aorta and the myocardium.
AS may cause syncope during exertion. Al-
Figure 8.9. Hemodynamic profile of aortic
stenosis. A systolic pressure gradient (shaded though left ventricular hypertrophy allows
area) is present between the left ventricle (LV) the chamber to generate a high pressure
and aorta. The second heart sound (S2) is dimin- and maintain a normal cardiac output at
ished in intensity, and there is a crescendo–de-
crescendo systolic murmur that does not extend rest, the ventricle cannot significantly in-
beyond S2. ECG, electrocardiogram. crease its cardiac output during exercise
because of the fixed stenotic aortic orifice.
systolic pressure gradients >100 mm Hg be- In addition, exercise leads to vasodilatation
tween the LV and the aorta. of the peripheral muscle beds. Thus, the
Since AS develops over a chronic course, combination of peripheral vasodilatation
the LV is able to compensate by undergoing and the inability to augment cardiac output
concentric hypertrophy in response to the contributes to decreased cerebral perfusion
high systolic pressure it must generate. Such pressure and, potentially, loss of conscious-
hypertrophy serves an important role in re- ness on exertion.
ducing ventricular wall stress (remember Finally, AS can result in symptoms of
from Chapter 6 that wall stress = (P × r) ÷ 2h, congestive heart failure. Early in the course
in which h represents wall thickness); how- of AS, an abnormally increased left atrial
ever, it also reduces the compliance of the pressure occurs primarily at the end of
ventricle. The resulting elevation of diastolic
LV pressure also causes the LA to hypertro-
phy in order to fill the “stiff” LV. Whereas TABLE 8.2. Median Survival Time in
left atrial contraction contributes only a Symptomatic Aortic Stenosis
small portion of the left ventricular stroke
volume in normal individuals, it may pro- Clinical Symptoms Median Survival
vide more than 25% of the stroke volume to Angina 5 years
the stiffened LV in AS patients. Thus, left Syncope 3 years
atrial hypertrophy is beneficial, and the loss Congestive heart failure 2 years
of effective atrial contraction (e.g., the de- Atrial fibrillation 6 months
velopment of atrial fibrillation) can cause Reprinted with permission from Ross J Jr, Braunwald E. Aortic
LINE SHORT marked clinical deterioration. stenosis. Circulation 1968;38(suppl v):61.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 212

212 Chapter Eight

diastole, when the LA contracts into the On the electrocardiogram, left ventricular
thickened noncompliant LV. As a result, hypertrophy is common in advanced AS, but
the mean left atrial pressure and the pul- echocardiography is a more sensitive tech-
monary venous pressure are not greatly af- nique to assess LV wall thickness. The trans-
fected early in the disease. However, with valvular pressure gradient and aortic valve
progression of the stenosis, the LV may de- area can be calculated by Doppler echocar-
velop contractile dysfunction because of diography (see Chapter 3). Cardiac catheteri-
the insurmountably high afterload, lead- zation is sometimes used to confirm the sever-
ing to increased left ventricular diastolic ity of AS and to define the coronary anatomy,
volume and pressure. The accompanying because concurrent coronary artery bypass
marked elevation of LA and pulmonary ve- surgery is often necessary at the time of aor-
nous pressures incites pulmonary alveolar tic valve replacement in patients with co-
congestion and the symptoms of congestive existing coronary disease.
heart failure.
The normal aortic valve cross-sectional
area is 3 to 4 cm2. When the valve area is re- Natural History and Treatment
duced to less than 2 cm2, a pressure gradient
The natural history of severe, symptomatic,
between the LV and aorta first appears (mild
uncorrected AS is very poor. Data from the
AS). Moderate AS is characterized by a valve
Mayo Clinic indicate that the 1-year sur-
area of 1.0 to 1.5 cm2. When the aortic valve
vival rate is 57% for patients with this con-
area is reduced to less than 1.0 cm2, severe
dition. The only effective treatment for ad-
valve obstruction is said to be present.
vanced AS is surgical replacement of the
valve.
Clinical Manifestations and Evaluation Aortic valve replacement (AVR) is indi-
cated when patients with severe AS develop
Presentation
symptoms, or when there is evidence of pro-
Angina, syncope, and congestive heart fail- gressive LV dysfunction in the absence of
ure may appear after many asymptomatic symptoms. The left ventricular ejection frac-
years of slowly progressive valve stenosis. tion almost always increases after valve re-
Once these symptoms develop, they confer placement, even in patients with impaired
a significantly decreased survival if surgical preoperative left ventricular function. The
correction of AS is not undertaken (see effect of AVR on the natural history of AS is
Table 8.2). dramatic, with the 10-year survival rate ex-
ceeding 75%.
Examination Unlike its successful role in mitral steno-
sis, percutaneous valvuloplasty has been dis-
Physical examination often permits accu-
appointing in the treatment of AS in adults.
rate detection and estimation of the sever-
Although balloon dilatation of the aortic
ity of AS. The key features of advanced
valve orifice can fracture calcified commis-
AS include (1) a coarse late-peaking systo-
lic ejection murmur and (2) a weakened sures leading to some immediate relief of
(parvus) and delayed (tardus) upstroke of outflow obstruction, up to 50% of patients
the carotid artery pulsations owing to the develop restenosis within 6 months. Valvu-
obstructed LV outflow. Other common loplasty is occasionally a suitable option for
findings on cardiac examination include patients who are poor surgical candidates or
the presence of an S4 (because of atrial con- as a temporizing measure in patients too ill
traction into the “stiff” LV) and reduced in- to proceed directly to valve replacement.
tensity, or complete absence, of the aortic Valvuloplasty is also sometimes effective in
component of the second heart sound (see young patients with congenitally bicuspid
Fig. 8.9). valves.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 213

Valvular Heart Disease 213

Mild, asymptomatic AS has a slow rate of (Fig. 8.10). In acute AR, the LV is of normal Fig. 10
progression such that over a 20-year period, size and relatively noncompliant. Thus, the
only 20% of patients will progress to severe or volume load of regurgitation causes the LV
symptomatic AS. Medical therapy for asymp- diastolic pressure to rise substantially. The
tomatic AS includes endocarditis antibiotic sudden high diastolic LV pressure is trans-
prophylaxis and cautious use of medica- mitted to the LA and pulmonary circulation,
tions that could result in hypotension in often producing dyspnea and pulmonary
this setting (e.g., vasodilators, diuretics, nitro- edema. Thus, severe acute AR is usually a sur-
glycerin). gical emergency, requiring immediate valve
replacement.
In chronic AR, the LV undergoes compen-
Aortic Regurgitation satory adaptation in response to the long-
standing regurgitation. AR subjects the LV
Etiology
primarily to volume overload but also to an
Aortic regurgitation (AR), also termed aortic excessive pressure load; therefore, the ven-
insufficiency, may result from (1) diseases of tricle compensates through dilatation and,
the aortic leaflets or (2) dilatation of the aor- to a lesser degree, both eccentric and con-
tic root. The most common causes of AR are centric hypertrophy. Over time, the dilata-
Tab. 3 listed in Table 8.3. tion increases the compliance of the LV and
allows it to accommodate a larger regurgitant
volume with less of an increase in diastolic
Pathophysiology pressure, reducing the pressure transmitted
In AR, abnormal regurgitation of blood from into the LA and pulmonary circulation. How-
the aorta into the LV occurs during diastole. ever, by accommodating the large regur-
Therefore, with each contraction, the LV gitant volume, the aortic (and therefore
must pump the regurgitant volume plus the systemic arterial) diastolic pressure drops
normal amount of blood returning from substantially. The combination of a high LV
the LA. Hemodynamic compensation relies stroke volume (and therefore high systolic
on the Frank-Starling mechanism to aug- arterial pressure) with a reduced aortic dias-
ment stroke volume. Factors influencing the tolic pressure produces a widened pulse pres-
severity of AR are analogous to those of MR: sure (the difference between arterial systolic
(1) the size of the regurgitant aortic orifice, and diastolic pressures), a hallmark of
(2) the pressure gradient across the aortic chronic AR (Fig. 8.11). As a result of the de- Fig. 11
valve during diastole, and (3) the duration creased aortic diastolic pressure, the coro-
of diastole. nary artery perfusion pressure falls, poten-
As in MR, the hemodynamic abnormalities tially reducing myocardial oxygen supply.
and symptoms differ in acute and chronic AR This, coupled with the increase in LV size
(which causes increased wall stress and myo-
cardial oxygen demand), can produce angina,
even in the absence of atherosclerotic coro-
TABLE 8.3. Causes of Aortic Regurgitation
nary disease.
Abnormalities of valve leaflets Because compensatory left ventricular
1. Congenital (bicuspid valve) dilatation and hypertrophy are generally
2. Endocarditis
adequate to meet the demands of chro-
3. Rheumatic
Dilatation of aortic root nic AR, affected patients are usually asymp-
1. Aortic aneurysm (inflammation; connective tomatic for many years. Gradually, how-
tissue disease, e.g., Marfan syndrome) ever, progressive remodeling of the LV
2. Aortic dissection occurs, resulting in systolic dysfunction.
3. Annuloaortic ectasia
This causes decreased forward cardiac out-
4. Syphilis
put as well as an increase in left atrial and
10090-08_CH08.qxd 8/31/06 4:51 PM Page 214

214 Chapter Eight

Pulmonary
congestion

Pressure
Pressure N-

Pressure Pressure
N-

Figure 8.10. Pathophysiology of acute and chronic aortic regur-


gitation (AR). Abnormal regurgitation of blood from the aorta into
the left ventricle (LV) is shown in each schematic drawing (large ar-
rows). In acute AR, the LV is of normal size and relatively low compli-
ance, such that its diastolic pressure rises significantly; this pressure in-
crease is reflected back to the left atrium (LA) and pulmonary
vasculature, resulting in pulmonary congestion or edema. In chronic
AR, adaptive LV and LA enlargement have occurred, such that a
greater volume of regurgitation can be accommodated with less of an
increase in diastolic LV pressure, so that pulmonary congestion is less
likely. N, normal.

pulmonary vascular pressures. At that point, after exhaling. In addition, a low-frequency


the patient develops symptoms of heart middiastolic rumbling sound may be aus-
failure. cultated at the cardiac apex in some pa-
tients with severe AR. Known as the Austin
Flint murmur, it is thought to reflect turbu-
Clinical Manifestations and Assessment lent blood flow across the mitral valve
Presentation during diastole owing to downward dis-
placement of the mitral anterior leaflet by
Common symptoms of chronic AR include the stream of AR. It can be distinguished
dyspnea on exertion, fatigue, decreased ex- from the murmur of MS by the absence of
ercise tolerance, and the uncomfortable sen- an opening snap or presystolic accentua-
sation of a forceful heartbeat associated with tion of the murmur.
high pulse pressure. In chronic AR, the chest radiograph shows
an enlarged left ventricular silhouette. This is
usually absent in acute AR, in which pul-
Examination
monary vascular congestion is the more likely
Physical examination may show bounding finding. Doppler echocardiography can identify
pulses and other stigmata of the widened and quantify the degree of AR and often can
Tab. 4 pulse pressure (Table 8.4), in addition to a identify its cause. Cardiac catheterization with
hyperdynamic LV impulse and a blowing contrast angiography is useful for evaluation
murmur of AR in early diastole along the of left ventricular function, quantification of
left sternal border (see Fig. 8.11). It is best the degree of AR, and assessment of coexist-
heard with the patient leaning forward, ing coronary artery disease. 1 LINE SHORT
10090-08_CH08.qxd 8/31/06 4:51 PM Page 215

Valvular Heart Disease 215

phy. Patients are instructed to follow anti-


ECG
biotic prophylaxis to prevent endocarditis,
as described in a later section. Patients with
asymptomatic severe AR and preserved
LV function may benefit from afterload re-
ducing vasodilators (e.g., a calcium channel
blocker or angiotensin-converting enzyme
inhibitor). For example, the vasodilating cal-
cium channel blocker nifedipine has been
shown in some, but not all, studies to re-
duce LV enlargement, increase the LV ejec-
tion fraction, and delay the need for valve
surgery in hypertensive patients with severe
AR who have normal LV contractile function.
Symptomatic patients, or asymptomatic
patients with severe AR and impaired LV
contractile function (i.e., an ejection frac-
tion <0.50), should be offered surgical cor-
rection to prevent progressive deteriora-
tion. Studies of patients with AR show that
Figure 8.11. Hemodynamic profile of aortic regur- without surgery, death usually occurs with-
gitation. During diastole, the aortic pressure falls in 4 years after the development of angina
rapidly (arrow), and left ventricular (LV) pressure rises as
blood regurgitates from the aorta into the LV. A dias- or 2 years after the onset of heart failure
tolic decrescendo murmur, beginning at the second symptoms.
heart sound (S2), corresponds with the abnormal regur-
gitant flow. ECG, electrocardiogram.

TRICUSPID VALVE DISEASE


Treatment Tricuspid Stenosis
Data from natural history studies indicate Tricuspid stenosis (TS) is rare and usually a
that clinical progression of patients with consequence of rheumatic heart disease.
asymptomatic chronic AR and normal LV The opening snap and diastolic murmur of
contractile function is very slow. Therefore, TS are similar to those of MS, but the mur-
asymptomatic patients are monitored with mur is heard closer to the sternum and it
periodic examinations and assessment of LV intensifies on inspiration because of in-
function, usually by serial echocardiogra- creased right-heart blood flow. In TS, the

TABLE 8.4. Physical Findings Associated With Widened Pulse Pressure


in Chronic Aortic Regurgitation

Name Description

Bisferiens pulse Double systolic impulse in carotid or brachial artery


Corrigan pulse “Water-hammer” pulses with marked distention and collapse
de Musset sign Head-bobbing with each systole
Duroziez sign To-and-fro murmur heard over femoral artery with light compression
Hill sign Popliteal systolic pressure more than 60 mm Hg greater than brachial systolic pressure
Müller sign Systolic pulsations of the uvula
Quincke sign Capillary pulsations visible at lip or proximal nail beds
Traube sign “Pistol-shot” sound auscultated over the femoral artery
LINE SHORT
10090-08_CH08.qxd 8/31/06 4:51 PM Page 216

216 Chapter Eight

neck veins are distended and show a large a PULMONIC VALVE DISEASE
wave as a result of right atrial contraction
against the stenotic tricuspid valve orifice. Pulmonic Stenosis
Patients may develop abdominal disten- Pulmonic stenosis (PS) is rare, and its cause
tion and hepatomegaly owing to passive is almost always congenital deformity of the
venous congestion. valve. Carcinoid syndrome, described in the
Symptoms of TS can be similar to those of previous section, is another rare etiology, in
MS, and the two conditions can coexist as which encasement and immobilization of
sequelae of rheumatic heart disease. Surgical the valve leaflets can occur.
therapy is usually required (valvuloplasty or Severe cases of pulmonic stenosis are as-
valve replacement). sociated with a peak systolic pressure gradi-
ent of greater than 80 mm Hg, moderate dis-
ease with a gradient of 40 to 80 mm Hg, and
Tricuspid Regurgitation
mild PS is said to be present when the trans-
Tricuspid regurgitation (TR) is usually func- valvular gradient is less than 40 mm Hg.
tional rather than structural (or organic); that Only patients with moderate to severe gra-
is, it develops because of right ventricular en- dients are symptomatic. In such cases, tran-
largement (e.g., owing to pressure or volume scatheter balloon valvuloplasty is usually
overload) and not because of primary valve effective therapy.
disease. Among patients with rheumatic MS,
20% have significant TR (of whom 80% have
functional TR because of pulmonary hyper- Pulmonic Regurgitation
tension with right ventricular enlargement, Pulmonic regurgitation most commonly de-
and 20% have organic TR resulting from velops in the setting of severe pulmonary
rheumatic involvement of the tricuspid hypertension and results from dilatation of
valve). Another rare cause of TR is carcinoid the valve ring by the enlarged pulmonary
syndrome, in which a type of tumor (usually artery. Auscultation reveals a high-pitched
in the small bowel or appendix, with metas- decrescendo murmur along the left sternal
tases to the liver) releases serotonin metabo- border that is often indistinguishable from
lites into the bloodstream. These metabolites AR (the two conditions are easily differenti-
are thought to be responsible for the forma- ated by Doppler echocardiography).
tion of endocardial plaques in the right side
of the heart. Involvement of the tricuspid
valve immobilizes the leaflets, often resulting PROSTHETIC VALVES
in substantial TR and, less often, tricuspid The patient who undergoes valve replace-
stenosis. ment surgery often benefits dramatically
The most sensitive physical signs of TR from hemodynamic and symptomatic im-
are prominent v waves in the jugular veins provement but also acquires a new set of po-
and a pulsatile liver because of regurgitation tential complications related to the valve
of right ventricular blood into the systemic prosthesis itself. Because all available valve
veins. The systolic murmur of TR is heard substitutes have certain limitations, valve re-
at the lower-left sternal border. It is often placement surgery is not a true “cure.”
soft but becomes louder on inspiration. The first successful valve replacements
Doppler echocardiography readily detects took place in the 1960s. Currently available
TR and can quantify it. The primary therapy valve substitutes include mechanical and bi-
of functional TR is directed at the conditions ologic (derived from animal or human tis-
responsible for the elevated right ventricular sue) devices (Fig. 8.12). Older mechanical Fig. 12
size or pressure as well as diuretic therapy; valves included a ball-in-cage design, with a
surgical repair of the valve is indicated in se- bulky shape that often left a significant
vere cases. valvular gradient and occasionally produced
10090-08_CH08.qxd 8/31/06 4:51 PM Page 217

Valvular Heart Disease 217

Figure 8.12. Examples of prosthetic heart valves. A. St. Jude mechanical bileaflet valve in the open position. (Cour-
tesy of St. Jude Medical, Inc., St. Paul, MN.) B. A bioprosthetic aortic valve with leaflets in the closed position. (Courtesy
of Medtronic, Inc., Minneapolis, MN.)

intravascular hemolysis from red blood cell because the mitral valve is forced closed
trauma. This valve type, however, had an during systolic contraction, resulting in
impressive record of durability, with some greater leaflet stress than that experienced
models functioning well for more than by aortic prostheses that close during dias-
30 years. Newer mechanical valves, such as tolic relaxation.
bileaflet prostheses, provide a lower profile The principal causes of bioprosthetic valve
and superior hemodynamics with no appar- failure are leaflet tears and calcification.
ent sacrifice of durability. One example is Conversely, the main advantage of biopros-
the St. Jude prosthesis, a hinged bileaflet theses is that they display a very low rate of
valve consisting of two Pyrolyte carbon thromboembolism and do not require long-
discs that open opposite one another (see term anticoagulation therapy. For patients
Fig. 8.12). Mechanical valves, while durable, with aortic valve endocarditis, aortic homo-
present foreign thrombogenic surfaces to graft replacements are especially useful be-
the circulating blood and require lifelong cause they have very low rates of subsequent
anticoagulation (usually with oral warfarin) infection.
to prevent thromboembolism. Common to all types of valve replace-
The most commonly used bioprostheses ment is the risk of infective endocarditis
are made from glutaraldehyde-fixed porcine (discussed in the next section), which occurs
valves secured in a support frame. In ad- at an incidence of 1% to 2% per patient per
dition, bovine pericardium and human year. If endocarditis occurs in the first 60 days
homograft (aortic valves harvested and after valve surgery, the mortality rate is ex-
cryopreserved from cadavers) prostheses ceedingly high (50% to 80%). If endocardi-
are used. Bioprosthetic valves have limited tis occurs later, mortality rates range from
durability compared with mechanical valves, 20% to 50%. Reoperation is usually required
and structural failure occurs in up to 50% of if endocarditis involves a mechanical pros-
valves at 10 years. Failure rates vary greatly thesis, because an adjacent abscess is almost
depending on the position of the valve. For always present (the organism cannot infect
example, bioprosthetic valves in the mitral the prosthetic material itself). Some cases of
position deteriorate more rapidly than bioprosthetic valve endocarditis respond to
those in the aortic position. This is likely antibiotic therapy alone.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 218

218 Chapter Eight

Given their respective advantages and as streptococci viridans are involved. SBE
disadvantages, the mortality and complica- most frequently occurs in individuals with
tion rates with mechanical and biopros- prior underlying valvular damage.
thetic valves are similar for the first 10 years The second means of classification of IE is
following replacement. In 20-year follow- according to the host substrate: (1) native
ups to long-term, randomized, controlled valve endocarditis, (2) prosthetic valve en-
trials, mechanical valves have been shown docarditis, or (3) endocarditis in the setting
to be superior to bioprosthetic valves in of intravenous drug abuse. Of these, native
event-free survival, except for bleeding com- valve endocarditis accounts for 60% to 80%
plications related to the required chronic of patients. Different microorganisms and
anticoagulation therapy. Therefore, the de- clinical courses are associated with each of
cision about which type of prosthesis to use these categories. For example, the skin cont-
in a patient often centers on (1) the patient’s aminant Staphylococcus epidermidis, is a com-
expected lifespan in comparison to the func- mon cause of prosthetic valve endocarditis,
tional longevity of the valve, (2) risk-versus- but that is rarely the case when endocarditis
benefit considerations of chronic anticoag- occurs on a native heart valve. Intravenous
ulation therapy, and (3) patient and surgeon drug users have a propensity for endocardi-
preferences. Mechanical valves are often rec- tis on the right-sided heart valves.
ommended for younger patients and for The third classification of IE is according
those who will be tolerant of, and compliant to the specific infecting microorganism (e.g.,
with, anticoagulant therapy. Bioprosthetic S. aureus endocarditis). Although the re-
valves are suitable choices for patients 65 mainder of this discussion focuses on the
years or older and for patients with con- endocarditis syndromes based on clinical
traindications to chronic anticoagulation course, it is important to recognize that all
therapy. three classifications of IE are used.

INFECTIVE ENDOCARDITIS Pathogenesis


Infection of the endocardial surface of the The pathogenesis of endocarditis requires
heart, including the cardiac valves, can lead several conditions: (1) endocardial surface
to extensive tissue damage and is often fatal. injury, (2) thrombus formation at the site of
Infective endocarditis (IE) carries an overall injury, (3) bacterial entry into the circula-
6-month mortality rate of 20% to 25%, even tion, and (4) bacterial adherence to the in-
with appropriate therapy, and a 100% mor- jured endocardial surface. The first two con-
tality rate if it is not recognized and treated ditions provide an environment favorable
correctly. to infection, whereas the latter two permit
There are three clinically useful ways to implantation of the organism on the endo-
classify IE: (1) by clinical course, (2) by host cardial surface. The most common cause of
substrate, or (3) by the specific infecting endothelial injury is turbulent blood flow
microorganism. In the first classification resulting from preexisting valvular disease;
scheme, IE is termed acute bacterial endo- approximately 70% of patients with endo-
carditis (ABE) when the syndrome pre- carditis have evidence of underlying struc-
sents as an acute, fulminant infection, and tural or hemodynamic abnormalities (Table Tab. 5
a highly virulent and invasive organism 8.5). Endothelial injury may also be incited
such as Staphylococcus aureus is implicated. by foreign material within the circulation,
Because of the aggressiveness of the respon- such as indwelling venous catheters or pros-
sible microorganism, ABE may occur on pre- thetic heart valves.
viously healthy heart valves. When IE pre- Once an endocardial surface is injured,
sents with a more insidious clinical course, platelets adhere to the exposed subendocar-
it is termed subacute bacterial endocardi- dial connective tissue and initiate the for-
tis (SBE), and less virulent organisms such mation of a sterile thrombus (termed a
10090-08_CH08.qxd 8/31/06 4:51 PM Page 219

Valvular Heart Disease 219

Gram-positive organisms account for ap-


TABLE 8.5. Cardiac Lesions That Predispose proximately 90% of cases of endocarditis,
to Endocarditis
largely because of their resistance to de-
Rheumatic valvular disease struction in the circulation by complement
Other acquired valvular lesions and their particular ability to adhere to en-
Calcific aortic stenosis dothelial and platelet surface proteins. The
Aortic regurgitation
ability of certain streptococcal species to
Mitral regurgitation
Mitral valve prolapse (if murmur auscultated or produce dextran, a bacterial cell wall com-
detected by Doppler) ponent that adheres to thrombus, correlates
Hypertrophic obstructive cardiomyopathy with their inciting endocarditis. Table 8.6 Tab. 6
Congenital heart disease lists the infectious agents reported to be the
Ventricular septal defect
most common causes of endocarditis in
Patent ductus arteriosus
Tetralogy of Fallot modern tertiary centers; staphylococci and
Aortic coarctation streptococci are the most frequent. It is im-
Bicuspid aortic valve portant to recognize that in more rural com-
Pulmonic stenosis munities with a low prevalence of intra-
Surgically implanted intravascular hardware
venous drug abuse, the percentage of viridans
Prosthetic heart valves
Pulmonary-systemic vascular shunts streptococcal infections tends to be greater
Ventriculoatrial shunts for hydrocephalus than that of S. aureus.
Once organisms adhere to the injured
surface, they may be protected from phago-
cytic activity by the overlying fibrin. The
vegetation) through fibrin deposition. This organisms are then free to multiply, which
process is referred to as nonbacterial throm- enlarges the infected vegetation. The latter
botic endocarditis (NBTE). NBTE makes the provides a source for continuous bacte-
endocardium more hospitable to microbes remia and can lead to several complica-
in two ways. First, the fibrin-platelet deposits tions, including (1) mechanical cardiac
provide a surface for adherence by bacteria. injury, (2) thrombotic or septic emboli, and
Second, the fibrin covers adherent organ- (3) immune injury mediated by antigenan-
isms and protects them from host defenses tibody deposition. For example, local ex-
by inhibiting chemotaxis and migration of tension of the infection within the heart
phagocytes. can result in progressive valvular damage,
When NBTE is present, the delivery of
microorganisms in the bloodstream to the
injured surface can lead to infective endo-
carditis. Three factors determine the ability TABLE 8.6. Microbiology of Infective
Endocarditis in Tertiary Centers
of an organism to induce IE: (1) access to the
bloodstream, (2) survival of the organism in Organism Incidence (%)
the circulation, and (3) adherence of the
Staphylococci
bacteria to the endocardium. Bacteria can be
S. aureus 31.6
introduced into the bloodstream whenever Coagulase-negative 10.5
a mucosal or skin surface harboring an or- Streptococci
ganism is traumatized, such as from the Viridans 18.0
mouth during dental procedures or from the Enterococci 10.6
S. bovis 6.5
skin during illicit intravenous drug use.
Other streptococci 5.1
However, while transient bacteremia is a rel- Other organisms (e.g., Gram-negative
atively common event, only microorgan- bacteria, fungi) 8.7
isms suited for survival in the circulation Culture negative or polymicrobial ∼9.4
and able to adhere to the platelet-fibrin
Modified from Fowler VG Jr, Miro JM, Hoen B, et al. Staphy-
mesh overlying the endocardial defect will lococcus aureus endocarditis: a consequence of medical
cause infective endocarditis. For example, progress. JAMA 2005;293:3012–3021.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 220

220 Chapter Eight

abscess formation, or erosion into the car- lar murmur (i.e., worsening regurgitation)
diac conduction system. Portions of a veg- over time may correspond with rapidly pro-
etation may embolize systemically, often gressive valvular damage. During the course
to the central nervous system, kidneys, or of endocarditis, severe valvular damage may
spleen, and incite infection or infarction of result in findings of congestive heart failure.
the target organs. Each of these is a poten- Other physical findings that may appear
tially fatal complication. Additionally, im- in IE are those associated with septic em-
mune complex deposition can result in bolism or immune complex deposition.
glomerulonephritis, arthritis, or vasculitis. Central nervous system emboli are seen in
The epidemiology of IE has evolved in up to 40% of patients, often resulting in new
recent decades, as bacteria resistant to anti- neurologic findings on physical examina-
biotics have become ubiquitous in the hos- tion. Injury to the kidneys, of embolic or
pital setting and have spread into the com- immunologic origin, may manifest as flank
munity. Antibiotic resistant strains such as pain, hematuria, or renal failure. Lung infarc-
methicillin-resistant S. aureus and vanco- tion (septic pulmonary embolism) or infec-
mycin-resistant enterococci have become tion (pneumonia) are particularly common
more common and are associated with in- in endocarditis that involves the right-sided
creased mortality rates from IE. valves.
Embolic infarction and seeding of the
vasa vasorum of arteries can cause localized
Clinical Manifestations aneurysm formation (termed a mycotic aneu-
rysm) that weakens the vessel wall and may
A patient with acute IE is likely to report an rupture. Mycotic aneurysms may be found
explosive and rapidly progressive illness in the aorta, viscera, or peripheral organs,
with high fever and shaking chills. In con- and are particularly dangerous in cerebral
trast, subacute IE presents less dramatically vessels, because rupture there can result in
with low-grade fever often accompanied by fatal intracranial hemorrhage.
nonspecific constitutional symptoms such Skin findings resulting from septic em-
as fatigue, anorexia, weakness, myalgia, and bolism or immune complex vasculitis are
night sweats. These symptoms are not spe- often collectively referred to as peripheral
cific for IE and could easily be mistaken for stigmata of endocarditis. For example, pe-
influenza or an upper respiratory tract in- techiae may appear as tiny, circular, red-
fection. Thus, the diagnosis of subacute brown discolorations on mucosal surfaces or
IE requires a high index of suspicion. A his- skin. Splinter hemorrhages, the result of sub-
tory of a valvular lesion or other condition ungual microemboli, are small, longitudinal
known to predispose to endocarditis is help- hemorrhages found beneath nails. Other
ful. A thorough history should also inquire peripheral stigmata of IE, which are now
about intravenous drug use, recent dental rarely encountered, include painless, flat, ir-
procedures, or other potential sources of bac- regular discolorations found on the palms
teremia. and soles called Janeway lesions; tender,
Cardiac examination may reveal a mur- pea-sized, erythematous nodules found pri-
mur representing the underlying valvular marily in the pulp space of the fingers and
pathology that predisposed the patient to IE, toes termed Osler nodes; and emboli to the
or a new murmur of valvular insufficiency retina that produce Roth spots, microinfarc-
owing to IE-induced damage. The develop- tions that appear as white dots surrounded
ment of right-sided valvular lesions (e.g., by hemorrhage.
tricuspid regurgitation), although rare in The systemic inflammatory response pro-
normal hosts, is particularly common in en- duced by the infection is responsible for
docarditis associated with intravenous drug fever and splenomegaly as well as for a num-
abuse. Serial examination in ABE may be es- ber of laboratory findings, including an ele-
pecially useful because changes in a particu- vated white blood cell count with a leftward
10090-08_CH08.qxd 8/31/06 4:51 PM Page 221

Valvular Heart Disease 221

shift (increase in proportion of neutrophils antibiotics may be used initially (after blood
and immature granulocytes), an elevated cultures are obtained) in patients who are se-
erythrocyte sedimentation rate or C-reactive verely ill or hemodynamically unstable, spe-
protein level, and in approximately 50% of cific, directed therapy is preferable once the
cases, an elevated serum rheumatoid factor. causative microorganism has been identified.
The electrocardiogram may help identify Surgical intervention, usually with valve re-
extension of the infection into the cardiac placement, is indicated for patients with per-
conduction system, manifest by various de- sistent bacteremia despite maximal antibiotic
grees of heart block or new arrhythmias. therapy, patients with severe valvular dys-
Echocardiography is used to visualize vegeta- function leading to heart failure, and patients
tions, valvular dysfunction, and associated who develop myocardial abscesses or experi-
abscess formation. Echocardiographic assess- ence recurrent thromboembolic events.
ment can consist of transthoracic echocar- Finally, an additional essential concept is
diography (TTE) or transesophageal echocar- prevention of endocarditis by administering
diography (TEE), as described in Chapter 3. antibiotics to susceptible individuals (i.e.,
TTE is useful in detecting large vegetations those with underlying structural heart dis-
and has the advantage of being noninva-
ease) before procedures that are likely to re-
sive and easy to obtain. However, while the
sult in bacteremia (Table 8.8). Although ran- Tab. 8
estimated specificity of TTE for vegetations
domized controlled trials have not been
is high, the sensitivity for finding vegeta-
performed to demonstrate the efficacy of an-
tions is less than 60%. TEE, on the other
tibiotic prophylaxis against IE, such practice
hand, is much more sensitive (>90%) for
is widespread.
the detection of small vegetations and can
be particularly useful for the evaluation of
infection involving prosthetic valves. SUMMARY
Central to the diagnosis and appropriate
treatment of endocarditis is the identifi- Valvular heart disease can be a significant
cation of the responsible microorganism source of disability and mortality. From sim-
by blood culture. Once positive culture re- ple bedside observations to complex hemo-
sults are obtained, treatment can be tailored dynamic measurements, much has been
to the causative organism according to its learned about the pathophysiology of these
antibiotic sensitivities. A specific etiologic conditions. A summary of the important
agent will be identified approximately findings associated with major valve lesions
95% of the time. However, blood cultures is presented in Table 8.9. Tab. 9
may fail to grow the responsible organism
if antibiotics had already been adminis-
tered or if the organism has unusual growth Acknowledgment
requirements. Contributors to the previous editions of this chapter
Even after a careful history, examination, were Edward Chan, MD; Elia Duh, MD; Stephen K.
and evaluation of laboratory data, the diag- Frankel, MD; Brian Stidham, MD; Patrick Yachimski,
nosis of IE can be elusive. Therefore, at- MD; John A. Bittl, MD; and Leonard S. Lilly, MD.
tempts have been made to standardize the
diagnosis, resulting in the now widely used
Tab. 7 Duke criteria (Table 8.7). By this standard,
Additional Reading
the diagnosis of endocarditis rests on the Bonow RO, Carabello BA, Chatterjee K, et al. ACC/
presence of either two major criteria, one AHA 2006 guidelines for the management of pa-
major and three minor criteria, or five minor tients with valvular heart disease: a report of the
American College of Cardiology/American Heart
criteria.
Association Task Force on Practice Guidelines.
Treatment of endocarditis entails 4 to J Am Coll Cardiol 2006;48:e1–148. Available at:
6 weeks of high-dose intravenous antibiotic http://www.acc.org/clinical/guidelines/valvular/
therapy. Although empiric broad-spectrum index.pdf. Accessed July 7, 2006.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 222

222 Chapter Eight

TABLE 8.7. Modified Duke Criteria for Diagnosis of Infective Endocarditis (IE)a

Major Criteria Minor Criteria

I. Positive blood culture, defined as either A or B Predisposing cardiac condition or intravenous


drug use
A. Typical microorganism for IE from two separate Fever (≥38.0° C)
blood cultures
1. Streptococci viridans, S. bovis, HACEK group; or Vascular phenomena (septic arterial or pul-
2. Staphylococcus aureus or enterococci, in the monary emboli, mycotic aneurysm, intracra-
absence of a primary focus nial hemorrhage, conjunctival hemorrhage,
Janeway lesions)
Immunologic phenomena (glomerulonephri-
tis, Osler’s nodes, Roth spots, rheumatoid
factor)
B. Microorganisms consistent with IE from Positive blood cultures not meeting major cri-
persistently positive blood cultures teria or serologic evidence of infection with
1. Blood cultures drawn >12 hours apart, or organism consistent with IE
2. All of three, or most of four separate cultures
drawn at least 1 hour apart
3. Single positive blood culture for Coxiella burnetii
or antiphase I IgG antibody titer >1:800
II. Evidence of endocardial involvement, defined
as A or B
A. Echocardiogram positive for endocarditis:
1. Oscillating intracardiac mass, or
2. Myocardial abscess, or
3. New partial detachment of prosthetic valve
B. New valvular regurgitation
a
Clinical diagnosis of definitive endocarditis requires two major criteria, one major criteria plus three minor criteria, or five
minor criteria.
HACEK, Haemophilus spp., Actinobacillus actinomycetemcomitans, Cardiobacterium hominis, Eikenella spp. and Kingella kingae.
Modified from Li JS, Sexton DJ, Mick N, et al. Proposed modifications to the Duke criteria for the diagnosis of infective
endocarditis. Clin Infect Dis 2000;30:633–638.

TABLE 8.8. Examples of Procedures That Warrant Endocarditis Prophylaxis

Dental manipulations that produce gingival bleeding


Upper respiratory tract procedures
Tonsillectomy and/or adenoidectomy
Surgical operations that involve respiratory mucosa
Bronchoscopy with a rigid bronchoscope
Genitourinary procedures
Urethral dilation
Cystoscopy
Prostatectomy
Gastrointestinal surgery, including cholecystectomy
TABLE 8.9. Summary of Major Valvular Conditions

Valve Lesion Causes Symptoms Physical Findings Murmur Diagram


10090-08_CH08.qxd

Mitral stenosis Sequelae of rheumatic Symptoms of left-sided • Loud S1 (in early MS)
(MS) fever (and later right- • Opening snap
sided) heart failurea • Diastolic rumble “MS”
8/31/06

(loudest in left lateral


decubitus position) S1 S2 OS S1
Mitral regurgitation Acute: Acute: • Holosystolic murmur
(MR) • Endocarditis • Pulmonary edema at apex (may be de-
• Ruptured chordae Chronic: crescendo in acute “MR”
4:51 PM

• Papillary muscle • Symptoms of left- MR)


dysfunction sided heart • Murmur accentuated
S1 S2
Chronic: failurea and low by clenching the fists
• Rheumatic cardiac output • Widely split S2
Page 223

• Mitral prolapse (e.g., fatigue)


• Calcified annulus
• LV dilatation
Aortic stenosis • Degenerative calcific • Chest pain • Carotids: delayed up-
• Congenital • Syncope stroke and decreased
• Rheumatic • Dyspnea on exertion volume
“AS”
• Palpation: supraster-
nal thrill
• Auscultation: S1 S2
• Soft A2
• Systolic ejection-
type murmur
(loudest at upper-
right sternal
border)
Aortic regurgitation • Congenital (e.g., • Dyspnea on exertion • Wide pulse pressure
bicuspid valve) • Chest pain (some- • Bounding pulses
• Endocarditis times) • Early diastolic de- “AR”
• Rheumatic crescendo murmur
• Aortic root dilatation (heard best at end ex- S1 S2
piration, with patient
leaning forward)

aSymptoms of left-sided heart failure include exertional dyspnea, orthopnea, paroxysmal nocturnal dyspnea; symptoms of right-sided heart failure include peripheral edema, abdominal

bloating, and right upper quadrant tenderness (hepatic enlargement).

223
A2, aortic portion of heart sound; LV, left ventricular; S1, first heart sound; S2, second heart sound.
10090-08_CH08.qxd 8/31/06 4:51 PM Page 224

224 Chapter Eight

Enriquez-Sarano M., Tajik AJ. Clinical practice. Aor- Li JS, Sexon DJ, Mick N, et al. Proposed modifications
tic regurgitation. N Engl J Med 2004;351:1539– to the Duke criteria for the diagnosis of infective
1546. endocarditis. Clin Infect Dis 2000;30:633–638.
Evangelista A, Tornos P, Sambola A, et al. Long-term Martin-Davila P, Fortun J, Navas E, et al. Nosocomial
vasodilator therapy in patients with severe aortic endocarditis in a tertiary hospital: an increasing
regurgitation. N Engl J Med 2005;353:1342–1349. trend in native valve cases. Chest 2005;128:
Ferrieri P. Jones Criteria Working Group. Proceed- 772–779.
ings of the Jones Criteria Workshop. Circulation Meira ZM, Goulart EM, Colosimo EA, et al. Long term
2002;106:2521–2523. follow up of rheumatic fever and predictors of se-
Filsoufi F, Anyanwu AC, Salzberg SP, et al. Long-term vere rheumatic valvar disease in Brazilian children
outcomes of tricuspid valve replacement in the and adolescents. Heart 2005;91:1019–1022.
current era. Ann Thorac Surg 2005;80:845–850. Murphy RT, Garcia MJ. Role of echocardiography in
Fowler VG Jr, Miro JM, Hoen B, et al. Staphylococcus diagnosis and management of endocarditis. Curr
Infect Dis Rep 2005;7:257–263.
aureus endocarditis: a consequence of medical
progress. JAMA 2005;293:3012–3021. Otto CM, Salerno CT. Timing of surgery in asymp-
tomatic mitral regurgitation. N Engl J Med 2005;
Infective endocarditis: diagnosis, antimicrobial ther-
352:928–929.
apy, and management of complications: a state-
ment for healthcare professionals from the Com- Rahimtoola SH. Valvular heart disease/cardiac sur-
mittee on Rheumatic Fever, Endocarditis, and gery. J Am Coll Cardiol 2005;45(suppl B):20B–
Kawasaki Disease; Council on Cardiovascular Dis- 23B.
ease in the Young; and the Councils on Clinical Rosenhek R. Statins for aortic stenosis. N Engl J Med
Cardiology, Stroke, and Cardiovascular Surgery 2005;352:2441–2443.
and Anesthesia, American Heart Association— Tawn Z, Himbert D, Brochet E, et al. Percutaneous
executive summary. Circulation 2005;111:3167– valve procedures: present and future. Int J Cardio-
3184. vasc Intervent 2005;7:14–20.
10090-08_CH08.qxd 8/31/06 4:51 PM Page a

Chapter 8—Author Queries


1. ED: Has permission been received?
2. ED: Has permission been received?
10090-09_CH09.qxd 8/31/06 4:52 PM Page 225

C H A P T E R

Heart Failure
Ravi Vikram Shah
Michael A. Fifer
9
PHYSIOLOGY CLINICAL MANIFESTATIONS
Determinants of Contractile Function in the Symptoms
Intact Heart Physical Signs
Pressure-Volume Loops Laboratory Tests
PATHOPHYSIOLOGY PROGNOSIS
Systolic Dysfunction TREATMENT
Diastolic Dysfunction Diuretics
Right-Sided Heart Failure Vasodilators
COMPENSATORY MECHANISMS Inotropic Drugs
Frank-Starling Mechanism β-Blockers
Neurohormonal Alterations Aldosterone Antagonist Therapy
Ventricular Hypertrophy and Remodeling Additional Therapies
Treatment of Diastolic Dysfunction
MYOCYTE LOSS AND CELLULAR DYSFUNCTION
ACUTE PULMONARY EDEMA
PRECIPITATING FACTORS

The heart normally accepts blood at low fill- roidism), in this chapter, only cardiac causes
ing pressures during diastole and then pro- of heart failure are considered.
pels it forward at higher pressures in systole. Heart failure may be the final and most
Heart failure is defined as the inability of the severe manifestation of nearly every form
heart to pump blood forward at a sufficient rate of cardiac disease, including coronary ather-
to meet the metabolic demands of the body (for- osclerosis, myocardial infarction, valvular
ward failure), or the ability to do so only if the diseases, hypertension, congenital heart dis-
cardiac filling pressures are abnormally high ease, and the cardiomyopathies. More than
(backward failure), or both. Although condi- 500,000 new cases of heart failure develop
tions outside the heart may cause this defi- in the United States each year, and approxi-
nition to be met through inadequate tissue mately 5 million people currently have this
perfusion (e.g., severe hemorrhage) or in- condition. It accounts for more than 12 mil-
creased metabolic demands (e.g., hyperthy- lion medical office visits annually and is the

225
10090-09_CH09.qxd 8/31/06 4:52 PM Page 226

226 Chapter Nine

most common diagnosis of hospitalized pa- A second observation from the isolated
tients aged 65 and older. The incidence of muscle experiments arises when the fibers
heart failure is actually increasing, partly be- are not tethered at a fixed length but are al-
cause the population is aging but also be- lowed to shorten during stimulation against
cause of interventions that prolong survival a fixed load (termed the afterload). In this
after acute cardiac insults such as myocar- situation (termed an isotonic contraction),
dial infarction. the final length of the muscle at the end of
Heart failure most commonly results from contraction is directly related to the magni-
conditions of impaired left ventricular func- tude of the load but is independent of the
tion. Thus, this chapter begins by review- length of the muscle before stimulation
ing the physiology of normal myocardial con- (see Fig. 9.1B). That is, (1) the tension gen-
traction and relaxation. erated by the fiber is equal to the fixed
load; (2) the greater the load opposing con-
traction, the less the muscle fiber can shorten;
PHYSIOLOGY
(3) if the fiber is stretched to a longer length
Experimental studies of isolated cardiac mus- before stimulation but the afterload is kept
cle segments have revealed several impor- constant, the muscle will shorten a greater
tant physiologic principles that can be ap- distance and will attain the same final length
plied to the intact heart. As an experimental at the end of contraction; and (4) the maxi-
muscle segment is stretched apart, the rela- mum tension that a fiber can produce during
tion between its length and the tension it isotonic contraction (i.e., such that the fiber
passively develops is curvilinear, reflecting is just unable to shorten) is the same as the
Fig. 1 its intrinsic elastic properties (Fig. 9.1A, force produced by an isometric contraction
lower curve). If the muscle is first passively for the applied preload.
stretched and then stimulated to contract The concept of afterload is also relevant
while its ends are held at fixed positions to the intact heart: the pressure generated by
(termed an isometric contraction), the total the ventricle and the size of the chamber at
tension (active plus passive tension) gener- the end of each contraction depend on the
ated by the fibers is proportional to the load against which the ventricle contracts
length of the muscle at the time of stimula- (i.e., largely the arterial pressure) but are in-
tion (see Fig. 9.1A, upper curve). That is, dependent of the stretch on the myocardial
stretching the muscle before stimulation fibers before contraction.
optimizes the overlap and interaction of A third key experimental observation re-
myosin and actin filaments, increasing the lates to myocardial contractility (also termed
number of cross bridges and the force of the inotropic state), which accounts for
contraction. Stretching cardiac muscle fibers changes in the force of contraction indepen-
also increases the sensitivity of the myofila- dent of the initial fiber length and afterload.
ments to calcium, which further augments Contractility generally reflects chemical and
force development. hormonal influences on cardiac contraction,
This relationship between the initial fiber such as exposure to catecholamines. When
length and force development is of great im- contractility is enhanced pharmacologically
portance in the intact heart: within a phys- (e.g., by a norepinephrine infusion), the re-
iologic range, the larger the ventricular vol- lation between initial fiber length and force
ume during diastole, the more the fibers are developed during contraction is shifted up-
stretched before stimulation and the greater ward (see Fig. 9.1C) such that a greater total
the force of the next contraction. This is the tension develops with isometric contraction
basis of the Frank-Starling relationship, at any given preload. Similarly, when con-
the observation that ventricular output tractility is augmented and the cardiac mus-
increases in relation to the preload (the cle is allowed to shorten against a fixed af-
stretch on the myocardial fibers before con- terload, the fiber contracts to a greater extent
traction). and achieves a shorter final fiber length com-
10090-09_CH09.qxd 8/31/06 4:52 PM Page 227

Heart Failure 227

c c
a a

g e

Figure 9.1. Physiology of normal cardiac muscle segments. A. Passive (lower curve) and total
(upper curve) length-tension relations for isolated cat papillary muscle. Lines ab and cd represent the
force developed during isometric contractions. Initial passive muscle length c is longer (i.e., has been
stretched more) than length a and therefore has a greater passive tension. When the muscle segments
are stimulated to contract, the muscle with the longer initial length generates greater total tension
(point d versus point b). B. If the muscle fiber preparation is allowed to shorten against a fixed load,
the length at the end of the contraction is dependent on the load but not the initial fiber length; stim-
ulation at point a or c results in the same final fiber length (e). Thus, the muscle that starts at length c
shortens a greater distance (∆Lc) than the muscle at length a (∆La). C. The uppermost curve is the
length-tension relation in the presence of the positive inotropic agent norepinephrine. For any given
initial length, an isometric contraction in the presence of norepinephrine generates greater force (point f )
than one in the absence of norepinephrine (point b). When contracting against a fixed load, the pres-
ence of norepinephrine causes greater muscle fiber shortening and a smaller final muscle length (point g)
compared with contraction in the absence of the inotropic agent (point e). (Adapted from Downing
SE, Sonnenblick EH. Cardiac muscle mechanics and ventricular performance: force and time parame-
ters. Am J Physiol 1964;207:705–715.)
10090-09_CH09.qxd 8/31/06 4:52 PM Page 228

228 Chapter Nine

pared with the normal state. Enhanced con-


tractility is likely induced by an increase in
the cycling rate of actin-myosin cross-bridge
formation.

Determinants of Contractile
Function in the Intact Heart
In a healthy person, cardiac output is
matched to the body’s total metabolic need.
Cardiac output (CO) is equal to the product
of stroke volume (SV, the volume of blood
ejected with each contraction) and the heart
Figure 9.2. Key mediators of cardiac output. Deter-
rate (HR): minants of the stroke volume include contractility, pre-
load, and afterload. Cardiac output = Heart rate × Stroke
CO = SV × HR volume.

The three major determinants of stroke vol-


ume are preload, afterload, and myocardial This relationship is illustrated graphically by
Fig. 2 contractility, as shown in Figure 9.2. the Frank-Starling curve, also known as the
ventricular function curve (Fig. 9.3). The Fig. 3
graph relates a measurement of cardiac per-
Preload
formance (such as cardiac output or stroke
Tab. 1 The concept of preload (Table 9.1) in the in- volume) on the vertical axis as a function of
tact heart was described by physiologists preload on the horizontal axis. As described
Frank and Starling a century ago. In expe- earlier, the preload can be thought of as the
rimental preparations, they showed that amount of myocardial stretch at the end of di-
within physiologic limits, the more a normal astole, just before contraction. Measurements
ventricle is distended (i.e., filled with blood) that correlate with myocardial stretch, and
during diastole, the greater the volume that is that are often used to indicate the preload on
ejected during the next systolic contraction. the horizontal axis, are the ventricular end-

TABLE 9.1. Terms Related to Cardiac Performance

Term Definition

Preload The ventricular wall tension at the end of diastole. In clinical terms, it is the stretch
on the ventricular fibers just before contraction, often approximated by the end-
diastolic volume or end-diastolic pressure.
Afterload The ventricular wall tension during contraction; the resistance that must be over-
come for the ventricle to eject its content. Often approximated by the systolic
ventricular (or arterial) pressure.
Contractility Property of heart muscle that accounts for changes in the strength of contraction,
(inotropic state) independent of the preload and afterload. Often reflects chemical or hormonal
influences (e.g., catecholamines) on the force of contraction.
Stroke volume (SV) Volume of blood ejected from the ventricle during systole. SV = End-diastolic
volume–End-systolic volume.
Ejection fraction (EF) The fraction of end-diastolic volume ejected from the ventricle during each systolic
contraction (normal range = 55–75%). EF = Stroke volume ÷ End-diastolic volume.
Cardiac output (CO) Volume of blood ejected from the ventricle per minute. CO = SV × Heart rate.
Compliance Intrinsic property of a chamber that describes its pressure-volume relationship dur-
ing filling. Reflects the ease or difficulty with which the chamber can be filled.
Strictl definition: Compliance = ∆ Volume ÷ ∆ Pressure.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 229

Heart Failure 229

Hypotension

Pulmonary congestion

Figure 9.3. Left ventricular (LV) performance (Frank-Starling) curves re-


late preload, measured as LV end-diastolic volume (EDV) or pressure
(EDP), to cardiac performance, measured as ventricular stroke volume
or cardiac output. On the curve of a normal heart (middle line), cardiac per-
formance continuously increases as a function of preload. States of increased
contractility (e.g., norepinephrine infusion) are characterized by an aug-
mented stroke volume at any level of preload (upper line). Conversely, de-
creased LV contractility (commonly associated with heart failure) is character-
ized by a curve that is shifted downward (lower line). Point a is an example of
a normal person at rest. Point b represents the same patient after developing
systolic dysfunction and heart failure (e.g., after a large myocardial infarction):
stroke volume has fallen, and the decreased LV emptying results in elevation
of the EDV. Because point b is on the ascending portion of the curve, the el-
evated EDV serves a compensatory role because it results in an increase in sub-
sequent stroke volume, albeit much less than if operating on the normal
curve. Further augmentation of LV filling (e.g., increased circulating volume)
in the heart failure patient is represented by point c, which resides on the rel-
atively flat part of the curve: stroke volume is only slightly augmented, but the
significantly increased EDP results in pulmonary congestion.

diastolic volume (EDV) or end-diastolic pres- overcome to empty its content. It is more
sure (EDP). Conditions that decrease intra- formally defined as the ventricular wall stress
vascular volume, and thereby reduce ven- that develops during systolic ejection. Wall
tricular preload (e.g., dehydration or severe stress (s), like pressure, is expressed as force
hemorrhage), result in a smaller EDV and per unit area, and for the left ventricle, may
hence a reduced stroke volume during con- be estimated from LaPlace’s relationship: AQ1
traction. Conversely, an increased volume
P ×r
within the left ventricle during diastole (e.g., s=
2h
a large intravenous fluid infusion) results in a
greater-than-normal stroke volume. in which P is ventricular pressure, r is ven-
tricular chamber radius, and h is ventricular
wall thickness. Thus, ventricular wall stress
Afterload
increases in response to a higher pressure
Afterload (see Table 9.1) in the intact heart load (e.g., hypertension) or an increased
reflects the resistance that the ventricle must chamber size (e.g., a dilated left ventricle).
10090-09_CH09.qxd 8/31/06 4:52 PM Page 230

230 Chapter Nine

Conversely, as would be expected from


LaPlace’s relationship, an increase in wall
thickness (h) serves a compensatory role in
reducing wall stress, because the force is dis-
tributed over a greater mass per unit surface
area of ventricular muscle.

Contractility
In the intact heart, as in the isolated mus-
cle preparation, contractility accounts for
changes in the force generated by the myo- Figure 9.4. Example of a normal
cardium for a given set of preload and af- left ventricular (LV) pressure-
terload conditions. By relating a measure of volume loop. At point a, the mi-
tral valve opens. During diastolic
ventricular performance (stroke volume or filling of the LV (line ab), the vol-
cardiac output) to preload (left ventricular ume increases in association with a
end-diastolic pressure or volume), each gradual rise in pressure. When
ventricular contraction commences
Frank-Starling curve is a reflection of the and its pressure exceeds that of the
heart’s current inotropic state (see Fig. 9.3). left atrium, the mitral valve (MV)
The effect on stroke volume by an alter- closes (point b) and isovolumetric
contraction of the LV ensues (the
ation in preload is reflected by a change in aortic valve is not yet open, and no
position along a particular Frank-Starling blood leaves the chamber), as
curve. A change in contractility, on the shown by line bc. When LV pres-
sure rises to that in the aorta, the
other hand, actually shifts the entire curve aortic valve (AV) opens (point c)
in an upward or downward direction. Thus, and ejection begins. The volume
when contractility is enhanced pharma- within the LV declines during ejec-
tion (line cd), but LV pressure con-
cologically (e.g., by an infusion of norepi- tinues to rise until ventricular relax-
nephrine), the ventricular performance ation commences. At point d, the
curve is displaced upward such that at any LV pressure during relaxation falls
below that in the aorta, and the
given preload, the stroke volume is in- AV closes, leading to isovolumetric
creased. Conversely, when a drug that re- relaxation (line da). As the LV pres-
duces contractility is administered or the sure falls further, the mitral valve
reopens (point a). Point b repre-
ventricle’s contractile function is impaired sents the end-diastolic volume
(as in many types of heart failure), the curve (EDV) and pressure, and point d is
shifts in a downward direction, leading to the end-systolic volume (ESV) and
pressure. Stroke volume is the dif-
reductions in the stroke volume and cardiac ference between the EDV and ESV.
output at any given preload.

rise in pressure, in accordance with the pas-


Pressure-Volume Loops
sive length-tension properties or compli-
Another useful graphic display to illustrate ance (see Table 9.1) of the myocardium,
the determinants of cardiac function is the analogous to the lower curve in Figure 9.1A
ventricular pressure-volume loop, which re- for an isolated muscle preparation.
lates changes in ventricular volume to cor- Next, the onset of left ventricular systolic
responding changes in pressure throughout contraction causes the ventricular pressure
Fig. 4 the cardiac cycle (Fig. 9.4). In the left ven- to rise. When the pressure in the left ventri-
tricle, filling of the chamber begins after the cle (LV) exceeds that of the left atrium (point
mitral valve opens in early diastole (point a). b), the mitral valve is forced to close. As the
The curve between points a and b represents pressure continues to increase, the ventricu-
diastolic filling. As the volume increases lar volume does not immediately change,
during diastole, it is associated with a small because the aortic valve has not yet opened;
10090-09_CH09.qxd 8/31/06 4:52 PM Page 231

Heart Failure 231

therefore, this phase is called isovolumetric long as contractility and afterload are kept
contraction. When the ventricular pressure constant.
reaches the aortic diastolic pressure, the aor- Although end-diastolic volume and end-
tic valve is forced to open (point c) and ejec- diastolic pressure are often used interchange-
tion of blood into the aorta commences. ably as markers of preload, the relationship
During ejection, the volume within the ven- between filling volume and pressure (known
tricle decreases, but its pressure continues to as ventricular compliance; see Table 9.1),
rise until ventricular relaxation begins. The largely governs the extent of ventricular fill-
pressure against which the ventricle ejects ing. If ventricular compliance is reduced
(afterload) is represented by the curve cd. (e.g., in severe LV hypertrophy), the slope of
Ejection ends during the relaxation phase, the diastolic filling curve (see segment ab in
when the ventricular pressure falls below Fig. 9.4) becomes steeper. A “stiff” or poorly
that of the aorta and the aortic valve closes compliant ventricle reduces the ability of the
(point d). chamber to fill during diastole, resulting in a
As the ventricle continues to relax, its lower-than-normal ventricular end-diastolic
pressure declines while its volume remains volume. In this circumstance, the stroke vol-
constant because the mitral valve has not ume will be reduced while the end-systolic
yet opened (this phase is known as isovolu- volume remains unchanged.
metric relaxation). When the ventricular
pressure falls below that of the left atrium,
Alterations in Afterload
the mitral valve opens again (point a) and
the cycle repeats. If preload and contractility are held con-
Note that point b represents the pressure stant and afterload is augmented (e.g., in
and volume at the end of diastole, whereas high-impedance states such as hyper ten-
point d represents the pressure and volume sion or aortic stenosis), the pressure gener-
at the end of systole. The difference between ated by the left ventricle during ejection
the EDV and end-systolic volume (ESV) rep- increases. In this situation, more ventri-
resents the quantity of blood ejected during cular work is expended in overcoming the
contraction (i.e., the stroke volume). resistance to ejection and less fiber short-
Changes in any of the determinants of ening takes place. As shown in Figure 9-5B,
cardiac function are reflected by alterations an increase in afterload results in a higher
in the pressure-volume loop. By analyzing ventricular systolic pressure and a greater-
the effects of a change in an individual pa- than-normal LV end-systolic volume. Thus,
rameter (preload, afterload, or contractility) in the setting of increased afterload, the
on the pressure-volume relationship, the re- ventricular stroke volume (EDV − ESV) is
sulting alterations in ventricular pressure reduced.
Fig. 5 and stroke volume can be predicted (Fig. 9.5). The dependence of the end-systolic vol-
ume on afterload is approximately linear:
the greater the afterload, the higher the end-
Alterations in Preload
systolic volume. This relationship is depicted
If afterload and contractility are held con- in Figure 9.5 as the end-systolic pressure vol-
stant but preload is caused to increase (e.g., ume relation (ESPVR) and is analogous to the
by administration of intravenous fluids), left total tension curve in the isolated muscle ex-
ventricular EDV rises. This increase in pre- periments described earlier.
load augments the stroke volume via the
Frank-Starling mechanism such that the
Alterations in Contractility
ESV achieved is the same as it was before in-
creasing the preload. This means that the The slope of the ESPVR line on the pressure-
normal left ventricle is able to adjust its volume loop graph is a function of cardiac
stroke volume and effectively empty its con- contractility. In conditions of increased con-
tent to match its diastolic filling volume, as tractility, the ESPVR slope becomes more
10090-09_CH09.qxd 8/31/06 4:52 PM Page 232

232 Chapter Nine

Figure 9.5. The effect of varying preload, afterload, and contractility on the pressure-volume
loop. A. When arterial pressure (afterload) and contractility are held constant, sequential increases
(lines 1, 2, 3) in preload (measured in this case as end-diastolic volume [EDV]) are associated with loops
that have progressively higher stroke volumes but a constant end-systolic volume (ESV). B. When the
preload (EDV) and contractility are held constant, sequential increases (points 1, 2, 3) in arterial pres-
sure (afterload) are associated with loops that have progressively lower stroke volumes and higher end-
systolic volumes. There is a nearly linear relationship between the afterload and ESV, termed the end-
systolic pressure-volume relation (ESPVR). C. A positive inotropic intervention shifts the end-systolic
pressure-volume relation upward and leftward from ESPVR-1 to ESPVR-2, resulting in loop 2, which
has a larger stroke volume and a smaller end-systolic volume than the original loop 1.

steep; that is, it shifts upward and toward independent of the end-diastolic volume be-
the left. Hence, at any given preload or after- fore contraction.
load, the ventricle empties more completely The important physiologic concepts in
(the stroke volume increases) and results in this section are summarized here:
a smaller-than-normal end-systolic volume 1. Ventricular stroke volume is a function
(see Fig. 9.5C). Conversely, in situations of of preload, afterload, and contractility.
reduced contractility, the ESPVR line shifts SV rises when there is an increase in pre-
downward, consistent with a decline in load, a decrease in afterload, or augmented
stroke volume and a higher end-systolic vol- contractility.
ume. Thus, the end-systolic volume is depen- 2. Ventricular end-diastolic volume (or end-
dent on the afterload against which the ven- diastolic pressure) is often used as a rep-
tricle contracts and the inotropic state but is resentation of preload. The end-diastolic
10090-09_CH09.qxd 8/31/06 4:52 PM Page 233

Heart Failure 233

volume is influenced by the chamber’s Figure 9.7A depicts the effects of systolic Fig. 7
compliance. dysfunction owing to impaired contractility
3. Ventricular end-systolic volume depends on the pressure-volume loop. The ESPVR is
on the afterload and contractility but not shifted downward such that systolic empty-
on the preload. ing ceases at a higher-than-normal end-
systolic volume. As a result, the stroke vol-
ume falls. When normal pulmonary venous
PATHOPHYSIOLOGY return is added to the increased end-systolic
Chronic heart failure may result from a volume that has remained in the ventricle
wide variety of cardiovascular insults. The because of incomplete emptying, the dias-
etiologies can be grouped into those that tolic chamber volume increases, resulting in
cause heart failure because of (1) impaired a higher-than-normal end-diastolic volume
ventricular contractility, (2) increased after- and pressure. While that increase in preload
load, or (3) impaired ventricular filling. induces a compensatory rise in stroke vol-
Heart failure that results from an abnormal- ume (via the Frank-Starling mechanism),
ity of ventricular emptying (owing to im- impaired contractility and the reduced ejec-
paired contractility or excessive afterload) is tion fraction cause the end-systolic volume
termed systolic dysfunction, whereas heart to remain elevated.
failure caused by abnormalities of diastolic During diastole, the persistently elevated
relaxation or ventricular filling is termed LV pressure is transmitted to the left atrium
diastolic dysfunction. Approximately two (through the open mitral valve) and to the
thirds of patients with heart failure demon- pulmonary veins and capillaries. An elevated
strate predominantly systolic dysfunction, pulmonary capillary hydrostatic pressure,
and the remainder primarily suffer from di- when sufficiently high (usually >20 mm Hg),
astolic dysfunction. However, there is much results in the transudation of fluid into the
overlap, and many patients demonstrate pulmonary interstitium and symptoms of
both systolic and diastolic abnormalities. pulmonary congestion.
Fig. 6 Figure 9.6 presents a general schema of car-
diac conditions that may result in heart fail-
Diastolic Dysfunction
ure. Although this schema applies to chronic
forms of heart failure, a sudden overwhelm- Approximately one third of patients with
ing cardiac load (as may occur with an acute clinical heart failure have normal ventricular
hypertensive crisis [see Chapter 13], acute contractile (systolic) function. Many of these
myocardial infarction [see Chapter 7], or people demonstrate abnormalities of diastolic
acute valvular insufficiency [see Chapter 8]) function: either impaired early diastolic relax-
can result in an acute form of heart failure ation (an active, energy-dependent process),
(e.g., pulmonary edema), as discussed later increased stiffness of the ventricular wall (a
in this chapter. passive property), or both. Acute myocardial
ischemia is an example of a condition that
transiently inhibits energy delivery and dias-
Systolic Dysfunction
tolic relaxation. Conversely, left ventricular
In systolic dysfunction, the affected ven- hypertrophy, fibrosis, or restrictive cardiomy-
tricle has a diminished capacity to eject opathy (see Chapter 10) causes the LV walls to
blood because of impaired myocardial become chronically stiffened. The effect of im-
contractility or pressure overload (i.e., ex- paired diastolic function is reflected in the
cessive afterload). Loss of contractility may pressure-volume loop (see Fig. 9.7B): in dias-
result from destruction of myocytes, ab- tole, filling of the ventricle occurs at higher-
normal myocyte function, or fibrosis. Pres- than-normal pressures because the lower part
sure overload impairs ventricular ejection of the loop is shifted upward as a result of re-
by significantly increasing resistance to duced chamber compliance. Patients with di-
flow. astolic dysfunction often present with signs of
10090-09_CH09.qxd 8/31/06 4:52 PM Page 234

234 Chapter Nine

Figure 9.6. Mechanisms and examples of conditions that cause left-sided


heart failure.

vascular congestion because the elevated di- the right ventricle (RV) is a thin-walled,
astolic pressure is transmitted retrograde to highly compliant chamber that accepts its
the pulmonary and systemic veins. blood volume at low pressures and ejects
against a low pulmonary vascular resistance.
As a result of its high compliance, the RV has
Right-Sided Heart Failure
little difficulty accepting a wide range of fill-
Whereas the physiologic principles systolic ing volumes without significant changes in
and diastolic dysfunction may be applied to its filling pressures. Conversely, the RV is
both right-sided and left-sided heart failure, quite susceptible to failure in situations that
the two ventricles have distinctly different present a sudden increase in afterload, such
functions. Compared with the left ventricle, as acute pulmonary embolism.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 235

Heart Failure 235

Figure 9.7. Systolic dysfunction and the pressure-volume loop. A. The normal pressure-volume loop (solid line)
is compared with one demonstrating systolic dysfunction (dashed line). In systolic dysfunction caused by decreased
cardiac contractility, the end-systolic pressure-volume relation is shifted downward and rightward (from line 1 to line
2). As a result, the end-systolic volume (ESV) is increased (arrow). As normal venous return is added to that greater-
than-normal ESV, there is an obligatory increase in the end-diastolic volume (EDV) and pressure (preload), which serves
a compensatory function by partially elevating stroke volume toward normal via the Frank-Starling mechanism.
B. The pressure-volume loop of diastolic dysfunction resulting from increased stiffness of the ventricle (dashed line).
The passive diastolic pressure-volume curve is shifted upward (from line 1 to line 2) such that at any diastolic volume,
the ventricular pressure is higher than normal. The result is a decreased EDV (arrow) because of reduced filling of the
stiffened ventricle at a higher-than-normal end-diastolic pressure.

The most common cause of right-sided When the right ventricle fails, the ele-
heart failure is actually the presence of left- vated diastolic pressure is transmitted retro-
Tab. 2 sided heart failure (Table 9.2). In this situa- grade to the right atrium with subsequent
tion, excessive afterload confronts the right congestion of the systemic veins, accompa-
ventricle because of the elevated pulmonary nied by signs of right-sided heart failure. In-
vascular pressures that result from LV dys- directly, isolated right-heart failure may also
function. Isolated right-heart failure is less influence left-heart function: the decreased
common and usually reflects increased RV af- right ventricular output reduces blood re-
terload owing to diseases of the lung paren- turn to the LV (a decline in preload), leading
chyma or pulmonary vasculature. Right-sided to a drop in left ventricular stroke volume.
heart disease that results from a primary pul-
monary process is known as cor pulmonale,
COMPENSATORY MECHANISMS
which often leads to right-heart failure.
Several natural compensatory mechanisms
are called into action in patients with heart
failure, serving to buffer the fall in cardiac
TABLE 9.2. Examples of Conditions That output and helping to maintain sufficient
Cause Right-Sided Heart Failure blood pressure to perfuse the vital organs.
Cardiac causes These mechanisms include (1) the Frank-
Left-sided heart failure Starling mechanism, (2) neurohormonal
Pulmonic valve stenosis alterations, and (3) the development of
Right ventricular infarction ventricular hypertrophy and remodeling AQ2
Parenchymal pulmonary disease
(Fig. 9.8). Fig. 8
Chronic obstructive pulmonary disease
Interstitial lung disease (e.g., sarcoidosis)
Adult respiratory distress syndrome Frank-Starling Mechanism
Chronic lung infection or bronchiectasis
Pulmonary vascular disease As shown in Figure 9.3, heart failure caused
Pulmonary embolism by impaired left ventricular contractile func-
Primary pulmonary hypertension
tion causes a downward shift of the ven-
10090-09_CH09.qxd 8/31/06 4:52 PM Page 236

236 Chapter Nine

Figure 9.8. Compensatory mechanisms in heart failure. Both the Frank-


Starling mechanism (which is invoked by the rise in ventricular end-diastolic
volume) and myocardial hypertrophy (in response to pressure or volume
overload) serve to maintain forward stroke volume (dashed lines). However,
the chronic rise in EDV by the former and increased ventricular stiffness by
the latter cause an increase in atrial pressure, which may result in manifes-
tations of “backward” failure (e.g., pulmonary congestion in the case of left-
sided heart failure).

tricular performance curve. Therefore, at a nervous system, (2) the renin-angiotensin- AQ3
given preload, stroke volume is decreased aldosterone system, and (3) increased pro-
compared with normal. The reduced stroke duction of antidiuretic hormone (ADH). In
volume results in incomplete chamber emp- part, these mechanisms serve to increase
tying; as a result, the volume of blood that systemic vascular resistance, which helps to
accumulates in the ventricle during diastole maintain arterial perfusion to vital organs,
is higher than normal (see Fig. 9.3, point b). even in the setting of a reduced cardiac out-
This increased stretch on the myofibers, act- put. That is, because blood pressure (BP) is
ing via the Frank-Starling mechanism, in- equal to the product of cardiac output (CO)
duces a greater stroke volume on subsequent and total peripheral resistance (TPR),
contraction, which helps to empty the en-
BP = CO × TPR
larged left ventricle and preserve forward
cardiac output (see Fig 9.8). a rise in TPR induced by these compensatory
This beneficial compensatory mechanism mechanisms can nearly balance the fall in
has its limits, however. In the case of severe CO and, in the early stages of heart failure,
heart failure with marked depression of maintain fairly normal BP. In addition,
contractility, the curve may be nearly flat neurohormonal activation results in salt
at higher diastolic volumes, reducing the and water retention, which in turn increases
augmentation of cardiac output achieved intravascular volume and left ventricular
by increased filling. Concurrently in such a preload, maximizing stroke volume via the
circumstance, marked elevation of the end- Frank-Starling mechanism.
diastolic volume and pressure (which is trans- Although the acute effects of neurohor-
mitted retrograde to the left atrium, pul- monal stimulation are compensatory and
monary veins, and capillaries) may result in beneficial, chronic activation of these mech-
pulmonary congestion and edema (see Fig. anisms often ultimately proves deleterious
9.3, point c). to the failing heart and contributes to a pro-
gressive downhill course, as described later
Neurohormonal Alterations in the chapter.

Neurohormonal activation encompasses


Adrenergic Nervous System
three important compensatory mechanisms
in heart failure in response to decreased The fall in cardiac output in heart failure
Fig. 9 cardiac output (Fig. 9.9): (1) the adrenergic is sensed as decreased perfusion pressure
10090-09_CH09.qxd 8/31/06 4:52 PM Page 237

Heart Failure 237

Figure 9.9. Compensatory neurohormonal stimulation develops in response


to the reduced forward cardiac output and blood pressure of heart failure. In-
AQ8 creased activity of the sympathetic nervous system, renin-angiotensin-aldosterone sys-
tem, and antidiuretic hormone serve to support the cardiac output and blood pres-
sure (boxes). However, adverse consequences of these activations (dashed lines)
include an increase in afterload from excessive vasoconstriction (which may then im-
pede cardiac output) and excess fluid retention, which contributes to peripheral
edema and pulmonary congestion.

by baroreceptors in the carotid sinus and put (see Fig. 9.2). Vasoconstriction of the ve-
aortic arch. These receptors decrease their nous and arterial circulations is also initially
rate of firing in proportion to the fall in beneficial. Venous constriction augments
BP, and the signal is transmitted by the blood return to the heart, which increases
ninth and tenth cranial nerves to the car- preload and raises stroke volume through
diovascular control center in the medulla. the Frank-Starling mechanism, as long as
As a result, sympathetic outflow to the heart the ventricle is operating on the ascending
and peripheral circulation is increased, portion of its ventricular performance curve
and parasympathetic tone is diminished. (see Fig. 9.3). Arteriolar constriction in-
Three immediate consequences arise (see creases the peripheral vascular resistance and
Fig. 9.9): (1) an increase in heart rate, (2) aug- therefore helps to maintain blood pressure
mentation of ventricular contractility, and (BP = CO × TPR). The regional distribution of
(3) vasoconstriction caused by stimulation α-receptors is such that during sympathetic
of α-receptors on the systemic veins and stimulation, blood flow is redistributed to
arteries. vital organs (e.g., heart and brain) at the
The increased heart rate and ventricular expense of the skin, splanchnic viscera, and
contractility directly augment cardiac out- kidneys.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 238

238 Chapter Nine

Renin-Angiotensin-Aldosterone System also appears to contribute to systemic vaso-


constriction.
This system is also activated early in patients
with heart failure (see Fig. 9.9), mediated by
Although each of these neurohormonal
increased renin release. The main stimuli for
alterations in heart failure is initially benefi-
renin secretion from the juxtaglomerular
cial, continued activation typically proves
cells of the kidney in heart failure patients harmful. For example, the increased circu-
include (1) decreased renal artery perfusion lating volume and augmented venous return
pressure secondary to low cardiac output, to the heart may ultimately worsen engorge-
(2) decreased salt delivery to the macula ment of the lung vasculature, exacerbating
densa of the kidney owing to alterations in congestive pulmonary symptoms. Further-
intrarenal hemodynamics, and (3) direct more, the elevated arteriolar resistance
stimulation of juxtaglomerular β2-receptors increases the afterload against which the
by the activated adrenergic nervous system. failing left ventricle contracts and may there-
Renin is an enzyme that cleaves circulat- fore impair stroke volume and reduce car-
ing angiotensinogen to form angiotensin I, diac output (see Fig. 9.9). In addition, the in-
which is then rapidly cleaved by endothelial creased heart rate augments metabolic
cell–bound angiotensin-converting enzyme demand and can therefore further reduce the
(ACE) to form angiotensin II (AII), a potent performance of the failing heart. Continuous
vasoconstrictor (see Chapter 13). Increased sympathetic activation results in downregu-
AII constricts arterioles and raises total pe- lation of cardiac β-adrenergic receptors and
ripheral resistance, thereby helping to main- upregulation of inhibitory G proteins, con-
tain systemic blood pressure. In addition, tributing to a decrease in the myocardium’s
AII acts to increase intravascular volume sensitivity to circulating catecholamines and
by two mechanisms: (1) at the hypothala- a reduced inotropic response.
mus, it stimulates thirst and therefore water Chronically elevated levels of AII and al-
intake; and (2) at the adrenal cortex, it acts dosterone have additional detrimental ef-
to increase aldosterone secretion. The latter fects. They provoke the production of cy-
hormone promotes sodium reabsorption tokines (small proteins that mediate cell-cell
from the distal convoluted tubule of the kid- communication and immune responses),
ney into the circulation (see Chapter 17), activate macrophages, and stimulate fibro-
serving to augment intravascular volume. blasts, resulting in fibrosis and adverse re-
The rise in intravascular volume increases modeling of the failing heart.
left ventricular preload and thereby aug- Because the undesired consequences of
ments cardiac output via the Frank-Starling chronic neurohormonal activation even-
mechanism in patients on the ascending por- tually outweigh their benefits, much of
tion of the ventricular performance curve today’s pharmacologic therapy of heart fail-
(see Fig. 9.3). ure is designed to moderate these “compen-
satory” mechanisms, as examined later in
the chapter.
Antidiuretic Hormone
Secretion of this hormone (also termed va-
Natriuretic Peptides
sopressin) by the posterior pituitary is in-
creased in many patients with heart failure, In contrast to the adverse consequences of
presumably mediated through arterial the neurohormonal alterations described
baroreceptors, and by increased levels of AII. in the previous sections, the natriuretic
ADH contributes to increased intravascular peptides are natural “beneficial” hormones
volume because it promotes water retention secreted in heart failure in response to in-
in the distal nephron. The increased intra- creased intracardiac pressures. The best stud-
vascular volume serves to augment left ven- ied of these are atrial natriuretic peptide
tricular preload and cardiac output. ADH (ANP) and B-type natriuretic peptide (BNP).
10090-09_CH09.qxd 8/31/06 4:52 PM Page 239

Heart Failure 239

ANP is stored in atrial cells and released in maintain contractile force and counteracts
response to atrial distention. BNP is not de- the elevated ventricular wall stress (recall
tected in normal hearts but produced when that wall thickness is in the denominator of
ventricular myocardium is subjected to he- the LaPlace wall stress formula). However,
modynamic stress (e.g., in heart failure or because of the increased stiffness of the
during myocardial infarction). Recent stud- hypertrophied wall, these benefits come at
ies have shown a close relationship between the expense of higher-than-normal diastolic
serum BNP levels and the clinical severity of ventricular pressures, which are transmitted
heart failure. to the left atrium and pulmonary vascula-
Actions of the natriuretic peptides are me- ture (see Fig. 9.8).
diated by specific natriuretic receptors and The pattern of compensatory hypertro-
are largely opposite to those of the other phy and remodeling that develops depends
hormone systems activated in heart failure. on whether the ventricle is subjected to
They result in excretion of sodium and water, chronic volume or pressure overload. Chro-
vasodilatation, inhibition of renin secretion, nic chamber dilatation owing to volume
and antagonism of the effects of AII on overload (e.g., chronic mitral or aortic regur-
aldosterone and vasopressin secretion. Al- gitation) results in the synthesis of new sar-
though these effects are beneficial to patients comeres in series with the old, causing the
with heart failure, they are usually not suffi- myocytes to elongate. The radius of the ven-
cient to fully counteract the vasoconstric- tricular chamber therefore enlarges, doing so
tion and volume-retaining effects of the in proportion to the increase in wall thick-
other activated hormonal systems. ness, and is termed eccentric hypertrophy.
Another recently recognized substance Chronic pressure overload (e.g., caused by
that is released in heart failure is endothelin hypertension or aortic stenosis) results in the
AQ4 1, a potent vasoconstrictor, derived from en- synthesis of sarcomeres in parallel with the
dothelial cells lining the vasculature (see old (i.e., the myocytes thicken), termed con-
Chapter 6). Drugs designed to inhibit en- centric hypertrophy. In this situation, the
dothelin-1 receptors (and therefore blunt wall thickness increases without propor-
adverse vasoconstriction) improve LV func- tional chamber dilatation, and wall stress
tion in heart failure, but long-term benefits may therefore be reduced substantially.
have not yet been demonstrated. Such hypertrophy and remodeling help to
reduce wall stress and maintain contractile
force, but ultimately, ventricular function de-
Ventricular Hypertrophy
teriorates and causes the chamber to dilate
and Remodeling
out of proportion to wall thickness. When
Ventricular hypertrophy and remodeling this occurs, the excessive hemodynamic
are important compensatory processes that burden on the contractile units produces a
develop over time in response to hemody- downward spiral of deterioration with pro-
namic burdens. Wall stress (as defined ear- gressive heart failure symptomatology.
lier and in Chapter 6) is often increased in
developing heart failure because of either LV
MYOCYTE LOSS AND
dilatation (increased chamber radius) or the
CELLULAR DYSFUNCTION
need to generate high systolic pressures to
overcome excessive afterload (e.g., in aortic Impairment of ventricular function in heart
stenosis or hypertension). A sustained in- failure may result from the actual loss of
crease in wall stress (along with neurohor- myocytes and/or impaired function of living
monal and cytokine alterations) stimulates myocytes. The loss of myocytes may result
the development of myocardial hypertro- from cellular necrosis (e.g., from myocardial in-
phy and deposition of extracellular matrix. farction or exposure to cardiotoxic drugs such
This increased mass of muscle fibers serves as doxorubicin) or apoptosis (programmed cell
as a compensatory mechanism that helps to death). In apoptosis, genetic instructions
10090-09_CH09.qxd 8/31/06 4:52 PM Page 240

240 Chapter Nine

activate intracellular pathways that cause the


cell to fragment and undergo phagocytosis TABLE 9.3. Factors That May Precipitate
Symptoms in Compensated
by other cells, without an inflammatory re- Heart Failure
sponse. Factors implicated in triggering apop-
tosis in heart failure include elevated cate- Increased metabolic demands
cholamines, AII, inflammatory cytokines, Fever
Infection
and mechanical strain on the myocytes
Anemia
owing to the augmented wall stress. Tachycardia
Even viable myocardium in heart failure Hyperthyroidism
is abnormal at the ultrastructural and mole- Pregnancy
cular levels. Mechanical wall stress, neuro- Increased circulating volume (increased preload)
Excessive sodium content in diet
hormonal activation, and inflammatory
Excessive fluid administration
cytokines, such as tumor necrosis factor α Renal failure
(TNF-α), are believed to activate changes in Conditions that increase afterload
the genetic expression of contractile pro- Uncontrolled hypertension
teins, ion channels, catalytic enzymes, sur- Pulmonary embolism (increased right ventricular
afterload)
face receptors, and secondary messengers in
Conditions that impair contractility
the myocyte. Recent experimental evidence Negative inotropic medications
has demonstrated such changes at the sub- Myocardial ischemia or infarction
cellular level that affect intracellular calcium Ethanol ingestion
handling by the sarcoplasmic reticulum, Failure to take prescribed heart failure
medications
decrease the responsiveness of the myo
Excessively slow heart rate
filaments to calcium, impair excitation-
contraction coupling, and alter cellular en-
ergy production. The cellular factors consid-
ered the most important contributors to cient increase in output by the failing heart,
dysfunction in heart failure are (1) a reduced so that symptoms of cardiac insufficiency
cellular ability to maintain calcium homeo- are precipitated. Tachyarrhythmias precipi-
stasis, and/or (2) changes in the production, tate heart failure by decreasing diastolic ven-
availability, and utilization of high-energy tricular filling time and by increasing myo-
phosphates. However, the exact subcellular cardial oxygen demand. Excessively low
alterations that result in heart failure have heart rates directly cause a drop in cardiac
not yet been elucidated, and this remains output (remember, cardiac output = stroke
one of the most active areas of cardiovascu- volume × heart rate). An increase in salt in-
lar research. gestion, renal dysfunction, or failure to take
prescribed diuretic medications may in-
crease the circulating volume, thus promot-
PRECIPITATING FACTORS
ing systemic and pulmonary congestion.
Many patients with heart failure remain Uncontrolled hypertension depresses sys-
asymptomatic for extended periods either tolic function because of excessive afterload.
because the impairment is mild or because A large pulmonary embolism results in both
cardiac dysfunction is balanced by the com- hypoxemia (and therefore decreased my-
pensatory mechanisms described earlier. ocardial oxygen supply) and a substantial
Often clinical manifestations occur only in increase in right ventricular afterload. Is-
the presence of precipitating factors that in- chemic insults (i.e., myocardial ischemia or
crease the cardiac workload and tip the bal- infarction), ethanol ingestion, or negative
anced state into one of decompensation. inotropic medications (e.g., large doses of
Common precipitating factors are listed β-blockers and certain calcium channel block-
Tab. 3 in Table 9.3. For example, conditions of in- ers) can all depress myocardial contractility
creased metabolic demand such as fever or and precipitate symptoms in the otherwise
infection may not be matched by a suffi- compensated congestive heart failure patient.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 241

Heart Failure 241

CLINICAL MANIFESTATIONS the absence of pulmonary congestion, be-


cause reduced forward blood flow to the
The clinical manifestations of heart failure overworked respiratory muscles and accu-
result from impaired forward cardiac output mulation of lactic acid may also contribute
and/or elevated venous pressures, and relate to that sensation. Heart failure may initially
Tab. 4 to which of the ventricles has failed (Table
cause dyspnea only on exertion, but more
9.4). A patient may present with the chronic severe dysfunction results in symptoms at
progressive symptoms of heart failure de- rest as well.
scribed here or, in certain cases, with sudden Other manifestations of low forward out-
decompensation of left-sided heart function put in heart failure may include dulled men-
(e.g., acute pulmonary edema, as described tal status because of reduced cerebral perfu-
later in the chapter). sion and impaired urine output during the day
because of decreased renal perfusion. The
latter often gives way to increased urinary
Symptoms
frequency at night (nocturia) when, while
The most prominent symptom of chronic supine, blood flow is redistributed to the
left ventricular failure is dyspnea (breathless- kidney, promoting renal perfusion and di-
ness) on exertion. Controversy regarding uresis. Reduced skeletal muscle perfusion
the cause of this symptom has centered on may result in fatigue and weakness.
whether it is primarily a manifestation of Other congestive manifestations of heart
pulmonary venous congestion or decreased failure include orthopnea, paroxysmal noctur-
forward cardiac output. A pulmonary ve- nal dyspnea (PND), and nocturnal cough. Or-
nous pressure that exceeds approximately thopnea is the sensation of labored breath-
20 mm Hg leads to transudation of fluid into ing while lying flat and is relieved by
the pulmonary interstitium and congestion sitting upright. It results from the redistri-
of the lung parenchyma. The resulting re- bution of intravascular blood from the
duced pulmonary compliance increases the gravity-dependent portions of the body (ab-
work of breathing to move the same volume domen and lower extremities) toward the
of air. Moreover, the excess fluid in the in- lungs after lying down. The degree of or-
terstitium compresses the walls of the bron- thopnea is generally assessed by the number
chioles and alveoli, increasing the resistance of pillows on which the patient sleeps to
to airflow and requiring greater effort of res- avoid breathlessness. Sometimes, orthopnea
piration. In addition, juxtacapillary recep- is so significant that the patient may try to
tors (J receptors) are stimulated and mediate sleep upright in a chair.
rapid shallow breathing. The heart failure PND is severe breathlessness that awak-
patient can also suffer from dyspnea even in ens the patient from sleep 2 to 3 hours after

TABLE 9.4. Most Common Symptoms and Physical Findings in Heart Failure

Symptoms Physical Findings

Left-sided
Dyspnea Diaphoresis (sweating)
Orthopnea Tachycardia, tachypnea
Paroxysmal nocturnal dyspnea Pulmonary rales
Fatigue Loud P2
Right-sided
Peripheral edema Jugular venous distention
Right upper quadrant discomfort Hepatomegaly
(owing to hepatic enlargement)
Peripheral edema
10090-09_CH09.qxd 8/31/06 4:52 PM Page 242

242 Chapter Nine

retiring to bed. This frightening symptom 9.4). Patients with only mild impairment
results from the gradual reabsorption into may appear well. However, a patient with se-
the circulation of lower extremity intersti- vere chronic heart failure may demonstrate
tial edema after lying down, with subse- cachexia (a frail, wasted appearance) owing
quent expansion of intravascular volume in part to poor appetite and to the metabolic
and increased venous return to the heart demands of the increased effort of breathing.
and lungs. A nocturnal cough is another In decompensated left-sided heart failure,
symptom of pulmonary congestion and is the patient may appear dusky (decreased
produced by a mechanism similar to orthop- cardiac output) and diaphoretic (sweating
nea. Hemoptysis (coughing up blood) may because of increased sympathetic nervous
result from rupture of engorged bronchial activity), and the extremities are cool be-
veins. cause of peripheral arterial vasoconstriction.
In right-sided heart failure, the elevated Tachypnea (rapid breathing) is common. The
systemic venous pressures can result in ab- pattern of Cheyne-Stokes respiration may
dominal discomfort because the liver becomes also be present in advanced heart failure,
engorged and its capsule stretched. Similarly, characterized by periods of hyperventila-
anorexia (decreased appetite) and nausea tion separated by intervals of apnea (absent
may result from edema within the gastroin- breathing). This pattern is related to the pro-
testinal tract. Peripheral edema, especially in longed circulation time between the lungs
the ankles and feet, also reflects increased and respiratory center of the brain in heart
hydrostatic venous pressures. Because of the failure that interferes with the normal feed-
effects of gravity, it tends to worsen while back mechanism of systemic oxygenation.
the patient is upright during the day and Sinus tachycardia (resulting from increased
is often improved by morning after lying sympathetic nervous system activity) is also
supine at night. Even before peripheral ede- common. Pulsus alternans (alternating strong
ma develops, the patient may note an unex- and weak contractions detected in the pe-
pected weight gain resulting from the accu- ripheral pulse) may be present as a sign of ad-
mulation of interstitial fluid. vanced ventricular dysfunction.
The symptoms of heart failure are com- In left-sided heart failure, the auscultatory
monly graded according to the New York finding of pulmonary rales is created by the
Heart Association (NYHA) classification “popping open” of small airways that had
Tab. 5 (Table 9.5). A newer system classifies pa- been closed off by edema fluid before inspi-
tients according to their stage in the course ration. This finding is initially apparent at
Tab. 6 of heart failure (Table 9.6). the lung bases, where hydrostatic forces are
greatest; however, more severe pulmonary
congestion is associated with additional rales
Physical Signs
higher in the lung fields. Compression of con-
The physical signs of heart failure depend on duction airways by pulmonary congestion
the severity and chronicity of the condition may produce coarse rhonchi and wheezing;
and can be divided into two groups based on the latter finding in heart failure is termed
left and right-heart dysfunction (see Table cardiac asthma.

TABLE 9.5. New York Heart Association Classification of Heart Failure

Class Definition

I No limitation of physical activity.


II Slight limitation of activity. Dyspnea and fatigue with moderate physical activity (e.g., walk-
ing up stairs quickly).
III Marked limitation of activity. Dyspnea with minimal activity (e.g., slowly walking up stairs).
IV Severe limitation of activity. Symptoms are present even at rest.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 243

Heart Failure 243

TABLE 9.6. Stages of Heart Failure

Stage Description

A Patient who is at risk of developing heart failure but has not yet developed structural cardiac
dysfunction (e.g., patient with coronary artery disease, hypertension, or family history of
cardiomyopathy)
B Patient who has structural heart disease associated with heart failure but has not yet developed
symptoms
C Patient who has current or prior symptoms of heart failure associated with structural heart disease
D Patient who has structural heart disease and marked heart failure symptoms despite maximal med-
ical therapy and requires advanced interventions (e.g., cardiac transplantation)

Modified from Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelines for the evaluation and management of chronic heart
failure in the adult: executive summary. Circulation 2001;104:2996–3007.

Depending on the cause of heart failure, veins as well as hepatic enlargement with ab-
palpation of the heart may show that the left dominal right upper quadrant tenderness.
ventricular impulse is not focal but diffuse (in Edema accumulates in the dependent por-
dilated cardiomyopathy), sustained (in pres- tions of the body, beginning in the ankles
sure overload states such as aortic stenosis or and feet of ambulatory patients and in the
hypertension), or lifting in quality (in vol- presacral regions of those who are bedridden.
ume overload states such as mitral regurgita- Pleural effusions may develop in either
tion). Because elevated left-heart filling pres- left- or right-sided heart failure, because the
sures result in increased pulmonary vascular pleural veins drain into both the systemic
pressures, the pulmonic component of the and pulmonary venous beds. The presence
second heart sound is often louder than of pleural effusions is suggested on physical
normal. An early diastolic sound (S3) is fre- examination by dullness to percussion over
quently heard in adults with systolic heart the posterior lung bases.
failure and is caused by abnormal filling of
the dilated chamber (see Chapter 2). A late di-
Diagnostic Studies AQ5
astolic sound (S4) results from forceful atrial
contraction into a stiffened ventricle and is Normally, the mean left atrial (LA) pressure
common in states of decreased LV compli- is ≤10 mm Hg. If the LA pressure exceeds ap-
ance (diastolic dysfunction). The murmur of proximately 15 mm Hg, the chest radio-
mitral regurgitation is sometimes auscultated graph shows upper-zone vascular redistribu-
in left-sided heart failure if LV dilatation has tion, such that the vessels supplying the
stretched the valve annulus and spread the upper lobes of the lung are larger than those
papillary muscles apart from one another, thus supplying the lower lobes (see Fig. 3.5). This
preventing full closure of the mitral leaflets is explained as follows: When a patient is in
in systole. the upright position, blood flow is normally
In right-sided heart failure, different phys- greater to the lung bases than to the apices
ical findings may be present. Cardiac exami- because of the effect of gravity. Redistribu-
nation may reveal a palpable parasternal right tion of flow occurs with the development of
ventricular heave, representing RV enlargement, interstitial and perivascular edema, which is
or a right-sided S3 or S4 gallop. The murmur of most prominent at the lung bases (where the
tricuspid regurgitation may be auscultated and hydrostatic pressure is the highest). There-
is owing to right ventricular enlargement, fore, the blood vessels in the lung bases are
analogous to mitral regurgitation that devel- compressed, whereas flow into the upper
ops in LV dilatation. The elevated systemic lung zones is less affected.
venous pressure produced by right-heart fail- When the LA pressure surpasses 20 mm
ure is manifested by distention of the jugular Hg, interstitial edema is usually manifested
10090-09_CH09.qxd 8/31/06 4:52 PM Page 244

244 Chapter Nine

on the chest radiograph as indistinctness of ity is owing to refractory heart failure, but
the vessels and the presence of Kerley B lines many patients die suddenly, presumably be-
(short linear markings at the periphery of cause of ventricular arrhythmias.
the lower lung fields indicating interlobular Ventricular dysfunction usually begins
edema). If the LA pressure exceeds 25 to with an inciting insult but is a progressive
30 mm Hg, alveolar pulmonary edema may process, contributed to by the maladaptive
develop, with opacification of the air spaces. activation of neurohormones, cytokines, and
The relationship between LA pressure and continuous ventricular remodeling. Thus,
chest radiograph findings is modified in pa- it should not be surprising that measures
tients with chronic heart failure because of of neurohormonal and cytokine stimulation
enhanced lymphatic drainage, such that predict survival in heart failure patients. For
higher pressures can be accommodated with example, adverse prognosis correlates with
fewer radiologic signs. the serum norepinephrine level (marker
Depending on the cause of heart failure, of sympathetic nervous system activity),
the chest radiograph may show cardio- serum sodium (reduced level reflects activa-
megaly, defined as a cardiothoracic ratio of tion of renin-angiotensin-aldosterone sys- AQ6
greater than 0.5 on the posteroanterior film. tem and alterations in intrarenal hemody-
A high right atrial pressure also causes en- namics), endothelin 1, and cytokine TNF-α
largement of the azygous vein silhouette. levels.
Pleural effusions may be present. Despite the generally bleak prognosis, re-
Assays for BNP, described earlier in the cent studies have demonstrated that survival
chapter, correlate well with the degree of LV in heart failure patients can be substantially
dysfunction and prognosis. Furthermore, an
prolonged by specific interventions, as dis-
elevated serum level of BNP can help distin-
cussed in the following section.
guish heart failure from other causes of dys-
pnea, such as pulmonary diseases.
The cause of heart failure is often evident TREATMENT
from the history, such as a patient who has
sustained a large myocardial infarction, or There are five main goals of therapy in pa-
by physical examination, as in a patient with tients with chronic heart failure:
the murmur of mitral stenosis. When the
1. Identification and correction of the under-
cause is not clear from clinical evaluation,
lying condition causing heart failure. In
the first step is to determine whether systolic
some patients, this may require surgical
ventricular function is normal or depressed
repair or replacement of dysfunctional
(see Fig. 9.6). Of the several noninvasive tests
cardiac valves, coronary artery revascu-
that can help make this determination,
larization, aggressive treatment of hy-
echocardiography is especially useful (as de-
pertension, or cessation of alcohol con-
scribed in Chapter 3). In a minority of cases,
sumption.
cardiac catheterization is necessary to deter-
mine the cause of heart failure, including 2. Elimination of the acute precipitating cause
specific valvular and ischemic etiologies. of symptoms in the patient with heart
failure who was previously in a com-
pensated state. This may include, for
PROGNOSIS example, treating acute infections or ar-
The prognosis of heart failure is dismal in rhythmias, removing sources of excessive
the absence of a correctable underlying salt intake, or eliminating drugs that can
cause. Only 50% of patients remain alive aggravate symptomatology (e.g., certain
5 years after the diagnosis is made. Patients calcium channel blockers, which have a
with severe symptoms (i.e., NYHA class III or negative inotropic effect, or nonsteroidal
IV) fare the least well, having a 1-year sur- anti-inflammatory drugs, which can con-
vival rate of only 40%. The greatest mortal- tribute to volume retention).
10090-09_CH09.qxd 8/31/06 4:52 PM Page 245

Heart Failure 245

3. Management of heart failure symptoms: Diuretics


a. Treatment of pulmonary and systemic
The mechanisms of action of diuretic drugs
vascular congestion. This is most readily
are summarized in Chapter 17. By promot-
accomplished by dietary sodium re-
ing the elimination of sodium and water
striction and diuretic medications.
through the kidney, diuretics reduce in-
b. Measures to increase forward cardiac travascular volume and thus venous return
output and perfusion of vital organs to the heart. As a result, the preload of the left
through the use of vasodilators and ventricle is decreased, and its diastolic pres-
positive inotropic drugs. sure falls out of the range that promotes pul-
4. Modulation of the neurohormonal response monary congestion (Fig. 9.10, point b). The Fig. 10
to help prevent adverse ventricular re- judicious use of diuretics does not signifi-
modeling in order to slow progression of cantly reduce cardiac output in this setting,
LV dysfunction. because the heart is operating on the “flat”
5. Improvement of long-term survival. There portion of a depressed Frank-Starling curve.
is strong evidence that longevity is en- The intent is to reduce the end-diastolic pres-
hanced by specific interventions, as de- sure (and therefore hydrostatic forces con-
scribed later in the chapter. tributing to pulmonary congestion) without
Hypotension

Pulmonary congestion

Figure 9.10. Examples of the effect of heart failure treatment on the


left ventricular (LV) Frank-Starling curve. Point a represents the failing
heart on a curve that is shifted downward compared with normal. The stroke
volume is reduced (bordering on hypotension), and the LV end-diastolic pres-
sure (LVEDP) is increased, resulting in symptoms of pulmonary congestion.
Therapy with a diuretic or pure venous vasodilator (point b on the same
Frank-Starling curve) reduces LV pressure without much change in stroke vol-
ume (SV). However, excessive diuresis or venous vasodilatation may result in
an undesired fall in SV with hypotension (point b′). Inotropic drug therapy
(point c) and arteriolar (or “balanced”) vasodilator therapy (point d) augment
SV, and because of improved LV emptying during contraction, the LVEDP
lessens. Point e represents the potential added benefit of combining an in-
otrope and vasodilator together. The dashed line shows one example of how
the Frank-Starling curve shifts upward during inotropic/vasodilator therapy
but does not achieve the level of a normal ventricle.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 246

246 Chapter Nine

a significant fall in stroke volume. However, The pharmacology of these drugs is described
overly vigorous diuresis can lower the LV in Chapter 17.
filling pressures into the steep portion of the Venous vasodilators (e.g., nitrates) increase
ventricular performance curve, resulting in venous capacitance, decrease venous return
an undesired fall in cardiac output (see Fig. to the heart, and therefore reduce left ven-
9.10, point b′). Thus, diuretics should be used tricular preload. Consequently, LV diastolic
only if there is evidence of pulmonary con- pressures fall and the pulmonary capillary
gestion (rales) or peripheral interstitial fluid hydrostatic pressure declines, similar to the
accumulation (edema). hemodynamic effects of diuretic therapy. As
Agents that act primarily at the renal loop a result, pulmonary congestion improves,
of Henle (e.g., furosemide, torsemide, and and as long as the heart failure patient is on
bumetanide) are the most potent diuretics in the relatively “flat” part of the depressed
heart failure. Thiazide diuretics (e.g., hydro- Frank-Starling curve (see Fig. 9.10), the car-
chlorothiazide and metolazone) are also diac output does not fall despite the reduc-
useful but are less effective in the setting of tion in ventricular filling pressure. However,
decreased renal perfusion, which is often venous vasodilatation in a patient who is
present in this condition. operating on the steeper part of the curve
The potential adverse effects of diuretics may result in an undesired fall in stroke vol-
are described in Chapter 17. The most im- ume, cardiac output, and blood pressure.
portant in heart failure patients include Pure arteriolar vasodilators (e.g., hydrala-
overly vigorous diuresis resulting in a fall in zine) reduce systemic vascular resistance and
cardiac output, and electrolyte disturbances therefore LV afterload, which in turn permits
(particularly hypokalemia and hypomagne- increased ventricular muscle fiber shortening
semia), which may contribute to dangerous during systole (see Fig. 9.5B). This results in
arrhythmias. When diuretics are used in pa- an augmented stroke volume and is repre-
tients with pure LV diastolic dysfunction to sented on the Frank-Starling diagram as a
relieve congestive symptoms, extra care shift in an upward direction (see Fig. 9.10).
must be taken to avoid overdiuresis because Although an arterial vasodilator might be
these patients require elevated diastolic fill- expected to reduce blood pressure—an un-
ing pressures to adequately fill their stiff- desired effect in patients with heart failure
ened left ventricles (see Fig. 9.7B). It is there- who may already be hypotensive—this gen-
fore often necessary to accept some degree erally does not happen. As resistance is re-
of chronically elevated filling pressures in duced by arteriolar vasodilatation, a concur-
patients with LV diastolic dysfunction. rent rise in cardiac output usually occurs,
such that blood pressure remains constant
or decreases only mildly.
Vasodilators
Some groups of drugs result in vasodi-
One of the most important cardiac advances latation of both the venous and arteriolar
in the late twentieth century was the intro- circuits (“balanced” vasodilators). Of these,
duction of vasodilator therapy for the treat- the most important are the ACE inhibitors,
ment of heart failure, particularly the class which inhibit the formation of AII, a vaso-
of agents known as angiotensin-converting constrictor whose production is stimulated
enzyme (ACE) inhibitors. As indicated ear- in heart failure patients. In addition, be-
lier, neurohormonal compensatory mecha- cause aldosterone levels fall in response to
nisms in heart failure often lead to excessive ACE inhibitor therapy, sodium elimination
vasoconstriction, volume retention, and ven- is facilitated, resulting in reduced intravas-
tricular remodeling. Vasodilator drugs help cular volume and improvement of systemic
to reverse these adverse consequences. More- and pulmonary vascular congestion. ACE
over, multiple studies have shown that cer- inhibitors also augment circulating levels
tain vasodilator regimens significantly ex- of bradykinin (see Chapter 17), which is
tend survival in patients with heart failure. thought to play an important vasodilatory
10090-09_CH09.qxd 8/31/06 4:52 PM Page 247

Heart Failure 247

role in heart failure. As a result of these ef- that the addition of H-ISDN to standard heart
fects, ACE inhibitors limit maladaptive ven- failure therapy (e.g., a diuretic, β-blocker,
tricular remodeling in patients with heart ACE inhibitor, or ARB) in black patients im-
failure and following acute myocardial in- proved functional status and survival.
farction (see Chapter 7). Nesiritide (human recombinant B-type
Supporting the beneficial hemodynamic natriuretic peptide) is an intravenous va-
and neurohormonal blocking effects of ACE sodilator drug available for hospitalized pa-
inhibitors, many large clinical trials have tients with decompensated heart failure.
shown that the drugs reduce heart failure It causes rapid and potent vasodilatation,
symptoms, improve stamina, reduce the need reduces elevated intracardiac pressures, aug-
for hospitalization, and most importantly, ments forward cardiac output, and lessens
extend survival in patients with chronic the activation of the renin-angiotensin-
heart failure. Thus, ACE inhibitors are stan- aldosterone and sympathetic nervous sys- AQ7
dard first-line chronic therapy for patients tems. It promotes diuresis, reduces heart fail-
with LV systolic dysfunction. ure symptoms, and can be combined with
The renin-angiotensin-aldosterone sys- diuretics and positive inotropic drugs. How-
tem can also be therapeutically inhibited by ever, it is an expensive drug, and recent evi-
angiotensin II receptor blockers (ARBs), dence has raised questions about its safety.
as described in Chapters 13 and 17. Since One analysis shows that patients treated
angiotensin II can be formed by pathways with nesiritide are more likely to die over
other than ACE, ARBs provide a more the following month than are those receiv-
complete inhibition of the system through ing traditional heart failure therapies. Cur-
blockade of the actual AII receptor (see Fig. rently, therefore, nesiritide is used primarily
17.6). Conversely, ARBs do not stimulate in patients who have not responded to or
the potentially beneficial rise in serum cannot tolerate other intravenous vasodila-
bradykinin. The net result is that the hemo- tors, such as intravenous nitroglycerin or
dynamic effects of ARBs in heart failure are nitroprusside (see Chapter 17).
similar to those of ACE inhibitors, and stud-
ies thus far have not shown any superiority
Inotropic Drugs
of these agents over ACE inhibitors in terms
of patient survival. Thus, they are prescribed The inotropic drugs include β-adrenergic
to heart failure patients mainly when ACE agonists, digitalis glycosides, and phospho-
inhibitors are not tolerated (e.g., because of diesterase inhibitors (see Chapter 17). By in-
the side effect of cough). creasing the availability of intracellular cal-
Chronic therapy using the combination of cium, each of these drug groups enhances
the venous dilator isosorbide dinitrate plus the force of ventricular contraction and
the arteriolar dilator hydralazine has also therefore shifts the Frank-Starling curve in
been shown to improve survival in patients an upward direction (see Fig. 9.10). As a re-
with moderate symptoms of heart failure. sult, stroke volume and cardiac output are
However, when administration of the ACE augmented at any given ventricular end-
inhibitor enalapril was compared with the diastolic volume. Therefore, these agents
hydralazine–isosorbide dinitrate (H-ISDN) may be useful in treating patients with sys-
combination, the ACE inhibitor was shown tolic ventricular dysfunction but not those
to produce the greater improvement in sur- with pure diastolic failure.
vival. Thus, H-ISDN is generally substituted The b-adrenergic agonists (e.g., dobuta-
when a patient cannot tolerate ACE inhibitor mine and dopamine) are administered in-
or ARB therapy (e.g., because of renal insuffi- travenously for temporary hemodynamic
ciency or hyperkalemia). However, H-ISDN support in acutely ill, hospitalized patients.
appears to have particular benefit in African Their long-term use is limited by the lack of
Americans with heart failure. The recent an oral form of administration and by the
African American Heart Failure trial showed rapid development of drug tolerance. The
10090-09_CH09.qxd 8/31/06 4:52 PM Page 248

248 Chapter Nine

latter refers to the progressive decline in ef- remains unclear but may relate to the drugs’
fectiveness during continued administration effect on reducing heart rate and blunting
of the drug, possibly owing to downregula- chronic sympathetic activation or to their
tion of myocardial adrenergic receptors. Like- anti-ischemic properties.
wise, the role of phosphodiesterase inhibi- In clinical trials of patients with all classes
tors (e.g. amrinone and milrinone) is limited of symptomatic heart failure, β-blockers have
to the intravenous treatment of congestive been well tolerated in stable patients (i.e.,
heart failure in acutely ill patients. Despite those without recent deterioration of symp-
the initial promise of effective oral phospho- toms or active signs of volume overload)
diesterase inhibitors, studies thus far demon- and have resulted in improved mortality
strate reduced survival among patients re- rates and fewer hospitalizations compared
ceiving this treatment. with placebo. Not all β-blockers have been
One of the oldest forms of inotropic ther- tested in heart failure. Those that have and
apy is digitalis (see Chapter 17), which can have been found to be beneficial include
be administered intravenously or orally. carvedilol (a nonselective β1- and β2-receptor
Digitalis preparations enhance contractility, blocker with weak β-blocking properties)
reduce cardiac enlargement, improve symp- and the β1-selective metoprolol (in a sus-
toms, and augment cardiac output in pa- tained-release formulation). Despite these
tients with systolic heart failure. Digitalis benefits, β-blockers must be used cautiously
also increases the sensitivity of the baro- in heart failure to prevent acute deterio-
receptors, so that the compensatory sym- ration owing to their potentially negative
pathetic drive in heart failure is blunted, a inotropic effect. Regimens should be started
desired effect that reduces left ventricular af- at low dosages and augmented gradually.
terload. By slowing AV nodal conduction
and thereby reducing the rate of ventricular
Aldosterone Antagonist Therapy
contractions, digitalis has an added benefit
in patients with congestive heart failure There is evidence that chronic excess of al-
who have concurrent atrial fibrillation. Al- dosterone levels in heart failure may con-
though digitalis can improve symptomatol- tribute to cardiac fibrosis and adverse ventric-
ogy in heart failure patients, it has not been ular remodeling. Antagonists of this hormone
shown to improve long-term survival. Its (which have been used for decades as mild di-
use is thus limited to patients who remain uretics) have shown clinical benefit in heart
symptomatic despite other standard thera- failure patients. For example, in a clinical trial
pies or to help slow the ventricular rate if of patients with advanced heart failure who
atrial fibrillation is also present. Digitalis is were already taking an ACE inhibitor and di-
not useful in the treatment of LV diastolic uretics, the antagonist spironolactone sub-
dysfunction because it does not improve stantially reduced mortality rates and im-
ventricular relaxation properties. proved heart failure symptoms. Eplerenone,
a newer aldosterone antagonist, has been
shown to improve survival of patients with
b-Blockers
congestive heart failure after an acute myo-
Historically, β-blockers have been contra- cardial infarction. Although aldosterone an-
indicated in patients with systolic dysfunc- tagonists are well tolerated in carefully con-
tion because the negative inotropic effect of trolled studies, heart failure patients’ serum
the drugs would be expected to worsen symp- potassium levels need to be closely monitored
tomatology. Paradoxically, recent studies to prevent hyperkalemia, especially if there is
have actually shown that β-blockers have im- renal impairment or concomitant ACE inhi-
portant benefits in heart failure, including bitor therapy.
augmented cardiac output, reduced hemo-
dynamic deterioration, and improved sur- In summary, standard therapy of chronic
vival. The explanation for this observation congestive heart failure associated with LV
10090-09_CH09.qxd 8/31/06 4:52 PM Page 249

Heart Failure 249

systolic dysfunction should include several systolic function (e.g., left ventricular ejec-
drugs, the cornerstones of which are an ACE tion fraction ≤35%), regardless of the pres-
inhibitor and a β-blocker. An accepted se- ence of ventricular arrhythmias, because the
quence of therapy is to start with an ACE in- approach reduces the likelihood of sudden
hibitor, as well as a diuretic if pulmonary or cardiac death in this population.
systemic congestive symptoms are present. If
the patient is unable to tolerate the ACE in-
Cardiac Resynchronization Therapy
hibitor, then an ARB (or hydralazine plus
isosorbide dinitrate) may be substituted. For Intraventricular conduction abnormalities
patients without recent clinical deterioration with widened QRS complexes (especially left
or volume overload, a β-blocker should be bundle branch block) are common in pa-
added. Those with advanced (NYHA class IV) tients with advanced heart failure. Such ab-
heart failure may benefit from the addition normalities can actually contribute to cardiac
of an aldosterone antagonist. For persistent symptoms because of the uncoordinated pat-
symptoms, digoxin can also be prescribed. terns of right and left ventricular contraction.
Advanced biventricular pacemakers have
therefore been developed that stimulate both
Additional Therapies
ventricles simultaneously, thus resynchro-
Other therapies commonly administered to nizing the contractile effort. This technique
patients with systolic dysfunction include of biventricular pacing, also termed cardiac
(1) anticoagulation to prevent intracardiac resynchronization therapy (CRT), has been
thrombus formation if LV systolic function shown to augment left ventricular systolic
is severely impaired (a controversial therapy function with an accompanying reduction
because clear benefit has not yet been de- of LV size, improve exercise capacity, reduce
monstrated by clinical trials) and (2) treat- the frequency of heart failure exacerbations,
ment of atrial and ventricular arrhythmias and reduce mortality related to progressive
that frequently accompany chronic heart heart failure. A wealth of evidence now in-
failure. For example, atrial fibrillation is very dicates that CRT is appropriate for selected
common in heart failure patients, and con- patients with advanced systolic dysfunction
version back to sinus rhythm can substan- (LV ejection fraction ≤35%) with a prolonged
tially improve cardiac output. Ventricular QRS duration (>120 msec) and continued
arrhythmias are also frequent in this popu- symptoms of heart failure despite appropri-
lation and may lead to sudden death. The ate pharmacologic therapies.
antiarrhythmic drug that is most effective Because patients who receive CRT are
and least likely to provoke dangerous arrhyth- typically also candidates for an ICD, mod-
mias in heart failure patients is amiodarone. ern devices combine both functions in a sin-
However, studies of amiodarone for treat- gle, small implantable unit.
ment of asymptomatic ventricular arrhyth-
mias in heart failure have not shown a
Cardiac Replacement Therapy
consistent survival benefit. In addition, heart
failure patients with symptomatic or sus- A patient with severe LV dysfunction whose
tained ventricular arrhythmias, or those condition remains refractory to maximal
with inducible ventricular tachycardia medical management may be a candidate for
during electrophysiologic testing, benefit cardiac transplantation. Because of a short-
more from insertion of an implantable age of donor hearts, only approximately
cardioverter-defibrillator (ICD; see Chapter 2,500 transplants are performed in the
11). Based on the results of recent large-scale United States each year. Thus, alternative
randomized trials, ICD therapy is recom- mechanical heart support therapies are un-
mended for many patients with chronic is- dergoing intense development, including
chemic or nonischemic dilated cardiomy- ventricular assist devices and totally im-
opathies and at least moderately reduced planted artificial hearts.
10090-09_CH09.qxd 8/31/06 4:52 PM Page 250

250 Chapter Nine

Treatment of to peripheral vasoconstriction in response to


Diastolic Dysfunction the increased sympathetic outflow. Tachyp-
nea and coughing of “frothy” sputum repre-
Correctable causes of impaired diastolic func- sent transudation of fluid into the alveoli.
tion should be considered and addressed. For Rales are present initially at the bases and
example, pericardiectomy would be under- then throughout the lung fields, sometimes
taken for constrictive pericarditis (see Chap- accompanied by wheezing because of edema
ter 14), or therapy should be directed at within the conductance airways.
coronary artery disease if transient ischemia Pulmonary edema is a life-threatening
is the mechanism of diastolic dysfunction. emergency that requires immediate im-
Inotropic drugs or vasodilators usually have provement of systemic oxygenation and
no role in the treatment of pure diastolic elimination of the underlying cause. The pa-
dysfunction. Diuretics may reduce pulmo- tient should be seated upright to permit
nary congestion and peripheral edema but pooling of blood within the systemic veins
must be used cautiously to avoid compro- of the lower body, thereby reducing venous
mising cardiac output and causing hypoten- return to the heart. Supplemental oxygen is
sion, because the stiffened left ventricle relies provided by a face mask. Morphine sul-
on higher-than-normal filling pressures to phate is administered intravenously to re-
maintain its output. The roles of other po- duce anxiety and as a venous dilator to fa-
tentially promising medications in diastolic cilitate pooling of blood peripherally. A
dysfunction, including β-blockers, calcium rapidly acting diuretic, such as intravenous
channel blockers, ACE inhibitors, and ARBs furosemide, is administered to further re-
currently remain undefined. duce LV preload and pulmonary capillary
hydrostatic pressure. Other means of reduc-
ing preload include administration of ni-
ACUTE PULMONARY EDEMA
trates (often intravenously) or, in extreme
A severe, acute form of left-sided heart failure cases, venous phlebotomy. Inotropic drugs
is cardiogenic pulmonary edema, in which (e.g., dopamine) can also be administered
elevated capillary hydrostatic pressure causes intravenously to increase forward cardiac
rapid accumulation of fluid within the inter- output. During resolution of the pulmonary
stitium and alveolar spaces of the lung. In the congestion and hypoxemia, attention should
presence of normal plasma oncotic pressure, be directed at identifying and treating the
pulmonary edema develops when the pul- underlying cause.
monary capillary wedge pressure, which re- An easy-to-remember mnemonic for acute
flects LV diastolic pressure, exceeds approxi- management of pulmonary edema is the al-
mately 25 mm Hg. phabetic sequence LMNOP:
This condition is frequently accompanied
Lasix (trade name for furosemide)
by hypoxemia because of shunting of pul-
monary blood flow through regions of hypo- Morphine
ventilated alveoli. Pulmonary edema may ap- Nitrates
pear suddenly in a previously asymptomatic Oxygen
person in, for example, the setting of an Position (sit upright)
acute myocardial infarction or in patients
with chronic compensated congestive heart
SUMMARY
failure following a precipitating event (see
Table 9.3). Pulmonary edema is a horrifying 1. Heart failure is present when cardiac out-
experience for the patient, resulting in severe put fails to meet the metabolic demands
dyspnea and anxiety while struggling to of the body or meets those demands only
breathe. if the cardiac filling pressures are abnor-
On examination, the patient is tachycardic mally high. It most often results from im-
and demonstrates cold, clammy skin owing paired left ventricular systolic function,
10090-09_CH09.qxd 8/31/06 4:52 PM Page 251

Heart Failure 251

but may also arise from ventricular dias- Bardy G, Lee K, Mark D, et al., for the SCD-HeFT
tolic dysfunction and other cardiac ab- investigators. Amiodarone or an implantable
cardioverter-defibrillator for congestive heart fail-
normalities that interfere with ventricular
ure. N Engl J Med 2005;352:225–237.
filling or emptying, such as pericardial or
Cleland J, Daubert J-C, Erdmann E, et al., for CARE-
valvular disease. HF investigators. The effect of cardiac resynchro-
2. Compensatory mechanisms in heart fail- nization on morbidity and mortality in heart fail-
ure that help maintain circulatory func- ure. N Engl J Med 2005;352:1539–1549.
tion include (a) preload augmentation Dec GW, editor. Heart Failure: A Comprehensive
with increased stroke volume via the Guide to Diagnosis and Treatment. New York:
Marcel Dekker, 2005.
Frank-Starling mechanism, (b) activation
De Lemos J, McGuire, D, Drazner M. B-type natri-
of neurohormonal systems, and (c) ven- uretic peptide in cardiovascular disease. Lancet
tricular hypertrophy. However, these 2003;362:316–322.
compensations eventually become mal- Foody JM, Farrell MH, Krumholz HM. β-Blocker ther-
adaptive, contributing to adverse ven- apy in heart failure: scientific review. JAMA 2002;
tricular remodeling and progressive dete- 287:883–889.
rioration of ventricular function. Goodfriend TL. Aldosterone—a hormone of cardio-
vascular adaption and maladaption. J Clin Hyper-
3. Symptoms of heart failure may be exacer-
tens (Greenwich) 2006;8:133–139.
bated by precipitating factors that in-
Hunt SA, Abraham WT, Chin MH, et al. ACC/AHA
crease metabolic demand, increase circu- 2005 guideline update for the diagnosis and man-
lating volume, raise afterload, or decrease agement of chronic heart failure in the adult. Cir-
contractility (summarized in Table 9.3). culation 2005;112:1825–1852.
4. Successful treatment of heart failure re- Jessup M, Brozena S. Heart failure. N Engl J Med
quires identification of the underlying 2003;348:2007–2018.
cause of the condition, elimination of Jong P, Demers C, McKelvie RS, et al. Angiotensin re-
ceptor blockers in heart failure: meta-analysis of
precipitating factors, and modulation of randomized controlled trials. J Am Coll Cardiol
neurohormonal activations. Standard 2002;39:463–470.
medical treatment includes an ACE in- Katz AM. Heart Failure: Pathophysiology, Molecular
hibitor, β-blocker and, as needed, di- Biology, Clinical Management. Philadelphia: Lip-
uretics and inotropic drugs. For patients pincott Williams & Wilkins, 2000.
who do not tolerate an ACE inhibitor, an Maisel A. The coming age of natriuretic peptides.
ARB or the combination of hydralazine J Am Coll Cardiol 2006;47:61–64.
plus nitrates can be substituted. The ad- Morita H, Seidman J, Seidman CE. Genetic causes of
human heart failure. J Clin Invest 2005;115:518–
dition of spironolactone should be con-
526.
sidered for patients with advanced heart
Pitt B, Rajagopalan S. Aldosterone receptor antago-
failure. nists for heart failure: current status, future indi-
5. In some patients, cardiac resynchroniza- cations. Cleve Clin J Med 2006;73:257–268.
tion therapy and/or insertion of an im- Sackner-Bernstein J, Kowalski M, Fox M, Aaronson K.
plantable cardioverter-defibrillator should Short-term risk of death after treatment with ne-
be considered. siritide for decompensated heart failure: a pooled
analysis of randomized controlled trials. JAMA
2005;293:1900–1905.
Acknowledgment Stevenson LW, Shekar P. Ventricular assist devices for
durable support. Circulation 2005;112:111–115.
Contributors to the previous editions of this chapter Taylor A, Ziesche S, Yancy C, et al. Combination of
were Arthur Coday Jr, MD; George S. M. Dyer, MD; isosorbide dinitrate and hydralazine in blacks
Stephen K. Frankel, MD; Vikram Janakiraman, MD; with heart failure. N Engl J Med 2004;351:2049–
and Michael A. Fifer, MD. 2057.
Walsh RA, ed. Molecular Mechanisms of Cardiac
Hypertrophy and Failure. New York: Taylor &
Additional Reading
Francis, 2005.
Aurigemma G, Gaasch W. Diastolic heart failure. N Ware LB, Matthay MA. Acute pulmonary edema.
Engl J Med 2004;351:1097–1105. N Engl J Med 2005;353:2788–2796.
10090-09_CH09.qxd 8/31/06 4:52 PM Page a

Chapter 9—Author Queries


1. AU: Edited for consistency with chapter 6
2. AU: Edit reflects wording in the subsection that comes later. OK?
3. AU: As in heading below?
4. AU: As in ch. 6.
5. AU: Similar headings in all chapters edited for a certain level of consistency.
6. AU: Correct?
7. AU: OK?
8. AU: OK?
10090-10_CH10.qxd 8/31/06 4:53 PM Page 252

C H A P T E R

The Cardiomyopathies
Marc N. Wein
G. William Dec
10
Leonard S. Lilly

DILATED CARDIOMYOPATHY Pathophysiology


Etiology Clinical Findings
Pathology Physical Examination
Pathophysiology Diagnostic Studies
Clinical Findings Treatment
Physical Examination Prognosis
Diagnostic Studies RESTRICTIVE CARDIOMYOPATHY
Treatment Pathophysiology
Prognosis Clinical Findings
HYPERTROPHIC CARDIOMYOPATHY Physical Examination
Etiology Diagnostic Studies
Pathology Treatment

The cardiomyopathies are a group of heart ance and abnormal physiology of the left Fig. 1
disorders in which the major structural ab- ventricle (Fig. 10.1). Dilated cardiomy-
normality is limited to the myocardium. opathy is characterized by ventricular cham-
These conditions often result in symptoms of ber enlargement with impaired systolic
heart failure, and although the underlying contractile function; hypertrophic cardio-
cause of myocardial dysfunction can some- myopathy, by an abnormally thickened
times be identified, the etiology frequently ventricular wall with abnormal diastolic re-
remains unknown. Excluded from the defin- laxation but usually intact systolic func-
ition of this group of diseases is heart muscle tion; and restrictive cardiomyopathy, by
impairment resulting from other known car- an abnormally stiffened myocardium (be-
diac conditions, such as hypertension, valvu- cause of fibrosis or an infiltrative process)
lar disorders, or coronary artery disease. leading to impaired diastolic relaxation,
Cardiomyopathies can be classified into but systolic contractile function is normal
three types based on the anatomic appear- or near normal.

252
10090-10_CH10.qxd 8/31/06 4:53 PM Page 253

The Cardiomyopathies 253

Figure 10.1. Anatomic appearance of the cardiomyopathies (CMPs). A. Normal


heart demonstrating left ventricle (LV) and left atrium (LA). B. Dilated CMP is charac-
terized by ventricular dilatation with only mild hypertrophy. C. Hypertrophic CMP
demonstrates significant ventricular hypertrophy, often predominantly involving the
intraventricular septum. D. Restrictive CMP is caused by infiltration or fibrosis of the ven-
tricles, usually without enlargement of the cavities. LA enlargement is common to all
three types of CMP.

DILATED CARDIOMYOPATHY Acute viral myocarditis generally afflicts


young, previously healthy people. Common
Etiology responsible infecting organisms include
Cardiac enlargement in dilated cardiomy- coxsackievirus group B and adenovirus. It is
opathy (DCM) is caused by ventricular di- usually a self-limited illness with full recov-
latation with only minor hypertrophy. Myo- ery, but for unclear reasons, some patients
cyte damage leading to this condition results progress to DCM. It is hypothesized that
from a wide spectrum of genetic, inflamma- myocardial destruction and fibrosis result
Tab. 1 tory, toxic, and metabolic causes (Table 10.1). from immune-mediated injury triggered by
Although most cases are idiopathic (i.e., the viral constituents. Nonetheless, immuno-
cause is undetermined), examples of condi- suppressive drugs have not been shown to
tions that are commonly recognized causes improve the prognosis of this condition.
of DCM include viral myocarditis, alcohol Transvenous right ventricular biopsy during
toxicity, and specific gene mutations. acute myocarditis may demonstrate active
10090-10_CH10.qxd 8/31/06 4:53 PM Page 254

254 Chapter Ten

similar to those of other dilated cardiomyo-


TABLE 10.1. Examples of Dilated pathies. Alcoholic cardiomyopathy is im-
Cardiomyopathies
portant to identify because its cause is re-
Idiopathic versible; cessation of ethanol consumption
Familial (genetic) can lead to dramatic improvement of ven-
Inflammatory tricular function. Other potentially reversible
Infectious (especially viral)
causes of DCM include other toxin expo-
Noninfectious
Connective tissue diseases sures, metabolic abnormalities (such as hy-
Peripartum cardiomyopathy pothyroidism), and some inflammatory eti-
Sarcoidosis ologies, such as sarcoidosis or connective
Toxic tissue diseases.
Chronic alcohol ingestion
Several familial forms of DCM have been
Chemotherapeutic agents (e.g., doxorubicin)
Metabolic identified and are believed to be responsi-
Hypothyroidism ble for 20% to 30% of what were once clas-
Chronic hypocalcemia or hypophosphatemia sified as idiopathic DCM. Autosomal dom-
Neuromuscular inant, autosomal recessive, X-linked, and
Muscular or myotonic dystrophy
mitochondrial patterns of inheritance have
been described, leading to defects in con-
tractile force generation, force transmission,
inflammation, and viral genomic DNA or energy production, and myocyte viability.
RNA sequences have been demonstrated in Mutations identified thus far occur in genes
some infected persons. that code predominantly for cardiac cyto-
Alcoholic cardiomyopathy develops in a skeletal proteins, including troponin T, myo-
few people who consume alcoholic bever- sin, actin, and dystrophin (Table 10.2). In Tab. 2
ages chronically. Although the pathophysi- certain families, associated phenotypical
ology of the condition is unknown, ethanol features have included auditory deficits, car-
is thought to impair cellular function by in- diac conduction system defects, and skeletal
hibiting mitochondrial oxidative phospho- muscle abnormalities. Recognition of affected
rylation and fatty acid oxidation. Its clini- persons and identification of their underly-
cal presentation and histologic features are ing genetic mutations can allow gene-based

TABLE 10.2. Examples of Disease-Related Genes in Familial Forms of Dilated and


Hypertrophic Cardiomyopathies

Gene Protein Function

Mutations observed in familial dilated cardiomyopathy


DES Desmin Transduction of contractile forces
SGCD ∂-Sarcoglycan Transduction of contractile forces
DMD Dystrophin Transduction of contractile forces
MYH7 β-Myosin heavy chain Muscle contraction
ACTC Cardiac actin Muscle contraction
TNNT2 Cardiac troponin T Muscle contraction
TTN Titin Scaffold for sarcomere
LMNA Lamin A/C Nuclear membrane protein
ABCC Sur2A Inwardly rectifying potassium channel
Mutations observed in familial hypertrophic cardiomyopathy:
MYH7 β-Myosin heavy chain Muscle contraction
MYBP3 Myosin-binding protein C Muscle contraction
TNNT2 Cardiac troponin T Muscle contraction
TNNI3 Cardiac troponin I Muscle contraction
TPM1 α-Tropomyosin Muscle contraction
MYL3 Essential myosin light chain Muscle contraction
10090-10_CH10.qxd 8/31/06 4:53 PM Page 255

The Cardiomyopathies 255

screening and diagnosis for family members volume increases the stretch of the myofibers,
and potentially earlier interventions to pre- thereby increasing the subsequent stroke
vent the development of symptoms and volume; and (2) neurohormonal activation,
complications. initially mediated by the sympathetic ner-
vous system (see Chapter 9). The latter con-
tributes to an increased heart rate and con-
Pathology
tractility, which help to buffer the fall in
Marked enlargement of all four cardiac cham- cardiac output. These compensations may
Fig. 2 bers is typical of DCM (Fig. 10.2), although render the patient asymptomatic during the
sometimes the disease is limited to the left early stages of ventricular dysfunction; how-
or right side of the heart. The thickness of ever, as progressive myocyte degeneration
the ventricular walls may be increased, but and volume overload ensue, clinical symp-
chamber dilatation is out of proportion to toms of heart failure develop.
any hypertrophy. Microscopically, there is With a persistent reduction of cardiac
evidence of myocyte degeneration with ir- output, the decline in renal blood flow
regular hypertrophy and atrophy of myo- prompts the kidneys to secrete increased
fibers. Interstitial and perivascular fibrosis is amounts of renin. This activation of the
often extensive. renin-angiotensin-aldosterone axis increases AQ1
peripheral vascular resistance (mediated
through angiotensin II) and intravascular
Pathophysiology
volume (because of increased aldosterone).
The hallmark of DCM is ventricular dilata- As described in Chapter 9, these effects are
tion with decreased contractile function (Fig. also initially helpful in buffering the fall in
Fig. 3
10.3). Most often in DCM, both ventricles cardiac output.
are impaired, but sometimes dysfunction is Ultimately, however, the “compensatory”
limited to the left ventricle (LV) and even effects of neurohormonal activation prove
less commonly to the right ventricle (RV). detrimental. Arteriolar vasoconstriction and
As ventricular stroke volume and cardiac increased systemic resistance render it more
output decline because of impaired myocyte difficult for the LV to eject blood in the for-
contractility, two compensatory effects are ward direction, and the rise in intravascular
activated: (1) the Frank-Starling mechanism, volume further burdens the ventricles, re-
in which the elevated ventricular diastolic sulting in pulmonary and systemic conges-
tion. In addition, chronically elevated levels
of angiotensin II and aldosterone directly
contribute to pathological myocardial re-
modeling and fibrosis.
As the cardiomyopathic process causes
the ventricles to enlarge over time, the mi-
tral and tricuspid valves may fail to coapt
properly in systole, and valvular regurgi-
tation ensues. This regurgitation has three
detrimental consequences: (1) excessive vol-
ume and pressure loads are placed on the
atria, causing them to dilate, often leading
to atrial fibrillation; (2) regurgitation of blood
Figure 10.2. Transverse sections of a normal heart into the left atrium further decreases for-
(left) and a heart from a patient with dilated car-
diomyopathy (DCM). In the DCM specimen, there is ward stroke volume into the aorta and sys-
biventricular dilatation without a proportional increase in temic circulation; and (3) when the regurgi-
wall thickness. LV, left ventricle; RV, right ventricle. (Mod- tant volume returns to the LV during each
ified from Emmanouilides GC, ed. Moss and Adams’
Heart Disease in Infants, Children, and Adolescents. 5th Ed. diastole, an even greater volume load is pre-
AQ3 Baltimore: Lippincott Williams & Wilkins, 1995:86.) sented to the dilated LV.
10090-10_CH10.qxd 8/31/06 4:53 PM Page 256

256 Chapter Ten

Figure 10.3. Pathophysiology of dilated cardiomyopathy. The reduced


ventricular stroke volume results in decreased forward cardiac output and in-
creased ventricular filling pressures. The listed clinical manifestations follow.
JVD, jugular venous distention.

Clinical Findings congestion results in auscultatory crackles


(rales), and basilar chest dullness to percus-
The clinical manifestations of DCM are those sion may be present because of pleural effu-
of congestive heart failure. The most common sions. Cardiac examination shows an en-
symptoms of low forward cardiac output in- larged heart with leftward displacement of a
clude fatigue, lightheadedness, and exer- diffuse apical impulse. On auscultation, a
tional dyspnea associated with decreased tis- third heart sound (S3) is common as a sign of
sue perfusion. Pulmonary congestion results poor systolic function. The murmur of mitral
in dyspnea, orthopnea, and paroxysmal noc- valve regurgitation is often present as a result
turnal dyspnea, whereas chronic systemic ve- of the significant left ventricular dilatation. If
nous congestion causes ascites and peripheral right ventricular heart failure has developed,
edema. Because these symptoms may develop signs of systemic venous congestion may in-
insidiously, the patient may complain only clude jugular vein distention, hepatomegaly,
of recent weight gain (because of interstitial ascites, and peripheral edema. Right ventric-
edema) and shortness of breath on exertion. ular enlargement and contractile dysfunc-
tion are often accompanied by the murmur
Physical Examination of tricuspid valve regurgitation.

Signs of decreased cardiac output are often


Diagnostic Studies
present and include cool extremities (owing
to peripheral vasoconstriction), low arterial The chest radiograph shows an enlarged car-
pressure, and tachycardia. Pulmonary venous diac silhouette. If heart failure has developed,
10090-10_CH10.qxd 8/31/06 4:53 PM Page 257

The Cardiomyopathies 257

then pulmonary vascular redistribution, output are essentially the same as standard
interstitial and alveolar edema, and pleural therapies for heart failure (see Chapter 9). Ini-
effusions are evident (see Fig. 3.5). tial therapy typically includes salt restriction
The electrocardiogram (ECG) usually de- and diuretics, vasodilator therapy with an
monstrates atrial and ventricular enlarge- angiotensin-converting enzyme (ACE) inhibitor
ment. Patchy fibrosis of the myofibers results or angiotensin II receptor blocker (ARB), and a AQ2
in a wide array of arrhythmias, most impor- β-blocker. In patients with advanced heart
tantly atrial fibrillation and ventricular tachy- failure, the potassium-sparing diuretic spiro-
cardia. Conduction defects (left or right bun- nolactone may be added later. These measures
dle branch block) occur in most cases. Diffuse have been shown to improve symptoms and
repolarization (ST segment and T wave) ab- reduce mortality in patients with dilated
normalities are common. In addition, regions cardiomyopathy. The oral inotropic agent
of dense myocardial fibrosis may produce lo- digoxin may also be added to further improve
calized Q waves, resembling the pattern of left ventricular function and reduce symp-
previous myocardial infarction. toms, but it has not been shown to prolong
Echocardiography is very useful in the diag- survival.
nosis of DCM. It typically demonstrates four-
chamber cardiac enlargement with little hy-
Prevention and Treatment
pertrophy and global reduction of systolic
of Arrhythmias
contractile function. Mitral and/or tricuspid
regurgitation is also frequently visualized. Atrial and ventricular arrhythmias are com-
Cardiac catheterization is often performed mon in advanced DCM, and approximately
to determine whether coexistent coronary 40% of deaths in this condition are caused
artery disease is contributing to the impaired by ventricular tachycardia or fibrillation. It
ventricular function. This procedure is most is important to maintain serum electrolytes
useful diagnostically in patients who describe (notably, potassium and magnesium) within
episodes of angina pectoris or have evidence their normal ranges, especially during di-
suggestive of previous myocardial infarction uretic therapy, to avoid provoking serious ar-
on the electrocardiogram. Typically, hemo- rhythmias. Studies have shown that avail-
dynamic measurements show elevated right- able antiarrhythmic drugs do not prevent
and left-sided diastolic pressures and dimin- death from ventricular arrhythmias in DCM.
ished cardiac output. In the catheterization In fact, when used in patients with poor LV
laboratory, a transvenous biopsy of the right function, many antiarrhythmic drugs may
ventricle is sometimes performed to clarify worsen the rhythm disturbance. Amiodarone
the etiology of the cardiomyopathy. The role is the contemporary antiarrhythmic studied
of this procedure is limited, however, because most extensively in patients with DCM.
it rarely is diagnostic in patients with DCM Whereas there is no convincing evidence
and infrequently alters therapeutic decisions. that it reduces mortality from ventricular
arrhythmias in DCM, it is the safest anti-
arrhythmic for treating atrial fibrillation and
Treatment
other supraventricular arrhythmias in this
The goal of therapy in DCM is to relieve population. In contrast to antiarrhythmic
symptoms, prevent complications, and im- drugs, the placement of an implantable
prove long-term survival. Thus, in addition cardioverter-defibrillator (ICD) does reduce
to treating any identified underlying cause arrhythmic deaths in patients with DCM.
of DCM, therapeutic considerations include Therefore, based on large-scale randomized
those described in the following sections. trials, ICD placement is a recommended ap-
proach for patients with chronic sympto-
matic dilated cardiomyopathy and at least
Medical Treatment of Heart Failure
moderately reduced systolic function (e.g.,
Approaches for the relief of vascular conges- LV ejection fraction ≤35%), regardless of the
tion and improvement in forward cardiac detection of ventricular arrhythmias, be-
10090-10_CH10.qxd 8/31/06 4:53 PM Page 258

258 Chapter Ten

cause such an approach reduces the likeli- survival rates after transplantation are 74%
hood of sudden cardiac death. and 55%, respectively. However, the scarcity
Many patients with DCM have electrical of donor hearts greatly limits the availability
conduction abnormalities that contribute to of this technique. Fewer than 2,500 trans-
dyssynchronous ventricular contraction and plants are performed in the United States an-
reduced cardiac output. Electronic pacemak- nually compared with approximately 20,000
ers capable of stimulating both ventricles si- patients who could potentially benefit from
multaneously have been devised to better the procedure. As a result, other mechanical
coordinate systolic contraction as an adjunct options have been explored and continue to
to medical therapy (the technique is known undergo experimental refinements, includ-
as cardiac resynchronization therapy). Demon- ing ventricular assist devices and completely
strated benefits of this approach include im- implanted artificial hearts.
proved quality of life and exercise tolerance
as well as decreased hospitalizations for heart
Prognosis
failure and reduced mortality, particularly in
those with pretreatment left bundle branch Despite advances in therapy, the prognosis
block or other conduction abnormalities with for patients with DCM who do not undergo
a markedly prolonged QRS duration. cardiac transplantation is poor—the average
5-year survival rate is less than 50%. Methods
to reduce progressive LV dysfunction by early
Prevention of Thromboembolic Events
intervention in asymptomatic or minimally
Patients with DCM are at increased risk of symptomatic patients and the prevention of
thromboembolic complications because of sudden cardiac death remain major research
(1) stasis in the ventricles resulting from poor goals in the treatment of this disorder.
systolic function, (2) stasis in the atria owing
to chamber enlargement or atrial fibrillation,
HYPERTROPHIC
(3) an abnormally thrombogenic endocardial
CARDIOMYOPATHY
surface, and (4) venous stasis caused by poor
forward flow. Peripheral venous or right ven- Hypertrophic cardiomyopathy (HCM) has
tricular thrombus may lead to pulmonary received notoriety in the lay press because it
emboli, whereas thromboemboli of left ven- is the most common cardiac abnormality
tricular origin may lodge in any systemic found in young athletes who die suddenly
artery, resulting in, for example, devastating during vigorous physical exertion. With an
cerebral, myocardial, or renal infarctions. The incidence of about 1 of 500 in the general
only definite indications for systemic antico- population, HCM is characterized by septal
agulation in DCM patients are atrial fibrilla- or left ventricular hypertrophy that is not
tion, a previous thromboembolic event, or caused by chronic pressure overload (i.e., not
an LV thrombus visualized by echocardiog- the result of systemic hypertension or aortic
raphy. Additionally, chronic oral anticoagu- stenosis). Other terms frequently used to de-
lation therapy (i.e., warfarin) is often admin- scribe this disease are hypertrophic obstruc-
istered to DCM patients who have severe tive cardiomyopathy and idiopathic hyper-
depression of ventricular function (e.g., LV trophic subaortic stenosis. In this condition,
ejection fraction <30%); however, prospec- systolic LV contractile function is vigorous
tive studies are lacking to evaluate the effec- but the thickened muscle is stiff, resulting in
tiveness of this therapy in DCM patients who impaired ventricular relaxation and high
are in sinus rhythm. diastolic pressures.

Cardiac Transplantation Etiology


In suitable patients, cardiac transplantation HCM is a familial disease in which inheri-
offers a substantially better 5-year prognosis tance follows an autosomal dominant pattern
than the standard therapies for DCM previ- with variable penetrance, and a large variety
ously described. The current 5- and 10-year of mutations in at least 10 different genes 1 LINE LONG
10090-10_CH10.qxd 8/31/06 4:53 PM Page 259

The Cardiomyopathies 259

have been implicated. The proteins encoded


by the responsible genes are all part of the
sarcomere complex and include β-myosin
heavy chain (β-MHC), cardiac troponin T,
and myosin-binding protein C (see Table
10.2). The incorporation of these mutated
peptides into the sarcomere is thought to
cause impaired contractile function. The re-
sultant increase in myocyte stress is then
hypothesized to lead to compensatory hy-
pertrophy and proliferation of fibroblasts.
The pathophysiology and natural history
of familial HCM are quite variable and ap-
pear related to particular mutations within
the disease-causing gene, rather than the ac-
tual gene involved. In fact, it has been
shown that the precise genetic mutation de-
termines the age of onset of hypertrophy,
the extent and pattern of cardiac remodel-
ing, and the person’s risk of developing IVS
symptomatic heart failure or sudden death.
For example, mutations in the β-MHC gene
that alter electrical charge in the encoded
protein are associated with worse prognoses
than other mutations. It is hoped that better Figure 10.4. Postmortem heart specimen from a
patient with hypertrophic cardiomyopathy. Signifi-
definition of the natural history of specific cant left ventricular hypertrophy is seen, especially of the
mutations will allow accurate risk stratifica- interventricular septum (IVS).
tion of patients and permit appropriate tim-
ing of therapeutic interventions.
Pathophysiology
Pathology
The predominant feature of HCM is marked
Although hypertrophy in HCM may involve ventricular hypertrophy that reduces the
any portion of the ventricles, asymmetric hy- compliance and relaxation (diastolic func-
Fig. 4 pertrophy of the ventricular septum (Fig. tion) of the chamber, such that filling be-
10.4) is most common (approximately 90% comes impaired (Fig. 10.6). Patients who Fig. 6
of cases). Less often, the hypertrophy involves have asymmetric hypertrophy of the prox-
the ventricular walls symmetrically or is lo- imal interventricular septum may display
calized to the apex or midregion of the LV. additional findings related to transient ob-
Unlike ventricular hypertrophy resulting struction of left ventricular outflow during
from hypertension, in which the myocytes systole. In this case, the mechanism of sys-
enlarge uniformly and remain orderly, the tolic obstruction is thought to involve ab-
histology of HCM is unusual. The myocar- normal motion of the anterior mitral valve
dial fibers are in a pattern of extensive disar- leaflet toward the LV outflow tract where
Fig. 5 ray (Fig. 10.5). Short, wide, hypertrophied the thickened septum protrudes (Fig. 10.7). Fig. 7
fibers are oriented in chaotic directions and The process is explained as follows: (1) dur-
surrounded by numerous cardiac fibroblasts ing ventricular contraction, ejection of
and extracellular matrix. This myocyte dis- blood past the upper septum is more rapid
array and fibrosis are characteristic of HCM than usual, because it must flow through
and play a role in the abnormal diastolic an outflow tract that is narrowed by the
stiffness and the arrhythmias common to thickened septum; (2) this rapid flow cre-
1 LINE LONG this disorder. ates Venturi forces that abnormally draw
10090-10_CH10.qxd 8/31/06 4:53 PM Page 260

260 Chapter Ten

Figure 10.5. Light microscopy of the hypertrophic myocardium. A. Normal myocardium. B. Hypertrophic myocytes
resulting from pressure overload in a patient with valvular heart disease. C. Disordered myocytes with fibrosis in a pa-
tient with hypertrophic cardiomyopathy. (Modified from Schoen FJ. Interventional and Surgical Cardiovascular Pathol-
ogy: Clinical Correlations and Basic Principles. Philadelphia: WB Saunders, 1989:181.)

Figure 10.6. Pathophysiology of hypertrophic cardiomyopathy. The disarrayed and hypertro-


phied myocytes may lead to ventricular arrhythmias (which can cause syncope or sudden death) and
impaired diastolic left ventricular (LV) relaxation (which causes elevated LV filling pressures and dys-
pnea). If dynamic left ventricular outflow obstruction is present, mitral regurgitation often accom-
panies it (which contributes to dyspnea), and the impaired ability to raise cardiac output with exer-
tion can lead to exertional syncope. The thickened LV wall and systolic outflow tract obstruction both
contribute to increased myocardial oxygen consumption (MVO2) and can precipitate angina. CO,
cardiac output; LVEDP, LV end-diastolic pressure; LVH, LV hypertrophy.
10090-10_CH10.qxd 8/31/06 4:53 PM Page 261

The Cardiomyopathies 261

MR

Figure 10.7. Pathophysiology of left ventricular (LV) outflow obstruction and mitral re-
gurgitation in hypertrophic cardiomyopathy (HCM). Left panel. The LV outflow tract is
abnormally narrowed between the hypertrophied interventricular septum and the anterior
leaflet of the mitral valve (AML). It is thought that the rapid ejection velocity along the narrowed
tract in early systole draws the AML toward the septum (small arrow). Right panel. As the mi-
tral valve abnormally moves anteriorly and contacts the septum, outflow into the aorta is tran-
siently obstructed. Because the mitral leaflets do not coapt normally in systole, mitral regurgita-
tion (MR) also results.

the anterior mitral leaflet toward the sep- HCM With Outflow Obstruction
tum during contraction; and (3) the ante-
In patients with outflow obstruction, eleva-
rior mitral leaflet approaches and tran-
ted left atrial and pulmonary capillary wedge
siently abuts the hypertrophied septum,
pressures result from both the decreased
causing brief obstruction of blood flow into
ventricular compliance and the outflow ob-
the aorta. It is useful to consider the patho-
struction during contraction. During systolic
physiology of HCM based on whether tran-
obstruction, a pressure gradient develops be-
sient systolic outflow tract obstruction is
tween the main body of the LV and the out-
present. flow tract distal to the obstruction (see Fig.
10.7). The elevated ventricular systolic pres-
HCM Without Outflow Tract Obstruction sure increases wall stress and myocardial
oxygen consumption, which can result in
Although systolic contraction of the left ven- anginal chest discomfort (see Fig. 10.6). In ad-
tricle is usually vigorous in HCM, the hyper- dition, because obstruction is caused by ab-
trophied walls result in increased stiffness normal motion of the anterior mitral leaflet
and impaired diastolic relaxation of the toward the septum (and therefore away from
chamber. The reduced ventricular compli- the posterior mitral leaflet), the mitral valve
ance alters the normal pressure-volume re- does not close properly during systole, and
lationship, causing the passive diastolic fill- mitral regurgitation may result. This further
ing curve to shift upward (see Fig. 9.7B). The elevates left atrial and pulmonary venous
associated rise in diastolic LV pressure is pressures and may worsen symptoms of dys-
transmitted backward, leading to elevated left pnea, as well as contribute to the develop-
atrial, pulmonary venous, and pulmonary ment of atrial fibrillation.
capillary pressures. Dyspnea, especially dur- The systolic pressure gradient observed
ing exertion, is thus a common symptom in in obstructive HCM is dynamic in that its
this disorder. magnitude varies during contraction and de-
10090-10_CH10.qxd 8/31/06 4:53 PM Page 262

262 Chapter Ten

pends, at any given time, on the distance be- ertion, when the pressure gradient is made
tween the anterior leaflet of the mitral valve worse by the increased force of contraction,
and the hypertrophied septum. Situations thereby causing a transient fall in cardiac out-
that decrease LV cavity size (e.g., reduced ve- put. Orthostatic lightheadedness is also com-
nous return because of intravascular volume mon in patients with outflow tract obstruc-
depletion) bring the mitral leaflet and sep- tion. This occurs because venous return to the
tum into closer proximity and promote ob- heart is reduced on standing by the gravita-
struction. Conversely, conditions that enlarge tional pooling of blood in the lower extremi-
the LV (e.g., augmented intravascular vol- ties. The LV thus decreases in size and outflow
ume) increase the distance between the ante- tract obstruction intensifies, transiently re-
rior mitral leaflet and septum and reduce the ducing cardiac output and cerebral perfusion.
obstruction. Positive inotropic drugs (which When arrhythmias occur, symptoms of
augment the force of contraction) also force HCM may be exacerbated. For example, atrial
the mitral leaflet and septum into closer pro- fibrillation is not well tolerated because the
ximity and contribute to obstruction, where- loss of the normal atrial “kick” further im-
as negative inotropic drugs (e.g., β-blockers, pairs diastolic filling and can worsen symp-
verapamil) have the opposite effect. toms of pulmonary congestion. Of greatest
Although dynamic systolic outflow tract concern, the first clinical manifestation of
obstruction creates impressive murmurs and HCM may be ventricular fibrillation, resulting
receives great attention, the symptoms of in sudden cardiac death, particularly in young
obstructive HCM appear to primarily stem adults with HCM during strenuous physical
from the increased LV stiffness and diastolic exertion. Risk factors for sudden death
dysfunction also present in the nonobstruc- among patients with known HCM include a
tive form. history of syncope, a family history of sud-
den death, certain high-risk mutations, and
extreme hypertrophy of the LV wall (>30 mm
Clinical Findings in thickness).
The symptoms of HCM vary widely, from
none to significant physical limitations. The
Physical Examination
average age of presentation is the mid-20s.
The most frequent symptom is dyspnea Patients with mild forms of HCM are often
owing to elevated diastolic LV (and therefore asymptomatic and have normal or near-
pulmonary capillary) pressures. This symp- normal physical exams. A common finding
tom is further exacerbated by the high systo- is the presence of a fourth heart sound (S4),
lic LV pressure and mitral regurgitation seen generated by left atrial contraction into the
in patients with outflow tract obstruction. stiffened LV (see Chapter 2). The forceful
Angina is often described by patients with atrial contraction may also result in a palpa-
HCM, even in the absence of obstructive ble presystolic impulse over the cardiac apex
coronary artery disease. Myocardial ischemia (creating what is known as a double apical
may be contributed to by (1) the high oxy- impulse).
gen demand of the increased muscle mass Other findings are common in patients
and (2) the narrowed small branches of the with systolic outflow obstruction. The caro-
coronary arteries within the hypertrophied tid pulse rises briskly in early systole but
ventricular wall. If outflow tract obstruction then quickly declines as obstruction to car-
is present, the high systolic ventricular pres- diac outflow appears. The characteristic sys-
sure also increases myocardial oxygen de- tolic murmur of LV outflow obstruction is
mand because of the increased wall stress. rough and crescendo-decrescendo in shape,
Syncope in HCM may result from cardiac heard best at the left lower sternal border
arrhythmias that develop because of the (because of turbulent flow through the nar-
structurally abnormal myofibers (see the next rowed outflow tract). In addition, as the
section). In patients with outflow tract ob- stethoscope is moved toward the apex, the
struction, syncope may also be induced by ex- holosystolic blowing murmur of mitral re-
10090-10_CH10.qxd 8/31/06 4:53 PM Page 263

The Cardiomyopathies 263

nent Q waves are common in the inferior


TABLE 10.3. Effect of Maneuvers on Murmurs and lateral leads, representing the forces of
of Aortic Stenosis (AS) and Hyper-
trophic Cardiomyopathy (HCM) initial depolarization of the hypertrophied
septum. In some patients, diffuse T wave
Valsalva Squatting Standing inversions are present, which can predate
HCM murmur ↑ ↓ ↑ clinical, echocardiographic, or other electro-
AS murmur ↓ ↑ ↓ cardiographic manifestations of HCM. Atrial
and ventricular arrhythmias are frequent,
especially atrial fibrillation. Ventricular ar-
rhythmias are particularly ominous because
gurgitation may be auscultated. Although they may herald ventricular fibrillation and
the LV outflow obstruction murmur may be sudden death, even in previously asymptom-
soft at rest, bedside maneuvers that alter pre-
atic patients.
load and afterload can dramatically increase
Echocardiography is most helpful in the
its intensity and help differentiate this mur-
evaluation of HCM. The degree of LV hyper-
mur from other conditions, such as aortic
trophy can be measured and regions of
Tab. 3 stenosis (Table 10.3).
A commonly used technique in this re- asymmetrical wall thickness readily identi-
gard is the Valsalva maneuver, produced by fied. Signs of left ventricular outflow obstruc-
asking the patient to “bear down” (techni- tion may also be demonstrated and include
cally defined as forceful exhalation with the abnormal anterior motion of the mitral valve
nose, mouth, and glottis closed). The Val- as it is drawn toward the hypertrophied sep-
salva maneuver increases intrathoracic pres- tum during systole, and partial closure of the
sure, which decreases venous return to the aortic valve in midsystole as flow across it is
heart and transiently reduces LV size. This transiently obstructed. Doppler recordings
action brings the hypertrophied septum and during echocardiography accurately measure
anterior leaflet of the mitral valve into closer the outflow pressure gradient and quantify
proximity, creating greater obstruction to any associated mitral regurgitation. Children
forward flow. Thus, during Valsalva, the and adolescents with apparently mild HCM
murmur of HCM increases in intensity. In should undergo serial echocardiographic
contrast, the murmur of aortic stenosis de- assessment over time, because the degree of
creases in intensity during Valsalva because hypertrophy may increase during puberty
of the reduced flow across the stenotic valve. and early adulthood.
Conversely, a change from standing to a Cardiac catheterization is reserved for pa-
squatting position suddenly augments venous tients for whom the diagnosis is uncertain or
return to the heart (which increases preload) if percutaneous septal ablation (described in
while simultaneously increasing the sys-
the next section) is planned. The major fea-
temic vascular resistance somewhat. The in-
ture in patients with obstruction is the find-
creased preload raises the stroke volume and
ing of a pressure gradient within the outflow
therefore causes the murmur of aortic steno-
portion of the left ventricle, either at rest or
sis to become louder. In contrast, the tran-
during maneuvers that transiently reduce LV
sient increase in LV size during squatting re-
duces the LV outflow tract obstruction in size and promote outflow tract obstruction.
HCM and softens the intensity of that mur- Myocardial biopsy at the time of catheteri-
mur. Sudden standing from a squatting posi- zation is not necessary, because histologic
tion has the opposite effect on each of these findings do not predict disease severity or
murmurs (see Table 10.3). long-term prognosis.
Although genetic testing for HCM is
not feasible on a wide-scale basis, future
Diagnostic Studies
genotyping may provide a noninvasive
The ECG typically shows left ventricular hy- technique for definitive diagnosis and risk
pertrophy and left atrial enlargement. Promi- stratification.
10090-10_CH10.qxd 8/31/06 4:53 PM Page 264

264 Chapter Ten

Treatment have survived a cardiac arrest, have experi-


enced episodes of syncope, display high-risk
β-Blockers are the standard therapy for HCM ventricular arrhythmias, or have a ventricu-
because they (1) reduce myocardial oxygen lar wall thickness >30 mm) should receive
demand by slowing the heart rate and the an ICD.
force of contraction (and therefore diminish Infective endocarditis can develop in pa-
angina and dyspnea); (2) lessen the LV out- tients with obstructive HCM because of tur-
flow gradient during exercise by reducing bulent blood flow through the narrowed LV
the force of contraction (allowing the cham- outflow tract and in association with the ac-
ber size to increase, thus separating the an- companying mitral regurgitation. Antibiotic
terior leaflet of the mitral valve from the prophylaxis (see Chapter 8) is therefore indi-
ventricular septum); (3) increase passive di- cated to prevent endocardial infection during
astolic ventricular filling time owing to the surgical procedures that result in bacteremia.
decreased heart rate; and (4) decrease the Some studies have shown clinical im-
frequency of ventricular ectopic beats. De- provement when patients with obstructive
spite their antiarrhythmic effect, β-blockers HCM are treated with a dual-chamber perma-
have not been shown to prevent sudden ar- nent pacemaker, the electrodes of which are
rhythmic death in this condition. placed in the right atrium and right ventricle.
Calcium channel antagonists can reduce The LV outflow gradient may become re-
ventricular stiffness and are sometimes useful duced by this procedure, possibly by altering
in improving exercise capacity in patients the normal sequence of ventricular contrac-
whose conditions fail to respond to β-block- tion, such that septal-mitral valve apposition
ers. Patients who develop pulmonary con- becomes less prominent. This technique
gestion may benefit from mild diuretic ther- seems to be useful for only a small percentage
apy, but these drugs must be administered of markedly symptomatic patients.
cautiously to avoid volume depletion; re- Surgical therapy (myomectomy) is consid-
duced intravascular volume decreases LV size ered for patients whose symptoms do not re-
and could exacerbate outflow tract obstruc- spond to pharmacologic therapy. This pro-
tion. Vasodilators (including nitrates) simi- cedure involves excision of portions of the
larly reduce LV size and should be avoided. hypertrophied muscle mass and usually im-
Atrial fibrillation is poorly tolerated in proves outflow tract obstruction, symptoms,
HCM and should be controlled aggressively, and exercise capacity. A less invasive alter-
most commonly with antiarrhythmic drugs. Ef- native is percutaneous septal ablation, per-
fective antiarrhythmics for atrial fibrillation formed in the cardiac catheterization labora-
in HCM include amiodarone and disopyra- tory, in which ethanol is injected directly
mide (a type IA antiarrhythmic drug that also into the first major septal coronary artery
possesses negative inotropic properties that (a branch of the left anterior descending
may help reduce LV outflow tract obstruc- artery), causing a small, controlled myocar-
tion; see Chapter 17). Digitalis should be dial infarction. This results in reduced septal
avoided in HCM because its positive inotropic thickness and can lessen outflow tract ob-
effect increases the force of contraction and struction. Randomized trials have not been
can worsen LV outflow tract obstruction. performed to compare the results of surgical
Sudden cardiac death has a propensity to myomectomy with ethanol septal ablation.
occur in patients with HCM in association Although septal ablation is an appropriate
with physical exertion; therefore, strenuous consideration for patients who cannot toler-
exercise and competitive sports should be ate surgery, surgical myomectomy remains
avoided. Sudden death in this syndrome is the current procedure of choice given its
almost always caused by ventricular tachy- 40-year history of experience and efficacy.
cardia or fibrillation. Although amiodarone Finally, genetic counseling should be pro-
may reduce the frequency of ventricular ar- vided to all patients with HCM. Because it is
rhythmias, HCM patients who are at high an autosomal dominant disease, children of
risk of sudden cardiac death (i.e., those who affected persons have a 50% chance of in- 1 LINE LONG
10090-10_CH10.qxd 8/31/06 4:53 PM Page 265

The Cardiomyopathies 265

heriting the abnormal gene. In addition,


first-degree relatives of patients with HCM TABLE 10.4. Examples of Restrictive
Cardiomyopathy
should be screened by physical examination,
electrocardiography, and echocardiography. Myocardial Endomyocardial
Even asymptomatic HCM patients are at in-
Noninfiltrative Endomyocardial fibrosis
creased risk of complications, including sud- Idiopathic Hypereosinophilic syndrome
den death, and must be monitored closely. Scleroderma Metastatic tumors
Infiltrative Radiation therapy
Amyloidosis
Prognosis Sarcoidosis
Storage diseases
The incidence of sudden death in HCM is Hemochromatosis
2% to 4% per year in adults and 4% to 6% in Glycogen storage
children and adolescents. It has become diseases
clear that different mutations have vastly
different phenotypes. Some cause extreme
hypertrophy in childhood without any clin-
ical symptoms until the occurrence of sud- by a plasma cell tumor (usually, multiple
den death; others manifest later in life with myeloma). In contrast, secondary amyloidosis is
heart failure symptoms. Most mutations characterized by the presence of AA amyloid
produce only mild hypertrophy and are as- (derived from the inflammatory marker serum
sociated with a normal life expectancy. As amyloid A) in a variety of chronic inflamma-
the clinical outcomes of specific mutations tory conditions, such as rheumatoid arthritis.
are better defined, the use and timing of spe- Less common is hereditary amyloidosis, an au-
cific therapeutic interventions will likely be tosomal dominant condition in which amy-
loid fibrils form from point mutations in the
clarified.
protein transthyretin (formerly known as pre-
albumin). Senile amyloidosis is common after
RESTRICTIVE CARDIOMYOPATHY age 80, in which amyloid deposits, derived
from transthyretin or other proteins, are found
The restrictive cardiomyopathies are less
scattered throughout the vascular system,
common than DCM and HCM. They are
muscles, kidney, and lung. In each form of
characterized by abnormally rigid (but not
amyloidosis, cardiac involvement is marked
necessarily thickened) ventricles with im-
by deposition of extracellular amyloid be-
paired diastolic filling but usually normal, or
tween myocardial fibers in the atria and ven-
near-normal, systolic function. This condi- tricles, in the coronary arteries and veins, and
tion results from either (1) fibrosis or scar- in the heart valves.
ring of the endomyocardium or (2) infiltra- Clinical manifestations of cardiac in-
tion of the myocardium by an abnormal volvement are most common in the pri-
Tab. 4 substance, such as amyloid (Table 10.4). mary (AL) form of the disease and typically
The most common recognized cause of relate to the development of restrictive car-
restrictive cardiomyopathy in nontropical diomyopathy (described later in the chap-
countries is amyloidosis. In this systemic ter) because of the infiltrating amyloid pro-
disease, insoluble misfolded amyloid fibrils tein. Heart failure resulting from systolic
deposit within tissues, including the heart, ventricular dysfunction also occurs, but less
causing organ dysfunction. Amyloid deposi- frequently. Orthostatic hypotension devel-
tion is diagnosed histologically by the Congo ops in about 10% of patients, likely con-
red stain, which displays amyloid fibrils with tributed to by amyloid deposition in the
a characteristic green birefringence. autonomic nervous system and peripheral
Amyloid fibrils can pathologically develop blood vessels. Infiltration of amyloid into
from a host of different proteins that distin- the cardiac conduction system can cause
guish the categories of the disease. Primary arrhythmias and conduction impairments,
amyloidosis is caused by deposition of immu- which can result in syncope or even sudden
1 LINE LONG noglobulin light chain AL fragments secreted death.
10090-10_CH10.qxd 8/31/06 4:53 PM Page 266

266 Chapter Ten

Pathophysiology paradoxically worsen with inspiration (the


Kussmaul sign) because the stiffened right
Reduced compliance of the ventricles owing ventricle cannot accommodate the increased
to fibrosis or infiltration results in an up- venous return.
ward shift of the passive ventricular filling
curve (see Fig. 9.7B), leading to abnormally
high intraventricular pressure throughout Diagnostic Studies
diastole. This condition has two major con- The chest radiograph usually shows a normal-
sequences: (1) elevated systemic and pul- sized heart with signs of pulmonary conges-
monary venous pressures, with signs of right- tion. The ECG often displays nonspecific ST
and left-sided vascular congestion, and (2) and T wave abnormalities; conduction dis-
reduced ventricular cavity size with de- turbances such as atrioventricular block or a
creased stroke volume and cardiac output. bundle branch block may be present.
The restrictive cardiomyopathies share
Clinical Findings nearly identical symptoms, physical signs,
and hemodynamic profiles with constrictive
It follows from the underlying pathophysi- pericarditis, as described in Chapter 14. How-
ology that signs of left- and right-sided heart ever, it is important to distinguish between
Fig. 8 failure are expected (Fig. 10.8). Decreased these two entities, because constrictive peri-
cardiac output is manifested by fatigue and carditis is a correctable condition, whereas
decreased exercise tolerance. Systemic con- the restrictive cardiomyopathies generally
gestion (often more prominent than pul- are not treatable.
monary congestion in this syndrome) leads The most useful diagnostic tools to dif-
to jugular venous distention, peripheral ferentiate restrictive cardiomyopathy from
edema, and ascites with a large, tender liver. constrictive pericarditis are transvenous en-
Arrhythmias such as atrial fibrillation are domyocardial biopsy, computed tomogra-
common. Infiltrative etiologies that involve phy (CT), and magnetic resonance imaging
the cardiac conduction system can cause (MRI). For example, in restrictive cardio-
various types of heart block. myopathy, a transvenous endomyocardial
biopsy may demonstrate the presence of
infiltrative matter such as amyloid, iron de-
Physical Examination posits (hemochromatosis), or metastatic
Signs of congestive heart failure are often pre- tumors. Conversely, CT or MRI scans are
sent, including pulmonary rales, distended useful to identify the thickened pericar-
neck veins, ascites, and peripheral edema. dium of constrictive pericarditis, a finding
Similar to constrictive pericarditis (see that is not expected in restrictive cardio-
Chapter 14), jugular venous distention may myopathy.

Hepatomegaly and ascites

Figure 10.8. Pathophysiology of restrictive cardiomyopathy. The rigid myocardium results in elevated
ventricular diastolic pressures and decreased ventricular filling. The resultant symptoms can be predicted
from these abnormalities. CO, cardiac output.
10090-10_CH10.qxd 8/31/06 4:53 PM Page 267

The Cardiomyopathies 267

Treatment forward cardiac output. Some restrictive


cardiomyopathies are prone to intraven-
Restrictive cardiomyopathy typically has a tricular thrombus formation, thus warrant-
very poor prognosis, except when treat- ing chronic oral anticoagulant therapy is
ment can target an underlying cause. For warranted. In the case of primary (AL) amy-
example, phlebotomy and iron chelation loidosis, chemotherapy followed by auto-
therapy may be helpful in the early stages logous bone marrow stem cell transplant
of hemochromatosis. Symptomatic therapy has proved effective in selected patients with
for all etiologies includes salt restriction and early cardiac involvement.
cautious use of diuretics to improve symp-
toms of systemic and pulmonary conges-
tion. Unlike the dilated cardiomyopathies,
SUMMARY
digitalis and vasodilators are not helpful 1. The cardiomyopathies are diseases of heart
because systolic function is usually pre- muscle classified by their pathophysio-
served. Maintenance of sinus rhythm is im- logic presentation into dilated, hypertro-
portant to maximize diastolic filling and phic, or restrictive types (Table 10.5). Tab. 5

TABLE 10.5. Summary of the Cardiomyopathies

Dilated Hypertrophic Restrictive


Cardiomyopathy Cardiomyopathy Cardiomyopathy

Ventricular Dilated LV with little Marked hypertrophy, Fibrotic or infiltrated


morphology hypertrophy often asymmetric myocardium

Symptoms Fatigue, weakness, dysp- Dyspnea, angina, syncope Dyspnea, fatigue


nea, orthopnea, PND
(symptoms of conges-
tive heart failure)
Physical exam Pulmonary rales, S3; if RV S4; if outflow obstruction Signs of RV failure: JVD,
failure present: JVD, he- present: systolic mur- hepatomegaly, periph-
patomegaly, peripheral mur loudest at left ster- eral edema
edema nal border, accompa-
nied by mitral
regurgitation
Pathophysiology Impaired systolic Impaired diastolic relax- “Stiff” LV with impaired
contraction ation; LV systolic diastolic relaxation but
function vigorous, normal systolic function
often with dynamic
obstruction
Cardiac size on Dilated Normal or dilated Usually normal
chest radiograph
Echocardiogram Dilated, poorly contrac- LV hypertrophy, often Usually normal systolic
tile LV more pronounced in contraction; “speckled”
septum; systolic anterior appearance in infiltra-
movement of MV with tive disorders
mitral regurgitation

JVD, jugular venous distension; LV, left ventricle; MV, mitral valve; PND, paroxysmal nocturnal dyspnea; RV, right ventricle.
10090-10_CH10.qxd 8/31/06 4:53 PM Page 268

268 Chapter Ten

2. Dilatedcardiomyopathiesarecharacterized Dec GW, Fuster VF. Idiopathic dilated cardiomyopa-


by ventricular dilatation with impaired thy. N Engl J Med 1994;331:1564–1575.
systolic function. Progressive left ventricu- Falk RH. Diagnosis and management of the cardiac
amyloidoses. Circulation 2005;112:2047–2060.
lar enlargement often leads to sympto-
Kushwaha SS, Fallon JT, Fuster V. Medical progress: re-
matic heart failure, ventricular arrhyth-
strictive cardiomyopathy. N Engl J Med 1997;336:
mias, and/or embolic complications. 267–276.
3. HCM is characterized by a thickened left Magnani JW, Dec GW. Myocarditis: current trends
ventriclewithimpaireddiastolicrelaxation. in diagnosis and treatment. Circulation 2006;113:
Dynamic LV outflow tract obstruction 876–890.
during systole may be present. The most Morita H, Seidman J, Seidman CE. Genetic causes
common symptoms are dyspnea and exer- of human heart failure. J Clin Invest 2005;115:
518–526.
tional angina. Ventricular arrhythmias
Nishimura RA, Holmes DR Jr. Hypertrophic obstruc-
may lead to sudden cardiac death.
tive cardiomyopathy. N Engl J Med 2004;350:1320–
4. The restrictive cardiomyopathies are un- 1327.
common and are characterized by impair- Richardson P, McKenna W, Bristow M, et al. Report of
ment of diastolic ventricular relaxation the 1995 World Health Organization/International
owing to an infiltrated or fibrotic myo- Society and Federation of Cardiology Task Force
cardium. Symptoms of heart failure are on the definition and classification of cardio-
myopathies. Circulation 1996;93:841–842.
typical.
Seidman J, Seidman CE. Proposal for contemporary
screening strategies in families with hypertrophic
Acknowledgment cardiomyopathy. J Am Coll Cardiol 2004;44:2125–
2132.
The authors thank Frederick Schoen, MD, for pro-
Skinner M, Sanchorawala V, Seldin DC, et al. High-
viding pathology specimens. Contributors to the
dose melphalan and autologous stem-cell trans-
previous editions of this chapter were Yi-Bin Chen,
MD; Kay Fang, MD; David Grayzel, MD; G. William plantation in patients with AL amyloidosis: an
Dec, MD; and Leonard S. Lilly, MD. 8-year study. Ann Intern Med 2004;140:85–93.
Yazdani K, Maraj S, Amanullah AM. Differentiating
constrictive pericarditis from restrictive cardiomy-
Additional Reading opathy. Rev Cardiovasc Med 2005;6:61–71.

Burkett EL, Hershberger RE. Clinical and genetic issues


in familial dilated cardiomyopathy. J Am Coll Car-
diol 2005;45:969–981.
10090-10_CH10.qxd 8/31/06 4:53 PM Page a

Chapter 10—Author Queries

1. AU: Chapter 9 only refers to this as a system, not an axis. Are both OK, or should termi-
nology be consistent?
2. AU: As in ch. 9. OK?
3. AU: There is a new edition of this book. Should figure/citation be updated?
10090-11_CH11.qxd 8/31/06 4:54 PM Page 269

C H A P T E R

Mechanisms of
Cardiac Arrhythmias
Hillary K. Rolls
11
William G. Stevenson
Gary R. Strichartz
Leonard S. Lilly

NORMAL IMPULSE FORMATION Abnormal Automaticity


Ionic Basis of Automaticity Triggered Activity
Native and Latent Pacemakers ALTERED IMPULSE CONDUCTION
Overdrive Suppression Conduction Block
Electrotonic Interactions Unidirectional Block and Reentry
ALTERED IMPULSE FORMATION APPROACHES TO ANTIARRHYTHMIC
Alterations in Sinus Node Automaticity TREATMENT
Escape Rhythms Bradyarrhythmias
Enhanced Automaticity of Latent Pacemakers Tachyarrhythmias

Normal cardiac function relies on the flow the atrium or AV node and designated
of electrical impulses through the heart in ventricular when they originate from the
an exquisitely coordinated fashion. Abnor- His-Purkinje system or ventricles. This chap-
malities of the electrical rhythm are known ter describes the mechanisms by which
as arrhythmias (also termed dysrhythmias) such arrhythmias develop, followed by a
and are among the most common clinical general description of their management.
problems encountered. The presentations of Chapter 12 summarizes specific rhythm
arrhythmias range from benign palpitations disorders and how to recognize and treat
to severe symptoms of low cardiac output them.
and death; therefore, a thorough under- Disorders of heart rhythm result from al-
standing of these disorders is important to terations of impulse formation, impulse
the daily practice of medicine. conduction, or both. This chapter first ad-
Abnormally slow rhythms are termed bra- dresses how alterations of impulse forma-
dycardias (or bradyarrhythmias). Fast rhythms tion and conduction occur and under what
are known as tachycardias (or tachyarrhyth- circumstances they cause arrhythmias. Fig- Fig. 1
mias). Tachycardias are further character- ure 11.1 provides an organizational schema
ized as supraventricular when they involve for this discussion.

269
10090-11_CH11.qxd 8/31/06 4:54 PM Page 270

270 Chapter Eleven

TACHYARRHYTHMIAS (Increased firing rate)

Automaticity Automaticity Abnormal Triggered Unidirectional block


of SA node of latent automaticity activity and reentry
pacemakers

Enhanced
automaticity

ALTERED ALTERED
IMPULSE IMPULSE
FORMATION CONDUCTION

Automaticity Conduction block


of SA node

BRADYARRHYTHMIAS (Decreased firing rate)

Figure 11.1. Arrhythmias result from alterations in impulse formation and/or impulse con-
duction. Tachyarrhythmias result from enhanced automaticity, unidirectional block with reentry, or trig-
gered activity. Bradyarrhythmias result from decreased automaticity or conduction block. SA, sinoatrial.

NORMAL IMPULSE FORMATION spontaneous diastolic depolarization reaches


the threshold voltage, an action potential
As presented in Chapter 1, electrical impulse is generated. An important ionic current
formation in the heart arises from the intrin- largely responsible for phase 4 spontaneous
sic automaticity of specialized cardiac cells. depolarization is known as the pacemaker
Automaticity refers to a cell’s ability to current (If). This current is activated by hy-
depolarize itself to a threshold voltage in a perpolarization (increasingly negative volt-
rhythmic, repeated fashion, such that spon- ages) and is carried mainly by sodium ions.
taneous action potentials are generated. Al- The channels that carry If open when the
though atrial and ventricular myocytes do membrane voltage becomes more negative
not have this property under normal condi- than approximately −50 mV and are differ-
tions, the cells of the specialized conducting ent from the fast sodium channels responsi-
system do possess natural automaticity and ble for rapid phase 0 depolarization in non-
are therefore termed pacemaker cells. The pacemaker cells. The inward flow of Na+
specialized conducting system includes the through these slow channels, driven by its
sinoatrial (SA) node, the atrioventricular (AV) concentration gradient and the negative in-
nodal region, and the ventricular conducting tracellular charge, forces the membrane po-
system. The latter is composed of the bundle tential to depolarize toward the threshold
of His, the bundle branches, and the Purkinje voltage.
fibers. In pathologic situations, myocardial cells In the pacemaker cells of the sinoatrial
outside the conducting system may also ac- node, alterations in two other ionic currents
quire the property of automaticity. also contribute to phase 4 depolarization:
(1) a slow inward calcium current, the chan-
nels of which become activated at voltages
Ionic Basis of Automaticity
reached near the end of phase 4, and (2) a
Cells with natural automaticity do not have progressive decline of an outward potassium
a static resting potential. Rather, they dis- current. Activation of the latter current is re-
play a gradual depolarization during phase 4 sponsible for cellular repolarization during
Fig. 2 of the action potential (Fig. 11.2) If this phase 3 of the action potential, and it pro-
10090-11_CH11.qxd 8/31/06 4:54 PM Page 271

Mechanisms of Cardiac Arrhythmias 271

If
influx

Figure 11.2. The action potential (AP) of a pacemaker cell. Notice


the slow phase 4 depolarization, largely caused by the If (pacemaker) cur-
rent through slow Na+ channels, which drives the cell to threshold po-
tential (approximately −40 mV). The upstroke of the AP is caused by the
slow inward current of Ca+ ions. Inactivation of the calcium channels and
K+ efflux through potassium channels are responsible for repolarization.
MDP, maximum negative diastolic potential; TP, threshold potential.

gressively diminishes during phase 4. The ization phase of pacemaker cells depends on
combination of the inward If, inward, Ca++ inactivation of the calcium channels and the
and reduced outward K+ currents acts to opening of voltage-gated potassium channels
gradually depolarize the SA nodal cells to that permit efflux of potassium from the cells.
the threshold potential.
When the membrane potential of the
Native and Latent Pacemakers
pacemaker cell reaches the threshold value,
the upstroke of the action potential is gen- The different populations of automatic cells
erated. In contrast to the phase 0 upstroke of in the specialized conduction pathway have
cells in the Purkinje system, that of cells in different intrinsic rates of firing. These rates
the sinus and AV nodes is much slower (see are determined by three variables that in-
Fig. 11.2; compare with Fig. 1.14). The rea- fluence how fast the membrane potential
son for the difference is that the membrane reaches threshold: (1) the rate (i.e., the slope)
potential determines the proportion of fast of phase 4 spontaneous depolarization, (2)
sodium channels that are in a resting state the maximum negative diastolic potential,
capable of depolarization, compared with and (3) the threshold potential. A more neg-
an inactivated state. The number of avail- ative maximum diastolic potential, or a less
able (or resting-state) fast sodium channels negative threshold potential, slows the rate
increases as the resting membrane potential of impulse initiation because it takes longer
becomes more negative. Because sinus and to reach that threshold value (Fig. 11.3). Con- Fig. 3
AV nodal cells have less negative maximum versely, the greater the If, the steeper the
diastolic membrane voltages (−50 to −60 mV) slope of phase 4 and the faster the cell de-
than do Purkinje cells (−90 mV), a greater polarizes. The rate of If depends on the num-
proportion of the fast sodium channels is in- ber and kinetics of the individual pacemaker
activated in these pacemaker cells. Thus, the channels through which this current flows.
action potential upstroke relies to a greater ex- Because all the healthy myocardial cells
tent on calcium ion inflow (through the rela- are electrically connected by gap junctions,
tively slower opening Ca++ channels) and its an action potential generated in one part of
slope is less steep in these cells. The repolar- the myocardium will ultimately spread to all
10090-11_CH11.qxd 8/31/06 4:54 PM Page 272

272 Chapter Eleven

a b c

a b

Figure 11.3. Determinants of cell-firing rates. A. Alterations in the pace-


maker current (If) and in the magnitude of the maximum diastolic potential
(MDP) alter the cell-firing rate. (a) The normal action potential (AP) of a pace-
maker cell. (b) Reduced If renders the slope of phase 4 less steep; thus, the
time required to reach threshold potential (TP) is increased. (c) The MDP is
more negative; therefore, the time required to reach TP is increased. B. Alter-
ations in TP change the firing rate of the cell. Compared with the normal TP
(a), the TP in b is less negative; thus, the duration of time to achieve thresh-
old is increased, and the firing rate decreases.

other regions. When an impulse arrives at a Other cells within the specialized conduction
cell that is not yet close to threshold, current system harbor the potential to act as pace-
from the depolarized cell will bring the makers if necessary and are therefore called
adjacent cell membrane potential to the latent pacemakers (or ectopic pacemak-
threshold level so that it will fire (regardless ers). In contrast to the SA node, the AV node
of how close its intrinsic If has brought it to and the bundle of His have intrinsic firing
threshold). Thus, the pacemaker cells with rates of 50 to 60 bpm, and cells of the Purk-
the fastest rate of depolarization set the inje system have rates of approximately 30 to
heart rate. In the normal heart, the domi- 40 bpm. These latent sites may initiate im-
nant pacemaker is the sinoatrial node, which pulses and take over the pacemaking func-
at rest initiates impulses at a rate of 60 to tion if the SA node slows or fails to fire, or if
100 bpm. Because the sinus node rate is conduction abnormalities block the normal
faster than that of the other tissues that pos- wave of depolarization from reaching them.
sess automaticity, its repeated discharges
prevent spontaneous firing of other poten-
Overdrive Suppression
tial pacemaker sites.
The SA node is known as the native pace- Not only does the cell population with the
maker because it normally sets the heart rate. fastest intrinsic rhythm preempt all other
10090-11_CH11.qxd 8/31/06 4:54 PM Page 273

Mechanisms of Cardiac Arrhythmias 273

automatic cells from spontaneously firing, it rent If, and further decreases the rate of spon-
also directly suppresses their automaticity. taneous depolarization. Thus, overdrive sup-
This phenomenon is called overdrive sup- pression decreases a cell’s automaticity when
pression. Cells maintain their transsarco- that cell is driven to depolarize faster than its
lemmal ion distributions because of the con- intrinsic discharge rate.
tinuously active Na+K+-ATPase pump that
extrudes three Na+ ions from the cell in ex- Electrotonic Interactions
change for two K+ ions transported in (Fig.
Fig. 4 11.4). Because its net transport effect is one In addition to overdrive suppression, anato-
positive charge in the outward direction, the mic connections between pacemaker and non-
Na+K+-ATPase pump creates a hyperpolarizing pacemaker cells are important in determining
current (i.e., it tends to make the inside of the how adjacent cells suppress latent pacemaker
foci. Myocardial cells that are not part of the
cell more negative). As the cell potential be-
specialized conducting system repolarize to a
comes increasingly negative, additional time
resting potential of −90 mV, whereas pace-
is required for spontaneous phase 4 depolar-
maker cells repolarize to a maximum dias-
ization to reach the threshold voltage (see Fig.
tolic potential of about −60 mV. When these
11.3A), and therefore the rate of spontaneous
two cell types are adjacent to one another,
firing is decreased. Although the hyperpolar- they are electrically coupled through low resis-
izing current antagonizes If, pacemaker cells tance gap junctions concentrated in their in-
firing at their own intrinsic rate have an If cur- tercalated discs. This coupling results in an
rent sufficiently large to overcome this hyper- equilibration of electrical potentials owing to
polarizing influence (see Fig. 11.4). electrotonic current flow between the cells,
The hyperpolarizing current increases causing relative hyperpolarization of the pace-
when a cell is forced to fire faster than its in- maker cell and relative depolarization of the
trinsic pacemaker rate. The more frequently nonpacemaker cell (Fig. 11.5). The hyperpo- Fig. 5
the cell is depolarized, the greater the quan- larizing current in the coupled pacemaker
tity of Na+ ions that enter the cell per unit cell competes with If and causes the slope of
time. As a result of the increased intracellular phase 4 diastolic depolarization to be less
AQ1 Na+, the Na+K+-ATPase pump becomes more steep, thereby reducing the cell’s automatic-
active, thereby tending to restore the normal ity. Electrotonic effects may be particularly
transmembrane Na+ gradient. This increased important in suppressing automaticity in the
pump activity provides a larger hyperpolar- AV node (via connections between atrial
izing current, opposing the depolarizing cur- myocytes and AV nodal cells) and in the dis-
tal Purkinje fibers (which are coupled to
nonautomatic ventricular myocardial cells).
In contrast, cells in the center of the SA node
are less tightly coupled to atrial myocytes;
thus, their automaticity is less subject to elec-
trotonic interactions.
Decoupling of normally suppressed cells,
such as those in the AV node (e.g., by ische-
mic damage), may reduce the inhibitory
electrotonic influence and enhance automa-
Figure 11.4. Competition between the de- ticity, producing ectopic rhythms by the la-
polarizing pacemaker current (If) and the tent pacemaker tissue.
AQ3 Na+K+-ATPase pump, which produces a hy-
perpolarizing current. The Na+K+-ATPase
pump transports three positive charges outside ALTERED IMPULSE FORMATION
the cell for every two it pumps in. The hyperpo-
larizing current acts to suppress automaticity by Arrhythmias may arise from altered impulse
antagonizing If and contributes to overdrive sup-
pression in cells that are stimulated more rapidly formation at the SA node or from other
than their intrinsic firing rate. sites, including the specialized conduction
10090-11_CH11.qxd 8/31/06 4:54 PM Page 274

274 Chapter Eleven

Figure 11.5. Electrotonic interaction between pacemaker (e.g., AV nodal) and nonpacemaker (myocardial)
cells. A. When pacemaker cells are not coupled to myocardial cells (as in the SA node), they have a maximum neg-
ative potential (MDP) of approximately −60 mV, whereas myocardial cells have a resting potential (RP) of approxi-
mately −90 mV. B. When pacemaker cells and myocytes are neighbors, they may be connected electrically by gap
junctions in their intercalated discs (e.g., in the AV node). In this situation, electrical current flows between the pace-
maker cell and the myocardial cell, tending to hyperpolarize the former and depolarize the latter, driving their mem-
brane potentials closer to one another. The more negative potential of atrial cells opposes If of the pacemaker cell,
such that the slope of phase 4 depolarization is less steep and therefore cellular automaticity is suppressed. If a dis-
ease state reduces coupling between cells, the influence of surrounding myocytes on the pacemaker cell is reduced,
allowing If to depolarize to threshold more readily and accelerating the rate of automaticity. TP, threshold potential.

pathways or regions of cardiac muscle. The acting through β1-adrenergic receptors, in-
main abnormalities of impulse initiation creases the probability of the pacemaker
that lead to arrhythmias are (1) altered au- channels being open (Fig. 11.6), through
tomaticity (of the sinus node or latent pace- which If can flow. The increase in If leads to
makers within the specialized conduction a steeper slope of phase 4 depolarization,
pathway), (2) abnormal automaticity in causing the SA node to reach threshold and
atrial or ventricular myocytes, and (3) trig- fire earlier than normal and the heart rate to
gered activity. increase.
In addition, sympathetic stimulation shifts
the action potential threshold to more-
Alterations in Sinus
negative voltages by increasing the proba-
Node Automaticity
bility that voltage-sensitive Ca++ channels
The rate of impulse initiation by the sinus are open (recall that calcium carries the cur-
node, as well as by the latent pacemakers of rent of phase 0 depolarization in pacemaker
the specialized conducting system, is regu- cells). Therefore, diastolic depolarization
lated primarily by neurohumoral factors. reaches the threshold potential earlier. Sym-
pathetic activity thus increases sinus node
automaticity both by causing the action po-
Increased Sinus Node Automaticity
tential threshold to become more negative
The most important modulator of normal and by increasing the rate of pacemaker de-
sinus node automaticity is the autonomic polarization via If. Examples of this normal
nervous system. Sympathetic stimulation, physiologic effect occur during exercise or
10090-11_CH11.qxd 8/31/06 4:54 PM Page 275

Mechanisms of Cardiac Arrhythmias 275

thus, the action potential threshold increases


to a more positive potential. Furthermore,
cholinergic stimulation increases the proba-
bility of the acetylcholine-sensitive K+ chan-
nels being open at rest. Positively charged K+
ions exit through these channels, producing
an outward current that drives the diastolic
potential to become more negative. The
overall effect of reduced If, a more negative
maximum diastolic potential, and a less
negative threshold level is a slowing of the
intrinsic firing rate and therefore a reduced
heart rate.
It follows that the use of pharmacologic
agents that modify the effects of the auto-
nomic nervous system will also affect the
Figure 11.6. The channels through which the
pacemaker current (If) flows are voltage
firing rate of the SA node. For example,
gated, opening at more negative membrane β-blocking drugs antagonize the β-adrener-
potentials. At any given voltage, there exists a gic sympathetic effect; therefore, they de-
probability between 0 and 1 that a specific chan-
nel will be open. Compared with normal baseline
crease the rate of phase 4 depolarization of
behavior (curve A), sympathetic stimulation (curve the SA node and slow the heart rate. Con-
B) or treatment with anticholinergic drugs shifts versely, atropine, an anticholinergic (anti-
this probability to a higher value for any given level
of membrane voltage, thus increasing the number
muscarinic) drug, has the opposite effect: by
of open channels and the rate at which the cell will blocking parasympathetic activity, the rate
fire. Curve C shows that parasympathetic stimula- of phase 4 depolarization increases and the
tion (or treatment with β-blockers) has the opposite
effect, decreasing the probability of a channel being
heart rate accelerates.
open, and therefore inhibiting depolarization.

Escape Rhythms
emotional stress, when sympathetic stimula- If the sinus node becomes suppressed and
tion appropriately increases the heart rate. fires less frequently than normal, the site of
impulse formation often shifts to a latent
pacemaker within the specialized conduc-
Decreased Sinus Node Automaticity
tion pathway. An impulse initiated by a la-
Normal decreases in SA node automaticity tent pacemaker because the SA node rate has
are mediated by reduced sympathetic stim- slowed is called an escape beat. Persistent
ulation and by increased activity of the para- impairment of the SA node will allow a con-
sympathetic nervous system. Whereas the tinued series of escape beats, termed an es-
sympathetic nervous system exerts a domi- cape rhythm. Escape rhythms are protec-
nant effect on the heart rate during times of tive in that they prevent the heart rate from
stress, the parasympathetic nervous system becoming too slow when SA node firing is
is the major mediator of the heart rate at impaired.
rest. As discussed in the previous section, sup-
Cholinergic (i.e., parasympathetic) stim- pression of sinus node activity may occur be-
ulation via the vagus nerve acts at the SA cause of increased parasympathetic tone.
node to reduce the probability of pacemaker Different regions of the heart have different
channels being open (see Fig. 11.6). Thus, If sensitivities to parasympathetic (vagal) stim-
and the slope of phase 4 depolarization are ulation. The SA node and the AV node are
reduced, and the intrinsic firing rate of the most sensitive to such an influence, followed
cell is slowed. In addition, the probability of by atrial tissue. The ventricular conducting
the Ca++ channels being open is decreased; system is the least sensitive. Therefore, mod-
10090-11_CH11.qxd 8/31/06 4:54 PM Page 276

276 Chapter Eleven

erate parasympathetic stimulation slows Because these myocardial cells have few
the sinus rate and allows the pacemaker to or no activated pacemaker channels, they
shift to another atrial site. However, very do not normally carry If. How injury allows
strong parasympathetic stimulation sup- such cells to spontaneously depolarize has
presses excitability at both the SA node and not been fully elucidated. However, when
atrial tissue, can cause conduction block at myocytes become injured, their membranes
the AV node, and may therefore result in become “leaky.” As such, they are unable to
the emergence of a ventricular escape pace- maintain the concentration gradients of
maker. ions, and the resting potential becomes less
negative (i.e., the cell partially depolarizes).
When a cell’s membrane potential is reduced
Enhanced Automaticity of
to a value less negative than −60 mV, grad-
Latent Pacemakers ual phase 4 depolarization can be demon-
Another means by which a latent pace- strated even among nonpacemaker cells.
maker can assume control of impulse for- This slow spontaneous depolarization is
mation is if it develops an intrinsic rate of probably related to a slow calcium current
depolarization faster than that of the sinus and by closure of a subset of K+ channels
node. Termed an ectopic beat, the impulse that normally help repolarize the cell.
is premature relative to the normal rhythm,
whereas an escape beat is late and terminates
Triggered Activity
a pause caused by a slowed sinus rhythm. A
sequence of similar ectopic beats is called an Under certain conditions, an action poten-
ectopic rhythm. tial can “trigger” abnormal depolarizations
Ectopic beats may arise in several cir- that result in extra heart beats or rapid ar-
cumstances. For example, high catechola- rhythmias. This process may occur when
mine concentrations can enhance the auto- the first action potential leads to oscillations
maticity of latent pacemakers, and if the of the membrane voltage known as afterde-
resulting rate of depolarization exceeds that polarizations. Unlike the spontaneous activity
of the sinus node, then an ectopic rhythm seen when enhanced automaticity occurs,
will develop. Ectopic beats are also com- this type of automaticity is stimulated by a
monly induced by hypoxemia, ischemia, preceding action potential. As illustrated in
electrolyte disturbances, and certain drug Figures 11.7 and 11.8, there are two types Fig. 7-8
toxicities (such as digitalis, as described in of afterdepolarizations depending on their
Chapter 17). timing after the inciting action potential:
early afterdepolarizations occur during the
repolarization phase of the inciting beat,
Abnormal Automaticity
whereas delayed afterdepolarizations occur
Cardiac tissue injury may lead to pathologic shortly after repolarization has been com-
changes in impulse formation whereby myo- pleted. In either case, abnormal action po-
cardial cells outside the specialized con- tentials are triggered if the afterdepolariza-
duction system acquire automaticity and tion reaches a threshold voltage.
spontaneously depolarize. Although such Early afterdepolarizations are changes
activity may appear similar to impulses orig- of the membrane potential in the positive di-
inating from latent pacemakers within the rection that interrupt normal repolarization
specialized conduction pathways, these ec- (see Fig 11.7). They can occur either during
topic beats arise from cells that do not usu- the plateau of the action potential (phase 2)
ally possess automaticity. If the rate of de- or during rapid repolarization (phase 3).
polarization of such cells exceeds that of the Early afterdepolarizations are more likely to
sinus node, they transiently take over the develop in conditions that prolong the ac-
pacemaker function and become the source tion potential duration (and therefore the
of an abnormal ectopic rhythm. electrocardiographic QT interval), as may
10090-11_CH11.qxd 8/31/06 4:54 PM Page 277

Mechanisms of Cardiac Arrhythmias 277

Figure 11.7. Triggered activity. An early afterdepolarization


(arrow) occurs before the action potential (AP) has fully repolarized.
Repetitive afterdepolarizations (dashed curve) may produce a rapid
sequence of triggered action potentials and hence a tachyarrhythmia.

occur during therapy with certain drugs (see be the initiating mechanism of the poly-
Chapter 17) and in the inherited long-QT morphic ventricular tachycardia known as
syndromes (see Chapter 12). torsades de pointes, which is described in
The ionic current responsible for an early Chapter 12.
afterdepolarization depends on the mem- Delayed afterdepolarizations may ap-
brane voltage at which the triggered event pear shortly after repolarization is complete
occurs. If the early afterdepolarization oc- (see Fig. 11.8). They most commonly de-
curs during phase 2 of the action potential, velop in states of high intracellular calcium, as
when most of the Na+ channels are in an in- may be present with digitalis intoxication
activated state, the upstroke of the triggered (see Chapter 17), or during marked cate-
beat relies on an inward Ca++ current. If, cholamine stimulation. It is thought that in-
however, it occurs during phase 3 (when the tracellular Ca++ accumulation causes the ac-
membrane voltage is more negative), there tivation of chloride currents or of the
is partial recovery of the fast Na+ channels, Na+-Ca++ exchanger that results in brief in-
which are then available to contribute to the ward currents that generate the delayed
current. afterdepolarization.
An early afterdepolarization-triggered ac- As with early afterdepolarizations, if the
tion potential can be self-perpetuating and amplitude of the delayed afterdepolarization
lead to a series of depolarizations (see Fig. reaches a threshold voltage, an action poten-
11.7). Early afterdepolarizations appear to tial will be generated. Such action potentials

Figure 11.8. Triggered activity. A delayed afterdepolarization (arrow)


arises after the cell has fully repolarized. If the delayed afterdepolarization
reaches the threshold voltage, a propagated action potential (AP) is triggered
(dashed curve).
10090-11_CH11.qxd 8/31/06 4:54 PM Page 278

278 Chapter Eleven

can be self-perpetuating and lead to tachy- sults in emergence of escape beats or escape
arrhythmias. For example, atrial and ven- rhythms, as the more distal sites assume the
tricular tachycardias associated with digi- pacemaker function.
talis toxicity are thought to be the result of AV block is common and a major reason
delayed afterdepolarizations, as described in for implantation of a permanent pacemaker,
Chapter 17. as discussed in Chapter 12.

ALTERED IMPULSE CONDUCTION Unidirectional Block and Reentry


Alterations in impulse conduction also lead A common mechanism by which a combi-
to arrhythmias. Conduction blocks gener- nation of conduction block and altered
ally slow the heart rate (bradyarrhythmias); impulse conduction leads to tachyarrhyth-
however, under certain circumstances, the mias is termed reentry. During a reentrant
process of reentry (described later) can ensue rhythm, an electrical impulse circulates re-
and produce abnormal fast rhythms (tachy- peatedly around a reentry path, recurrently
arrhythmias). depolarizing a region of cardiac tissue.
During normal cardiac conduction, each
electrical impulse that originates in the SA
Conduction Block
node travels in an orderly, sequential fash-
A propagating impulse is blocked when it en- ion through the rest of the heart, ultimately
counters a region of the heart that is electri- depolarizing all the myocardial fibers. The
cally unexcitable. Conduction block can be refractory period of each cell prevents im-
either transient or permanent and may be mediate reexcitation from adjacent depolar-
unidirectional (i.e., conduction proceeds ized cells, so that the impulse stops when all
when the involved region is stimulated from of the heart muscle has been excited. How-
one direction, but not when stimulated from ever, conduction blocks that prevent rapid
the opposite direction) or bidirectional (con- depolarization of parts of the myocardium
duction is blocked in both directions). Vari- can create an environment conducive to
ous conditions may cause conduction block, continued impulse propagation and reen-
including ischemia, fibrosis, inflammation, try, as illustrated in Figure 11.9. Fig. 9
and certain drugs. When conduction block The figure depicts electrical activity as it
occurs because a propagating impulse en- flows through a branch point anywhere with-
counters cardiac cells that are still refractory in the conduction pathways. Panel A shows
(from a previous depolarization), the block is propagation of a normal action potential. At
said to be functional. A propagating impulse point x, the impulse reaches two parallel
that arrives later may be able to be conducted. pathways (α and β) and travels down each
For example, antiarrhythmic drugs that into the more distal conduction tissue. In the
prolong action potential duration tend to normal heart, the α and β pathways have
produce functional conduction block. When similar conduction velocities and refractory
conduction block is caused by a barrier im- periods such that the wave fronts that pass
posed by fibrosis or scarring that replaces myo- through them collide in the distal conduc-
cytes, conduction block is fixed. tion tissue and extinguish each other.
Conduction block within the specialized Panel B shows what happens if conduc-
conducting system of the AV node or the tion is blocked in one limb of the pathway.
His-Purkinje system prevents normal propa- In this example, the action potential is ob-
gation of the cardiac impulse from the sinus structed when it encounters the β pathway
node to more distal sites. This atrioventricu- from above and therefore propagates only
lar block (AV block) removes the normal down the α tract into the distal tissue. As
overdrive suppression that keeps latent the impulse continues to spread, it encoun-
pacemakers in the His-Purkinje system in ters the distal end of the β pathway (at point
check. Thus, conduction block usually re- y). If the tissue in the distal β tract is also
10090-11_CH11.qxd 8/31/06 4:54 PM Page 279

Mechanisms of Cardiac Arrhythmias 279

Figure 11.9. Mechanism of reentry. A. Normal


conduction. When an action potential (AP)
reaches a branch in the conduction pathway
(point x), the impulse travels down both fibers (α
and β) to excite distal conduction tissue. B. Uni-
directional block. Forward passage of the im-
pulse is blocked in the β pathway but proceeds
normally down the α pathway. When the im-
pulse reaches point y, if retrograde conduction of
the β pathway is intact, the AP can enter β from
below and conduct in a retrograde fashion. C.
When point x is reached again, if the α pathway
has not had sufficient time to repolarize, then the
impulse stops. D. However, if conduction
through the retrograde pathway is sufficiently
slow (jagged line), it reaches point x after the α
pathway has recovered. In that circumstance, the
impulse is able to excite the α pathway again and
a reentrant loop is formed.

unable to conduct, the impulse simply con- of the necessary conditions for reentry has
tinues to propagate into the deeper tissues been met.
and reentry does not occur. However, if the When an action potential can conduct in
impulse at point y is able to propagate ret- a retrograde direction in a conduction path-
rogradely (backward) into pathway β, one way, whereas it had been prevented from
10090-11_CH11.qxd 8/31/06 4:54 PM Page 280

280 Chapter Eleven

doing so in the forward direction, unidirec- ever, with slower conduction velocities, a
tional block is said to be present. Unidirec- smaller reentry circuit is possible. Most
tional block tends to occur in regions where clinical cases of reentry occur within small
the refractory periods of adjacent cells are regions of tissue because the conduction
heterogeneous, such that some cells recover velocity within the reentrant loop is, in
before others. In addition, unidirectional fact, abnormally slow.
block may occur in states of cellular dys- In summary, the two critical conditions
function, and in regions where fibrosis has for reentry are (1) unidirectional block and
altered the myocardial structure. (2) slowed conduction through the reentry
As shown in panel C of Figure 11.9, if the path. These conditions commonly occur in
impulse is able to propagate retrogradely regions where fibrosis has developed, such
up the β pathway, it will again arrive at as infarction scars. In some cases, reentry oc-
point x. At that time, if the β pathway has curs over an anatomically fixed circuit or
not yet repolarized from the previous ac- path, such as AV reentry using an accessory
tion potential that had occurred moments pathway (as discussed in the following sec-
earlier, that limb is refractory to repeat tion). Reentry around distinct anatomic
stimulation and the returning impulse sim- pathways usually appears as a monomorphic
ply stops there. tachycardia on the electrocardiogram (ECG;
However, panel D illustrates what hap- i.e., in the case of ventricular tachycardia, all
pens if the velocity of retrograde conduc- the QRS complexes have the same appear-
tion in the diseased β path is not normal ance). This is because the reentry path is the
but slower than normal. In that case, suffi- same from beat to beat, producing a stable,
cient time may elapse for the α pathway regular tachycardia.
to repolarize before the returning impulse Other types of reentry do not require a
reaches point x from the β limb. Then stable, fixed path. For example, reentry can
the impulse is free to stimulate the α path- occur in electrophysiologically heteroge-
way once again, and the cycle repeats itself. neous myocardium, in which waves of re-
This circular stimulation can continue in- entrant excitation spiral through the tissue,
definitely, and each pass of the impulse continually changing direction. Ischemic
through the loop excites cells of the distal myocardium provides such a setting because
conduction tissue, which propagates to the the affected tissue is a mosaic of unexcitable
rest of the myocardium, resulting in a tachy- and partially excitable zones with reduced
arrhythmia. conduction velocities. When reentrant ven-
For the mechanism of reentry to occur, tricular tachycardia develops in an area of
the propagating impulse must continu- ischemia, the reentry circuit incessantly
ously encounter excitable tissue. Thus, the changes and the QRS complexes typically
time it takes for the impulse to travel vary from beat to beat, causing a polymor-
around the reentrant loop must be greater phic ventricular tachycardia pattern on the
than the time required for recovery (the re- ECG. As described in Chapter 12, fibrillation
fractory period) of the tissue, and this must of the atria or ventricles is likely caused by
be true for each point in the circuit. If the multiple circulating reentry wave fronts.
conduction time is shorter than the recov-
ery time, the impulse will encounter refrac-
tory tissue and stop. Because normal con-
Accessory Pathways and the
Wolff-Parkinson-White Syndrome
duction velocity is approximately 50 cm/sec
and the average effective refractory period The concept of reentry is dramatically illus-
is about 0.2 sec, a reentry path circuit tratedbytheWolff-Parkinson-White (WPW)
would need to be at least 10 cm long for syndrome. In the normal heart, the impulse
reentry to occur in a normal ventricle. How- generated by the SA node propagates through 1 LINE SHORT
10090-11_CH11.qxd 8/31/06 4:54 PM Page 281

Mechanisms of Cardiac Arrhythmias 281

atrial tissue to the AV node, where expected mal conduction over the AV node proceeds
slower conduction causes a short delay be- concurrently, ventricular depolarization
fore continuing on to the ventricles. How- represents a combination of the electrical
ever, approximately 1 in 1,000 people has impulse traveling via the accessory tract
the WPW syndrome and is born with an ad- and that conducted through the normal
ditional connection between the atrium and conduction pathway. As a result, the QRS
ventricle. Termed an accessory pathway or complex in patients with WPW is wider
bypass tract, this connection allows con- than normal and demonstrates an abnor-
duction between the atria and ventricles to mally slurred initial upstroke, known as a
bypass the AV node. The most common delta wave (Fig. 11.10).
type of accessory pathway consists of mi- During sinus rhythm, simultaneous con-
croscopic fibers that span the atrioventricu- duction through the accessory pathway and
lar groove somewhere along the mitral or AV node creates an interesting ECG appear-
tricuspid annuli (known as a bundle of Kent), ance but causes no symptoms. The presence
Fig. 10 as shown in Figure 11.10. of the abnormal pathway, however, creates
Because accessory pathway tissue con- an ideal condition for reentry because the
ducts impulses faster than the AV node, refractory period of the pathway is usually
stimulation of the ventricles during sinus different from that of the AV node. An ap-
rhythm begins earlier than normal, and the propriately timed abnormal impulse (e.g., a
PR interval of the ECG is therefore shortened premature beat) may encounter blockage in
(usually <0.12 sec, or <3 small boxes). In the accessory pathway but conduction over
this situation, the ventricles are said to be the AV node, or vice versa. If the propagat-
“preexcited.” However, the accessory path- ing impulse then finds that the initially
way connects to ventricular myocardium blocked pathway has recovered (unidirec-
rather than to the Purkinje system, such tional block), it can conduct in a retrograde
that the subsequent spread of the impulse direction up to the atrium and then down
through the ventricles from that site is the other pathway back to the ventricles.
slower than usual. In addition, because nor- Thus, a large anatomic loop is established,

Normal ECG

QRS
AV
node P T
SA
node

ECG with bypass tract

Bypass Widened
tract QRS

Delta
Shortened PR wave

Figure 11.10. Accessory pathway (also termed the bypass tract). Example of an atrioventricu-
lar bypass tract (bundle of Kent), shown schematically, which can conduct impulses from the atrium
directly to the ventricles, bypassing the AV node. The ECG demonstrates a short PR interval and a
“delta wave” caused by early excitation of the ventricles via the accessory pathway. ECG, electro-
1 LINE SHORT cardiogram; SA, sinoatrial.
10090-11_CH11.qxd 8/31/06 4:54 PM Page 282

282 Chapter Eleven

with the accessory pathway serving as one potension or cardiac arrest, it must be im-
limb and the normal conduction pathway mediately terminated to restore effective
through the AV node as the other. The clin- cardiac function. Therapy for termination
ical characteristics of the Wolff-Parkinson- may include electrical cardioversion (an
White syndrome, including the types of electrical “shock”) for tachycardias, cardiac
reentrant tachycardia associated with it, are pacing for bradycardias, or administration
described in Chapter 12. of medications.
Additional therapy to prevent recurrences
The mechanisms of altered impulse for- is guided by the etiology of the rhythm dis-
mation and conduction form the basis of all turbance. Correctable factors that contribute
common arrhythmias, both abnormally slow to abnormal impulse formation and con-
rhythms (bradyarrhythmias) and abnormally duction (such as ischemia or electrolyte ab-
fast ones (tachyarrhythmias). Table 11.1 normalities) should be corrected. If there is
lists the underlying mechanisms and exam- a risk of recurrent arrhythmia, medications
ples of their commonly associated rhythm that alter automaticity, conduction and/or
disturbances. refractoriness may be administered. Some-
times, catheter or surgical ablation of con-
duction pathways is undertaken to physi-
APPROACHES TO
cally disrupt the region responsible for the
ANTIARRHYTHMIC TREATMENT
arrhythmia. Other advanced options in-
The treatment of a rhythm disorder de- clude implantation of a permanent pace-
pends on its severity and likely mechanism. maker for serious bradyarrhythmias or an
When an arrhythmia produces severe hy- internal ICD to automatically terminate ma-

TABLE 11.1. Mechanisms of Arrhythmia Development

Abnormality Mechanism Examples

Bradyarrhythmias
Altered impulse formation
• Decreased automaticity Decreased phase 4 depolarization Sinus bradycardia
(e.g., parasympathetic stimulation)
Altered impulse conduction
• Conduction blocks Ischemic, anatomic, or drug- First-, second-, and third-degree AV
induced impaired conduction blocks
Tachyarrhythmias
Altered impulse formation
• Enhanced automaticity
Sinus node Increased phase 4 depolarization Sinus tachycardia
(e.g., sympathetic stimulation)
Ectopic focus Acquires phase 4 depolarization Ectopic atrial tachycardia
• Triggered activity
Early afterdepolarization Prolonged action potential duration Torsades de pointes
Delayed afterdepolarization Intracellular calcium overload APBs, VPBs, digitalis-induced
(e.g., digitalis toxicity) arrhythmias
Altered impulse conduction
• Reentry Unidirectional block plus slowed
conduction
Anatomical Atrial flutter, AV nodal reentrant
tachycardia
Functional Atrial fibrillation, ventricular
fibrillation

AV, atrioventricular; APB, atrial premature beat; VPB, ventricular premature beat.
10090-11_CH11.qxd 8/31/06 4:54 PM Page 283

Mechanisms of Cardiac Arrhythmias 283

lignant tachyarrhythmias should they recur. manent basis. Temporary units are used to
The following sections summarize the com- stabilize patients who are awaiting implan-
mon therapeutic modalities, and Chapter 12 tation of a permanent pacemaker or to treat
describes how they are used to address spe- transient bradyarrhythmias, such as those
cific rhythm disorders. caused by reversible drug toxicities.
There are two types of temporary pace-
makers. External transthoracic pacemakers
Bradyarrhythmias
deliver electrical pulses to the patient’s chest
Not all slow heart rhythms require specific through large adhesive electrodes placed on
treatment. For those that do, pharmacologic the skin. The advantage of this technique is
therapy can increase the heart rate acutely, that it can be applied rapidly. Unfortu-
but their effect is transient. Electronic pace- nately, because the current used must be
makers are used when a more sustained ac- sufficient to initiate a cardiac depolariza-
tion is needed. tion, it stimulates thoracic nerves and skele-
tal muscle, which can be quite uncomfort-
able. Therefore, this form of pacing is
Pharmacologic Therapy
usually used only on an emergency basis
Pharmacologic therapy of bradyarrhythmias until another means of treating the ar-
modifies the autonomic input to the heart rhythmia can be implemented.
in one of two ways: The other option for temporary pacing is
a transvenous unit. In this case, an electrode-
1. Anticholinergic drugs (i.e., antimuscarinic
tipped catheter is inserted percutaneously
agents such as atropine). Vagal stimula-
into the venous system, passed into the
tion reduces the rate of sinus node depo-
heart, and connected to an external power
larization (which slows the heart rate)
source (termed a pulse generator). Electrical
and decreases conduction through the
pulses are applied directly to the heart
AV node, through the release of acetyl-
through the electrode catheter, which is typ-
choline onto muscarinic receptors. Anti-
ically placed in the right ventricle or right
cholinergic drugs competitively bind to
atrium. This type of pacing is not painful
muscarinic receptors and thereby reduce
and can be effective for days. There is, how-
the vagal effect. This results in an in-
ever, a risk of infection and/or thrombosis
creased heart rate and enhanced AV nodal
that increases with time.
conduction.
Permanent pacemakers are more sophis-
2. β1-receptor agonists (e.g., isoproterenol).
ticated than the temporary variety. Various
Mimicking the effect of endogenous cat-
configurations can sense and capture the
echolamines, these drugs increase heart
electrical activity of the atria and/or ventri-
rate and speed AV nodal conduction.
cles. One or more wires (known as leads)
Atropine and isoproterenol are adminis- with pacing electrodes are passed through an
tered intravenously. Although these drugs axillary or subclavian vein into the right ven-
are useful in treating certain slow heart tricle or right atrium or through the coro-
rhythms emergently, it is not practical to nary sinus into a cardiac vein (to stimulate
continue them over the long term for per- the left ventricle). The pulse generator, simi-
sistent bradyarrhythmias. lar in size to two silver dollars stacked on top
of one another, is connected to the leads and
then implanted under the skin, typically in
Electronic Pacemakers
the infraclavicular region. The pacemaker
Electronic pacemakers apply repeated elec- battery typically lasts about 10 years.
trical stimulation to the heart to initiate de- Modern permanent pacemakers sense
polarizations at a desired rate, thereby as- cardiac activity and pace only when needed.
suming control of the rhythm. Pacemakers They incorporate complex functions to
may be installed on a temporary or a per- track the patient’s normal heart rate and can
10090-11_CH11.qxd 8/31/06 4:54 PM Page 284

284 Chapter Eleven

stimulate beats automatically in response to 3. Make the threshold potential less nega-
activity. They can also record useful data, tive
such as whether fast rates have been sensed
(that might indicate a tachyarrhythmia), the Desired Antiarrhythmic Effects to Interrupt
amount of pacing that has been required, Reentrant Circuits
and other parameters of pacemaker function.
1. Decrease conduction in the reentry cir-
An external radio frequency programming
cuit to the point that conduction fails,
device is used to “interrogate” the pacemaker thus stopping the reentry impulse
to obtain the recorded information and ad-
2. Increase the refractory period within the
just the pacing functions.
reentrant circuit so that a propagating
Although the most common indications
impulse finds tissue within the loop un-
for permanent pacemakers are bradyarrhyth-
excitable and the impulse stops
mias, pacemakers that incorporate a left ven-
3. Suppress premature beats that can initi-
tricular pacing lead are also used to improve
ate reentry
cardiac performance in some patients with
heart failure (see Chapter 9).
Desired Drug Effects to Eliminate
Triggered Activity
Tachyarrhythmias 1. Shorten the action potential duration (to
The treatment of tachyarrhythmias is di- prevent early afterdepolarizations)
rected at (1) protection of the patient from 2. Correct conditions of calcium overload
the consequences of the arrhythmia and (to prevent delayed afterdepolarizations)
(2) the specific mechanism responsible for
Drugs used to achieve the goals modulate
the abnormal rhythm. Pharmacologic agents
the action potential through interactions
and cardioversion/defibrillation are com- with ion channels, surface receptors, and
monly used approaches, but innovative transport pumps. Many drugs have multiple
electrical devices and transvenous catheter- effects and may attack arrhythmias through
based techniques have revolutionized treat- more than one mechanism. The commonly
ment of these disorders. used antiarrhythmic drugs and their actions
are described in Chapter 17.
Pharmacologic Therapy It is extremely important to recognize
that in addition to suppressing arrhythmias,
Pharmacologic management of tachyarr- these drugs have the potential to aggravate
hythmias is directed against the underlying or provoke certain rhythm disturbances. This
mechanism (abnormal automaticity, reen- undesired consequence is referred to as pro-
trant circuits, or triggered activity). Many an- arrhythmia and is a major limitation of
tiarrhythmic drugs are available, the phar- contemporary antiarrhythmic drug therapy.
macology and actions of which are addressed For example, antiarrhythmic agents that act
in Chapter 17. The choice of drug relies on therapeutically to prolong the action poten-
the cause of the specific arrhythmia. From tial duration can, as an undesired effect,
consideration of the arrhythmia mechanisms cause early afterdepolarizations, the mecha-
presented in this chapter, the following strate- nism underlying the polymorphic ventricular
gies emerge. tachycardia known as torsades de pointes (see
Chapter 12). In addition, most agents used to
Desired Drug Effects to Eliminate Rhythms treat tachyarrhythmias have the potential to
Caused by Increased Automaticity aggravate bradyarrhythmias, and all antiar-
rhythmics have potentially toxic noncardiac
1. Reduce the slope of phase 4 spontaneous side effects. These shortcomings have led to
depolarization of the automatic cells an increased reliance on nonpharmacologic
2. Make the diastolic potential more nega- treatment options, as described in the follow-
tive (hyperpolarize) ing sections.
10090-11_CH11.qxd 8/31/06 4:54 PM Page 285

Mechanisms of Cardiac Arrhythmias 285

Vagotonic Maneuvers External defibrillation is performed to


terminate ventricular fibrillation, employing
Many tachycardias involve transmission of
the same equipment as that used for cardio-
impulses through the AV node, a structure
version. However, during fibrillation, there is
that is sensitive to vagal modulation. Vagal
no organized QRS complex on which to syn-
tone can be transiently increased by a num-
chronize the electrical discharge, so it is de-
ber of bedside maneuvers, and performing
livered using the “asynchronous” mode of
these may slow conduction, which termi-
the device.
nates some reentrant tachyarrhythmias. For
example, carotid sinus massage is performed
by rubbing firmly for a few seconds over the Implantable Cardioverter-Defibrillators
carotid sinus, located at the bifurcation of the
ICDs automatically terminate dangerous
internal and external carotid arteries on either
ventricular arrhythmias using internal car-
side of the neck. This maneuver stimulates
dioversion or defibrillation, or a technique
the baroreceptor reflex (see Chapter 13),
which elicits the desired increase in vagal known as antitachycardia pacing. These de-
tone and withdrawal of sympathetic tone. vices are implanted, in a manner similar to
This maneuver should be performed on only that of permanent pacemakers, in patients
one carotid sinus at a time (to prevent inter- at high risk of sudden cardiac death from
ference with brain perfusion) and is best ventricular arrhythmias. The device contin-
avoided in patients with known atherosclero- uously monitors cardiac activity, and if the
sis involving the carotid arteries. heart rate exceeds a certain programmable
threshold for a specified time (e.g., >12
beats), the ICD delivers an appropriate in-
Electrical Cardioversion tervention, such as an electrical shock. In-
and Defibrillation ternal cardioversion or defibrillation requires
Cardioversion and defibrillation involve the substantially less energy than external defib-
application of an electric shock to terminate rillation but is still uncomfortable if the pa-
a tachycardia. A shock with sufficient en- tient is conscious.
ergy depolarizes the bulk of excitable myo- In addition, many monomorphic ven-
cardial tissue, interrupts reentrant circuits, tricular tachycardias can be terminated by
establishes electrical homogeneity, and al- an ICD with a rapid burst of electrical im-
lows the sinus node (the site of fastest spon- pulses, termed antitachycardia pacing (ATP).
taneous discharge) to regain pacemaker con- The goal is to artificially pace the heart at a
trol. Tachyarrhythmias that are caused by rate faster than the tachycardia to prema-
reentry can usually be terminated by this turely depolarize a portion of a reentrant cir-
procedure, whereas arrhythmias owing to cuit, thereby rendering it refractory to fur-
abnormal automaticity may simply persist. ther immediate stimulation. Consequently,
External cardioversion is used to termi- when a reentrant impulse returns to the
nate supraventricular tachycardias or orga- zone that has already been depolarized by
nized ventricular tachycardias. It is per- the device, it encounters unexcitable tissue,
formed by briefly sedating the patient and it cannot propagate further, and the circuit
then placing two large electrode paddles (or is broken. An advantage of ATP is that, un-
adhesive electrodes) against the chest on like internal cardioversion, it is painless.
either side of the heart. The electrical dis- However, ATP is not effective for terminat-
charge is electronically synchronized to occur ing ventricular fibrillation.
at the time of a QRS complex (i.e., when
ventricular depolarization occurs). This pre-
Catheter Ablation
vents the possibility of discharge during the
relative refractory period of the ventricle If an arrhythmia originates from distinct
(see Fig. 1.16), which could induce ventric- anatomical reentry circuits or automatic foci,
ular fibrillation. electrophysiologic mapping techniques can
10090-11_CH11.qxd 8/31/06 4:54 PM Page 286

286 Chapter Eleven

be used to localize the region of myocardium 4. Bradyarrhythmias are usually treated with
or conduction tissue responsible for the dis- drugs that accelerate the rate of sinus
turbance. It is then often possible to ablate node discharge and enhance AV nodal
the site via a catheter that applies radio- conduction (atropine, isoproterenol) or
frequency current to heat and destroy the tis- electronic pacemakers.
sue. These procedures have revolutionized the 5. Pharmacologic therapy for tachyarrhyth-
management of patients with many types of mias is directed at the mechanism re-
supraventricular tachycardias, because they sponsible for the rhythm disturbance.
often offer a permanent therapeutic solution For refractory tachyarrhythmias, or in
that spares patients from undergoing chronic emergency situations, electrical cardio-
antiarrhythmic drug therapy. For patients version or defibrillation is used. Catheter-
AQ2 with recurrent ventricular tachycardias caus- based ablative techniques are useful for
ing defibrillator shocks, ablation is often ef- long-term control of certain tachy-
fective in reducing the frequency of episodes. arrhythmias. ICDs are lifesaving devices
implanted in patients at high risk of sud-
den cardiac death because of ventricular
SUMMARY tachyarrhythmias.
1. Arrhythmias result from disorders of im- Chapter 12 summarizes the diagnosis and
pulse formation, impulse conduction, or management of the most common arrhyth-
both. mias. Chapter 17 describes currently avail-
2. Bradyarrhythmias develop because of de- able antiarrhythmic drugs.
creased impulse formation (e.g., sinus bra-
dycardia) or decreased impulse conduc-
Acknowledgment
tion (e.g., AV nodal conduction blocks).
3. Tachyarrhythmias result from increased Contributors to the previous editions of this chapter
were Wendy Armstrong, MD; Nicholas Boulis, MD;
automaticity (of the SA node, latent pace- Jennifer E. Ho, MD; Mark S. Sabatine, MD; Elliott M.
makers, or abnormal myocardial sites), Antman, MD; Leonard I. Ganz, MD; Gary R. Strichartz,
triggered activity, or reentry. PhD; and Leonard S. Lilly, MD.
10090-11_CH11.qxd 8/31/06 4:54 PM Page a

Chapter 11—Author Queries


1. AU: Correct to add?
2. AU: Correct meaning of VT acronym?
3. AU: Correct to add? See also below.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 287

C H A P T E R

Clinical Aspects of
Cardiac Arrhythmias
Hillary K. Rolls
12
William G. Stevenson
Leonard S. Lilly

BRADYARRHYTHMIAS TACHYARRHYTHMIAS
Sinoatrial Node Supraventricular Arrhythmias
Escape Rhythms Ventricular Arrhythmias
Atrioventricular Conduction System

Arrhythmias appear frequently in patients BRADYARRHYTHMIAS


with cardiac disease and can occur in
The normal resting heart rate, resulting from
healthy people as well. Chapter 11 presented
repetitive depolarization of the sinus node,
the mechanisms by which abnormal heart
ranges from 60 to 100 bpm. Bradyarrhyth-
rhythms develop. This chapter describes how
mias are rhythms in which the heart rate is
Tab. 1 to recognize and treat them. Table 12.1 cate-
<60 bpm. They arise from disorders of im-
gorizes the common disorders considered in
pulse formation or impaired impulse con-
this chapter.
duction, as discussed in Chapter 11.
There are five basic questions to consider
when confronted with a patient with an ab-
normal heart rhythm, as detailed in the sec- Sinoatrial Node
tions that follow: Sinus Bradycardia

1. Definition: What is the arrhythmia? Sinus bradycardia (Fig. 12.1) is simply a slow- Fig. 1
ing of the normal heart rhythm, as a result of
2. Pathogenesis: What is the underlying
decreased firing of the sinoatrial (SA) node,
mechanism?
to a rate <60 bpm. Sinus bradycardia at rest
3. Precipitating factors: What conditions or during sleep is normal and a benign find-
provoke it? ing in many people. It is therefore incum-
4. Clinical presentation: What symptoms bent on the physician to decide whether this
and signs accompany the arrhythmia? rhythm is appropriate or pathologic in a par-
5. Treatment: What to do about it? ticular patient, and whether treatment is re-

287
10090-12_CH12.qxd 8/31/06 4:55 PM Page 288

288 Chapter Twelve

TABLE 12.1. Common Arrhythmias

Location Bradyarrhythmias Tachyarrhythmias

SA node Sinus bradycardia Sinus tachycardia


Sick sinus syndrome
Atria Atrial premature beats
Atrial flutter
Atrial fibrillation
Paroxysmal supraventricular tachycardias
Ectopic atrial tachycardia
Multifocal atrial tachycardia
AV node Conduction blocks Paroxysmal reentrant tachycardias (AV or AV nodal)
Junctional escape rhythm
Ventricles Ventricular escape rhythm Ventricular premature beats
Ventricular tachycardia
Torsades de pointes
Ventricular fibrillation

AV, atrioventricular; SA, sinoatrial.

quired. This decision can be made on the Mild sinus bradycardia is usually asymp-
basis of a patient’s age, underlying heart dis- tomatic and does not require treatment.
ease, level of physical activity, symptoms, However, a pronounced reduction of the
and whether the heart rate increases appro- heart rate can produce a fall in cardiac out-
priately with exercise. put with fatigue, light-headedness, confu-
Sinus bradycardia can result from either sion, or syncope. In such cases, any extrin-
intrinsic SA node disease or extrinsic factors sic provocative factors should be corrected,
that affect the node. Depressed intrinsic au- and specific therapy, as described in the next
tomaticity can be caused by aging or any dis- section, may be needed.
ease process that affects the atrium, including
ischemic heart disease or cardiomyopathy.
Sick Sinus Syndrome
Extrinsic factors that suppress SA nodal ac-
tivity include medications (e.g., many anti- Intrinsic SA node dysfunction that causes
arrhythmic drugs, including β-blockers and periods of inappropriate bradycardia is
certain calcium channel blockers) and meta- known as sick sinus syndrome (SSS). This
bolic causes (e.g., hypothyroidism). Highly condition often produces symptoms of dizzi-
trained athletes often have elevated vagal ness, confusion, or syncope. Patients with
tone, which results in physiologic (i.e., ap- this syndrome (or any cause of symptomatic
propriate) resting sinus bradycardia. How- sinus bradycardia) can be treated with intra-
ever, transient periods of high vagal tone venous anticholinergic drugs (e.g., atropine)
can also occur as a reflex response to pain or β-adrenergic agents (e.g., isoproterenol),
or fear resulting in inappropriate sinus brady- which transiently accelerate the heart rate. If
cardia. the problem is chronic and not corrected by

Figure 12.1. Sinus bradycardia. The P wave and QRS complexes are normal but the rate is <60 bpm.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 289

Clinical Aspects of Cardiac Arrhythmias 289

Figure 12.2. Bradycardia-tachycardia syndrome. A brief irregular tachycardia is followed by slow


sinus node discharge.

removal of aggravating factors, placement of ity but typically have slower firing rates than
a permanent pacemaker is required. the sinus node and are therefore suppressed
SSS is common in elderly patients, who during normal sinus rhythm. However, if SA
are also susceptible to supraventricular node activity becomes impaired or if there is
tachycardias, most commonly, atrial fibril- conduction block of the impulse from the SA
lation. This combination is known as the node, escape rhythms can emerge from the
Fig. 2 bradycardia-tachycardia syndrome (Fig. 12.2) more distal latent pacemakers (Fig. 12.3). Fig. 3
and is thought to result from atrial fibrosis Junctional escape beats arise from the AV
that impairs function of the SA node and node or proximal bundle of His. They are
predisposes to atrial fibrillation and flutter. characterized by a normal, narrow QRS com-
During the tachyarrhythmia, overdrive sup- plex, and when they occur in sequence
pression of the SA node occurs, and when (termed a junctional escape rhythm), ap-
the tachycardia terminates, profound peri- pear at a rate of 40 to 60 bpm. The QRS com-
ods of sinus bradycardia ensue. Treatment plexes are not preceded by normal P waves
generally requires the combination of an- because the impulse originates below the
tiarrhythmic drug therapy to suppress the atria. However, retrograde P waves may be ob-
tachyarrhythmias plus a permanent pace- served as an impulse propagates from the
maker to prevent bradycardia. more distal pacemaker backward to the at-
rium. Retrograde P waves typically follow the
QRS complex and are inverted (negative de-
Escape Rhythms
flection on the electrocardiogram [ECG]) in
Cells in the atrioventricular (AV) node and limb leads II, III, and aVF, indicating activa-
His-Purkinje system are capable of automatic- tion of the atria from the inferior direction.

Figure 12.3. Escape rhythms. No P waves are evident. A. Junctional escape rhythm with normal-width
QRS complexes. B. Wide QRS complexes typical of a ventricular escape rhythm.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 290

290 Chapter Twelve

Ventricular escape rhythms are charac- First-Degree AV Block


terized by even slower rates (30 to 40 bpm)
First-degree AV block, shown in Figure 12.4, Fig. 4
and widened QRS complexes. The complexes
indicates prolongation of the normal delay
are wide because the ventricles are not de-
between atrial and ventricular depolariza-
polarized by the normal rapid simultaneous
tion, such that the PR interval is lengthened
conduction over the right and left bundle
(>0.2 sec, which is >5 small boxes on the
branches but rather from a more distal point
ECG). However, the 1:1 relationship between
in the conduction system. The morphology
P waves and QRS complexes is preserved. The
that the QRS shows depends on the site of
impairment of conduction is usually within
origin of the escape rhythm. For example, the AV node itself and can be caused by a
an escape rhythm originating from the left transient influence or a structural defect. Re-
bundle branch will cause a right bundle versible causes include heightened vagal tone,
branch block QRS pattern, because the im- transient AV nodal ischemia, and drugs that
pulse depolarizes the left ventricle first and depress conduction through the AV node, in-
then spreads more slowly through the right cluding digitalis glycosides, β-blockers, cer-
ventricle. Conversely, an escape rhythm orig- tain calcium channel antagonists, and other
inating in the right bundle branch causes antiarrhythmic medications. Structural causes
the QRS to appear with a left bundle branch of first-degree AV block include myocardial
block configuration. Escape rhythms that infarction and chronic degenerative diseases
originate more distally, in the ventricular of the conduction system, which commonly
myocardium itself, are characterized by even occur with aging.
wider QRS complexes because such impulses Generally, first-degree AV block is a be-
are conducted outside the rapidly propagat- nign, asymptomatic condition that does not
ing Purkinje fibers. require treatment. However, it can indicate
Junctional and ventricular escape rhythms disease in the AV node associated with sus-
are protective backup mechanisms that ceptibility to higher degrees of AV block if
maintain a heart rate and cardiac output drugs are administered that further impair
when the sinus node or normal AV conduc- AV conduction, or if the conduction disease
tion fail. The clinical findings and treat- progresses.
ment of bradycardia associated with escape
rhythms are identical to those of SSS de-
Second-Degree AV Block
scribed earlier.
Second-degree AV block is characterized by
intermittent failure of AV conduction, result-
Atrioventricular
ing in some P waves not followed by a QRS
Conduction System
complex (i.e., intermittent failure of 1:1 atri-
The AV conduction system includes the AV oventricular conduction). There are two
node, bundle of His, and the left and right forms of second-degree AV block. In Möbitz
bundle branches. Impaired conduction be- type I block (also termed Wenckebach
tween the atria and ventricles can cause three block), shown in Figure 12.5, the degree of Fig. 5
degrees (types) of AV conduction block. AV delay gradually increases with each beat

P P P P P

Figure 12.4. First-degree AV block. The PR interval is prolonged.


10090-12_CH12.qxd 8/31/06 4:55 PM Page 291

Clinical Aspects of Cardiac Arrhythmias 291

P P P P P P

Figure 12.5. Second-degree AV block: Möbitz type I (Wenckebach). The P wave rate is constant,
but the PR interval progressively lengthens until a QRS is completely blocked (after fourth P wave).

until an impulse is completely blocked and QRS complexes), in which case it is known as
ventricular stimulation does not follow a P high-grade AV block (Fig. 12.7). Möbitz type Fig. 7
wave for a single beat. Therefore, the ECG II block is usually caused by conduction
shows a progressive increase in the PR inter- block beyond the AV node (in the bundle of
val from one beat to the next until a single His or more distally in the Purkinje system),
QRS complex is absent, after which the PR and the QRS pattern usually is widened in a
interval shortens to its initial length, and pattern of right or left bundle branch block.
the cycle starts anew. Möbitz type I block al- This type of block may arise from extensive
most always results from impaired conduc- myocardial infarction involving the septum
tion in the AV node (rather than more dis- or from chronic degeneration of the His-
tally in the conduction system). It is usually Purkinje system. It usually indicates severe
benign and may be seen in children, trained disease and is more dangerous than Möbitz
athletes, and people with high vagal tone, type I block. Möbitz type II may progress to
particularly during sleep. It may also occur third-degree block without warning; there-
during an acute myocardial infarction be- fore, treatment with a pacemaker is usually
cause of increased vagal tone or ischemia of warranted, even in asymptomatic patients.
the AV node, but the block is usually tran-
sient. Treatment is typically not necessary,
but in symptomatic cases, administration of Third-Degree AV Block
intravenous atropine or isoproterenol usu- Third-degree AV block, also termed com-
ally improves AV conduction transiently. plete heart block (Fig. 12.8), is present when Fig. 8
Placement of a permanent pacemaker is re- there is complete failure of conduction be-
quired for a symptomatic block that does tween the atria and ventricles. In adults, the
not resolve spontaneously or persists despite most common causes are acute myocardial
the correction of aggravating factors. infarction, drug toxicity (especially digitalis),
Möbitz type II second-degree AV block is and chronic degeneration of the conduction
characterized by the sudden intermittent loss pathways with age. Third-degree AV block
of AV conduction, without preceding gradual electrically disconnects the atria and ventri-
Fig. 6 lengthening of the PR interval (Fig. 12.6). cles; there is no relationship between the P
The block may persist for two or more beats waves and QRS complexes because the atria
(i.e., two sequential P waves not followed by depolarize in response to SA node activity,

P P P P P

Figure 12.6. Second-degree AV block: Möbitz type II. A QRS complex is blocked (after the fourth
P wave) without gradual lengthening of the preceding PR intervals.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 292

292 Chapter Twelve

P P P P P P

Figure 12.7. High-grade AV block. Sequential QRS complexes are blocked (after the second and
third P waves).

while a more distal escape rhythm drives the ferentiated by (1) the width of the QRS com-
ventricles independently. Thus, the P waves plex, (2) the morphology and rate of the
“march out” at a rate that is not related to the P waves, (3) the relationship between the
intervals at which QRS complexes appear. P waves and the QRS complexes, and (4) the
Depending on the site of the escape rhythm, response of the rhythm to vagal maneuvers
the QRS complexes may be of normal width such as carotid sinus massage (Fig. 12.9). Fig. 9
and occur at 40 to 60 bpm (originating from
the AV node) or may be widened and occur Supraventricular Arrhythmias
at slower rates (originating from the His-
Purkinje system). As a result of the slow rate, Sinus Tachycardia
patients may experience light-headedness or Sinus tachycardia is characterized by an SA
syncope. Permanent pacemaker therapy is al- node discharge rate >100 bpm (typically 100
most always necessary. to 180 bpm) with normal P waves and QRS
The term atrioventricular dissociation complexes (Fig. 12.10). This rhythm most Fig. 10
is a general description that refers to any sit- often results from increased sympathetic
uation in which the atria and ventricles beat and/or decreased vagal tone. Sinus tachycar-
independently, without any direct relation- dia is an appropriate physiologic response to
ship between P waves and QRS complexes. exercise. However, it may also result from
Third-degree AV block is one example of AV sympathetic stimulation in pathologic con-
dissociation. ditions, including fever, hypoxemia, hyper-
thyroidism, hypovolemia, and anemia. In
TACHYARRHYTHMIAS disease states, sinus tachycardia is usually a
sign of the severity of the primary patho-
Whenever the heart rate is >100 bpm for physiologic process and treatment is directed
3 beats or more, a tachyarrhythmia is said to at the underlying cause.
be present. Tachyarrhythmias result from
one of three mechanisms: enhanced auto-
Atrial Premature Beats
maticity, reentry, or triggered activity (see
Chapter 11). Tachyarrhythmias are catego- Atrial premature beats (APBs) are common in
rized into those that arise above the ventri- healthy as well as diseased hearts (Fig. 12.11). Fig. 11
cles (supraventricular) and those that arise They originate from automaticity or reentry
within the ventricles and can usually be dif- in an atrial focus outside the SA node and are

P P P P P P P P

Figure 12.8. Third-degree AV block. The P wave and QRS rhythms are independent of one another.
The QRS complexes are widened as they originate within the distal ventricular conduction system, not
at the bundle of His. The second and fourth P waves are superimposed on normal T waves. 1 LINE LONG
10090-12_CH12.qxd 8/31/06 4:55 PM Page 293

Clinical Aspects of Cardiac Arrhythmias 293

Sustained
tachycardia
Normal (narrow) QRS WiQRSde

Regular Irregularly No Constant


rhythm irregular relationship relationship
(constant rhythm between between
P-P intervals) QRS and QRS and
P waves P waves,
and
P is upright
No
in II, III,
distinct
aVF
P waves

Multifocal Atrial Ventricular Supraventricular


atrial fibrillation tachycardia tachycardias
tachycardia with “arantbe ”
uctiond

Si n su Reentra Ecatrilopc Atrial


ta c h ayc r adi SVT ta c h ayc r adi fluter
(A orV A dal)noV

At r lia r )(bpmeat 100-180 140-250 130-250 180-350

P w a v moe r olgyph No r lma V a r ia b le V a r ia b le “S a thedow- ”


pedsha

Ma bay r ytlup AV b ol c k AV b ol c k
dtiarocensRp At r lia r eat r e v e r ot m a reasciny ; m a y
si n esagmu m a lsy o w no r lma doesn ’tyusal aseincr
re v e rt

Figure 12.9. Differentiation of the common tachyarrhythmias.

often exacerbated by sympathetic stimula- duction through the atria). The QRS complex
tion. APBs are usually asymptomatic but may that follows the P wave is usually normal, re-
cause palpitations. On the ECG, an APB ap- sembling the QRS during sinus rhythm, be-
pears as an earlier-than-expected P wave with cause ventricular conduction is not impaired.
an abnormal shape (the impulse does not arise However, if the abnormal atrial focus fires
from the SA node, indicating abnormal con- very soon after the previous beat, the impulse

1 LINE LONG Figure 12.10. Sinus tachycardia. The P wave and QRS complexes are normal, but the rate is >100 bpm.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 294

294 Chapter Twelve

APB

Figure 12.11. Atrial premature beat (APB). The P wave occurs earlier than expected, and its shape
is abnormal.

may encounter an AV node that is refractory resulting in a slower ventricular rate, often
to excitation, indicating a blocked impulse an even fraction of the atrial rate. Thus, if
that does not conduct to the ventricles. The the atrial rate is 300 bpm and 2:1 block oc-
premature P wave is then not followed by a curs at the AV node (i.e., every other atrial
QRS complex and is termed a blocked APB. impulse finds the AV node refractory), the
Similarly, if the ectopic focus fires just a bit ventricular rate is 150 bpm. Because vagal
later in diastole, it may conduct through the maneuvers (e.g., carotid sinus massage) de-
AV node but encounter portions of the His- crease AV nodal conduction, they increase
Purkinje system that are still refractory. As a the degree of block, temporarily slowing the
result, the impulse is conducted through ventricular rate, which allows better visual-
those territories and to the ventricular my- ization of the underlying atrial activity. In
ocytes more slowly than normal, producing general, atrial flutter is caused by reentry
QRS complexes that are abnormally wide. over a large anatomically fixed circuit. In
APBs require treatment only if they are the common form of atrial flutter, this cir-
symptomatic. Because caffeine, alcohol, cuit is the atrial tissue along the tricuspid
and adrenergic stimulation (e.g., emotional valve annulus: the circulating depolariza-
stress) can all predispose to APBs, it is im- tion wave propagates up the interatrial sep-
portant to remove these factors when possi- tum, across the roof and down the free wall
ble. β-Blockers are the initial preferred phar- of the right atrium and finally along the
macologic treatment if needed. floor of the right atrium between the tricus-
pid valve annulus and inferior vena cava.
Because large parts of the atrium are depo-
Atrial Flutter
larized throughout the cycle, P waves often
Atrial flutter is characterized by rapid, regu- have a sinusoidal or “sawtooth” appearance.
lar atrial activity at a rate of 180 to 350 bpm Large flutter circuits can occur in other parts
Fig. 12 (Fig. 12.12). Many of these fast impulses of the right or left atrium as well, usually as-
reach the AV node during its refractory pe- sociated with areas of atrial scarring from
riod and do not conduct to the ventricles, disease, prior heart surgery, or ablation pro-

Figure 12.12. Atrial flutter is typified by rapid “saw-toothed” atrial activity (arrows).
10090-12_CH12.qxd 8/31/06 4:55 PM Page 295

Clinical Aspects of Cardiac Arrhythmias 295

cedures (e.g., those performed for treatment 3. Patients without an immediate need for
of atrial fibrillation). cardioversion can begin pharmacologic
Atrial flutter generally occurs in patients therapy. First, the ventricular rate is slowed
with preexisting heart disease. It may be by drugs that increase AV block: β-block-
paroxysmal and transient, persistent (lasting ers, calcium channel blockers (e.g., vera-
for days or weeks), or permanent. Symptoms pamil, diltiazem), or digoxin. Once the
of atrial flutter depend on the accompanying rate is effectively slowed, attempts are
ventricular rate. If the rate is <100 bpm, the made to restore sinus rhythm using an-
patient may be asymptomatic. Conversely, tiarrhythmic drugs that slow conduction
faster rates often cause palpitations, dyspnea, or prolong the refractory period of the
or weakness. Paradoxically, antiarrhythmic atrial myocardium (class IA, IC, or III
medications that reduce the rate of atrial flut- agents; see Chapter 17). Should these
ter by slowing conduction in the atrium may drugs fail to convert the rhythm, elective
paradoxically make the rhythm more dan- electrical cardioversion can then be un-
gerous by allowing the AV node more time to dertaken. Once sinus rhythm has been re-
recover between impulses. In this situation, stored, class IA, IC, or III antiarrhythmic
the AV node may begin to conduct in a 1:1 drugs may be administered chronically to
fashion, producing very rapid ventricular prevent recurrences.
rates. For example, a patient with atrial flut- 4. When chronic therapy is required to pre-
ter at a rate of 280 bpm and 2:1 conduction
vent recurrences, catheter ablation is
block at the AV node would have a ventricu-
often a better alternative than pharma-
lar rate of 140 bpm. If the atrial rate then
cologic approaches. In this method, an
slows to 220 bpm, the AV node may be able
electrode catheter is inserted into the
to recover sufficiently between depolariza-
femoral vein, passed via the inferior vena
tions to conduct every atrial impulse (i.e., 1:1
cava to the right atrium, and used to lo-
conduction), causing the ventricular rate to
calize and cauterize (ablate) part of the
accelerate to 220 bpm. In patients with lim-
reentrant loop to permanently interrupt
ited cardiac reserve, this acceleration may
the flutter circuit.
result in a profound reduction of cardiac out-
put and hypotension. Atrial flutter is associ-
ated with a risk of atrial thromboembolism, Atrial Fibrillation
as discussed later in the chapter.
Atrial fibrillation (AF) is a chaotic rhythm
Several approaches to the treatment of
with an atrial rate so fast (350 to 600 bpm) AQ1
atrial flutter are available:
that discrete P waves are not discernible on
1. For symptomatic patients with recent- the ECG (Fig. 12.13). As with atrial flutter, Fig. 13
onset atrial flutter, the most expeditious many of the atrial impulses encounter re-
therapy is electrical cardioversion to re- fractory tissue at the AV node, causing only
store sinus rhythm. This technique is also some of the depolarizations to be conducted
used to revert chronic atrial flutter that has to the ventricles in a very irregular fashion
not responded to other approaches. (indicated by the characteristic “irregularly
2. Flutter can be terminated by rapid atrial irregular” rhythm). The average ventricular
stimulation (burst pacing) using a tem- rate in untreated AF is approximately 140 to
porary or permanent pacemaker (see 160 bpm. Because discrete P waves are not
Chapter 11). This procedure can be used visible on the ECG, the baseline shows low-
when temporary atrial pacing wires are amplitude undulations punctuated by QRS
already present, as in the period after car- complexes and T waves.
diac surgery. In addition, certain types of The mechanism of AF probably involves
permanent pacemakers and implanted multiple “wandering” reentrant circuits
defibrillators can be programmed to per- within the atria, and in some patients, the
form burst pacing automatically when rhythm repetitively shifts between fibrilla-
atrial flutter occurs. tion and atrial flutter. When fibrillation is
10090-12_CH12.qxd 8/31/06 4:55 PM Page 296

296 Chapter Twelve

Figure 12.13. Atrial fibrillation is characterized by chaotic atrial activity without organized P waves and
by irregularity of the ventricular (QRS) rate.

paroxysmal (i.e., sudden, unpredictable epi- tricular rate. Digitalis is less effective for this
sodes), it is often initiated by rapid firing of purpose, although it may be useful in pa-
foci in paths of atrial muscle that extend tients with accompanying impairment of
along the pulmonary veins. For multiple ventricular contractile function. For those
reentry loops to be sustained in atrial fibril- who remain symptomatic despite adequate
lation, a minimum number of reentrant cir- rate control, conversion to sinus rhythm is
cuits is needed, and an enlarged atrium in- usually attempted. AF that persists for more
creases the potential for this to occur. Thus, than 48 hours may indicate that intraatrial
AF is often associated with right or left atrial thrombus has formed, and systemic antico-
enlargement. Accordingly, diseases that in- agulation (for at least 3 weeks) is usually
crease atrial pressure and size promote atrial warranted prior to cardioversion to reduce
fibrillation, including heart failure, hyper- the risk of thromboembolism. Alternatively,
tension, coronary artery disease, and pul- a transesophageal echocardiogram can be
monary disease. Thyrotoxicosis and acute al- performed to evaluate for thrombus; if none
cohol consumption can precipitate AF in is found, cardioversion can proceed sooner.
some people. Cardioversion to sinus rhythm can be at-
AF is a potentially dangerous arrhythmia tempted chemically by administration of class
for two reasons: (1) rapid ventricular rates IA, IC, or III antiarrhythmic drugs (see Chap-
may compromise cardiac output, resulting ter 17). Alternatively, electrical cardioversion
in hypotension and pulmonary congestion can be undertaken. Following successful con-
(especially in patients with a hypertrophied version to sinus rhythm, antiarrhythmic
or “stiff” left ventricle in whom the loss of drugs are often continued to prevent recur-
normal atrial contraction can significantly rences. Although antiarrhythmic drugs do
reduce left ventricular filling and stroke vol- not always maintain sinus rhythm during
ume), and (2) the absence of organized atrial long-term follow-up, they can reduce the
contraction promotes blood stasis in the number of AF episodes. Note that these drugs
atria, which increases the risk of thrombus can also cause serious, sometimes lethal,
formation, particularly in the left atrial ap- side effects (see Chapter 17). Thus, in pa-
pendage. Embolization of left atrial thrombi tients with asymptomatic AF, it is often safer
is an important cause of stroke. Treatment to simply continue chronic anticoagulation
of AF therefore focuses on three aspects of therapy and to control the ventricular rate
the arrhythmia: (1) ventricular rate control, with β-blockers, calcium channel blockers,
(2) attempts to restore sinus rhythm, and or digoxin.
(3) assessment of the need for anticoagula- Because the efficacies and toxicities of an-
tion to prevent thromboembolism. tiarrhythmic drugs have been disappointing,
Antiarrhythmic drug treatment of AF is nonpharmacologic options for management
similar to that of atrial flutter. β-Blockers or of AF have been devised. For example, the
certain Ca++ channel antagonists (diltiazem, surgical “maze procedure” places multiple
verapamil) are administered to promote incisions in the left and right atria to prevent
block at the AV node and to reduce the ven- the formation of reentry circuits. A less in-
10090-12_CH12.qxd 8/31/06 4:55 PM Page 297

Clinical Aspects of Cardiac Arrhythmias 297

vasive approach is percutaneous catheter ab- tricle. Enhanced automaticity and triggered
lation of regions around the pulmonary activity in the atrium or AV node are less
veins in the left atrium. This procedure abol- common causes.
ishes atrial fibrillation in some patients, pos-
sibly by ablating the triggering atrial foci that
AV Nodal Reentrant Tachycardia
initiate the fibrillation, as well as by damag-
ing areas that may promote atrial reentry. Atrioventricular nodal reentrant tachycar-
It requires extensive catheter manipulation dia (AVNRT) is the most common form of
and ablation in the left atrium, and risks or PSVT in adults. In the normal heart, the AV
the procedure include systemic thrombo- node is a lobulated structure that consists of
embolism and cardiac perforation that can a compact portion and several atrial exten-
cause tamponade (see Chapter 14). Thus, sions. The latter constitute two (or more)
catheter ablation for AF is usually reserved potential pathways for conduction through
for patients who remain quite symptomatic the AV node (Fig. 12.15). In some people, Fig. 15
despite pharmacologic approaches. these extensions conduct at different veloc-
When sinus rhythm cannot be maintained ities, providing both slow-conducting and
and the heart rate cannot be controlled ade- fast-conducting pathways. The fast pathway
quately with medications, catheter ablation is characterized by a rapid conduction ve-
of the AV node is another procedure that can locity, whereas the slow pathway demon-
be undertaken. This method intentionally strates slower conduction but typically has a
creates complete heart block as a means to shorter refractory period than the fast path-
permanently slow the ventricular rate. As a way. Thus, although the fast pathway con-
result, permanent pacemaker placement is ducts rapidly, it takes longer to recover be-
also required to generate an adequate ven- tween impulses compared with the slow
tricular rate. pathway. Normally, a stimulus arriving at
the AV node travels down both pathways,
but the impulse traveling down the fast
Paroxysmal Supraventricular
pathway reaches the bundle of His first. By
Tachycardias
the time the impulse traversing the slow path-
Paroxysmal supraventricular tachycardias way reaches the bundle of His, it encounters
(PSVTs) are manifested by (1) sudden onset refractory tissue and is extinguished. Thus,
and termination, (2) atrial rates between under normal conditions, only the fast path-
140 and 250 bpm, and (3) narrow (normal) way impulse makes its way forward to the
QRS complexes, unless aberrant conduction is ventricles (see Fig 12.15A).
Fig. 14 present, as described later (Fig. 12.14). The In contrast, consider what happens when
mechanism of PSVTs is most often reentry an atrial premature beat (APB) spontaneously
involving the AV node, atrium, or an acces- occurs (Fig 12.15B). Because the refractory
sory pathway between an atrium and ven- period of the fast pathway is relatively long,

Figure 12.14. Paroxysmal supraventricular tachycardia caused by AV nodal reen-


try. Retrograde P waves in this example occur simultaneously with, and are “hidden” in,
the QRS complexes.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 298

298 Chapter Twelve

Figure 12.15. Common mechanism of AV nodal reentry. In most patients, the AV


node (gray region in the drawing) is a lobulated structure consisting proximally of sev-
eral atrial extensions and distally of a compact node portion. A. In patients with AV
nodal reentry, two functionally distinct tracts exist within the AV node (termed the
slow and fast pathways). The slow pathway conducts slowly and has a short refrac-
tory period, whereas the fast pathway conducts more rapidly but has a long refractory
period. Impulses from above conduct down both pathways; because the fast pathway
impulse reaches the distal common pathway first, it continues to the bundle of His.
Conversely, the slow pathway impulse arrives later and encounters refractory tissue.
B. An atrial premature beat arrives at the entrance of the two pathways. The fast path-
way is still refractory from the preceding beat and the impulse is blocked, but the slow
pathway has repolarized and is able to conduct. When the impulse reaches the distal
portion of the fast pathway after traveling down the slower pathway, the fast pathway
has repolarized and is able to conduct the impulse in a retrograde direction (exempli-
fying unidirectional block). The impulse can then travel through the atrium and back to
the slow pathway, and a reentrant loop is initiated.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 299

Clinical Aspects of Cardiac Arrhythmias 299

an APB would find that pathway unex- heart disease, more severe symptoms may
citable and unable to conduct the impulse. result, such as syncope, angina, or pul-
However, the impulse is able to conduct monary edema.
over the slow pathway (which is excitable Acute treatment of AVNRT is aimed at ter-
because it has a shorter refractory period minating reentry by impairing conduction
than the fast pathway and has already repo- in the AV node. Transient increases in vagal
larized when the APB arrives). By the time tone produced by the Valsalva maneuver or
this impulse travels down the slowly con- carotid sinus massage (see Chapter 11) may
ducting pathway and reaches the compact block AV conduction, terminating the tachy-
portion of the AV node, the distal end of the cardia. The most rapidly effective pharmaco-
fast pathway may have had time to repolar- logic treatment is intravenous adenosine,
ize, and the impulse is able to propagate which impairs AV nodal conduction and
both distally (to the bundle of His and ven- often aborts the reentrant rhythm (see Chap-
tricles) and backward to the atria, up the fast ter 17). Other drug options include intra-
pathway in a retrograde direction. On reach- venous calcium channel antagonists (vera-
ing the atria, the impulse can then circulate pamil and diltiazem) or β-blockers.
back down the slow pathway, completing Most patients with AVNRT have infre-
the reentrant loop and initiating tachycar- quent episodes that terminate with vagal
dia as this sequence repeats. Thus, the fun- maneuvers and do not require other specific
damental conditions for reentry in AVNRT interventions. Frequent symptomatic
in this example are transient unidirectional episodes, particularly when requiring visits
block in the fast pathway (an APB encoun- to the emergency department for treatment,
tering refractory tissue) and relatively slow warrant preventive therapy: oral β-blockers,
conduction through the other pathway. calcium channel blockers, or digoxin are
The ECG in AVNRT shows a regular tachy- often successful for this purpose. Catheter
cardia with normal-width QRS complexes. ablation of the slow AV nodal pathway is
P waves may not be apparent, because retro- also an effective option but is associated
grade atrial depolarization typically occurs si- with a small risk (approximately 1% to 2%)
multaneously with ventricular depolariza- of heart block owing to unintended damage
tion. Thus, the retrograde P wave and QRS are to the fast AV nodal pathway, which re-
inscribed at the same time and the P is typi- quires permanent pacemaker implantation.
cally “hidden” in the QRS complex. When Chronic class IA or IC antiarrhythmic drugs
P waves are visible, they are superimposed on are also effective but are often less desirable
the terminal portion of the QRS complex and than catheter ablation because of associated
inverted (negative deflection) in limb leads II, potential toxicities.
III, and aVF because of the caudocranial di-
rection of atrial activation.
Atrioventricular Reentrant Tachycardias
Rarely, the reentrant loop revolves in the
reverse direction, with anterograde conduc- Atrioventricular reentrant tachycardias
tion down the fast pathway, and retrograde (AVRTs) are similar to AVNRTs except that
conduction up the slow pathway. This is in the former, one limb of the reentrant
known as uncommon AVNRT and, unlike the loop is constituted by an accessory path-
more common rhythm, results in clearly way (bypass tract), rather than by separate
visible retrograde P waves following the QRS fast and slow pathways within the AV node
complex on the ECG. itself. As described in Chapter 11, an acces-
AVNRT often presents in teenagers or sory pathway is an abnormal band of myo-
young adults. It is usually well tolerated but cytes that spans the AV groove and con-
causes palpitations that many patients find nects atrial to ventricular tissue separately
frightening, and rapid tachycardias can cause from the normal conduction system (see
light-headedness or shortness of breath. In Fig. 11.10). Approximately 1 in 1,500 peo-
elderly patients or those with underlying ple has such a pathway.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 300

300 Chapter Twelve

Accessory pathways allow an impulse to one from the normal His-Purkinje system
conduct from atrium to ventricle (antero- (Figs. 12.16 and 12.17). Fig. 16-17
grade conduction), from ventricle to atrium Patients with WPW syndrome are predis-
(retrograde conduction), or in both direc- posed to PSVTs because the accessory path-
tions. Depending on the characteristics of way provides a potential limb of a reentrant
the pathway, one of two characteristic enti- loop. The most common PSVT in these
ties can result: (1) the ventricular preexcita- patients is orthodromic AVRT. During this
tion syndrome, or (2) PSVT resulting from a tachycardia, an impulse travels anterogradely
concealed bypass tract. Some pathways do down the AV node to the ventricles and
not conduct impulses at rates sufficient to then retrogradely up the accessory tract back
cause tachycardias and cause no symptoms to the atria (see Fig. 12.17B). Because the
at all. ventricles in this situation are depolarized
exclusively via the normal conduction sys-
tem (through the AV node and bundle of
Ventricular Preexcitation Syndrome
His), there is no delta wave during the
In patients with ventricular preexcitation tachycardia and the width of the QRS is usu-
(also termed Wolff-Parkinson-White [WPW] ally normal. Retrograde P waves are often
syndrome; see Chapter 11), atrial impulses visible soon after each QRS complex because
can pass in an anterograde direction to the the atria are stimulated from below via ret-
ventricles through both the AV node and rograde conduction through the accessory
the accessory pathway. Because conduction pathway.
through the accessory pathway is usually In fewer than 10% of patients with AVRT
faster than that via the AV node, the ventri- involving an accessory pathway, the re-
cles are stimulated earlier (preexcited) than entrant arrhythmia travels in the opposite di-
by normal conduction over the AV node. rection. Impulses travel anterogradely down
During sinus rhythm, activation of the ven- the accessory pathway and retrogradely up the
tricle from the accessory pathway causes a AV node (see Fig. 12.17C). Termed antidromic
characteristic ECG appearance: (1) the PR AVRT, its ECG pattern is characterized by a
interval is short (<0.12 sec) because ventric- wide QRS complex because the ventricles are
ular stimulation begins earlier than normal activated entirely from anterograde conduc-
through the accessory pathway, (2) the QRS tion over the accessory pathway. From the
has a slurred rather than a sharp upstroke ECG alone, such antidromic tachycardia
(referred to as a delta wave) because initial is difficult to distinguish from ventricular
ventricular activation by the accessory path- tachycardia (described later in the chapter).
way is slower than activation over the Purk- A third type of arrhythmia encountered
inje system, and (3) the QRS complex is in patients with WPW syndrome is antero-
widened because it represents fusion of two grade conduction over the accessory path-
excitation wave fronts through the ventri- way when atrial fibrillation or flutter is pre-
cles, one from the accessory pathway and sent. Some accessory pathways have short

Figure 12.16. Wolff-Parkinson-White syndrome. The delta wave (arrow) indicates preexcitation of the ven-
tricles. (Courtesy of Dr. Eric Isselbacher, Massachusetts General Hospital, Boston.)
10090-12_CH12.qxd 8/31/06 4:55 PM Page 301

Clinical Aspects of Cardiac Arrhythmias 301

Figure 12.17. Wolff-Parkinson-White syndrome. A. During normal sinus rhythm, the shortened PR interval, delta
wave, and widened QRS complex indicate fusion of ventricular activation via the AV node and accessory pathway. B. An
atrial premature beat can trigger an orthodromic atrioventricular reentrant tachycardia, in which impulses are conducted
anterogradely down the AV node and retrogradely up the accessory pathway. Retrograde P waves are visible immedi-
ately after the QRS complex. There is no delta wave because anterograde ventricular stimulation passes exclusively
through the AV node. C. Antidromic atrioventricular reentrant tachycardia in which impulses are conducted antero-
gradely down the accessory tract and retrogradely up the AV node. The QRS complex is very widened because the ven-
tricles are stimulated by abnormal conduction through the accessory pathway. SA, sinoatrial.

refractory periods that allow faster rates of duction. Therefore, the drugs could precipi-
ventricular stimulation than does the AV tate even faster ventricular rates (and hemo-
node. Thus, during AF or atrial flutter, ven- dynamic collapse) when administered to pa-
tricular rates as fast as 300 bpm may result. tients with WPW syndrome who develop
Such rates are poorly tolerated and can lead atrial fibrillation or flutter. In contrast,
to ventricular fibrillation and cardiac arrest, sodium+ channel blockers (specifically, class
even in a young, otherwise healthy patient. IA and IC antiarrhythmics) and some class
Pharmacologic management of arrhyth- III antiarrhythmic drugs slow conduction
mias in patients with the WPW syndrome and prolong the refractory period of acces-
requires greater caution than those with sory pathways as well as the AV node; there-
AVNRTs. Although digitalis, β-blockers, and fore, these are the preferred pharmacologic
certain calcium++ channel blockers are effec- agents for this condition.
tive at blocking conduction through the When a patient with WPW presents with a
AV node, they do not slow conduction over wide QRS tachycardia, acute therapy depends
most accessory pathways. Sometimes these on the patient’s hemodynamic tolerance of
drugs actually shorten the refractory period the arrhythmia. If accompanied by hemody-
of the accessory pathway, thus speeding con- namic collapse, immediate cardioversion is
10090-12_CH12.qxd 8/31/06 4:55 PM Page 302

302 Chapter Twelve

required. Conversely, if the patient is hemo- recurrent episodes is catheter ablation of the
dynamically stable, intravenous administra- accessory pathway, which permanently pre-
tion of procainamide (a class IA agent that vents recurrences in most patients.
slows conduction in the accessory pathway)
will often terminate the arrhythmia.
Ectopic Atrial Tachycardia
Patients who have WPW with symptom-
atic arrhythmias should generally under- Ectopic atrial tachycardia (AT) results from
go an electrophysiologic study with radio- either automaticity of an atrial focus or re-
frequency catheter ablation of the accessory entry. The ECG has the appearance of sinus
pathway. Ablation abolishes conduction tachycardia, with a P wave before each QRS
over the pathway, curing the condition. If complex, but the P wave morphology is dif-
this procedure is not an option, chronic oral ferent from that of sinus rhythm, indicating
therapy should include a drug that slows ac- depolarization of the atrium from an abnor-
cessory pathway conduction (i.e., a class IA, mal site. The arrhythmia can be paroxysmal
IC, or III agent). and of limited duration, or it can persist.
Another form of ventricular preexcitation Short, asymptomatic bursts of atrial tachy-
is the Lown-Ganong-Levine syndrome, an cardia are commonly observed on 24-hour
uncommon condition also characterized by ECG recordings, even in otherwise healthy
a short PR interval but a normal, narrow QRS people.
complex (i.e., no delta wave during sinus Atrial tachycardia can be caused by digi-
rhythm). Although it has been suggested that talis toxicity and is also aggravated by ele-
a short accessory pathway connects the atria vated sympathetic tone (e.g., during exertion
directly to the His-Purkinje system in this or periods of illness). Initial treatment in-
condition, most patients just have enhanced cludes correction of any contributing factors.
conduction through the normal AV node, Unlike reentrant supraventricular tachycar-
thus shortening the PR interval. When PSVT dias, vagal maneuvers (such as carotid sinus
occurs in these patients, it is usually owing to massage) may have no effect on atrial dis-
AV nodal reentry. charges from an ectopic pacemaker focus.
However, β-blockers; calcium channel block-
ers; and class IA, IC, and III antiarrhythmic
Concealed Accessory Pathways
drugs can be effective. Catheter ablation is
Accessory pathways do not always result in also an option for symptomatic patients.
ECG findings of ventricular preexcitation
(i.e., short PR, delta wave). Many are capable
Multifocal Atrial Tachycardia
of only retrograde conduction. In this case,
during sinus rhythm, the ventricles are depo- In multifocal atrial tachycardia (MAT), the
larized normally through the AV node alone ECG shows an irregular rhythm with multi-
and the ECG is normal (i.e., the accessory ple (at least three) P wave morphologies,
pathway is concealed). However, because the and the average atrial rate is >100 bpm (Fig.
accessory pathway is capable of retrograde 12.18). An isoelectric (i.e., “flat”) baseline Fig. 18
conduction, it can form a limb of a reentrant between P waves distinguishes MAT from
circuit and result in orthodromic AVRT. the chaotic baseline of AF. This rhythm is
Management of patients with tachycardia likely caused by either abnormal automatic-
involving a concealed accessory pathway is ity in several foci within the atria or trig-
the same as for patients with AVNRT. Be- gered activity and occurs most often in the
cause the reentrant circuit travels antero- setting of severe pulmonary disease and hy-
gradely down the AV node, vagal maneuvers poxemia. Because patients with this rhythm
and drugs that interrupt conduction over are often critically ill from the underlying
the AV node (e.g., adenosine, verapamil, dil- disease, the mortality rate is high, and treat-
tiazem, and β-blockers) can terminate the ment is aimed at the causative disorder. The
tachycardia. Another option in patients with calcium channel blocker verapamil is often
10090-12_CH12.qxd 8/31/06 4:55 PM Page 303

Clinical Aspects of Cardiac Arrhythmias 303

Figure 12.18. Multifocal atrial tachycardia. Each QRS is preceded by a P wave (arrows) of varying
morphology. (Courtesy of Dr. Eric Isselbacher, Massachusetts General Hospital, Boston.)

effective at slowing the ventricular rate as a VPBs can also occur in repeating patterns.
temporizing measure. When every alternate beat is a VPB, the
rhythm is termed bigeminy. When two nor-
mal beats precede every VPB, trigeminy is
Ventricular Arrhythmias
said to be present. Three preceding normal
The common ventricular arrhythmias are beats are referred to as quadrigeminy, and
(1) ventricular premature beats, (2) ventricu- so on. Consecutive VPBs are referred to as
lar tachycardia,and (3) ventricular fibrillation. couplets (two in a row) or triplets (three in
Ventricular arrhythmias are usually more a row).
dangerous than supraventricular rhythm dis- VPBs are not dangerous by themselves,
orders and are responsible for many of the ap- and in patients without heart disease, they
proximately 300,000 sudden cardiac deaths confer no added risk of a life-threatening ar-
that occur every year in the United States. rhythmia. They can, however, be an indica-
tion of an underlying cardiac disorder and
take on added significance in that case. For
Ventricular Premature Beats
example, in patients with structural heart
Similar to APBs, ventricular premature beats disease, VPBs generally increase in frequency
(VPBs) are common even among healthy in relation to the severity of depressed ven-
people and are often asymptomatic and be- tricular contractility. They have been associ-
Fig. 19 nign (Fig. 12.19). A VPB arises when an ec- ated with an increased risk of sudden death
topic ventricular focus fires an action poten- in patients with heart failure or prior my-
tial. On the ECG, a VPB appears as a widened ocardial infarction. Therefore, the discovery
QRS complex, because the impulse travels of VPBs warrants assessment for underlying
from its ectopic site through the ventricles heart conditions.
via slow cell-to-cell connections rather than In otherwise healthy persons, treatment
through the normal rapidly conducting His- of VPBs mainly involves reassurance and,
Purkinje system. Furthermore, the ectopic if needed, symptomatic control using β-
beat is not related to a preceding P wave. blockers. In patients with advanced structural

Figure 12.19. Ventricular premature beats (arrows).


10090-12_CH12.qxd 8/31/06 4:55 PM Page 304

304 Chapter Twelve

heart disease who are at risk of life-threaten- to beat, the VT is referred to as polymorphic.
ing ventricular arrhythmias, placement of Multiple ectopic foci or a continually chang-
an implantable cardioverter-defibrillator ing reentry circuit is the cause. Torsades de
(ICD) is typically recommended. pointes (discussed later in the chapter) and
acute myocardial ischemia or infarction are
Ventricular Tachycardia the most common causes of polymorphic
VT. Rare, inherited predispositions to poly-
Ventricular tachycardia (VT) is a series of morphic ventricular tachycardia and sudden
Fig. 20 three or more VPBs (Fig. 12.20). VT is divided death arise from abnormalities of cardiac
arbitrarily into two categories. If it persists potassium and sodium channels (e.g., the
for more than 30 seconds, produces severe long-QT syndromes and the Brugada syn-
symptoms such as syncope, or requires ter- drome), as described in Box 12.1. Sustained
mination by cardioversion or administration polymorphic VT usually degenerates to
of an antiarrhythmic drug, it is designated as ventricular fibrillation.
sustained VT; self-terminating episodes are The symptoms of VT vary depending on
termed nonsustained VT. Both forms of VT the rate of the tachycardia, its duration, and
are found most commonly in patients with the underlying condition of the heart. Sus-
structural heart disease, including myocar-
tained VT can cause low cardiac output re-
dial ischemia and infarction, heart failure,
sulting in loss of consciousness (syncope),
ventricular hypertrophy, primary electrical
pulmonary edema, or progress to cardiac
Box 1 diseases (e.g., long-QT syndromes; see Box
arrest. These severe consequences of VT are
12.1), valvular heart diseases, and congenital
most likely in patients who have under-
cardiac abnormalities.
lying depressed contractile function. Con-
The QRS complexes of VT are typically
versely, If sustained VT is relatively slow
wide (>0.12 sec) and occur at a rate of 100
(e.g., <130 bpm) it may be well tolerated
to 200 bpm or sometimes faster. VT is fur-
and cause only palpitations.
ther categorized according to its QRS mor-
phology. When every QRS complex ap-
pears the same and the rate is regular, it is Distinguishing Monomorphic VT
referred to as monomorphic VT (see Fig. from Supraventricular Tachycardia
12.20). Sustained monomorphic VT usu-
ally indicates a structural abnormality that Ventricular tachycardia can usually be dis-
supports a reentry circuit, most commonly tinguished from supraventricular tachycar-
a region of myocardial scar from an old in- dia (SVT) by the width of the QRS com-
farction or cardiomyopathy. Occasionally, plex: it is routinely wide in the former and
sustained monomorphic VT occurs as a re- narrow (i.e., normal) in the latter, as indi-
sult of an ectopic ventricular focus in an cated in Figure 12.9. However, under cer-
otherwise healthy person, usually referred tain circumstances, arrhythmias that orig-
to as idiopathic VT. inate from sites above the ventricles can
When the QRS complexes continually result in wide QRS complexes and may ap-
change in shape and the rate varies from beat pear similar to monomorphic VT. This sit-

Figure 12.20. Monomorphic ventricular tachycardia.


10090-12_CH12.qxd 8/31/06 4:55 PM Page 305

Clinical Aspects of Cardiac Arrhythmias 305

uation is termed supraventricular tachycar- be managed as though they have VT until


dia with aberrant ventricular conduction, or proven otherwise.
simply SVT with aberrancy, and may arise
in three scenarios: (1) a patient has an un-
Management of Patients with VT
derlying conduction abnormality (e.g., a
bundle branch block), such that the QRS is Sustained episodes of VT are dangerous be-
abnormally wide even when in normal cause they can produce syncope or deterio-
sinus rhythm; (2) repetitive rapid ventric- rate into ventricular fibrillation, which is
ular stimulation during an SVT finds one fatal if not quickly corrected. Acute treat-
of the bundle branches refractory (because ment usually consists of electrical cardiover-
of insufficient time to recover from the sion. Intravenous administration of certain
previous depolarization), such that the im- antiarrhythmic drugs, such as amiodarone,
pulse propagates abnormally through the procainamide, or lidocaine, can be con-
ventricles, causing the QRS to be distorted sidered if the patient is hemodynamically
and wide; or (3) a patient develops an- stable.
tidromic tachycardia through an accessory After sinus rhythm is restored, a patient
pathway (described earlier). who has had an episode of sustained VT re-
Certain clinical and electrocardiographic quire careful evaluation to define whether
features can help to distinguish wide QRS underlying structural heart disease is pre-
complexes of monomorphic VT from those sent and to correct any aggravating factors,
of supraventricular rhythms with aberrant such as acute myocardial ischemia, electro-
conduction. In patients with a history of lyte disturbances, or drug toxicities. Patients
prior myocardial infarction, congestive who have suffered VT in the setting of struc-
heart failure, or left ventricular dysfunc- tural heart disease have a high risk of recur-
tion, a wide complex tachycardia is more rence and sudden cardiac death; implan-
likely to be VT rather than SVT with aber- tation of an ICD is usually warranted to
rancy. At the bedside, SVT is more probable automatically and promptly terminate fu-
if vagal maneuvers (such as carotid sinus ture episodes.
massage) affect the rhythm, as indicated in Patients who experience VT in the ab-
Figure 12.9. sence of underlying structural heart disease
Electrocardiographically, a supraventricu- are usually found to have idiopathic VT.
lar tachyarrhythmia is more likely if the This type of arrhythmia tends to originate
morphology of the QRS at the rapid rate is from foci in the right ventricular outflow
similar to that on the patient’s ECG tracing tract or in the septal portion of the left ven-
obtained while in sinus rhythm (i.e., the tricle. It is rarely life threatening. β-Blockers,
complex is widened because of an underly- calcium channel blockers, or catheter abla-
ing bundle branch block). Conversely, ven- tion are commonly effective to control symp-
tricular tachycardia is more likely if (1) there tomatic episodes of idiopathic VT.
is no relationship between the QRS com-
plexes and any observed P waves (atrioven-
Torsades de Pointes
tricular dissociation) or (2) the QRS com-
plexes in each of the chest leads (V1 through Torsades de pointes (“twisting of the points”)
V6) all have a similar appearance, with a is a form of polymorphic VT presenting as
dominant positive or negative deflection varying amplitudes of the QRS, as if the
(i.e., there is “concordance” of the precor- complexes are “twisting” about the base-
dial QRS complexes). These features are line (Fig. 12.21). It can be produced by Fig. 21
Tab. 2 summarized in Table 12.2. Other morpho- early afterdepolarizations (triggered activ-
logic ECG features have been used to distin- ity), particularly in patients who have a pro-
guish VT from SVT with aberrancy, but the longed QT interval. QT prolongation (which
distinction is often very difficult. Most pa- indicates a lengthened action potential du-
tients with wide QRS tachycardia should ration) can result from electrolyte distur-
10090-12_CH12.qxd 8/31/06 4:55 PM Page 306

306 Chapter Twelve

Box 12.1 Genetic Mutations and Ventricular Arrhythmias

Genetic causes of ventricular arrhythmias occur either in association with various types of
structural heart disease or as isolated conditions. Examples of inherited structural disease
that can be complicated by life-threatening arrhythmias include hypertrophic cardiomyo-
pathy and the familial dilated cardiomyopathies, both described in Chapter 10. Ar-
rhythmogenic right ventricular dysplasia (ARVD) is another form of cardiomyopathy
associated with reentrant ventricular tachycardia, typically originating from the right ven-
tricle (RV). This condition is characterized by replacement of portions of the RV with adi-
pose and fibrous tissue, and approximately one third of patients display a familial pattern
with autosomal dominant inheritance. The most common mutations involve genes that
encode components of cell membrane desmosomes (structures involved in cell-to-cell ad-
hesion) and the cardiac ryanodine receptor (see Chapter 1). ARVD may be suspected on
routine electrocardiogram (ECG) by the presence of inverted T waves in leads V1 through
V3 and occasionally an epsilon wave, a terminal notch of the QRS complex in lead V1 (see
arrow in the accompanying figure), which reflects abnormal
RV activation. The abnormal RV morphology can be identified V1
by noninvasive imaging techniques in some patients. Treat-
ment typically includes an implantable cardioverter-defibrilla-
tor (ICD) because the disease is progressive and ventricular
tachycardia is common, which can lead to sudden death.
Several other inherited arrhythmic disorders occur in the
absence of structural cardiac disease. These occur infrequently but are important because
they can cause life-threatening arrhythmias in young, otherwise healthy people without
prior warning. The most common of these conditions are (1) the Brugada syndrome,
(2) the congenital long-QT syndromes, and (3) familial catecholaminergic polymorphic
ventricular tachycardia.
The Brugada syndrome is believed to be responsible for a large percentage of idio-
pathic ventricular fibrillation. It is inherited in an autosomal dominant fashion and has
been linked in some (but not all) families to mutations in a sodium channel subunit gene
(SCN5A). A clue to the presence of this syndrome is a specific ECG abnormality: right bun-
dle branch block with prominent ST elevation in leads V1
through V3 (see accompanying figure). This pattern may be
V1
present chronically or intermittently; in the latter case, the
syndrome may be unmasked by administering certain an-
tiarrhythmic drugs (e.g., flecainide, procainamide). Brugada
syndrome is a potentially lethal condition, and ICD implan-
tation is the most effective way to prevent an arrhythmic
death.
The congenital long-QT syndromes (LQTS) are associated with prolonged ventric-
ular repolarization (hence the long QT interval), which can lead to life-threatening poly-
morphic ventricular tachycardia (i.e., torsades de pointes). Mutations in at least seven
genes result in LQTS (see the accompanying table) by prolonging the action potential
duration. As shown in the table, identified mutations either enhance the depolarizing
Na+ current, impair the repolarizing K+ current, or in one case (LQT4), result in an ab-
normal cellular structural protein. There are two general phenotypes of LQTS: (1) the
Romano-Ward syndrome, transmitted in an autosomal dominant fashion (an inheritance
pattern that occurs with mutations in any of the listed genes); and (2) Jervelle and Lange-
10090-12_CH12.qxd 8/31/06 4:55 PM Page 307

Clinical Aspects of Cardiac Arrhythmias 307

Genetic Basis of Congenital Long-QT Syndromes

Gene Mechanism of Prolonged


Type (location) Protein Repolarization Inheritance

LPT1 KCNQ1 KvLQT1 ↓Outward K+ current AD and AR


(11p15) (α subunit of IKs K+ channel)
LQT2 KCNH2 HERG ↓Outward K+ current AD
(7q35) (α subunit of IKr K+ channel)
LQT3 SCN5A Nav 1.5 ↑Inward Na+ current AD
(3p21) (Na+ channel)
LQT4 ANK2 Ankyrin-B Not yet known AD
(4q25) (structural protein)
LQT5 KCNE1 MinK ↓Outward K+ current AD and AR
(21q22) (β subunit of IKs K+ channel)
LQT6 KCNE2 MiRP1 ↓Outward K+ current AD
(21q22) (β subunit of IKr K+ channel)
LQT7 KCNJ2 IK1 ↓outward K+ current AD
(17q23) (Kir2.1 inward rectifier K+ channel)

AD, autosomal dominant (i.e., Romano-Ward syndrome); AR, autosomal recessive (i.e., Jervell and Lange-Nielsen
syndrome [long QT and sensorineural deafness]).

Nielsen syndrome, an autosomal recessive condition associated with QT prolongation


combined with sensorineural hearing loss that results from mutations in two of the
seven loci.
The symptomatology of patients with LQTS is highly variable, even for patients with the
same mutation. The degree of QT prolongation, and in some cases the patient’s gender,
are predictors of arrhythmic risk when a mutation is present. An affected patient may be
asymptomatic and come to medical attention only as a result of the abnormal ECG, or
because of a family member with this condition. Others present with syncope or even sud-
den death caused by torsades de pointes. The most common forms (LQT1 and LQT2) are
associated with ventricular arrhythmias during physical exercise or emotional stress. Con-
versely, those with LQT3 are much more likely to experience cardiac events at rest or dur-
ing sleep.
Other acquired conditions that prolong the QT interval can trigger life-threatening
arrhythmias in patients with LQTS, including hypokalemia, hypomagnesemia, hypocal-
cemia, and several medications (including many antiarrhythmic drugs). Conversely,
β-blockers reduce the risk of arrhythmias in many forms of LQTS, even though they do not
shorten the QT interval (see discussion of torsades de pointes later in the chapter). For pa-
tients at high risk of life-threatening arrhythmias, ICD implantation is warranted.
Familial catecholaminergic polymorphic ventricular tachycardia, inherited in
autosomal dominant and recessive patterns, is marked by ventricular tachycardia and/or
ventricular fibrillation during exercise or emotional arousal. The mechanism is thought to
be triggered activity resulting from delayed afterdepolarizations (see Chapter 11). Muta-
tions in affected families have been demonstrated in at least two genes involved in intra-
cellular calcium handling, including a missense mutation in the locus that codes for the car-
diac ryanodine receptor. β-Blockers are effective for some patients; otherwise, an ICD is
implanted.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 308

308 Chapter Twelve

TABLE 12.2. Differentiation of Wide Complex Tachycardias

Supports SVT with Aberrant Conduction Supports Ventricular Tachycardia

QRS morphology same as when in No relationship between P wave and QRS


sinus rhythm complexes
Rhythm responds to vagal Concordance of QRS complexes in the chest leads
maneuvers (see Fig. 12.9) (V1–V6)

SVT, supraventricular tachycardia.

bances (especially hypokalemia or hypo- terenol) and accelerating the heart rate via an
magnesemia), persistent bradycardia, and artificial pacemaker. Paradoxically, when tor-
drugs that block cardiac potassium currents, sades de pointes results from congenital pro-
including many antiarrhythmic agents longation of the QT interval, β-blocking drugs
(particularly the class III drugs sotalol, ibu- are often the treatment of choice because
tilide, and dofetilide and some class I sympathetic stimulation actually aggravates
drugs, including quinidine, procainamide, the arrhythmia. An implantable defibrillator
and disopyramide). Many medications given is often appropriate for these patients.
for noncardiac illnesses can prolong the
QT interval and predispose to torsades de
Ventricular Fibrillation
pointes, including erythromycin, phenoth-
iazines, haloperidol, and methadone. A rare Ventricular fibrillation (VF) is an immedi-
group of hereditary ion channel abnormal- ately life-threatening arrhythmia (Fig. 12.22). Fig. 22
ities produce congenital QT prolongation, It results in disordered, rapid stimulation of
which can also lead to torsades de pointes the ventricles with no coordinated contrac-
(see Box 12.1). tion. The result is essentially cessation of
Torsades de pointes is usually sympto- cardiac output and death if not quickly re-
matic, causing light-headedness or syncope, versed. This rhythm most often occurs in
but is frequently self-limited. Its main danger patients with severe underlying heart dis-
results from degeneration into ventricular ease and is the major cause of mortality in
fibrillation. When it is drug or electrolyte in- acute myocardial infarction.
duced, correcting the underlying cause abol- VF is often initiated by an episode of ven-
ishes the arrhythmia. Administration of tricular tachycardia, which degenerates, it is
intravenous magnesium often suppresses believed, by the breakup of excitation waves
recurrent episodes. Other preventive strate- into multiple smaller wavelets of reentry
gies are aimed at shortening the QT interval that wander through the myocardium. On
by increasing the underlying heart rate. Such the ECG, VF is characterized by a chaotic ir-
strategies include administering intravenous regular appearance without discrete QRS
β-adrenergic stimulating agents (e.g., isopro- waveforms.

Figure 12.21. Torsades de pointes. The widened polymorphic QRS complexes demonstrate a waxing and
waning pattern; the QT interval was prolonged before the onset of the arrhythmia (not shown).
10090-12_CH12.qxd 8/31/06 4:55 PM Page 309

Clinical Aspects of Cardiac Arrhythmias 309

Figure 12.22. Ventricular fibrillation.

Untreated, VF rapidly leads to death. The Each of the ECG texts listed at the end of
only effective therapy is prompt electrical Chapter 4 provides additional examples
defibrillation. As soon as the heart has been of the rhythm disorders presented in this
converted to a safe rhythm, the underlying chapter.
precipitant of the arrhythmia (e.g., elec-
trolyte imbalances, hypoxemia, or acidosis)
Acknowledgments
should be sought and corrected to prevent
further episodes. Intravenous antiarrhyth- Contributors to the previous editions of this chapter
mic drug therapy may be administered to were Wendy Armstrong, MD; Nicholas Boulis, MD;
Jennifer E. Ho, MD; Marc S. Sabatine, MD; Elliott M.
prevent immediate recurrences. If no re-
Antman, MD; Leonard I. Ganz, MD; and Leonard S.
versible inciting precipitant is found, sur- Lilly, MD.
vivors of VF usually receive an ICD.

Additional Reading
SUMMARY Ackerman MJ. Molecular basis of congenital and ac-
quired long QT syndromes. J Electrocardiol 2004;
1. Disorders of impulse formation and con-
37(suppl):1–6.
duction result in bradyarrhythmias and
Delacretaz E. Supraventricular Tachycardia. N Engl
tachyarrhythmias. Through careful analy- J Med 2006;354:1039–1051.
sis of the ECG, it is usually possible to dis- Goldberger Z, Lambert R. Implantable cardioverter-
tinguish among the array of rhythm dis- defibrillators. JAMA 2006;295:809–818.
orders so that appropriate therapy can be Gregoratos G, Abrams J, Epstein AE, et al. ACC/AHA/
administered. NASPE 2002 guideline update for implantation of
cardiac pacemakers and antiarrhythmia devices:
2. When evaluating a patient with a slow
summary article. A report of the American College
heart rhythm (Figs. 12.1–12.8), two key of Cardiology/American Heart Association task
questions should be addressed: force on practice guidelines (ACC/AHA/NASPE
a. Are P waves present? committee to update the 1998 pacemaker guide-
lines). Circulation 2002;106:2145.
b. What is the relationship between the
Iqbal MB, Taneja AK, Lip GY, Flather M. Recent de-
P waves and the QRS complexes? velopments in atrial fibrillation. BMJ 2005;330:
3. Differentiation of tachyarrhythmias re- 238–43.
quires assessment of the following: Kléber AG, Rudy Y. Basic mechanisms of cardiac im-
a. The width of the QRS complex (nor- pulse propagation and associated arrhythmias.
Physiol Rev 2004; 84:431–488.
mal or wide)
McNamara RL, Tamariz LJ, Segal JB, et al. Manage-
b. The morphology and rate of the ment of atrial fibrillation: review of the evidence
P waves for the role of pharmacologic therapy, electrical
c. The relationship between the P waves cardioversion, and echocardiography. Ann Intern
Med 2003;139:1018.
and the QRS complexes
Morady F. Catheter ablation of supraventricular ar-
d. The response to vagal maneuvers (see rhythmias: state of the art. J Cardiovasc Electro-
Fig. 12.9) physiol 2004;15:124–139.
10090-12_CH12.qxd 8/31/06 4:55 PM Page 310

310 Chapter Twelve

Roberts R. Genomics and cardiac arrhythmias. J Am Shah M, Akar FG, Tomaselli GF. Molecular basis of
Coll Cardiol 2006;47:9–21. arrhythmias. Circulation 2005;112:2517–2529.
Roden DM. Drug-induced prolongation of the QT Trohman RG, Kim MH, Pinski SL. Cardiac pacing:
interval. N Engl J Med 2004;350:1013–1022. the state of the art. Lancet 2004;364:1701–1719.
Schwartz PJ, Spazzolini C, Crotti L, et al. The Jervell Wellens HJ. Twenty-five years of insights into the
and Lange-Nielson Syndrome—natural history, mechanisms of supraventricular arrhythmias.
molecular basis, and clinical outcome. Circulation J Cardiovasc Electrophysiol 2003;14:1020–1025.
2006;113:783–790.
10090-12_CH12.qxd 8/31/06 4:55 PM Page a

Chapter 12—Author Query


1. AU: Previously, atrial rate given in bpm. Correct here, too? If so, is 350–600 still OK?
10090-13_CH13.qxd 8/31/06 4:55 PM Page 311

C H A P T E R

Hypertension
Payman Zamani
Gordon H. Williams
13
Leonard S. Lilly

WHAT IS HYPERTENSION? Renal Causes


Mechanical Causes
HOW IS BLOOD PRESSURE REGULATED?
Endocrine Causes
Four Regulatory Systems
Blood Pressure Reflexes CONSEQUENCES OF HYPERTENSION
Clinical Signs and Symptoms
ESSENTIAL HYPERTENSION
Organ Damage Caused by Hypertension
Epidemiology and Genetics
Experimental Findings HYPERTENSIVE CRISIS
Natural History TREATMENT OF HYPERTENSION
SECONDARY HYPERTENSION Nonpharmacologic Treatment
Patient Evaluation Pharmacologic Treatment
Exogenous Causes

More than 50 million Americans have hyper- blood pressure is usually asymptomatic
tension—a blood pressure high enough to until an acute cardiovascular event strikes,
be a danger to their well-being. That num- screening for hypertension is a critical aspect
ber will undoubtedly rise; data from the of preventive medicine.
Framingham Heart Study indicate that 90% Hypertension is also a scientific problem
of people over age 55 will develop hyper- of unexpected complexity. In almost 95% of
tension during their lifetimes. Thus, this affected patients, the cause of the blood
condition represents a great public health pressure elevation is unknown, a condition
concern because it is a major risk factor for termed primary or essential hypertension
coronary artery disease, stroke, heart failure, (EH). Evidence suggests that the causes of EH
renal disease, and peripheral vascular dis- are multiple and diverse, and considerable in-
ease. Surprisingly, more than two thirds of sight into those causes can be achieved by
hypertensive persons are either unaware studying the normal physiology of blood
of their elevated blood pressure or are not pressure control, as examined in this chapter.
treated adequately to minimize the cardio- High blood pressure attributed to a defin-
vascular risk. Moreover, because elevated able cause is termed secondary hyperten-

311
10090-13_CH13.qxd 8/31/06 4:55 PM Page 312

312 Chapter Thirteen

sion. Although far less common than EH, recent evidence suggests that systolic pres-
conditions that cause secondary hyper- sure more accurately predicts cardiovascu-
tension are important because many are lar complications.
amenable to permanent cure. Following the
discussions of essential and secondary hyper-
HOW IS BLOOD
tension, this chapter considers the clinical
PRESSURE REGULATED?
consequences of elevated blood pressure
and approaches to treatment. Four Regulatory Systems
Blood pressure (BP) is the product of cardiac
WHAT IS HYPERTENSION? output (CO) and total peripheral resistance
(TPR):
Blood pressure values vary widely in the
population. For example, diastolic pres- BP = CO × TPR
sures follow the smooth bell-shaped distri-
Fig. 1 bution shown in Figure 13.1. In addition, And CO is the product of cardiac stroke vol-
blood pressure levels tend to increase with ume (SV) and heart rate (HR):
Fig. 2 age, as illustrated in Figure 13.2. The risk of CO = SV × HR
a vascular complication increases progres-
sively and linearly with higher blood pres- Stroke volume is determined by (1) cardiac
sure values, so the exact cutoff points to contractility; (2) the venous return to the
define stages of hypertension are some- heart (i.e., the preload, as described in Chap-
what arbitrary. The currently established ter 9); and (3) the resistance the left ventri-
Tab. 1 criteria are listed in Table 13.1. By this clas- cle must overcome to eject blood into the
sification, a diastolic pressure consistently aorta (i.e., afterload).
at or above 90 mm Hg or a systolic pressure It follows that at least four systems are di-
at or above 140 mm Hg establishes the rectly responsible for blood pressure regula-
diagnosis of hypertension. Those with tion: the heart, which supplies the pumping
prehypertension have an increased risk of pressure; blood vessel tone, which largely
developing definite hypertension and are determines systemic resistance; the kidney,
encouraged to undertake lifestyle modifica- which regulates intravascular volume; and
tions. Although the emphasis has histori- hormones, which modulate the function the
cally been on the level of diastolic pressure, other three systems. Figure 13.3 shows how

Figure 13.1. Distribution of diastolic blood pressure values in the


30- to 69-year age group (n 5 158,906). Hypertension is arbitrarily de-
fined as diastolic blood pressure ≥90 mm Hg. (Modified from Hyperten-
sion Detection and Follow-up Program. A progress report. Circ Res 1977;
40(suppl1):106.)
10090-13_CH13.qxd 8/31/06 4:55 PM Page 313

Hypertension 313

one typically improves the blood pressure.


Similarly, surgical placement of a kidney
from a genetically hypertensive rat into a
previously normotensive one usually leads
to hypertension.
In the presence of normally functioning
kidneys, an increase in blood pressure leads
to augmented urine volume and sodium ex-
cretion, which then returns the blood pres-
sure back to normal. This process, known as
pressure natriuresis, is blunted in the kidneys
of hypertensive patients; thus, higher pres-
sures are required to excrete a given sodium
and water load. Current evidence suggests at
least two possible reasons for this blunted
response. First, microvascular and tubulo-
interstitial injury within the kidneys of hy-
pertensive patients impairs sodium excre-
Figure 13.2. Relationship between blood pressure tion, although the mechanism of this injury
and age (n 5 1,029). Systolic (upper curves) and di- is still under investigation. Second, the de-
astolic (lower curves) values are shown. Notice that fect may lie with hormonal factors critical to
by age 60, the average systolic pressure of women ex-
ceeds that of men. (Modified from Kotchen JM, McKean
appropriate renal reactions to the sodium
HE, Kotchen TA. Blood pressure trends with aging. Hy- and intravascular volume environment
pertension 1982;4(suppl 3):111–129.) (e.g., the renin-angiotensin system, as de-
scribed later in the chapter). In contrast to
the first possibility, abnormalities of hor-
factors related to these systems contribute to monal regulation are amendable to correc-
CO and TPR. tion with appropriate therapy.
The renal component of blood pressure
regulation deserves special mention, in light
Blood Pressure Reflexes
of the temptation to view hypertension sim-
ply as a cardiovascular problem. No matter The cardiovascular system is endowed with
how high the CO or TPR, renal excretion has feedback mechanisms that continuously
the capacity to completely return blood pres- monitor arterial pressure: they sense when
sure to normal by reducing intravascular vol- the pressure becomes excessively high or low
ume. Therefore, the maintenance of chronic and then respond rapidly to those changes.
hypertension requires renal participation. One such mechanism is the baroreceptor
Transplantation studies have confirmed this reflex, which is mediated by receptors in the
point: the implantation of a kidney from a walls of the aortic arch and the carotid si-
normotensive person into a hypertensive nuses. The baroreceptors monitor changes in

TABLE 13.1. Classification of Blood Pressure in Adults

Category Systolic Pressure (mm Hg) Diastolic Pressure (mm Hg)

Normal <120 And <80


Prehypertension 120–139 Or 80–99
Stage 1 hypertension 140–159 Or 90–99
Stage 2 hypertension ≥160 Or ≥100

Modified from The seventh report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of
High Blood Pressure. JAMA 2003;289:2560–2572.
10090-13_CH13.qxd 8/31/06 4:55 PM Page 314

314 Chapter Thirteen

Figure 13.3. Regulation of systemic blood pressure. The small arrows indicate whether there is a stimulatory (↑)
or inhibitory (↓) effect on the boxed parameters. ADH, antidiuretic hormone; CC, cardiac contractility; HR, heart rate;
NP, natriuretic peptides; PSNS, parasympathetic nervous system; SNS, sympathetic nervous system; SV, stroke volume;
VR, venous return.

pressure by sensing the stretch and deforma- signals from the aortic arch receptors are
tion of the arteries. If the arterial pressure carried by the vagus nerve (cranial nerve X).
rises, the baroreceptors are stimulated, in- These nerve fibers converge at the tractus
creasing their transmission of impulses to the solitarius in the medulla, where the barore-
central nervous system (i.e., the medulla). ceptor impulses inhibit sympathetic nervous
Negative feedback signals are then sent back system outflow and excite parasympathetic
to the circulation via the autonomic nervous effects. The net result is (1) a decline in pe-
system, causing the blood pressure to fall ripheral vascular resistance (i.e., vasodila-
back to its baseline level. tion) and (2) a reduction in cardiac output
The higher the blood pressure rises, the (because of a lower heart rate and reduced
more the baroreceptors are stretched and force of cardiac contraction). Each of these
the greater the impulse transmission rate to effects tends to lower arterial pressure back
the medulla. Signals from the carotid sinus toward its baseline. Conversely, when a fall
receptors are carried by the glossopharyn- in systemic pressure is sensed by the barore-
geal nerve (cranial nerve IX), whereas the ceptors, fewer impulses are transmitted to
10090-13_CH13.qxd 8/31/06 4:55 PM Page 315

Hypertension 315

the medulla, leading to a reflexive increase in Epidemiology and Genetics


blood pressure back to its baseline.
Heredity appears to play an important role
The main effect of the baroreceptor me-
in EH, but definite genetic markers have not
chanism is to modulate moment-by-moment
been consistently identified. It seems most
variations in systemic blood pressure. How-
likely that essential hypertension is a com-
ever, the baroreceptor reflex is not involved
plex genetic disorder, involving several loci
in the long-term regulation of blood pres-
interacting with environmental factors. Evi-
sure and does not prevent the development
dence for a genetic role is suggested by the
of chronic hypertension. The reason for
higher rate of elevated blood pressures among
this is that the baroreceptors constantly
first-degree relatives of hypertensive patients
reset themselves. After a day or two of expo-
than in the general population. Concordance
sure to higher-than-baseline pressures, the between identical twins is high and signifi-
baroreceptor-firing rate slows back to its con- cantly greater than that for dizygotic twins.
trol value. Furthermore, an uneven distribution of EH
exists among different racial groups. For ex-
ESSENTIAL HYPERTENSION ample, in most age distributions, blacks are
significantly more likely to be hypertensive
Almost 95% of hypertensive patients have than persons of other races.
blood pressures that are elevated for no Although no gene has been consistently
readily definable reason, a condition termed linked to essential hypertension, several loci
essential hypertension. The diagnosis of EH have demonstrated positive associations.
is one of exclusion; it is the option left to the For example, autosomal dominant con-
clinician after considering the causes of sec- tributors to elevated blood pressure have
ondary hypertension described later in this been discovered, usually involving defects
chapter. of renal sodium channels. However, these
EH is more a description than a diagnosis, abnormalities are rare and are thought to be
indicating only that a patient manifests a present in only a small fraction of hyper-
specific physical finding (high blood pres- tensive patients. Genes regulating the renin-
sure) for which no cause has been found. In angiotensin-aldosterone axis have been most AQ1
all likelihood, different underlying defects thoroughly studied in hypertensive patients
are responsible for the elevated pressure because of the central role of this system in
in different subpopulations of patients. Be- determining intravascular volume and vas-
cause the exact nature of these defects is un- cular tone. Within this group, certain poly-
known, to understand EH is to understand morphisms in the gene for angiotensinogen
the possibilities: what could go wrong with confer an increased risk of hypertension.
normal physiology to produce chronically Additionally, polymorphisms in the gene
elevated blood pressure? for adducin, a cytoskeletal protein, may be
This discussion of EH therefore reflects involved in a subgroup of EH patients. Fi-
what is currently known about its epidemi- nally, as described later in the chapter, sig-
ology and genetics, experimental findings, nificant associations exist between hyper-
and natural history. The picture that emerges tension and obesity, insulin resistance, and
is that EH likely results from multiple diabetes. The pathophysiologic and genetic
defects of blood pressure regulation that links among these four conditions are areas
interact with environmental stressors. The of very active investigation.
regulatory defects may be acquired or
genetically determined and may be inde- Experimental Findings
pendent of one another. As a result, EH pa-
System Abnormalities
tients exhibit varied combinations of ab-
normalities and therefore have various Multiple defects in blood pressure regulation
physiologic bases for their elevated blood have been found in EH patients and their rel-
pressures. atives. By themselves, or in conjunction with
10090-13_CH13.qxd 8/31/06 4:55 PM Page 316

316 Chapter Thirteen

one another, these abnormalities may con- which directly cause sodium retention; or
tribute to chronic blood pressure elevation. (3) inappropriate hormonal regulation. For
The heart can contribute to a high cardiac example, the renin-angiotensin-aldosterone
output–based hypertension owing to sympa- axis is an important hormonal regulator of
thetic overactivity. For example, when tested peripheral vascular resistance. Renin levels
under psychologically stressful conditions, in EH patients (compared with those in nor-
hypertensive patients (and their first-degree motensive persons) are subnormal in 30%,
relatives) often develop excessive heart rate normal in 60%, and high in 10%. Because
acceleration compared with control sub- renin secretion should be suppressed by high
jects, suggesting an excessive sympathetic blood pressure, even “normal” levels are in-
response. appropriate in hypertensive patients. Thus,
The blood vessels may contribute to pe- abnormalities of this system’s regulation
ripheral vascular resistance-based hyperten- may play a role in some persons with EH.
sion by constricting in response to (1) in- Figure 13.4 highlights these and other Fig. 4
creased sympathetic activity; (2) abnormal potential mechanisms of EH. Note that al-
regulation of vascular tone by local factors, though the heart, blood vessels, and kidneys
including nitric oxide, endothelin, and na- are the organs ultimately responsible for
triuretic factors; or (3) ion channel defects in producing the pressure, primary defects may
contractile vascular smooth muscle. be located elsewhere as well (e.g., the central
The kidney can induce volume-based hy- nervous system, arterial baroreceptors, and
pertension by retaining excessive sodium and adrenal hormone secretion). Yet, although
water as a result of (1) failure to regulate renal abnormal regulation at these sites can con-
blood flow appropriately; (2) ion channel de- tribute to elevated blood pressure, it is im-
fects (e.g., reduced basolateral Na+K+-ATPase), portant to remember that without renal

CNS
• ↑ Basal sympathetic tone
Blood vessel
• Abnormal stress response
Functional: • Abnormal response to signals from
• ↓ Nitric oxide secretion baroreceptors and volume receptors
• ↑ Endothelin production
• Ca2+ or Na+/K+ channel defects Pressure / volume receptors
• Hyperresponsiveness to • Desensitization
catecholamines
Structural:
• Exaggerated medial
hypertrophy

Adrenal Kidney
• Catecholamine leak • RAA dysfunction
or malregulation • Ion channel defects (e.g., Na+/ K+/ 2Cl –
cotransporter, basolateral Na+/ K+ ATPase,
Ca 2+ ATPase)

Figure 13.4. Potential primary abnormalities in essential hypertension. These defects are supported by experi-
mental evidence, but their specific contributions to essential hypertension remain unclear. CNS, central nervous sys-
tem; RAA, renin-angiotensin-aldosterone system.
10090-13_CH13.qxd 8/31/06 4:55 PM Page 317

Hypertension 317

complicity, malfunction of other systems more than 60% of Americans older than 60
would not produce sustained hypertension, years of age are hypertensive. In addition,
because the normal kidney is capable of the hemodynamic characteristics of blood
eliminating sufficient volume to return the pressure elevation in EH tend to change over
blood pressure to normal. time. Even in the absence of disease, systolic
pressures tend to increase throughout adult
life. Diastolic blood pressure, on the other
Insulin Resistance, Obesity, and the
hand, rises until the age of 50 and declines
Metabolic Syndrome
slightly from then on (see Fig. 13.2). Ac-
Recent research has suggested that the hor- cordingly, diastolic hypertension is more
mone insulin may play a role in the develop- common in young people, while a substan-
ment of EH. In many people with hyperten- tial number of hypertensive patients over
sion, especially those who are obese or have age 50 have isolated systolic hypertension
type 2 diabetes, there is impaired insulin- and normal diastolic values.
dependent transport of glucose into many In younger persons with hypertension,
tissues (termed insulin resistance). As a result, elevated blood pressure tends to be driven
serum glucose levels rise, stimulating the by high cardiac output in the setting of
pancreas to release additional insulin. Ele- relatively normal peripheral vascular resis-
vated insulin levels may contribute to hyper- tance, termed the hyperkinetic phase of EH
tension via increased sympathetic activation (Fig. 13.5). With advancing age, however, Fig. 5
or by stimulation of vascular smooth muscle the effect of cardiac output declines, perhaps
cell hypertrophy, which increases vascular because of the development of left ventricu-
resistance. Smooth muscle cell hypertrophy lar hypertrophy and its consequent reduced
may be caused by a direct mitogenic effect diastolic filling (which in turn reduces stroke
of insulin or through enhanced sensitivity volume and cardiac output). Conversely,
to platelet-derived growth factor, another vascular resistance increases with age owing
known smooth muscle cell growth factor. to medial hypertrophy as the vessels adapt to
Obesity itself has been directly associated the prolonged pressure stress. Thus, younger
with hypertension. Possible explanations for hypertensive patients often display aug-
this relationship include (1) the release of an- mented cardiac output as the principal ab-
giotensinogen from adipocytes as substrate normality, and older patients tend to have
for the renin-angiotensin system, (2) aug- elevated total peripheral resistance as the
mented blood volume related to increased major hemodynamic finding.
body mass, and (3) increased blood viscosity
caused by adipocyte release of profibrinogen In summary, EH is a syndrome that may
and plasminogen activator inhibitor 1. The arise from many potential abnormalities, but
current epidemic of obesity has led to a dra- it exhibits a characteristic hemodynamic pro-
matic increase in the number of people with file and natural history. It is likely that mul-
metabolic syndrome. As described in Chapter tiple defects, separately inherited or acquired,
5, this condition represents a clustering of act individually or together to chronically
atherogenic risk factors, including hyperten- raise blood pressure. Although we may not
sion, hypertriglyceridemia, low serum HDL, understand the precise underlying mecha-
a tendency toward glucose intolerance, and nisms in individual hypertensive patients,
truncal obesity. Current evidence suggests we can at least describe what kind of patho-
that insulin resistance is central to the patho- physiology might be at fault. EH, although
genesis of this clustering. idiopathic, is not entirely a “black box.”

Natural History SECONDARY HYPERTENSION


EH characteristically arises after young adult- Although EH dominates the clinical picture,
hood. Its prevalence increases with age and a defined structural or hormonal cause for
10090-13_CH13.qxd 8/31/06 4:55 PM Page 318

318 Chapter Thirteen

Figure 13.5. Hemodynamic progression of essential hypertension. Schematic representa-


tion of the changing contribution of cardiac output (CO) and total peripheral resistance (TPR) as
age increases in many patients with essential hypertension.

hypertension may be found in about 5% of pertension, there are clinical clues that a given
patients. Thus, cases of such secondary hy- patient may have one of the correctable con-
pertension are relatively uncommon, but ditions (Table 13.2):
their identification is important because the
underlying conditions are often curable and 1. Age. If a patient develops hypertension
may require therapy different from that ad- before age 20 or after age 50 (outside the
ministered for EH. Moreover, if secondary usual range of EH), secondary hyperten-
hypertension is left uncontrolled, adaptive sion is more likely.
cardiovascular changes may develop analo- 2. Severity. Secondary hypertension often
gous to those of long-standing EH that causes blood pressure to rise dramati-
could cause the elevated pressures to persist cally, whereas most EH patients usually
even after the underlying cause is corrected. have mild-to-moderate hypertension.
Although secondary forms should be con- 3. Onset. Secondary forms of hypertension
sidered in the workup of all patients with hy- often present abruptly in a patient who

TABLE 13.2. Causes of Hypertension

Percent of Hypertensive
Type Patients Clinical Clues

Essential 95% • Age of onset: 20–50 years


• Family history of hypertension
• Normal serum K+, urinalysis
Chronic renal disease 2–4% • ↑ Creatinine, abnormal urinalysis
Primary aldosteronism 1–2% • ↓ Serum K+
Renovascular 1% • Abdominal bruit
• Sudden onset (especially if age >50 or <20)
• ↓ Serum K+
Pheochromocytoma 0.2% • Paroxysms of palpitations, diaphoresis, and anxiety
• Episodic hypertension in one-third of patients
Coarctation of the aorta 0.1% • Blood pressure in arms > legs, or right arm > left arm
• Midsystolic murmur between scapulae
• CXR: aortic indentation, rib-notching due to
collaterals
Cushing syndrome 0.1% • “Cushingoid” appearance (e.g., central obesity,
hirsutism)
10090-13_CH13.qxd 8/31/06 4:55 PM Page 319

Hypertension 319

was previously normotensive, rather cholesterol, and triglyceride levels, as part of


than gradually progressing over years as the global vascular risk screen; and (5) an
is the usual case in EH. electrocardiogram (for evidence of left ven-
4. Associated signs and symptoms. The pro- tricular hypertrophy caused by chronic
cess that induces hypertension may give hypertension).
rise to other characteristic abnormalities, If no abnormalities are found that suggest
identified by the history and physical a secondary form of hypertension, the pa-
examination. For example, a renal arte- tient is presumed to have EH and treated ac-
ry bruit (swishing sound caused by tur- cordingly. If, however, the patient’s blood
bulent blood flow through a stenotic pressure continues to be elevated despite
artery) may be heard on abdominal ex- standard treatments, then more detailed di-
amination in a patient with renal artery agnostic testing is undertaken to search for
stenosis. specific secondary causes.
5. Family history. EH patients often have hy-
pertensive first-degree relatives, whereas Exogenous Causes
secondary hypertension more commonly Several medications can elevate blood pres-
occurs sporadically. sure. For example, oral contraceptives may
cause secondary hypertension in some wo-
Patient Evaluation men. The mechanism is likely related to in-
creased activity of the renin-angiotensin
The usual clinical evaluation of a patient system. Estrogens increase the hepatic syn-
with recently diagnosed hypertension be- thesis of angiotensinogen, leading to greater
gins with a careful history and physical ex- production of angiotensin II (Fig. 13.6). An- Fig. 6
amination, including a search for clues to giotensin II raises blood pressure by several
the secondary forms. For example, repeated mechanisms, most notably by direct vaso-
urinary tract infections may suggest the constriction and by stimulating the adrenal
presence of chronic pyelonephritis with release of aldosterone. The latter hormone
renal damage as the cause of hypertension. causes renal sodium retention and therefore
Excessive weight loss may be an indicator of increased intravascular volume.
pheochromocytoma, whereas weight gain Other medications that can raise blood
may point to the presence of Cushing syn- pressure include glucocorticoids, cyclospo-
drome. The history also should include an rine (an antirejection drug used in patients
assessment of lifestyle behaviors that may with organ transplants), erythropoietin (a
contribute to hypertension, such as exces- hormone that increases bone marrow red
sive alcohol consumption, and the patient’s blood cell formation and elevates blood
medications should be reviewed because pressure by increasing blood viscosity and
certain drugs (e.g., glucocorticoids and es- reversing local hypoxic vasodilatation), and
trogen) may elevate blood pressure. sympathomimetic drugs (which are com-
Laboratory tests commonly performed in mon in over-the-counter cold remedies).
the evaluation of the hypertensive patient, Two other substances that may contribute
including general screening for secondary to hypertension are cocaine and chronic ex-
causes, are (1) urinalysis and serum concen- cessive ethanol consumption. Both of these
tration of creatinine and blood urea nitro- are associated with increased sympathetic
gen to evaluate for renal abnormalities; nervous system activity.
(2) serum potassium level (abnormally low
in renovascular hypertension or aldostero-
Renal Causes
nism); (3) blood glucose level (elevated in
diabetes, which is strongly associated with Given the crucial role of the kidney in the
hypertension and renal disease); (4) serum control of blood pressure, it is not surprising
cholesterol, high-density lipoprotein (HDL) that renal dysfunction can lead to hyper-
10090-13_CH13.qxd 8/31/06 4:55 PM Page 320

320 Chapter Thirteen

Figure 13.6. The renin-angiotensin-aldosterone system. Liver-derived angiotensinogen is


cleaved in the circulation by renin (of kidney origin) to form angiotensin I (AI). AI is rapidly con-
verted to the potent vasoconstrictor angiotensin II (AII) by angiotensin-converting enzyme. AII also
modulates the release of aldosterone from the adrenal cortex. Aldosterone in turn acts to reab-
sorb Na+ from the distal nephron, resulting in increased intravascular volume. The other listed
effects of AII receptor stimulation may also contribute to the development and maintenance of
hypertension.

tension. In fact, renal disease contributes to intravascular volume. Damaged nephrons


two leading endogenous causes of secondary are unable to excrete normal amounts of
hypertension: renal parenchymal disease, sodium and water, leading to a rise in in-
accounting for 2% to 4% of hypertensive pa- travascular volume, elevated cardiac output,
tients, and renal arterial stenosis, which ac- and hence increased blood pressure.
counts for approximately 1%. If renal function is only mildly impaired,
blood pressure may stabilize at a level at
which the higher systemic pressure (and
Renal Parenchymal Disease
therefore renal perfusion pressure) enables
Parenchymal damage to the kidney can re- sodium excretion to balance sodium intake.
sult from diverse pathologic processes. The Conversely, if a patient has end-stage renal
major mechanism by which injury leads to failure, the glomerular filtration rate may be
elevated blood pressure is through increased so greatly decreased that the kidney simply
10090-13_CH13.qxd 8/31/06 4:55 PM Page 321

Hypertension 321

cannot excrete sufficient volume, and malig- by impeding the formation of angiotensin
nant-range blood pressures may follow. Note II (see Chapter 17).
that renal parenchymal disease may con-
tribute to hypertension even if the glome- Mechanical Causes
rular filtration rate is not greatly reduced,
through the excessive elaboration of renin. Coarctation of the Aorta
Coarctation is an infrequent congenital nar-
Renovascular Hypertension rowing of the aorta typically located just dis-
tal to the origin of the left subclavian artery
Stenosis of one or both renal arteries leads to (see Chapter 16). As a result of the relative
hypertension. Although emboli, vasculitis, obstruction to flow, the blood pressure in
and external compression of the renal arte- the aortic arch, head, and arms is higher than
ries can be responsible, the two most com- that in the descending aorta and its branches
mon causes of renovascular hypertension and in the lower extremities. Sometimes the
(RH) are atherosclerosis and fibromuscular coarctation involves the origin of the left
dysplasia. Atherosclerotic lesions arise from subclavian artery, causing lower pressure in
extensive plaque formation either within the left arm than in the right.
the renal artery or in the aorta at the origin Hypertension in this condition arises by
of the renal artery. This form accounts for two mechanisms. First, reduced blood flow to
about two thirds of cases of RH and occurs the kidneys stimulates the renin-angiotensin
most commonly in elderly men. In contrast, system, resulting in vasoconstriction (via an-
fibromuscular lesions consist of discrete re- giotensin II). Second, high pressures proxi-
gions of fibrous or muscular proliferation, mal to the coarctation stiffen the aortic arch
generally within the arterial media. Fibro- through medial hyperplasia and accelerated
muscular dysplasia accounts for one third of atherosclerosis, blunting the normal baro-
cases of RH and characteristically occurs in receptor response to elevated intravascular
young women. pressure.
The elevated blood pressure in RH arises Clinical clues to the presence of coarcta-
from reduced renal blood flow to the af- tion include symptoms of inadequate blood
fected kidney, which responds to the lower flow to the legs or left arm, such as claudi-
perfusion pressure by secreting renin. The cation or fatigue, or the finding of weakened
latter raises the blood pressure through the or absent femoral pulses. A midsystolic mur-
subsequent actions of angiotensin II (vaso- mur associated with the stenotic segment of
constriction) and aldosterone (sodium re- the aorta may be auscultated, especially over
tention), as shown in Figure 13.6. the back, between the scapulae. The chest
The diagnosis of RH is suggested by an radiograph may show indentation of the
abdominal bruit, which can be identified aorta at the level of the coarctation. It may
in 40% to 60% of patients, or by the pres- also demonstrate a notched appearance of
ence of unexplained hypokalemia (owing the ribs secondary to the enlargement of col-
to excessive renal excretion of potassium as lateral intercostal arteries, which shunt blood
a result of elevated aldosterone levels). RH around the aortic narrowing. Treatment op-
is a correctable form of hypertension that tions include angioplasty or surgery to cor-
often is treated successfully by percutaneous rect the stenosis. However, the hypertension
catheter interventions or surgical recon- may not abate completely after mechanical
struction of the stenosed vessel. Medical correction, possibly because of persistent de-
therapy, particularly with angiotensin- sensitization of the arterial baroreceptors.
converting enzyme (ACE) inhibitors, can
also be effective initial therapy in patients
Endocrine Causes
with unilateral renal artery disease. ACE in-
hibitors negate the hypertensive effects of Circulating hormones play an important
elevated circulating renin in this situation role in the control of normal blood pressure,
10090-13_CH13.qxd 8/31/06 4:55 PM Page 322

322 Chapter Thirteen

so it should not be surprising that endocrine as well as drugs that inhibit catecholamine
diseases may cause hypertension. When biosynthesis (e.g., α-methyltyrosine).
suspected, the presence of such endocrine
conditions is evaluated in four ways:
Adrenocortical Hormone Excess
1. History of characteristic signs and symp-
Among the hormones produced by the ad-
toms
renal cortex are mineralocorticoids and
2. Measurement of hormone levels glucocorticoids. Excess of either of these can
3. Assessment of hormone secretion in res- result in hypertension.
ponse to stimulation or inhibition Mineralocorticoids, primarily aldos-
4. Imaging studies to identify tumors sec- terone, increase blood volume by stimulat-
reting the excessive hormone ing reabsorption of sodium into the circula-
tion by the distal portions of the nephron.
This occurs in exchange for potassium ex-
Pheochromocytoma
cretion into the urine, and the resulting hy-
Pheochromocytomas are catecholamine- pokalemia is an important marker of miner-
secreting tumors of neuroendocrine cells alocorticoid excess. Primary aldosteronism,
(usually in the adrenal medulla) that cause found in approximately 1% to 2% of hyper-
approximately 0.2% of cases of hyperten- tensive patients, is generally the result of an
sion. The release of epinephrine and norepi- adrenal adenoma (termed Conn syndrome),
nephrine by the tumor results in intermittent but may also result from bilateral hyperpla-
or chronic vasoconstriction, tachycardia, sia of the adrenal glands. Because the disease
and other sympathetic-mediated effects. may be asymptomatic, diagnosis relies on
A characteristic presentation consists of the detection of hypokalemia and is con-
paroxysmal rises in blood pressure accom- firmed by measurement of excessive aldos-
panied by “autonomic attacks” caused by terone secretion and suppressed plasma
the increased catecholamine levels: severe renin levels. Therapy includes either surgical
throbbing headaches, profuse sweating, pal- removal of the adenoma or medical man-
pitations, and tachycardia. Although some agement with aldosterone receptor antago-
patients are actually normotensive between nists (e.g., spironolactone). Glucocorticoid-
attacks, most have sustained hypertension. remediable aldosteronism (GRA), an uncom-
Ten percent of pheochromocytomas are mon hereditary (autosomal dominant) form
malignant. of primary aldosteronism, results from a ge-
Determination of plasma catecholamine netic rearrangement in which aldosterone
levels, or urine catecholamines and their synthesis abnormally comes under the reg-
metabolites (e.g., vanillylmandelic acid and ulatory control of adrenocorticotropic hor-
metanephrine), obtained under controlled mone (ACTH). This condition typically pre-
circumstances, are used to identify this con- sents as severe hypertension in childhood or
dition. Because some pheochromocytomas young adulthood, as opposed to more com-
secrete only episodically, diagnosis may re- mon forms of primary aldosteronism, which
quire measurement of catecholamines im- are generally diagnosed in the third through
mediately following an attack. sixth decades. Unlike other forms of hyper-
Pharmacologic therapy of pheochromo- tension, GRA-related blood pressure ele-
cytomas includes the combination of an α- vation responds to glucocorticoid therapy,
receptor blocker (e.g., phenoxybenzamine) which suppresses ACTH release from the pi-
combined with a β-blocker. However, once tuitary gland.
the tumor is localized by computed tomog- Secondary aldosteronism can result from
raphy, magnetic resonance imaging, or an- increased angiotensin II production stimu-
giography, the definitive therapy is surgical lated by the rare renin-secreting tumor. More
resection. For patients with inoperable dis- commonly, secondary elevation of aldos-
ease, treatment consists of α- and β-blockade terone is a result of augmented circulating
10090-13_CH13.qxd 8/31/06 4:55 PM Page 323

Hypertension 323

angiotensin II in women taking oral contra- sympathetic and adrenal activation in hy-
ceptives (which stimulate hepatic produc- pothyroidism.
tion of angiotensinogen, as described ear-
lier) or because of impaired angiotensin II
CONSEQUENCES OF
degradation in chronic liver diseases.
HYPERTENSION
Glucocorticoids, such as cortisol, elevate
blood pressure when present in excess Whatever the cause of blood pressure eleva-
amounts, likely via blood volume expansion tion, the ultimate consequences are similar.
and stimulated synthesis of components of High blood pressure itself is generally asymp-
the renin-angiotensin system. In addition, tomatic but can result in devastating effects
though mineralocorticoids are more potent on many organs, especially the blood vessels,
activators of mineralocorticoid receptors in heart, kidney, and retina.
the renal tubules, excess glucocorticoids may
also activate them.
Clinical Signs and Symptoms
Nearly 80% of patients with Cushing syn-
drome, a disorder of glucocorticoid excess, In the past, “classic” symptoms of hyper-
have some degree of hypertension. These pa- tension were considered to include head-
tients often present with classic “cushin- ache, epistaxis (nose bleeds), and dizziness.
goid” features: a characteristic rounded facial The usefulness of these symptoms has been
appearance, central obesity, proximal mus- called into question, however, by studies
cle weakness, and hirsutism. The cause of the that indicate that they are found no more
excess glucocorticoids may be either a pitu- frequently among hypertensive patients
itary ACTH–secreting adenoma, a peripheral than in the general population. Other symp-
ACTH–secreting tumor (either of which causes toms, such as flushing, sweating, and blurred
adrenal cortical hyperplasia), or an adrenal vision, do seem more common in the hy-
cortisol–secreting adenoma. The diagnosis of pertensive population. In general, however,
Cushing syndrome is made by a 24-hour most hypertensive patients are asympto-
urine collection for the measurement of matic and are diagnosed simply by blood
cortisol, or by a dexamethasone test, which pressure measurement during routine phys-
evaluates whether exogenous glucocorti- ical examinations.
coids can suppress cortisol secretion. Several physical signs of hypertension
discussed in the following sections directly
result from elevated pressure, including left
Thyroid Hormone Abnormalities
ventricular hypertrophy and retinopathy. In
Approximately one third of hyperthyroid and addition, hypertension complicated by ather-
one fourth of hypothyroid patients have sig- osclerosis can manifest by arterial bruits, par-
nificant hypertension. Thyroid hormones ticularly in the carotid and femoral arteries.
exert their cardiovascular effects by (1) in-
ducing sodium-potassium ATPases in the
heart and vessels; (2) increasing blood vol- Organ Damage Caused
ume; and (3) stimulating tissue metabolism by Hypertension
and oxygen demand, with secondary accu- Target organ complications of hypertension
mulation of metabolites that modulate local reflect the degree of chronic blood pressure
vascular tone. Hyperthyroid patients de- elevation. Such organ damage can be attrib-
velop hypertension through cardiac hyper- uted to (1) the increased workload of the
activity and an increase in blood volume. heart and (2) arterial damage resulting from
Hypothyroid patients demonstrate predom- the combined effects of the elevated pres-
inantly diastolic hypertension and an sure itself (weakened vessel walls) and accel-
increase in peripheral vascular resistance. erated atherosclerosis (Fig. 13.7). Abnormal- Fig. 7
Though the precise mechanism is unclear, ities of the vasculature that result from
the latter effect appears to be mediated by elevated pressure include smooth muscle
10090-13_CH13.qxd 8/31/06 4:55 PM Page 324

324 Chapter Thirteen

Figure 13.7. Pathophysiology of the major consequences of hypertension. LVH, left ventricular hypertrophy.

hypertrophy, endothelial cell dysfunction, the heart must contract and accelerated ath-
and fatigue of elastic fibers. Chronic hyper- erosclerosis within the coronary arteries.
tensive trauma to the endothelium pro-
motes atherosclerosis possibly by disrupting
normal protective mechanisms, such as the
Left Ventricular Hypertrophy and
secretion of nitric oxide. Arteries lined by Diastolic Dysfunction
atherosclerotic plaque may thrombose or The high arterial pressure (heightened after-
may serve as a source of cholesterol emboli load) increases the wall tension of the left
that occlude distal vessels, causing organ in- ventricle, which compensates through hyper-
farction (such as cerebrovascular occlusion, trophy. Concentric hypertrophy (without di-
resulting in stroke). In addition, atheroscle- latation) is the normal pattern of compen-
rosis of large arteries hinders their elasticity,
resulting in systolic pressure spikes that can
further traumatize endothelium or provoke
events such as aneurysm rupture. TABLE 13.3. Target Organ Damage in
Hypertension
The major target organs for the destruc-
tive complications of chronic hypertension Organ System Manifestations
are the heart, the cerebrovascular system,
Heart • Left ventricular hyper-
the aorta and peripheral vascular system,
trophy
Tab. 13 the kidney, and the retina (Table 13.3). Left • Heart failure
untreated, approximately 50% of hyperten- • Myocardial ischemia and
sive patients die of coronary artery disease infarction
or congestive heart failure, about 33% suc- Cerebrovascular • Stroke
cumb to stroke, and 10% to 15% die from Aorta and peripheral • Aortic aneurysm and/or
vascular dissection
complications of renal failure. • Arteriosclerosis
Kidney • Nephrosclerosis
• Renal failure
Heart Retina • Arterial narrowing
The major cardiac effects of hypertension re- • Hemorrhages, exudates,
papilledema
late to the increased afterload against which
10090-13_CH13.qxd 8/31/06 4:55 PM Page 325

Hypertension 325

sation, although conditions that elevate myocardial infarction complications such as


blood pressure by virtue of increased circu- rupture of the ventricular wall, LV aneurysm
lating volume (e.g., primary aldosteronism) formation, and congestive heart failure. In
may instead cause eccentric hypertrophy fact, 60% of patients who die of transmural
with chamber dilatation (see Chapter 9). myocardial infarctions have a history of hy-
Left ventricular hypertrophy (LVH) results pertension.
in increased stiffness of the left ventricle
with diastolic dysfunction, manifest by ele-
Cerebrovascular System
vation of LV filling pressure that can result
in pulmonary congestion. Hypertension is the major modifiable risk
Physical findings of LVH may include factor for strokes, also termed cerebrovas-
a heaving LV impulse on chest palpation, cular accidents (CVAs). Although diastolic
indicative of increased muscle mass. It is pressure is important, it is the magnitude of
frequently accompanied by a fourth heart the systolic pressure that has been most
sound, as the left atrium contracts into the closely linked to CVAs. The presence of iso-
stiffened left ventricle (see Chapter 2). lated systolic hypertension more than dou-
LVH is one of the strongest predictors of bles a person’s risk for this complication.
cardiac morbidity in hypertensive patients. Hypertension-induced strokes can be he-
The degree of hypertrophy correlates with morrhagic or, more commonly, athero-
the development of congestive heart failure, thrombotic. Hemorrhagic CVAs result from
angina, arrhythmias, myocardial infarction, rupture of microaneurysms induced in cere-
and sudden cardiac death. bral parenchymal vessels by long-standing
hypertension. Atherothrombotic (also called
thromboembolic) CVAs arise when portions
Systolic Dysfunction
of atherosclerotic plaque within the carotids
Although LVH initially serves a compen- or major cerebral arteries, or thrombi that
satory role, later in the course of systemic form on those plaques, break off and em-
hypertension, the increased LV mass may be bolize to smaller distal vessels. Additionally,
insufficient to balance the high wall tension intracerebral vessels may directly occlude
caused by the elevated pressure. As LV con- by local atherosclerotic plaque rupture and
tractile capacity deteriorates, findings of sys- thrombosis.
tolic dysfunction become evident (i.e., re- Occlusion of small penetrating brain ar-
duced cardiac output and pulmonary teries can result in multiple tiny infarcts. As
congestion). Systolic dysfunction is also pro- these lesions soften and are absorbed by
voked by the accelerated development of phagocytic cells, small (≤3 mm diameter)
coronary artery disease with resultant peri- cavities form, termed lacunae. These lacunar
ods of myocardial ischemia. infarctions are seen almost exclusively in
patients with long-standing hypertension
and are usually localized to the penetrating
Coronary Artery Disease
branches of the middle and posterior circu-
Chronic hypertension is a major contribu- lation of the brain.
tor to the development of myocardial is- The generalized arterial narrowing found
chemia and infarction. These complications in hypertensive patients reduces collateral
reflect the combination of accelerated coro- flow to ischemic tissues and imposes struc-
nary atherosclerosis (decreased myocardial tural requirements for higher perfusion
oxygen supply) and the high systolic work- pressure to maintain adequate tissue flow.
load (increased oxygen demand). Not only This leaves the hypertensive patient vulner-
is acute myocardial infarction more com- able to cerebral infarcts in areas supplied by
mon among hypertensive patients than the distal ends of arterial branches (“water-
among normotensive people, but also the shed” infarcts) if blood pressure should fall
former also have a higher incidence of post- suddenly.
10090-13_CH13.qxd 8/31/06 4:55 PM Page 326

326 Chapter Thirteen

Effective treatment of hypertension di- come thickened with a hyaline infiltrate,


minishes the risk of stroke and has con- known as hyaline arteriolosclerosis (Fig. Fig. 8
tributed to a 50% reduction in deaths at- 13.8). Greater levels of hypertension can in-
tributed to cerebrovascular events in recent duce smooth muscle hypertrophy and even
decades. necrosis of capillary walls, termed fibrinoid
necrosis. These changes result in reduced
vascular supply and subsequent ischemic at-
Aorta and Peripheral Vasculature
rophy of tubules and, to a lesser extent,
The accelerated atherosclerosis associated glomeruli. Because intact nephrons can
with hypertension may result in plaque for- usually compensate for those damaged by
mation and narrowing throughout the arte- patchy ischemia, mild hypertension rarely
rial vasculature. In addition to the coronary leads to renal insufficiency in the absence of
arteries, lesions most commonly appear with- other insults to the kidney. However, malig-
in the aorta and the major arteries to the nant levels of hypertension can inflict per-
lower extremities, neck, and brain. manent damage to the point that dialysis
Chronic hypertension may lead to the becomes necessary.
development of aneurysms, particularly of One of the consequences of hypertensive
the abdominal aorta (see Chapter 15). An renal failure is perpetuation of elevated blood
abdominal aortic aneurysm (AAA) repre- pressure. For example, progressive renal dys-
sents prominent dilatation of the vessel, function compromises the ability of the kid-
usually located below the level of the renal ney to regulate blood volume, which con-
arteries, aided by the mechanical stress of the tributes further to chronic hypertension.
high pressure on an arterial wall already
weakened by medial damage and atheroscle-
Retina
rosis. Aneurysms greater than 6 cm in diam-
eter have a very high likelihood of rupture The retina is the only location where sys-
within 2 years if not surgically corrected. temic arteries can be directly visualized
Another life-threatening vascular conse- by physical examination. High blood pres-
quence of high blood pressure is aortic dis- sure induces abnormalities that are collec-
section (see Chapter 15). Elevated blood tively termed hypertensive retinopathy.
pressure, especially in the highest ranges, ac- Although vision may be compromised
celerates degenerative changes in the media when the damage is extensive, more com-
of the aorta. When the weakened wall is fur- monly the changes serve as an asympto-
ther exposed to high pressure, the intima matic clinical marker for the severity of hy-
may tear, allowing blood to dissect into the pertension and its duration.
aortic media and propagate in either direc- Severe hypertension that is acute in onset
tion within the vessel wall, “clipping off” (e.g., uncontrolled and/or malignant hyper-
and obstructing major branch vessels along tension) may burst small retinal vessels,
the way (e.g., coronary or carotid arteries). causing hemorrhages, exudation of plasma
The mortality rate of aortic dissection is lipids, and areas of local infarction. If ische-
greater than 90% unless treated emergently, mia of the optic nerve develops, patients
usually by surgical repair if the proximal may describe generalized blurred vision.
aorta is involved. Rigorous control of hyper- Retinal ischemia caused by hemorrhage
tension is essential. leads to more patchy loss of vision. Papil-
ledema, or swelling of the optic disc with
blurring of its margins, may arise from high
Kidney
intracranial pressure when the blood pres-
Hypertension-induced kidney disease (neph- sure reaches malignant levels and cerebro-
rosclerosis) is a leading cause of renal failure vascular autoregulation begins to fail.
that results from damage to the organ’s vas- Chronically elevated blood pressure results
culature. Histologically, the vessel walls be- in a different set of retinal findings. Pa-
10090-13_CH13.qxd 8/31/06 4:55 PM Page 327

Hypertension 327

Figure 13.8. Histologic effects of chronic hypertension on the kidney.


The arteriolar walls are thickened by hyaline infiltrate (short arrows). The
glomeruli (long arrow) appear partially sclerosed because of reduced vascu-
lar supply. (Courtesy of Dr. Helmut G. Rennke, Brigham and Women’s Hos-
pital, Boston, MA.)

pilledema is absent, but vasoconstriction sive crisis is more often caused by an acute
results in arterial narrowing, and medial hy- hemodynamic insult (e.g., acute renal dis-
pertrophy thickens the vessel wall, which ease) superimposed on a chronic hyperten-
“nicks” (indents) crossing veins. With more sive state. As a result of rapid pathologic
severe chronic hypertension, arterial sclero- changes (fibrinoid necrosis) within the blood
sis is evident as an increased reflection of vessels and kidney, a spiraling increase in
light through the ophthalmoscope (termed blood pressure evolves. Further volume ex-
“copper” or “silver” wiring. Although these pansion and vasoconstriction occur as renal
changes are not in themselves of major perfusion drops and serum renin and an-
functional importance, they indicate that giotensin levels rise.
the patient has had long-standing, poorly Severe blood pressure elevation results in
controlled hypertension. increased intracranial pressure, and patients
may present with hypertensive encepha-
lopathy manifested by headache, blurred
HYPERTENSIVE CRISIS
vision, confusion, somnolence, and some-
A hypertensive crisis is a medical emergency times coma. When hypertension results in
characterized by a severe elevation of blood acute damage to retinal vessels, accelerated-
pressure. In the past, this type of elevation malignant hypertension is said to be pre-
was usually a consequence of inadequate sent. Funduscopic examination shows the
blood pressure treatment. Now a hyperten- effects of the rapid pressure rise as hemor-
10090-13_CH13.qxd 8/31/06 4:55 PM Page 328

328 Chapter Thirteen

rhages, exudates, and sometimes papilledema. factors, a more aggressive approach to phar-
The increased load on the left ventricle dur- macologic therapy is usually warranted to
ing a hypertensive crisis may precipitate reduce the risk burden.
angina (because of increased myocardial oxy- For most hypertensive patients, drug the-
gen demand) or pulmonary edema. rapy is ultimately the most effective way
A hypertensive crisis requires rapid ther- to prevent future complications, but that
apy to reduce blood pressure and prevent should not deter consideration of other ben-
permanent vascular complications. Correc- eficial lifestyle changes.
tion of blood pressure is generally followed
by reversal of the acute pathologic changes, Nonpharmacologic Treatment
including papilledema and retinal exuda-
tion, although renal damage often persists. Weight Reduction
Studies have consistently found obesity and
TREATMENT OF HYPERTENSION hypertension to be highly correlated, espe-
cially when the obesity is of a central (ab-
The therapeutic approach to the hyperten- dominal) distribution. Blood pressure reduc-
sive patient should be tempered by two con- tion follows weight loss in a large portion of
siderations. First, a single elevated blood pres- hypertensive patients who are more than
sure measurement does not establish the 10% above their ideal weights. Each 10 kg of
diagnosis of hypertension because blood weight loss is associated with a 5 to 20 mm
pressure varies considerably from day to day. Hg fall in systolic blood pressure.
Moreover, blood pressure measurement in
the hospital or doctor’s office may be affected
by the “white coat” effect resulting from pa- Exercise
tient anxiety. The average of multiple read- Sedentary normotensive people have a 20%
ings taken at two or three office visits and/or to 50% higher risk of developing hyperten-
in the home provides a more reliable basis for sion than their more active peers. Regular
labeling a patient as hypertensive. There is aerobic exercise, such as walking, jogging, or
also evidence that automatic ambulatory bicycling, has been shown to contribute to
blood pressure measurements, taken over the blood pressure reduction over and above
course of 24 hours while the patient follows any resulting weight loss. A hypertensive pa-
a daily routine, are more predictive of cardio- tient who becomes physically conditioned
vascular mortality than traditional in-clinic manifests a lower resting heart rate and re-
measurements. duced levels of circulating catecholamines
Second, although even mild hyperten- than before training, suggesting a fall in sym-
sion is a major public health problem be- pathetic tone.
cause of its widespread prevalence, for the
person with stage 1 hypertension (see Table
Diet
13.1), the risks are small. For example, the
additional risk of a stroke is approximately 1 In addition to caloric restriction for weight
in 850 per year. Hence, observation over loss, changes in the composition of a pa-
time to determine whether the low-level tient’s diet may be important for blood pres-
hypertension persists, or whether lifestyle sure reduction. For example, a diet high in
changes can reduce the pressure, is often fruits, vegetables, and low-fat dairy products
a recommended alternative to immediate has been shown to significantly reduce blood
drug therapy. This is especially true in the pressure.
absence of other cardiovascular risk factors
such as smoking, diabetes, or high serum
Sodium
cholesterol. However, for patients with es-
tablished cardiovascular disease or for those Salt restriction for people with high blood
who have other major atherosclerotic risk pressure is a controversial issue, but there
10090-13_CH13.qxd 8/31/06 4:55 PM Page 329

Hypertension 329

are several epidemiologic and clinical trials unclear. Caffeine ingestion transiently in-
that support the benefit of moderating so- creases blood pressure (as much as 5 to 15
dium intake. In normotensive persons, ex- mm Hg after two cups of coffee), but routine
cess salt ingestion is simply excreted by the use does not seem to produce chronic pres-
kidneys, but approximately 50% of patients sure elevation.
with essential hypertension are found to
have blood pressures that vary with sodium
Smoking
intake, suggesting a defect in natriuresis.
Sensitivity to sodium levels is more com- Cigarette smoking transiently increases blood
mon in African American and elderly hy- pressure, likely because of the effect of nico-
pertensive patients. Because low-salt diets tine on the autonomic ganglia, and is a risk
tend to increase the effectiveness of antihy- factor for the development of sustained hy-
pertensive medications in general, the cur- pertension. In addition, the atherogenic
rent recommendation is to limit salt intake effect of smoking may contribute to the de-
to <6 g of sodium chloride (<2.3 g sodium) velopment of renovascular hypertension.
per day, which is one-third less than the Cigarette usage is associated with many other
average U.S. consumption. health hazards, and all patients should be
discouraged from smoking.
Potassium
Relaxation Therapy
Total body potassium content tends to de-
crease when a person eats a diet low in fruits Blood pressure frequently rises under condi-
and vegetables or takes potassium-wasting tions of stress. In addition, essential hyper-
diuretics. Potassium deficiency has several tensive patients and their relatives often
theoretical effects that may raise blood pres- show higher than normal basal sympathetic
sure and contribute to adverse cardiovascu- tone and exaggerated autonomic responses
lar outcomes, such that dietary supplements to mental stress. Hence, relaxation tech-
are routinely recommended to help replete niques have been advocated as a method to
low serum K+ levels. There is no convincing control hypertension. Available methods in-
evidence that prescribing potassium supple- clude biofeedback and meditation. The ef-
ments to a normokalemic hypertensive pa- fectiveness of such therapy has not been
tient will lower blood pressure. consistently demonstrated in clinical trials
and seems to depend on the patient’s atti-
tude and long-term compliance.
Alcohol
The chronic intake of alcoholic beverages In summary, nonpharmacologic therapy
correlates with high blood pressure and re- offers a wide range of options that do not
sistance to antihypertensive medications. have the expense and potential side effects of
Moreover, experimental evidence shows that prescribed drug use. The effectiveness of
blood pressure (especially systolic) may rise these therapies should come as no surprise,
acutely following alcohol consumption. The given the extent to which environmental fac-
reason for this link remains incompletely un- tors play a role in hypertension. Therefore,
derstood, but decreasing chronic alcohol in- behavior-based interventions are recom-
take has been shown to lower blood pressure. mended as first-line therapy in any patient
whose hypertension is not an immediate
danger to life and well-being.
Other
Low calcium intake and magnesium deple-
Pharmacologic Treatment
tion have been associated with elevated
blood pressure, but the responsible mecha- Antihypertensive medications are the stan-
nisms and the implications for therapy are dard means to lower chronically elevated
10090-13_CH13.qxd 8/31/06 4:55 PM Page 330

330 Chapter Thirteen

blood pressure and are indicated if nonphar- glucose, cholesterol, and triglyceride levels.
macologic treatment proves inadequate. More In addition, hypokalemia, hyperuricemia,
than 100 drug preparations are available to and decreased sexual function are potential
treat hypertension, but fortunately the most side effects. However, when diuretics are
commonly used medications fall into four prescribed in low dosages, it is often possible
classes: diuretics, sympatholytics, vasodila- to accrue the desired antihypertensive effect
tors, and drugs that interfere with the renin- while minimizing adverse complications.
Tab. 4 angiotensin system (Table 13.4). The individ- Sympatholytic agents include (1) β-
ual actions of these groups on the physiologic blockers, (2) central α-adrenergic agonists,
abnormalities in hypertension are shown and (3) systemic α-adrenergic-blocking drugs.
Fig. 9 in Figure 13.9. The pharmacology and use of b-Blockers, such as propranolol, are be-
the antihypertensive drugs are described in lieved to lower blood pressure through sev-
greater detail in Chapter 17. eral mechanisms, including (1) reducing
Diuretics have been used for decades to cardiac output through a decrease in heart
treat hypertension. They reduce circulatory rate and mild decrease in contractility and
volume, cardiac output, and mean arterial (2) decreasing the secretion of renin (and
pressure and are most effective in patients therefore levels of angiotensin II), which
with mild-to-moderate hypertension who leads to a decrease in total peripheral resis-
have normal renal function. They are espe- tance. β-Blockers are less effective than di-
cially effective in African American or elderly uretics in elderly and African American
persons, who tend to be salt sensitive. In clin- hypertensive patients. Adverse effects of
ical trials, diuretics have reduced the risk of β-blockers include bronchospasm (because
strokes and cardiovascular events in hyper- of bronchiolar β2-receptor blockade), fa-
tensive patients and are inexpensive com- tigue, impotence, and hyperglycemia. They
pared with other agents. Thiazide diuretics may also adversely alter lipid metabolism.
(e.g., hydrochlorothiazide) and potassium- Most β-blockers cause an increase in serum
sparing diuretics (e.g., spironolactone) pro- triglyceride levels and a decrease in “good”
mote Na+ and Cl− excretion in the nephron. HDL cholesterol levels. However, β-blockers
Loop diuretics (e.g., furosemide) are gener- with intrinsic sympathomimetic activity
ally too potent and their actions too short (see Chapter 17) or those with combined
lived for use as antihypertensive agents; β-blocking properties (such as labetalol) do
however, they are useful in lowering blood not adversely affect HDL levels.
pressure in patients with renal insufficiency, Centrally acting a2-adrenergic agonists,
who often do not respond to other diuretics. such as methyldopa and clonidine, reduce
Diuretics may result in adverse metabolic sympathetic outflow to the heart, blood ves-
side effects, including elevation of serum sels, and kidneys. These are rarely used owing

TABLE 13.4. Classes of Antihypertensive Medications

Drug Class Types (see Chapter 17)

Diuretics Thiazides
Aldosterone-antagonists
Sympatholytics β-Blockers
Combined α- and β-blockers
Central α2-agonists
Peripheral α1-blockers
Vasodilators Calcium channel blockers
Direct vasodilators (e.g., hydralazine, minoxidil)
Renin-angiotensin system antagonists Angiotensin-converting enzyme inhibitors
Angiotensin II receptor blockers
10090-13_CH13.qxd 8/31/06 4:55 PM Page 331

Hypertension 331

Figure 13.9. Physiologic effects of antihypertensive medications. Notice that some antihypertensive medications
work at multiple sites. CC, cardiac contractility; CCB, calcium++ channel blockers; HR, heart rate; RAS blockers, renin-
angiotensin system blockers (i.e., angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers); SV,
stroke volume; VR, venous return.

to their high frequency of side effects (e.g., reducing cardiac contractility and total pe-
dry mouth, sedation). Systemic a1-antago- ripheral resistance (see Chapter 17). Clinical
nists, such as prazosin, terazosin, and doxa- trials in patients with hypertension have
zosin, cause a decrease in total peripheral shown that calcium channel blockers re-
resistance through relaxation of vascular duce the risk of myocardial infarction and
smooth muscle. They may be beneficial for stroke. Thus long-acting (i.e., sustained-
hypertension in some older men because the release drugs taken once a day) members of
drugs also improve symptoms of prostatic en- this group are frequently used to treat
largement. However, they are otherwise not hypertension. The shorter-acting calcium
often recommended because a major clinical channel blocker preparations are no longer
trial showed that an α-blocker was associated used for this purpose; they are less conve-
with a greater number of adverse cardio- nient and have been associated with adverse
vascular events compared with a diuretic. cardiovascular outcomes (see Chapter 6).
Peripheral vasodilators include calcium Hydralazine and minoxidil lower blood
channel blockers, hydralazine, and minoxi- pressure by directly relaxing vascular smooth
dil. Calcium channel blockers reduce the muscle of precapillary resistance vessels.
influx of Ca++ responsible for cardiac and However, this action can result in a reflex
vascular smooth muscle contraction, thus increase in heart rate, so that combined
10090-13_CH13.qxd 8/31/06 4:55 PM Page 332

332 Chapter Thirteen

β-blocker therapy is frequently necessary. which drug to use as initial therapy in an in-
The use of these direct vasodilators in treat- dividual patient can seem daunting. The
ing hypertension has waned with the ad- first-line drugs for uncomplicated hyperten-
vent of newer agents with fewer side effects. sion recommended by the Joint National
Drugs that interfere with the renin- Committee on Detection, Evaluation, and
angiotensin system include ACE inhibitors Treatment of High Blood Pressure are thi-
and angiotensin II receptor blockers. ACE in- azide diuretics because of their proven long-
hibitors decrease blood pressure by blocking term benefits at reducing morbidity and
the conversion of angiotensin I to angio- mortality as well as their low cost. In certain
tensin II (see Fig. 17.6), thereby reducing the circumstances, or if initial therapy with a di-
vasopressor activity of angiotensin II and the uretic is not sufficient, an ACE inhibitor, an-
secretion of aldosterone. Thus, reductions in giotensin receptor blocker, calcium channel
total peripheral resistance and sodium reten- blocker, or β-blocker is substituted or added.
tion by the kidney occur. An additional anti- For example, an ACE inhibitor should be
hypertensive effect of ACE inhibitors is to in- given prime consideration in patients with
crease the concentration of the circulating concurrent heart failure, diabetes, or LV
vasodilator bradykinin (see Fig. 17.6). ACE dysfunction following myocardial infarc-
inhibitors are important drugs that have tion. A β-blocker would be an appropriate
been shown to reduce mortality rates in pa- first choice in a patient with ischemic heart
tients following an acute myocardial infarc- disease.
tion, in patients with chronic symptomatic There are some other guiding principles.
systolic heart failure (see Chapter 9), and First, the chosen drug regimen should
even in people at high risk for developing
conform to the patient’s specific needs. For
cardiovascular disease. The drugs also slow
example, an anxious young patient in the
the deterioration of renal function in pa-
throes of the hyperkinetic phase of EH
tients with diabetic nephropathy. The most
might be best treated with a β-blocker,
common side effect of ACE inhibitors is the
whereas a more effective choice for the
development of a reversible dry cough (likely
same patient many years later, after the
related to the increased bradykinin effect);
pressure becomes more dependent on pe-
hyperkalemia and azotemia may also occur,
ripheral vascular resistance, could be a va-
as described in Chapter 17.
sodilator (e.g., long-acting calcium channel
Angiotensin II receptor blockers (ARBs)
are the most recently introduced class of blocker). Because therapy is likely to con-
antihypertensive agents. The action of this tinue for many years, consideration of
group is to block the binding of angiotensin adverse effects and impact of drug therapy
II to its receptors (i.e., subtype AT1 receptors) on the patient’s quality of life are very
in blood vessels and other targets (see Fig. important.
17.6). By inhibiting the effects of angiotensin Another helpful principle of antihyper-
II (and thereby causing vasodilatation and re- tensive drug therapy concerns the use of
duced secretion of aldosterone), blood pres- multiple agents. The effects of one drug, act-
sure falls. In clinical trials, the antihyperten- ing at one physiologic control point, can be
sive efficacy of this group is similar to that of defeated by natural compensatory mecha-
ACE inhibitors. However, they are very well nisms. For example, the drop in renal perfu-
tolerated, and unlike ACE inhibitors, cough sion by a direct vasodilator can activate the
is not a common side effect. ARBs have been renin-angiotensin system, prompting the kid-
shown to reduce cardiovascular event rates ney to retain more volume, thereby blunting
(including myocardial infarction and stroke) the antihypertensive benefit. Combination
in high-risk patients. drug therapy is aimed at preventing such an
action by using agents acting at different
Given the large number of effective anti- complementary sites. In this example, a di-
hypertensive drugs available, the choice of rect vasodilator is often paired with a low-
10090-13_CH13.qxd 8/31/06 4:55 PM Page 333

Hypertension 333

dose diuretic to avoid the undesired volume Acknowledgment


expansion effect.
Contributors to the previous editions of this chapter
In conclusion, hypertension emerges as
were Rahul Deshmukh, MD; Rajesh S. Magrulkar,
a tremendously important clinical problem MD; Rajeev Malhotra, MD; Peter Allison McDo-
because of its prevalence and potentially nough, MD; A. Nigrovic, MD; Thomas J. Moore, MD;
devastating consequences. The evaluation and Leonard S. Lilly, MD.
and treatment of a patient with hyperten-
sion require methodical consideration of Additional Reading
the ways in which normal cardiovascular
physiology may have gone awry. Because Agarwal A, Williams GH, Fisher ND. Genetics of
human hypertension. Trends Endocrinol Metab
most patients still fall into the idiopathic
2005;16:127–133.
category of EH, there is still much room for
Chobanian AV, Bakris GL, Black HR, et al. The sev-
creative thought and research in this area. enth report of the joint national committee on
prevention, detection, evaluation, and treatment
of high blood pressure: the JNC 7 report. JAMA
SUMMARY 2003;289:2560–2571.
Dolan E, Stanton A, Thijs L, et al. Superiority of am-
1. Hypertension is defined as a chronic dias- bulatory over clinic blood pressure measurement
tolic blood pressure ≥90 mm Hg and/or in predicting mortality. Hypertension 2005;46:
systolic blood pressure ≥140 mm Hg. 156–161.
Eckel RH, Grundy SM, Zimmet PZ. The metabolic
2. Hypertension is of unknown etiology in
syndrome. Lancet 2005;365:1415–1428.
95% of patients (EH). Secondary hyper-
Elley CR, Arroll B. Refining the exercise prescription
tension may arise from diverse causes, in- for hypertension. Lancet 2005;366:1248–1249.
cluding (a) renal abnormalities (e.g., renal Fisher NDL, Williams GH. Hypertensive vascular dis-
parenchymal disease and renal artery ease. In: Kasper DL, Braunwald E, Fauci AS, et al.,
stenosis); (b) coarctation of the aorta; and eds. Harrison’s Principles of Internal Medicine.
(c) endocrine abnormalities (e.g., pheo- 16th Ed. New York: McGraw-Hill, 2004:1463–1480.
Giles TD, Berk BC, Black HR, et al. Expanding the de-
chromocytoma, primary or secondary
finition and classification of hypertension. J Clin
aldosteronism, Cushing syndrome, and Hypertens 2005;7:505–512.
thyroid abnormalities). Hajjar I, Kotchen TA. Trends in prevalence, aware-
3. Most hypertensive patients remain asymp- ness, treatment, and control of hypertension in
tomatic until complications arise. Poten- the United States, 1988–2000. JAMA 2003;290:
199–206.
tial complications include (a) stroke,
Haslam DW, James WP. Obesity. Lancet 2005;366:
(b) myocardial infarction, (c) heart fail- 1197–1209.
ure, (d) aortic aneurysm and dissection, Kaplan NM. Kaplan’s Clinical Hypertension. 9th Ed.
(e) renal damage, and (f) retinopathy. Philadelphia: Lippincott Williams & Wilkins, 2006.
4. Treatment of hypertension includes life- Weber MA. Hypertension treatment and impli-
style and dietary improvements, followed cations of recent cardiovascular outcome trials.
J Hypertens 2006;24(suppl 2):S37–S44.
by pharmacologic therapy. Commonly
White WB. Update on the drug treatment of hyper-
used antihypertensive drugs include thi-
tension in patients with cardiovascular disease.
azide diuretics, β-blockers, ACE inhibi- Am J Med 2005;118:695–705.
tors, angiotensin receptor blockers, and Williams B. Recent hypertension trials. J Am Coll
long-acting calcium channel blockers. Cardiol 2005;45:813–827.
10090-13_CH13.qxd 8/31/06 4:55 PM Page a

Chapter 13—Author Queries

1. AU: The chapter also refers to the renin-angiotensin system. Are both terms correct? Or
should they be consistent?
10090-14_CH14.qxd 8/31/06 4:56 PM Page 334

C H A P T E R

Diseases of the
Pericardium
Yanerys Ramos
14
Leonard S. Lilly

ANATOMY AND FUNCTION CARDIAC TAMPONADE


Etiology
ACUTE PERICARDITIS
Pathophysiology
Etiology
Clinical Features
Pathogenesis
Diagnostic Studies
Pathology
Treatment
Clinical Features
Diagnostic Studies CONSTRICTIVE PERICARDITIS
Treatment Etiology and Pathogenesis
Pathology
PERICARDIAL EFFUSION
Pathophysiology
Etiology
Clinical Features
Pathophysiology
Diagnostic Studies
Clinical Features
Treatment
Diagnostic Studies
Treatment

Diseases of the pericardium form a spectrum ceral pericardium) adheres to the outer wall of
that ranges from benign, self-limited peri- the heart and is reflected back on itself, at the
carditis to life-threatening cardiac tamponade. level of the great vessels, to line the tough fi-
The clinical manifestations of these disorders brous outer layer (parietal pericardium). A thin
and the approaches to their management can film of pericardial fluid slightly separates the
be predicted from an understanding of peri- two layers and decreases the friction between
cardial anatomy and pathophysiology, as pre- them.
sented in this chapter. The pericardium appears to serve three
functions: (1) fixing the heart within the
mediastinum and limits its motion, (2) pre-
ANATOMY AND FUNCTION
venting extreme dilatation of the heart dur-
The pericardium is a two-layered sac that en- ing sudden rises of intracardiac volume, and
circles the heart. The inner serosal layer (vis- (3) possibly functioning as a barrier to limit

334
10090-14_CH14.qxd 8/31/06 4:56 PM Page 335

Diseases of the Pericardium 335

the spread of infection from the adjacent by echovirus or coxsackievirus group B. Al-
lungs. However, patients with complete ab- though a viral origin could be confirmed in
sence of the pericardium (either congeni- infected patients by comparing antiviral
tally or after surgical removal) are generally titers of acute and convalescent serum, this
asymptomatic, casting doubt on its actual is rarely done in the clinical setting because
importance in normal physiology. Yet like the patient has usually recovered by the
the unnecessary appendix, the pericardium time those results would be available. Thus,
can become diseased and cause great harm. idiopathic and viral pericarditis are consid-
In the healthy heart, intrapericardial pres- ered similar clinical entities, and the terms
sure varies during the respiratory cycle from are used interchangeably.
−5 mm Hg (during inspiration) to +5 mm Hg Other viruses known to cause pericarditis
(during expiration) and nearly equals the include those responsible for influenza, vari-
pressure within the pleural space. However, cella, mumps, hepatitis B, and infectious
pathologic changes in pericardial stiffness, mononucleosis. Pericarditis has been found
or the accumulation of fluid within the with increased frequency among patients
pericardial sac, may profoundly increase this with AIDS, possibly related to HIV itself, but
pressure. often owing to superimposed tuberculous or
other bacterial infections in this immuno-
compromised population.
ACUTE PERICARDITIS
The most common affliction of the peri- Tuberculous Pericarditis
cardium is acute pericarditis, which refers to
inflammation of its layers. Many disease Although tuberculosis remains a worldwide
states and etiologic agents can produce this problem, its incidence in the United States is
Tab. 1 syndrome (Table 14.1), the most common of low. It is, however, an important cause of
which are described here. pericarditis in immunosuppressed patients,
such as those with AIDS. Tuberculous peri-
carditis arises from reactivation of the organ-
Etiology ism in mediastinal lymph nodes, with spread
Infectious into the pericardium. It can also extend di-
rectly from a site of tuberculosis within the
Idiopathic and Viral Pericarditis lungs, or the organism can arrive at the peri-
Acute pericarditis is most often of idiopathic cardium by hematogenous dissemination.
origin, meaning that the actual cause is
unknown. However, serologic studies have
Nontuberculous Bacterial Pericarditis
demonstrated that many such episodes are (Purulent Pericarditis)
actually caused by viral infection, especially
Bacterial pericarditis has become rare since
the advent of antibiotics. Pneumococcus and
staphylococci are responsible most fre-
TABLE 14.1. Most Common Causes of
Acute Pericarditis quently, whereas Gram-negative infection
occurs less often. Mechanisms by which bac-
Infectious terial invasion of the pericardium develops
Viral include (1) perforating trauma to the chest
Tuberculosis (e.g., stab wound); (2) contamination during
Pyogenic bacteria
Noninfectious
chest surgery; (3) extension of an intracardiac
Postmyocardial infarction infection (i.e., infective endocarditis); (4) ex-
Uremia tension of pneumonia or a subdiaphragmatic
Neoplastic disease infection; and (5) hematogenous spread from
Radiation-induced a remote infection. Bacterial pericarditis is a
Connective tissue diseases
fulminant illness but is rare in otherwise
Drug induced
healthy persons; it is most likely to occur in
10090-14_CH14.qxd 8/31/06 4:56 PM Page 336

336 Chapter Fourteen

immunocompromised patients, including phoma. Primary tumors of the pericardium


those with severe burns and malignancies. are rare. Neoplastic effusions are usually large
and hemorrhagic and frequently lead to car-
Noninfectious diac tamponade, a life-threatening complica-
tion described later in the chapter.
Pericarditis Following
Myocardial Infarction
Radiation-Induced Pericarditis
There are two forms of pericarditis associated
with acute myocardial infarction (MI). The Pericarditis may complicate radiation ther-
early form occurs within the first few days apy to the thorax (e.g., administered for the
after an MI. It likely results from inflamma- treatment of certain tumors), especially if
tion extending from the epicardial surface the cumulative dose has exceeded 4,000 rads
of the injured myocardium to the adjacent (40 Gy). Radiation-induced damage causes a
pericardium; therefore, it is most common local inflammatory response that can result
in patients with transmural (as opposed to in pericardial effusions and ultimately fi-
subendocardial) infarctions. The prognosis brosis. Cytologic examination of the peri-
following acute MI is not affected by the pres- cardial fluid helps to distinguish radiation-
ence of pericarditis; its major importance is induced pericardial damage from that of
in distinguishing it from the pain of recur- tumor invasion.
rent myocardial ischemia. This form of peri-
carditis occurs in fewer than 5% of patients
Pericarditis Associated with Connective
with acute MI who are treated with acute
Tissue Diseases
reperfusion strategies (see Chapter 7) but it is
more common in those who are not (and Pericardial involvement is common in many
who therefore sustain larger infarctions). connective tissue diseases, including sys-
The second form of post-MI pericarditis is temic lupus erythematosus (SLE), rheuma-
known as Dressler’s syndrome, which can de- toid arthritis, and progressive systemic scle-
velop 2 weeks to several months following rosis. For example, 20% to 40% of patients
an acute infarction. Its cause is unknown, with SLE experience clinically detectable
but it is thought to be of autoimmune origin, pericarditis during the course of the disease.
possibly directed against antigens released Customary treatment of the underlying con-
from necrotic myocardial cells. A clinically nective tissue disease usually ameliorates the
similar form of pericarditis may occur weeks pericarditis as well.
to months following heart surgery (termed
postpericardiotomy pericarditis).
Drug-Induced Pericarditis
Uremic Pericarditis Several pharmaceutical agents have been re-
ported to cause pericarditis as a side effect,
Pericarditis is a serious complication of chro-
often by inducing a syndrome similar to SLE
nic renal failure, but its pathogenesis in this
setting is unknown. Studies have shown no (Table 14.2). These drugs include the antiar- Tab. 2
correlation between the plasma level of ni- rhythmic procainamide and the vasodilator
trogen waste products and the incidence of hydralazine. Drug-induced pericarditis usu-
pericarditis, and it may even develop in pa- ally abates when the causative agent is dis-
tients during the first few months of dialysis continued.
therapy.
Pathogenesis
Neoplastic Pericarditis
Similar to other inflammatory processes,
Tumors within the pericardium most com- pericarditis is characterized by three stages:
monly results from metastatic spread or local (1) local vasodilation with transudation of
invasion by cancer of the lung, breast, or lym- protein-poor, cell-free fluid into the pericar-
10090-14_CH14.qxd 8/31/06 4:56 PM Page 337

Diseases of the Pericardium 337

ened and fused. Occasionally, this process


TABLE 14.2. Examples of Drug-Induced leads to a dense scar that restricts movement
Pericarditis
and diastolic filling of the cardiac chambers,
Related to drug-induced SLE-like syndrome as described later in the chapter.
Procainamide Suppurative (or purulent) pericarditis is
Hydralazine an intense inflammatory response associated
Methyldopa
most commonly with bacterial infection.
Isoniazid
Phenytoin
The serosal surfaces are erythematous and
Not related to drug-induced SLE-like syndrome coated with purulent exudate. Hemorrhagic
Anthracycline antineoplastic agents (doxorubicin, pericarditis refers to a grossly bloody form
daunorubicin) of pericardial inflammation and is most
Minoxidil often caused by tuberculosis or malignancy.
SLE, systemic lupus erythematosus.
Clinical Features
History
dial space; (2) increased vascular permeability, The most frequent symptoms of acute peri-
with leak of protein into the pericardial space; carditis are chest pain and fever (Table 14.3). Tab. 3
and (3) leukocyte exudation, initially by neu- The pain may be severe and most often local-
trophils, followed later by mononuclear cells. izes to the retrosternal area and left pre-
The leukocytes are of critical importance cordium; it may also radiate to the back and
because they help contain or eliminate the ridge of the left trapezius muscle. What dif-
offending infectious or autoimmune agent. ferentiates it from myocardial ischemia or in-
However, metabolic products released by farction is that the pain of pericarditis is typi-
these cells may prolong inflammation, cause cally sharp and pleuritic (it is aggravated by
pain and local cellular damage, and mediate inspiration and coughing) and positional (e.g.,
somatic symptoms such as fever. Therefore, sitting and leaning forward often lessen the
the immune response to pericardial injury discomfort). Dyspnea is common during acute
may significantly contribute to tissue dam- pericarditis but is not exertional and probably
age and symptomatology. results from a reluctance of the patient to
breathe deeply because of pleuritic pain.
Patients with idiopathic or viral peri-
Pathology carditis are typically young and previously
The pathologic appearance of the peri- healthy. Pericarditis of other causes should
cardium depends on the underlying cause be suspected in patients with the underlying
and severity of inflammation. Serous peri- conditions listed in Table 14.1 who develop
carditis is characterized by scant polymor- the typical sharp, pleuritic chest pains and
phonuclear leukocytes, lymphocytes, and fever.
histiocytes. The exudate is a thin fluid se-
creted by the mesothelial cells lining the Physical Examination
serosal surface of the pericardium. This likely
A scratchy pericardial friction rub is com-
represents the early inflammatory response
mon in acute pericarditis and is produced
common to all types of acute pericarditis.
Serofibrinous pericarditis is the most
commonly observed morphologic pattern
TABLE 14.3. Clinical Features of
in patients with pericarditis. The pericardial
Acute Pericarditis
exudate contains plasma proteins, including
fibrinogen, yielding a grossly rough and Pleuritic chest pain
shaggy appearance (termed “bread and but- Fever
ter” pericarditis). Portions of the visceral Pericardial friction rub
ECG abnormalities
and parietal pericardium may become thick-
10090-14_CH14.qxd 8/31/06 4:56 PM Page 338

338 Chapter Fourteen

by the movement of the inflamed pericar- most important ECG pattern, which reflects
dial layers against one another. Ausculta- inflammation of the adjacent myocardium,
tion of the rub is best heard using the di- consists of diffuse ST segment elevation in
aphragm of the stethoscope with the patient most of the ECG leads, usually with the
leaning forward while exhaling (which exception of aVR and V1 (Fig. 14.1). In addi-
brings the pericardium closer to the chest tion, PR segment depression in several leads
wall and stethoscope). In its full form, the is often evident, reflecting abnormal atrial
rub consists of three components, corre- repolarization related to atrial epicardial
sponding to the phases of greatest cardiac inflammation. These abnormalities are in
movement: ventricular contraction, ventri- contrast to the ECG of acute ST segment el-
cular relaxation, and atrial contraction. evation MI, in which the ST segments are el-
Characteristically, the pericardial rub is evated only in the leads overlying the re-
evanescent, coming and going from one ex- gion of infarction, and PR depression is not
amination to the next. expected.
Blood studies typically reveal signs of
acute inflammation, including an increased
Diagnostic Studies
white blood cell count (usually a mild lym-
The presence of pleuritic, positional chest phocytosis in acute viral/idiopathic peri-
pain and the characteristic pericardial fric- carditis) and elevation of the erythrocyte
tion rub implicate the presence of acute sedimentation rate. Some patients with
pericarditis. However, certain laboratory acute pericarditis also demonstrate elevated
studies are helpful to confirm the diagnosis serum cardiac biomarkers (e.g., cardiac tro-
and to assess for impending complications. ponin I), suggesting inflammation of the
The electrocardiogram (ECG) is abnormal neighboring myocardium.
in 90% of patients with acute pericarditis Further testing in acute pericarditis often
and helps to distinguish it from other forms includes echocardiography to evaluate for the
of cardiac disease, such as an acute MI. The presence and hemodynamic significance of

Figure 14.1. Electrocardiogram in acute pericarditis. Diffuse ST segment elevation is present. Also notice
depression of the PR segment (arrow). 1 LINE SHORT
10090-14_CH14.qxd 8/31/06 4:56 PM Page 339

Diseases of the Pericardium 339

a pericardial effusion. Additional studies dial disease usually indicates widely meta-
that may be useful in individual cases, to de- static cancer, and therapy is unfortunately
fine the cause of pericarditis include (1) pu- only palliative.
rified protein derivative skin test for tuber-
culosis, (2) serologic tests (antinuclear
PERICARDIAL EFFUSION
antibodies and rheumatoid factor) to screen
for connective tissue diseases, and (3) a care- Etiology
ful search for malignancy, especially of the
The normal pericardial space contains 15 to
lung and breast (physical examination sup-
plemented by chest radiograph and mam- 50 mL of pericardial fluid, a plasma ultrafil-
mogram). The yield of diagnostic pericardio- trate secreted by the mesothelial cells that
centesis (removal of pericardial fluid through line the serosal layer. A larger volume of
a needle) in uncomplicated acute pericardi- fluid may accumulate in association with
tis is low and should be reserved for patients any of the forms of acute pericarditis previ-
with very large effusions or evidence of car- ously discussed.
diac chamber compression, as discussed later In addition, noninflammatory serous ef-
in the chapter. fusions may result from conditions of (1) in-
creased capillary permeability (e.g., severe
hypothyroidism); (2) increased capillary hy-
Treatment drostatic pressure (e.g., congestive heart fail-
Idiopathic or viral pericarditis is a self-lim- ure); or (3) decreased plasma oncotic pressure
ited disease that usually runs its course in 1 (e.g., cirrhosis or the nephrotic syndrome).
to 3 weeks. Management consists of rest, to Chylous effusions may occur in the pres-
reduce the interaction of the inflamed peri- ence of lymphatic obstruction of pericardial
cardial layers, and pain relief by analgesic and drainage, most commonly caused by neo-
anti-inflammatory drugs (aspirin and other plasms and tuberculosis.
nonsteroidal anti-inflammatory agents). Oral
corticosteroids are often effective for severe Pathophysiology
or recurrent pericardial pain but should not
Because the pericardium is a relatively stiff
be used in uncomplicated cases because of
potentially severe side effects and because structure, the relationship between its inter-
even gradual withdrawal of this form of the- nal volume and pressure is not linear, as
rapy often leads to recurrent symptoms of shown in curve A of Figure 14.2. Notice that Fig. 2
pericarditis. the initial portion of the curve is nearly flat,
The forms of pericarditis related to myo- indicating that at the low volumes normally
cardial infarction are treated in a similar present within the pericardium, a small in-
fashion, with rest and aspirin. Other nons- crease in volume leads to only a small rise in
teroidal anti-inflammatory agents are often pressure. However, when the intrapericar-
avoided immediately following an MI be- dial volume expands beyond a critical level
cause of experimental evidence linking them (see Fig. 14.2, arrow), a dramatic increase in
to delayed healing of the infarct. pressure is incited by the nondistensible sac.
Purulent pericarditis requires more aggres- At that point, even a minor increase in vol-
sive treatment, including catheter drainage ume can translate into an enormous com-
of the pericardium and intensive antibiotic pressive force on the heart.
therapy. Nevertheless, even with such ther- Three factors determine whether a peri-
apy, the mortality rate is very high. Tuber- cardial effusion remains clinically silent or
culous pericarditis requires prolonged mul- whether symptoms of cardiac compression
tidrug antituberculous therapy. Pericarditis ensue: (1) the volume of fluid, (2) the rate at
in the setting of uremia often resolves fol- which the fluid accumulates, and (3) the com-
1 LINE SHORT lowing intensive dialysis. Neoplastic pericar- pliance characteristics of the pericardium.
10090-14_CH14.qxd 8/31/06 4:56 PM Page 340

340 Chapter Fourteen

Figure 14.2. Schematic representation of the volume-pressure re-


lationship of the normal pericardium. A. At the very lowest levels, a
small rise in volume results in a small rise in pressure. However, when the
limits of pericardial stretch are reached (arrow), the curve becomes very
steep, and a further small rise in intrapericardial volume results in signifi-
cantly increased pressure. B. Chronic slow accumulation of volume allows
the pericardium to gradually stretch over time; thus, the curve shifts to
the right and much larger volumes are accommodated at lower pressures.
(Modified from Freeman GL, LeWinter MM. Pericardial adaptations dur-
ing chronic dilation in dogs. Circ Res 1984;54:294.)

A sudden increase of pericardial volume, as present with findings of cardiac tamponade,


may occur in chest trauma with intraperi- as described later in the chapter. In addition,
cardial hemorrhage, results in marked eleva- the effusion may cause symptoms resulting
tion of pericardial pressure (see Fig. 14.2, from compression of adjacent structures,
steep portion of curve A) and the potential such as dysphagia (difficult swallowing be-
for severe cardiac chamber compression. cause of esophageal compression), dyspnea
Even lesser amounts of fluid may cause sig- (shortness of breath resulting from lung com-
nificant elevation of pressure if the peri- pression), hoarseness (caused by to recurrent
cardium is pathologically noncompliant and laryngeal nerve compression), or hiccups (re-
stiff, as may occur in the presence of tumor sulting from phrenic nerve stimulation).
or fibrosis of the sac. In contrast, if the peri- On examination (Table 14.4), a large peri- Tab. 4
cardial effusion accumulates slowly, over cardial fluid “insulates” the heart from the
weeks to months, the pericardium gradually chest wall, and the heart sounds may be muf-
stretches, such that the volume-pressure rela- fled. In fact, a friction rub that had been pre-
tionship curve shifts toward the right (see sent during the acute phase of pericarditis
Fig. 14.2, curve B). With this adaptation, the may disappear if a large effusion develops and
pericardium can accommodate larger vol- separates the inflamed layers from one an-
umes (e.g., 1 to 2 L) without marked eleva- other. Dullness to percussion of the left lung
tion of intrapericardial pressure. over the angle of the scapula may be present

Clinical Features
TABLE 14.4. Clinical Features of Large
A spectrum of possible symptoms is associ- Pericardial Effusion
ated with pericardial effusions. For example,
the patient with a large effusion may be Soft heart sounds
asymptomatic, may complain of a dull con- Reduced intensity of friction rub
Ewart sign (dullness over posterior left lung)
stant ache in the left side of the chest, or may
10090-14_CH14.qxd 8/31/06 4:56 PM Page 341

Diseases of the Pericardium 341

(known as the Ewart sign) owing to compres- order (e.g., intensive dialysis for uremic ef-
sive atelectasis by the enlarged pericardial sac. fusion). If the cause is not evident, the clini-
cal state of the patient determines whether
pericardiocentesis (removal of pericardial
Diagnostic Studies
fluid) should be undertaken. An asympto-
The chest radiograph may be normal if only a matic effusion, even of large volume, can be
small pericardial effusion is present. How- observed for long periods without specific
ever, if more than approximately 250 mL intervention. However, if serial examina-
has accumulated, the cardiac silhouette en- tion demonstrates a precipitous rise in peri-
larges in a globular, symmetric fashion. In cardial volume or if hemodynamic compres-
large effusions, the ECG may demonstrate sion of the cardiac chambers becomes
reduced voltage of the complexes. In the evident, then pericardiocentesis should be
presence of extremely large effusions, the performed for therapeutic drainage and for
height of the QRS complex may vary from analysis of the fluid.
beat to beat (electrical alternans), a result of a
constantly changing electrical axis as the
CARDIAC TAMPONADE
heart swings from side to side within the
large pericardial volume. At the opposite end of the spectrum from
One of the most useful laboratory tests in the asymptomatic pericardial effusion is car-
the evaluation of an effusion is echocardiog- diac tamponade. In this condition, pericar-
Fig. 3 raphy (Fig. 14.3), which can identify pericar- dial fluid accumulates under high pressure,
dial collections as small as 20 mL. This non- compresses the cardiac chambers, and se-
invasive technique can quantify the volume verely limits filling of the heart. As a result,
of pericardial fluid, determine whether ven- ventricular stroke volume and cardiac out-
tricular filling is compromised, and when put decline, potentially leading to hypoten-
necessary, help direct the placement of a sive shock and death.
pericardiocentesis needle.
Etiology
Treatment
Any etiology of acute pericarditis (see Table
If the cause of the effusion is known, ther- 14.1) can progress to cardiac tamponade,
apy is directed toward the underlying dis- but the most common causes are neoplastic,
postviral, and uremic pericarditis. Acute he-
morrhage into the pericardium is also an
important cause of tamponade, which can
result from (1) blunt or penetrating chest
trauma, (2) rupture of the left ventricular
(LV) free wall following MI (see Chapter 7),
or (3) as a complication of a dissecting aor-
tic aneurysm (see Chapter 15).
LV
PE
Pathophysiology
As a result of the surrounding tense pericar-
dial fluid, the heart is compressed, and the
diastolic pressure within each chamber becomes
elevated and equal to the pericardial pressure.
The pathophysiologic consequences of this
are illustrated in Figure 14.4. Because the Fig. 4
Figure 14.3. Two-dimensional echocardiogram (para-
sternal short-axis view) of a pericardial effusion (PE) compromised cardiac chambers cannot ac-
surrounding the heart. LV, left ventricle. commodate normal venous return to the
10090-14_CH14.qxd 8/31/06 4:56 PM Page 342

342 Chapter Fourteen

Cardiac tamponade Constrictive pericarditis

Hypotension

Jugular venous distention Pulmonary Reflex tachycardia


rales
pHe toa ge m lya ±siteac
re P phi alre emad

Figure 14.4. Pathophysiology of cardiac tamponade and constrictive peri-


carditis. The symptoms and signs (boxes) arise from impaired diastolic filling of the
ventricles in both conditions.

heart, the systemic and pulmonary venous ops signs and symptoms of systemic vascu-
pressures rise. The increase of systemic ve- lar congestion and decreased cardiac output
nous pressure results in signs of right-sided (Table 14.5). The key physical findings in-
heart failure (e.g., jugular venous disten- clude (1) jugular venous distention; (2) sys-
tion), whereas elevated pulmonary venous temic hypotension; and (3) a “small, quiet
pressure leads to pulmonary congestion. In heart” on physical examination, a result of
addition, reduced filling of the ventricles the insulating effects of the effusion. Other
during diastole decreases the systolic stroke signs include sinus tachycardia and pulsus
volume, and the cardiac output declines. paradoxus. Dyspnea and tachypnea reflect
These derangements trigger compensatory pulmonary congestion and decreased oxy-
mechanisms aimed at maintaining tissue gen delivery to peripheral tissues.
perfusion, initially through activation of the If tamponade develops suddenly, symp-
sympathetic nervous system. Nonetheless, toms of profound hypotension are evident,
failure to evacuate the effusion leads to inad-
equate perfusion of vital organs, shock, and
ultimately death.
TABLE 14.5. Clinical Features of
Cardiac Tamponade
Clinical Features Jugular venous distention
Cardiac tamponade should be suspected in Hypotension with pulsus paradoxus
Quiet precordium on palpation
any patient with known pericarditis, peri-
Sinus tachycardia
cardial effusion, or chest trauma who devel-
10090-14_CH14.qxd 8/31/06 4:56 PM Page 343

Diseases of the Pericardium 343

including confusion and agitation. How- Fig. 3.13). In addition, echocardiography can
ever, if the effusion develops more slowly, differentiate between cardiac tamponade and
over a period of weeks, then fatigue (caused other causes of low cardiac output, such as
by low cardiac output) and peripheral edema ventricular contractile dysfunction.
(owing to right-sided heart failure) may be The definitive diagnostic procedure for
the presenting complaints. cardiac tamponade is cardiac catheterization
Pulsus paradoxus is an important phys- with measurement of intracardiac and in-
ical sign in cardiac tamponade that can be trapericardial pressures, usually combined
recognized at the bedside using a blood pres- with therapeutic pericardiocentesis, as de-
sure cuff. It refers to a decrease of systolic blood scribed in the next section.
pressure (more than 10 mm Hg) during normal
inspiration. Treatment
Pulsus paradoxus is not really “paradoxi-
cal”; it is just an exaggeration of appropriate Removal of the high-pressure pericardial
cardiac physiology. Normally, expansion of fluid is the only intervention that reverses
the thorax during inspiration causes the in- the life-threatening physiology of this con-
trathoracic pressure to become more nega- dition. Pericardiocentesis is best performed
tive compared with the expiratory phase. in the cardiac catheterization laboratory,
This facilitates systemic venous return to the where the hemodynamic effect of fluid re-
moval can be assessed. The patient is posi-
chest and augments filling of the right ven-
tioned head up at a 45° angle to promote
tricle (RV). The transient increase in RV size
pooling of the effusion, and a needle is in-
shifts the interventricular septum toward the
serted into the pericardial space through the
left, which slightly diminishes left ventricu-
skin, just below the xiphoid process (which
lar filling. As a result, in normal persons, LV
is the safest location to avoid piercing a
stroke volume and systolic blood pressure
coronary artery). A catheter is then threaded
decline slightly following inspiration.
into the pericardial space and connected to
In cardiac tamponade, this situation is
a transducer for pressure measurement. An-
exaggerated because both ventricles share a
other catheter is threaded through a sys-
reduced, fixed volume as a result of external
temic vein into the right side of the heart,
compression by the tense pericardial fluid.
and simultaneous recordings of intracardiac
In this case, the inspiratory increase of right and intrapericardial pressures are compared.
ventricular volume and bulging of the in- In tamponade, the pericardial pressure is el-
terventricular septum toward the left have a evated and is equal to the diastolic pressures
proportionally greater effect on the limita- within the cardiac chambers; the diastolic
tion of LV filling. Thus, in tamponade there pressures are elevated to the same degree in AQ1
is a more substantial reduction of LV stroke all the chambers because of the surrounding
volume (and therefore systolic blood pres- compressive force of the effusion.
sure) following inspiration. In addition, the right atrial pressure trac-
Pulsus paradoxus may also be manifest ing, which is equivalent to the jugular ve-
by other conditions in which inspiration is nous pulsation observed on physical exami-
exaggerated, including severe asthma and nation, displays a characteristic abnormality
chronic obstructive airway disease. (Fig. 14.5). During early diastole in a normal Fig. 5
person, as the right ventricular pressure falls
Diagnostic Studies and the tricuspid valve opens, blood quickly
flows from the right atrium into the RV,
Echocardiography is the most useful noninva- leading to a rapid decline in the right atrial
sive technique to evaluate whether pericar- pressure tracing (y descent). In tamponade,
dial effusion has led to cardiac tamponade however, the pericardial fluid compresses
physiology. An important indicator of high- the right ventricle and prevents its rapid
pressure pericardial fluid is compression of expansion. Thus, the right atrium cannot
the RV and right atrium during diastole (see empty quickly, and the y descent is blunted.
10090-14_CH14.qxd 8/31/06 4:56 PM Page 344

344 Chapter Fourteen

pericardial fluid include cell counts (e.g.,


white cell count is elevated in bacterial infec-
tions and other inflammatory conditions)
and protein and lactate dehydrogenase (LDH)
levels. If the concentration ratio of pericardial
protein to serum protein is >0.5, or that of
pericardial LDH to serum LDH is >0.6, then
the fluid is consistent with an exudate; other-
wise, it is more likely a transudate. When tu-
berculosis is suspected, it is also useful to mea-
sure the level of adenosine deaminase in the
pericardial fluid. Some studies have indicated
that an elevated level is highly sensitive and
specific for tuberculosis.
If cardiac tamponade recurs following
pericardiocentesis, the procedure can be re-
peated. In some cases, a more definitive sur-
gical undertaking (removal of part or all of
the pericardium) is required to prevent reac-
cumulation of the effusion.

Figure 14.5. Schematic diagrams of right atrial (or CONSTRICTIVE PERICARDITIS


jugular venous) pressure electrocardiogram (ECG)
recordings. A. Normal. The initial a wave represents The other major potential complication of
atrial contraction. The v wave reflects passive filling of the pericardial diseases is constrictive pericardi-
atria during systole, when the tricuspid and mitral valves tis. This is a condition not often encoun-
are closed. After the tricuspid valve opens, the right atrial
pressure falls (y descent) as blood empties into the right tered but important to understand, because
ventricle. B. Cardiac tamponade. High-pressure pericar- it can masquerade as other, more common
dial fluid compresses the heart, impairing right ventricu- disorders. In addition, it is an affliction that
lar filling, so that the y descent is blunted. C. Constrictive
pericarditis. The earliest phase of diastolic filling is not im- may cause profound symptoms yet is fully
paired so that the y descent is not blunted. The y descent correctable if recognized.
appears accentuated because it descends from a higher
than normal right atrial pressure. The right atrial c wave
(described in Chapter 2) is not shown. Etiology and Pathogenesis
In the early part of the twentieth century,
tuberculosis was the major cause of con-
Following successful pericardiocentesis, strictive pericarditis, but that is much less
the pericardial pressure falls to normal and common today in industrialized societies.
is no longer equal to the pressures within The most frequent cause now is idiopathic
the heart chambers, which also decline to (i.e., months to years following presumed
their normal levels. After initial aspiration idiopathic or viral acute pericarditis). How-
of fluid, the pericardial catheter may be left ever, any cause of pericarditis can lead to this
in place for 1 to 2 days to allow more com- complication (see Table 14.1).
plete drainage.
When pericardial fluid is obtained for di-
Pathology
agnostic purposes, it should be stained and
cultured for bacteria, fungi, and acid-fast Following an episode of acute pericarditis,
bacilli (tuberculosis), and cytologic examina- any pericardial effusion that has accumu-
tion should be performed to evaluate for ma- lated usually undergoes gradual resorption.
lignancy. Other common measurements of However, in patients who later develop con- 1 LINE SHORT
10090-14_CH14.qxd 8/31/06 4:56 PM Page 345

Diseases of the Pericardium 345

strictive pericarditis, the fluid undergoes or- sudden cessation of ventricular diastolic fill-
ganization, with subsequent fusion of the ing imposed by the rigid pericardial sac.
pericardial layers, followed by fibrous scar In contrast to cardiac tamponade, peri-
formation. In some patients, calcification of cardial constriction results in pulsus para-
the adherent layers ensues, further stiffen- doxus much less frequently. Recall that in
ing the pericardium. tamponade, this finding reflects inspiratory
augmentation of RV filling, at the expense
of LV filling. However, in constrictive peri-
Pathophysiology
carditis, the negative intrathoracic pressure
The pathophysiologic abnormalities in con- generated by inspiration is not easily trans-
strictive pericarditis occur during diastole; mitted through the rigid pericardial shell to
systolic contraction of the ventricles is usu- the right-sided heart chambers; therefore,
ally normal. In this condition, a rigid, scarred inspiratory augmentation of RV filling is
pericardium encircles the heart and inhibits more limited. Rather, when a patient with
normal filling of the cardiac chambers. For ex- severe pericardial constriction inhales, the
ample, as blood passes from the right atrium negative intrathoracic pressure draws blood
into the right ventricle during diastole, the toward the thorax, where it cannot be ac-
RV size expands and quickly reaches the commodated by the constricted right-sided
limit imposed by the constricting peri- cardiac chambers. As a result, the increased
cardium. At that point, further filling is sud- venous return accumulates in the intratho-
denly arrested, and venous return to the racic systemic veins, causing the jugular
right heart ceases. Thus, systemic venous veins to become more distended during in-
pressure rises, and signs of right-sided heart spiration (Kussmaul sign). This is the oppo-
failure ensue. In addition, the impaired fill- site of normal physiology, in which inspira-
ing of the left ventricle causes a reduction in tion results in a decline in jugular venous
stroke volume and cardiac output, which pressure, as venous return is drawn into the
leads to lower blood pressure. heart. Thus, typical findings in pericardial
disease can be summarized as follows:

Clinical Features Constrictive Cardiac


The symptoms and signs of constrictive peri- Pericarditis Tamponade
carditis usually develop over months to Pulsus paradoxus + +++
Kussmaul sign +++ −
years. They result from (1) reduced cardiac
output (fatigue, hypotension, reflex tachy-
cardia) and (2) elevated systemic venous
Diagnostic Studies
pressures (jugular venous distention, hepa-
tomegaly with ascites, and peripheral edema). The chest radiograph in constrictive peri-
Because the most impressive physical find- carditis shows a normal or mildly enlarged
ings are often the insidious development of cardiac silhouette. Calcification of the peri-
hepatomegaly and ascites, patients may be cardium can be detected in some patients
mistakenly suspected of having hepatic cir- with severe chronic constriction. The ECG
rhosis or an intra-abdominal tumor. How- generally shows only nonspecific ST and T
ever, careful inspection of the jugular veins wave abnormalities, although atrial arrhyth-
can point to the correct diagnosis of con- mias are common.
strictive pericarditis. Echocardiographic evidence of constriction
On cardiac examination, an early dias- is subtle. The pericardium, if well imaged, is
tolic “knock” may follow the second heart thickened; the ventricular cavities are small
sound (S2; see Chapter 2) in patients with se- and contract vigorously, and diastolic ven-
1 LINE SHORT vere calcific constriction. It represents the tricular filling terminates abruptly in early
10090-14_CH14.qxd 8/31/06 4:56 PM Page 346

346 Chapter Fourteen

diastole, as the chambers reach the limit im- 14.5). After the tricuspid valve opens,
posed by the surrounding rigid shell. the right atrium quickly empties into the
Computed tomography or magnetic resonance RV (and its pressure rapidly falls) during
imaging is superior to echocardiography in the very brief period before filling is ar-
the assessment of pericardial anatomy and rested. This is in contrast to cardiac tam-
thickness. The presence of normal pericardial ponade, in which the external compres-
thickness (<2 mm) by these modalities is gen- sive force throughout the cardiac cycle
erally a reliable indication that constrictive prevents rapid ventricular filling, even in
pericarditis is not present. early diastole, such that the y descent is
The diagnosis of constrictive pericarditis blunted.
can be confirmed by cardiac catheterization, 4. During the respiratory cycle, there is dis-
which reveals four key features: cordance in the RV and LV systolic pres-
sures (the RV systolic pressure rises with
1. Elevation and equalization of the diastolic
inspiration, while that of the LV tends to
pressures in each of the cardiac chambers.
decline). In normal persons, the negative
2. An early diastolic “dip and plateau” con- intrathoracic pressure induced by inspi-
figuration in the right and left ventricu- ration causes the systolic pressure of both
Fig. 6 lar tracings (Fig. 14.6). This pattern re- ventricles to decline slightly. In contrast,
flects blood flow into the ventricles at the in constrictive pericarditis, the heart is
very onset of diastole, just after the tri- isolated from the rest of the thorax by the
cuspid and mitral valves open, followed surrounding rigid shell. In this circum-
by sudden cessation of filling as further stance, negative intrathoracic pressure
expansion of the ventricles is arrested by associated with inspiration decreases the
the surrounding rigid pericardium. pressure in the pulmonary veins but not
3. A prominent y descent shown in the in the left-sided cardiac chambers. This
right atrial pressure tracing (see Fig. causes a decline in the pressure gradient
driving blood back to the left side of the
heart, such that left ventricle filling is re-
ECG duced. Less ventricular filling reduces the
stroke volume and therefore results in a
100
lower LV systolic pressure during inspira-
LV tion (thus causing pulsus paradoxus in
some patients). Simultaneously, because
80 the two ventricles share a fixed space lim-
Pressure (mm Hg)

ited by the rigid pericardium, the reduced


60 LV volume allows the interventricular
septum to shift towards the left, which
RV enlarges the RV (a behavior termed ven-
40
tricular interdependence). The resultant in-
Plateau crease in RV filling augments its stroke
20 volume and systolic pressure during in-
spiration. During expiration, the situa-
Early diastolic
tion is reversed, with the RV systolic
filling wave
pressure declining and that of the LV
TIME
increasing.
Figure 14.6. Schematic tracings of left ventricular
(LV) and right ventricular (RV) pressures in constric- The clinical and hemodynamic findings of
tive pericarditis. Early diastolic ventricular filling abruptly constrictive pericarditis are often similar
halts as the volume in each ventricle quickly reaches the to those of restrictive cardiomyopathy (see
limit imposed by the constricting pericardium. Through-
out most of diastole, the LV and RV pressures are abnor- Chapter 10), another uncommon condition.
mally elevated and equal. Distinguishing between these two syndromes
10090-14_CH14.qxd 8/31/06 4:56 PM Page 347

Diseases of the Pericardium 347

TABLE 14.6. Differences Between Constrictive Pericarditis and Restrictive Cardiomyopathy

Feature Constrictive Pericarditis Restrictive Cardiomyopathy

Chest radiography
• Pericardial calcifications Yes (25–30% of patients) Absent
CT or MRI
• Thickened pericardium Yes No
Echocardiography
• Thickened pericardium Yes (but difficult to visualize) No
• Respiratory cycle effect on transvalvular Exaggerated variations Normal
Doppler velocities
Cardiac catheterization
• Equalized LV and RV diastolic pressures Yes Often, LV > RV
• Elevated PA systolic pressure Uncommon Common
• Effect of inspiration on ystolic pressures Discordant : LV↓, RV↑ Concordant: LV↓, RV↓
Endomyocardial biopsy Normal Abnormal (e.g., amyloid)

CT, computed tomography; LV, left ventricle; MRI, magnetic resonance imaging; PA, pulmonary artery; RV, right ventricle.

is important because pericardial constriction ST segment elevation on the ECG, often


is correctable, whereas most cases of restric- accompanied by PR segment depression.
tive cardiomyopathy have very limited ef- 3. Complications of pericarditis include car-
Tab. 6 fective treatments (Table 14.6). An endomyo- diac tamponade (accumulation of peri-
cardial biopsy is sometimes necessary to cardial fluid under high pressure, which
distinguish between these (the biopsy results compresses the cardiac chambers) and
are normal in constriction but usually ab- constrictive pericarditis (restricted filling
normal in restrictive cardiomyopathy; see of the heart because of surrounding rigid
Chapter 10). pericardium).

Treatment Acknowledgment
The only effective treatment of severe con- Contributors to the previous editions of this chapter
strictive pericarditis is surgical removal of were Angela Fowler, MD; Kathy Glatter, MD; Thomas
the pericardium. Symptoms and signs of G. Roberts, MD; Alan Braverman, MD; and Leonard
S. Lilly, MD.
constriction may not resolve immediately
because of the associated stiffness of the
neighboring outer walls of the heart, but Additional Reading
eventual symptomatic improvement is ex-
Bertog SC, Thambidorai SK, Parakh K, et al. Con-
pected in the majority of patients who
strictive pericarditis: etiology and cause-specific
undergo this procedure. survival after pericardiectomy. J Am Coll Cardiol
2004;43:1445–1452.
Gaya AM, Ashford RF. Cardiac complications of ra-
SUMMARY
diation therapy. Clin Oncol 2005;17(3):153–159.
1. Acute pericarditis is most often of idio- Imazio M, Bobbio M, Cecchi E, et al. Colchicine in
pathic or viral cause and is usually a self- addition to conventional therapy for acute peri-
limited illness. More serious forms of peri- carditis: Results of the COlchicine for acute
PEricarditis (COPE) Trial. Circulation 2005;112:
carditis arise from the conditions listed in
2012–2016.
Table 14.1.
Lange RA, Hillis LD. Clinical practice. Acute peri-
2. Common findings in acute pericarditis carditis. N Engl J Med 2004;351:2195.
include (a) pleuritic chest pain; (b) fever; Little WC, Freeman GL. Pericardial disease. Circula-
(c) pericardial friction rub; and (d) diffuse tion 2006;113:1622–1632.
10090-14_CH14.qxd 8/31/06 4:56 PM Page 348

348 Chapter Fourteen

Maisch B, Seferovic PM, Ristic AD, et al. Guidelines Shabetai R. The Pericardium. Boston: Kluwer Aca-
on the diagnosis and management of pericardial demic, 2003.
diseases executive summary; the task force on the Spodick DH. Acute cardiac tamponade. N Engl J Med
diagnosis and management of pericardial diseases 2003;349:684–690.
of the European society of cardiology. Eur Heart J Troughton R, Asher CR, Klein AL. Pericarditis. Lancet
2004;25:587. 2004;363:717–727.
Permayer-Miralda G. Acute pericardial disease: ap-
proach to aetiologic diagnosis. Heart 2004;90:
252–254.
10090-14_CH14.qxd 8/31/06 4:56 PM Page a

Chapter 14—Author Query


1. AU: Correct interpretation of “all of the latter”?
10090-15_CH15.qxd 8/31/06 4:57 PM Page 349

C H A P T E R

Diseases of the
Peripheral Vasculature
Arash Mostaghimi
15
Mark A. Creager

DISEASES OF THE AORTA DISEASE CAUSING ARTERIAL SPASM:


Aortic Aneurysms RAYNAUD PHENOMENON
Aortic Dissection VENOUS DISEASE
OCCLUSIVE ARTERIAL DISEASES Varicose Veins
Peripheral Arterial Disease Venous Thrombosis
Acute Arterial Occlusion
Vasculitic Syndromes

Peripheral vascular disease is an umbrella term tion of immune cells to traumatized or in-
that includes a number of diverse pathologic fected tissues. Disease states of the periph-
entities that affect arteries, veins, and lym- eral vasculature interfere with these essen-
phatics. Although this terminology makes a tial functions.
distinction between the “central” coronary Peripheral vascular diseases result from
and “peripheral” systemic vessels, the vascu- processes that can be grouped into three cat-
lature as a whole comprises a dynamic, inte- egories: (1) structural changes in the vessel wall
grated, and multifunctional organ system secondary to degenerative conditions, infec-
that does not naturally comply with this se- tion, or inflammation that lead to dilation,
mantic division. aneurysm, dissection, or rupture; (2) narrow-
Blood vessels serve many critical func- ing of the vascular lumen caused by athero-
tions. First, they regulate the differential dis- sclerosis, thrombosis, or inflammation; and
tribution of blood and delivery of nutrients (3) spasm of vascular smooth muscle. These
and oxygen to tissues. Second, blood vessels processes can occur in isolation or one may
actively synthesize and secrete vasoactive lead to another.
substances that regulate vascular tone and
antithrombotic substances that maintain
DISEASES OF THE AORTA
the fluidity of blood and vessel patency (see
Chapters 6 and 7). Third, the vessels play an The aorta is the largest conductance vessel of
integral role in the transport and distribu- the vascular system. In adults, its diameter is

349
10090-15_CH15.qxd 8/31/06 4:57 PM Page 350

350 Chapter Fifteen

approximately 3 cm at its origin at the base Diseases of the aorta most commonly
of the heart. The ascending aorta, 5 to 6 cm appear as one of three clinical conditions:
in length, leads to the aortic arch, from aneurysm, dissection, or obstruction.
which arise three major branches: the bra-
chiocephalic (which bifurcates into the right
Aortic Aneurysms
common carotid and subclavian arteries),
the left common carotid, and the left sub- An aneurysm is an abnormal localized di-
clavian arteries. As the descending aorta latation of an artery. In the aorta, aneurysms
continues beyond the arch, its diameter nar- are distinguished from diffuse ectasia, which
rows to approximately 2 to 2.5 cm in healthy is a generalized yet lesser increase of the
adults. As the aorta pierces the diaphragm, it aortic diameter. Ectasia develops in older
becomes the abdominal aorta, providing patients as elastic fibers fragment, smooth
arteries to the abdominal viscera before bi- muscle cells decrease in number, and acid
furcating into the left and right common mucopolysaccharide ground substance ac-
iliac arteries, which supply the pelvic organs cumulates within the vessel wall.
and lower extremities. The term aneurysm is applied when the di-
The aorta, like other arteries, is composed ameter of a portion of the aorta has increased
Fig. 1 of three layers (Fig. 15.1). At the luminal sur- by at least 50% compared with normal. A
face, the intima is composed of endothelial true aneurysm represents a dilatation of all
cells overlying the internal elastic lamina. The three layers of the aorta, creating a large
bulge of the vessel wall. True aneurysms are
endothelial layer is a functional interface be-
characterized as either fusiform or saccular,
tween the vasculature and circulating blood
depending on the extent of the vessel’s cir-
cells and plasma. The media is composed of
cumference within the aneurysm (see Fig.
smooth muscle cells and a matrix that in-
15.1). A fusiform aneurysm, the more com-
cludes collagen and elastic fibers. Collagen
mon type, is characterized by symmetrical di-
provides tensile strength that enables the ves-
lation of the entire circumference of a seg-
sels to withstand high-pressure loads. Elastin
ment of the aorta. A saccular aneurysm is a
is capable of stretching to 250% of its original
localized outpouching involving only a por-
length and confers a distensible quality on
tion of the circumference.
vessels that allows them to recoil under pres- In contrast, a pseudoaneurysm, or false
sure. The adventitia is primarily composed of aneurysm, is a contained rupture of the ves-
collagen fibers, perivascular nerves, and vasa sel wall that develops when blood leaks out
vasorum, a rich vascular network that sup- of the vessel lumen through a hole in the in-
plies oxygenated blood to the aorta. timal and medial layers and is contained by
The aorta is subject to injury from me- a layer of adventitia or perivascular or-
chanical trauma because it is continuously ganized thrombus (see Fig. 15.1). Pseudo-
exposed to high pulsatile pressure and shear aneurysms develop at sites of vessel injury
stress. The predominance of elastin in the caused by infection or trauma, such as punc-
media (2:1 over collagen) allows the aorta to ture of the vessel during surgery or percuta-
expand during systole and recoil during di- neous catheterization. They are very unsta-
astole. The recoil of the aorta against the ble and are prone to rupture.
closed aortic valve contributes to the distal Aneurysms may be confined to the abdo-
propagation of blood flow during the phase minal aorta (most common), the thoracic
of left ventricular relaxation. With advanc- aorta, or both locations. They may also ap-
ing age, the elastic component of the aorta pear in peripheral and cerebral arteries.
and its branches degenerates, and as colla-
gen becomes more prominent, the arteries
Etiology and Pathogenesis
stiffen. Systolic blood pressure therefore
of True Aortic Aneurysms
tends to rise with age because less energy is
dissipated into the aorta during left ventric- The etiology of aortic aneurysm formation is
ular contraction. multifactorial and varies depending on the
10090-15_CH15.qxd 8/31/06 4:57 PM Page 351

Diseases of the Peripheral Vasculature 351

Intima Intima
Media Media
Adventitia Adventitia

lumen
A Aortic wall

Fusiform Saccular

Hematoma
contained by
adventitia or
perivascular clot

Hole in intima
and media
C

Figure 15.1. Classification of aortic aneurysms. A. The normal arterial wall consists of three
layers: the intima, media, and adventitia. B. True aneurysms represent localized dilatation of all
three layers of the arterial wall. Fusiform aneurysms involve the entire circumference of the aorta,
whereas saccular aneurysms are a localized bulge of only a portion of the circumference. C. A false
aneurysm (or pseudoaneurysm) is actually a hole in the intima and media, with hematoma con-
tained by a thin layer of adventitia or perivascular clot.

location of the lesion. Ascending thoracic aor- hypertension and occurs in certain inher-
tic aneurysms typically are characterized by ited disorders of connective tissue, includ-
cystic medial necrosis, a process of degen- ing Marfan syndrome and Ehlers-Danlos
eration and fragmentation of elastic fibers, syndrome.
with accumulation of collagenous and mu- Aneurysms of the descending thoracic and
coid material within the medial layer. This abdominal aorta are associated with athero-
process is associated with aging and with sclerosis and its risk factors, including smok-
10090-15_CH15.qxd 8/31/06 4:57 PM Page 352

352 Chapter Fifteen

ing, hypertension, dyslipidemia, male gen- in cough, dyspnea, or pneumonia. Com-


der, and advanced age. However, it is unlikely pression of the esophagus can result in dys-
that atherosclerosis alone is responsible for phagia, and involvement of the recurrent
such aneurysm development. Rather, impor- laryngeal nerve may lead to hoarseness.
tant contributions appear to derive from ge- Aneurysms of the ascending aorta may di-
netic predisposition (on a polygenetic basis), late the aortic ring, resulting in aortic regur-
local vessel inflammation, and an imbal- gitation and symptoms of congestive heart
ance between synthesis and degradation of failure. Abdominal aortic aneurysms may
extracellular matrix proteins such as colla- cause abdominal or back pain or nonspecific
gen and elastin. For example, matrix metal- gastrointestinal symptoms.
loproteinases (see Chapter 5) capable of de- Aortic aneurysms are often first suspected
grading the components of the extracellular when dilatation of the vessel is observed on
matrix are found in increased quantities chest or abdominal radiographs, particu-
within aortic aneurysms. Aneurysm forma- larly if the wall is calcified. Aneurysms of the
tion is also associated with markers of inflam- abdominal aorta or of the large peripheral
mation, including C-reactive protein (CRP) arteries may also be discovered by careful
and cytokines such as interleukin 6 (IL-6). palpation during physical examination. The
Levels of both CRP and IL-6 have been shown diagnosis is confirmed by ultrasonography,
to correlate with the size of aneurysms, and contrast-enhanced computed tomography
inflammatory cells such as B and T lympho- (CT), or magnetic resonance angiography
cytes and macrophages are frequently found (Fig. 15.2). Fig. 2
on histologic examination. The most devastating consequence of an
Infrequent causes of aortic aneurysms aortic aneurysm is rupture, which can be
Tab. 1 (Table 15.1) include weakness of the media fatal. An aneurysm may leak slowly or burst
from infections of the vessel wall by Salmo- suddenly, resulting in profound blood loss
nella species, staphylococci, streptococci, tu- and hypotension. Thoracic aortic aneurysms
berculosis, syphilis, or fungi. Inflammatory may rupture into the pleural space, medi-
diseases such as Takayasu arteritis or giant astinum, or bronchi. Abdominal aortic aneu-
cell arteritis (both described later in the rysms may rupture into the retroperitoneal
chapter) may similarly weaken the vessel
and result in aneurysm formation.

Clinical Presentation and Diagnosis


Most aneurysms are asymptomatic, though
some patients, especially those with abdom-
inal aortic aneurysms, may be aware of a
pulsatile mass. Others present with symp-
toms related to compression of neighboring
structures by an expanding aneurysm. Tho-
racic aortic aneurysms may compress the
trachea or mainstream bronchus, resulting

TABLE 15.1. Conditions Associated


with True Aortic Aneurysms

1. Cystic medical necrosis


2. Atherosclerosis Figure 15.2. Abdominal aortic aneurysm. Computed
3. Infections of arterial wall tomographic angiogram (CTA) of an abdominal aortic
4. Vasculitis aneurysm, indicated by the arrow. (Courtesy of Dr. Frank
Rybicki, Brigham and Women’s Hospital, Boston, MA).
10090-15_CH15.qxd 8/31/06 4:57 PM Page 353

Diseases of the Peripheral Vasculature 353

space or abdominal cavity or erode into the Surgical repair of abdominal aortic aneu-
intestines, resulting in massive gastrointesti- rysms involves placement of a prosthetic
nal bleeding. graft. The operative mortality for such pro-
Natural history studies have shown that cedures at high-volume institutions is 1% to
the risk of rupture is related to the size of the 2%. Percutaneous endovascular repair of in-
aneurysm, as predicted by LaPlace’s relation- frarenal abdominal aortic aneurysms with
ship (wall tension is proportional to the prod- stent grafts can be performed in selected pa-
uct of pressure and radius). The mean rates of tients with less acute morbidity, and results
thoracic and abdominal aortic aneurysms ex- appear to be similar to that of surgical re-
pansion are 0.1 and 0.4 cm/year, respectively. pair, but long-term outcomes are still being
Thoracic aneurysms rupture at an annual rate defined.
of 2% for aneurysms <5 cm in diameter, 3%
for aneurysms 5 to 5.9 cm, and 7% for
Aortic Dissection
aneurysms >6 cm. Abdominal aneurysms
<4 cm, 4 to 4.9 cm, and 5 to 5.9 cm have an Aortic dissection is a life-threatening condi-
annual rates of rupture of 0.3%, 1.5%, and tion in which a blood-filled channel divides
6.5%, respectively. Abdominal aneurysms the medial layers of the aorta, splitting (or
>6 cm have a markedly higher risk of rupture. dissecting) the intima from the adventitia
along various lengths of the vessel.
Treatment
Etiology, Pathogenesis, and Classification
Treatment of an aortic aneurysm is based
on its size and the patient’s overall medical Aortic dissection is thought to arise from a
condition. Once an aneurysm is identified, tear in the intimal layer of the vessel wall
its dimensions should be closely moni- that allows blood from the lumen, under
tored through repeated imaging every 6 the driving force of the systemic pressure, to
to 12 months. In general, surgical treat- enter into the media and propagate along
ment is considered for ascending aortic the plane of the muscle layer. Another pos-
aneurysms >5.5 to 6.0 cm. Ascending aor- tulated origin of aortic dissection relates to
tic aneurysms in patients with Marfan syn- rupture of vasa vasorum with hemorrhage
drome (in whom the risk of complications into the media, forming a hematoma in the
is greater) should be considered for surgical arterial wall that subsequently tears through
repair if the diameter is >5 cm. Surgical the intima and into the vessel’s lumen.
repair is generally recommended for de- Any condition that interferes with the
scending thoracic aortic aneurysms mea- normal integrity of the elastic or muscular
suring 6.5 to 7.0 cm, for abdominal aortic components of the medial layer can predis-
aneurysms measuring 5.5 cm or more, and pose to aortic dissection. Such degeneration
for smaller aneurysms that enlarge at a rate may arise from chronic hypertension, aging,
>1.0 cm/year. and/or cystic medial degeneration (which,
The mortality associated with elective sur- as described earlier, is a feature of certain
gical repair of thoracic aortic aneurysms is hereditary connective tissue disorders, such
3% to 5%. Patients are maintained on car- as the Marfan syndrome and Ehlers-Danlos
diopulmonary bypass as the aneurysm is re- syndrome). In addition, traumatic insult to
sected and replaced with a prosthetic Dacron the aorta (e.g., blunt chest trauma or acci-
graft. Patients with aneurysms involving mul- dental vessel damage during intra-arterial
tiple aortic segments have staged repairs, in catheterization or cardiac surgery) can also
which one segment is corrected at a time. incite this condition.
Some patients may be candidates for mini- Aortic dissection is most common in the
mally invasive repair, in which a translumi- sixth and seventh decades and occurs more
nally placed endovascular stent graft is posi- frequently in men. More than two thirds of
tioned across the aneurysm. patients have a history of hypertension. Dis-
10090-15_CH15.qxd 8/31/06 4:57 PM Page 354

354 Chapter Fifteen

section most commonly involves the as- may also be classified as acute or chronic,
cending thoracic aorta (65%) and descend- with acute dissections presenting with symp-
ing thoracic aorta (20%), while the aortic toms of less than 2 weeks’ duration.
arch (10%) and abdominal aortic (5%) seg-
ments are less commonly affected.
Clinical Presentation and Diagnosis
Dissections are commonly classified into
two categories (Stanford types A and B), de- The most common symptom of aortic dis-
pending on their location and extent (Fig. section is sudden, severe pain with a “tear-
Fig. 3 15.3). In a type A dissection (proximal), the ing” or “ripping” quality in the anterior
ascending aorta is involved, regardless of the chest (typical of type A dissections) or be-
site of the primary tear. A type B dissection tween the scapulae (type B dissections). The
(distal) does not involve the ascending aorta pain travels as the dissection propagates
or arch and is therefore confined to the de- along the aorta and can radiate anywhere in
scending thoracic and abdominal aorta. This the thorax or abdomen. Painless dissection
distinction is important because treatment is possible but uncommon (6.4% of cases).
strategies and prognoses are determined by Other symptoms relate to the catastrophic
location. Proximal aortic involvement tends complications that can occur at the time of
to be the more devastating form because of presentation or thereafter (Table 15.2) and Tab. 2
its potential for extension into the coronary include (1) rupture through the adventitia
and arch vessels, the support structures of anywhere along the aorta (often into the left
the aortic valve, or the pericardial space. Ap- pleural space or pericardium); (2) occlusion
proximately two thirds of dissections are of major branches of the aorta by the prop-
type A and one third are type B. Dissections agating hematoma within the vessel wall,

Figure 15.3. Aortic dissection. Type A involves the ascending aorta,


whereas type B does not. (Reprinted with permission from Cotran RS, Kumar
V, Robbins SL. Robbin’s Pathologic Basis of Disease. Philadelphia: WB Saun-
AQ3 ders, 1989.)
10090-15_CH15.qxd 8/31/06 4:57 PM Page 355

Diseases of the Peripheral Vasculature 355

local expertise. In emergency situations, CT


TABLE 15.2. Complications of Aortic Dissection scanning or TEE can generally be obtained
Rupture rapidly and offer excellent sensitivity and
Pericardial tamponade specificity for the diagnosis.
Hemomediastinum
Hemothorax (usually left sided)
Occlusion of aortic branch vessels Treatment
Carotid (stroke)
Coronary (myocardial infarction) The goal of acute treatment is to arrest pro-
Splanchnic (organ infarction) gression of the dissecting channel. Suspi-
Renal (acute renal failure) cion of acute aortic dissection warrants im-
AQ2 Iliac, brachiocephalic, subclavian (limb ischemia) mediate medical therapy to reduce systolic
Distortion of aortic annulus
blood pressure (aiming for a systolic pres-
Aortic regurgitation
sure of 100 to 120 mmHg) and to decrease
the force of left ventricular contraction and
thus minimize aortic wall shear stress. Use-
which compresses the lumen and can result ful pharmacologic agents in this regard in-
in myocardial infarction (coronary artery in- clude β-blockers (to reduce the force of con-
volvement), stroke (carotid artery involve- traction and heart rate as well as to lower
ment), visceral ischemia, renal failure, or blood pressure) and vasodilators such as so-
loss of pulse in an extremity; and (3) exten- dium nitroprusside (to rapidly reduce blood
sion into the aortic root, with disruption of pressure). In proximal (type A) dissections,
the aortic valve support apparatus causing early surgical correction has been shown to
aortic regurgitation. improve outcomes compared with medical
Several important physical findings may therapy alone. Surgical therapy involves re-
be present. Hypertension is frequently de- pairing the intimal tear, suturing the edges
tected, either as an underlying cause of dis- of the false channel, and if necessary, in-
section, a result of the sympathetic nervous serting a synthetic aortic graft.
system response to the severe pain, or be- In contrast, patients with uncomplicated
cause of diminished renal vascular flow, with type B dissections are initially managed with
activation of the renin-angiotensin system. aggressive medical therapy alone; early sur-
If the dissection has occluded one of the gical intervention does not improve the out-
subclavian arteries, a difference in systolic come in these patients. Surgery is indicated,
blood pressure between the arms is noted. however, if there is clinical evidence of
Neurologic deficits may accompany dissec- propagation of the dissection, compromise
tion into the carotid vessels. If a type A dis- of major branches of the aorta, impending
section results in aortic regurgitation, an rupture, or continued pain. Percutaneous
early diastolic murmur can be detected on catheter-based repair with endovascular
auscultation. Leakage from a type A dissec- stent grafts has been used successfully in se-
tion into the pericardial sac may produce lected stable patients with type B dissec-
signs of cardiac tamponade (see Chapter 14). tions. The graft seals the entry site of the dis-
The diagnosis of aortic dissection must section, resulting in thrombosis of the false
not be delayed, because catastrophic com- lumen.
plications or death may rapidly ensue. The
confirmatory imaging techniques most use-
OCCLUSIVE ARTERIAL DISEASES
ful in detecting dissection include contrast-
enhanced CT, transesophageal echocardiog- Arterial occlusion may result from athero-
raphy (TEE), magnetic resonance imaging, sclerosis, thromboembolism, or vasculitis
and contrast angiography. Each of these (inflammation of the vessel wall). The clini-
techniques has specific advantages and dis- cal presentation of these disorders results
advantages, and the decision of which to from decreased perfusion to the affected limb
employ is often guided by the hospital’s or organs.
10090-15_CH15.qxd 8/31/06 4:57 PM Page 356

356 Chapter Fifteen

Peripheral Arterial Disease degree of vessel narrowing by the stenosis


(i.e., the change in r) has the greatest impact
Etiology and Pathogenesis on flow. For example, if the radius is reduced
The formation of atherosclerotic plaques in by one half, the flow will be reduced to one
large and medium-sized arteries may result sixteenth of its baseline. The equation also
in chronic occlusive arterial disease, with indicates that for stenoses of the same
progressive stenosis and obstruction of length and radius, higher flow rates corre-
blood flow. This disorder is often referred to spond to greater pressure drops across the
as peripheral arterial disease (PAD), and al- stenoses. That is, as the flow velocity in-
though it may affect many vascular beds, it creases across a stenotic vessel, the blood
generally refers to atherosclerotic disease in turbulence results in a loss of kinetic energy.
arteries of the pelvis or lower limbs. Its The result is a decline in perfusion pressure
symptoms result from ischemia distal to the distal to the stenosis.
stenosis. It is a very prevalent vascular dis- During exercise, products of skeletal mus-
order, affecting approximately 4% of per- cle metabolism (e.g., adenosine) act locally
sons over age 40 and 15% to 20% of those to dilate arterioles. The resulting decrease in
over age 70. vascular resistance serves to increase blood
The pathology of PAD is identical to that flow to the active muscle (recall that flow =
of atherosclerotic coronary artery disease pressure ÷ resistance). In turn, the increased
(CAD), and the major coronary risk factors flow stimulates healthy arterial endothelium
(e.g., cigarette smoking, diabetes mellitus, to release vasodilating factors such as nitric
dyslipidemia, and hypertension) are also as- oxide, thereby increasing the radii of up-
sociated with PAD. Approximately 40% of stream conduit vessels. However, in PAD, ob-
patients with PAD actually have clinically structed arteries cannot respond to the va-
significant CAD. As a consequence of the sodilating stimuli, thereby limiting flow
systemic nature of atherosclerosis, patients increases. In addition, dysfunctional athero-
with PAD have a twofold to fivefold in- sclerotic endothelium does not release nor-
creased risk of cardiovascular death com- mal amounts of vasodilating substances (see
pared with patients who do not have this Chapter 6). Thus, the physical properties of
condition. a stenosis and the reduced vasodilator activ-
The pathophysiology of PAD is also simi- ity imposed by diseased endothelium pre-
lar to that of CAD. Ischemia of the affected vent adequate blood flow from reaching
region occurs when the balance between distal tissues and contribute to ischemia.
oxygen supply and demand is upset; exer- Hemodynamic changes alone cannot ac-
cise raises the demand for blood flow in the count for the dramatic reductions in exer-
limbs’ skeletal muscle, and a stenosed or ob- cise capacity experienced by PAD patients;
structed artery cannot provide an adequate changes in muscle structure and function
supply. Rest improves symptoms as the bal- are also seen. One such change is the dener-
ance between oxygen supply and demand is vation and dropout of muscle fibers, which
restored. is thought to occur as an adaptation to in-
Recall from Chapter 6 that the degree of termittent ischemia. The loss of such fibers
blood flow reduction relates closely to the can explain the reduced muscle strength
extent of vessel narrowing, the length of the and atrophy that occur in PAD patients.
stenosis, and blood viscosity. Pouseille’s Even viable muscle fibers in affected limbs
equation describes this relationship: may show abnormalities of mitochondrial
∆P πr 4 oxidative metabolism.
Q= In summary, atherosclerotic lesions pro-
8ηL
duce stenoses in peripheral conduit vessels
in which Q = flow, ∆P = pressure drop across and limit blood flow to the affected extrem-
the stenosis, r = vessel radius, η = blood vis- ity. Mechanisms normally in place to com-
cosity, and L = length of stenosis. Thus, the pensate for increased demand, such as en-
10090-15_CH15.qxd 8/31/06 4:57 PM Page 357

Diseases of the Peripheral Vasculature 357

dogenous release of vasodilators during ex- (Fig. 15.4). Patients with PAD may therefore Fig. 4
ercise and recruitment of microvessels, fail develop buttock, thigh, or calf discomfort
in the face of endothelial dysfunction and precipitated by walking and relieved by rest.
diminished flow velocity. Thus, states of in- This classic symptom of exertional limb fa-
creased oxygen demand are not met with tigue and pain is known as claudication. In
adequate supply, producing limb ischemia. severe PAD, patients may experience pain at
Adaptations to ischemia include changes in rest, usually affecting the feet or toes. The
muscle fiber metabolism and muscle fiber chronically reduced blood flow in this case
dropout. Together, these physical and bio- predisposes the extremity to ulceration, in-
chemical changes result in weak lower limbs fection, and skin necrosis (Fig. 15.5). Patients Fig. 5
that suffer ischemic discomfort during exer- who smoke or have diabetes mellitus are at
cise. Severe peripheral atherosclerosis may high risk of these complications.
reduce limb blood flow to such an extent The location of claudication corresponds
that it cannot satisfy resting metabolic re- to the diseased artery, with the femoral and
quirements. This results in critical limb ische- popliteal arteries being the most common
mia, which may progress to tissue necrosis sites (Table 15.3). The arteries of the upper Tab. 3
and gangrene that may threaten viability of extremities are less frequently affected, but
the extremity. brachiocephalic or subclavian artery disease
can cause arm claudication.
Physical examination generally reveals
Clinical Presentation and Diagnosis
loss of pulses distal to the stenotic segment.
PAD may affect the aorta or the iliac, fe- Bruits (swishing sounds auscultated over a re-
moral, popliteal, and tibioperoneal arteries gion of turbulent blood flow) may be audible

Figure 15.4. An angiogram demonstrating atherosclerotic disease of the iliac vessels. No-
tice the severe stenosis of the left external iliac artery (arrow).
10090-15_CH15.qxd 8/31/06 4:57 PM Page 358

358 Chapter Fifteen

TABLE 15.3. Relation of Stenotic Site


to Claudication Symptoms

Location of
Site Claudication Symptoms

Distal aorta or iliac Buttocks, hips, thighs,


arteries or calves
Femoral and popliteal Calves
arteries
Subclavian or axillary Arms
arteries

Ischemic ulcers resulting from PAD often


begin as small traumatic wounds in areas of
increased pressure or in regions prone to in-
jury, such as the tips of toes and lateral
malleolus (see Fig. 15.5A). These often-
painful ulcers fail to heal owing to the inad-
equate blood supply. Diabetic patients with
peripheral sensory neuropathies are particu-
larly susceptible to ulcers at sites of trauma
or pressure from ill-fitting footwear. Ische-
mic ulcers can be distinguished from venous
insufficiency ulcers, which develop more
proximally and on the medial portion of the
leg. Venous ulcers are also associated with
reddish-brown pigmentation and varicose
veins (see Fig. 15.5B).
In the evaluation of PAD, it is helpful to
measure the ratio of blood pressure in the
ankles to that in the arms (termed the ankle-
brachial index [ABI]) using a blood pressure
cuff and a Doppler instrument to detect
blood flow. A normal ABI is ≥1.0 (i.e., the
ankle pressure is equal to, or slightly greater
than, that in the arms). An index <0.9 is di-
Figure 15.5. Ulcerations caused by vascular insuffi- agnostic of PAD and may be associated with
ciency A. Arterial insufficiency. Ulceration (arrow) affect-
ing the great toe in a patient with severe peripheral arte- symptoms of claudication, whereas and
rial disease. B. Venous insufficiency ulcer near the medial index <0.5 is often observed in patients with
malleolus of the right leg. Notice the pigmentation of the rest pain and severe arterial compromise of
surrounding skin.
the affected extremity. Other testing to as-
sess peripheral perfusion includes limb seg-
in the abdomen (because of stenoses within mental systolic pressure measurements (using
the mesenteric or renal arteries) or over iliac, pneumatic cuffs placed along the extremity)
femoral, or subclavian arterial stenoses. In and pulse volume recordings (i.e., graphical
patients with chronic severe ischemia, the measurement of volume changes in seg-
lack of blood perfusion results in muscle at- ments of the extremity with each pulse). Du-
rophy, pallor, cyanotic discoloration, hair plex ultrasonography is a commonly used
loss, and occasionally gangrene and necrosis noninvasive method to visualize and assess
of the foot and digits. the extent of arterial stenoses and the corre-
10090-15_CH15.qxd 8/31/06 4:57 PM Page 359

Diseases of the Peripheral Vasculature 359

sponding reductions in blood flow. Other therapy in cases of severe limb ischemia. In
more advanced imaging studies (e.g., mag- severe cases, therapy is directed at healing
netic resonance angiography, computed tomo- ischemic ulcerations and preventing limb
graphic angiography, or intra-arterial contrast loss. Catheter-based interventions, such as
angiography) are obtained when revascular- percutaneous transluminal angioplasty and
ization procedures are planned. stent implantation, can be performed on se-
lected patients with low morbidity. Surgical
procedures include bypass operations to cir-
Treatment
cumvent the occluded arteries using saphe-
For all patients with PAD, antiplatelet ther- nous vein or prosthetic grafts. However, am-
apy and risk factor modification (including putation may be necessary if blood flow
smoking cessation, lipid lowering, and con- cannot be satisfactorily reestablished to main-
trol of diabetes and hypertension) are impor- tain limb viability.
tant in reducing the likelihood of coronary
events. Platelet inhibitors such as aspirin
Acute Arterial Occlusion
have been shown to reduce cardiovascular
morbidity and mortality in patients with Acute arterial occlusion is caused either by
PAD. It has not been established if anti- embolization from a cardiac or proximal
platelet agents reduce symptoms or prevent vascular site or by thrombus formation in
thrombotic complications of PAD itself. situ. The origin of arterial emboli is most
Specific treatment of PAD includes sup- often the heart, usually resulting from dis-
portive care of the feet to prevent trauma or orders involving intracardiac stasis of flow
restriction of blood flow. Exercise, particu- (Table 15.4). Emboli may also originate from Tab. 4
larly walking, improves endurance in part thrombus or atheromatous material overly-
by increasing metabolic efficiency in the ing a segment of the aorta. Rarely, arterial
skeletal muscle of the legs. A formal exercise emboli originate from the venous circula-
program is considered first-line therapy in tion. If a venous clot travels to the right-
the management of PAD. heart chambers and is able to pass through
Certain medical therapies are sometimes an abnormal intracardiac communication
useful in the treatment of claudication. (e.g., an atrial septal defect), it then enters
Cilostazol is a selective phosphodiesterase the systemic arterial circulation (a condition
inhibitor that increases cyclic adenosine known as a paradoxical embolism). Pri-
monophosphate and has vasodilator and mary arterial thrombus formation may ap-
platelet inhibiting properties; it has been
shown to improve exercise capacity in pa-
tients with PAD. Pentoxifylline is a drug pur-
ported to improve the deformability of red TABLE 15.4. Origins of Arterial Emboli
and white blood cells and may improve clau-
dication symptoms in some patients. Note Cardiac origin
that most vasodilator drugs (see Chapter 17) Stagnant left atrial flow (e.g., atrial fibrillation,
mitral stenosis)
are not helpful in relieving claudication. Left ventricular mural thrombus (e.g., dilated
More effective medical therapies for PAD cardiomyopathy, myocardial infarction, ventric-
are on the horizon. Advances in angiogene- ular aneurysm)
sis research and ongoing clinical trials pro- Valvular lesions (endocarditis, mitral stenosis,
vide hope that pharmacologic revasculariza- thrombus on prosthetic valve)
Left atrial myxoma (mobile tumor in left atrium)
tion with angiogenic growth factors, such as Aortic origin
vascular endothelial growth factor and basic Thrombus material overlying atherosclerotic
fibroblast growth factor may be possible. segment
Mechanical revascularization is indicated Venous origin
when medical therapy has failed for patients Paradoxical embolism travels through intracardiac
shunt
with disabling claudication and as first-line
10090-15_CH15.qxd 8/31/06 4:57 PM Page 360

360 Chapter Fifteen

pear at sites of endothelial damage or ather- Vasculitic Syndromes


osclerotic stenoses, or within bypass grafts.
The extent of tissue damage from throm- Vasculitis (vessel wall inflammation) results
boembolism depends on the site of the oc- from immune complex deposition or cell-
cluded artery, the duration of occlusion, mediated immune reactions directed
and the degree of collateral circulation against the vessel wall. Immune complexes
serving the tissue beyond the obstruction. activate the complement cascade with sub-
Common symptoms and signs that may sequent release of chemoattractants and
develop from reduced blood supply include anaphylatoxins that direct neutrophil mi-
pain, pallor, paralysis, paresthesia, and gration to the vessel wall and increase vas-
pulselessness (termed the “five Ps”). A sixth cular permeability. Neutrophils injure the
P, poikilothermia (coolness), is also often vessel by releasing lysosomal contents and
manifest. producing toxic oxygen-derived free radi-
Patients with a proven acute arterial oc- cals. In cell-mediated immune reactions,
clusion should be treated with anticoagu- T lymphocytes bind to vascular antigens and
lant agents such as heparin (followed by release lymphokines that attract additional
warfarin) to prevent propagation of the lymphocytes and macrophages to the vessel
clot and to reduce the likelihood of addi- wall. These inflammatory processes can
tional embolic events. A revascularization cause end-organ ischemia through vascular
procedure (catheter-based thrombolysis or necrosis or local thrombosis.
thrombectomy, surgical embolectomy, or The cause of most of the vasculitic syn-
bypass surgery) is indicated if limb viability dromes is unknown, but they often can be
is at risk. distinguished from one another by the pat-
Atheroembolism is the condition of pe- tern of involved vessels and by histologic
AQ1 ripheral arterial occlusion by atheromatous characteristics (Table 15.5). Tab. 5
material (i.e., cholesterol, platelets, and fibrin) Takayasu arteritis is a chronic vasculitis
derived from more proximal vascular sites, of unknown etiology that targets the aorta
such as atherosclerotic lesions or aneurysms. and its major branches. The estimated an-
The emboli lodge distally, resulting in occlu- nual incidence is 1.2 to 2.6 cases per million.
sion of small arteries in the muscle and skin. Between 80% and 90% of affected persons
Patients typically present with acute pain and are women, with onset typically between
tenderness at the involved site. Occlusion of the ages of 10 and 40. Most reported cases
digital vessels may result in the “blue toe” have been from Asia and Africa, but it is a
syndrome, culminating in gangrene and worldwide disease. Patients typically present
necrosis. Other findings may include livedo with systemic complaints such as malaise
reticularis (purplish mottling of involved and fever; focal symptoms are related to
skin), kidney failure (caused by renal ath- inflammation of the affected vessel and
eroembolism), and intestinal ischemia. Al- include cerebrovascular ischemia (brachio-
though an estimated 50% to 60% of cases are cephalic or carotid artery involvement),
spontaneous, atheroembolism may occur myocardial ischemia (coronary artery), arm
after intra-arterial procedures (e.g., cardiac claudication (brachiocephalic or subclavian
catheterization) when atherosclerotic mate- artery), or hypertension (renal artery). The
rial is unintentionally dislodged from the carotid and limb pulses are diminished or
proximal vasculature. Ischemia resulting absent in nearly 85% of patients at the time
from atheroemboli is difficult to manage be- of diagnosis; hence, this condition is often
cause the heterogeneous composition and termed a “pulseless” disease. Takayasu ar-
distribution of emboli often precludes sur- teritis is also an uncommon cause of aortic
AQ2 gical removal or fibrinobolytic therapy. aneurysm or aortic dissection. Histologic
Surgical intervention to remove or bypass examination of affected vessels reveals con-
the source of emboli may be necessary to tinuous or patchy granulomatous inflam-
prevent recurrences. mation with lymphocytes, histiocytes, and
10090-15_CH15.qxd 8/31/06 4:57 PM Page 361

Diseases of the Peripheral Vasculature 361

TABLE 15.5. Vasculitic Syndromes

Type Arteries Commonly Affected Histology

Takayasu arteritis Aorta and its branches Granulomatous arteritis with fibrosis; sig-
nificant luminal narrowing
Giant cell arteritis Medium to large size (especially cranial Lymphocyte infiltration, intimal fibrosis,
vessels as well as aortic arch and its granuloma formation
branches)
Thromboangiitis Small size (especially distal arteries of Inflammation and thrombosis without
obliterans extremities) necrosis
(Buerger disease)

multinucleated giant cells, resulting in inti- echoic halo around the involved arterial
mal proliferation, disruption of the elastic lumen with vessel stenosis and/or occlu-
lamina, and fibrosis. Antiendothelial anti- sion. The diagnosis can be confirmed by
bodies may also play a role in the disease. biopsy of an involved vessel, usually a tem-
Steroid and cytotoxic drugs may reduce vas- poral artery, but treatment should not wait
cular inflammation and alleviate symptoms for biopsy results. High-dose systemic steroids
of Takayasu arteritis. Surgical bypass of ob- are effective in treating vasculitis and pre-
structed vessels may be helpful in severe venting visual complications. Giant cell ar-
cases. The 5-year survival rate is 80% to 90%. teritis usually has a self-limited course of
Giant cell arteritis (also termed temporal 1 to 5 years.
arteritis) is a chronic vasculitis of medium- Thromboangiitis obliterans (Buerger
sized to large arteries that most commonly disease) is a segmental inflammatory dis-
involve the cranial vessels or the aortic arch ease of small and medium-sized arteries,
and its branches. It is an uncommon dis- veins, and nerves involving the distal vessels
ease, with an incidence of 24 per 100,000, of the upper and lower extremities. It is
and the typical onset is after age 50; 65% of most prevalent in the Far and Middle East
patients are female. Giant cell arteritis may and has a very strong association with ciga-
be associated with the inflammatory condi- rette smoking. It is most common in men
tion known as polymyalgia rheumatica. His- younger than age 45; only 10% to 25% of
tologic findings in affected vessels include patients are female. There is an increased in-
lymphocyte and macrophage infiltration, cidence of human leukocyte antigen A9
intimal fibrosis, and focal necrosis, with (HLA-A9) and HLA-B5 in affected persons.
granulomas containing multinucleated giant Thromboangiitis obliterans presents with
cells. a triad of symptoms and signs: distal arter-
Symptoms and signs of giant cell arteritis ial occlusion, Raynaud phenomenon (de-
depend on the distribution of affected arter- scribed in the next section), and migrating
ies and may include diminished temporal superficial vein thrombophlebitis. Arterial
pulses, prominent headache (temporal artery occlusion results in arm and foot claudica-
involvement), or facial pain and claudica- tion as well as ischemia of the digits. Tradi-
tion of the jaw while chewing (facial artery tional laboratory markers of inflammation
involvement). Ophthalmic artery arteritis and autoimmune disease are usually not de-
leads to impaired vision, with permanent tected. Arteriographic features of involved
partial or complete loss in 15% to 20% of pa- arteries include areas of stenosis interspersed
tients. In giant cell arteritis, the erythrocyte with normal-appearing vessels with more
sedimentation rate and C-reactive protein severe disease distally, collateral vessels
are invariably elevated as markers of inflam- with a “corkscrew” appearance around the
mation. Ultrasound examination can sup- stenotic regions, and lack of atherosclerosis
port the diagnosis by demonstrating a hypo- in proximal arteries. The diagnosis can be
10090-15_CH15.qxd 8/31/06 4:57 PM Page 362

362 Chapter Fifteen

boangiitis obliterans is smoking cessation,


which usually prevents progression of the
disease and its complications. Debridement
of necrotic tissue may be necessary in ad-
vanced cases. Revascularization is not usu-
ally an option because of the distal location
of the arterial lesions.

DISEASE CAUSING ARTERIAL


SPASM: RAYNAUD PHENOMENON
Raynaud phenomenon is a vasospastic dis-
Figure 15.6. Histologic section of an artery display- ease of the digital arteries that occurs in sus-
ing thromboangiitis obliterans. Endothelial cell and fi- ceptible people when exposed to cool tem-
broblast proliferation appears in the vessel wall, and
thrombus is present in the vessel lumen (arrow).
peratures or sometimes during emotional
stress. Vasospasm is an extreme vasocon-
strictor response that temporarily obliterates
established by tissue biopsy, although this is the vascular lumen, inhibiting blood flow.
rarely needed. Biopsy specimens of affected Typically, such episodes are characterized by
vessels reveal an occlusive, highly cellular, a triphasic color response. First, the fingers
inflammatory thrombus, with limited in- and/or toes blanch to a distinct white as
volvement of the vessel wall and preserva- blood flow is interrupted (Fig. 15.7). The sec- Fig. 7
Fig. 6 tion of the internal elastic lamina (Fig. 15.6). ond phase is characterized by cyanosis, re-
The most important treatment for throm- lated to local accumulation of desaturated

Figure 15.7. Raynaud phenomenon. The fourth digit (arrow) is


blanched (phase 1 of the tricolor response).
10090-15_CH15.qxd 8/31/06 4:57 PM Page 363

Diseases of the Peripheral Vasculature 363

hemoglobin, followed by a third phase of rial occlusive disease, the digital vascular
ruddy color as blood flow resumes. The lumen is largely obliterated by sclerosis or
color response may be accompanied by inflammation, resulting in lower intralumi-
numbness, paresthesias, or pain of the af- nal pressure and greater susceptibility to
fected digits. sympathetically mediated vasoconstriction.
This condition may occur as an isolated Treatment of Raynaud phenomenon in-
disorder, termed primary Raynaud phenome- volves avoiding cold environments, dress-
non or Raynaud disease. Patients are predom- ing in warm clothes, and wearing insulated
inantly women between the ages of 20 and gloves or footwear. There has also been
40. Primary Raynaud phenomenon most some success in preventing vasospasm with
often manifests in the fingers, but 40% of pharmacologic agents that relax vascular
patients also have involvement of the toes. tone, including calcium channel blockers
The prognosis of primary Raynaud phe- and α-adrenergic blockers (see Chapter 17),
nomenon is relatively benign, with only a but such therapies are reserved for severe
minority reporting a worsening of symp- cases.
toms over time.
Secondary Raynaud phenomenon may
VENOUS DISEASE
appear as a component of other conditions.
Common causes include connective tissue Veins are high-capacitance vessels that con-
diseases (e.g., scleroderma and systemic lupus tain more than 70% of the total blood vol-
erythematosus) and arterial occlusive dis- ume. In contrast to the muscular structure
orders. Other causes of secondary Raynaud of arteries, the subendothelial layer of veins
phenomenon include carpal tunnel syn- is thin, and the tunica media comprises
drome, thoracic outlet syndrome, blood fewer, smaller bundles of smooth muscle
dyscrasias, certain drugs, and thermal or cells intermixed with reticular and elastic
vibration injury. fibers. Whereas veins of the extremities pos-
Even in healthy vessels, cold exposure sess intrinsic vasomotor activity, transport
normally produces a vasoconstrictor re- of blood back to the heart relies greatly on
sponse. Cooling stimulates the sympathetic external compression by the surrounding
nervous system, resulting in local discharge skeletal muscles and on a series of one-way
of norepinephrine, which binds to vascular endothelial valves.
adrenergic receptors. In the fingers and toes, Veins of the extremities are classified as
only vasoconstricting α-receptors are pres- either deep or superficial. In the lower ex-
ent; other regional circulations have both tremities, where most peripheral venous dis-
constrictor and dilator adrenergic responses. orders occur, the deep veins generally course
Thus, a modest vasoconstriction of the dig- along the arteries, whereas the superficial
its results when healthy people are exposed veins are located subcutaneously. The super-
to cooling. In contrast, in Raynaud phe- ficial vessels drain into deeper veins through
nomenon, cold exposure induces severe vaso- a series of perforating connectors, ultimately
constriction. returning blood to the heart.
A variety of mechanisms have been pro-
posed to explain the vasospastic response to
Varicose Veins
cold and stress in patients with primary Ray-
naud phenomenon, including an exagger- Varicose veins (Fig. 15.8) are dilated, tortu- Fig. 8
ated sympathetic discharge in response ous superficial vessels that often develop in
to cold, heightened vascular sensitivity to the lower extremities. Clinically apparent
adrenergic stimuli, or excessive release of varicose veins occur in 10% to 20% of the
vasoconstrictor stimuli, such as serotonin, general population. They affect women two
thromboxane, and endothelin. In patients to three times more frequently than men,
with secondary Raynaud phenomenon and roughly half of patients have a family
caused by connective tissue diseases or arte- history of this condition. Varicosities can
10090-15_CH15.qxd 8/31/06 4:57 PM Page 364

364 Chapter Fifteen

Figure 15.8. A patient with extensive venous varicosities of the


right leg.

occur in any vein in the body but are most ondary varicose veins occur when an abnor-
common in the saphenous veins of the leg mality in the deep venous system is the
and their tributaries. They may also develop cause of superficial varicosities. These may
in the anorectal area (hemorrhoids), in the develop in the setting of deep venous insuf-
lower esophageal veins (esophageal varices), ficiency or occlusion, or when the perforat-
and in the spermatic cord (varicocele). ing veins are incompetent. In such cases,
Varicosity is thought to result from in- deep venous blood is shunted retrogradely
trinsic weakness of the vessel wall, from in- through perforating channels into super-
creased intraluminal pressure, or from con- ficial veins, increasing intraluminal pres-
genital defects in the structure and function sure and volume and causing dilatation and
of venous valves. Varicose veins in the lower varicosity formation.
extremities are classified as either primary Many people with varicose veins are
or secondary. Primary varicose veins origi- asymptomatic but seek treatment for cos-
nate in the superficial system, and factors metic reasons. When symptoms do develop,
that contribute to their development in- they include a dull ache, “heaviness,” or a
clude pregnancy, prolonged standing, and pressure sensation in the legs after pro-
obesity. During pregnancy or prolonged longed standing. Superficial venous insuffi-
standing, the high venous pressure within ciency may result when venous valves are
the legs contributes to varicosities when unable to function normally in the dilated
there is underlying weakness of the vessel veins. This can cause swelling and skin ul-
walls. In obese patients, the adipose tissue ceration that is particularly severe near the
surrounding vessel walls offers less struc- ankle. Stasis of blood within varicose veins
tural support to veins than lean mass. Sec- can promote superficial vein thrombosis,
10090-15_CH15.qxd 8/31/06 4:57 PM Page 365

Diseases of the Peripheral Vasculature 365

and varicosities can also rupture, causing a sequences of deep venous thrombosis are
localized hematoma. pulmonary embolism and postphlebitic syn-
Varicose veins are usually treated conser- drome. Pulmonary embolism can supervene
vatively. Patients should elevate their legs when a clot, most often from a DVT in the
while supine, avoid prolonged standing, proximal veins of the lower extremities, dis-
and wear external compression stockings lodges and travels through the inferior
that counterbalance the increased venous vena cava and right-heart chambers, finally
hydrostatic pressure. Small symptomatic reaching and obstructing a portion of the
varicosities are sometimes treated by injec- pulmonary vasculature (Fig. 15.9; also see Fig. 9
tion of a sclerosing agent into the vein. Laser Fig. 3.21). This complication may be her-
treatments can be used to improve the cos- alded by pleuritic chest pain, tachypnea,
metic appearance of small affected vessels. cough, and/or dyspnea. Pulmonary em-
Endovenous laser therapy, radiofrequency bolism is common (incidence of approxi-
ablation procedures, and surgical vein liga- mately 600,000 per year in the United
tion and removal are used for patients who States) and is often fatal, with an untreated
are very symptomatic, suffer recurrent mortality rate of 30% to 40%.
superficial vein thrombosis, or develop skin Postphlebitic syndrome, or chronic deep
ulcerations. venous insufficiency, results from venous
valvular damage and/or persistent occlusion
by deep venous thrombosis. This may lead
Venous Thrombosis to chronic leg swelling, stasis pigmentation,
The term venous thrombosis or thrombophle- and skin ulcerations.
bitis is used to describe thrombus within a In 1856, Virchow described a triad of fac-
superficial or deep vein and the inflamma- tors that predispose to venous thrombosis:
tory response in the vessel wall that it in- (1) stasis of blood flow, (2) hypercoagulabil-
cites. Thrombi in the lower extremities are ity, and (3) vascular damage. Stasis disrupts
classified by location as either deep venous laminar flow and brings platelets into con-
thrombi or superficial venous thrombi. tact with the endothelium. This allows co-
Initially, the venous thrombus is com- agulation factors to accumulate and retards
posed principally of platelets and fibrin. Later, the influx of clotting inhibitors. Factors that
red blood cells become interspersed within slow venous flow and induce stasis include
the fibrin, and the thrombus tends to propa- immobilization (e.g., prolonged bed rest
gate in the direction of blood flow. The after surgery, or sitting in a car or an airplane
changes in the vessel wall can be minimal or for a long trip), cardiac failure, and hyper-
can include granulocyte infiltration, loss of viscosity syndromes (Table 15.6). Tab. 6
endothelium, and edema. Thrombi may di- Various clinical disorders cause systemic
minish or obstruct vascular flow, or they hypercoagulability, including resistance of
may dislodge and form thromboemboli. coagulation factor V to activated protein C,
a prothrombin gene mutation, and inher-
ited deficiencies of antithrombin III, protein
Deep Venous Thrombosis C, and protein S. Pancreatic, lung, stomach,
breast, and genitourinary tract adenocarci-
Epidemiology, Etiology,
nomas are associated with a high prevalence
and Pathogenesis
of venous thrombosis. This is thought to
Deep venous thrombosis (DVT) occurs most occur in part because necrotic tumor cells re-
commonly in the veins of the calves but lease thrombogenic factors. Other condi-
may also develop initially in more proximal tions that contribute to hypercoagulability
veins such as the popliteal, femoral, and are listed in Table 15.6.
iliac vessels. If left untreated, 20% to 30% of Vascular damage, either by external in-
DVTs that occur in the calves may propagate jury or intravenous catheters, can denude
to these proximal veins. The two major con- the endothelium and expose subendothelial
10090-15_CH15.qxd 8/31/06 4:57 PM Page 366

366 Chapter Fifteen

Figure 15.9. Pulmonary angiogram displaying a massive pulmonary embolism. There is


a large filling defect in the left main pulmonary artery (arrow), additional filling defects in the
lower pulmonary artery branches, and a paucity of vessels in the left midlung region (a result
of obstructed flow).

TABLE 15.6. Conditions That Predispose collagen. Exposed collagen acts as a sub-
to DVT strate for the binding of von Willebrand fac-
tor and platelets and initiates the clotting
Stasis of blood flow cascade, leading to clot formation. Less se-
Prolonged inactivity (following surgery, long travel
vere damage can cause endothelial dysfunc-
by car or plane)
Immobilized extremity (following bone fracture) tion that contributes to thrombosis by dis-
Heart failure (with systemic venous congestion) rupting the production of vasodilating and
Hyperviscosity syndromes (e.g., polycythemia vera) antiplatelet substances (e.g., nitric oxide
Hypercoagulable states and prostacyclin) and antithrombotic mol-
Inherited disorders of coagulation
ecules such as thrombomodulin and hep-
Resistance to activated protein C (factor V Leiden)
Prothrombin gene mutation (PT G20210A) aran sulfate.
Antithrombin III deficiency The risk of venous thrombosis is partic-
Deficiency of protein C or protein S ularly high after fractures of the spine,
Antiphospholipid antibodies/lupus anticoagulant pelvis, and bones of the lower extremities.
Neoplastic disease (e.g., pancreatic, lung, stomach,
The risk following bone fracture may be re-
or breast cancers)
Pregnancy and oral contraceptive use lated to stasis of blood flow, increased coag-
Myeloproliferative diseases ulability, and possibly traumatic endothelial
Smoking damage. In addition, venous thrombosis oc-
Vascular damage curs frequently in patients following surgi-
Instrumentation (e.g., intravenous catheters)
cal procedures, particularly major orthope-
Trauma
dic operations.
10090-15_CH15.qxd 8/31/06 4:57 PM Page 367

Diseases of the Peripheral Vasculature 367

Women have a several-fold increase in only 75% sensitive for diagnosing sympto-
the incidence of venous thrombus forma- matic calf vein thrombi. This technique uses
tion during late pregnancy and the early real-time ultrasound scanning to image the
postpartum period. In the third trimester, vein and pulsed Doppler ultrasound to assess
the fetus compresses the inferior vena cava blood flow within it (Fig. 15.10). Criteria used Fig. 10
and can cause stasis of blood flow, and for diagnosis of DVT with duplex ultra-
high levels of circulating estrogen may in- sonography include the inability to compress
duce a hypercoagulable state. Oral contra- the vein with direct pressure (suggesting the
ceptives and other pharmacologic estrogen presence of an intraluminal thrombus), di-
products may also predispose to thrombus rect visualization of the thrombus, and ab-
formation. sence of blood flow within the vessel.
Other diagnostic techniques are some-
times used. For example, magnetic resonance
Clinical Presentation venography can aid in the diagnosis of proxi-
mal DVT, particularly pelvic vein thrombi,
Patients with DVT may be asymptomatic.
which are difficult to detect by ultrasound.
Symptomatic patients often describe calf or
Contrast venography is an invasive imaging
thigh discomfort, particularly when stand- technique that can provide a definitive diag-
ing or walking, or report unilateral leg nosis. Radiocontrast material is administered
swelling. The physical signs of proximal into a foot vein, and images are obtained as
DVT include edema of the involved leg and the contrast ascends through the venous sys-
occasionally localized warmth and erythema. tem of the leg. DVT is diagnosed by the pres-
Tenderness may be present over the course ence of a filling defect (see Fig. 15.10).
of the phlebitic vein, and a deep venous
cord (induration along the thrombosed ves-
sel) is occasionally palpable. Calf pain pro- Treatment
duced by dorsiflexion of the foot (Homans
Elevation of the affected extremity above
sign) is a nonspecific and unreliable sign
the level of the heart helps reduce edema
of DVT.
and tenderness, and anticoagulation pre-
vents extension of the thrombus and pul-
Diagnosis monary embolism. Initial anticoagulation
usually consists of subcutaneous low molec-
The primary laboratory tests for the diagno- ular weight heparin (LMWH). Intravenous un-
sis of DVT include measurement of the fractionated heparin is a cost-effective alter-
serum D-dimer level and venous compres- native that has been used successfully for
sion ultrasonography. D-dimer, a byproduct this purpose for many years, although some
of fibrin degradation that can be measured studies have shown superior outcomes with
in a peripheral blood sample, is highly sen- LMWH, which is also more convenient to
sitive for the diagnosis of DVT and/or acute administer (see Chapter 17). Warfarin, an
pulmonary embolism. Because D-dimer oral anticoagulant, is prescribed for long-
may also be elevated in many other condi- term management and is usually continued
tions (such as cancer, inflammation, infec- for 6 months or more, depending on the un-
tion, and necrosis), a positive test result is derlying cause of DVT. Catheter-based
not specific for DVT. Thus, a normal D-dimer thrombolysis may be useful for selected pa-
value can help exclude the presence of DVT, tients with iliofemoral deep vein thrombo-
but an elevated level does not confirm the sis. In patients with proximal DVT who can-
diagnosis. not be treated with anticoagulants because
Venous compression duplex ultrasonography of a bleeding disorder, an intravascular filter
is a readily available noninvasive technique can be percutaneously inserted into the in-
that is 95% sensitive for the diagnosis of ferior vena cava to prevent emboli from
symptomatic DVT in a proximal vein but reaching the lungs.
10090-15_CH15.qxd 8/31/06 4:57 PM Page 368

368 Chapter Fifteen

Figure 15.10. Diagnostic imaging of deep venous thrombosis. A. Normal venogram. Contrast material was
injected into a foot vein and fills the leg veins in this radiograph. B. Venogram demonstrating extensive thrombo-
sis of the deep calf veins, popliteal vein, and superficial femoral vein. Arrow indicates a filling defect in the superfi-
cial femoral vein (which is actually a deep vein despite its name) owing to the presence of thrombus. The deep calf
veins are filled with thrombus and cannot be visualized. C. Ultrasound indicating deep venous thrombosis. The
thrombus appears as an echogenic area (arrow) within the femoral vein (V). A healthy vein would be easily com-
pressible by the ultrasound transducer. This vein, however, has the same diameter at baseline (top panel) and after
compression (bottom panel), confirming the presence of thrombus within it. Art, artery.

Treatment of patients with calf vein termittent external pneumatic compression


thrombosis is more controversial because of the legs to prevent venous stasis.
pulmonary emboli from that site are un-
common. Some physicians advocate serial
Superficial Thrombophlebitis
noninvasive monitoring to determine if
the thrombus propagates into proximal Superficial thrombophlebitis is a benign dis-
veins, whereas others treat such thrombo- order associated with inflammation and
sis with heparin (unfractionated or low thrombosis of a superficial vein, just below
molecular weight) followed by warfarin for the skin. It may occur, for example, as a
3 to 6 months. complication of an in-dwelling intravenous
Prophylaxis against DVT is mandatory in catheter. It is characterized by erythema,
clinical situations in which the risk of de- tenderness, and edema over the involved
veloping the condition is high, such as dur- vein. Treatment consists of local heat and
ing bed rest following surgery. Prophylactic rest of the involved extremity. Aspirin or
measures include subcutaneous unfraction- other anti-inflammatory medications may
ated heparin or low molecular weight heparin relieve the associated discomfort. Unlike
(fondaparinux; see Chapter 17), low-dose oral DVT, superficial thrombophlebitis does not
warfarin, compression stockings, and/or in- lead to pulmonary embolism.
10090-15_CH15.qxd 8/31/06 4:57 PM Page 369

Diseases of the Peripheral Vasculature 369

SUMMARY intracardiac shunt (e.g., atrial septal de-


fect). Therapeutic options include anti-
1. Aortic aneurysms are of two types: coagulation, thrombolysis, and surgical
true aneurysms and false (pseudo) aneu- or endovascular interventions.
rysms. True aneurysms are caused by de-
6. Vasculitic syndromes are inflammatory
generative changes in the aortic wall.
diseases of blood vessels that impair ar-
Cystic medial necrosis is associated with
terial flow and result in localized and
ascending thoracic aortic aneurysms,
systemic symptoms. They are distin-
and atherosclerosis is commonly found
guished from one another by the pat-
in descending thoracic and abdominal
tern of vessel involvement and mor-
aortic aneurysms. A false aneurysm rep-
phologic findings (see Table 15.5).
resents a hole in the arterial intima and
media contained by a layer of adventitia 7. Raynaud phenomenon is an episodic
or perivascular clot. vasospasm of arteries that supply the dig-
its of the upper and lower extremities. It
2. Symptoms of aortic aneurysms relate to
may be a primary condition (Raynaud
compression of adjacent structures (back
disease) or may appear in association
pain, dysphagia, respiratory symptoms)
with other disorders such as connective
or blood leakage. The most severe
tissue diseases or blood dyscrasias.
consequence is aneurysm rupture. An
aneurysm can be repaired either with 8. Varicose veins are dilated tortuous ves-
an open surgical procedure or by inser- sels that may present cosmetic prob-
tion of an endovascular graft. lems. Occasionally, they cause discom-
fort, become thrombosed, or lead to
3. An aortic dissection results from a split-
venous insufficiency. Initial manage-
ting apart of a weakened medial layer
ment is conservative, with periodic leg
of the aorta, often in the setting of
elevation and compression stockings.
advanced age, hypertension, or other
Severe symptomatic varicose veins can
causes of medial degeneration (cystic
be treated with radiofrequency ablation,
medial necrosis). Type A (proximal) aor-
laser therapy, or surgical ligation and
tic dissections involve the ascending
removal.
aorta, whereas type B dissections are
confined to the descending aorta. The 9. Venous thrombosis results from stasis of
former are more common, more devas- blood flow, hypercoagulability, and vas-
tating, and require surgical treatment. cular damage. The major complication of
Type B dissections are often managed by deep venous thrombosis is pulmonary
embolism. A chronic complication is
medical therapy alone.
venous insufficiency causing chronic leg
4. PAD is a common atherosclerotic disease
swelling and skin ulceration.
of large and medium-sized arteries, often
10. D-dimer assay and venous compression
resulting in claudication of the limbs. It
ultrasonography are the primary tools
is treated by risk factor modification,
used to diagnose deep venous throm-
exercise, antiplatelet agents, and some-
bosis. Anticoagulation therapy with
times cilostazol, a selective phospho-
low molecular weight heparin or un-
diesterase inhibitor. Catheter-based or
fractionated intravenous heparin, fol-
surgical revascularization procedures are
lowed by oral warfarin, is the usual
implemented in patients with disabling
treatment.
symptoms or critical limb ischemia.
5. Acute arterial occlusion results from
thrombus formation in situ or from ar- Acknowledgment
terial embolism. The latter arises from
Contributors to the previous editions of this chapter
thrombus within the heart, from proxi- were Michael Diminick, MD; Mary Beth Gordon,
mal arterial sites, or paradoxically from MD; Stuart Kaplan, MD; Geoffrey McDonough, MD;
the systemic veins in the presence of an Jesse Salmeron, MD; and Mark A. Creager, MD.
10090-15_CH15.qxd 8/31/06 4:57 PM Page 370

370 Chapter Fifteen

Additional Reading Hirsch AT, Haskal ZJ, Hertzer NR, et al. ACC/AHA
2005 guidelines for the management of patients
Bates SM, Ginsburg JS. Treatment of deep vein throm- with peripheral arterial disease (lower extremity,
bosis. N Engl J Med 2004;351:268–277. renal, mesenteric, and abdominal aortic). J Am
Creager MA, ed. Management of Peripheral Arterial Coll Cardiol 2006;47:1239–1312. Available at:
Disease: Medical, Surgical and Interventional http://www.cardiosource.com/guidelines/guide-
Aspects. London: Remedica, 2000. lines/pad/pad_execsum.pdf. Accessed July 25, 2006.
Creager MA, Dzau VJ. Vascular diseases of the ex- Isselbacher EM. Thoracic and abdominal aortic
tremities. In: Braunwald E, Fauci AS, Kasper D, aneurysms. Circulation 2005;111:816–828.
et al., eds. Harrison’s Principles of Internal Medi- Marso SP, Hiatt WR. Peripheral arterial disease in pa-
cine. 16th Ed. New York: McGraw-Hill, 2005. tients with diabetes. J Am Coll Cardiol 2006;47:
Creager MA, Dzau VJ, Loscalzo J, eds. Vascular Med- 921–929.
icine: A Companion to Braunwald’s Heart Disease. Olin JW. Thromboangiitis obliterans (Buerger’s dis-
Philadelphia: Elsevier Saunders, 2006. ease). N Engl J Med 2000;343:864–869.
Dzau VJ, Creager MA. Diseases of the aorta. In: Smith SC, Allen J, Blair SN, et al. AHA/ACC guide-
Braunwald E, Fauci AS, Kasper D, et al., eds. Harri- lines for secondary prevention for patients with
son’s Principles of Internal Medicine. 16th Ed. coronary and other atherosclerotic vascular dis-
New York: McGraw-Hill, 2005. ease: 2006 update. J Am Coll Cardiol 2006;47:
Faxon DP, Creager MA, Smith SC Jr, et al. Athero- 2130–2139.
sclerotic Vascular Disease Conference executive Tsai TT, Nienaber CA, Eagle KA. Acute aortic syn-
summary. Circulation 2004;109:2595–2604. dromes. Circulation 2005;112:3802–3813.
Hankey GJ, Norman PE, Eikelboom JW. Medical treat- Wigley FM. Raynaud’s phenomenon. N Engl J Med
ment of peripheral arterial disease, JAMA 2006; 2002;347:1001–1008.
295:547–553.
10090-15_CH15.qxd 8/31/06 4:57 PM Page a

Chapter 15—Author Queries


1. AU: Is “and” correct here, or should it be “or”?
2. AU: Is this the correct term?
3. AU: Please provide page number.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 371

C H A P T E R

Congenital Heart
Disease
Vijay G. Sankaran
16
David W. Brown

NORMAL DEVELOPMENT OF COMMON CONGENITAL HEART LESIONS


THE CARDIOVASCULAR SYSTEM Acyanotic Lesions
Development of the Heart Tube Cyanotic Lesions
Formation of the Heart Loop
EISENMENGER SYNDROME
Septation
Development of the Cardiac Valves
FETAL AND TRANSITIONAL CIRCULATIONS
Fetal Circulation
Transitional Circulation

Congenital heart diseases are the most com- eases and substantial improvements in the
mon form of birth defects and are the lead- ability to evaluate and treat those afflicted.
ing cause of death from birth abnormalities Research has shown that genetic mutations,
in the first year of life. These conditions af- environmental factors, or maternal illness
fect approximately 8 of 1,000 live births, or ingestion of toxins can contribute to car-
and an estimated 1 million people in the diac malformations. However, specific eti-
United States have congenital heart lesions. ologies remain unknown in most cases.
Some abnormalities are severe and require The survival of children with congenital
immediate medical attention, whereas many heart disease has also improved dramati-
are less pronounced and have minimal clin- cally in recent decades, largely because of
ical consequences. Although congenital heart better diagnostic and interventional tech-
defects are present at birth, milder defects niques. However, the lifelong needs of af-
may remain inapparent for weeks, months, fected patients include guidance regarding
or years and, not infrequently, escape detec- physical activity, pregnancy, endocarditis
tion until adulthood. prophylaxis, insurance, and employment.
The past half century has witnessed tre- Formation of the cardiovascular system be-
mendous growth in the understanding of gins during the third week of embryonic de-
the pathophysiology of congenital heart dis- velopment. Soon after, a unique circulation

371
10090-16_CH16.qxd 8/31/06 4:57 PM Page 372

372 Chapter Sixteen

develops that allows the fetus to mature in


the uterus, using the placenta as the primary
organ of gas, nutrient, and waste exchange.
At birth, the fetal lungs inflate and become Dorsal aorta
functional, making the placenta unneces-
sary and dramatically altering circulation
patterns to allow the neonate to adjust to A
life outside the womb. Given the remarkable
complexity of these processes, it is easy to Endocardial heart tube
envision ways that cardiovascular malfunc-
tions could develop.
This chapter begins with an overview of
fetal cardiovascular development and then
describes the most common forms of con-
genital heart disease.
Foregut

NORMAL DEVELOPMENT OF
THE CARDIOVASCULAR SYSTEM
During the third week of gestation, the nu-
trient and gas exchange needs of the rapidly
B
growing embryo can no longer be met by Fusing endocardial heart tubes
diffusion alone, and the tissues begin to rely
on the developing cardiovascular system to
deliver these substances over long distances.

Development of the Heart Tube


In the middle of the third week of embryo-
genesis, mesodermal cells proliferate at the
cranial end of the early embryonic disc. They Foregut
eventually form two longitudinal cell clusters
known as angioblastic cords. These cords
Pericardial
canalize and become paired endothelial heart
cavity
Fig. 1 tubes (Fig. 16.1). Lateral embryonic folding
gradually causes these two tubes to oppose
one another and allows them to fuse in the
ventral midline, forming a single endocardial
C
tube by day 22. From inside to outside, the Endocardial heart tube
layers of this primitive heart tube are an en- Figure 16.1. Embryonic transverse sections illustrat-
dothelial lining that becomes the endo- ing fusion of the two heart tubes into a single endo-
cardium, a layer of gelatinous connective tis- cardial heart tube. A. 18 days. B. 21 days. C. 22 days.
sue (cardiac jelly), and a thick muscular layer
that is derived from the splanchnic meso-
derm and develops into the myocardium. oping cardiac area eventually becomes the
The endocardial tube is continuous with the pericardial cavity, housing the future heart.
aortic arch system rostrally and with the ve-
nous system caudally. The primitive heart be-
Formation of the Heart Loop
gins to beat around day 22 or 23, causing
blood to circulate by the beginning of the As the tubular heart grows and elongates, it
fourth week. The space overlying the devel- develops a series of alternate constrictions
10090-16_CH16.qxd 8/31/06 4:57 PM Page 373

Congenital Heart Disease 373

and dilations, creating the first sign of the sixth weeks. Although these events are de-
primitive heart chambers—the truncus arte- scribed separately here, they actually occur si-
riosus, the bulbus cordis, the primitive ven- multaneously.
tricle, the primitive atrium, and the sinus
Fig. 2 venosus (Fig. 16.2). Continued growth and Septation of the Atria
elongation within the confined pericardial
cavity force the heart tube to bend on itself The primary atrial septum, also known as
on day 23, eventually forming a U-shaped the septum primum, begins as a ridge of
loop with the round end pointing ventrally tissue on the roof of the common atrium
and to the right by day 28. The result of this that grows downward into the atrial cavity
looping is placement of the atrium and (Fig. 16.4). As the septum primum advances, Fig. 4
sinus venosus above and behind the truncus it leaves a large opening known as the os-
arteriosus, bulbus cordis, and ventricle (Fig. tium primum between the crescent-shaped
Fig. 3 16.3). At this point, neither definitive septa leading edge of the septum and the endo-
between the developing chambers nor de- cardial cushions surrounding the AV canal.
finitive valvular tissue have formed. The The ostium primum allows passage of blood
connection between the primitive atrium between the forming atria. Eventually, the
and ventricle is termed the atrioventricular septum primum fuses with the superior as-
(AV) canal. In time, the AV canal becomes pect of the endocardial cushions (described
two separate canals, one housing the tricus- in more detail in the next section), obliter-
pid valve and the other the mitral valve. The ating the ostium primum. However, before
sinus venosus is eventually incorporated closure of the ostium primum is complete,
into the right atrium, forming both the small perforations appear in the center of
coronary sinus and a portion of the right the septum primum that ultimately coa-
atrial wall. The bulbus cordis and truncus ar- lesce to form the ostium secundum, pre-
teriosus contribute to the future ventricular serving a pathway for blood flow between
outflow tracts, forming parts of the proxi- the atria (see Fig. 16.4). Following closure of
mal aorta and pulmonary artery. the ostium primum, a second, more muscu-
lar membrane, the septum secundum, be-
gins to develop immediately to the right of
Septation
the superior aspect of the septum primum.
Septation of the developing atrium, AV canal, This septum grows downward and overlaps
and ventricle occurs between the fourth and the ostium secundum. The septum secun-

[Right and left atria]

Figure 16.2. The straight heart tube at approximately 22 days. The structures that will
ultimately form from each segment are listed in brackets.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 374

374 Chapter Sixteen

A B C
Truncus
arteriosus
Truncus
arteriosus Bulbus Primitive Primitive
cordis right left
atrium atrium
Primitive Conus cordis
Bulboventricular Primitive
atrium
sulcus right Primitive
ventricle left
ventricle
Figure 16.3. Formation of the heart loop. A, B. By day 24, continued growth and elongation within the confined
pericardial space necessitate bending of the heart tube on itself, forming a U-shaped loop that points ventrally and
to the right. C. Looping eventually places the atria above and behind the primitive ventricles.

dum eventually fuses with the endocardial the valve to close, as described later in the
cushions, although only in a partial fashion, chapter.
leaving an oval-shaped opening known as
the foramen ovale. The superior edge of the
Septation of the Atrioventricular Canal
septum primum then gradually regresses,
leaving the lower edge to act as a “flaplike” Growth of the endocardial cushions con-
valve that allows only right-to-left flow tributes to atrial septation and, as described
Fig. 5 through the foramen ovale (Fig. 16.5). Dur- later, to the membranous portion of the in-
ing gestation, blood passes from the right terventricular septum. Endocardial cushions
atrium to the left atrium because the pres- initially begin as swellings of the gelatinous
sure in the fetal right atrium is greater than connective tissue layer within the AV canal.
that of the left atrium. This pressure gradi- They are then populated by migrating cells
ent changes direction postnatally, causing from the primitive endocardium and subse-

Septum
Secundum Ostium
Secundum
Septum
Ostium Primum
Primum
Endocardial Interventricular
Cushion Foramen
A B

Septum Ostium
Secundum Secundum
Septum Foramen
Secundum Septum
Ovale Primum
Septum
Interventricular Interventricular
Primum
Foramen Septum
Interventricular Membranous
Septum
C Muscular D
Figure 16.4. Atrial septal formation at 30 days (A), 33 days (B), and 37 days (C) of develop-
ment as well as in the newborn (D). As the septum primum grows toward the ventricles, the
opening between it and the AV canal is the ostium primum. Before the ostium primum completely
closes, perforations within the upper portion of the septum primum form the ostium secundum. A
second ridge of tissue, the septum secundum, grows downward to the right of the septum primum,
partially covering the ostium secundum. The foramen ovale is an opening of the septum secundum
that is covered by the “flap valve” of the lower septum primum. (Modified from Moss AJ, Adams FH.
Heart Disease in Infants, Children, and Adolescents. Baltimore: Williams & Wilkins, 1968:16.) AQ2
10090-16_CH16.qxd 8/31/06 4:57 PM Page 375

Congenital Heart Disease 375

A right and left canals that later give rise to the


tricuspid and mitral orifices, respectively.

Septation of the Ventricles and


Shunt
Ventricular Outflow Tracts
At the end of the fourth week, the primitive
ventricle begins to grow, leaving a median
muscular ridge, the primitive interventricular
septum. Most of the early increase in height
of the septum results from dilation of the two
new ventricles forming on either side of it.
Only later does new cell growth in the sep-
B tum itself contribute to its size. The free edge
of the muscular interventricular septum does
not fuse with the endocardial cushions; the
opening that remains and allows communi-
cation between the right and left ventricles is
the interventricular foramen (Fig. 16.7). This Fig. 7
remains open until the end of the seventh
week of gestation, when the fusion of tissue
from the right and left bulbar ridges and the
Figure 16.5. Diagrammatic depiction of the flap- endocardial cushions forms the membranous
type valve of the foramen ovale. A. Before birth, the portion of the interventricular septum.
valve permits only right-to-left flow of blood from the During the fifth week, neural crest-
higher-pressured right atrium (RA) to the lower-pressured
left atrium (LA). B. Following birth, the pressure in the LA derived mesenchymal proliferation occur-
becomes greater than that in the RA, causing the septum ring in the bulbus cordis and truncus arte-
primum to close firmly against the septum secundum. riosus creates a pair of protrusions known as
(Modified from Moore KL, Persaud TVN. The Developing
AQ3 Human. Philadelphia: WB Saunders, 1993:318.) the bulbar ridges (Fig. 16.8). These ridges Fig. 8
fuse in the midline and undergo a 180° spi-
raling process, forming the aorticopulmonary
quently transform into mesenchymal tissue. septum. This septum divides the bulbus cor-
Tissue growth occurs primarily in the hori- dis and the truncus arteriosus into two arte-
zontal plane, resulting in septation of the AV rial channels, the pulmonary artery and the
canal through the continued growth of the aorta, the former continuous with the right
lateral, superior, and inferior endocardial ventricle (RV) and the latter with the left
Fig. 6 cushions (Fig. 16.6). Septation creates the ventricle (LV).

Superior Left
endocardial atrioventricular
cushion canal

Lateral Lateral
endocardial endocardial
cushion cushion
Common Inferior Right
atrioventricular endocardial atrioventricular
canal cushion canal

Figure 16.6. The progression of septal formation in the atrioventricular canal through successive stages.
The septum forms through growth of the superior, inferior, and lateral endocardial cushions. The endocardial cush-
ions are masses of mesenchymal tissue that surround the atrioventricular canal and aid in the formation of the ori-
fices of the mitral and tricuspid valves, as well as the upper interventricular septum and lower interatrial septum.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 376

376 Chapter Sixteen

Aorta Pulmonary
artery

Left bulbar
ridge

Interventricular Right bulbar Interventricular


foramen ridge foramen
Muscular
Muscular
part of
interventricular interventricular
septum septum
Fused endocardial
A cushions

Figure 16.7. The interventricular septum and the in-


terventricular foramen. (Modified from Moore KL, Per-
saud TVN. The Developing Human. Philadelphia: WB
Saunders, 1993:325.)

Development of
the Cardiac Valves
Semilunar Valve Development Right atrio- Interventricular
(Aortic and Pulmonary Valves) ventricular foramen
canal Left atrio-
The semilunar valves start to develop just ventricular
canal
before the completion of the aorticopul-
monary septum. The process begins when B Endocardial cushion
three outgrowths of subendocardial mes-
enchymal tissue form around both the aor-
tic and pulmonary orifices. These growths
are ultimately shaped and excavated by the
joint action of programmed cell death and Pulmonary
artery
blood flow to create the three thin-walled
cusps of both the aortic and pulmonary Aorticopulmonary
valves. septum
Right
ventricle

Atrioventricular Valve Development Membranous Muscular


(Mitral and Tricuspid Valves) part of part of
interventricular interventricular
septum septum
After the endocardial cushions fuse to form
the septa between the right and left AV canals, C
the surrounding subendocardial mesen-
Figure 16.8. Formation of the aorticopulmonary sep-
chymal tissue proliferates and develops out- tum occurs via fusion of the bulbar ridges, resulting
growths similar to those of the semilunar in division of the bulbus cordis and truncus arterio-
valves. These are also sculpted by pro- sus into the aorta and pulmonary artery (A, 5 weeks;
B, 6 weeks; C, 7 weeks). The bulbus cordis becomes the
grammed cell death that occurs within the right ventricular outflow tract. Fusion of tissue from the
inferior surface of the nascent leaflets and in endocardial cushions, the aorticopulmonary septum, and
the ventricular wall. This process leaves be- the muscular interventricular septum creates the mem-
branous interventricular septum. (Modified from Moore
hind only a few fine muscular strands to KL, Persaud TVN. The Developing Human. Philadelphia:
connect the valves to the ventricular wall WB Saunders, 1993:322.)
Fig. 9 (Fig. 16.9). The superior portions of these
strands eventually degenerate and are re-
placed by strings of dense connective tissue,
becoming the chordae tendineae.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 377

Congenital Heart Disease 377

FETAL AND TRANSITIONAL


CIRCULATIONS
The fetal circulation elegantly serves the
needs of in utero development. At birth, the
circulation automatically undergoes modifi-
Interventricular cations that establish the normal blood flow
foramen
pattern of a newborn infant.
Lumen of left
ventricle
Fetal Circulation
A In fetal life, oxygenated blood leaves the
placenta through the umbilical vein (Fig. Fig. 10
16.10). Approximately half of this blood is
shunted through the fetal ductus venosus,
bypassing the hepatic vasculature, and pro-
ceeding directly into the inferior vena cava
(IVC). The remaining blood passes through
Developing mitral the portal vein to the liver and then into the
valve IVC through the hepatic veins. IVC blood is
therefore a mixture of well-oxygenated um-
Membranous part
of interventricular bilical venous blood and the blood of low
septum oxygen tension returning from the systemic
veins of the fetus. Because of this mixture,
Trabeculae the oxygen tension of inferior vena caval
B carneae
blood is higher than that of blood returning
to the fetal right atrium from the superior
vena cava. This distinction is important be-
cause these two streams of blood are partially
Atrioventricular node
separated within the right atrium to follow
different circulatory paths. The consequence
Mitral valve
of this separation is that the fetal brain
and myocardium receive blood of relatively
Chordae tendineae higher oxygen content, whereas the more
poorly oxygenated blood is diverted to the
placenta (via the descending aorta and um-
Papillary muscle bilical arteries) for subsequent oxygenation.
Ventricular conduction Most IVC blood entering the right atrium
C system
is directed to the left atrium through the
Figure 16.9. Proliferation of mesenchymal tissue sur- foramen ovale. This intracardiac shunt of
rounding the atrioventricular canals forms the atri- relatively well-oxygenated blood is facili-
AQ4 oventricular valves. Degeneration of myocardium and
replacement by connective tissue forms the chordae tated by the inferior border of the septum se-
tendineae; their muscular attachments to the ventricular cundum, termed the crista dividens, which
wall are the papillary muscles. (Modified from Moore KL, is positioned such that it overrides the open-
Persaud TVN. The Developing Human. Philadelphia: WB
Saunders, 1993:325.) ing of the IVC into the right atrium. This
shunted blood then mixes with the small
amount of poorly oxygenated blood return-
ing to the left atrium through the fetal pul-
monary veins (remember that the lungs are
not ventilated in utero; the developing pul-
monary tissues actually remove oxygen from
the blood). From the left atrium, blood flows
10090-16_CH16.qxd 8/31/06 4:57 PM Page 378

378 Chapter Sixteen

Superior vena cava Arch of aorta


Ductus arteriosus

Lung Pulmonary trunk


Pulmonary veins

Foramen ovale Left atrium

Right atrium

Inferior vena cava


Left hepatic vein
Right hepatic
vein
Ductus venosus

Descending aorta

inus
tal s Sphincter
Por

Portal vein
Umbilical vein
Umbilicus Oxygen saturation
Urinary of blood:
bladder
High
Umbilical Medium
arteries
Low

Placenta Legs Internal iliac


artery
Figure 16.10. The fetal circulation. Arrows indicate the direction of blood flow. Three shunts
(ductus venosus, foramen ovale, and ductus arteriosus) allow most of the blood to bypass the lungs
and liver during fetal life but cease to function shortly after birth. (Modified from Moore KL,
Persaud TVN. The Developing Human. Philadelphia: WB Saunders, 1993:344.)

into the LV and is then pumped into the as- The remaining well-oxygenated inferior
cending aorta. This well-oxygenated blood vena caval blood entering the right atrium
is distributed primarily to three territories: mixes with poorly oxygenated blood from
(1) approximately 9% enters the coronary the superior vena cava and passes to the RV.
arteries and perfuses the myocardium, In the fetus, the RV is the actual “work-
(2) 62% travels in the carotid and subclavian horse” of the heart, providing two thirds of
vessels to the upper body and brain, and the total cardiac output. This output flows
(3) 29% passes into the descending aorta to into the pulmonary artery and from there
the rest of the fetal body. either through the ductus arteriosus into
10090-16_CH16.qxd 8/31/06 4:57 PM Page 379

Congenital Heart Disease 379

the descending aorta (88% of RV output) or creases, causing left atrial pressure to rise. At
through the pulmonary arteries and into the the same time, cessation of umbilical ve-
lungs (12% of RV output). This unequal dis- nous flow and constriction of the ductus
tribution of right ventricular outflow is ac- venosus cause a fall in IVC and right atrial
tually quite efficient. Bypassing the lungs is pressures. As a result, the left atrial pressure
desired because the fetal lungs are filled with becomes greater than that in the right
amniotic fluid and are incapable of gas ex- atrium, and the valve of the foramen ovale
change. The low oxygen tension of this fluid is forced against the septum secundum,
causes constriction of the pulmonary ves- eliminating the previous flow between the
sels, which increases pulmonary vascular re- atria (see Fig. 16.5).
sistance and facilitates shunting of blood With oxygenation now occurring in the
through the ductus arteriosus to the systemic newborn lungs, the ductus arteriosus be-
circulation. From the descending aorta, blood comes superfluous and begins to constrict.
is distributed to the lower body and to the During fetal life, a high circulating level of
umbilical arteries, leading back to the pla- prostaglandin E1 (PGE1) is generated in re-
centa for gas exchange. sponse to relative hypoxia, which causes the
smooth muscle of the ductus arteriosus to
relax, keeping it patent. After birth, PGE1
Transitional Circulation
levels decline as the oxygen tension rises
Immediately following birth, the neonate and the ductus constricts. The responsive-
rapidly adjusts to life outside the womb. The ness of the ductus to vasoactive substances
newly functioning lungs replace the pla- depends on the gestational age of the fetus.
centa as the organ of gas exchange, and the The ductus often fails to constrict in prema-
three shunts (ductus venosus, foramen ture infants, resulting in the congenital
ovale, and ductus arteriosus) that operated anomaly known as patent ductus arteriosus
during gestation close. This shift in the site (described later in the chapter).
of gas exchange and the resulting changes With the anatomic separation of the cir-
in cardiovascular architecture allow the culatory paths of the right and left sides of
newborn to survive independently. the heart now complete, the stroke volume
As the umbilical cord is clamped or con- of the LV increases and that of the RV de-
stricts naturally, the low-resistance placen- creases, equalizing the cardiac output from
tal flow is removed from the arterial system, both ventricles. The augmented pressure
resulting in an increase in systemic vascular and volume load placed on the LV induces
resistance. Simultaneously, pulmonary vas- the myocardial cells of that chamber to
cular resistance falls for two reasons: (1) the hypertrophy, while the decreased pressure
mechanical inflation of the lungs after birth and volume loads on the RV result in grad-
stretches the lung tissues, causing pulmonary ual regression of RV wall thickness.
artery expansion and wall thinning, and
(2) vasodilatation of the pulmonary vascula-
COMMON CONGENITAL
ture occurs in response to the rise in blood
HEART LESIONS
oxygen tension accompanying aeration of
the lungs. This reduction in pulmonary re- Congenital heart defects are generally well
sistance results in a dramatic rise in pul- tolerated before birth. The fetus benefits
monary blood flow. It is most marked within from shunting of blood through the ductus
the first day after birth but continues for the arteriosus and the foramen ovale, allowing
next several weeks until adult levels of pul- the bypass of most defects. It is only after
monary resistance are achieved. birth, when the neonate has been separated
As pulmonary resistance falls and more from the maternal circulation and the oxy-
blood travels to the lungs through the pul- genation it provides and the fetal shunts
monary artery, venous return from the pul- have closed, that congenital heart defects
monary veins to the left atrium also in- usually become manifest.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 380

380 Chapter Sixteen

Congenital heart lesions can be catego- flow to supervene, accompanied by the


rized as cyanotic or acyanotic. Cyanosis refers physical findings of hypoxemia and cyano-
to a blue-purple discoloration of the skin sis. The development of pulmonary vascular
and mucous membranes caused by an ele- disease as a result of a chronic large left-to-
vated blood concentration of deoxygenated right shunt is known as Eisenmenger syn-
hemoglobin (at least 4 g/dL, which corre- drome and is described in greater detail in
sponds to an arterial O2 saturation of ap- the final section of the chapter.
proximately 80% to 85%). In congenital
heart disease, cyanosis results from defects Acyanotic Lesions
that allow poorly oxygenated blood from the
Atrial Septal Defect
right side of the heart to be shunted to the
left side, bypassing the lungs. An atrial septal defect (ASD) is a persistent
Acyanotic lesions include intracardiac or opening in the interatrial septum after birth
vascular stenoses, valvular regurgitation, that allows direct communication between
and defects that result in left-to-right shunt- the left and right atria. ASDs are relatively
ing of blood. Large left-to-right shunts at the common, occurring with an incidence of 1
atrial, ventricular, or great vessel level (all in 1,500 live births. They can occur any-
described in the following sections) cause where along the atrial septum, but the most
the pulmonary artery volume and pressure common site is at the region of the foramen
to increase and can be associated with the ovale, termed an ostium secundum ASD (Fig. Fig. 11
later development of pulmonary arteriolar 16.11). This defect arises from excessive re-
hypertrophy and increased resistance to sorption or inadequate development of the
flow. Over time, the elevated pulmonary septum primum, inadequate formation of
resistance may force the direction of the the septum secundum, or a combination.
original shunt to reverse, causing right-to-left Less commonly, an ASD appears in the infe-

ASD

Figure 16.11. Atrial septal defect (ASD), ostium secundum type. A. The arrow indicates shunted
flow from the left atrium (LA) toward the right atrium (RA). B. Schematic representation of blood flow
through an uncomplicated ASD, resulting in enlargement of the RA, right ventricle (RV), and pulmonary
artery (PA). Ao, aorta; IVC, inferior vena cava; LV, left ventricle; SVC, superior vena cava.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 381

Congenital Heart Disease 381

rior portion of the interatrial septum, adja- size and the filling properties (compliance)
cent to the AV valves. Named an ostium pri- of the ventricles into which the atria pass
mum defect, this abnormality results from their contents. Normally after birth, right
failure of the septum primum to fuse with ventricular compliance becomes greater
the endocardial cushions and is typically as- than that of the LV owing to the regression
sociated with abnormal development of the of right ventricular wall thickness, facilitat-
mitral and tricuspid valves. A third type of ing a left-to-right directed shunt. The result
ASD occurs in the superior portion of the is volume overload and enlargement of the
atrial septum near the entry of the superior right atrium and RV (see Fig. 16.11B). If
vena cava and is termed a sinus venosus ASD. right ventricular compliance decreases over
It results from incomplete absorption of the time (because of the excessive load), the left-
sinus venosus into the right atrium and is to-right shunt may lessen. Occasionally, if
often accompanied by the anomalous drain- severe pulmonary vascular disease develops
age of pulmonary veins from the right lung (e.g., Eisenmenger syndrome), the direction
into the right atrium. of the shunt may actually reverse (causing
In contrast, a patent foramen ovale (PFO), right-to-left flow), such that desaturated
which is thought to be present in approxi- blood enters the systemic circulation, re-
mately 20% of the general population, is not sulting in hypoxemia and cyanosis.
a true ASD. As described earlier, the foramen
ovale functionally shuts in the days after
Symptoms
birth, and it anatomically closes by the age
of 6 months through fusion of the atrial Most infants with ASDs are asymptomatic.
septa. PFOs arise this fusion fails to occur. The condition is typically detected by the
A PFO is usually clinically silent because presence of a murmur on routine physical
the one-way valve, though not sealed, re- examination during childhood or adoles-
mains functionally closed because the LA cence. If symptoms do occur, they include
pressure is higher compared with that in dyspnea on exertion, fatigue, and recurrent
the right atrium. However, a PFO takes on lower respiratory tract infections. The most
added significance if the right atrial pressure common symptoms in adults are decreased
becomes elevated (e.g., in states of pulmo- stamina and palpitations owing to atrial
nary hypertension or right-heart failure), re- tachyarrhythmias resulting from right atrial
sulting in pathologic right-to-left intracardiac enlargement.
shunting. In that case, deoxygenated blood
passes directly into the arterial circulation.
Physical Examination
Occasionally, a PFO can be implicated in
a patient who has suffered a systemic em- A prominent systolic impulse may be pal-
bolism (e.g., a stroke). This situation, termed pated along the lower-left sternal border,
paradoxical embolism, occurs when throm- representing contraction of the dilated RV
bus in a systemic vein breaks loose, travels (termed RV heave). The second heart sound
to the right atrium, passes across the PFO to (S2) demonstrates a widened, fixed splitting
the left atrium (if right-to-left shunting is pattern (see Chapter 2), because the normal
present because of elevated right-heart pres- respiratory variation in systemic venous re-
sures), and then into the systemic arterial turn is countered by reciprocal changes in
circulation. the volume of blood shunted across the
ASD. The increased volume of blood flowing
across the pulmonary valve often creates a
Pathophysiology
systolic murmur at the upper-left sternal
In the case of an uncomplicated ASD, oxy- border. A middiastolic murmur may also be
genated blood from the left atrium is shunted present at the lower-left sternal border owing
into the right atrium, but not vice versa. to the increased flow across the tricuspid
Flow through the defect is a function of its valve. Blood traversing the ASD itself does
10090-16_CH16.qxd 8/31/06 4:57 PM Page 382

382 Chapter Sixteen

not produce a murmur because of the ab- sure or with a pericardial or synthetic patch.
sence of a significant pressure gradient be- In children and young adults, morphologic
tween the two atria. changes in the right heart often return to
normal after repair. Percutaneous ASD re-
pair, using a closure device deployed via an
Diagnostic Studies
intravenous catheter, is a less invasive alter-
On chest radiographs, the heart is usually native to surgery in some patients.
enlarged because of right atrial and right
ventricular dilatation, and the pulmonary
Ventricular Septal Defect
artery is prominent with increased pulmo-
nary vascular markings. The electrocardio- A ventricular septal defect (VSD) is an ab-
gram (ECG) shows right ventricular hyper- normal opening in the interventricular sep-
trophy, often with right atrial enlargement tum (Fig. 16.12). VSDs are relatively com- Fig. 12
and incomplete or complete right bundle mon, having an incidence of 1.5 to 3.5 per
branch block. In patients with the ostium 1,000 live births. They are most often lo-
primum type of ASD, left axis deviation is cated in the membranous (70%) and mus-
common and is thought to be a result of dis- cular (20%) portions of the septum, with
placement and hypoplasia of the left bundle few defects occurring just below the aortic
branch’s anterior fascicle. Echocardiography valve or adjacent to the AV valves.
demonstrates right atrial and right ventricu-
lar enlargement; the ASD may be visualized
Pathophysiology
directly, or its presence may be implied by
the demonstration of a transatrial shunt by The hemodynamic changes that accom-
Doppler flow interrogation. The magnitude pany VSDs depend on the size of the defect
and direction of shunt flow and an estima- and the relative resistances of the pulmonary
tion of right ventricular systolic pressure can and systemic vasculatures. In small VSDs,
be determined. the defect itself offers more resistance to
Given the high sensitivity of echocardio- flow than the pulmonary or systemic vascu-
graphy, it is rarely necessary to perform car- lature; thus, the magnitude of the shunt de-
diac catheterization to confirm the presence pends on the size of the hole. Conversely,
of an ASD. Catheterization may be useful to with larger “nonrestrictive” defects, the
assess the pulmonary vascular resistance volume of the shunt is determined by the
and to diagnose concurrent coronary artery relative pulmonary and systemic vascular
disease in older adults. In a normal person resistances. In the perinatal period, the pul-
undergoing cardiac catheterization, the oxy- monary vascular resistance approximates
gen saturation measured in the right atrium the systemic vascular resistance, and mini-
is similar to that in the superior vena cava. mal shunting occurs between the two ventri-
However, an ASD with left-to-right shunting cles. After birth, however, as the pulmonary
of well-oxygenated blood causes the satura- vascular resistance falls, an increasing left-
tion in the right atrium to be much greater to-right shunt through the defect develops.
than that of the superior vena cava. When this shunt is large, the RV, pulmonary
circulation, left atrium, and LV experience a
relative volume overload. Initially, the in-
Treatment
creased blood return to the LV augments
Most patients with ASDs remain asympto- stroke volume (via the Frank-Starling mech-
matic. However, if the volume of shunted anism); but over time, the increased volume
blood is large (even in the absence of symp- load can result in chamber dilatation, sys-
toms), elective surgical repair is recom- tolic dysfunction, and symptoms of heart
mended to prevent the development of failure. In addition, the augmented circula-
heart failure or pulmonary vascular disease. tion through the pulmonary vasculature can
The defect is repaired by direct suture clo- cause pulmonary vascular disease as early as
10090-16_CH16.qxd 8/31/06 4:57 PM Page 383

Congenital Heart Disease 383

VSD

Figure 16.12. Ventricular septal defect (VSD). A. The arrow indicates shunted flow from the left ven-
tricle (LV) toward the right ventricular (RV) outflow tract. B. Schematic representation of blood flow
through an uncomplicated VSD. The dashed lines represent increased blood return to the left side of the
heart as a result of the shunt, which causes enlargement primarily of the left atrium (LA) and LV. Ao, aorta;
IVC, inferior vena cava; PA, pulmonary artery; RA, right atrium; SVC, superior vena cava.

2 years of age. As pulmonary vascular resis- at the left sternal border. Smaller defects
tance increases, the intracardiac shunt may tend to have the loudest murmurs because
reverse its direction (Eisenmenger syndrome), of the great turbulence of flow that they
leading to systemic hypoxemia and cyanosis. cause. A systolic thrill can commonly be
palpated in the region of the murmur. In
addition, a middiastolic rumble can often
Symptoms
be heard at the apex owing to the increased
Patients with small VSDs typically remain flow across the mitral valve. If pulmonary
symptom free. Conversely, 10% of infants vascular disease develops, the holosystolic
with VSDs have large defects and will de- murmur diminishes as the pressure gra-
velop early symptoms of congestive heart dient across the defect decreases. In such
failure, including tachypnea, poor feeding, patients, an RV heave, a loud pulmonic
failure to thrive, and frequent lower respi- closure sound (P2), and cyanosis may be
ratory tract infections. Patients with VSDs evident.
complicated by pulmonary vascular disease
and reversed shunts may present with dysp-
Diagnostic Studies
nea and cyanosis. Bacterial endocarditis (see
Chapter 8) can develop, regardless of the On chest radiographs, the cardiac silhouette
size of the VSD. may be normal in patients with small de-
fects, but in those with large shunts, cardio-
megaly and prominent pulmonary vascular
Physical Examination
markings are present. If pulmonary vascular
The most common physical finding is a disease has developed, enlarged pulmonary
harsh holosystolic murmur that is best heard arteries with peripheral tapering may be
10090-16_CH16.qxd 8/31/06 4:57 PM Page 384

384 Chapter Sixteen

evident. The ECG shows left atrial enlarge- vascular disease but with significant volume
ment and left ventricular hypertrophy in overload can be corrected later in childhood.
those with a large shunt, and right ventricular Less invasive catheter-based treatments are
hypertrophy is usually evident if pulmonary still investigational. Medical management
vascular disease is present. Echocardiography includes endocarditis prophylaxis for all
with Doppler studies can accurately deter- patients with VSDs.
mine the location of the VSD, identify the
direction and magnitude of the shunt, and
Patent Ductus Arteriosus
provide an estimate of right ventricular sys-
tolic pressure. Cardiac catheterization demon- The ductus arteriosus is the vessel that con-
strates increased oxygen saturation in the nects the left pulmonary artery to the de-
RV compared with the right atrium, the re- scending aorta during fetal life. Patent duc-
sult of shunting of highly oxygenated blood tus arteriosus (PDA) results when the ductus
from the LV into the RV. fails to close after birth, resulting in a persis-
tent connection between the great vessels
(Fig. 16.13). It has an overall incidence of Fig. 13
Treatment
about 1 in 2,500 to 5,000 live term births.
By age 2, at least 50% of small and moderate- Risk factors for its presence include first
sized VSDs undergo sufficient partial or com- trimester maternal rubella infection, prema-
plete spontaneous closure to make inter- turity, and birth at a high altitude.
vention unnecessary. Surgical correction of
the defect is recommended in the first few
Pathophysiology
months of life for children with congestive
heart failure or pulmonary vascular disease. As described earlier, the smooth muscle of
Moderate-sized defects without pulmonary the ductus arteriosus usually constricts after

PDA

Figure 16.13. Patent ductus arteriosus (PDA). A. The arrow indicates shunted flow from the de-
scending aorta (Ao) toward the pulmonary artery (PA). B. Schematic representation of blood flow
through an uncomplicated PDA. The dashed lines represent increased blood return to the left side of
the heart as a result of the shunt, which causes enlargement of the left atrium (LA), left ventricle (LV),
and Ao. IVC, inferior vena cava; RA, right atrium; RV, right ventricle; SVC, superior vena cava.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 385

Congenital Heart Disease 385

birth owing to the sudden rise in blood oxy- Physical Examination


gen tension and a reduction in the level of
The most common finding in a patient with
circulating prostaglandins. Over the next
a left-to-right shunt through a PDA is a con-
several weeks, intimal proliferation and fi-
tinuous, machinelike murmur (see Fig. 2.10),
brosis result in permanent closure. Failure of
heard best at the left subclavicular region. The
the ductus to close results in a persistent
murmur is present throughout the cardiac
shunt between the descending aorta and the
cycle because a pressure gradient exists be-
left pulmonary artery. The magnitude of
tween the aorta and pulmonary artery in both
flow through the shunt depends on the
systole and diastole. However, if pulmonary
cross-sectional area and length of the ductus
vascular disease develops, the gradient be-
itself as well as the relative resistances of the tween the aorta and the pulmonary artery de-
systemic and pulmonary vasculatures. Pre- creases, leading to diminished flow through
natally, when the pulmonary vascular resis- the PDA, and the murmur becomes shorter
tance is high, the blood is diverted away (the diastolic component may disappear). If
from the immature lungs to the aorta. As Eisenmenger syndrome develops, lower ex-
the pulmonary resistance drops postna- tremity cyanosis and clubbing may be present
tally, the shunt reverses direction and blood on examination because poorly oxygenated
flows from the aorta into the pulmonary blood is shunted to the descending aorta.
circulation instead. Because of this left-to-
right shunt, the pulmonary circulation, left
atrium, and LV become volume overloaded. Diagnostic Studies
This can lead to left ventricular dilatation With a large PDA, the chest radiograph shows
and left-sided heart failure, whereas the an enlarged cardiac silhouette (left atrial and
right heart remains normal unless pulmo- left ventricular enlargement) with promi-
nary vascular disease ensues. If the latter nent pulmonary vascular markings. In adults,
does develop, Eisenmenger syndrome results, calcification of the ductus may be visual-
with reversal of the shunt causing blood to ized. The ECG shows left atrial enlargement
flow from the pulmonary artery, through and left ventricular hypertrophy when a
the ductus, to the descending aorta. The re- large shunt is present. Echocardiography with
sulting flow of desaturated blood to the Doppler imaging can visualize the defect,
lower extremities causes cyanosis of the feet; demonstrate flow through it, and estimate
the upper extremities are not cyanotic, be- right-sided systolic pressures. Cardiac catheter-
cause they receive normally saturated blood ization is usually unnecessary for diagnostic
from the proximal aorta. purposes. When performed in patients with
a left-to-right shunt, it demonstrates a step
up in oxygen saturation in the pulmonary
Symptoms
artery compared with the RV, and angio-
Children with small PDAs are generally graphy shows the abnormal flow of blood
asymptomatic. Those with large left-to- through the PDA.
right shunts develop early congestive heart
failure with tachycardia, poor feeding, slow
Treatment
growth, and recurrent lower respiratory
tract infections. Moderate-sized lesions can In the absence of other congenital cardiac
present with fatigue, dyspnea, and palpita- abnormalities or severe pulmonary vascular
tions in adolescence and adult life. Atrial disease, a PDA should generally be therapeu-
fibrillation may occur owing to left atrial tically occluded. Although many sponta-
dilatation. Turbulent blood flow across neously close during the first months after
the defect can set the stage for endovascu- birth, this rarely occurs later. Given the
lar infection, similar to endocarditis (see constant risk of endarteritis and the minimal
Chapter 8) but more accurately termed complications of corrective procedures, even
endarteritis. a small asymptomatic PDA is commonly re-
10090-16_CH16.qxd 8/31/06 4:57 PM Page 386

386 Chapter Sixteen

ferred for closure. For neonates and prema- stenotic opening through which blood is
ture infants with congestive heart failure, a ejected. Bicuspid aortic valves are common,
trial of prostaglandin synthesis inhibitors appearing in approximately 2% to 4% of the
(e.g., indomethacin) can be administered in population. Although they rarely result in
an attempt to constrict the ductus. Definitive congenital AS, they are a common cause of
closure can be accomplished by surgical divi- AS in adults because the leaflets fibrose and
sion or ligation of the ductus or by trans- calcify over time (see Chapter 8).
catheter techniques in which an occluding
coil or other vascular occlusion device is
Pathophysiology
placed.
Because the valvular orifice is significantly
narrowed, left ventricular systolic pressure
Congenital Aortic Stenosis
must increase to pump blood across the
Congenital aortic stenosis (AS) is most often valve into the aorta. In response to this in-
caused by abnormal development of the creased pressure load, the LV hypertrophies.
Fig. 14 valve (Fig. 16.14). It occurs in 5 of 10,000 The high-velocity jet of blood that passes
live births and is four times as common in through the stenotic valve may impact on
males as in females. Twenty percent of pa- the proximal aortic wall and contribute to
tients have an additional abnormality, most dilatation of that vessel.
commonly coarctation of the aorta (dis-
cussed later in the chapter). The aortic valve
Symptoms
in congenital AS usually has a bicuspid
leaflet structure instead of the normal three- The clinical picture of AS depends on the
leaflet configuration, causing an eccentric severity of the lesion. Fewer than 10% of in-

Figure 16.14. Congenital aortic valve stenosis. A. The arrow points to the narrowed aortic valve.
B. Schematic representation of obstructed flow through the narrowed aortic valve (jagged arrow). Left
ventricular (LV) hypertrophy results from the chronic increased pressure load. Poststenotic dilatation of
the aorta (Ao) is common. IVC, inferior vena cava; LA, left atrium; PA, pulmonary artery; RA, right
atrium; RV, right ventricle; SVC, superior vena cava.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 387

Congenital Heart Disease 387

fants experience symptoms of heart failure obstruction of the aortic valve during in-
before age 1, but if they do, they manifest fancy may mandate immediate surgical or
tachycardia, tachypnea, failure to thrive, transcatheter balloon valvuloplasty. Gen-
and poor feeding. Most older children with erally, valvuloplasty in infancy is only pal-
congenital AS are asymptomatic and de- liative and future additional catheter bal-
velop normally. When symptoms do occur, loon dilation or surgical revision is usually
they are similar to those of adult AS and in- needed.
clude becoming fatigued easily, exertional
dyspnea, angina pectoris, and syncope (see
Pulmonic Stenosis
Chapter 8).
Isolated pulmonic stenosis (Fig. 16.15) may Fig. 15
occur at the level of the pulmonic valve (e.g.,
Physical Examination
from congenitally fused valve commissures),
Auscultation reveals a harsh crescendo- within the body of the RV (obstruction in
decrescendo systolic murmur, loudest at the the RV outflow tract), or in the pulmonary
base of the heart with radiation to the neck. artery itself. Valve stenosis is the most com-
It is often preceded by a systolic ejection mon form (>90% of cases). Pulmonic steno-
click (see Chapter 2), especially when a bi- sis is also seen in 10% of patients with other
cuspid valve is present. Unlike the murmurs forms of congenital heart disease.
of ASD, VSD, or PDA, the murmur of con-
genital AS is characteristically present from
Pathophysiology
birth because it does not depend on a de-
cline in pulmonary vascular resistance. With The consequence of pulmonic stenosis is ob-
advanced disease, the ejection time becomes struction to right ventricular systolic ejec-
longer, causing the peak of the murmur to tion, which leads to increased right ventric-
occur later in systole. In severe disease, the ular pressures and chamber hypertrophy.
significantly prolonged ejection time The clinical course is determined by the
causes a delay in closure of the aortic valve severity of the obstruction. In the presence
such that A2 occurs after P2—a phenome- of normal cardiac output, a peak systolic
non known as reversed splitting of S2 (see transvalvular pressure gradient >50 mm Hg
Chapter 2). is considered mild pulmonic stenosis, be-
tween 50 and 80 mm Hg is moderate steno-
sis, and severe stenosis is defined by a peak
Diagnostic Studies
gradient >80 mm Hg.
The chest radiograph of an infant with AS
may show an enlarged LV and a dilated as-
Symptoms
cending aorta. The ECG often shows left
ventricular hypertrophy. Echocardiography Children with mild or moderate pulmonary
can identify the structure of the aortic valve stenosis are asymptomatic. The diagnosis is
and the degree of left ventricular hyper- often first made on discovery of a murmur
trophy. Doppler assessment can accurately during a routine physical examination. Se-
measure the pressure gradient across the vere stenosis may cause manifestations such
stenotic valve and allow calculation of the as dyspnea with exertion, exercise intoler-
valve area. Cardiac catheterization confirms ance, and with decompensation, symptoms
the pressure gradient across the valve. of right-sided heart failure such as abdomi-
nal fullness and pedal edema.
Treatment
Physical Examination
In its milder forms, AS does not need to
be corrected, but endocarditis prophylaxis The physical findings in pulmonic stenosis
should be followed (see Chapter 8). Severe depend on the severity of the obstruction. If
10090-16_CH16.qxd 8/31/06 4:57 PM Page 388

388 Chapter Sixteen

Figure 16.15. Congenital pulmonary valve stenosis. A. The arrow points to the narrowed pul-
monary valve. B. Schematic representation of obstructed flow through the narrowed pulmonary valve
(jagged arrow). Right ventricular hypertrophy results from the chronically increased pressure load.
Ao, aorta; IVC, inferior vena cava; LA, left atrium; LV, left ventricle; PA, pulmonary artery; RA, right
atrium; RV, right ventricle; SVC, superior vena cava.

the stenosis is severe with accompanying leaflets into the pulmonary artery prior to
right ventricular hypertrophy, a prominent RV contraction, preempting the rapid tens-
jugular venous a wave can be observed (see ing in early systole that is thought to pro-
Chapter 2) and an RV heave is palpated over duce the sound.
the sternum. A loud, late-peaking, crescendo-
decrescendo systolic ejection murmur is
Diagnostic Studies
heard at the upper-left sternal border, often
associated with a palpable thrill. Widened The chest radiograph may demonstrate an en-
splitting of the S2 with a soft P2 component is larged right atrium and ventricle with post-
caused by the delayed closure of the stenotic stenotic pulmonary artery dilation (caused
pulmonary valve. by the impact of the high-velocity jet of
In more moderate stenosis, a pulmonic blood against the wall of the pulmonary
ejection sound (a high-pitched “click”) fol- artery). The ECG shows right ventricular
lows S1 and precedes the systolic murmur. It hypertrophy and right axis deviation. Echo-
occurs during the early phase of right ven- cardiography with Doppler imaging assesses
tricular contraction as the stenotic valve the pulmonary valve morphology, deter-
leaflets suddenly reach their maximum level mines the presence of right ventricular
of ascent into the pulmonary artery, just be- hypertrophy, and accurately measures the
fore blood ejection. Unlike other sounds pressure gradient across the obstruction.
and murmurs produced by the right side of
the heart, the pulmonic ejection sound di-
Treatment
minishes in intensity during inspiration.
This occurs because with inspiration, the Mild pulmonic stenosis usually does not
augmented right-sided filling elevates the progress or require treatment. Moderate or
10090-16_CH16.qxd 8/31/06 4:57 PM Page 389

Congenital Heart Disease 389

severe valvular obstruction at the valvular


level can be relieved by dilating the stenotic
valve through transcatheter balloon valvu-
loplasty. Long-term results of this procedure
have been uniformly excellent, and right
ventricular hypertrophy usually regresses
subsequently. Antibiotic prophylaxis for en-
docarditis is required, however, even after
valvuloplasty.

Coarctation of the Aorta


Coarctation of the aorta typically consists of
a discrete narrowing of the aortic lumen.
This anomaly has an incidence of 1 in 6,000
live births and often occurs in patients with Figure 16.17. Magnetic resonance imaging of coarc-
Turner syndrome (45, XO). Two types of tation of the aorta. This lateral view demonstrates a
preductal coarctation, manifest as a focal aortic narrow-
coarctation are distinguished according to ing (white arrow). AA, ascending aorta; DA, descending
the location of the aortic narrowing in rela- aorta; LA, left atrium; RA, right atrium; TAA, transverse
tion to the ductus arteriosus: preductal (2%) aortic arch.
and postductal (98%), as shown in Figure
Fig. 16 16.16. Preductal coarctation, in which nar-
rowing occurs proximal to the ductus, re- ductal coarctation is most likely the result of
sults when an intracardiac anomaly during muscular ductal tissue that extends into the
fetal life decreases blood flow through the aorta during fetal life. When ductal tissue
left side of the heart, leading to hypoplastic constricts following birth, the ectopic tissue
Fig. 17 development of the aorta (Fig. 16.17). Post- within the aorta also constricts, creating an
obstruction.

Pathophysiology
In both types of coarctation, the LV faces an
Preductal
Aorta coarctation increased pressure load. Blood flow to the
head and upper extremities is preserved be-
Ductus cause the vessels supplying these areas usu-
Pulmonary arteriosus
ally branch off the aorta proximal to the ob-
artery
struction, but flow to the descending aorta
A and lower extremities may be diminished. If
coarctation is not corrected, compensatory
alterations include (1) development of left
ventricular hypertrophy and (2) dilatation
of collateral blood vessels from the inter-
costal arteries that bypass the coarctation
Aorta and provide blood to the descending aorta.
Postductal
coarctation Eventually, these collateral vessels enlarge
Pulmonary and can erode the undersurface of the ribs.
artery

Constricted ductus Symptoms


B
Figure 16.16. Coarctation of the aorta. A. Preductal Patients with preductal and severe postduc-
coarctation. B. Postductal coarctation. tal coarctation usually present very shortly
10090-16_CH16.qxd 8/31/06 4:57 PM Page 390

390 Chapter Sixteen

after birth with symptoms of heart failure. ized. The ECG shows left ventricular hyper-
Infants with preductal coarctation may also trophy resulting from the pressure load
exhibit differential cyanosis if the ductus placed on that chamber. Doppler echocardio-
arteriosus remains open. The upper half graphy confirms the diagnosis of coarctation
of the body, supplied by the LV and the and assesses the pressure gradient across the
ascending aorta, is perfused with well- lesion. Magnetic resonance imaging demon-
oxygenated blood; however, the lower half strates in detail the length and severity of
appears cyanotic because it is largely sup- coarctation (see Fig. 16.17). Diagnostic
plied by right-to-left flow of poorly oxy- catheterization and angiography are rarely
genated blood from the pulmonary artery, necessary.
across the ductus arteriosus, and into the de-
scending aorta.
When the coarctation is less severe, as in Treatment
most postductal cases, it may be suspected
In neonates with severe obstruction, pro-
by finding upper extremity hypertension on
staglandin infusion is administered to keep
physical examination during childhood.
the ductus arteriosus patent, thus maintain-
ing blood flow to the descending aorta be-
Physical Examination fore surgery is undertaken. In children, elec-
tive repair is usually performed to prevent
On examination, the femoral pulses are systemic hypertension. Several effective sur-
weak and delayed. An elevated blood pres- gical procedures are available, including ex-
sure in the upper body is the most common cision of the narrowed aortic segment with
presentation. If the coarctation occurs dis- end-to-end reanastomosis and direct repair
tal to the takeoff of the left subclavian of the coarctation, sometimes using syn-
artery, the systolic pressure in the arms is thetic patch material. For older children,
greater than that in the legs. If the coarcta- adults, and patients with recurrent coarcta-
tion occurs proximal to the takeoff of the left tion after previous repair, transcatheter in-
subclavian artery, the systolic pressure in terventions (balloon dilatation with or with-
the right arm may exceed that in the left out stent placement) is usually successful.
arm. A systolic pressure in the right arm Antibiotic prophylaxis to prevent endarteri-
that is 15 to 20 mm Hg greater than that tis (like the prophylaxis against endocarditis
in a leg is sufficient to suspect coarctation, described in Chapter 8) is necessary even
because normally the systolic pressure in after repair.
the legs is higher than that in the arms. A
midsystolic ejection murmur (caused by
flow through the coarctation) may be audi- Cyanotic Lesions
ble over the chest and/or back. A prominent
tortuous collateral arterial circulation may
Tetralogy of Fallot
create continuous murmurs over the chest Tetralogy of Fallot results from a single de-
in adults. velopmental defect: an abnormal anterior
and cephalad displacement of the infun-
dibular (outflow tract) portion of the inter-
Diagnostic Studies
ventricular septum. As a consequence, four
In adults with uncorrected coarctation of anomalies arise that characterize this condi-
the aorta, chest radiography generally reveals tion, as shown in Figure 16.18: (1) a VSD Fig. 18
notching of the inferior surface of the pos- caused by malalignment of the interventric-
terior ribs owing to enlarged intercostal ular septum, (2) subvalvular pulmonic
vessels supplying collateral circulation to stenosis because of obstruction from the in-
the descending aorta. An indented aorta at fundibular septum, (3) an overriding aorta
the site of coarctation may also be visual- that receives blood from both ventricles,
10090-16_CH16.qxd 8/31/06 4:57 PM Page 391

Congenital Heart Disease 391

and (4) right ventricular hypertrophy owing


to the high pressure load placed on the RV
by the pulmonic stenosis. Tetralogy of Fallot
is the most common form of cyanotic con-
genital heart disease after infancy, occurring
in 5 of 10,000 live births, and is often asso-
ciated with other cardiac defects, including
a right-sided aortic arch (25% of patients),
ASD (10% of patients), and less often,
anomalous origin of the left coronary artery.
A microdeletion in chromosome 22 (22q11)
has been identified in patients with a syn-
drome that includes tetralogy of Fallot as
one of the cardiovascular manifestations
(see Box 16.1). Box 1

Pathophysiology
Increased resistance by the subvalvular pul-
monic stenosis causes deoxygenated blood
returning from the systemic veins to be di-
Figure 16.18. Tetralogy of Fallot is characterized by verted from the RV, through the VSD, to the
four associated anomalies. 1) A ventricular septal de-
fect (hollow arrow), 2) obstruction to right ventricular LV, and into the systemic circulation, result-
outflow (solid arrow), 3) an overriding aorta that receives ing in systemic hypoxemia and cyanosis.
blood from both ventricles, and 4) right ventricular The magnitude of shunt flow across the VSD
hypertrophy. Ao, aorta; IVC, inferior vena cava; LA, left
atrium; LV, left ventricle; PA, pulmonary artery; RA, right is primarily a function of the severity of the
atrium; RV, right ventricle; SVC, superior vena cava. pulmonary stenosis, but acute changes in

Box 16.1 Genetic Abnormalities in Congenital Heart Disease

Progress in the understanding of genetic influences on cardiac development and con-


genital heart disease is proceeding at a brisk pace, aided by the Human Genome Project.
Although nearly all cardiac congenital anomalies can occur as isolated findings, the clus-
tering of certain forms with heritable syndromes and known genetic abnormalities pro-
vides clues to the underlying basis for some defects. As with other congenital conditions,
a complex interplay between genes and the embryonic environment can result in an array
of phenotypes.
Among infants with Down syndrome (trisomy 21) the incidence of congenital heart
defects is nearly a 40%. Many of these are common abnormalities such as atrial septal de-
fect, ventricular septal defect, and patent ductus arteriosus. However, there is also a high
incidence of a rarer condition known as common atrioventricular canal, which consists of
a large atrial and ventricular septal defect and a common (undivided) atrioventricular valve
above the two ventricles. This portion of heart tissue is usually formed by the interaction
between the endocardial cushions and cells of neural crest origin, which are known to
have abnormal migration patterns in patients with trisomy 21.
Turner syndrome (45, XO) is a rare heritable condition of girls and women that is also
associated with congenital heart disease. Left-sided obstructive congenital heart lesions
10090-16_CH16.qxd 8/31/06 4:57 PM Page 392

392 Chapter Sixteen

are common in patients with this syndrome, including bicuspid aortic valve, coarctation
of the aorta and occasionally the hypoplastic left heart syndrome (underdevelopment of
the left ventricle and aorta). The specific genes responsible for these abnormalities have
not yet been elucidated.
In contrast, discrete gene abnormalities have been identified in other syndrome-asso-
ciated forms of congenital heart disease. For example, patients with Williams syndrome
(characterized by mental retardation, hypercalcemia, renovascular hypertension, facial ab-
normalities, and short stature) have a high incidence of supravalvular aortic stenosis, a
ringlike obstruction above the aortic valve. Some patients have a more diffuse arteriopathy
of the aorta as well as pulmonary artery obstruction. The genetic abnormality in Williams
syndrome is a deletion on chromosome 7 (7q11.23), a region that includes the elastin
gene. Abnormalities in the production of elastin, a critical component of the arterial wall,
may be responsible for the observed arteriopathy.
AQ5 DiGeorge syndrome (manifested by a characteristic facial appearance, pharyngeal de-
fects, absent parathyroid glands with hypocalcemia, and hypoplasia of the thymus with
defective immune T-cell function) is associated with congenital abnormalities of the car-
diac outflow tracts, such as tetralogy of Fallot, truncus arteriosus (a large VSD over which
a single large outflow vessel arises), and interrupted aortic arch. Most patients with Di-
George syndrome have a microdeletion within chromosome 22 (22q11). Recent research
has implicated a single gene (TBX1) in the cardiac manifestations. This gene encodes a
transcription factor that appears to play a critical role in developmental patterning of the
cardiac outflow tracts.
Several other transcription factors of importance in heart development likely contribute
to congenital heart disease. Some families with heritable forms of atrial septal defects have
mutations in the transcription factor gene Nkx2.5. An associated transcription factor gene
(GATA4) appears to collaborate with Nkx2.5 and has been found to be involved in familial
septal defect syndromes. Mutations in TBX5, yet another transcription factor gene, are re-
sponsible for the Holt-Oram syndrome (also known as the heart-hand syndrome), an au-
tosomal dominant disorder characterized by abnormal development of the upper extrem-
ities and cardiac defects, most commonly secundum ASDs and ventricular septal defects.
There are also examples of loci involved in specific cellular processes, mutations of
which have been implicated in congenital heart disease. One example is the gene PTPN11,
which functions as a critical regulator of signal transduction pathways. This gene is mu-
tated in many patients with Noonan syndrome, features of which include short stature,
a dysmorphic facial appearance, chest deformities, and congenital heart defects, most
commonly valvular and supravalvular pulmonary stenosis.
Continuing advances in knowledge about the genome will undoubtedly lead to a
greater understanding of cardiac development and how molecular defects in these
processes lead to congenital heart abnormalities.

systemic and pulmonary vascular resistances when systemic vasodilatation results in an


can affect it as well. increased right-to-left shunt. Manifestations
of such spells include irritability, cyanosis,
hyperventilation, and occasionally syncope
Symptoms
or convulsions. Children learn to alleviate
Children with tetralogy of Fallot often ex- their symptoms by squatting down, which
perience dyspnea on exertion. “Spells” may is thought to increase systemic vascular re-
occur following exertion, feeding, or crying sistance by “kinking” the femoral arteries,
10090-16_CH16.qxd 8/31/06 4:57 PM Page 393

Congenital Heart Disease 393

thereby decreasing the right-to-left shunt branches) to the pulmonary artery and cre-
and directing more blood from the RV to the ating a left-to-right shunt to increase pul- AQ1
lungs. monary blood flow. Such procedures are oc-
casionally used today in infants for whom
definitive repair is planned at an older age.
Physical Examination
Complete surgical correction of tetralogy of
Children with tetralogy of Fallot and mod- Fallot involves closure of the VSD and en-
erate pulmonary stenosis often have mild largement of the subpulmonary infundibu-
cyanosis, most notably of the lips, mucous lum with the use of a pericardial patch. Elec-
membranes, and digits. Infants with severe tive repair is usually recommended around
pulmonary stenosis may present with pro- 1 year of age to decrease the likelihood of fu-
found cyanosis in the first few days of life. ture complications. Most patients who have
Chronic hypoxemia caused by the right-to- undergone successful repair grow to become
left shunt commonly results in clubbing of asymptomatic adults. However, antibiotic
the fingers and toes. Right ventricular hy- prophylaxis to prevent endocarditis is still
pertrophy may be appreciated on physical required.
examination as a palpable heave along the
left sternal border. The S2 is single, composed
Transposition of the Great Arteries
of a normal aortic component; the pulmo-
nary component is soft and usually inaudi- In transposition of the great arteries (TGA)
ble. A systolic ejection murmur heard best at each great vessel inappropriately arises from
the upper-left sternal border is created by the opposite ventricle; that is, the aorta orig-
turbulent blood flow through the stenotic inates from the RV and the pulmonary
right ventricular outflow tract. There is usu- artery originates from the LV (Fig. 16.19). Fig. 19
ally no distinct murmur related to the VSD,
because it is typically large and thus gener-
ates little turbulence.

Diagnostic Studies
Chest radiography demonstrates prominence
of the RV and decreased size of the main pul-
monary artery segment, giving the appear-
ance of a “boot-shaped” heart. Pulmonary
vascular markings are typically diminished
because of decreased flow through the pul-
monary circulation. The ECG shows right
ventricular hypertrophy with right axis de-
viation. Echocardiography details the right
ventricular outflow tract anatomy, the mis-
aligned VSD, right ventricular hypertrophy,
and other associated defects, as does cardiac
catheterization.

Treatment
Before definitive surgical correction of tet-
ralogy of Fallot was developed, several forms Figure 16.19. Transposition of the great arteries. The
of palliative therapy were undertaken. These aorta (Ao) and pulmonary artery (PA) arise abnormally
from the right ventricle (RV) and left ventricle (LV), re-
involved creating anatomic communica- spectively. IVC, inferior vena cava; LA, left atrium; RA,
tions between the aorta (or one of its major right atrium; SVC, superior vena cava.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 394

394 Chapter Sixteen

This anomaly accounts for approximately and is pumped out the pulmonary artery.
7% of congenital heart defects, affecting 40 Most of the pulmonary artery flow travels
of 100,000 live births. Whereas tetralogy of through the ductus arteriosus into the aorta,
Fallot is the most common etiology of instead of the high-resistance pulmonary
cyanosis after infancy, TGA is the most com- vessels, and then oxygen is provided to the
mon cause of cyanosis in the neonatal period. developing tissues.
The precise cause of transposition re- After birth, normal physiologic closure of
mains unknown. Historically, failure of the the ductus and the foramen ovale elimi-
aorticopulmonary septum to spiral in a nates the shunt between the parallel circu-
normal fashion during fetal development lations and, without intervention, would re-
was considered the underlying problem. It sult in death because oxygenated blood does
has been suggested that the defect may be not reach the systemic tissues. However, if
the result of abnormal growth and absorp- the ductus arteriosus and foramen ovale re-
tion of the subpulmonary and subaortic main patent (either naturally or with exoge-
infundibuli during the division of the nous prostaglandins or surgical interven-
truncus arteriosus. Normally, reabsorption tion), communication between the parallel
of the subaortic infundibulum places the circuits is maintained, and sufficiently oxy-
forming aortic valve posterior and inferior genated blood may be provided to the brain
to the pulmonary valve and in continuity and other vital organs.
with the LV. In TGA, the process of in-
fundibular reabsorption may be reversed,
Symptoms and Physical Examination
placing the pulmonary valve over the LV
instead. Infants with transposition appear blue, with
the intensity of the cyanosis dependent on
the degree of intermixing between the par-
Pathophysiology
allel circuits. In most cases, generalized
TGA separates the pulmonary and systemic cyanosis is apparent on the first day of life
circulations by placing the two circuits in and progresses rapidly as the ductus arte-
parallel rather than in series. This arrange- riosus closes. Palpation of the chest reveals
ment forces desaturated blood from the sys- a right ventricular impulse at the lower
temic venous system to pass through the sternal border as the RV faces systemic
RV and then return to the systemic circula- pressures. Auscultation may reveal an ac-
tion through the aorta without undergoing centuated S2, which reflects closure of the
normal oxygenation in the lungs. Similarly, anteriorly placed aortic valve just under
oxygenated pulmonary venous return passes the chest wall. Prominent murmurs are un-
through the LV and then back through the common and may signal an additional
pulmonary artery to the lungs without im- defect.
parting oxygen to the systemic circulation.
The result is an extremely hypoxic, cyanotic
Diagnostic Studies
neonate. Without intervention to create
mixing between the two circulations, TGA is Chest radiography is usually normal, although the
a lethal condition. base of the heart may be narrow owing to
TGA is compatible with life in utero be- the more anterior-posterior orientation of
cause flow through the ductus arteriosus the aorta and pulmonary artery. The ECG
and foramen ovale allows communication demonstrates right ventricular hypertrophy,
between the two circulations. Oxygenated reflecting the fact that the RV is the systemic
fetal blood flows from the placenta through “high-pressure” pumping chamber. The de-
the umbilical vein to the right atrium, and finitive diagnosis of transposition can be
then most of it travels into the left atrium made by echocardiography, which demon-
through the foramen ovale. The oxygenated strates the abnormal orientation of the great
blood in the left atrium passes into the LV vessels.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 395

Congenital Heart Disease 395

Treatment ture of the pulmonary vessels can result in


hemoptysis.
TGA is a medical emergency. Initial treat-
On examination, a patient with Eisen-
ment includes maintenance of the ductus ar-
menger syndrome appears cyanotic with
teriosus by prostaglandin infusion and cre-
digital clubbing. A prominent a wave in the
ation of an interatrial communication using jugular venous pulsation represents elevated
a balloon catheter (the Rashkind procedure). right-sided pressure during atrial contrac-
These procedures allow adequate mixing of tion. A loud P2 is common. The murmur of
the two circulations until definitive correc- the inciting left-to-right shunt is usually ab-
tive surgery can be performed. The current sent, because the original pressure gradient
corrective procedure of choice is the “arterial across the lesion is negated by the elevated
switch” operation (Jatene procedure), which right-heart pressures.
involves transection of the great vessels Chest radiography in Eisenmenger syn-
above the semilunar valves and origin of the drome is notable for proximal pulmonary
coronary arteries. The great vessels are then artery dilatation with peripheral tapering.
switched to the natural configuration, so the Calcification of the pulmonary vasculature
aorta arises from the LV and the pulmonary may be seen. The ECG demonstrates right
artery arises from the RV. The coronary ar- ventricular hypertrophy and right atrial en-
teries are then relocated to the new aorta. largement. Echocardiography with Doppler
studies can usually identify the underlying
EISENMENGER SYNDROME cardiac defect and quantitate the pulmo-
nary artery systolic pressure.
Eisenmenger syndrome is the condition of Treatment includes the avoidance of ac-
severe pulmonary vascular obstruction that tivities that can exacerbate the right-to-left
results from chronic left-to-right shunting shunt. These include strenuous physical ac-
through a congenital cardiac defect. The el- tivity, high altitude, and the use of periph-
evated pulmonary vascular resistance causes eral vasodilator drugs. Pregnancy is espe-
reversal of the original shunt (to the right- cially dangerous; the rate of spontaneous
to-left direction) and systemic cyanosis. abortion is 20% to 40% and the incidence of
The mechanism by which increased pul- maternal mortality is 45%.
monary flow causes this condition is un- No medical therapy offers a reliably effec-
known. Histologically, the pulmonary arte- tive way to reduce the elevated pulmonary
riolar media hypertrophies and the intima vascular resistance. Supportive measures
proliferates, reducing the cross-sectional include endocarditis prophylaxis, manage-
area of the pulmonary vascular bed. Over ment of rhythm disturbances, and phlebo-
time, the vessels become thrombosed, and tomy for patients with symptomatic eryth-
the resistance of the pulmonary vasculature rocytosis. The only effective long-term
rises, causing the original left-to-right shunt strategy for severely affected patients is lung
to decrease. Eventually, if the resistance of or heart-lung transplantation. Fortunately,
the pulmonary circulation exceeds that of the with the dramatic advances that have been
systemic vasculature, the direction of shunt made in the detection and early correction
flow reverses. of severe congenital heart defects, Eisen-
With reversal of the shunt to the right- menger syndrome has become less common.
to-left direction, symptoms arise from hy-
poxemia, including exertional dyspnea and
SUMMARY
fatigue. Reduced hemoglobin saturation stim-
ulates the bone marrow to produce more red 1. The significance of congenital heart le-
blood cells (erythrocytosis), which can lead to sions can be predicted from an under-
hyperviscosity, symptoms of which include standing of cardiovascular embryonic de-
fatigue, headaches, and stroke (caused by velopment and the transition to
cerebrovascular occlusion). Infarction or rup- postnatal circulatory pathways.
10090-16_CH16.qxd 8/31/06 4:57 PM Page 396

396 Chapter Sixteen

2. Cardiac malformations occur in 0.8% of Brickner ME, Hillis LD, Lange RA. Congenital heart
live births. Such lesions can be grouped disease in adults, part I of II. N Engl J Med 2000;
342(4):256–263.
into cyanotic or acyanotic defects, de-
Brickner ME, Hillis LD, Lange RA. Congenital heart
pending on whether the abnormality re-
disease in adults, part II of II. N Engl J Med 2000;
sults in pulmonary-to-systemic (right-to- 342(5):334–342.
left) shunting of blood. Gatzoulis MA, Swan L, Therrien J. Adult Congenital
3. Acyanotic defects often result in either Heart Disease: A Practical Guide. Malden, MA: BMJ
volume overload (ASD, VSD, PDA) or pres- Books, 2005.
sure overload (AS, pulmonic stenosis, Gelb BD. Genetic basis of congenital heart disease.
Curr Opin Cardiol 2004;19:110–115.
coarctation of the aorta). Chronic volume
Goldmuntz E. The genetic contribution to congeni-
overload resulting from a large left-to-
tal heart disease. Pediatr Clin North Am 2004;51:
right shunt can ultimately result in in- 1721–1737.
creased pulmonary vascular resistance, re- Kearns-Jonker M. Congenital Heart Disease: Molecu-
versal of the direction of shunt flow, and lar Diagnostics. Totowa, NJ: Humana Press, 2006.
subsequent cyanosis (Eisenmenger syn- Moodie DS. Diagnosis and management of congen-
drome). ital heart disease in the adult. Cardiol Rev 2001;9:
276–281.
4. Among the most common cyanotic de-
Moore KL, Persaud TVN. The Developing Human:
fects are tetralogy of Fallot and TGA.
Clinically Oriented Embryology. 7th Ed. Philadel-
phia: WB Saunders, 2003.
Acknowledgment Park MK, Troxler RG. Pediatric Cardiology for Prac-
titioners. 4th Ed. St. Louis: Mosby, 2002.
Contributors to the previous editions of this chapter Perloff JK. The Clinical Recognition of Congenital
were Yi-Bin Chen, MD; Douglas W. Green, MD; Heart Disease. 5th Ed. Philadelphia: WB Saunders,
Lakshmi Halasyamani, MD; Andrew Karson, MD; 2003.
Raymond Tabibiazar, MD; Michael D. Freed, MD.; Rudolph AM. Congenital Diseases of the Heart:
and Richard Liberthson, MD. Clinical-Physiological Considerations. 2nd Ed.
New York: Futura, 2001.
Sadler TW. Langman’s Medical Embryology. 9th Ed.
Additional Reading
Philadelphia: Lippincott Williams & Wilkins,
Allen HD, Gutgesell HP, Clark EB, et al., eds. Moss 2004.
and Adams’ Heart Disease in Infants, Children, Tworetzky W, Marshall AC. Fetal interventions for
and Adolescents. 6th Ed. Baltimore: Lippincott cardiac defects. Pediatr Clin North Am 2004;51:
Williams & Wilkins, 2001. 1503–1513.
10090-16_CH16.qxd 8/31/06 4:57 PM Page a

Chapter 16—Author Queries


1. AU: Should this be “or” instead?
2. Additional Readings section cites a 2001 edition (6th). Should this citation (and possibly
the figure) be updated?
3. Additional Readings section cites a 2003 edition (7th). Should this citation (and possibly
the figure) be updated? See also Figures 16.7–10.
4. AU: The figure shows A, B, and C panels. Should this caption be so labeled?

Chapter 16—Author Query (Box)


5. AU: Edit correct?
10090-17_CH17.qxd 8/31/06 4:58 PM Page 397

C H A P T E R

Cardiovascular Drugs
Martin W. Schoen
Elliott M. Antman
17
Gary R. Strichartz
Leonard S. Lilly

INOTROPIC DRUGS AND VASOPRESSORS Class IB Antiarrhythmics


Digitalis Glycosides Class IC Antiarrhythmics
Sympathomimetic Amines Class II Antiarrhythmics
Phosphodiesterase Inhibitors Class III Antiarrhythmics
Vasopressin Class IV Antiarrhythmics
Adenosine
VASODILATOR DRUGS
Angiotensin-Converting Enzyme Inhibitors DIURETICS
Angiotensin II Type 1 Receptor Antagonists Loop Diuretics
Direct-Acting Vasodilators Thiazide Diuretics
Calcium Channel Blockers Potassium-Sparing Diuretics
Organic Nitrates ANTITHROMBOTIC DRUGS
Natriuretic Peptides Platelet Inhibitors
Sildenafil Anticoagulant Drugs
ANTIADRENERGIC DRUGS LIPID-REGULATING DRUGS
Central Adrenergic Inhibitors HMG CoA Reductase Inhibitors
Sympathetic Nerve-Ending Antagonists Bile Acid–Binding Agents
Peripheral α-Adrenergic Receptor Antagonists Cholesterol Absorption Inhibitors
β-Adrenergic Receptor Antagonists Niacin
ANTIARRHYTHMIC DRUGS Fibrates
Class IA Antiarrhythmics

This chapter reviews the physiologic basis INOTROPIC DRUGS


and clinical use of cardiovascular drugs. Al- AND VASOPRESSORS
though a multitude of drugs are available to
treat cardiac disorders, these agents can for- Inotropic drugs are used to increase the force
tunately be grouped by their pharmacologic of ventricular contraction when myocardial
actions into a small number of categories. systolic function is impaired. The pharmaco-
Additionally, many drugs are useful in more logic agents in this category include the car-
than one form of heart disease. diac glycosides, sympathomimetic amines,

397
10090-17_CH17.qxd 8/31/06 4:58 PM Page 398

398 Chapter Seventeen

and phosphodiesterase inhibitors. Although chanical effect) and 2) to prolong the refrac-
they work through different mechanisms, tory period of the atrioventricular (AV) node
they are all thought to improve cardiac con- in patients with supraventricular arrhyth-
traction by increasing the intracellular cal- mias (electrical effect).
cium concentration, thus augmenting actin
and myosin interactions. The hemodynamic Mechanical Effect
effect is to shift a depressed ventricular per-
formance curve (Frank-Starling curve) in an The action by which digitalis improves
Fig. 1 upward direction (Fig. 17.1), so that for a contractility appears to be inhibition of the
given ventricular filling pressure, stroke vol- sarcolemmal Na+K+-ATPase “pump,” nor-
ume and cardiac output are increased. mally responsible for maintaining trans-
membrane Na+ and K+ gradients. By bind-
ing to and inhibiting this pump, digitalis
Digitalis Glycosides
causes the intracellular [Na+] to rise. As AQ1
The cardiac glycosides are often called “dig- shown in Figure 17.2, an increase in intra- Fig. 2
italis” because commonly used drugs of this cellular sodium content reduces Ca++ extru-
class are based on extracts of the foxglove sion from the cell by the Na+-Ca++ exchanger.
plant, Digitalis purpurea. In this discussion, Consequently, more Ca++ is pumped into
the term digitalis is used to describe the en- the sarcoplasmic reticulum, and when
tire group of cardiac glycosides, including subsequent action potentials excite the
digoxin, digitoxin, and ouabain. cell, a greater-than-normal amount of Ca++
is released to the myofilaments, thereby en-
hancing the force of contraction. The mag-
Mechanism of Action
nitude of the positive inotropic effect cor-
The two desired effects of digitalis are (1) to relates with the degree of Na+K+-ATPase
improve contractility of the failing heart (me- inhibition.

Normal
Cardiac Output

Inotropic
therapy

Heart failure
Hypotension

Diuretic
therapy

Pulmonary congestion

Left ventricular end-diastolic pressure


Figure 17.1. Ventricular performance (Frank-Starling) curve. In heart failure,
the curve is displaced downward, so that at a given left ventricular end-diastolic
pressure (LVEDP), the cardiac output is lower than in a normal heart. Diuretics re-
duce LVEDP but do not change the position of the curve; thus, pulmonary con-
gestion improves but cardiac output may fall. Inotropic drugs displace the curve
upward, toward normal, so that at any LVEDP, the cardiac output is higher.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 399

Cardiovascular Drugs 399

ple, in atrial and ventricular Purkinje fibers,


a high digitalis concentration has three im-
portant actions that may lead to dangerous
arrhythmias (Fig. 17.3): Fig. 3
DIGITALIS
1. Less negative resting potential. Inhibition of
the Na+ K+-ATPase causes the resting po-
tential to become less negative. Since the
Na+ K+-ATPase normally removes three
Na+ ions from the cell in exchange for two
inwardly moving K+ ions; inhibition of
the pump results in a decrease of this out-
ward current and a resulting depolariza-
tion of the cell. Consequently, there is a
voltage-dependent partial inactivation of
the fast Na+ channels, which leads to a
slower rise of phase 0 depolarization and
Figure 17.2. Mechanism of action of digitalis (in- reduction in conduction velocity (see
otropic effect). A. Digitalis inhibits the sarcolemmal Fig. 1.17). The slowed conduction, if pre-
Na+K+-ATPase, causing intracellular [Na+] to rise. B. In-
creased cytosolic [Na+] reduces the transmembrane Na+ sent heterogeneously among neighboring
gradient; thus, the Na+-Ca+ exchanger drives less Ca++ out cells, enhances the possibility of reentrant
of the cell. C. The increased [Ca++] is stored in the sar- arrhythmias.
coplasmic reticulum, such that with subsequent action
potentials, greater-than-normal Ca++ is released to the 2. Decreased action potential duration. At high
contractile elements in the cytoplasm, intensifying the digitalis concentrations, the cardiac action
force of contraction.
potential shortens. This relates in part to
the digitalis-induced elevated intracellu-
lar [Ca++], which increases the activity of
Electrical Effect a Ca++-dependent K+ channel. The open-
ing of this channel promotes K+ efflux
The major therapeutic electrical effect of dig-
and more rapid repolarization. In addition,
italis occurs at the AV node, where it slows
high intracellular [Ca++] inactivates the
conduction velocity and increases refractori-
Ca++ channels, decreasing the inward de-
Tab. 1 ness (Table 17.1). Digitalis affects the elec-
polarizing Ca++ current. The decrease in
trical properties of cardiac tissue directly,
action potential duration and the associ-
but more importantly, it modifies autonomic
ated shortened refractory period increase
nervous system output by enhancing vagal
the time during which cardiac fibers are
tone and reducing sympathetic activity. As
responsive to external stimulation, allow-
a result, digitalis decreases the frequency of
ing greater opportunity for propagation
transmission of atrial impulses through the
of arrhythmic impulses.
AV node to the ventricles. This is beneficial
in reducing the rate of ventricular stimula- 3. Enhanced automaticity. Digitalis enhances
tion in patients with rapid supraventricular cellular automaticity and may generate
tachycardias such as atrial fibrillation or atrial ectopic rhythms by two mechanisms:
flutter. In addition, by enhancing the re- a. The less negative membrane resting
fractoriness of the AV node, digitalis may potential may induce phase 4 gradual
convert supraventricular reentrant arrhyth- depolarization, even in nonpacemaker
mias to normal rhythm. cells (see Chapter 11), and an action
However, if digitalis concentrations rise potential is triggered each time the
into the toxic range, further enhancement threshold voltage is reached.
of vagal tone and more extreme inhibition b. The digitalis-induced increase in intra-
of the Na+ K+-ATPase pump can result in ad- cellular [Ca++] may trigger delayed after-
verse electrophysiologic effects. For exam- depolarizations (see Fig. 17.3). If an
10090-17_CH17.qxd 8/31/06 4:58 PM Page 400

400 Chapter Seventeen

TABLE 17.1. Electrophysiologic Effects of Digitalis

Region Mechanism of Action Effect

Therapeutic effects
AV node Vagal effect • ↓ Rate of transmission of atrial im-
pulses to the ventricles in supraven-
tricular tachyarrhythmias
↓ Conduction velocity • ↓ Conduction velocity and ↑ refrac-
tory period may interrupt reentrant
circuits passing through the AV node
↑ Effective refractory period
Toxic effects
Sinoatrial node ↑ Vagal and direct suppression • Sinus bradycardia
• Sinoatrial block (impulse not trans-
mitted from SA node to atrium)
Atrium Delayed afterdepolarizations (triggered • Atrial premature beats
activity), ↑ slope of phase 4 depolar-
ization (↑ automaticity)
Variable effects on conduction velocity • Nonreentrant SVT (ectopic rhythm)
and ↑ refractory period (can fragment
conduction and lead to reentry)
• Reentrant PSVT
AV node Direct and vagal-mediated conduction • AV block (first, second or third degree)
block
AV junction Delayed afterdepolarizations (triggered • Accelerated junctional rhythm
(between AV node activity), ↑ slope of phase 4 depolar-
and His bundle) ization (↑ automaticity)
Purkinje fibers and Delayed afterdepolarizations (triggered • Ventricular premature beats
ventricular muscle activity), ↓ conduction velocity and ↑
refractory period (can lead to reentry)
↑ slope of phase 4 depolarization • Ventricular tachycardia
(↑ automaticity)

AV, atrioventricular; PSVT, paroxysmal supraventricular tachycardia.

afterdepolarization reaches the thresh-


old voltage, an action potential (ecto-
B pic beat) is generated. Ectopic beats
may lead to additional afterdepolar-
A C izations and self-sustaining arrhyth-
mias such as ventricular tachycardia.
Thus, digitalis in toxic concentrations may
lead to several types of ectopic or reentrant
Figure 17.3. Direct effects of digitalis on the Purkinje rhythms (see Table 17.1). In addition, the
cell action potential. The solid tracing represents depo-
larization and repolarization of a normal cell; the dashed
augmented direct and indirect vagal effects
tracing demonstrates the effects of digitalis. A. The max- of toxic doses of digitalis slow conduction
imum diastolic potential is less negative, and there is an through the AV node, such that high de-
increase in the slope of phase 4 depolarization, endowing
the cell with intrinsic automaticity, and the potential for grees of AV block, including complete heart
ectopic rhythms. B. Because depolarization of the cell oc- block, can occur.
curs at a more positive voltage, the rate of rise of phase 0
is decreased, and conduction velocity is slowed, which, if
present heterogeneously among neighboring cells, can
produce conditions for reentry. C. Delayed afterdepolar- Clinical Uses
izations may develop at high concentrations of digitalis in
association with an increased intracellular calcium con- The most common use of digitalis is as an in-
centration and can result in triggered tachyarrhythmias. otropic agent to treat heart failure caused by
10090-17_CH17.qxd 8/31/06 4:58 PM Page 401

Cardiovascular Drugs 401

decreased ventricular contractility (see Chap- Pharmacokinetics and Toxicity


ter 9). Digitalis increases the force of contrac-
The most commonly used form of digitalis is
tion and augments cardiac output, thereby
digoxin, which is excreted unchanged by the
improving left ventricular emptying, reduc-
kidney. A series of loading doses of digoxin is
ing left ventricular size, and decreasing the el-
necessary to raise the drug’s concentration
evated ventricular filling pressures typical of
into the therapeutic range. If a loading dose
patients with systolic dysfunction. Digitalis is
is not given, the steady-state concentration
not beneficial in forms of heart failure associ-
is established in approximately 7 days. The
ated with normal ventricular contractility
maintenance dosage depends on the patient’s
(e.g., high-output failure associated with thy-
ability to excrete the drug (i.e., renal function).
rotoxicosis, pulmonary congestion caused by
The potential for digitalis toxicity is sig-
mitral stenosis, or in the setting of pure di-
nificant because of a low toxic-to-therapeutic
astolic dysfunction).
drug concentration ratio. Although many
Once the mainstay of therapy in con-
side effects are minor, life-threatening
gestive heart failure (CHF), the use of digi-
arrhythmias may result. Extracardiac signs
talis has waned in the face of newer, more of acute digitalis toxicity are often gastro-
effective therapies (as discussed later in the intestinal (e.g., nausea, vomiting, anorexia),
chapter; see also Chapter 9). Nonetheless, thought to be mediated by the action of
digitalis continues to be useful in treating digoxin on the area postrema of the brain
patients with CHF complicated by atrial stem. Cardiac toxicity includes a host of
fibrillation (it has the added benefit of slow- arrhythmias (see Table 17.1) that may pre-
ing the ventricular heart rate), or when cede extracardiac warning symptoms. The
symptoms do not respond adequately to most frequently encountered rhythm dis-
angiotensin-converting enzyme (ACE) in- turbance is the development of ventricular
hibitors, β-blockers, and diuretics. Unlike extrasystoles. In addition, various degrees of
ACE inhibitors and β-blockers, digitalis does AV block may occur because of the direct
not prolong the life expectancy of patients and vagal effects on AV nodal conduction.
with chronic heart failure. Digitalis toxicity is the most common cause
The second-most common use of digitalis of nonreentrant types of supraventricular
is as an antiarrhythmic agent in the treatment tachycardia (i.e., caused by enhanced auto-
of atrial fibrillation, atrial flutter, and parox- maticity or delayed afterdepolarizations).
ysmal supraventricular tachycardia (PSVT). Many factors contribute to digitalis intox-
In atrial fibrillation and flutter, digitalis re- ication, the most common of which is hypo-
duces the number of impulses transmitted kalemia, often caused by the concurrent
across the AV node, thereby slowing the administration of diuretics. Hypokalemia
ventricular rate. Digitalis may terminate re- exacerbates digitalis toxicity because it fur-
entrant supraventricular tachycardias, likely ther inhibits the Na+ K+-ATPase pump. Other
through enhancement of vagal tone, which conditions that promote digitalis toxicity in-
slows impulse conduction, prolongs the ef- clude hypomagnesemia and hypercalcemia.
fective refractory period, and can therefore In addition, the concurrent administration
interrupt reentrant circuits that pass through of other drugs (e.g., quinidine) may raise
the AV node. the serum digoxin concentration by decreas-
The use of digitalis as an antiarrhythmic ing its excretion and reducing its volume of
has also become less frequent in recent years distribution.
because other agents such as β-blockers, cal- The treatment of digitalis-induced tachy-
cium channel blockers, and amiodarone are arrhythmias includes administration of po-
often more effective. Nonetheless, for the tassium (if hypokalemia is present) and often
treatment of supraventricular tachyarrhyth- intravenous lidocaine (discussed later in the
mias in the presence of CHF, digitalis remains chapter). High-grade AV block may require
an important option. temporary pacemaker therapy. In patients
10090-17_CH17.qxd 8/31/06 4:58 PM Page 402

402 Chapter Seventeen

with severe intoxication, administration of Dopamine is an endogenous catechol-


Fab fragments of antidigitalis antibodies may amine and the precursor of norepinephrine.
be life saving. It possesses an unusual combination of ac-
tions that make it attractive in the treat-
ment of heart failure associated with hy-
Sympathomimetic Amines
potension and poor renal perfusion. There
Sympathomimetic amines are inotropic drugs are various types of receptors with various
that bind to cardiac β1-receptors. Stimulation affinities for dopamine. At low dosages,
of these receptors increases the activity of <2 µg/kg per minute, dopamine interacts
adenylate cyclase, causing increased forma- primarily with dopaminergic receptors dis-
tion of cyclic adenosine monophosphate tributed in the renal and mesenteric vascu-
Fig. 4 (cAMP; Fig. 17.4). Increased cAMP activates lar beds. Stimulation of these receptors causes
protein kinases, which promote intracellular local vasodilation and increases renal blood
calcium influx by phosphorylating the slow flow and glomerular filtration, facilitating
L-type calcium channels. The increased cal- diuresis.
cium entry triggers a corresponding rise in Medium dosages of dopamine, 2 to 10 µg/kg
Ca++ release from the sarcoplasmic reticulum, per minute, increase inotropy by stimulation
which enhances the force of contraction. of cardiac β1-receptors directly and indi-
Intravenous dopamine and dobutamine are rectly by promoting release of norepineph-
commonly used sympathomimetic amines in rine from sympathetic nerve terminals. This
the treatment of acute heart failure. Norepi- action increases heart rate, cardiac contrac-
nephrine, epinephrine, and isoproterenol are tility, and stroke volume, all of which aug-
used in special circumstances, as described in ment cardiac output.
Tab. 2 the following paragraphs. Table 17.2 sum- At high dosages, >10 µg/kg per minute, do-
marizes the receptor actions and major hemo- pamine also stimulates systemic α-receptors,
dynamic effects of these agents. thereby causing vasoconstriction and elevat-

Adenylate
cyclase

GS protein
ATP
cAMP

PD
AMP Inactive
protein Active
kinases protein
kinases
Myocyte

Figure 17.4. Mechanism by which b-adrenergic stimulation increases intra-


cellular Ca11. β1-Receptor stimulation acts through G proteins (guanine nucleotide
regulatory proteins) to activate adenylate cyclase. The latter increases cyclic adenosine
monophosphate (cAMP) production, which mediates protein kinase phosphorylation
of cellular proteins, including ion channels. Phosphorylation of the slow Ca++ channel
increases calcium influx. cAMP is degraded by phosphodiesterase (PD).
10090-17_CH17.qxd 8/31/06 4:58 PM Page 403

Cardiovascular Drugs 403

TABLE 17.2. Sympathomimetic Drug Effects

Receptor Stimulation

Drug D1 (↑ renal perfusion) a (vasoconstriction) b1 (↑contractility) b2 (vasodilatation)

Dopamine + ++++ ++++


(low dose) (high dose) (mid or high dose) (mid dose)
Dobutamine 0 + ++++ +
Norepinephrine 0 ++++ ++++ 0
Epinephrine 0 ++++ ++++
Isoproterenol 0 0 ++++++++

ing systemic resistance. High-dose dopamine rine has positive inotropic and chronotropic
is indicated in hypotensive states such as effects. Acting at peripheral α-receptors, it is
shock. However, these doses are inappro- also a potent vasoconstrictor. The increase
priate in most patients with cardiac failure in total peripheral resistance causes the mean
because the peripheral vasoconstriction in- arterial blood pressure to rise.
creases the resistance against which the heart With this combination of effects, norepi-
must contract (i.e., higher afterload), further nephrine is useful in patients suffering from
impairing left ventricular output. “warm shock,” in which the combination of
The major toxicity of dopamine arises in cardiac contractile dysfunction and periph-
patients who are treated with high-dose ther- eral vasodilatation lower blood pressure.
apy. The most important side effects are However, the intense vasoconstriction elic-
acceleration of the heart rate (which increases ited by this drug makes it less attractive than
oxygen consumption) and stimulation of others in treating most other cases of shock.
tachyarrhythmias. Norepinephrine’s side effects include pre-
Dobutamine is a synthetic analog of dopa- cipitation of myocardial ischemia (because
mine that stimulates β1-, β2-, and α-receptors. of the augmented afterload and force of con-
It increases cardiac contractility by virtue of traction) and tachyarrhythmias.
the β1 effect but does not increase peripheral Epinephrine, the predominant endoge-
resistance because of the balance between nous catecholamine produced in the adrenal
α-mediated vasoconstriction and β2-mediated medulla, is formed by the decarboxylation
vasodilation. Thus, it is useful in the treat- of norepinephrine. As indicated in Table 17.2,
ment of heart failure not accompanied by epinephrine is an agonist of α-, β1-, and
hypotension. Unlike dopamine, dobutamine β2-receptors. Administered as an intravenous
does not stimulate dopaminergic receptors infusion at low dosages (<0.01 µg/kg per
(i.e., no renal vasodilating effect), nor does it minute), its stimulation of the β1-receptor in-
facilitate the release of norepinephrine from creases ventricular contractility and speeds
peripheral nerve endings. Like dopamine, it impulse generation. As a result, stroke vol-
is useful for short-term therapy (<1 week), ume, heart rate, and cardiac output increase.
after which time it loses its efficacy, presum- However, at this dosage range, β2-mediated
ably because of downregulation of adrenergic vasodilation may reduce total peripheral re-
receptors. The major adverse effect is the sistance and blood pressure.
provocation of tachyarrhythmias. At higher dosages, epinephrine is a po-
Norepinephrine is an endogenous cate- tent vasopressor because α-mediated con-
cholamine synthesized from dopamine in striction dominates over β2-mediated vaso-
adrenergic postganglionic nerves and in dilation. In this case, the effects of positive
adrenal medullary cells (where it is both a inotropy, positive chronotropy, and vaso-
final product and the precursor of epineph- constriction act together to raise the arte-
rine). Through its β1 activity, norepineph- rial blood pressure.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 404

404 Chapter Seventeen

Epinephrine is therefore used most often Amrinone and milrinone are used in the
when the combination of inotropic and treatment of acute heart failure only if there
chronotropic stimulation is desired, such as has been insufficient improvement with con-
in the setting of cardiac arrest. The α-associ- ventional vasodilators, digitalis, and diuret-
ated vasoconstriction may also help support ics. This is because of the high incidence of
blood pressure in that setting. The most adverse effects, including serious ventricular
common toxic effect is the precipitation arrhythmias. Amrinone has not been shown
of tachyarrhythmias. Epinephrine should to improve the clinical state with chronic use
be avoided in patients receiving β-blocker in heart failure patients, and chronic milri-
therapy, because unopposed α-mediated none therapy has actually demonstrated an
vasoconstriction could produce acute severe increase in mortality rates. Roles for these
hypertension. agents are therefore limited to short-term
Isoproterenol is a synthetic epinephrine therapy in hospitalized patients.
analog. Unlike norepinephrine and epineph- Table 17.3 summarizes the actions and Tab. 3
rine, it is a “pure” β-agonist, having activity toxicities of commonly used inotropic drugs.
almost exclusively at β1- and β2-receptors,
with almost no α-receptor effect. In the
Vasopressin
heart, isoproterenol has positive inotropic
and chronotropic effects, thereby increasing Vasopressin, the endogenous antidiuretic
cardiac output. In peripheral vessels, stimu- hormone secreted by the posterior pituitary,
lation of β2-receptors results in vasodilation primarily functions to maintain water bal-
and reduced peripheral resistance, which ance (see Chapter 9). It acts as a potent non-
may cause blood pressure to fall. adrenergic vasoconstrictor when adminis-
Isoproterenol is sometimes used in emer- tered intravenously at higher-than-natural
gency circumstances to increase the heart doses by directly stimulating vascular smooth
rate in patients with bradycardia or heart muscle V1 receptors. It has proved useful for
block (e.g., as a temporizing measure before maintaining blood pressure in patients with
pacemaker implantation). It may also be types of vasodilatory shock, such as septic
useful in patients with systolic dysfunction shock. It may also be beneficial during cardiac
and slow heart rates with high systemic vas- arrest resuscitation because it increases coro-
cular resistance (a situation sometimes en- nary perfusion pressure, augments blood flow
countered after cardiac surgery in patients to vital organs, and improves the likelihood
who had previously been receiving β-blocker of successful resuscitation in patients with
therapy). Isoproterenol should be avoided in ventricular fibrillation.
patients with myocardial ischemia, in whom
the increased heart rate and inotropic stim-
VASODILATOR DRUGS
ulation would further increase myocardial
oxygen consumption. Vasodilator drugs play a central role in the
treatment of heart failure and hypertension.
As described in Chapter 9, the fall in car-
Phosphodiesterase Inhibitors
diac output in heart failure triggers impor-
Amrinone and milrinone are nondigitalis, tant compensatory pathways, including the
noncatecholamine inotropic agents. They adrenergic nervous system and the renin-
exert their positive inotropic actions by in- angiotensin-aldosterone system (see Fig. 9.9). AQ2
hibiting phosphodiesterase in cardiac myo- As a result of activating these pathways,
cytes (see Fig. 17.4). This inhibition reduces two potent vasoconstrictors are released into
the breakdown of intracellular cAMP, the ul- the circulation: norepinephrine and angio-
timate result of which is enhanced Ca++ entry tensin II. These hormones bind to receptors
into the cell and increased force of contrac- in arterioles and veins, where they cause
tion. These agents also have vasodilating vasoconstriction. Initially, vasoconstriction
properties. is beneficial in heart failure because it maxi-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 405

Cardiovascular Drugs 405

TABLE 17.3. Commonly Used Inotropic Drugs

Drug Mechanism of Action Major Adverse Effects

Cardiac glycosides Inhibition of sarcolemmal Na+K+-ATPase Gastrointestinal: nausea, vomiting


Digoxin Cardiac: atrial, nodal, and ventricular
tachyarrhythmias; high-degree AV
block
Enhanced vagal tone
Sympathomimetic
amines
Dopamine Low dosage (<2 µg/kg per minute): D1 Tachycardia, arrhythmias, hypertension,
receptor stimulation results in mes- drug tolerance
enteric and renal arterial dilatation
(facilitates diuresis)
Medium dosage (2–10 µg/kg per minute):
β1-receptor stimulation and release of
norepinephrine from sympathetic
nerve terminals (inotropic effect)
High dosage (>10 µg/kg per minute):
α-receptor stimulation (peripheral
vasoconstriction)
Dobutamine β1-, β2-, and α-receptor stimulation Tachyarrhythmias, drug tolerance
Phosphodiesterase Increased intracellular cAMP due to inhi- Gastrointestinal: nausea, vomiting
inhibitors bition of its breakdown by phospho-
diesterase
Amrinone Cardiac: arrhythmias
Milrinone (Amrinone only): thrombocytopenia

AV, atrioventricular; cAMP, cyclic adenosine monophosphate; D1, dopamine 1.

mizes left ventricular preload (through ve- Individual vasodilator drug classes act at
nous constriction) and maintains systemic specific vascular sites (Fig. 17.5). Nitrates, for Fig. 5
blood pressure (by arterial constriction). example, are primarily venodilators, whereas
However, venous constriction may ulti- hydralazine is a pure arteriolar dilator. Some
mately cause excessive venous return to the drugs, such as the ACE inhibitors, α-blockers,
heart, with a rise in the pulmonary capil- sodium nitroprusside, and nesiritide are bal-
lary hydrostatic pressure and development anced vasodilators that act on both sides of
of pulmonary congestion. In addition, ex- the circulation.
cessive arteriolar constriction increases the
resistance against which the left ventricle
Angiotensin-Converting
must contract and therefore ultimately im-
Enzyme Inhibitors
pedes forward cardiac output. Vasodilator
therapy is directed at modulating the ex- The renin-angiotensin system plays a critical
cessive constriction of veins and arterioles, role in cardiovascular homeostasis. The major
thus reducing pulmonary congestion and effector of this pathway (Fig. 17.6) is angio- Fig. 6
augmenting forward cardiac output (see tensin II (AII), which is formed by the cleav-
Fig. 9.10). age of angiotensin I by ACE. All the actions of
Vasodilators are also useful antihyperten- AII known to affect blood pressure control are
sive drugs. Recall from Chapter 13 that mediated by its binding to angiotensin II re-
blood pressure is the product of cardiac out- ceptors of the AT1 subtype (see Fig. 13.6).
put and total peripheral resistance (BP = CO Interaction with this receptor generates a
× TPR). Vasodilator drugs decrease arteriolar series of intracellular reactions that cause,
resistance and therefore lower elevated blood among other effects, vasoconstriction and
pressure. the adrenal release of aldosterone, which
10090-17_CH17.qxd 8/31/06 4:58 PM Page 406

406 Chapter Seventeen

Nesiritide
Figure 17.5. Examples of vasodilator drugs and their sites of action: the venous bed, the
arteriolar bed, or both. ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker.

promotes Na+ reabsorption from the distal the distal nephron), and reduce adverse ven-
nephron. As a result of these actions on vas- tricular remodeling (see Chapter 9).
cular tone and sodium homeostasis, AII plays Another action of ACE inhibitors, which
a major role in blood pressure and blood vol- likely contributes to their hemodynamic ef-
ume regulation. By blocking the formation of fects, is related to bradykinin (BK) metabo-
AII, ACE inhibitors decrease the systemic arte- lism, as shown in Figure 17.6. The natural
rial pressure (by decreasing vasoconstriction), vasodilator BK is normally degraded to in-
facilitate natriuresis (e.g., by decreasing aldo- active metabolites by ACE. Because ACE in-
sterone and reducing Na+ reabsorption from hibitors impede that degradation, BK accu-

Vasodilatation

Actions enhanced
by ACE inhibitor

AT1
receptor ACE
antagonist inhibitor

Actions impaired by ACE inhibitor


or AT1 receptor antagonist

Figure 17.6. The renin-angiotensin system. Angiotensin-converting enzyme (ACE) gener-


ates angiotensin II, which results in actions that include vasoconstriction, sodium retention, and
increased sympathetic activity. ACE inhibitors and angiotensin II type 1 (AT1) receptor antago-
nists impair these effects. ACE also promotes the degradation of the natural vasodilator
bradykinin; thus, ACE inhibition—but not AT1 receptor inhibition—results in accumulation of
bradykinin and enhanced vasodilatation.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 407

Cardiovascular Drugs 407

mulates and contributes to the antihyperten- crease preload), and increase cardiac output.
sive effect, likely by stimulating the endothe- The rise in cardiac output usually matches
lial release of nitric oxide and biosynthesis of the fall in peripheral resistance such that
vasodilating prostaglandins. blood pressure tends not to fall (remember,
BP = CO × TPR), except in patients whose
intravascular volume is depleted as might
Clinical Uses result from overly vigorous diuretic ther-
Hypertension apy. The augmented cardiac output reduces
the drive for compensatory neurohormonal
In hypertensive patients, ACE inhibitors stimulation in CHF (see Chapter 9), such
lower blood pressure with little change in car- that elevated levels of norepinephrine fall.
diac output or heart rate. One might assume In addition, clinical trials have shown that
that because this class of drug interferes ACE inhibitors significantly improve sur-
with the renin-angiotensin system, it would vival in patients with chronic heart failure
be effective only in patients with “high- (see Chapter 9) and following myocardial
renin” hypertension, but that is not the case. infarction (see Chapter 7). Some studies
Rather, they are effective in most hyperten- have shown that ACE inhibition also re-
sive patients, regardless of serum renin lev- duces the risk of myocardial infarction and
els. The reason for this is not clear but may death in patients with chronic vascular
relate to the additional antihypertensive disease, including coronary artery disease
effects of BK and vasodilatory prostaglan- (CAD), even if left ventricular function is
dins previously discussed. In addition, renin- not impaired.
angiotensin activity has been demonstrated The available ACE inhibitors are listed in
within tissues outside the circulation, in- Table 17.4. The primary excretory pathway Tab. 4
cluding the walls of the vasculature, where of most of these agents is through the urine,
ACE inhibitors may exert a vasodilatory so their dosages should generally be reduced
effect independent of the circulating renin in patients with renal dysfunction.
concentration.
ACE inhibitors increase renal blood flow,
usually without altering the glomerular
filtration rate (GFR), because of dilation of TABLE 17.4. Drugs That Interfere With
both the afferent and efferent glomerular ar- the Renin-Angiotensin System
terioles. Used alone in hypertension, ACE Drug Major Elimination Pathway
inhibitors show similar efficacy as diuretics
and β-blockers. They do not adversely affect ACE inhibitors
Benazepril Renal
serum glucose or lipid concentrations, and Captopril Renal
unlike diuretics, they do not result in hypo- Enalapril Renal
kalemia. ACE inhibitors are often recom- Fosinopril Hepatic/renal
mended therapy in diabetic hypertensive Lisinopril Renal
Moexipril Hepatic/renal
patients, because the drugs slow the devel-
Perindopril Renal
opment of diabetic nephropathy (a syn- Quinapril Renal
drome of progressive renal deterioration, Ramipril Renal
proteinuria, and hypertension) through fa- Trandolapril Hepatic/renal
vorable effects on intraglomerular pressure. Angiotensin II receptor antagonists
Candesartan Hepatic/renal
Eprosartan Hepatic/renal
Heart Failure Irbesartan Hepatic/renal
Losartan Hepatic/renal
In heart failure, ACE inhibitors reduce pe- Olmesartan Hepatic/renal
ripheral vascular resistance (decrease after- Telmisartan Hepatic
Valsartan Hepatic/renal
load), reduce cardiac filling pressures (de-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 408

408 Chapter Seventeen

AQ3 Toxicity high efferent glomerular arteriolar resistance


(which is highly dependent on AII) to main-
ACE inhibitors are generally well tolerated,
tain intraglomerular pressure and filtration.
and the adverse effects described here are not
Administering an ACE inhibitor abruptly de-
common.
creases efferent arteriolar tone and glomer-
ular hydrostatic pressure and may therefore
Hypotension worsen GFR in this setting.
This is a rare side effect when ACE inhibitors
are used to treat hypertension. It is more Cough
likely to occur in heart failure patients in
Irritation of the upper airways resulting in a
whom intravascular volume depletion has dry cough has been reported in up to 15% of
resulted from vigorous diuretic use. Such patients receiving ACE inhibitor therapy. Its
patients have significant activation of the mechanism has not been established but
renin-angiotensin system; therefore, blood may relate to the increased bradykinin con-
pressure is largely maintained by the vaso- centration provoked by ACE inhibitor ther-
constricting actions of circulating AII. The apy. This side effect may last several weeks
administration of an ACE inhibitor in that after the drug is discontinued.
setting may result in hypotension because
of the sudden reduction of AII levels. This
side effect is avoided by temporarily reduc- Other Effects
ing the diuretic regimen and starting the ACE Very rare adverse reactions to the ACE in-
inhibitor at a low dosage. hibitors include angioedema and agranulo-
cytosis. ACE inhibitors should not be used
Hyperkalemia in pregnancy because they have been shown
to cause fetal injury in the second and third
Because ACE inhibitors indirectly reduce the trimesters.
serum aldosterone concentration, the serum
potassium concentration may rise, but only
rarely into the clinically important hyper-
Angiotensin II Type 1
kalemic range. Conditions that can further Receptor Antagonists
increase serum potassium levels and may re- Angiotensin II type 1 (AT1) receptor antag-
sult in dangerous hyperkalemia during ACE onists, also termed angiotensin receptor
inhibitor use include renal insufficiency, blockers (ARBs), are a second group of drugs
diabetes (caused by hyporeninemic hypo- that interfere with the renin-angiotensin
aldosteronism, a condition often present in system. There are at least two distinct types
elderly diabetics), and concomitant use of of AII receptors: AT1 and AT2. All the actions
potassium-sparing diuretics. of AII known to affect blood pressure control
(e.g., vasoconstriction, aldosterone release,
renal Na+ reabsorption, and sympathetic
Renal Insufficiency
nervous system stimulation) are mediated
Administration of an ACE inhibitor to pa- by its binding to receptors of the AT1 sub-
tients whose intravascular volume is depleted type. The AT2 receptor subtype is abundant
may result in hypotension with decreased during fetal development and has been lo-
renal perfusion and azotemia. Correction cated in some adult tissues, but its precise
of the volume depletion or reduction of the actions are unknown.
ACE inhibitor dosage usually corrects this ARBs compete with AII for AT1 receptors
complication. and therefore inhibit AII-mediated effects AQ4
ACE inhibitor therapy can also precipitate (see Fig. 17.6), thus lowering the blood pres-
renal failure in patients with bilateral renal sure of hypertensive patients. ARBs provide
artery stenosis because these patients rely on a more substantial blockade of the renin-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 409

Cardiovascular Drugs 409

angiotensin system than ACE inhibitors, be- Studies in patients with type 2 diabetes
cause the latter do not completely block for- have demonstrated that ARBs slow the pro-
mation of AII (some AI is converted to AII by gression of kidney disease, an effect that also
circulating enzymes other than ACE). Un- has been demonstrated with ACE inhibitors.
like ACE inhibitors, the AT1 receptor antag-
onists do not affect serum bradykinin levels.
Direct-Acting Vasodilators
The available ARBs are listed in Table 17.4.
Each of these is excreted primarily in the Hydralazine, minoxidil, sodium nitroprus-
bile but most are also partly excreted in the side, and diazoxide are examples of direct-
urine. Trials have demonstrated that ARBs acting vasodilators (Table 17.5). Hydralazine Tab. 5
are as effective as ACE inhibitors in treating and minoxidil are used primarily as long-
hypertension, and they are among the best- term oral vasodilators, whereas nitroprusside
tolerated antihypertensive drugs. As with and diazoxide are administered intravenously
ACE inhibitors, the antihypertensive effect in more-acute settings. Fenoldopam is a
of ARBs is enhanced by concurrent use of a newer arterial vasodilator administered intra-
thiazide diuretic. Also like ACE inhibitors, venously for severe hypertension.
ARBs have the potential side effects of hypo- Hydralazine acts as a potent and direct ar-
tension and hyperkalemia (owing to reduced teriolar dilator at the level of the precapillary
aldosterone levels). Unlike ACE inhibitors, arterioles and has no effect on systemic veins.
ARBs typically do not cause cough. The cellular mechanism of its effect is un-
In the setting of moderate to severe heart known. The fall in blood pressure following
failure, ARBs display hemodynamic bene- arteriolar dilation results in a baroreceptor-
fits similar to those of ACE inhibitors but mediated increase in sympathetic outflow
have not demonstrated superiority over the and cardiac stimulation (e.g., reflex tachy-
latter group (see Chapter 9). Thus, ARBs are cardia), which could precipitate myocardial
generally recommended in heart failure for ischemia in patients with underlying CAD.
patients who are intolerant of ACE inhibi- Therefore, hydralazine is often combined
tors (e.g., because of ACE inhibitor–induced with a β-blocker to blunt this undesired
cough). response.

TABLE 17.5. Direct Vasodilators

Drug Clinical Use Route of Administration Major Adverse Effects

Hydralazine • Hypertension (chronic Oral, intravenous bolus, • Hypotension, tachycardia


and acute therapy) intramuscular
• CHF • Headache, flushing
• Angina
• Drug-induced lupus
Minoxidil • Chronic therapy of Oral • Reflex tachycardia
hypertension • Na+ retention
• Hypertrichosis
• Hypotension
Nitroprusside • Hypertensive emergencies Intravenous infusion
• Acute CHF • Cyanide and thiocyanate
toxicity
Fenoldopam • Hypertensive emergencies Intravenous infusion • Hypotension
• Increased intraocular
pressure
Diazoxide • Hypertensive emergencies Intravenous bolus • Hypotension
• Na+ retention
• Hyperglycemia

CHF, congestive heart failure.


10090-17_CH17.qxd 8/31/06 4:58 PM Page 410

410 Chapter Seventeen

As newer drugs have emerged, hydralazine renal perfusion often results in fluid reten-
is now used only occasionally as an anti- tion, so that a diuretic usually must be ad-
hypertensive, often in combination with ministered concurrently.
other drugs. It can be prescribed concur- Minoxidil’s primary clinical indication is
rently with the venodilator isosorbide dini- in the treatment of severe or intractable
trate to treat heart failure in patients with hypertension. It is especially useful in pa-
systolic dysfunction. This combination im- tients with renal failure who are often re-
proves symptoms in patients with mild-to- fractory to other antihypertensive regimens.
moderate heart failure and has been shown It is well absorbed from the gastrointestinal
to reduce morbidity and mortality rates, most tract and is metabolized primarily by he-
dramatically in African American patients patic glucuronidation, but approximately
(see Chapter 9). one fifth is excreted unchanged by the kid-
Hydralazine possesses low bioavailability ney. Although it has a short half-life, its
because of extensive first-pass hepatic metab- pharmacologic effects persist even after
olism. However, such metabolism depends serum drug concentration falls, probably
on whether the patient displays fast or slow because, like hydralazine, the drug binds
hepatic acetylation; on average, 50% of Amer- avidly to vascular tissue.
icans are fast and 50% are slow acetylators. Side effects of minoxidil, in addition to re-
Slow acetylators show less hepatic degrada- flex sympathetic stimulation and fluid reten-
tion, higher bioavailability, and increased tion, include hypertrichosis (excessive hair
antihypertensive effects, whereas fast acety- growth) and occasional pericardial effusion
lators demonstrate the opposite responses. (unknown mechanism).
Hydralazine has a short half-life (2 to 4 hours) Sodium nitroprusside, a potent dilator
in the circulation, but its effect persists as long of both arterioles and veins, is used intra-
as 12 hours because the drug binds avidly to venously to treat hypertensive emergencies
vascular tissue. and, in intensive care settings, for blood
The most common side effects of hydra- pressure control. It is also prescribed for pre-
lazine include headache (increased cerebral load and afterload modulation in severe
vasodilatation), palpitations (reflex tachy- CHF. Sodium nitroprusside is a complex of
cardia), flushing (increased systemic vaso- iron, cyanide groups, and a nitroso moiety,
dilatation), nausea, and anorexia. As pre- and its metabolism by red blood cells results
viously indicated, tachycardia caused by in the liberation of nitric oxide (Fig. 17.7). Fig. 7
reflex adrenergic stimulation may precipi- Nitric oxide causes vasodilation through
tate anginal attacks in patients with CAD if activation of guanylate cyclase in vascular
hydralazine is not jointly administered with smooth muscle (as described later in this
a β-blocker. Finally, a syndrome similar to chapter; see also Chapter 6).
systemic lupus (characterized by arthralgias, Sodium nitroprusside’s hemodynamic ef-
myalgia, skin rashes, and fever) may de- fects result from its ability to decrease arte-
velop, especially in patients who are slow rial resistance and to increase venous capaci-
acetylators. tance. In patients with normal left ventricular
Minoxidil also results in arteriolar vaso- function, it can actually decrease cardiac out-
dilatation without significant venodilation. put because of the reduction in venous return
Its mechanism of action may involve an in- (see Fig. 9.10). However, in a patient with im-
crease in potassium channel permeability, paired left ventricular contractile function,
which results in smooth muscle cell hyper- the decreased systemic resistance induced by
polarization and relaxation. Like other agents sodium nitroprusside (i.e., decreased after-
that selectively cause arteriolar dilation, re- load) augments forward cardiac output, while
flex adrenergic stimulation leads to increased venous dilation reduces return of blood to the
heart rate and contractility, an undesired heart. The latter decreases pulmonary capil-
effect that can be blunted by coadministra- lary hydrostatic pressure and improves symp-
tion of a β-blocker. In addition, decreased toms of pulmonary congestion.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 411

Cardiovascular Drugs 411

Figure 17.7. Sodium nitroprusside is a complex of iron, cyanide


(CN), and a nitroso group. Erythrocyte metabolism liberates CN and the
active vasodilator nitric oxide. The CN is metabolized in the liver to thio-
cyanate, which is eliminated by the kidneys.

Sodium nitroprusside is often the treat- intravenous antihypertensive agents, it ben-


ment of choice for hypertensive emergen- eficially maintains or enhances renal per-
cies because of its great potency and rapid fusion, and its activation of renal tubular
action. A β-blocker is often administered con- D1 receptors facilitates natriuresis. Unlike
currently to counteract the reflex increase in dopamine, fenoldopam does not stimulate
sympathetic outflow that may accompany α- or β-adrenergic receptors.
the use of this drug. Fenoldopam is administered by continu-
Sodium nitroprusside is administered by ous intravenous infusion. Its onset of ac-
continuous intravenous infusion. Its onset tion is rapid, achieving 50% of maximal ef-
of action begins within 30 sec, and its peak fect within 15 min and steady-state in 30 to
effect is achieved in 2 min. Its effectiveness 60 min. It is metabolized by the liver to in-
dissipates within minutes of its discontinua- active substances that are excreted through
tion. After sodium nitroprusside is metabo- the kidney. It has a rapid offset of action
lized into nitric oxide and cyanide, the liver, after discontinuation (an elimination half-
in the presence of a sulfhydryl donor, trans- life of <10 min), which is a desirable effect
forms cyanide into thiocyanate; the thio- that minimizes the risk of excessive blood
cyanate, in turn, is excreted by the kidney. pressure reduction during the treatment of
Thiocyanate accumulation and toxicity, mani- hypertensive emergencies. These pharma-
fested by blurred vision, tinnitus, disorienta- cologic properties also make fenoldopam
tion, and/or nausea, may occur with contin- useful for controlling hypertension in the
ued use, especially in the setting of renal postoperative setting. However, nitroprus-
impairment. Thus, it is important to moni- side works even faster and remains more
tor serum levels of thiocyanate if sodium popular for this purpose. Unlike nitroprus-
nitroprusside is administered for more than side, fenoldopam does not cause thiocyanate
24 hours. In addition, excessive infusion rates toxicity. The most common side effects are
of sodium nitroprusside, or a deficiency in headache, dizziness, and tachycardia. Fenol-
hepatic thiosulfate stores, can result in lethal dopam also increases intraocular pressure
cyanide toxicity, the early signs of which in- (probably by slowing aqueous humor drain-
clude metabolic acidosis, headache, and nau- age) and should be avoided in patients with
sea, followed by loss of consciousness. glaucoma.
Fenoldopam is a rapidly acting potent Diazoxide is a potent arteriolar dilator
arteriolar vasodilator used intravenously to that is now infrequently used. Its mecha-
treat episodes of severe hypertension. It is a nism of action involves activation of ATP-
selective agonist of peripheral dopamine 1 sensitive potassium channels, leading to ar-
(D1) receptors, the activation of which re- teriolar smooth muscle hyperpolarization
sults in arteriolar vasodilatation through a and vasodilatation. The fall in resistance
cAMP-dependent mechanism. Unlike other leads to a reflex activation of the adrenergic
10090-17_CH17.qxd 8/31/06 4:58 PM Page 412

412 Chapter Seventeen

nervous system with tachycardia, and to fluid Ca++-calmodulin complex. This complex
retention because of activation of the renin- stimulates myosin light chain kinase, which
angiotensin system. It also inhibits pan- phosphorylates myosin light chains and al-
creatic insulin secretion and can result in lows myosin and actin to interact and cause
hyperglycemia. This drug is administered contraction. CCBs promote relaxation of vas-
intravenously and has been used primarily cular smooth muscle by inhibiting Ca++ entry
for hypertensive emergencies. However, its through the voltage-gated channels. Other
use has declined in favor of newer and better- organs possessing smooth muscle (includ-
tolerated agents. ing gastrointestinal, uterine, and bronchio-
lar tissues) are also susceptible to this relax-
ing effect.
Calcium Channel Blockers
The calcium channel blockers (CCBs) are
Cardiac Cells
discussed here as a group, but differences
exist among the drugs of this class. The com- Cardiac muscle also depends on Ca++ influx
mon property of CCBs is their ability to im- during depolarization for contractile protein
pede the influx of Ca++ through membrane interactions, but by a different mechanism
channels in cardiac and smooth muscle cells. than that in vascular smooth muscle. Ca++
Two principal types of voltage-gated Ca++ entry into the cardiac cell during depolar-
channels have been identified in cardiac ization triggers additional intracellular Ca++
tissue, termed L and T. The L-type channel release from the sarcoplasmic reticulum,
is responsible for the Ca++ entry that main- leading to contraction (see Chapter 1). By
tains phase 2 of the action potential (the blocking Ca++ entry, CCBs interfere with
“plateau” in Fig. 1.14). The T-type Ca++ excitation-contraction coupling and decrease
channel likely plays a role in the initial de- the force of contraction. Because the pace-
polarization of nodal tissues. It is the L-type maker tissues of the heart (e.g., sinoatrial [SA]
channel that is antagonized by currently and AV node) are the most dependent on
available CCBs. the inward Ca++ current for depolarization,
one would expect that CCBs would reduce
the rate of sinus firing and AV nodal con-
Mechanisms of Action
duction. Some, but not all, CCBs have this
The cellular mechanism of CCBs has been property (Table 17.6). The effect on cardiac Tab. 6
partly delineated. Increased concentrations conduction appears to depend not only on
of intracellular Ca++ lead to augmented con- whether the specific CCB reduces the in-
tractile force in both myocardium and vas- ward Ca++ current but also on whether it de-
cular smooth muscle. At both sites, the net lays recovery of the Ca++ channel to its pre-
effect of Ca++ channel blockade is to decrease activated state. Verapamil and diltiazem
the amount of Ca++ available to the contrac- have this property, whereas nifedipine and
tile proteins within these cells, which trans- the other dihydropyridine CCBs do not (as
lates into vasodilatation of vascular smooth discussed later).
muscle and a negative inotropic effect in
cardiac muscle.
Clinical Uses
As a result of their actions on vascular
Vascular Smooth Muscle
smooth muscle and cardiac cells, CCBs are
Contraction of vascular smooth muscle de- useful in several cardiovascular disorders
pends on the cytoplasmic Ca++ concentra- through the mechanisms summarized in
tion, which is regulated by the transmem- Table 17.7. In angina pectoris, they exert Tab. 7
brane flow of Ca++ through voltage-gated beneficial effects by reducing myocardial
channels during depolarization. Intracellu- oxygen consumption as well as by potentially
lar Ca++ interacts with calmodulin to form a increasing oxygen supply through coronary
10090-17_CH17.qxd 8/31/06 4:58 PM Page 413

Cardiovascular Drugs 413

TABLE 17.6. Calcium Channel Blockers

Negative Suppress AV
Drug Vasodilation Inotropic Effect Node Conduction Major Adverse Effects

Verapamil + +++ +++ • Hypotension


• Bradycardia, AV block
• CHF
• Constipation
Diltiazem ++ ++ ++ • Hypotension
• Peripheral edema
• Bradycardia
Dihydropyridines
Amlodipine +++ 0 to + 0 • Hypotension
Felodipine • Headache, flushing
Isradipine • Peripheral edema
Nicardipine
Nifedipine
Nisoldipine

AV, atrioventricular; CHF, congestive heart failure.

dilatation. The latter effect is also useful in the excessive vasodilatation) and ankle edema
management of coronary artery vasospasm. (caused by local vasodilatation of peripheral
CCBs are often used to treat hypertension. vascular beds). Verapamil and diltiazem may
More so than β-blockers or ACE inhibitors, result in bradyarrhythmias and should be
CCBs are particularly effective in elderly pa- used with caution in patients already receiv-
tients. Nifedipine and the other dihydro- ing β-blocker therapy.
pyridines are the most potent vasodilators The safety of short-acting CCBs has been
of this class. called into question. In several observa-
CCBs are usually administered orally, and tional studies, a higher incidence of myocar-
once-a-day formulations are available for dial infarction or death has been reported in
most of these agents. Routes of excretion patients with hypertension or coronary dis-
vary. For example, nifedipine and verapamil ease taking such agents. In contrast, these
are eliminated primarily in the urine, whereas adverse outcomes have not been demon-
diltiazem is excreted through the liver. Com- strated with long-acting CCBs (i.e., formula-
mon side effects (see Table 17.6) include tions meant for once-a-day ingestion). Thus,
the development of hypotension (owing to only the long-acting versions should be used

TABLE 17.7. Clinical Effects of Calcium Channel Blockers

Condition Mechanism

Angina pectoris ↓ Myocardial oxygen consumption AQ12


↓ blood pressure
↓ contractility
↓ heart rate (verapamil and diltiazem)
↑ Myocardial oxygen supply
↑ coronary dilatation
Coronary artery spasm Coronary artery vasodilatation
Hypertension Arteriolar smooth muscle relaxation
Supraventricular arrhythmias (Verapamil and diltiazem): Decrease conduction velocity
and increase refractoriness of atrioventricular node via
block-ade of slow inward Ca++ current
10090-17_CH17.qxd 8/31/06 4:58 PM Page 414

414 Chapter Seventeen

in most cases. Also, recall from Chapter 6 Hemodynamic and Antianginal Effects
that β-blockers and/or nitrates are preferred
At low doses, nitroglycerin, the prototypical
over CCBs for initial therapy in patients
organic nitrate, produces greater dilation of
with CAD.
veins than of arterioles. The venodilation re-
sults in venous pooling, diminished venous
Organic Nitrates return, and hence decreased right and left
ventricular filling. Systemic arterial resistance
The nitrates constitute one of the oldest is generally unaffected, but cardiac output
treatments of angina pectoris. They are also may fall because of the diminished preload,
used in other ischemic syndromes and in especially in patients with intravascular vol-
heart failure. The main physiologic action of ume depletion (see Fig. 9.10). Arterial dila-
the nitrates is vasodilatation, particularly of tion occurs to some extent in the coronary
the systemic veins. arteries and may also occur in the facial ves-
sels and the meningeal arterioles, giving rise
to the side effects of flushing and headache,
Mechanism of Action
respectively.
Nitrates produce vascular smooth muscle re- At high doses, nitrates result in widespread
laxation. The proposed mechanism involves arteriolar dilation and venodilation. Arteri-
the conversion of the administered drug to olar dilation may result in systemic hypo-
nitric oxide at or near the plasma membrane tension and reflex tachycardia. However,
Fig. 8 of vascular smooth muscle cells (Fig. 17.8). the increase in heart rate is not typically
Nitric oxide, in turn, activates guanylate manifest in patients with heart failure, be-
cyclase to produce cyclic guanosine mono- cause decreasing afterload in that situation
phosphate (cGMP), and the intracellular may actually improve cardiac output and re-
accumulation of cGMP leads to smooth mus- duce the sympathetic drive.
cle relaxation. This mechanism of vascular The major use of nitrates is in the treat-
smooth muscle relaxation is similar to that as- ment of angina pectoris, in which the re-
sociated with nitroprusside and endogenous duction of left ventricular filling reduces pre-
endothelial-derived nitric oxide. load. The smaller left ventricular size lowers

Figure 17.8. Organic nitrates incite vascular smooth muscle (SM)


relaxation by conversion to nitric oxide (NO) at or near the cell
membrane. Nitroprusside and endothelial-dependent vasodilators also
promote NO delivery to vascular smooth muscle and cause relaxation. In
the SM, NO stimulates formation of cyclic guanosine monophosphate
(cGMP), which mediates relaxation.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 415

Cardiovascular Drugs 415

ventricular wall stress and myocardial oxy- Toxicity AQ5


gen consumption, which alleviates the oxy-
The most common adverse effects of the
gen imbalance in ischemic states. Nitrates are
nitrates include hypotension, reflex tachy-
also useful in patients with coronary artery
cardia, headache, and flushing.
spasm (Prinzmetal variant angina) by dilat-
ing the coronary arterioles.
Natriuretic Peptides
Agents and Pharmacokinetics
As described in Chapter 9, natriuretic pep-
Many formulations of nitrates are available. tides are naturally secreted from atrial and
When the relief of acute angina is the objec- ventricular myocardium in patients with
tive, rapid onset of action is essential. How- heart failure. Among their beneficial physi-
ever, in the long-term prevention of angi- ologic effects, these peptides promote vaso-
nal attacks in a patient with chronic CAD, dilation and result in sodium and water ex-
duration of action and predictability of ef- cretion. The pharmacologic agent nesiritide
fect are more crucial than the speed of drug (human recombinant B-type natriuretic pep-
effect. tide) is available for intravenous administra-
Sublingual nitroglycerin tablets or sprays tion to hospitalized patients with decompen-
are used in the treatment of acute angina at- sated heart failure. It results in vasodilation,
tacks. The peak action of these agents occurs augmented cardiac output, and reduction
within 3 min, because they are rapidly ab- of the undesired activation of the renin-
sorbed into the bloodstream via the oral angiotensin and sympathetic nervous sys-
mucosa; their effect, however, diminishes tems that are typical in heart failure. In some
rapidly, falling off within 15 to 30 min, as the patients, it promotes diuresis.
drug is deactivated in the liver. These forms of Nesiritide binds to G protein–coupled re-
nitroglycerin are also effective when taken ceptors in multiple tissues, including the
prophylactically, immediately before situa- blood vessels (resulting in vasodilation), kid-
tions known by the patient to produce angina neys, and adrenals. In the kidney, natriuresis
(e.g., before walking up a hill). is a consequence of several effects of the
The “long-acting” nitrates are used to pre- drug. An augmented glomerular filtration
vent chest pain in the chronic management rate results from dilation of the afferent re-
of angina and must be given in sufficient nal arterioles and constriction of the effer-
dosage to saturate the liver’s deactivating ca- ent renal arterioles, thereby increasing the
pacity. In this situation, high oral doses of filtered load of sodium. In the proximal
sustained-release nitroglycerin, isosorbide tubule, AII-mediated sodium uptake is in-
dinitrate, or isosorbide mononitrate are hibited. Because the proximal tubule is where
routinely used. These agents have a dura- the vast majority of sodium is reabsorbed
tion of action of 2 to 14 hours. Transdermal (as described later in the chapter), this inter-
nitroglycerin patches or nitroglycerin paste ruption in uptake results in sodium excre-
applied to the skin also deliver a sustained tion. In the distal tubule, natriuretic peptides
release of nitroglycerin. Of note, the efficacy appear to further reduce sodium reabsorp-
of long-acting nitrate therapy is attenuated tion through epithelial sodium channels.
by the rapid development of drug tolerance In the adrenal zona glomerulosa, the drug
with continuous use. For this reason, it is inhibits aldosterone synthesis, which leads
important that the dosing regimens allow a to enhanced sodium excretion in the distal
drug-free interval of several hours each day nephron.
to maintain efficacy. Despite these benefits, the clinical role of
Intravenous nitroglycerin is adminis- nesiritide is still being defined, because its
tered by continuous infusion is most useful use has not been shown to improve survival
in the treatment of hospitalized patients with in heart failure patients and in one study was
unstable angina or acute heart failure. actually associated with increased mortality.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 416

416 Chapter Seventeen

Sildenafil therefore, these groups of drugs should not


be prescribed concurrently.
Sildenafil, a phosphodiesterase type 5 in-
hibitor used to treat erectile dysfunction, has
been shown to decrease pulmonary vascular ANTIADRENERGIC DRUGS
resistance in patients with primary pulmo-
nary hypertension (PPH). It inhibits the Drugs that interfere with the sympathetic
breakdown of cGMP in the pulmonary vas- nervous system are used commonly to treat
culature, which enhances vasodilation and cardiovascular disorders. These agents act
oxygenation. Other phosphodiesterase in- at different loci, including the central ner-
hibitors have not been shown to be effective vous system (CNS), postganglionic sympa-
in PPH. When combined with nitrates, thetic nerve endings, and peripheral α- and
sildenafil can cause severe hypotension; β-receptors (Fig. 17.9). Fig. 9

Figure 17.9. Sites of action of the antiadrenergic drugs. Note that receptors at the sympathetic nerve
ending bind norepinephrine (NE) and provide feedback: the β-receptor stimulates, and the α2-receptor in-
hibits, further NE releases. CNS, central nervous system.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 417

Cardiovascular Drugs 417

Normally, when a sympathetic nerve is used antihypertensive drugs but have largely
stimulated, norepinephrine is released, tra- given way to better-tolerated agents. They
verses the synapse, and stimulates postsynap- are not sufficiently potent to serve as vaso-
tic α- and β-receptors. The consequences of dilators in the treatment of heart failure.
receptor stimulation depend on the organ The drugs in this group are listed in Fig-
Tab. 8 involved (Table 17.8). The effect of α-receptor ure 17.9. They are all available as oral prepa-
stimulation on vascular smooth muscle is rations, and clonidine can also be prescribed
vasoconstriction, whereas β2 stimulation as a skin patch that is applied and left in
causes vasodilatation. In the CNS, α2 stim- place for 1 week at a time, facilitating drug
ulation inhibits sympathetic outflow to the compliance in the treatment of hyperten-
periphery, thereby contributing to vaso- sion. Side effects of CNS α2-agonists include
dilatation. sedation, dry mouth, bradycardia, and if the
In addition, norepinephrine within the drug is stopped suddenly, the possibility of
synapse can bind to presynaptic β- and α2- a sudden, paradoxical rise in blood pressure.
receptors, which provides a feedback mech-
anism that modulates further release of the
Sympathetic Nerve-Ending
hormone. The β-receptor increases and the
Antagonists
α2-receptor inhibits further norepinephrine
release. Reserpine was the first drug found to inter-
fere with the sympathetic nervous system. It
inhibits the uptake of norepinephrine into
Central Adrenergic Inhibitors
storage vesicles in postganglionic and central
α2-Receptors are located in the presynaptic neurons, leading to norepinephrine degra-
neurons of the CNS. When stimulated by an dation. The antihypertensive effect results
α2-agonist, they lead to diminished sympa- from the depletion of catecholamines, which
thetic outflow from the medulla. This action causes the force of myocardial contraction
reduces peripheral vascular resistance and and total peripheral resistance to decrease.
decreases cardiac stimulation, resulting in a Reserpine’s CNS toxicity represents its
fall in blood pressure and heart rate. Thus, chief drawback. It often produces sedation
drugs known as central adrenergic inhibitors and can impair concentration. The most seri-
are actually agonists of the CNS α2-receptors. ous potential toxicity is psychotic depres-
They were once among the most commonly sion, and patients with a history of depressive

TABLE 17.8. Responses to Adrenergic Receptor Stimulation


Receptor
Type Distribution Response

α1 Vascular smooth muscle (arterioles and veins) Vasoconstriction


α2 Presynaptic adrenergic nerve terminals Inhibition of NE release
Vascular smooth muscle (coronary and renal arterioles) Vasoconstriction
β1 Heart Increases heart rate
Increases contractility
Speeds AV node conduction
Kidney (JG cells) Increases renin release
Presynaptic adrenergic nerve terminals Increases NE release
Adipose tissue Stimulates lipolysis
β2 Vascular smooth muscle (arterioles, except skin and Vasodilation
cerebral)
Bronchial smooth muscle Bronchodilation
Liver Stimulates glycogenolysis

AV, atrioventricular; NE, norepinephrine.


10090-17_CH17.qxd 8/31/06 4:58 PM Page 418

418 Chapter Seventeen

disorders should not receive this drug. than those treated with a thiazide diuretic.
Newer, better-tolerated antihypertensive Thus, α1-antagonists have fallen out of favor
agents have largely supplanted the use of re- in the management of hypertension. The
serpine and other sympathetic nerve-ending drugs have also been evaluated in the treat-
antagonists. ment of heart failure; however, they lose
their effectiveness over time (i.e., display
drug tolerance) and, unlike other vasodila-
Peripheral a-Adrenergic
tor regimens (e.g., ACE inhibitors or hydra-
Receptor Antagonists lazine plus nitrates), do not reduce mortal-
Tab. 9 Peripheral α-antagonists (Table 17.9) are di- ity rates in chronic CHF. Terazosin and
vided into those that act on both α1- and doxazosin are mainly used today to treat the
α2-receptors, and those that inhibit α1 alone. symptoms of benign prostatic hyperplasia,
α1-Selective receptor antagonists (prazosin, because the drugs also beneficially relax
terazosin, doxazosin) are occasionally pre- prostatic smooth muscle tone.
scribed in the treatment of hypertension. Phentolamine and phenoxybenzamine
Their selectivity for the α1-receptor explains are nonselective α-blockers. They are used
their ability to produce less reflex tachy- in the treatment of pheochromocytoma, a
cardia than nonselective agents. Normally, tumor that abnormally secretes catechol-
drug-induced vasodilatation results in baro- amines into the circulation (see Chapter 13).
receptor-mediated stimulation of the sym- Otherwise, these drugs are rarely used be-
pathetic nervous system and an undesired cause the α2-blockade impairs the normal
increase in heart rate. This effect is ampli- feedback inhibition of norepinephrine re-
fied by drugs that block the presynaptic lease, an undesired effect, as indicated earlier.
α2-receptor, because feedback inhibition of
norepinephrine release is prevented. How-
b-Adrenergic
ever, α1-selective agents do not block the
Receptor Antagonists
negative feedback on the α2-receptor. Thus,
further norepinephrine release and reflex The β-adrenergic antagonists are used for
sympathetic side effects are blunted. a number of cardiovascular conditions, in-
Historically, the principal indication for cluding ischemic heart disease, hyperten-
β1-antagonists has been in the treatment of sion, heart failure, and tachyarrhythmias.
hypertension. One of their advantages is Because catecholamines increase inotropy,
that they do not adversely affect the serum chronotropy, and conduction velocity in the
concentrations of cholesterol and triglyc- heart, it follows that β-receptor antagonists
erides as can other antihypertensives, such decrease inotropy, slow the heart rate, and
as diuretics and β-blockers. However, in a decrease conduction velocity. When stimula-
large prospective, randomized trial, patients tion of the β-receptors is low, as in a normal
treated with the α1-antagonist doxazosin ex- resting person, the effect of blocking agents
perienced more adverse cardiac outcomes is likewise mild. However, when the sympa-

TABLE 17.9. a-Receptor Antagonists

Mechanism/Drug Indications Major Adverse Effects

Selective peripheral a1-blockade • Hypertension • Postural hypotension


Prazosin • Benign prostatic hyperplasia • Headache, dizziness
Terazosin • No reflex tachycardia
Doxazosin
Nonselective a-blockade • Pheochromocytoma • Postural hypotension
Phentolamine • Reflex tachycardia
Phenoxybenzamine • Arrhythmias
10090-17_CH17.qxd 8/31/06 4:58 PM Page 419

Cardiovascular Drugs 419

thetic nervous system is activated (e.g., dur- to decrease myocardial oxygen demand (see
ing exercise), these antagonists can sub- Chapter 6). They reduce the heart rate, blood
stantially diminish catecholamine-mediated pressure (afterload), and contractility. The
effects. negative inotropic effect is directly related to
The β-blockers can be distinguished from blockade of the cardiac β-receptor, which
one another by specific properties (Table results in decreased calcium influx into the
Tab. 10 17.10): (1) the relative affinity of the drug myocyte (see Fig. 17.4). β-Blockers also im-
for β1- and β2-receptors, (2) whether partial prove survival and reduce the rate of reinfarc-
β-agonist activity is present, (3) whether tion following an acute myocardial infarc-
α1-receptors are also blocked, and (4) differ- tion. Agents with intrinsic sympathomimetic
ences in pharmacokinetic properties. The activity are less beneficial in this regard than
goal of β1-selective agents is to achieve myo- β-blockers without it.
cardial receptor blockade, with less effect on
bronchial and vascular smooth muscle (tis- Hypertension
sues that exhibit β2-receptors), thus produc-
ing less bronchospasm and vasoconstriction β-Blocking agents are effective antihyperten-
in susceptible patients. Agents with partial sive agents. Despite their widespread use in
β-agonist effects (also termed intrinsic sym- this capacity, the mechanisms responsible
pathomimetic activity) tend to slow the heart for blood pressure lowering are not com-
rate less than other β-blockers. pletely understood. With initial use, the anti-
During short-term use, nonselective hypertensive action is thought to result from
β-antagonists tend to reduce cardiac output a decrease in cardiac output, in association
because they decrease heart rate and con- with slowing of the heart rate and mild de-
tractility as well as slightly increase periph- crease in contractility. However, with
eral resistance (via β2-receptor blockade). β- chronic administration, other mechanisms
antagonists that have partial agonist activity are likely at work, including reduced renal
(such as pindolol) or those that possess some secretion of renin and possibly CNS effects.
α-blocking activity (such as labetalol) can ac-
tually lower peripheral resistance by interact- Heart Failure
ing with their respective β2- and α-receptors.
The negative inotropic effect of β-blockade
would be expected to worsen heart failure
Clinical Uses symptoms in patients with underlying left
ventricular systolic dysfunction. However,
Ischemic Heart Disease
trials in patients with all classes of clini-
The beneficial effects of β-blockers in isch- cally stable heart failure have actually shown
emic heart disease are related to their ability a survival benefit with chronic β-blocker

TABLE 17.10. b-Adrenergic Blockers

Activity Nonselective b-Blockers b1-Selective b-Blockers

No β-agonist activity Carvedilola Atenolol


Labetalola Betaxolol
Propranolol Bisoprolol
Nadolol Esmololb
Timolol Metoprolol
β-agonist activity Carteolol Acebutolol
Penbutolol
Pindolol
a
Also has α1-adrenergic blocking properties.
b
Administered intravenously only.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 420

420 Chapter Seventeen

administration using carvedilol, metoprolol, hypoglycemia, such as tachycardia. If β-


or bisoprolol (see Chapter 9). The mech- blockers are used in diabetics, β1-selective
anism may relate to blunting of the cardio- agents are generally preferred.
toxic effects of excessive circulating cate-
Other potential side effects include insom-
cholamines. Because of the potential risk of
nia, depression, and impotence. Finally, β-
actually transiently worsening heart failure in
antagonists should be used with caution in
tenuous patients, β-blocker therapy should
combination with nondihydropyridine CCBs
be started at low dosage, augmented slowly,
(verapamil or diltiazem), because both types
and carefully monitored.
of drugs can impair myocardial contractility
Other conditions that benefit from β-
and AV nodal conduction, possibly precipitat-
blocker therapy include tachyarrhythmias
ing heart failure or AV conduction blocks.
(as discussed later in the chapter) and hyper-
trophic cardiomyopathy (see Chapter 10).
ANTIARRHYTHMIC DRUGS
Toxicity Drug therapy is a common approach to
treat cardiac tachyarrhythmias. However,
Fatigue may occur during β-blocker therapy
despite their benefits, antiarrhythmic drugs
and is most likely a CNS side effect. β-Blockers
are among the most dangerous pharmaco-
with less lipid solubility (e.g., nadolol) do not
logic agents because of their frequent seri-
penetrate the blood-brain barrier and may
ous adverse effects. Therefore, a thorough
have fewer CNS adverse effects than more
understanding of their mechanisms of action,
lipid-soluble drugs, such as propranolol.
indications, and toxicities is of particular
Other potential adverse effects relate to the
importance.
predictable consequences of β-blockade:
Although a number of classification sys-
1. β2-Blockade associated with use of non- tems for these agents exist, antiarrhythmic
selective agents (or large doses of β1- drugs are commonly separated into four
selective blockers) can exacerbate broncho- groups based on their electrophysiologic
spasm, worsening preexisting asthma or mechanisms of action (Table 17.11): Tab. 11
chronic obstructive lung disease.
1. Class I drugs block the fast sodium chan-
2. The impairment of AV nodal conduction
nel responsible for phase 0 depolarization
by β1-blockade can provoke conduction
of the action potential. They are further
blocks.
divided into three subtypes based on the
3. β2-Blockade can precipitate arterial vaso- degree of sodium channel blockade and
spasm, which can result in Raynaud phe- the effect of the drug on the cell’s action
nomenon or worsen symptoms of pe- potential duration.
ripheral vascular disease.
2. Class II drugs are β-adrenergic receptor
4. Abrupt withdrawal of a β-antagonist after
antagonists (β-blockers).
chronic use could precipitate myocardial
3. Class III drugs significantly prolong the
ischemia in patients with CAD.
action potential with little effect on the
5. Undesirable reduction of high-density
rise of phase 0 depolarization. The main
lipoprotein (HDL) cholesterol and eleva-
mechanism is blockade of the repolariz-
tion of triglycerides can occur through an
ing K+ current.
unknown mechanism. This effect appears
to be less pronounced with blockers that 4. Class IV drugs block the slow L-type
have partial β-agonist activity or com- calcium channel.
bined β- and α-blocking properties. Drugs that do not conveniently fit into these
6. β2-Blockade may impair recovery from classes (and are discussed separately) include
hypoglycemia in diabetics suffering an in- adenosine and the digitalis glycosides.
sulin reaction. In addition, β-blockers may Regardless of the class, the goal of anti-
mask the sympathetic warning signs of arrhythmic therapy is to abolish the mech-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 421

Cardiovascular Drugs 421

TABLE 17.11. Classification of Antiarrhythmic Drugs

Class General Mechanism Examples

I Na1 channel blockade


IA Moderate block (↓↓Phase 0 upstroke rate; prolonged AP duration) Quinidine
Procainamide
Disopyramide
IB Mild block (↓Phase 0 upstroke rate; shortened AP duration) Lidocaine
Mexiletine
IC Marked block (↓↓↓Phase 0 upstroke rate; no change in AP duration) Flecainide
Propafenone
II b-adrenergic receptor blockade Propranolol
Esmolol
Metoprolol
Others
III Prolongation of action potential duration Amiodarone
(predominantly via K+ channel blockade) Sotalol
Bretylium
Ibutilide
Dofetilide
IV Ca11 channel antagonists Verapamil
Diltiazem

AP, action potential

anisms by which tachyarrhythmias occur the refractory period is pharmacologically


(see Chapter 11). These mechanisms are prolonged, a propagating impulse confronts
(1) increased automaticity of pacemaker or inactive sodium channels, cannot conduct
nonpacemaker cells, (2) reentrant pathways, further, and is extinguished.
and (3) triggered activity. A second means to interrupt reentrant
In the case of arrhythmias caused by in- circuits is to additionally impair impulse pro-
creased automaticity, treatment is aimed at pagation within the already slowed retro-
lowering the maximum frequency at which grade limb. This is accomplished via phar-
cardiac action potentials can occur by (1) re- macologic blockade of the Na+ channels
ducing the slope of spontaneous phase 4 di- responsible for phase 0 depolarization. Such
astolic depolarization and/or (2) prolonging blockade fully abolishes the compromised
the effective refractory period. These actions impulse conduction within the retrograde
reduce or extinguish abnormally high rates limb and breaks the self-sustaining loop.
of firing.
Antiarrhythmic drugs inhibit reentrant
rhythms by a different mechanism. The ini-
A.
tiation of a reentrant circuit relies on a re- B.
Prolong
gion of unidirectional block and slowed con- Impair conduction
refractory
even more
Fig. 10 duction (Fig. 17.10). For a reentrant rhythm period
to sustain itself, the length of time it takes
for an impulse to propagate around the cir-
cuit must exceed the effective refractory pe-
riod of the tissue. If an impulse returns to an
area of myocardium that was depolarized
moments earlier but has not yet recovered Figure 17.10. Two strategies to interrupt reentry. A.
excitability, it cannot restimulate that tis- Prolonging the tissue refractory period causes returning
impulses to find the tissue unexcitable. B. Further reduc-
sue. Thus, one strategy to stop reentry is to ing conduction causes the impulse to “die out” in the
lengthen the tissue’s refractory period. When slow retrograde limb of the circuit.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 422

422 Chapter Seventeen

The elimination of the third type of duces cellular and tissue conduction veloc-
tachyarrhythmia, triggered activity, requires ities. If impaired sufficiently within a re-
suppression of early and delayed after- entrant circuit, the impulse will die out
depolarizations. within the already slowed retrograde limb,
An ideal pharmacologic agent would sup- aborting the rhythm. In addition, class IA
press ectopic foci and interrupt reentrant agents prolong the cell’s refractory period,
loops without affecting normal conduction both by lengthening the action potential and
pathways. Unfortunately, when the con- by dissociating relatively slowly from Na+
centrations of antiarrhythmic drugs exceed channels after repolarization (see Fig. 17.11).
their narrow therapeutic ranges, even nor- Thus, an impulse traveling in the reentrant
mal electrical activity may become sup- loop encounters unexcitable tissue and is
pressed. In addition, most antiarrhythmic extinguished.
drugs have the potential to aggravate rhythm
disturbances (termed proarrhythmic effect).
Effect on the Electrocardiogram
For example, this may occur when an anti-
arrhythmic drug prolongs the action poten- Because the conduction velocity is decreased
tial and induces early afterdepolarizations, and the action potential duration and repo-
resulting in a triggered-type of arrhythmia, larization are prolonged, the effect of class
such as torsades de pointes (see Chapter 12). IA agents is to mildly prolong the QRS and
Drug-induced proarrhythmia occurs most QT intervals (Table 17.12). At higher dos- Tab. 12
often in patients with left ventricular dys- ages, the drugs may substantially lengthen
function or in those with an increased QT these intervals, potentially setting the stage
interval (a sign that the action potential is for afterdepolarizations and drug-induced
prolonged). arrhythmias.

Class IA Antiarrhythmics Clinical Uses


Mechanisms of Action Class IA drugs are effective in treating vari-
ous reentrant and ectopic supraventricular
Effect on Arrhythmias Caused
and ventricular tachycardias (Table 17.13). Tab. 13
by Increased Automaticity
However, their use has declined because of
Class IA agents produce moderate blockade the development of more effective and less
of the fast sodium channels, thus raising proarrhythmic strategies, as discussed later
the threshold potential and slowing the up- in the chapter.
stroke (phase 0) of the action potential. In
addition, perhaps by inhibition of pacemaker
Specific Class IA Drugs
channels, the slope of phase 4 depolariza-
Fig. 11 tion is depressed (Fig. 17.11) so that it takes Quinidine displays the electrophysiologic
longer to reach threshold and fire the action effects inherent to class IA agents but also
potential. These effects are most pronounced has anticholinergic properties that may aug-
at Purkinje fibers and abnormal ectopic pace- ment conduction at the AV node, thus antag-
makers. Because IA agents have little effect onizing its direct suppressant effect. Because
on the automaticity of the SA node, the lat- of this increased AV nodal conduction,
ter can resume its function as the cardiac quinidine is often combined with a negative
pacemaker after ectopic foci are suppressed. chronotropic agent such as a β-blocker, verap-
amil, diltiazem, or digoxin. Quinidine also
displays an α-adrenergic blocking action that
Effect on Reentrant Arrhythmias
may cause hypotension, especially with par-
Because sodium channel blockade slows the enteral intravenous administration. There-
rate of phase 0 depolarization by reducing fore, it is administered only by the oral route.
the magnitude of the inward current, it re- Because quinidine is metabolized primarily
10090-17_CH17.qxd 8/31/06 4:58 PM Page 423

Cardiovascular Drugs 423

Figure 17.11. Electrophysiologic effects of the class I antiarrhythmic drugs. A. Effect on the Purkinje cell action
potential. B. Effect on pacemaker cell action potential.

by the liver, its dosage must be reduced in pa-


TABLE 17.12. Effect of Antiarrhythmic
tients with hepatic dysfunction.
Drugs on Electrocardiographic
Intervals Cardiac and noncardiac side effects occur
frequently during quinidine therapy. The
Class PR QRS QT most common are related to the gastro-
IA 0 ↑ ↑ intestinal tract, including diarrhea in one
IB 0 0 0 or ↓ third of patients. Quinidine can cause ex-
IC ↑ ↑ 0 or ↑ cessive prolongation of the QT interval,
II 0 or ↑ 0 0 or ↓ which may lead to the life-threatening
III 0 or ↑ 0 or ↑ ↑
ventricular tachyarrhythmia torsades de
IV ↑ 0 0
pointes, described in Chapter 12. In patients
10090-17_CH17.qxd 8/31/06 4:58 PM Page 424

424 Chapter Seventeen

changed in the urine; the remainder under-


TABLE 17.13. Common Clinical Uses goes acetylation by the liver to form N-acetyl
of Antiarrhythmic Drugs
procainamide (NAPA), which is subsequently
Class Use excreted by the kidneys. In renal failure, or
in patients who are rapid acetylators, high
IA • Atrial fibrillation and flutter
• Paroxysmal SVT serum levels of NAPA may accumulate.
• Ventricular tachycardia NAPA shares procainamide’s ability to pro-
IB • Ventricular tachycardia long the action potential and refractory pe-
• Digitalis-induced arrhythmias riod, but it does not alter the rate of phase 4
IC • Atrial fibrillation and paroxysmal SVT
II • Atrial or ventricular premature beats
depolarization or the slope of phase 0 up-
• Paroxysmal SVT stroke of the action potential.
• Atrial fibrillation and flutter Noncardiac side effects of procainamide
• Ventricular tachycardia (ischemia-related) are common and include fever and rash. Ap-
III • Ventricular tachycardia (amiodarone and
proximately one third of patients develop a
sotalol)
• Atrial fibrillation and flutter systemic lupus–like syndrome after 6 months
• Bypass-tract mediated paroxysmal SVT of therapy, manifested by arthralgias, rash,
(amiodarone) and connective tissue inflammation. It most
IV • Paroxysmal SVT often occurs among patients who are slow
• Atrial fibrillation and flutter (↓ VR)
• Multifocal atrial tachycardia (↓ VR)
acetylators and is reversible on cessation of
drug therapy.
SVT, supraventricular tachycardia; VR, ventricular rate. Disopyramide’s electrophysiologic and
antiarrhythmic effects are similar to those
of quinidine. However, disopyramide causes
much fewer gastrointestinal side effects
taking digoxin, quinidine raises the blood
and does not increase serum digoxin levels.
level of that drug because of reductions in
Also, disopyramide has a much greater anti-
the body’s clearance and volume of distri-
cholinergic effect, so that common side ef-
bution. Thus, it is important to reduce the
dose of digoxin when quinidine is added. fects include constipation, urinary retention,
The electrophysiologic effects of pro- and exacerbation of glaucoma. More so than
cainamide are similar to those of quinidine, quinidine or procainamide, disopyramide
though procainamide does not prolong has a pronounced negative inotropic effect
the action potential (and therefore QT in- and must be used with caution in patients
terval) as much. Nonetheless, the danger- with left ventricular systolic dysfunction.
ous arrhythmia torsades de pointes can still Disopyramide is administered orally. The
be provoked. Procainamide also has less primary excretory pathway is via the kid-
pronounced anticholinergic effects than neys, and toxic levels may accumulate in
quinidine, so that facilitation of AV nodal patients with renal insufficiency. QT pro-
conduction is less significant. longation and precipitation of ventricular
Procainamide has mild ganglionic block- arrhythmias (including torsades de pointes)
ing effects that may cause peripheral vaso- can occur.
dilatation and a negative cardiac inotropic
effect, particularly when the drug is ad-
Class IB Antiarrhythmics
ministered intravenously. However, because
hypotension associated with intravenous Class IB drugs inhibit the fast sodium chan-
procainamide is much less common than nel, but unlike IA agents, they typically
with quinidine, it is used when an intra- shorten the action potential duration and the
venous class IA agent is desired. refractory period. Such shortening is attrib-
Procainamide can be administered by uted to blockade of small sodium currents
mouth, intramuscularly, or intravenously. that normally continue through phase 2 of
More than 50% of the drug is excreted un- the action potential.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 425

Cardiovascular Drugs 425

Class IB drugs at therapeutic concentra- half-life of the drug depends greatly on he-
tions do not substantially alter the electrical patic blood flow. Reduced flow (as in heart
activity of normal tissue; rather, they pref- failure or in older individuals) or intrinsic
erentially act on diseased or ischemic cells. liver disease can greatly increase serum li-
Conditions present during ischemia—such docaine concentrations and toxic effects;
as acidosis, faster rates of cell stimulation, therefore, the infusion rate should be low-
and increased extracellular potassium con- ered in such patients.
centration (and consequently a less negative The most common side effects of lido-
diastolic membrane potential)—increase the caine are not cardiac; rather, they are related
ability of class IB drugs to block the sodium to the CNS and include confusion, dizzi-
channel. This blockade promotes conduc- ness, and seizures. These effects are dosage
tion block in ischemic cells by reducing the related and can be prevented by monitoring
slope of phase 0 depolarization and slow- serum levels of the drug or preemptively re-
ing the conduction velocity, thus inhibit- ducing the infusion rate when liver disease
ing reentrant arrhythmias (see Fig. 17.11). or decreased hepatic blood flow is suspected.
Similar to other class I drugs, the auto- Mexiletine is structurally similar to lido-
maticity of ectopic pacemakers is also sup- caine and shares its electrophysiologic prop-
pressed by decreasing phase 4 spontaneous erties, but mexiletine is administered orally.
depolarization and (in the case of some Ninety percent of mexiletine is metabolized
drugs of this class) by raising the threshold in the liver to inactive products, and the
potential. In addition, intravenous lidocaine, dosage of the drug should be reduced in
a member of this class, suppresses delayed patients with hepatic dysfunction. Dose-
afterdepolarizations. related side effects of mexiletine are com-
The most common use of class IB drugs is mon, especially of the CNS (dizziness, tre-
in the suppression of ventricular arrhyth- mor, slurred speech) and the gastrointestinal
mias, especially those that appear in asso- tract (nausea, vomiting).
ciation with ischemia or digitalis toxicity.
Conversely, they have little effect on atrial
Class IC Antiarrhythmics
tissue at therapeutic concentrations because
of the shorter action potential duration of The class IC drugs are the most potent sod-
atrial cells, which allows less time for the ium channel blockers. They markedly de-
drug to bind and block the Na+ channel. crease the upstroke of the action potential
Thus, these agents are ineffective in atrial and conduction velocity in atrial, ventricu-
fibrillation, atrial flutter, and supraventricu- lar, and Purkinje fibers (see Fig. 17.11). Al-
lar tachycardias. though they have little effect on the duration
Because the QT interval is not prolonged of the action potential or refractory period of
by class IB drugs, early afterdepolarizations Purkinje fibers, they significantly prolong the
do not occur, and torsades de pointes is not refractory period within the AV node and in
an expected complication. accessory bypass tracts.
The group IC agents were originally de-
veloped to treat ventricular arrhythmias.
Specific Class IB Drugs
However, that use has diminished because
Lidocaine is an antiarrhythmic drug com- studies have shown an increased mortality
monly used acutely to suppress ventricular rate in patients taking class IC drugs for ven-
arrhythmias in hospitalized patients. It is tricular ectopy following myocardial infarc-
administered intravenously only, because tion and in those who have survived cardiac
oral administration results in unpredictable arrest. In patients with underlying left ven-
plasma levels. As a result of rapid distribu- tricular dysfunction, class IC drugs can pre-
tion and hepatic metabolism, lidocaine must cipitate heart failure. Thus, drugs of this sub-
be administered as a continuous infusion class should be avoided in patients who
following two or three loading boluses. The have other underlying heart abnormalities,
10090-17_CH17.qxd 8/31/06 4:58 PM Page 426

426 Chapter Seventeen

such as CAD or ventricular dysfunction. Class II Antiarrhythmics


Class IC drugs have been shown to be bene-
The class II drugs are β-adrenergic receptor
ficial (and reasonably safe) in preventing
antagonists, which are used in the man-
supraventricular arrhythmias in patients who
agement of both supraventricular and ven-
have otherwise structurally normal hearts
tricular arrhythmias. Most of their anti-
(see Table 17.13).
arrhythmic properties can be attributed to
Flecainide is well absorbed after oral ad-
inhibition of cardiac sympathetic activity.
ministration. Approximately 40% of the drug Additional actions of some β-blockers, such as
is excreted unchanged in the urine, and the β1-cardioselectivity or a membrane-stabilizing
remainder is converted to inactive metabo- effect, seem to make no contribution to anti-
lites by the liver. Cardiac toxicities include arrhythmic activity.
the aggravation of ventricular arrhythmias Chapter 11 describes how β-adrenergic
and precipitation of CHF in patients with stimulation results in a more rapid upslope of
underlying left ventricular dysfunction. Non- phase 4 depolarization and an increased fir-
cardiac side effects are referable to the CNS ing rate of the SA node. β-Adrenergic antago-
and include confusion, dizziness, and blurred nists inhibit these effects, thus reducing auto-
vision. maticity (Fig. 17.12). This action extends to Fig. 12
The electrophysiologic properties of pro- the cardiac Purkinje fibers, where arrhythmias
pafenone are similar to those of flecainide, due to enhanced automaticity are inhibited.
but additionally it has a weak β-adrenergic In addition, because afterdepolarizations
blocking action. Propafenone is metabolized may be caused by excessive catecholamines,
by the liver, but because the level of genetic β-blockers may prevent triggered arrhythmias
variation is high, a patient’s dosage must be induced by that mechanism. All β-blockers
carefully titrated by observing the drug’s ef- increase the effective refractory period of the
fect. Extracardiac side effects are not com- AV node; therefore, these drugs are effective
mon and include dizziness and disturbances at interrupting reentrant rhythms that pass
of taste. through it.

Figure 17.12. Electrophysiologic effects of the class II antiarrhythmic drugs on the pacemaker cell action
potential.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 427

Cardiovascular Drugs 427

β-Blockers may also have a beneficial anti- AV node. In addition, β-blockers may termi-
arrhythmic effect by decreasing myocardial nate reentrant supraventricular arrhythmias
oxygen demand, thus reducing myocardial in which the AV node constitutes one limb
ischemia. Several drugs from this group have of the reentrant pathway.
been shown to reduce mortality following β-Blockers are effective in suppressing
myocardial infarction (see Chapter 7), which ventricular premature beats and other ven-
may in part relate to their antiarrhythmic tricular arrhythmias, especially when in-
effect. Since the AV nodal conduction time duced by exercise. They are also effective in
is prolonged by β-blockers, the PR interval treating ventricular arrhythmias related to
on the ECG may become prolonged (see prolongation of the QT interval because, un-
Table 17.12). The QRS and QT intervals are like group IA agents, they do not pathologi-
usually unaffected. cally prolong that interval.

Clinical Uses Class III Antiarrhythmics


β-Blockers are most useful in suppressing Class III drugs are structurally dissimilar
tachyarrhythmias induced by excessive cat- from one another but share the property of
echolamines (e.g., during exercise or emo- significantly prolonging the action poten-
tional stimulation). They are also frequently tial of Purkinje and ventricular muscle
used to slow the ventricular rate in atrial fibers (Fig. 17.13), predominantly by block- Fig. 13
flutter and fibrillation by impairing conduc- ing the outward K+ current that supports
tion and increasing the refractoriness of the phase 3 repolarization. Unlike class I agents,

Figure 17.13. Electrophysiologic effects of the class III antiarrhythmic drugs on the Purkinje cell action
potential.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 428

428 Chapter Seventeen

class III antiarrhythmics generally have little in tissues, and undergoes very slow hepatic
effect on phase 0 depolarization or conduc- metabolism. Its elimination half-life is long
tion velocity. and variable, averaging 25 to 60 days. The
Amiodarone is a powerful antiarrhythmic drug is excreted by the biliary tract, lacrimal
with many potential adverse reactions. Its glands, and skin but not by the kidney; thus,
major therapeutic effect is to prolong the ac- its dosage does not need to be adjusted in
tion potential duration and refractoriness of patients with renal failure. However, because
all cardiac fibers. However, it also shares ac- the drug’s action has a delayed onset and
tions with each of the other antiarrhythmic very long duration, amiodarone is difficult
classes. The slope of phase 0 depolarization to regulate if side effects ensue.
may be depressed through sodium channel There are numerous potential side effects
blockade (class I effect), it exerts a β-blocking of amiodarone. The most serious is pulmo-
effect (class II), and also demonstrates weak nary toxicity, manifest by pneumonitis lead-
calcium channel blockade (class IV). As a ing to pulmonary fibrosis. Its origin is unclear
result, the electrophysiologic effects of amio- but may represent a hypersensitivity reaction
darone are to decrease the sinus node fir- and, if recognized early, is reversible.
ing rate, suppress automaticity, interrupt Other life-threatening side effects of
reentrant circuits, and prolong the PR, QRS, amiodarone relate to cardiac toxicity: symp-
and QT intervals on the ECG. tomatic bradycardia and aggravation of ven-
tricular arrhythmia each occur in approx-
In addition, amiodarone is a vasodilator
imately 2% of patients. Because amiodarone
(because of α-receptor and calcium channel
significantly prolongs the QT interval, early
blocking effects) and a negative inotrope
afterdepolarizations and torsades de pointes
(β-blocker and CCB effects). The resulting
can occur, but this happens only rarely.
vasodilatation is more prominent than the
Intravenously administered amiodarone
negative inotropic effect, so that cardiac out-
occasionally precipitates heart failure.
put does not usually suffer in patients treated
Abnormalities of thyroid function are
with this drug.
common during amiodarone treatment, be-
Amiodarone is more effective than most
cause the drug contains a significant iodine
other antiarrhythmic drugs for a wide spec- load and because it inhibits the peripheral
trum of ventricular and supraventricular conversion of T4 to T3. During the first few
tachyarrhythmias. These include atrial fi- weeks of therapy, it is common to observe
brillation, atrial flutter, ventricular tachycar- transient abnormalities of thyroid biochem-
dia, ventricular flutter, and supraventricular ical tests without clinical findings of thyroid
tachycardias, including those involving by- disease: serum TSH and T4 rise, and serum T3
pass tracts. It is a first-line agent for the emer- falls. Over time, some patients develop overt
gency treatment of ventricular arrhythmias hypothyroidism (owing mostly to the anti-
during cardiac resuscitation (including ven- thyroid effects of iodine) or hyperthyroid-
tricular fibrillation and ventricular tachy- ism (because of either an iodine effect in
cardia refractory to electrical shocks), and is iodine-deficient communities or a direct thy-
more effective than lidocaine for this pur- roid inflammatory process incited by amio-
pose. It is commonly used to treat arrhyth- darone in susceptible patients).
mias in patients with ventricular systolic Gastrointestinal side effects of amiodarone
dysfunction because it causes fewer pro- include anorexia, nausea, and elevation of
arrhythmic complications in that popula- liver function tests, all of which improve
tion than other agents. In addition, low- with lower doses of the drug. Neurologic
dose amiodarone is effective for long-term side effects include proximal muscle weak-
suppression of atrial fibrillation and flutter. ness, peripheral neuropathy, ataxia, tremors,
Amiodarone is absorbed slowly from the and sleep disturbances. Commonly, corneal
gastrointestinal tract, requiring 5 to 6 hours microdeposits can be detected in patients re-
to reach peak plasma concentrations. It is ceiving chronic amiodarone therapy, but
highly lipophilic, is extensively sequestered these rarely affect vision.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 429

Cardiovascular Drugs 429

As a result of these potential adverse release can transiently aggravate arrhythmias,


effects, ECGs, thyroid and liver function but continued therapy lengthens the ac-
blood tests, chest radiographs, and some- tion potential duration and refractoriness of
times pulmonary function studies are per- atrial, ventricular, and Purkinje fibers. As a
formed on a regular basis in patients receiv- result, the threshold for ventricular fibrilla-
ing chronic therapy. Amiodarone interacts tion is substantially raised.
with and increases the activity of certain Immediately after bretylium administra-
drugs, including warfarin and digoxin, such tion, blood pressure may rise because of the
that the dosages of those agents must be catecholamine release. However, significant
adjusted. Because amiodarone prolongs the orthostatic hypotension may follow because
QT interval, other drugs that do the same of the drug’s antiadrenergic actions.
should be used with great caution. Pharma- Ibutilide is an intravenous antiarrhyth-
ceuticals that possess negative chronotropic mic agent used for the acute conversion of
AQ6 or negative inotropic effects (e.g., β-blockers, atrial fibrillation or atrial flutter of recent
verapamil, and diltiazem) should also gen- onset. This agent prolongs the action po-
erally be avoided. tential duration and increases atrial and
Sotalol is actually a nonselective β-blocker, ventricular refractoriness. The mechanism
but it is used in practice because of its addi- relates to activation of a slow inward current
tional class III antiarrhythmic properties. It that prolongs the plateau (phase 2) of the
prolongs the duration of the action poten- action potential, rather than blockade of
tial, increases the refractory period of atrial potassium currents that is typical of other
and ventricular tissue, and inhibits conduc- class III drugs. In clinical trials, the success
tion in accessory bypass tracts. The phase 0 rate for conversion of atrial flutter is ap-
upstroke velocity is not altered in the usual proximately 60% but only 30% for those in
dosage range. It is effective in the treatment atrial fibrillation.
of both supraventricular and ventricular Because ibutilide prolongs the QT inter-
arrhythmias. val, the torsades de pointes can be precipi-
Because sotalol is excreted exclusively by tated, especially in patients with underlying
the kidneys, its dosage should be adjusted in ventricular dysfunction. Therefore, careful
the presence of renal disease. Potential side electrocardiographic monitoring is necessary
effects include those of the β-blockers de- for several hours after drug administration.
scribed earlier. Because the drug prolongs Dofetilide acts by blocking the outward
the QT interval, the most serious potential potassium current, causing prolongation of
adverse effect is provoking the ventricular the action potential duration and an increase
arrhythmia torsades de pointes. This com- in the effective refractory period. It is used
plication occurs in approximately 2% of pa- orally to convert atrial fibrillation and atrial
tients and is more common in patients with flutter to sinus rhythm and to maintain sinus
a history of heart failure and in women (for rhythm after conversion. QT prolongation
unclear reasons). complicated by torsades de pointes is the
Bretylium tosylate is an intravenously major potential adverse effect. Thus, like most
administered class III drug used on occasion antiarrhythmics, drug administration should
to treat life-threatening ventricular tachy- be initiated in the hospital with electrocar-
cardia or fibrillation, when all other attempts diographic monitoring. Dofetilide is excreted
at resuscitation have failed. Its mechanism by the kidney, and its dose must be adjusted
of action is different from that of other anti- in patients with renal failure.
arrhythmic agents in that it acts at post-
ganglionic adrenergic nerve terminals, where
Class IV Antiarrhythmics
it initially releases norepinephrine but then
inhibits subsequent discharge. Thus, after Class IV drugs exert their electrophysiologic
initial stimulation, sympathetic activity of effects by selective blockade of the slow
the heart decreases. The initial catecholamine L-type cardiac calcium channels and include
10090-17_CH17.qxd 8/31/06 4:58 PM Page 430

430 Chapter Seventeen

verapamil and diltiazem, but not nifedipine most important side effect of verapamil and
or the other dihydropyridine CCBs. They are diltiazem, when administered intravenously,
most potent in tissues in which the action po- is hypotension. In addition, these agents
tential depends on calcium currents, such should be avoided in patients receiving β-
as the SA and AV nodes. Within nodal tis- blocker therapy, because the combined neg-
sue, calcium channel blockade elevates the ative inotropic and chronotropic effects may
threshold potential, decreases the rate of rise precipitate heart failure.
of phase 0 depolarization and conduction ve-
locity, and lengthens the refractory period of
Fig. 14 the AV node (Fig. 17.14). These electrophys-
Adenosine
iologic actions translate into their clinical ef- Adenosine is an endogenous nucleoside with
fects: (1) the heart rate slows; (2) transmis- a very short half-life. Administered intra-
sion of rapid atrial impulses through the AV venously, it is the most effective drug for the
node to the ventricles decreases, thus slowing rapid termination of reentrant PSVT.
the ventricular rate in atrial fibrillation and Adenosine has substantial electrophysio-
atrial flutter; and (3) reentrant rhythms trav- logic effects on specialized conduction tis-
eling through the AV node may terminate. sues, especially the SA and AV nodes. By
A primary antiarrhythmic use of class IV binding to specific adenosine receptors, it
drugs is in the treatment of reentrant PSVT. activates potassium channels (Fig. 17.15). Fig. 15
At one time, intravenous verapamil was the The resultant increase in the outward potas-
treatment of choice for acute episodes of sium current hyperpolarizes the membrane,
this rhythm, but intravenous adenosine (de- which suppresses spontaneous depolariza-
scribed in the next section) has assumed tion of the SA node and slows conduction
that role. The class IV antiarrhythmics are through the AV node.
also often used to slow the ventricular rate In addition, adenosine decreases the in-
in patients with atrial fibrillation or flutter. tracellular cAMP concentration by inhibit-
The pharmacology and toxicities of CCBs ing adenylate cyclase. The result is a decrease
were presented earlier in this chapter. The in the inward pacemaker current (If) and

Figure 17.14. Electrophysiologic effects of the class IV antiarrhythmic drugs on the pacemaker cell action
potential.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 431

Cardiovascular Drugs 431

ADENOSINE

Figure 17.15. Mechanism of antiarrhythmic action of adenosine. Stimulation of the


myocyte adenosine receptor activates potassium channels, and the resultant outward K+ cur-
rent hyperpolarizes the membrane (resulting in decreased automaticity). Adenosine also
inhibits membrane adenylate cyclase activity; the subsequent reduction in active protein
kinases decreases the inward pacemaker (If) and Ca++ currents (resulting in decreased auto-
maticity and decreased conduction through the AV node). Xanthines compete for the adeno-
sine receptor, blocking these effects. Dipyridamole interferes with cellular uptake and degra-
dation of adenosine and therefore amplifies its effect.

the inward calcium current (see Fig. 17.15). ists with most agents. Whenever antiarrhyth-
Thus, the net effect of adenosine is to slow mic drugs are used, patients must be followed
the SA node firing rate and to decrease AV closely, the effectiveness of the drug demon-
nodal conduction. By inducing transient AV strated, and surveillance for toxicity contin-
nodal block, adenosine terminates reentrant ued over the long term.
pathways that include the AV node as part
of the circuit. Ventricular myocytes are rel-
DIURETICS
atively immune to these effects, in part be-
cause the specific potassium channels re- Diuretics are most often used to treat heart
sponsive to adenosine are not present in failure and hypertension. In heart failure,
those cells. enhanced renal reabsorption of sodium and
With a half-life of only 10 sec, adenosine water, with subsequent expansion of the ex-
has very transient side effects (headache, tracellular volume, contributes to peripheral
chest pain, flushing, bronchoconstriction). edema and pulmonary congestion. Diuretics
Because methylxanthines (caffeine, theo- eliminate excess sodium and water through
phylline) competitively antagonize the ade- renal excretion and are therefore the cor-
nosine receptor, higher doses of adenosine nerstone of therapy to relieve congestive
may be necessary in patients using those symptoms (see Chapter 9). In the treatment
substances. Conversely, dipyridamole inhibits of hypertension, diuretics eliminate intra-
the breakdown of adenosine and amplifies its vascular volume and in some cases promote
effect. vascular dilatation.
In the kidney, the rate of glomerular filtra-
In summary, antiarrhythmic drugs have tion typically averages 135 to 180 L/day in
complex actions and display multiple car- normal adults. Most of the filtered Na+ is re-
diac and noncardiac toxicities. The poten- absorbed by the renal tubules, leaving only a
tial of inducing dangerous arrhythmias ex- small quantity in the final urine (Fig. 17.16). Fig. 16
10090-17_CH17.qxd 8/31/06 4:58 PM Page 432

432 Chapter Seventeen

Carbonic
anhydrase
inhibitors
K+
sparing
diuretics

Loop Thiazides
diuretics

Figure 17.16. Schematic diagram of the renal tubules. Approximately 70% of filtered sodium is reabsorbed in the
proximal convoluted tubule, 25% in the thick ascending limb of the loop of Henle, 5% in the distal convoluted tubule,
and 1% to 2% in the cortical collecting tubule (mediated by the action of aldosterone). Antidiuretic hormone (ADH)
increases the permeability of the distal nephron for water. Diuretics are secreted into the proximal convoluted tubule
and act at the sites shown.

Approximately 65% to 70% of the filtered Na+ moted by an antidiuretic hormone and
is reabsorbed isosmotically in the proximal driven by the osmotic gradient between the
tubule by active transport. In the thick as- tubule and the hypertonic interstitium. Sub-
cending limb of the loop of Henle, an ad- stances that interfere with the antidiuretic
ditional 25% of the filtered sodium is re- hormone, such as ethanol consumption,
absorbed, through a Na+-K+ cotransport therefore have diuretic actions.
system coupled to the uptake of two Cl− ions. The three most commonly used groups
Because this region is impermeable to the re- of diuretics are the loop diuretics, thiazide
absorption of water, it is the site of hypotonic diuretics, and potassium-sparing diuretics
tubular fluid formation, and the surrounding (Table 17.14 and Fig. 17.16). These classes Tab. 14
interstitium becomes hypertonic. In the dis- are generally distinguished by the site of
tal convoluted tubule, an additional small the kidney tubule where they act and by
fraction of NaCl is reabsorbed (approximately their potency. Loop diuretics impair ab-
5%). In the cortical collecting duct, Na+ per- sorption in the thick ascending limb of
meability is modulated by an aldosterone- the loop of Henle, thiazide diuretics act on
sensitive mechanism, such that Na+ is re- the distal tubule and collecting segment,
absorbed into the tubular cells in the presence and potassium-sparing diuretics act on the
of aldosterone, creating a lumen-negative po- aldosterone-sensitive region of the cortical
tential difference that enhances K+ and H+ ex- collecting tubule. A fourth group, the car-
cretion. Approximately 1% to 2% of sodium bonic anhydrase inhibitors, are weak diuret-
reabsorption takes place at this location. ics rarely used in the treatment of hyperten-
Most of the distal tubule is impermeable sion or heart failure. They act at the proximal
to water. In the collecting tubule, however, convoluted tubule, resulting in a loss of bi-
water permeability and reabsorption are pro- carbonate (and sodium) in the urine.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 433

Cardiovascular Drugs 433

TABLE 17.14. Commonly Used Diuretics

Method of Onset of Duration of


Diuretic Administration Action (hours) Action (hours) Potential Adverse Effects

Thiazides Hypokalemia, hypomagne-


Chlorothiazide PO 1 6–12 semia, hyponatremia,
IV 0.25 2 hypercalcemia, hyper-
Hydrochlorothiazide PO 2 12 glycemia, hyperuricemia,
Chlorthalidone PO 2 24 hypercholesterolemia,
Metolazone PO 1 12–24 hypertriglyceridemia, meta-
Indapamide PO 1–2 16–36 bolic alkalosis
Loop diuretics Hypotension, hypokalemia,
Furosemide PO 1 6 hypomagnesemia, hyper-
IV 5 min 2 glycemia, hyperuricemia,
Bumetanide PO 0.5–1 4–6 metabolic alkalosis
IV 0.25 0.5–1
Torsemide PO <1 6–8
IV 10 min 6–8
Ethacrynic acid IV 0.25 3
K1-sparing diuretics
Spironolactone PO >24 2–3 days Hyperkalemia, GI distur-
Eplerenone PO >24 24 bances; gynecomastia
Triamterene PO 2 12–16 (spironolactone only)
Amiloride PO 2 24

GI, gastrointestinal; IV, intravenous; PO, by mouth.

Diuretics, as active pharmacologic agents, dilatation, which is also beneficial in reduc- AQ7
are secreted into the proximal renal tubule, ing venous return and pulmonary conges-
and their major sites of action are shown tion (see Chapter 9). The mechanism of ve-
schematically in Figure 17.16. nous vasodilatation appears to involve drug-
induced prostaglandin and nitric oxide gen-
eration from endothelial cells, which act to
Loop Diuretics
relax vascular smooth muscle.
These agents are so named because they act The most common side effects of the loop
principally on the thick ascending limb of diuretics are intravascular volume depletion,
the loop of Henle. They are powerful diuret- hypokalemia, and metabolic alkalosis. Hypo-
ics that result in the excretion of 20% to 25% kalemia arises because (1) these agents impair
of the filtered Na+ load through inhibition of the reabsorption of sodium in the loop of
the Na+-2Cl−-K+ cotransport system. Because Henle, such that an increased amount of Na+
inhibition at this site impairs the generation is delivered to the distal tubule, where it
of a hypertonic interstitium, the gradient for prompts greater-than-normal exchange for
passive water movement out of the collecting potassium (and therefore more K+ excretion
duct is diminished and water diuresis results. into the urine); and (2) diuretic-induced in-
Loop diuretics are of great importance in travascular volume depletion activates the
the acute management of pulmonary edema renin-angiotensin system. The subsequent
(administered intravenously) and in the rise in aldosterone promotes additional Na+-
treatment of chronic heart failure or periph- K+ exchange and hence potassium loss into
eral edema (taken orally). Unlike other di- the urine.
uretics, they tend to be effective in the set- Metabolic alkalosis during loop diuretic
ting of impaired renal function. In addition therapy results from two mechanisms: (1) in-
to the diuretic effect, and even preceding it, creased H+ secretion into the distal tubule
drugs of this class may induce venous vaso- (and therefore into the urine) owing to the
10090-17_CH17.qxd 8/31/06 4:58 PM Page 434

434 Chapter Seventeen

secondary hyperaldosteronism described ear- This class of drugs acts at the distal tubule,
lier, and (2) contraction alkalosis, in which where they block the reabsorption of ap-
reduced intravascular volume promotes in- proximately 3% to 5% of the filtered sodium
creased sodium bicarbonate reabsorption by (see Fig. 17.16). Na+ reabsorption at this site
the proximal tubule (see Fig. 17.16). is mediated through a Na+-Cl− cotransporter
Additional side effects may also occur dur- on the luminal membrane. The thiazides in-
ing continued loop diuretic therapy. Hypo- hibit this carrier by a mechanism that has
magnesemia may result, because magnesium not been elucidated but may involve com-
reabsorption depends on NaCl transport in petition for the Cl− site. The antihyperten-
the thick ascending limb of the loop of sive effect is initially associated with a de-
Henle, the action blocked by these drugs. crease in cardiac output owing to reduced
Ototoxicity (cranial nerve VIII toxicity) occa- intravascular volume and with unchanged
sionally develops, impairing hearing and peripheral resistance. With long-term thi-
vestibular function. It is thought to arise azide use, however, cardiac output often re-
from electrolyte disturbances of the endo- turns to normal as total peripheral resistance
lymphatic system, most likely because of becomes reduced by vascular dilatation.
Na+-2Cl−-K+ cotransport inhibition by the Indapamide is unique among this class in
diuretic at that site. that it displays a particularly prominent vaso-
The most commonly used loop diuretic is dilating effect.
furosemide, the oral form of which demon- Thiazides are typically administered orally.
strates reliable gastrointestinal absorption Diuresis occurs after 1 to 2 hours, but the
but a short duration of action (4 to 6 hours) full antihypertensive effect of continued
that limits its usefulness in the chronic treat- therapy may not become manifest for up to
ment of hypertension. Bumetanide is simi- 12 weeks (possibly related to the vasodilator
lar to furosemide and shares its actions and mechanism alluded to in the previous para-
adverse effects but has greater potency and graph). Chlorothiazide, the parent com-
bioavailability. It also appears to have a lower pound, has a low lipid solubility and hence
incidence of ototoxicity than the other drugs low bioavailability: higher doses are there-
of this class. Bumetanide is sometimes use- fore required to achieve therapeutic levels
ful in heart failure patients when edema is compared with the more commonly used
refractory to other agents and in some pa- hydrochlorothiazide. Chlorthalidone is
tients allergic to furosemide. Torsemide is slowly absorbed and hence has a long dura-
also similar to furosemide, with more com- tion of action. Metolazone, unlike other
plete bioavailability. Ethacrynic acid is the drugs of this class, is sometimes effective in
only nonsulfonamide loop diuretic, so it can patients with reduced renal function.
be prescribed to patients who are intolerant Clinically, the thiazides differ from the
of sulfonamide compounds. It is otherwise loop diuretics in that they are less potent,
not widely used because of its high incidence have a longer duration of action, and (with
of ototoxicity. the exception of metolazone) demonstrate
poor diuretic efficacy in the setting of im-
paired renal function: generally, they are not
Thiazide Diuretics
effective when the GFR is <25 mL/min.
Thiazides and related compounds (chlortha- Thiazides serve as the cornerstone of anti-
lidone, indapamide, and metolazone) are hypertensive therapy because of their low
commonly used diuretics because they de- cost, effectiveness, and proven benefits in re-
monstrate excellent gastrointestinal absorp- ducing the risk of stroke and cardiac events.
tion when administered orally and are usu- They are sometimes used in heart failure,
ally well tolerated. They are less potent than generally for patients with mild chronic
the loop diuretics but, because of their sus- congestive symptoms. In addition, they can
tained actions, are useful in chronic condi- be combined with a loop diuretic for pa-
tions such as hypertension and mild CHF. tients who have become refractory to the di-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 435

Cardiovascular Drugs 435

uretic effect of the latter. The combination of none); and (2) direct inhibitors of Na+ per-
the two classes lowers the dose-dependent ad- meability in the collecting duct, which act in-
verse effects that accompany each drug; be- dependently of aldosterone (e.g., triamterene
cause they act on sequential segments of the and amiloride).
renal tubule, a more profound natriuretic ef- Na+ and K+ exchange in the collecting
fect ensues than with either agent used alone. tubules accounts for only a small percentage
Among the most important potential ad- of sodium reuptake, preventing potent di-
verse effects of thiazides are (1) hypokalemia uresis from occurring when these agents are
and metabolic alkalosis, which result in part used alone. Therefore, they are often used
from increased Na+ delivery to the distal in combination with the loop or thiazide
tubule, where exchange for K+ and H+ takes classes for additive diuretic effect and to pre-
place, and partly from volume contraction vent clinically important hypokalemia.
and secondary hyperaldosteronism, as pre- Spironolactone is a synthetic steroid that
viously described for the loop diuretics; competes for the cytoplasmic aldosterone re-
(2) hyponatremia, during prolonged treat- ceptor, thereby inhibiting the aldosterone-
ment because of continued Na+ excretion in sensitive Na+ channel in the kidney. Because
the setting of chronic free water consump- Na+ reabsorption through the sodium chan-
tion; (3) hyperuricemia (and possible precipi- nel is inhibited, no lumen-negative poten-
tation of gout) owing to decreased clearance tial exists to drive K+ and H+ ion excretion at
of uric acid; (4) hyperglycemia, because of the distal nephron sites; thus, K+ and H+ ions
either impaired pancreatic insulin release are retained in the circulation. Spironolac-
or decreased peripheral glucose utilization; tone also displays beneficial cardiac anti-
(5) alterations in serum lipids (at least tran- remodeling effects in patients with heart fail-
siently), characterized by increased low- ure (see Chapter 9). In a trial of patients with
density lipoprotein (LDL) cholesterol and severe heart failure, spironolactone (added
triglycerides; and (6) weakness, fatigability, to an ACE inhibitor and a loop diuretic, with
and paresthesias, which can occur with long- or without digoxin) improved heart failure
term use because of volume depletion and symptoms and reduced mortality rates.
hypokalemia. In addition, serum calcium The most serious potential complication
levels often rise slightly during thiazide ther- of spironolactone is hyperkalemia, resulting
apy, but this is rarely clinically significant. from the impaired excretion of that ion.
In past decades, the standard thiazide Thus, caution should be observed when ad-
dosage was excessive compared with current ministering K+ supplements, ACE inhibitors,
practice. By using lower dosages, it is possi- or angiotensin receptor blockers concurrent
ble to accrue the benefits of this class of di- with potassium-sparing diuretics because
uretics while minimizing adverse effects. they could contribute to this complication.
Spironolactone also displays antiandrogenic
activity that may produce gynecomastia in
Potassium-Sparing Diuretics
men and menstrual irregularities in women.
These agents are relatively weak diuretics Eplerenone is a more specific inhibitor
that antagonize physiologic Na+ reabsorption of the aldosterone receptor that does not
at the distal convoluted tubule and cortical have the systemic anti-androgenic effects
collecting tubule. Potassium-sparing diuret- (i.e., gynecomastia) of spironolactone. Like
ics reduce K+ excretion; thus, unlike other spironolactone, it is used in the treatment
diuretics, hypokalemia is not a side effect. of hypertension and chronic systolic heart
They are used when maintenance of serum failure. In patients with clinical evidence of
potassium levels is crucial and in states char- heart failure following a myocardial infarc-
acterized by aldosterone excess (e.g., primary tion, eplerenone has also been shown to
or secondary hyperaldosteronism). Two types improve survival when added to usual care.
of drugs make up this group: (1) aldosterone Triamterene and amiloride are struc-
antagonists (e.g., spironolactone, and eplere- turally related potassium-sparing diuretics
10090-17_CH17.qxd 8/31/06 4:58 PM Page 436

436 Chapter Seventeen

that act independently of aldosterone. At the tion of platelet products and activation of
distal tubules, they inhibit the reabsorption key surface receptors), and (3) aggregation.
of Na+, which subsequently diminishes the For example, following blood vessel injury,
excretion of K+ and H+. Triamterene is me- platelets quickly adhere to exposed sub-
tabolized by the liver, and its active prod- endothelial collagen by means of membrane
uct is secreted into the proximal tubule by glycoprotein receptors, a process that de-
the organic cation transport system. Amilo- pends on von Willebrand factor. Following
ride is secreted directly into the proximal adhesion to the vessel wall, platelets release
tubule and appears unchanged in the urine. the preformed contents of their granules in
As with spironolactone, the most impor- response to agonists (including collagen and
tant potential adverse effect of these drugs thrombin) that bind to platelet receptors.
is the development of hyperkalemia. Among these repackaged substances are ade-
nosine diphosphate (ADP), serotonin, fib-
rinogen, growth factors, and procoagulants.
ANTITHROMBOTIC DRUGS
Concurrently within the activated platelet,
Platelets and the coagulation proteins play a there is de novo synthesis and secretion of
key role in the pathogenesis of many cardio- thromboxane A2 (TXA2), a powerful vaso-
vascular disorders, including the acute coro- constrictor (Fig. 17.17). Fig. 17
nary syndromes, deep venous thrombosis, Certain agonists, including ADP, throm-
and thrombi that may complicate atrial fib- bin, and TXA2, stimulate platelets to aggre-
rillation, dilated cardiomyopathy, or mecha- gate and form the primary hemostatic plug,
nical prosthetic heart valves. Therefore, the as additional platelets are recruited from the
modulation of platelet function and of the circulation. During this process, the platelet
coagulation pathway is often critically im- membrane glycoprotein (GP) IIb/IIIa re-
portant in cardiovascular therapeutics. ceptors undergo a critical conformational
The formation of a thrombus, whether in change. This alteration allows the previ-
normal hemostasis or in pathologic clot for- ously inactive GP IIb/IIIa receptor to bind AQ8
mation, requires three events: (1) exposure of fibrinogen molecules, an action that tightly
circulating blood elements to thrombogenic links platelets to one another and consti-
material (e.g., unmasking of subendothelial tutes the final common pathway of platelet
collagen after atherosclerotic plaque rupture); aggregation. The developing clump of plate-
(2) activation of platelets; and (3) triggering lets is stabilized and tethered to the site of
of the coagulation cascade, ultimately re- injury by a developing mesh of fibrin, which
sulting in a fibrin clot. Hemostasis effected is produced by the simultaneous activation
by platelets and the coagulation system are of the coagulation protein cascade.
closely interlinked: activated platelets accel- Platelet activation is regulated to a great
erate the coagulation pathway, and certain extent by release of stored Ca++ from the
coagulation proteins (e.g., thrombin) contri- platelet-dense tubular system. This action re-
bute to platelet aggregation. sults in an increase in the cytosolic calcium
This section focuses first on drugs that concentration, with activation of protein ki-
interfere with platelet function and then on nases and phosphorylation of intraplatelet
those that inhibit the coagulation cascade. regulatory proteins. The increase in cytosolic
Fibrinolytic agents, which dissolve clots Ca++ also stimulates phospholipase A2 (PLA2),
that have already formed, are described in causing the release of arachidonic acid, the
Chapter 7. precursor of TXA2 (see Fig. 17.17).
The critical release of calcium is modu-
lated by several factors. Acting at their
Platelet Inhibitors
respective platelet membrane receptors,
Platelets are responsible for primary hemo- thrombin and other agonists generate inter-
stasis by a three-part process: (1) adhesion to mediaries that stimulate the release of cal-
the site of injury, (2) release reaction (secre- cium from the dense tubules. TXA2 increases
10090-17_CH17.qxd 8/31/06 4:58 PM Page 437

Cardiovascular Drugs 437

THIENOPYRIDINES

Prostacyclin TXA2 ADP Serotonin Thrombin


P2Y12 P2Y1
Gs AC Gi Gi PIP2
PLA2 + – PLC

ATP
cAMP IP3
Arachidonic – +
acid +
DIPYRIDAMOLE
Cyclooxygenase
Ca++ Ca++

TXA2 DENSE
TUBULES
ASPIRIN
PLATELET
ACTIVATION
GP IIb/IIIa
RECEPTOR
Granule Activation of ANTAGONISTS
secretion glycoprotein (prevent fibrinogen
Cytoskeletal binding)
IIb/IIIa –
reorganization
receptor

Figure 17.17. Platelet activation is mediated by cytosolic Ca11. Factors that promote and inhibit calcium re-
lease from the platelet dense tubules are shown. Thrombin and serotonin, acting at their specific receptors, stimu-
late the formation of inositol triphosphate (IP3) from phosphatidylinositol diphosphate (PIP2) by phospholipase C
(PLC). IP3 subsequently enhances the intracellular release of calcium. Thromboxane A2 (TXA2) also facilitates calcium
release. It inhibits adenyl cyclase (AC) and reduces cyclic adenosine monophosphate (cAMP) formation. Because
cAMP normally prevents Ca++ release from the dense tubules, the reduction of this effect by TXA2 increases Ca++ re-
lease into the cytosol. Conversely, endothelial-derived prostacyclin has the opposite effect. It reduces intraplatelet
calcium release because it stimulates AC activity and cAMP formation. ADP also stimulates calcium release via its
two receptors (see text for details). Calcium promotes the action of phospholipase A2 (PLA2), which generates the
precursors of TXA2 from the cell membrane. Platelet activation modulated by [Ca++] ultimately results in granule se-
cretion, cytoskeletal reorganization, and the critical conformational change in glycoprotein IIb/IIIa receptors that is
necessary for platelet aggregation. The sites of action of commonly used antiplatelet drugs are shown in color. P2Y1
and P2Y12, purinoceptors.

the intracellular Ca++ by binding to its nopyridines and GP IIb/IIIa inhibitors, have
platelet receptor, which inhibits the activity rapidly expanded.
of adenylate cyclase and thereby reduces
cAMP formation, an action that augments Aspirin
the release of Ca++ from the dense tubules
(see Fig. 17.17). Conversely, endothelial cell- As described in the previous section, TXA2
derived prostacyclin (PGI2) stimulates adeny- is an important mediator of platelet activa-
late cyclase activity and increases platelet tion and clot formation. Aspirin (acetylsali-
cAMP concentration, which inhibits Ca++ re- cylic acid) acts by irreversibly acetylating
lease from the dense tubular system. (and thus blocking the action of) cyclooxy-
Antiplatelet drugs interfere with platelet genase, an enzyme essential to TXA2 produc-
function at various points along the se- tion from arachidonic acid (see Fig. 17.17).
quence of activation and aggregation (see The form of the enzyme found in platelets is
Fig. 17.17). The most commonly used anti- cyclooxygenase 1 (COX-1), which is effec-
platelet drug is aspirin, but the roles of tively inhibited by the nonselective action
newer antiplatelet drugs, especially the thie- of aspirin (but it is not inhibited by selective
10090-17_CH17.qxd 8/31/06 4:58 PM Page 438

438 Chapter Seventeen

COX-2 antagonists, such as celecoxib). Be- Clinical Uses


cause platelets lack nuclei and therefore
Aspirin therapy has many proven clinical
cannot synthesize new proteins (including
benefits in patients with cardiovascular dis-
cyclooxygenase), aspirin permanently disables Tab. 15
ease (Table 17.15). In individuals with un-
TXA2 production in exposed cells. stable angina, acute myocardial infarction,
The prostaglandin PGI2, a major antago- or a history of myocardial infarction, aspirin
nist of TXA2 that is produced by endothelial conclusively reduces the incidence of future
cells, shares a dependency on cyclooxyge- fatal and nonfatal coronary events. Simi-
nase activity for its formation, and aspirin, larly, in patients with chronic stable angina
at high doses, impairs its synthesis as well. without a history of myocardial infarction,
Unlike platelets, however, endothelial cells aspirin lessens the occurrence of subsequent
are able to generate new cyclooxygenase to myocardial infarction and mortality. In pa-
replace what has been deactivated by acety- tients who have suffered a minor stroke or
lation. Thus, when used at low doses, aspirin transient cerebral ischemic attacks, aspirin
effectively inhibits platelet TXA2 synthesis reduces the rate of future stroke and cardio-
without significantly interfering with the vascular events. Additionally, aspirin lowers
presence and beneficial actions of PGI2. the likelihood of graft occlusion in patients
Because the antiplatelet effect of aspirin who have undergone coronary artery bypass
is limited to inhibition of TXA2 formation, surgery.
platelet aggregation induced by other factors Less clear is the benefit of aspirin for pri-
(e.g., ADP) is not significantly impeded. Thus, mary prevention (i.e., in individuals without
aspirin is not a “complete” antithrombotic a history of cardiovascular events or symp-
agent. toms). When tested in a large cohort of

TABLE 17.15. Cardiovascular Uses of Antithrombotic Drugs

Unstable Mechanical
Chronic Angina/ Heart Atrial
Drug Angina NSTEMI STEMI DVT Valve Fibrillation PCI HIT

Platelet inhibitors
Aspirin + + + (1) (2) +
Thienopyridines + + (3)
GP IIb/IIIa inhibitors + (4) +
Dipyridamole (5)
Anticoagulants
UFH + + + (6) (6) +
LMWH + + +
Direct thrombin inhibitors (7) + +
Fondaparinux +
Warfarin (8) + + +

(1) Sometimes used in combination with warfarin.


(2) If patient has a low risk of stroke, or if warfarin is contraindicated.
(3) When intracoronary stent is implanted.
(4) If PCI undertaken.
(5) Sometimes used in combination with warfarin for recurrent embolism.
(6) For hospitalized patients unable to take warfarin.
(7) Emerging use.
(8) For 3–6 months after MI if large akinetic segment is present.
DVT, deep venous thrombosis; HIT, heparin-induced thrombocytopenia; LMWH, low-molecular-weight heparin; NSTEMI,
non–ST-elevation myocardial infarction (MI); PCI, percutaneous coronary intervention; STEMI, ST-elevation MI; UFH, un-
fractionated heparin.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 439

Cardiovascular Drugs 439

healthy American middle-aged men, aspirin can be ameliorated by lowering the dosage
was associated with a reduced incidence of and/or using enteric-coated or buffered
nonfatal myocardial infarction but an in- tablets. More serious potential side effects
creased rate of nonfatal hemorrhagic stroke include gastrointestinal bleeding, hemor-
and gastrointestinal bleeding; there was no rhagic strokes, allergic reactions, and asthma
effect on total vascular mortality. Subsequent exacerbation in aspirin-sensitive patients.
meta-analyses of clinical trials have simi- Because aspirin is excreted by the kidneys
larly concluded that aspirin is effective for and competes with uric acid for the renal
primary prevention of myocardial infarction proximal tubule organic anion transporter,
in patients with coronary risk factors, but it it may also occasionally exacerbate gout.
also increases the risk of hemorrhagic stroke.
In the prospective, primary prevention trial
Thienopyridines
known as the Women’s Health Initiative, as-
pirin lowered the risk of ischemic stroke in The thienopyridines inhibit ADP-mediated
women but did not reduce the incidence of activation of platelets (see Fig. 17.17). Nor-
MI or death from cardiovascular disease. mally, extracellular ADP activates platelets
Thus, whereas aspirin plays an extremely by binding to two types of surface purino-
important role in patients with known car- ceptors. The first (termed P2Y1) acts through
diovascular disease, it is not evident that phospholipase C to increase intraplatelet
otherwise healthy people should routinely [Ca++], which potentiates platelet activation.
take aspirin for “cardiovascular protection.” The second purinoceptor (P2Y12) is linked to
Current recommendations are that as- an inhibitory G protein and reduces cAMP
pirin (at a dosage of 75 to 325 mg/day) be production when activated, thus also raising
administered to patients with clinical man- intraplatelet [Ca++] (see Fig. 17.17). ADP-in-
ifestations of coronary disease in the ab- duced platelet aggregation requires that ADP
sence of contraindications (i.e., aspirin al- simultaneously activate both P2Y1 and P2Y12
lergy or complications described in the next purinoceptors. The thienopyridines, after
section). It should not be prescribed rou- conversion to active metabolites in the liver,
tinely for primary prevention purposes in irreversibly block the P2Y12 purinoceptor by
completely healthy individuals. However, binding directly to it or to a nearby mem-
pending the results of ongoing research, brane protein. As a result, platelet aggrega-
many physicians believe it is appropriate to tion is inhibited.
recommend aspirin use in men and women Clopidogrel and ticlopidine and are the
older than age 50 who have at least one thienopyridines approved for clinical use.
major atherosclerosis risk factor (see Chap- Both are well absorbed orally and have good
ter 5). In addition, the American Diabetes bioavailability. Meta-analyses of the use of
Association recommends that all diabetics ticlopidine or clopidogrel in patients prone
with at least one other coronary risk factor to coronary syndromes have concluded that
take aspirin for cardiovascular protection. these drugs are modestly superior to aspirin
Finally, aspirin is not as beneficial as war- in reducing the risk of myocardial infarction,
farin (described later in the chapter) for the stroke, or vascular deaths, but at an increased
prevention of stroke in high-risk patients risk of side effects and at a higher economic
with atrial fibrillation and should only be cost. Studies evaluating the combination
used in that setting when warfarin cannot of aspirin plus clopidogrel in patients with
be safely administered. unstable angina, non–ST-elevation and ST-
elevation myocardial infarction have de-
monstrated a benefit in cardiovascular out-
AQ9 Toxicity
comes compared with aspirin alone, but at
The most common adverse effects of aspirin an increased bleeding risk.
are related to the gastrointestinal system, in- Important uses of the thienopyridines
cluding dyspepsia and nausea, which often (especially clopidogrel) are as an antiplatelet
10090-17_CH17.qxd 8/31/06 4:58 PM Page 440

440 Chapter Seventeen

substitute in patients allergic to aspirin and been developed but have not demonstrated
to prevent thrombotic complications fol- beneficial outcomes in clinical trials.
lowing percutaneous coronary stenting (see The major side effects of the GP IIb/IIIa
Chapter 6). The combination of clopidogrel receptor inhibitors are bleeding (in 1% to
plus aspirin is also approved for use in pa- 10% of patients) and thrombocytopenia (in
tients with unstable angina or non–ST- approximately 2% of patients treated with
elevation myocardial infarction to reduce abciximab and less commonly with the other
the rate of recurrent cardiac events. A recent agents). Abciximab has a short plasma half-
study showed that clopidogrel is also bene- life (30 min); thus, its effects can be reversed
ficial for this purpose in patients with ST- by discontinuing the drug or by administer-
elevation myocardial infarction treated with ing a platelet transfusion. Because the other
fibrinolytic therapy (see Chapter 7). GP IIb/IIIa receptor antagonists have longer
Side effects of the thienopyridines include half-lives, they may inactivate transfused
dyspepsia and diarrhea. In addition, the use platelets. However, bleeding complications
of ticlopidine has been limited by potentially are infrequent using current protocols and
life-threatening adverse reactions: severe careful dosing.
neutropenia (occurring in 0.8% to 2.5%
of patients) and thrombotic thrombocyto-
Dipyridamole
penic purpura (in approximately 0.02% of
treated patients). These hematologic effects The antiplatelet drug dipyridamole is occa-
are much rarer with clopidogrel, which is sionally prescribed to patients who are in-
therefore the preferred agent of this class. tolerant to aspirin, but it is not as effective.
Its mechanism of antiplatelet action is un-
clear, but it may act, in part, by increasing
Glycoprotein IIb/IIIa
platelet cAMP levels, either by inhibiting
Receptor Antagonists
the enzyme phosphodiesterase or by block-
The GP IIb/IIIa receptor antagonists consti- ing cellular uptake and destruction of endo-
tute one of the most potent classes of anti- genous adenosine. As a result, cytosolic
platelet agents. This group reversibly inhibits [Ca++] falls, which inhibits platelet aggrega-
the critical and final common pathway of tion (see Fig. 17.17). By itself, dipyridamole
platelet aggregation—the binding of acti- has no proven benefits. It is used sometimes
vated platelet GP IIb/IIIa receptors to fib- in combination with warfarin for an aug-
rinogen and von Willebrand factor. As a re- mented antithrombotic effect in patients
sult, platelets are inhibited from “sticking” with recurrent thromboembolism from pros-
to one another, impairing the formation of thetic heart valves, but the combination of
a hemostatic plug. Three types of GP IIb/IIIa aspirin plus warfarin is more effective. Its
receptor antagonists have been developed: most common current use is actually as an
(1) monoclonal antibodies (e.g., abcix- agent in pharmacologic stress testing (see
imab, which is the Fab fragment of a Chapters 3 and 6). AQ10
chimeric human-mouse monoclonal anti-
body); (2) synthetic peptide antagonists
Anticoagulant Drugs
(e.g., eptifibatide); and (3) synthetic non-
peptide antagonists (e.g., tirofiban). As de- Anticoagulant drugs (see Table 17.15) inter-
scribed in Chapters 6 and 7, the GP IIb/IIIa fere with the coagulation cascade and im-
antagonists represent a major advance in pair secondary hemostasis. Because the final
improving outcomes of patients under- step in both the intrinsic and extrinsic co-
going percutaneous coronary interventions agulation pathways is the formation of a fi-
and in high-risk acute coronary syndromes. brin clot by the action of thrombin, major
All the GP IIb/IIIa receptor inhibitors in goals of anticoagulant therapy are to inhibit
current use must be given intravenously. thrombin activation from prothrombin by
Oral GP IIb/IIIa receptor inhibitors have factor Xa (e.g., using unfractionated hepa-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 441

Cardiovascular Drugs 441

rin, low molecular weight forms of heparin, fixed low dosages of subcutaneous UFH are
or fondaparinux); to inhibit thrombin it- often administered to prevent deep venous
self (e.g., with unfractionated heparin or di- thrombosis.
rect thrombin inhibitors); or to decrease The most important side effect of heparin
the production of functional prothrombin is bleeding. An overdose of UFH can be treated
(e.g., using warfarin). with intravenous protamine sulfate, which
forms a stable complex with UFH and im-
mediately reverses the anticoagulation effect.
Unfractionated Heparin
Heparin-induced thrombocytopenia (HIT) is
Unfractionated heparin (UFH) is a hetero- another potential major adverse effect and
geneous mixture of highly charged muco- can occur in two forms. The more common
polysaccharide polymers. Although it has lit- type, thought to result from direct heparin-
tle anticoagulant action by itself, it associates induced platelet aggregation, occurs in up to
with antithrombin III (AT III) in the circula- 15% of patients and is usually asympto-
tion, greatly increasing its effect. AT III (also matic, dose dependent, and self-limited. This
termed simply antithrombin) is a natural mild HIT rarely causes severe reductions in
protein that inhibits the action of thrombin platelet counts and usually does not require
and other clotting factors. When UFH com- cessation of heparin.
plexes with AT III, the affinity of AT III for The less common, much more dangerous
thrombin increases 1,000-fold, markedly re- form of HIT is immune mediated, a condi-
ducing thrombin’s ability to generate fibrin tion that affects 3% of UFH-treated patients.
from fibrinogen. The UFH–AT III complex It can lead to life-threatening bleeding and,
also inhibits activated factor X, additionally paradoxically, to thrombosis. Thrombosis
contributing to the anticoagulant action. is caused by the formation of antibodies di-
Furthermore, UFH has antiplatelet properties rected against heparin-platelet complexes,
by binding to, and blocking the action of, resulting in platelet activation, aggrega-
von Willebrand factor. tion, and clot production. In the immune-
UFH is administered parenterally because mediated form of HIT, the platelet count can
it is not absorbed from the gastrointestinal fall markedly and is not dependent on the
tract. For most acute indications, an intra- dose of heparin. Therapy requires immediate
venous bolus is followed by a continuous cessation of heparin and substitution by
infusion of the drug. The bioavailability of alternate antithrombotic therapy to prevent
UFH varies from patient to patient because further clotting (e.g., a direct thrombin in-
it is a heterogeneous collection of molecules hibitor, described later in the chapter).
that bind to plasma proteins, macrophages, Patients receiving long-term UFH therapy
and endothelial cells. The dosage–effect re- are also prone to a dose-dependent form of
lationship is often not predictable; thus, osteoporosis through an unclear mechanism.
frequent blood samples are required to mon-
itor the degree of anticoagulation (most
Low Molecular Weight Heparins
commonly, measurement of the activated
partial thromboplastin time), so that the in- Some of the shortcomings of UFH (e.g., short
fusion rate can be adjusted properly. half-life and unpredictable bioavailability)
The usual cardiovascular settings in which have been addressed by the development of
intravenous UFH is indicated include (1) un- low molecular weight heparins (LMWHs),
stable angina (see Chapter 6), (2) acute myo- examples of which are enoxaparin, dalte-
cardial infarction after fibrinolytic therapy parin and tinzaparin. As the name im-
or if an extensive wall motion abnormality plies, LMWH molecules are approximately
is present (see Chapter 7), and (3) pulmonary one-third the size of UFH molecules. LMWHs
embolism or deep venous thrombosis (see Chap- also interact with AT III, but unlike UFH,
ter 15). Among hospitalized or bed-ridden the LMWH–AT III complex preferentially
patients not receiving intravenous heparin, inhibits factor Xa more potently than it in-
10090-17_CH17.qxd 8/31/06 4:58 PM Page 442

442 Chapter Seventeen

hibits thrombin (thrombin inhibition re- ity independently of AT III and are effective
quires heparin molecules larger than those against both circulating and clot-bound
in LMWHs). thrombin. They do not cause thrombo-
Advantages of LMWHs over UFH include cytopenia and are used to maintain anti-
(1) inhibition of platelet-bound factor Xa, coagulation and prevent thrombosis in
contributing to a more prominent anticoagu- patients with heparin-induced thrombo-
lant effect; (2) less binding to plasma proteins cytopenia. Bivalirudin is approved for use as
and endothelial cells, resulting in more pre- an anticoagulant in patients with unstable
dictable bioavailability and a longer half-life; angina undergoing percutaneous coronary
(3) fewer bleeding complications; and (4) a interventions. All the direct thrombin in-
lower incidence of immune-mediated HIT. hibitors are potent anticoagulants and the
From a practical standpoint, the major ad- major adverse effect is bleeding.
vantages of LMWH formulations are the ease
of use and more consistent level of anticoag-
Fondaparinux
ulation. They can be administered as sub-
cutaneous injections once or twice a day in The anticoagulant fondaparinux is a syn-
fixed doses, without the frequent blood mo- thetic analog of heparin that specifically in-
nitoring required for UFH. In rare cases in hibits factor Xa, thereby reducing thrombin
which monitoring the anticoagulant effect is activation. Like heparin, fondaparinux binds
necessary (e.g., in patients with renal dys- to AT III but with very high affinity, which
function, because LMWHs are cleared via the greatly increases the ability of AT III to in-
kidneys), a factor Xa inhibition assay is used. activate factor Xa. Unlike UFH, fondapari-
In clinical trials, LMWH therapy is at least nux does not inactivate formed thrombin,
as effective as UFH in preventing deep venous nor does it interfere with platelet actions or
thrombosis and treating unstable angina. It cause heparin-induced thrombocytopenia.
also has a better safety profile than UFH, It is administered by subcutaneous injec-
with lower rates of bleeding, thrombocyto- tion, and its half-life is sufficiently long (17 to
penia, and osteoporosis. LMWHs should 21 hours) that it can be prescribed just once
not, however, be used in patients with a his- a day. There are no known antidotes to its
tory of heparin-induced thrombocytopenia, anticoagulant effect.
and unlike UFH, the effects of LMWHs Fondaparinux is currently approved for
cannot be completely reversed by protamine. prevention of deep venous thrombosis in pa-
Current clinical indications for LMWH ther- tients undergoing orthopedic surgery of the
apy are (1) prophylaxis against deep venous hip and knees, and as treatment for deep ve-
thrombosis following hip, knee, or abdomi- nous thrombosis and pulmonary embolism.
nal surgery; (2) treatment of deep venous
thrombosis (with or without pulmonary em-
Warfarin
bolism); and (3) management of acute coro-
nary syndromes. Warfarin is an oral agent prescribed for long-
term anticoagulation. It acts by antagonizing
an enzyme (vitamin K epoxide reductase) that
Direct Thrombin Inhibitors
is required for usual vitamin K metabolism.
The anticoagulation effects of UFH and Normally, the reduced form of vitamin K
LMWH are limited because their activity de- promotes the carboxylation of a glutamic
pends, at least in part, on AT III, and they in- acid residue within specific coagulation fac-
hibit only circulating thrombin. The large tors (factors II, VII, IX, and X), an action that
heparin–AT III complex cannot inactivate is necessary for the factors to subsequently
thrombin that is already bound to fibrin bind calcium, become functional, and parti-
within a clot. In contrast, the direct throm- cipate in coagulation (Fig. 17.18). By interfer- Fig. 18
bin inhibitors (lepirudin, bivalirudin, arga- ing with the formation of reduced vitamin K,
troban, and others) inhibit thrombin activ- warfarin indirectly inhibits carboxylation of
10090-17_CH17.qxd 8/31/06 4:58 PM Page 443

Cardiovascular Drugs 443

Precursors of
Epoxide factors II, VII, IX, X
Oxidized reductase Reduced
form of form of
vitamin K – vitamin K Carboxylated
factors II, VII, IX, X
(able to bind Ca++)
WARFARIN

Figure 17.18. Mechanism of action of warfarin. Normally, co-


agulation factors II, VII, IX, and X are converted to functional
forms by carboxylation in the liver, in the presence of reduced vi-
tamin K. During this reaction, vitamin K undergoes oxidation and
must be regenerated back to the reduced state for the sustained
synthesis of functional clotting factors. Warfarin inhibits the for-
mation of reduced vitamin K by antagonizing the enzyme epox-
ide reductase, such that the conversion of the coagulation fac-
tors does not occur and they remain nonfunctional.

the coagulation factors, rendering them in- other antiplatelet agents increases the risk of
active. Because certain natural coagulation a bleeding complication.
inhibitors (protein C and protein S) are also If serious bleeding arises during warfarin
vitamin K dependent, warfarin impairs their therapy, the drug’s effect can be reversed
functions as well, which in some cases may within hours by the administration of vita-
counteract the drug’s anticoagulant effect. min K (or even more quickly by transfusing
Warfarin’s anticoagulation action has a fresh-frozen plasma, which directly replen-
delayed onset of 2 to 7 days; thus, if an im- ishes functional circulating clotting factors).
mediate effect is needed, UFH or LMWH In patients with mechanical heart valves,
must be used concurrently at first. The half- vitamin K should be avoided unless life-
life of warfarin is long (37 hours), and the threatening bleeding occurs, because of the
drug’s dosage must be individualized to possibility of rebound valve thrombosis.
achieve a therapeutic effect while minimiz- Warfarin is teratogenic and should not be
ing the risk of bleeding complications. The taken during pregnancy, especially in the
extent of anticoagulation is monitored by first trimester.
measuring the prothrombin time in blood
samples, reported as an International Nor-
LIPID-REGULATING DRUGS
malized Ratio (INR). There are two “target”
ranges of anticoagulant intensity. For pa- As described in Chapter 5, abnormal serum
tients at greatest risk of pathologic throm- lipid levels play a critical role in the patho-
bosis (e.g., those with certain types of me- genesis of atherosclerosis. Drugs that im-
chanical heart valves), the desired INR is 2.5 prove lipid abnormalities are cardioprotec-
to 3.5. For others (e.g., those with uncom- tive; they inhibit the progression (and in
plicated atrial fibrillation), the target INR is high doses may induce the regression) of
2.0 to 3.0. atherosclerosis, improve cardiovascular out-
Many factors can influence the anticoagu- comes, and in high-risk patients, reduce
lation effect of warfarin and require alter- mortality rates. The most commonly used
ations in its dosage. For example, liver disease lipid-regulating drugs are described in this
or heart failure reduces the warfarin require- section.
ment, whereas a high dietary ingestion of
foods containing vitamin K (e.g., green leafy
HMG CoA Reductase Inhibitors
vegetables) increases the dosage need. Simi-
larly, many pharmaceuticals alter warfarin’s The HMG CoA reductase inhibitors, com-
Tab. 16 anticoagulation effect (Table 17.16). Finally, monly known as statins, are the most effec-
the combined use of warfarin with aspirin or tive drugs for reducing LDL cholesterol. By
10090-17_CH17.qxd 8/31/06 4:58 PM Page 444

444 Chapter Seventeen

TABLE 17.16. Drugs That Alter the Anticoagulation Effect of Warfarin

Reduced Anticoagulation Effect Increased Anticoagulation Effect

Hepatic enzyme induction Hepatic enzyme inhibition


Barbiturates Amiodarone
Rifampin Cephalosporins
Carbamazepine Cimetidine
Nafcillin Erythromycin
Warfarin malabsorption Fluconazole
Cholestyramine Isoniazid
Sucralfate Ketoconazole
Metronidazole
Propafenone
Trimethoprim-sulfamethoxazole
Displacement from protein binding sites
Allopurinol
Gemfibrozil
Phenytoin
Altered vitamin K production by gut flora
Ciprofloxacin
Piperacillin

virtue of their potency, excellent tolerabil- Statins are competitive inhibitors of


ity, and mortality benefits, they are the most the enzyme HMG CoA reductase, a rate-
widely prescribed lipid-regulating drugs. The controlling factor in cholesterol biosynthe-
available agents of this group, in increasing sis (Fig. 17.19). By inhibiting cholesterol Fig. 19
order of potency, are fluvastatin, lovasta- production in the liver, statins lower serum
tin, pravastatin, simvastatin, atorvastatin, LDL cholesterol through three mechanisms:
Tab. 17 and rosuvastatin (Table 17.17). (1) the reduced intrahepatic cholesterol con-

TABLE 17.17. Lipid-Regulating Drugs

Triglyceride
Class LDL Effect HDL Effect Effect Adverse Effects AQ13
HMG CoA reductase inhibitors ↓20–55% ↑5–15% ↓10–30% Transaminitis, myopathy
(in increasing order of potency)
Fluvastatin
Lovastatin
Pravastatin
Simvastatin
Atorvastatin
Rosuvastatin
Bile acid–binding agents ↓15–30% ↑3–5% May ↑ Constipation, bloating
Cholestyramine
Colestipol
Colesevelam
Cholesterol absorption inhibitor ↓15–20% ↑1–2% ↓0–5% Rare allergic reaction
Ezetimibe
Niacin ↓10–25% ↑15–35% ↓20–50% Flushing, hepatotoxicity,
hyperglycemia, hyper-
uricemia,
Fibric acid derivatives ↓0–20% or ↑10–20% ↓20–50% Nausea, gallstones
Fenofibrate ↑0–10%
Gemfibrozil
10090-17_CH17.qxd 8/31/06 4:58 PM Page 445

Cardiovascular Drugs 445

LDL LDL

Hepatocyte
VLDL
HMG remnants
Fibrates
COA
Statins – LPL
TG Niacin
Cholesterol VLDL
APO (mostly TG)
Bile acids

Intestine
Bile-acid
binding agents

Figure 17.19. Major sites of action of lipid-regulating drugs. The


statins inhibit cholesterol biosynthesis in the liver by competing with
the enzyme HMG CoA reductase. This action depletes intrahepatic
cholesterol stores, which results in increased expression of surface low-
density lipoprotein (LDL) receptors. The latter enhance clearance of LDL
and very low-density lipoprotein (VLDL) remnants from the circulation.
The lower intrahepatic cholesterol content leads to reduction in VLDL
synthesis. Ezetimibe selectively inhibits cholesterol uptake in the small
intestine, thereby recuing chylomicron production and cholesterol de-
livery to the liver. Bile acid–binding agents interrupt the enterohepatic
circulation of bile acids in the intestine, causing more hepatic choles-
terol to be diverted to new bile acid production. In response to reduced
availability of intrahepatic cholesterol, LDL receptor expression and LDL
clearance increase. Niacin inhibits VLDL production. It also increases
lipoprotein lipase (LPL) activity, thus promoting triglyceride (TG) clear-
ance from circulating VLDL particles. Niacin raises circulating high-
density lipoprotein (HDL) by impairing hepatic uptake of apo AI, a
major HDL apoprotein (not shown). Fibrates enhance VLDL catabolism
by increasing the synthesis of lipoprotein lipase via peroxisome
proliferator-activated receptor α (PPAR-α), a nuclear transcription
factor. They also raise HDL by stimulating the production of HDL-
associated apoproteins (not shown). Apo, apoproteins.

tent induces increased expression of the LDL causes the formation of LDL, lowering VLDL
receptor gene, causing a greater number of production also decreases circulating LDL
LDL receptors to appear on the surface of the levels. The reduced production of VLDL is
hepatocyte, which facilitates the binding also likely responsible for the triglyceride-
and clearance of LDL from the circulation; lowering effect of statins, because this lipo-
(2) circulating LDL precursors (known as protein is the major carrier of triglycerides
very low density lipoprotein [VLDL] rem- in the circulation.
nants and intermediate-density lipoprotein The overall effect is that statins reduce
particles) are cleared more rapidly from the serum LDL levels by 20% to 55%, depending
circulation because of their cross-recognition on which agent is used. Statins also decrease
with the hepatic LDL receptor; and (3) hepa- plasma triglyceride levels by 7% to 30%, and
tic VLDL production falls due to the reduced by an unclear mechanism, HDL levels in-
availability of intracellular cholesterol for crease by 5% 15%.
lipoprotein assembly. Because the catabo- The lowering of LDL reduces the lipid con-
lism of VLDL in the circulation ultimately tent of atherosclerotic lesions and promotes
10090-17_CH17.qxd 8/31/06 4:58 PM Page 446

446 Chapter Seventeen

plaque stability. This lessens the vulnerabil- cle) with myoglobinuria and renal failure.
ity of plaque to rupture, thus decreasing the When this occurs, muscle-derived creatine
likelihood of thrombus formation and vas- kinase levels in the serum rise to more than
cular occlusion. In addition to their lipid- 10 times the upper limit of normal. This se-
modulating properties, statins have other vere complication occurs in fewer than 0.1%
potentially cardioprotective effects. They im- of patients taking statins alone. However,
prove endothelial function as evidenced by the incidence is increased significantly by
enhanced synthesis of nitric oxide. They fur- concomitant therapy with certain other
ther promote plaque stability by inhibiting drugs, including other lipid-lowering agents
monocyte penetration into the arterial wall (i.e., niacin and the fibric acid derivatives).
and reducing macrophage secretion of met- The incidence is also increased by concur-
alloproteinases, enzymes that degrade and rent administration of drugs that inhibit
weaken the fibrous caps of plaques. Statins the 3A4 isoform of cytochrome P-450, which
also diminish the vulnerability of lipopro- is responsible for hepatic metabolism of
teins to oxidation, thus inhibiting the unreg- most statins. Such drugs include macrolide
ulated uptake of modified LDL cholesterol by antibiotics (e.g., erythromycin, clarithromy-
macrophages. Finally, they appear to suppress cin); azole antifungal agents (e.g., ketocona-
inflammation, thought to be a key aspect of zole, itraconazole); cyclosporine; and many
atherogenesis. HIV protease inhibitors. Notably, pravastatin
Statins are widely prescribed for patients and fluvastatin are not substantially depen-
with CAD, because major trials have shown dent on the cytochrome P-450 3A4 isoform
that they substantially reduce mortality, car- for their metabolism and appear to be less
diac events, and strokes in this population, likely to cause myopathy in combination
whether LDL cholesterol is elevated or even with these other drugs.
in the “normal” range. In studies of patients
not known to have CAD, statin therapy has
Bile Acid–Binding Agents
been shown to reduce coronary events in
high-risk individuals—those with elevated This group includes the resins cholestyra-
LDL cholesterol levels or those with average mine and colestipol and the hydrophilic
total cholesterol but low HDL values. polymer colesevelam. These drugs are large,
Statins are well-tolerated drugs. Mild highly positively charged molecules that bind
gastrointestinal upset or sleep disturbances bile acids (which are negatively charged) in
occasionally occur. The most significant the intestine and prevent their normal re-
potential adverse effects are hepatotoxicity absorption to the liver through the entero-
and myotoxicity (skeletal muscle toxicity), hepatic circulation (see Fig. 17.19). To make
and these are rare. Hepatotoxicity is dose re- up for the loss, more hepatic cholesterol is
lated and occurs in fewer than 1% of pa- converted into newly produced bile acids.
tients. Those affected may experience fatigue, This action depletes intrahepatic cholesterol
anorexia, and weight loss. More commonly, stores, thus stimulating the production of
the patient is asymptomatic, but results of LDL receptors. Similar to the effect of the
routine laboratory studies show an increase statins, an increased number of hepatic LDL
in transaminase levels (ALT, AST). Symp- receptors bind a greater amount of circulat-
toms disappear almost immediately after the ing LDL, reducing the circulating concen-
drug is discontinued, but transaminase lev- tration of the lipoprotein. However, unlike
els may remain elevated for weeks. The risk statins, new hepatic cholesterol production
of statin-associated hepatic toxicity is higher is also stimulated by the reduced intrahepatic
in patients who drink excessive amounts of cholesterol content. The boost in choles-
alcohol. terol synthesis augments VLDL production,
Myotoxicity is characterized by intense which likely explains the commonly observed
myalgias and muscle weakness and can rise in serum triglyceride levels during ther-
lead to rhabdomyolysis (destruction of mus- apy with a bile acid–binding agent.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 447

Cardiovascular Drugs 447

In one of the first drug trials of men with about 18%. However, when combined with
hypercholesterolemia, cholestyramine sig- statin therapy, marked lowering of LDL (by
nificantly reduced the risk of fatal and non- up to 60%) has been reported. Unlike bile
fatal myocardial infarctions. However, drugs acid–binding agents, side effects from eze-
of this class are difficult for patients to take timibe therapy appear to be rare. When com-
(e.g., cholestyramine and colestipol are un- bined with a statin, the incidence of trans-
appealing gritty powders that must be mixed aminase elevation is only slightly greater
with liquids) and their potency is inferior to than that of statin therapy alone. The addi-
that of statins. Thus, the bile acid–binding tion of ezetimibe to a statin regime does not
agents are only occasionally used today, appear to increase the risk of statin-associated
mainly as second-line lipid-regulating drugs, myopathy.
in combination with statin therapy. Because
they can cause elevations of the serum tri-
glyceride level, they should be avoided in
Niacin
patients with hypertriglyceridemia. Niacin is one of the oldest lipid-regulating
The bile acid–binding agents interfere with drugs and has favorable effects on all the cir-
the absorption of fat-soluble vitamins and culating lipid fractions. It is the most effec-
certain drugs (e.g., warfarin, digoxin, propra- tive agent available for raising HDL choles-
nolol, and thyroid hormones). Thus, other terol (by 15% to 35%), and it reduces LDL
medications should be consumed 1 hour be- cholesterol (by 5% to 25%) and triglyceride
fore or 3 to 4 hours after these agents. The levels (by 20% to 50%). Furthermore, unlike
main side effects are gastrointestinal: bloat- most other lipid-lowering drugs, niacin sub-
ing, constipation, and nausea. Because bile stantially reduces the circulating level of lipo-
acid–binding agents are not absorbed into protein (a), an LDL-like lipoprotein that car-
the circulation, they do not cause systemic ries an independent risk of cardiovascular
adverse reactions. disease (see Chapter 5).
Niacin modifies lipid levels through multi-
Cholesterol Absorption ple mechanisms. It inhibits the release of
fatty acids from adipose tissue. As a result,
Inhibitors
fewer fatty acids are transported to the liver
Ezetimibe, the first member of this class of and hepatic triglyceride synthesis declines.
agents, is a selective inhibitor of cholesterol Impaired triglyceride production by the liver
uptake at the brush border of epithelial cells reduces VLDL secretion into the circula-
in the small intestine. It is believed to com- tion; consequently, less LDL is formed (see
petitively inhibit a transporter known as the Fig. 17.19). Niacin also enhances the clear-
Niemann-Pick C1–like 1 protein. Normally, ance of triglycerides from circulating VLDL
a portion of dietary and biliary cholesterol by promoting the activity of lipoprotein li-
taken up in this manner is esterified and in- pase, the enzyme that processes VLDL parti-
corporated into chylomicrons, which then cles by hydrolyzing the triglyceride core at
enter into the circulation and are trans- adipose and muscle cells. The net effect of
ported to the liver (see Box 5.1). By inhi- these actions is a reduction in serum triglyc-
biting cholesterol uptake (see Fig. 17.19), eride and LDL levels. In addition, the drug
ezetimibe results in reduced chylomicron reduces the proportion of small, dense LDL
production and therefore less cholesterol particles (which are thought to promote
delivery to the liver. The reduced cholesterol atherogenesis) in favor of larger and more
content stimulates compensatory hepatic buoyant forms. Niacin raises circulating HDL
production of LDL receptors, which augment cholesterol levels by decreasing the hepatic
clearance of circulating LDL particles. The uptake of its apoprotein, apo A1, thus reduc- AQ11
net result is lowering of circulating LDL (and ing clearance of HDL particles from the cir-
therefore serum cholesterol levels). culation. This mechanism does not disturb
Used alone at standard dosage (10 mg/ hepatic retrieval (and disposal) of cholesterol
day), ezetimibe reduces LDL cholesterol by from the HDL particles.
10090-17_CH17.qxd 8/31/06 4:58 PM Page 448

448 Chapter Seventeen

In a major study of niacin in men who but again total mortality was not signifi-
had experienced a prior myocardial infarc- cantly affected. In a 5-year study of patients
tion, niacin reduced the risk of future car- with type 2 diabetes, fenofibrate reduced the
diac events and lowered the mortality rate incidence of nonfatal myocardial infarction
in long-term follow-up. That study was per- but not the total cardiovascular mortality.
formed before the better-tolerated and more Fibrates are thought to exert their anti-
effective statin drugs were available. Niacin lipid effects through interactions with per-
is now mainly used to treat patients with oxisome proliferator–activated receptor α
low serum HDL levels and/or elevated serum (PPAR-α), a nuclear transcription factor. Ac-
triglycerides. tivation of PPAR-α leads to a decrease in tri-
Niacin has several common side effects. glycerides, at least in part by augmenting
Transient cutaneous flushing episodes occur fatty acid oxidation and increasing the syn-
in most patients. These episodes are pro- thesis of lipoprotein lipase (see Fig. 17.19).
staglandin mediated and can be minimized The latter results in increased VLDL catabo-
by taking aspirin prior to the niacin dose. lism, which may actually augment the circu-
Gastrointestinal side effects include nausea lating LDL level, especially in patients with
and exacerbation of peptic ulcer disease. baseline hypertriglyceridemia. Fibrates raise
Hepatotoxicity can occur, manifested by fa- HDL cholesterol levels via PPAR-α activation
tigue, weakness, and elevated serum trans- of the genes for apoproteins AI and AII,
aminases (ALT, AST). Niacin should be used which are key constituents of HDL particles.
cautiously in diabetic patients because it can Fibrates are primarily used to lower tri-
induce insulin resistance and contribute to glyceride levels and raise HDL cholesterol lev-
hyperglycemia. Niacin also raises serum uric els. They are metabolized by hepatic glucu-
acid levels and can precipitate gout in sus- ronidation with subsequent renal excretion.
ceptible patients. Rare cases of myopathy Thus, they should be avoided or prescribed at
have been reported with niacin therapy. The lower dosages for patients with impaired liver
incidence is increased when niacin is pre- or kidney function.
scribed concurrently with a statin. Fibrates are generally well tolerated. Poten-
tial side effects include dyspepsia, gallstones,
and myalgias. When used in combination
Fibrates
with a statin, the risk of rhabdomyolysis is
The fibric acid derivatives, or fibrates, in- increased. Therefore, if these drugs are pre-
clude gemfibrozil and fenofibrate. They are scribed concurrently, it is recommended that
the most powerful agents to reduce serum the serum creatine kinase (a marker of mus-
triglyceride levels (by up to 50%), and they cle inflammation or necrosis) be monitored
raise HDL cholesterol levels (by up to 20%). every several months. Fibrates augment
However, their effect on LDL cholesterol is the effect of warfarin by displacing it from
more variable: they can actually increase LDL albumin-binding sites, possibly necessitating
in patients with preexisting hypertriglyc- a decrease in the anticoagulant dosage. In a
eridemia. Fibrates shift the proportion of similar fashion, fibrates also enhance the
LDL from smaller and denser sizes to more effects of oral hypoglycemic drugs.
buoyant, larger, and presumably less athero- Table 17.17 summarizes the expected re-
genic particles. sults and potential side effects of the com-
A large study of men who had hyper- monly used lipid-altering drugs.
cholesterolemia but were not known to have
coronary disease showed that gemfibrozil re-
SUMMARY
duced the number of subsequent myocardial
infarctions (without affecting the total death This chapter has presented an overview of
rate). In another study of men with known the most commonly used cardiovascular
CAD, normal LDL levels, and low HDL levels, drugs. These agents are covered in greater de-
the rate of coronary events was decreased, tail in the references listed under “Additional
10090-17_CH17.qxd 8/31/06 4:58 PM Page 449

Cardiovascular Drugs 449

Reading.” It is hoped that the tables, figures, of cardiovascular disorders. Circulation 2006;113:
and brief explanations presented here will be 2556–2564.
useful to the reader now and again when Hirsh J, Fuster V, Ansell J, et al. American Heart
Association/American College of Cardiology
considering the basic pathophysiology of
Foundation guide to warfarin therapy. Circulation
heart disease while caring for patients. 2003;107:1692–1711.
Jorde UP. Suppression of the renin-angiotensin-
aldosterone system in chronic heart failure: choice
Acknowledgments of agents and clinical impact. Cardiol Rev 2006;
14:81–87.
The authors thank Dr. Robert Handin for his helpful
suggestions. Contributors to the previous editions of Krismer AC, Dunser MW, Lindner KH, et al. Vaso-
this chapter were Mark Friedberg, MD; Andrew C. pressin during cardiopulmonary resuscitation and
Hecht, MD; Steven N. Kalkanis, MD; Steven P. Leon, different shock states: a review of the literature.
MD; Chiadi E. Ndumele, MD; David Sloane, MD; Am J Cardiovasc Drugs 2006;6:51–68.
Ralph A. Kelly, MD; Gary R. Strichartz, MD; and Lafuente-Lafuente C, Mouly S, Longas-Tejero MA,
Leonard S. Lilly, MD. et al. Antiarrhythmic drugs for maintaining sinus
rhythm after cardioversion of atrial fibrillation.
Arch Intern Med 2006;166:719–728.
Additional Reading Levine TB, Levine AB. Rationale for the use of angio-
tensin II receptor blockers in patients with left ven-
Antman EM. Cardiovascular Therapeutics: A Com- tricular dysfunction. Clin Cardiol 2005;28:215–218.
panion to Braunwald’s Heart Disease. 3rd Ed. Members of the Sicilian Gambit. New approaches to
Philadelphia: Elsevier Saunders, 2006. antiarrhythmic therapy: emerging therapeutic ap-
Bhatt DL, Fox KAA, Hacke W, et al. Clopidogrel and plications of the cell biology of cardiac arrhythmias.
aspirin versus aspirin alone for the prevention of Circulation 2001;104:2865–2873, 2990–2994.
atheroembolic events. N Engl J Med 2006;354: Nissen SE, Nicholls SJ, Sipahi I, et al. Effect of very
1706–1717. high-intensity statin therapy on regression of coro-
Berger JS, Roncaglioni MC, Avanzini F, et al. Aspirin nary atherosclerosis: the ASTEROID Trial. JAMA
for the primary prevention of cardiovascular events 2006;295:1556–1565.
in women and men. JAMA 2006;295:306–313. Opie LH, Gersh BJ, eds. Drugs for the Heart. 6th Ed.
Danchin N, Cucherat M, Thuillez C, et al. Angio- Philadelphia: WB Saunders, 2004.
tensin-converting enzyme inhibitors in patients Pitt B, White H, Nicolau J, et al. Eplerenone reduces
with coronary artery disease and absence of heart mortality 30 days after randomization following
failure or left ventricular systolic dysfunction. Arch acute myocardial infarction in patients with left
Intern Med 2006;166:787–796. ventricular systolic dysfunction and heart failure.
Daviglus ML, Lloyd-Jones DM, Pirzada A. Preventing J Am Coll Cardiol 2005;46:425–431.
cardiovascular disease in the 21st century: thera- Ridker PM, Cook NR, Lee IM, et al. A randomized
peutic and preventive implications of current evi- trial of low-dose aspirin in the primary prevention
dence. Am J Cardiovasc Drugs 2006;6:87–101. of cardiovascular disease in women. N Engl J Med
Di Nisio M, Middeldorp S, Buller HR. Direct throm- 2005;352:1293–1304.
bin inhibitors. N Engl J Med 2005;353:1028–1040. Sabatine MS, Cannon CP, Gibson CM, et al. Effect of
Egan BM, Basile J, Chilton RJ, et al. Cardioprotection: clopidogrel pretreatment before percutaneous coro-
the role of β-blocker therapy. J Clin Hypertens nary intervention in patients with ST-elevation
2005;7:409–416. myocardial infarction treated with fibrinolytics: the
Expert Panel on Detection, Evaluation, and Treat- PCI-CLARITY Study. JAMA 2005;294:1224–1232.
ment of High Blood Cholesterol in Adults. Execu- Savi P, Herbert JM. Clopidogrel and ticlopidine: P2Y12
tive summary of the Third Report of the National adenosine diphosphate-receptor antagonists for
Cholesterol Education Program Expert Panel on the prevention of atherothrombosis. Sem Thromb
Detection, Evaluation, and Treatment of High Hemost 2005;31:174–183.
Blood Cholesterol in Adults (Adult Treatment Weir R, McMurray JJ. Treatments that improve out-
Panel III). JAMA 2001;285:2486–2497. come in the patient with heart failure, left ventric-
Garcia-Calvo M, Lisnock J, Bull HG, et al. The target ular systolic dysfunction, or both after acute myo-
of ezetimibe is Niemann-Pick C1-Like 1 (NPC1L1). cardial infarction. Heart 2005;91:ii17–ii20.
Proc Natl Acad Sci USA 2005;102:8132–8137. Yan AT, Goodman SG. Low-molecular-weight hepa-
Gheorghiade M, vanVeldhuisen DJ, Colucci WS. rins in ischemic heart disease. Curr Opin Cardiol
Contemporary use of digoxin in the management 2004;19:309–316.
10090-17_CH17.qxd 8/31/06 4:58 PM Page a

Chapter 17—Author Queries


1. AU: Unclear why elements sometimes appear in brackets. Please confirm that usage is
correct.
2. AU: As queried in Chapter 9, should this term be added? Please ensure consistency of
terminology throughout this chapter.
3. AU: Other headings use “Side Effects” or “Adverse Effects,” and tables use “Adverse Ef-
fects.” Perhaps there should be some consistency in terminology, at least in headings.
4. AU: Already listed above
5. AU: For consistency with other sections?
6. AU: Correct, or should it be i.e.?
7. AU: Please confirm correct terminology.
8. AU: Correct to add?
9. AU: For consistency with other sections?
10. AU: Correct to add ch. 6?
11. AU: Later in this chapter (and in Ch. 5) the term used is apo AI (using the letter “I”
rather than the number 1. Are both apo AI and apo A1 correct?
12. AU: Correct symbol?
13. AU: Correct?

Você também pode gostar