You are on page 1of 23

Heart Failure: Practice Essentials, Background, Pathophysiology

1 of 23

http://emedicine.medscape.com/article/163062-overview

Heart Failure
Author: Ioana Dumitru, MD; Chief Editor: Henry H Ooi, MD, MRCPI more...
Updated: Jul 13, 2015

Practice Essentials
Heart failure develops when the heart, via an abnormality of cardiac function
(detectable or not), fails to pump blood at a rate commensurate with the
requirements of the metabolizing tissues or is able to do so only with an elevated
diastolic filling pressure. See the image below.

This chest radiograph shows an enlarged cardiac silhouette and edema at the lung bases,
signs of acute heart failure.

Signs and symptoms

Signs and symptoms of heart failure include the following:


Exertional dyspnea and/or dyspnea at rest
Orthopnea
Acute pulmonary edema
Chest pain/pressure and palpitations
Tachycardia
Fatigue and weakness
Nocturia and oliguria
Anorexia, weight loss, nausea
Exophthalmos and/or visible pulsation of eyes
Distention of neck veins
Weak, rapid, and thready pulse
Rales, wheezing
S 3 gallop and/or pulsus alternans
Increased intensity of P 2 heart sound
Hepatojugular reflux
Ascites, hepatomegaly, and/or anasarca
Central or peripheral cyanosis, pallor

See Clinical Presentation for more detail.

Diagnosis

Heart failure criteria, classification, and staging

The Framingham criteria for the diagnosis of heart failure consists of the concurrent
presence of either 2 major criteria or 1 major and 2 minor criteria.[1]
Major criteria include the following:

Paroxysmal nocturnal dyspnea


Weight loss of 4.5 kg in 5 days in response to treatment
Neck vein distention
Rales
Acute pulmonary edema
Hepatojugular reflux
S 3 gallop
Central venous pressure greater than 16 cm water
Circulation time of 25 seconds

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

2 of 23

http://emedicine.medscape.com/article/163062-overview

Radiographic cardiomegaly
Pulmonary edema, visceral congestion, or cardiomegaly at autopsy

Minor criteria are as follows:

Nocturnal cough
Dyspnea on ordinary exertion
A decrease in vital capacity by one third the maximal value recorded
Pleural effusion
Tachycardia (rate of 120 bpm)
Bilateral ankle edema

The New York Heart Association (NYHA) classification system categorizes heart
failure on a scale of I to IV,[2] as follows:

Class I: No limitation of physical activity


Class II: Slight limitation of physical activity
Class III: Marked limitation of physical activity
Class IV: Symptoms occur even at rest; discomfort with any physical activity

The American College of Cardiology/American Heart Association (ACC/AHA)


staging system is defined by the following 4 stages[3, 4] :
Stage A: High risk of heart failure but no structural heart disease or
symptoms of heart failure
Stage B: Structural heart disease but no symptoms of heart failure
Stage C: Structural heart disease and symptoms of heart failure
Stage D: Refractory heart failure requiring specialized interventions

Testing

The following tests may be useful in the initial evaluation for suspected heart
failure[3, 5, 6] :
Complete blood count (CBC)
Urinalysis
Electrolyte levels
Renal and liver function studies
Fasting blood glucose levels
Lipid profile
Thyroid stimulating hormone (TSH) levels
B-type natriuretic peptide levels
N-terminal pro-B-type natriuretic peptide
Electrocardiography
Chest radiography
2-dimensional (2-D) echocardiography
Nuclear imaging [7]
Maximal exercise testing
Pulse oximetry or arterial blood gas

See Workup for more detail.

Management

Treatment includes the following:

Nonpharmacologic therapy: Oxygen and noninvasive positive pressure


ventilation, dietary sodium and fluid restriction, physical activity as
appropriate, and attention to weight gain
Pharmacotherapy: Diuretics, vasodilators, inotropic agents, anticoagulants,
beta blockers, and digoxin

Surgical options

Surgical treatment options include the following:


Electrophysiologic intervention
Revascularization procedures
Valve replacement/repair
Ventricular restoration
Extracorporeal membrane oxygenation
Ventricular assist devices
Heart transplantation
Total artificial heart

See Treatment and Medication for more detail.

Background

Heart failure is the pathophysiologic state in which the heart, via an abnormality of
cardiac function (detectable or not), fails to pump blood at a rate commensurate with
the requirements of the metabolizing tissues or is able to do so only with an elevated
diastolic filling pressure.

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

3 of 23

http://emedicine.medscape.com/article/163062-overview

Heart failure (see the images below) may be caused by myocardial failure but may
also occur in the presence of near-normal cardiac function under conditions of high
demand. Heart failure always causes circulatory failure, but the converse is not
necessarily the case, because various noncardiac conditions (eg, hypovolemic
shock, septic shock) can produce circulatory failure in the presence of normal,
modestly impaired, or even supranormal cardiac function. To maintain the pumping
function of the heart, compensatory mechanisms increase blood volume, cardiac
filling pressure, heart rate, and cardiac muscle mass. However, despite these
mechanisms, there is progressive decline in the ability of the heart to contract and
relax, resulting in worsening heart failure.

This chest radiograph shows an enlarged cardiac silhouette and edema at the lung bases,
signs of acute heart failure.

A 28-year-old woman presented with acute heart failure secondary to chronic hypertension.
The enlarged cardiac silhouette on this anteroposterior (AP) radiograph is caused by acute
heart failure due to the effects of chronic high blood pressure on the left ventricle. The heart
then becomes enlarged, and fluid accumulates in the lungs (ie, pulmonary congestion).

Signs and symptoms of heart failure include tachycardia and manifestations of


venous congestion (eg, edema) and low cardiac output (eg, fatigue). Breathlessness
is a cardinal symptom of left ventricular (LV) failure that may manifest with
progressively increasing severity.
Heart failure can be classified according to a variety of factors (see Heart Failure
Criteria and Classification). The New York Heart Association (NYHA) classification
for heart failure comprises 4 classes, based on the relationship between symptoms
and the amount of effort required to provoke them, as follows[2] :

Class I patients have no limitation of physical activity


Class II patients have slight limitation of physical activity
Class III patients have marked limitation of physical activity
Class IV patients have symptoms even at rest and are unable to carry on any
physical activity without discomfort

The American College of Cardiology/American Heart Association (ACC/AHA) heart


failure guidelines complement the NYHA classification to reflect the progression of
disease and are divided into 4 stages, as follows[3, 4] :

Stage A patients are at high risk for heart failure but have no structural heart
disease or symptoms of heart failure
Stage B patients have structural heart disease but have no symptoms of

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

4 of 23

http://emedicine.medscape.com/article/163062-overview

heart failure
Stage C patients have structural heart disease and have symptoms of heart
failure
Stage D patients have refractory heart failure requiring specialized
interventions

Laboratory studies for heart failure should include a complete blood count (CBC),
electrolytes, and renal function studies. Imaging studies such as chest radiography
and 2-dimensional echocardiography are recommended in the initial evaluation of
patients with known or suspected heart failure. B-type natriuretic peptide (BNP) and
N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels can be useful in
differentiating cardiac and noncardiac causes of dyspnea. (See the Workup Section
for more information.)

In acute heart failure, patient care consists of stabilizing the patient's clinical
condition; establishing the diagnosis, etiology, and precipitating factors; and initiating
therapies to provide rapid symptom relief and survival benefit. Surgical options for
heart failure include revascularization procedures, electrophysiologic intervention,
cardiac resynchronization therapy (CRT), implantable cardioverter-defibrillators
(ICDs), valve replacement or repair, ventricular restoration, heart transplantation,
and ventricular assist devices (VADs). (See the Treatment Section for more
information.)

The goals of pharmacotherapy are to increase survival and to prevent complications.


Along with oxygen, medications assisting with symptom relief include diuretics,
digoxin, inotropes, and morphine. Drugs that can exacerbate heart failure should be
avoided (nonsteroidal anti-inflammatory drugs [NSAIDs], calcium channel blockers
[CCBs], and most antiarrhythmic drugs). (See the Medication Section for more
information.)
For further information, see the Medscape Reference articles Pediatric Congestive
Heart Failure, Congestive Heart Failure Imaging, Heart Transplantation, Coronary
Artery Bypass Grafting, and Implantable Cardioverter-Defibrillators.

Pathophysiology

The common pathophysiologic state that perpetuates the progression of heart failure
is extremely complex, regardless of the precipitating event. Compensatory
mechanisms exist on every level of organization, from subcellular all the way
through organ-to-organ interactions. Only when this network of adaptations
becomes overwhelmed does heart failure ensue.[8, 9, 10, 11, 12]

Adaptations

Most important among the adaptations are the following[13] :

The Frank-Starling mechanism, in which an increased preload helps to


sustain cardiac performance
Alterations in myocyte regeneration and death
Myocardial hypertrophy with or without cardiac chamber dilatation, in which
the mass of contractile tissue is augmented
Activation of neurohumoral systems

The release of norepinephrine by adrenergic cardiac nerves augments myocardial


contractility and includes activation of the renin-angiotensin-aldosterone system
[RAAS], the sympathetic nervous system [SNS], and other neurohumoral
adjustments that act to maintain arterial pressure and perfusion of vital organs.
In acute heart failure, the finite adaptive mechanisms that may be adequate to
maintain the overall contractile performance of the heart at relatively normal levels
become maladaptive when trying to sustain adequate cardiac performance.[14]

The primary myocardial response to chronic increased wall stress is myocyte


hypertrophy, death/apoptosis, and regeneration.[15] This process eventually leads to
remodeling, usually the eccentric type. Eccentric remodeling further worsens the
loading conditions on the remaining myocytes and perpetuates the deleterious cycle.
The idea of lowering wall stress to slow the process of remodeling has long been
exploited in treating heart failure patients.[16]
The reduction of cardiac output following myocardial injury sets into motion a
cascade of hemodynamic and neurohormonal derangements that provoke activation
of neuroendocrine systems, most notably the above-mentioned adrenergic systems
and RAAS.[17]
The release of epinephrine and norepinephrine, along with the vasoactive
substances endothelin-1 (ET-1) and vasopressin, causes vasoconstriction, which
increases calcium afterload and, via an increase in cyclic adenosine
monophosphate (cAMP), causes an increase in cytosolic calcium entry. The
increased calcium entry into the myocytes augments myocardial contractility and
impairs myocardial relaxation (lusitropy).
The calcium overload may induce arrhythmias and lead to sudden death. The
increase in afterload and myocardial contractility (known as inotropy) and the

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

5 of 23

http://emedicine.medscape.com/article/163062-overview

impairment in myocardial lusitropy lead to an increase in myocardial energy


expenditure and a further decrease in cardiac output. The increase in myocardial
energy expenditure leads to myocardial cell death/apoptosis, which results in heart
failure and further reduction in cardiac output, perpetuating a cycle of further
increased neurohumoral stimulation and further adverse hemodynamic and
myocardial responses.

In addition, the activation of the RAAS leads to salt and water retention, resulting in
increased preload and further increases in myocardial energy expenditure.
Increases in renin, mediated by decreased stretch of the glomerular afferent
arteriole, reduce delivery of chloride to the macula densa and increase beta1adrenergic activity as a response to decreased cardiac output. This results in an
increase in angiotensin II (Ang II) levels and, in turn, aldosterone levels, causing
stimulation of the release of aldosterone. Ang II, along with ET-1, is crucial in
maintaining effective intravascular homeostasis mediated by vasoconstriction and
aldosterone-induced salt and water retention.

The concept of the heart as a self-renewing organ is a relatively recent


development.[18] This new paradigm for myocyte biology has created an entire field
of research aimed directly at augmenting myocardial regeneration. The rate of
myocyte turnover has been shown to increase during times of pathologic stress.[15]
In heart failure, this mechanism for replacement becomes overwhelmed by an even
faster increase in the rate of myocyte loss. This imbalance of hypertrophy and death
over regeneration is the final common pathway at the cellular level for the
progression of remodeling and heart failure.

Ang II

Research indicates that local cardiac Ang II production (which decreases lusitropy,
increases inotropy, and increases afterload) leads to increased myocardial energy
expenditure. Ang II has also been shown in vitro and in vivo to increase the rate of
myocyte apoptosis.[19] In this fashion, Ang II has similar actions to norepinephrine in
heart failure.
Ang II also mediates myocardial cellular hypertrophy and may promote progressive
loss of myocardial function. The neurohumoral factors above lead to myocyte
hypertrophy and interstitial fibrosis, resulting in increased myocardial volume and
increased myocardial mass, as well as myocyte loss. As a result, the cardiac
architecture changes, which, in turn, leads to further increase in myocardial volume
and mass.

Myocytes and myocardial remodeling

In the failing heart, increased myocardial volume is characterized by larger myocytes


approaching the end of their life cycle.[20] As more myocytes drop out, an increased
load is placed on the remaining myocardium, and this unfavorable environment is
transmitted to the progenitor cells responsible for replacing lost myocytes.
Progenitor cells become progressively less effective as the underlying pathologic
process worsens and myocardial failure accelerates. These featuresnamely, the
increased myocardial volume and mass, along with a net loss of myocytesare the
hallmark of myocardial remodeling. This remodeling process leads to early adaptive
mechanisms, such as augmentation of stroke volume (Frank-Starling mechanism)
and decreased wall stress (Laplace's law), and, later, to maladaptive mechanisms
such as increased myocardial oxygen demand, myocardial ischemia, impaired
contractility, and arrhythmogenesis.
As heart failure advances, there is a relative decline in the counterregulatory effects
of endogenous vasodilators, including nitric oxide (NO), prostaglandins (PGs),
bradykinin (BK), atrial natriuretic peptide (ANP), and B-type natriuretic peptide
(BNP). This decline occurs simultaneously with the increase in vasoconstrictor
substances from the RAAS and the adrenergic system, which fosters further
increases in vasoconstriction and thus preload and afterload. This results in cellular
proliferation, adverse myocardial remodeling, and antinatriuresis, with total body
fluid excess and worsening of heart failure symptoms.

Systolic and diastolic failure

Systolic and diastolic heart failure each result in a decrease in stroke volume. This
leads to activation of peripheral and central baroreflexes and chemoreflexes that are
capable of eliciting marked increases in sympathetic nerve traffic.
While there are commonalities in the neurohormonal responses to decreased stroke
volume, the neurohormone-mediated events that follow have been most clearly
elucidated for individuals with systolic heart failure. The ensuing elevation in plasma
norepinephrine directly correlates with the degree of cardiac dysfunction and has
significant prognostic implications. Norepinephrine, while directly toxic to cardiac
myocytes, is also responsible for a variety of signal-transduction abnormalities, such
as down-regulation of beta1-adrenergic receptors, uncoupling of beta2-adrenergic
receptors, and increased activity of inhibitory G-protein. Changes in beta1adrenergic receptors result in overexpression and promote myocardial hypertrophy.

ANP and BNP

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

6 of 23

http://emedicine.medscape.com/article/163062-overview

ANP and BNP are endogenously generated peptides activated in response to atrial
and ventricular volume/pressure expansion. ANP and BNP are released from the
atria and ventricles, respectively, and both promote vasodilation and natriuresis.
Their hemodynamic effects are mediated by decreases in ventricular filling
pressures, owing to reductions in cardiac preload and afterload. BNP, in particular,
produces selective afferent arteriolar vasodilation and inhibits sodium reabsorption
in the proximal convoluted tubule. It also inhibits renin and aldosterone release and,
therefore, adrenergic activation. ANP and BNP are elevated in chronic heart failure.
BNP, in particular, has potentially important diagnostic, therapeutic, and prognostic
implications.
For more information, see the Medscape Reference article Natriuretic Peptides in
Congestive Heart Failure.

Other vasoactive systems

Other vasoactive systems that play a role in the pathogenesis of heart failure include
the ET receptor system, the adenosine receptor system, vasopressin, and tumor
necrosis factor-alpha (TNF-alpha).[21] ET, a substance produced by the vascular
endothelium, may contribute to the regulation of myocardial function, vascular tone,
and peripheral resistance in heart failure. Elevated levels of ET-1 closely correlate
with the severity of heart failure. ET-1 is a potent vasoconstrictor and has
exaggerated vasoconstrictor effects in the renal vasculature, reducing renal plasma
blood flow, glomerular filtration rate (GFR), and sodium excretion.
TNF-alpha has been implicated in response to various infectious and inflammatory
conditions. Elevations in TNF-alpha levels have been consistently observed in heart
failure and seem to correlate with the degree of myocardial dysfunction. Some
studies suggest that local production of TNF-alpha may have toxic effects on the
myocardium, thus worsening myocardial systolic and diastolic function.

In individuals with systolic dysfunction, therefore, the neurohormonal responses to


decreased stroke volume result in temporary improvement in systolic blood pressure
and tissue perfusion. However, in all circumstances, the existing data support the
notion that these neurohormonal responses contribute to the progression of
myocardial dysfunction in the long term.

Heart failure with normal ejection fraction

In diastolic heart failure (heart failure with normal ejection fraction [HFNEF]), the
same pathophysiologic processes occur that lead to decreased cardiac output in
systolic heart failure, but they do so in response to a different set of hemodynamic
and circulatory environmental factors that depress cardiac output.[22]

In HFNEF, altered relaxation and increased stiffness of the ventricle (due to delayed
calcium uptake by the myocyte sarcoplasmic reticulum and delayed calcium efflux
from the myocyte) occur in response to an increase in ventricular afterload (pressure
overload). The impaired relaxation of the ventricle then leads to impaired diastolic
filling of the left ventricle (LV).
Morris et al found that RV subendocardial systolic dysfunction and diastolic
dysfunction, as detected by echocardiographic strain rate imaging, are common in
patients with HFNEF. This dysfunction is potentially associated with the same fibrotic
processes that affect the subendocardial layer of the LV and, to a lesser extent, with
RV pressure overload. This may play a role in the symptomatology of patients with
HFNEF.[23]

LV chamber stiffness

An increase in LV chamber stiffness occurs secondary to any one of, or any


combination of, the following 3 mechanisms:
Rise in filling pressure
Shift to a steeper ventricular pressure-volume curve
Decrease in ventricular distensibility

A rise in filling pressure is the movement of the ventricle up along its pressurevolume curve to a steeper portion, as may occur in conditions such as volume
overload secondary to acute valvular regurgitation or acute LV failure due to
myocarditis.

A shift to a steeper ventricular pressure-volume curve results, most commonly, not


only from increased ventricular mass and wall thickness (as observed in aortic
stenosis and long-standing hypertension) but also from infiltrative disorders (eg,
amyloidosis), endomyocardial fibrosis, and myocardial ischemia.
Parallel upward displacement of the diastolic pressure-volume curve is generally
referred to as a decrease in ventricular distensibility. This is usually caused by
extrinsic compression of the ventricles.

Concentric LV hypertrophy

Pressure overload that leads to concentric LV hypertrophy (LVH), as occurs in aortic


stenosis, hypertension, and hypertrophic cardiomyopathy, shifts the diastolic

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

7 of 23

http://emedicine.medscape.com/article/163062-overview

pressure-volume curve to the left along its volume axis. As a result, ventricular
diastolic pressure is abnormally elevated, although chamber stiffness may or may
not be altered.

Increases in diastolic pressure lead to increased myocardial energy expenditure,


remodeling of the ventricle, increased myocardial oxygen demand, myocardial
ischemia, and eventual progression of the maladaptive mechanisms of the heart that
lead to decompensated heart failure.

Arrhythmias

While life-threatening rhythms are more common in ischemic cardiomyopathy,


arrhythmia imparts a significant burden in all forms of heart failure. In fact, some
arrhythmias even perpetuate heart failure. The most significant of all rhythms
associated with heart failure are the life-threatening ventricular arrhythmias.
Structural substrates for ventricular arrhythmias that are common in heart failure,
regardless of the underlying cause, include ventricular dilatation, myocardial
hypertrophy, and myocardial fibrosis.

At the cellular level, myocytes may be exposed to increased stretch, wall tension,
catecholamines, ischemia, and electrolyte imbalance. The combination of these
factors contributes to an increased incidence of arrhythmogenic sudden cardiac
death in patients with heart failure.

Etiology

Most patients who present with significant heart failure do so because of an inability
to provide adequate cardiac output in that setting. This is often a combination of the
causes listed below in the setting of an abnormal myocardium. The list of causes
responsible for presentation of a patient with heart failure exacerbation is very long,
and searching for the proximate cause to optimize therapeutic interventions is
important.
From a clinical standpoint, classifying the causes of heart failure into the following 4
broad categories is useful:

Underlying causes: Underlying causes of heart failure include structural


abnormalities (congenital or acquired) that affect the peripheral and coronary
arterial circulation, pericardium, myocardium, or cardiac valves, thus leading
to increased hemodynamic burden or myocardial or coronary insufficiency
Fundamental causes: Fundamental causes include the biochemical and
physiologic mechanisms, through which either an increased hemodynamic
burden or a reduction in oxygen delivery to the myocardium results in
impairment of myocardial contraction
Precipitating causes: Overt heart failure may be precipitated by progression
of the underlying heart disease (eg, further narrowing of a stenotic aortic
valve or mitral valve) or various conditions (fever, anemia, infection) or
medications (chemotherapy, NSAIDs) that alter the homeostasis of heart
failure patients
Genetics of cardiomyopathy: Dilated, arrhythmic right ventricular and
restrictive cardiomyopathies are known genetic causes of heart failure.

Underlying causes

Specific underlying factors cause various forms of heart failure, such as systolic
heart failure (most commonly, left ventricular systolic dysfunction), heart failure with
preserved LVEF, acute heart failure, high-output heart failure, and right heart failure.
Underlying causes of systolic heart failure include the following:

Coronary artery disease


Diabetes mellitus
Hypertension
Valvular heart disease (stenosis or regurgitant lesions)
Arrhythmia (supraventricular or ventricular)
Infections and inflammation (myocarditis)
Peripartum cardiomyopathy
Congenital heart disease
Drugs (either recreational, such as alcohol and cocaine, or therapeutic drugs
with cardiac side effects, such as doxorubicin)
Idiopathic cardiomyopathy
Rare conditions (endocrine abnormalities, rheumatologic disease,
neuromuscular conditions)

Underlying causes of diastolic heart failure include the following:


Coronary artery disease
Diabetes mellitus
Hypertension
Valvular heart disease (aortic stenosis)
Hypertrophic cardiomyopathy
Restrictive cardiomyopathy (amyloidosis, sarcoidosis)
Constrictive pericarditis

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

8 of 23

http://emedicine.medscape.com/article/163062-overview

Underlying causes of acute heart failure include the following:

Acute valvular (mitral or aortic) regurgitation


Myocardial infarction
Myocarditis
Arrhythmia
Drugs (eg, cocaine, calcium channel blockers, or beta-blocker overdose)
Sepsis

Underlying causes of high-output heart failure include the following:


Anemia
Systemic arteriovenous fistulas
Hyperthyroidism
Beriberi heart disease
Paget disease of bone
Albright syndrome (fibrous dysplasia)
Multiple myeloma
Pregnancy
Glomerulonephritis
Polycythemia vera
Carcinoid syndrome

Underlying causes of right heart failure include the following:


Left ventricular failure
Coronary artery disease (ischemia)
Pulmonary hypertension
Pulmonary valve stenosis
Pulmonary embolism
Chronic pulmonary disease
Neuromuscular disease

Precipitating causes of heart failure


A previously stable, compensated patient may develop heart failure that is clinically
apparent for the first time when the intrinsic process has advanced to a critical point,
such as with further narrowing of a stenotic aortic valve or mitral valve. Alternatively,
decompensation may occur as a result of failure or exhaustion of the compensatory
mechanisms but without any change in the load on the heart in patients with
persistent, severe pressure or volume overload. In particular, consider whether the
patient has underlying coronary artery disease or valvular heart disease.
The most common cause of decompensation in a previously compensated patient
with heart failure is inappropriate reduction in the intensity of treatment, such as
dietary sodium restriction, physical activity reduction, or drug regimen reduction.
Uncontrolled hypertension is the second most common cause of decompensation,
followed closely by cardiac arrhythmias (most commonly, atrial fibrillation).
Arrhythmias, particularly ventricular arrhythmias, can be life threatening. Also,
patients with one form of underlying heart disease that may be well compensated
can develop heart failure when a second form of heart disease ensues. For
example, a patient with chronic hypertension and asymptomatic LVH may be
asymptomatic until a myocardial infarction (MI) develops and precipitates heart
failure.

Systemic infection or the development of unrelated illness can also lead to heart
failure. Systemic infection precipitates heart failure by increasing total metabolism as
a consequence of fever, discomfort, and cough, increasing the hemodynamic burden
on the heart. Septic shock, in particular, can precipitate heart failure by the release
of endotoxin-induced factors that can depress myocardial contractility.
Cardiac infection and inflammation can also endanger the heart. Myocarditis or
infective endocarditis may directly impair myocardial function and exacerbate
existing heart disease. The anemia, fever, and tachycardia that frequently
accompany these processes are also deleterious. In the case of infective
endocarditis, the additional valvular damage that ensues may precipitate cardiac
decompensation.

Patients with heart failure, particularly when confined to bed, are at high risk of
developing pulmonary emboli, which can increase the hemodynamic burden on the
right ventricle by further elevating right ventricular (RV) systolic pressure, possibly
causing fever, tachypnea, and tachycardia.
Intense, prolonged physical exertion or severe fatigue, such as may result from
prolonged travel or emotional crisis, is a relatively common precipitant of cardiac
decompensation. The same is true of exposure to severe climate change (ie, the
individual comes in contact with a hot, humid environment or a bitterly cold one).
Excessive intake of water and/or sodium and the administration of cardiac
depressants or drugs that cause salt retention are other factors that can lead to
heart failure.

Because of increased myocardial oxygen consumption and demand beyond a


critical level, the following high-output states can precipitate the clinical presentation

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

9 of 23

http://emedicine.medscape.com/article/163062-overview

of heart failure:

Profound anemia
Thyrotoxicosis
Myxedema
Paget disease of bone
Albright syndrome
Multiple myeloma
Glomerulonephritis
Cor pulmonale
Polycythemia vera
Obesity
Carcinoid syndrome
Pregnancy
Nutritional deficiencies (eg, thiamine deficiency, beriberi)

Longitudinal data from the Framingham Heart Study suggests that antecedent
subclinical left ventricular systolic or diastolic dysfunction is associated with an
increased incidence of heart failure, supporting the notion that heart failure is a
progressive syndrome.[24, 25] Another analysis of over 36,000 patients undergoing
outpatient echocardiography reported that moderate or severe diastolic dysfunction,
but not mild diastolic dysfunction, is an independent predictor of mortality.[26]

Genetics of cardiomyopathy

Autosomal dominant inheritance has been demonstrated in dilated cardiomyopathy


and in arrhythmic right ventricular cardiomyopathy. Restrictive cardiomyopathies are
usually sporadic and associated with the gene for cardiac troponin I. Genetic tests
are available at major genetic centers for cardiomyopathies.[27]
In families with a first-degree relative who has been diagnosed with a
cardiomyopathy leading to heart failure, the at-risk patient should be screened and
followed.[27] The recommended screening consists of an electrocardiogram and an
echocardiogram. If the patient has an asymptomatic left ventricular dysfunction, it
should be treated.[27]

Epidemiology

United States statistics


According to the American Heart Association, heart failure affects nearly 5.7 million
Americans of all ages[28] and is responsible for more hospitalizations than all forms
of cancer combined. It is the number 1 cause of hospitalization for Medicare
patients. With improved survival of patients with acute myocardial infarction and with
a population that continues to age, heart failure will continue to increase in
prominence as a major health problem in the United States.[29, 30, 31, 32]
Analysis of national and regional trends in hospitalization and mortality among
Medicare beneficiaries from 1998-2008 showed a relative decline of 29.5% in heart
failure hospitalizations[33] ; however, wide variations are noted between states and
races, with black men having the slowest rate of decline. A relative decline of 6.6%
in mortality was also observed, although the rate was uneven across states. The
length of stay decreased from 6.8 days to 6.4 days, despite an overall increase in
the comorbid conditions.[33]
Heart failure statistics for the United States are as follows:

Heart failure is the fastest-growing clinical cardiac disease entity in the United
States, affecting 2% of the population
Heart failure accounts for 34% of cardiovascular-related deaths [28]
Approximately 670,000 new cases of heart failure are diagnosed each year

[28]

About 277,000 deaths are caused by heart failure each year [28]
Heart failure is the most frequent cause of hospitalization in patients older
than 65 years, with an annual incidence of 10 per 1,000 [28]
Rehospitalization rates during the 6 months following discharge are as much
as 50% [34]
Nearly 2% of all hospital admissions in the United States are for
decompensated heart failure, and the average duration of hospitalization is
about 6 days
In 2010, the estimated total cost of heart failure in the United States was
$39.2 billion, [35] representing 1-2% of all health care expenditures

The incidence and prevalence of heart failure are higher in blacks, Hispanics, Native
Americans, and recent immigrants from developing nations, Russia, and the former
Soviet republics. The higher prevalence of heart failure in blacks, Hispanics, and
Native Americans is directly related to the higher incidence and prevalence of
hypertension and diabetes. This problem is particularly exacerbated by a lack of
access to health care and by substandard preventive health care available to the
most indigent of individuals in these and other groups; in addition, many persons in
these groups do not have adequate health insurance.

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

10 of 23

http://emedicine.medscape.com/article/163062-overview

The higher incidence and prevalence of heart failure in recent immigrants from
developing nations are largely due to a lack of prior preventive health care, a lack of
treatment, or substandard treatment for common conditions, such as hypertension,
diabetes, rheumatic fever, and ischemic heart disease.

Men and women have the same incidence and the same prevalence of heart failure.
However, there are still many differences between men and women with heart
failure, such as the following:

Women tend to develop heart failure later in life than men do


Women are more likely than men to have preserved systolic function
Women develop depression more commonly than men do
Women have signs and symptoms of heart failure similar to those of men, but
they are more pronounced in women
Women survive longer with heart failure than men do

The prevalence of heart failure increases with age. The prevalence is 1-2% of the
population younger than 55 years and increases to a rate of 10% for persons older
than 75 years. Nonetheless, heart failure can occur at any age, depending on the
cause.

International statistics

Heart failure is a worldwide problem. The most common cause of heart failure in
industrialized countries is ischemic cardiomyopathy, with other causes, including
Chagas disease and valvular cardiomyopathy, assuming a more important role in
developing countries. However, in developing nations that have become more
urbanized and more affluent, eating a more processed diet and leading a more
sedentary lifestyle have resulted in an increased rate of heart failure, along with
increased rates of diabetes and hypertension. This change was illustrated in a
population study in Soweto, South Africa, where the community transformed into a
more urban and westernized city, followed by an increase in diabetes, hypertension,
and heart failure.[36]
In terms of treatment, one study showed few important differences in uptake of key
therapies in European countries with widely differing cultures and varying economic
status for patients with heart failure. In contrast, studies of sub-Saharan Africa,
where health care resources are more limited, have shown poor outcomes in
specific populations.[37, 38] For example, in some countries, hypertensive heart
failure carries a 25% 1-year mortality rate, and human immunodeficiency virus
(HIV)associated cardiomyopathy generally progresses to death within 100 days of
diagnosis in patients who are not treated with antiretroviral drugs.
While data regarding developing nations are not as robust as studies of Western
society, the following trends in developing nations are apparent:

Causes tend to be largely nonischemic


Patients tend to present at a younger age
Outcomes are largely worse where health care resources are limited
Isolated right heart failure tends to be more prominent, with a variety of
causes having been postulated, ranging from tuberculous pericardial disease
to lung disease and pollution

Prognosis

In general, the mortality following hospitalization for patients with heart failure is
10.4% at 30 days, 22% at 1 year, and 42.3% at 5 years, despite marked
improvement in medical and device therapy.[28, 39, 40, 41, 42, 43] Each
rehospitalization increases mortality by about 20-22%.[28]

Mortality is greater than 50% for patients with NYHA class IV, ACC/AHA stage D
heart failure. Heart failure associated with acute MI has an inpatient mortality of
20-40%; mortality approaches 80% in patients who are also hypotensive (eg,
cardiogenic shock). (See Heart Failure Criteria and Classification).

Numerous demographic, clinical and biochemical variables have been reported to


provide important prognostic value in patients with heart failure, and several
predictive models have been developed.[44]

A study by van Diepen et al suggests that patients with heart failure or atrial
fibrillation have a significantly higher risk of noncardiac postoperative mortality than
patients with coronary artery disease; this risk should be considered even if a minor
procedure is planned.[45]

A study by Bursi et al found that among community patients with heart failure,
pulmonary artery systolic pressure (PASP), assessed by Doppler echocardiography,
can strongly predict death and can provide incremental and clinically significant
prognostic information independent of known outcome predictors.[46]
Higher concentrations of galectin-3, a marker of cardiac fibrosis, were associated
with an increased risk for incident heart failure (hazard ratio: 1.28 per 1 SD increase
in log galectin-3) in the Framingham Offspring Cohort. Galectin-3 was also
associated with an increased risk for all-cause mortality (multivariable-adjusted

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

11 of 23

http://emedicine.medscape.com/article/163062-overview

hazard ratio: 1.15).[47]

Patient Education
To help prevent recurrence of heart failure in patients in whom heart failure was
caused by dietary factors or medication noncompliance, counsel and educate such
patients about the importance of proper diet and the necessity of medication
compliance. Dunlay et al examined medication use and adherence among
community-dwelling patients with heart failure and found that medication adherence
was suboptimal in many patients, often because of cost.[48] A randomized controlled
trial of 605 patients with heart failure reported that the incidence of all-cause
hospitalization or death was not reduced in patients receiving multi-session self-care
training compared to those receiving a single session intervention. The optimum
method for patient education remains to be established. It appears that more
intensive interventions are not necessarily better.[49]
For patient education information, see the Heart Health Center, Cholesterol Center,
and Diabetes Center, as well as Congestive Heart Failure, High Cholesterol, Chest
Pain, Heart Rhythm Disorders, Coronary Heart Disease, and Heart Attack.
Clinical Presentation

Contributor Information and Disclosures

Author
Ioana Dumitru, MD Associate Professor of Medicine, Division of Cardiology, Founder and Medical Director, Heart
Failure and Cardiac Transplant Program, University of Nebraska Medical Center; Associate Professor of
Medicine, Division of Cardiology, Veterans Affairs Medical Center
Ioana Dumitru, MD is a member of the following medical societies: American College of Cardiology, International
Society for Heart and Lung Transplantation, Heart Failure Society of America
Disclosure: Nothing to disclose.

Coauthor(s)
Mathue M Baker, MD Cardiologist, BryanLGH Heart Institute and Saint Elizabeth Regional Medical Center
Mathue M Baker, MD is a member of the following medical societies: American College of Cardiology
Disclosure: Nothing to disclose.

Specialty Editor Board


Mary L Windle, PharmD Adjunct Associate Professor, University of Nebraska Medical Center College of
Pharmacy; Editor-in-Chief, Medscape Drug Reference
Disclosure: Nothing to disclose.

Chief Editor
Henry H Ooi, MD, MRCPI Director, Advanced Heart Failure and Cardiac Transplant Program, Nashville Veterans
Affairs Medical Center; Assistant Professor of Medicine, Vanderbilt University School of Medicine
Disclosure: Nothing to disclose.

Acknowledgements
Barry E Brenner, MD, PhD, FACEP Professor of Emergency Medicine, Professor of Internal Medicine, Program
Director, Emergency Medicine, Case Medical Center, University Hospitals, Case Western Reserve University
School of Medicine

Barry E Brenner, MD, PhD, FACEP is a member of the following medical societies: Alpha Omega Alpha,
American Academy of Emergency Medicine, American College of Chest Physicians, American College of
Emergency Physicians, American College of Physicians, American Heart Association, American Thoracic Society,
Arkansas Medical Society, New York Academy of Medicine, New York AcademyofSciences,and Society for
Academic Emergency Medicine
Disclosure: Nothing to disclose.

David FM Brown, MD Associate Professor, Division of Emergency Medicine, Harvard Medical School; Vice Chair,
Department of Emergency Medicine, Massachusetts General Hospital
David FM Brown, MD is a member of the following medical societies: American College of Emergency Physicians
and Society for Academic Emergency Medicine

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

12 of 23

http://emedicine.medscape.com/article/163062-overview

Disclosure: Nothing to disclose.

William K Chiang, MD Associate Professor, Department of Emergency Medicine, New York University School of
Medicine; Chief of Service, Department of Emergency Medicine, Bellevue Hospital Center
William K Chiang, MD is a member of the following medical societies: American Academy of Clinical Toxicology,
American College of Medical Toxicology, and Society for Academic Emergency Medicine
Disclosure: Nothing to disclose.

Joseph Cornelius Cleveland Jr, MD Associate Professor, Division of Cardiothoracic Surgery, University of
Colorado Health Sciences Center

Joseph Cornelius Cleveland Jr, MD is a member of the following medical societies: Alpha Omega Alpha,
American Association for the Advancement of Science, American College of Cardiology, American College of
Chest Physicians, American College of Surgeons, American Geriatrics Society, American Physiological Society,
American Society of Transplant Surgeons, Association for Academic Surgery, Heart Failure Society of America,
International Society for Heart and Lung Transplantation, Phi Beta Kappa, Society of Critical Care Medicine,
Society of Thoracic Surgeons, and Western Thoracic Surgical Association
Disclosure: Thoratec Heartmate II Pivotal Tria; Grant/research funds Principal Investigator - Colorado; Abbott
Vascular E-Valve E-clip Honoraria Consulting; Baxter Healthcare Corp Consulting fee Board membership;
Heartware Advance BTT Trial Grant/research funds Principal Investigator- Colorado; Heartware Endurance DT
trial Grant/research funds Principal Investigator-Colorado

Shamai Grossman, MD, MS Assistant Professor, Department of Emergency Medicine, Harvard Medical School;
Director, The Clinical Decision Unit and Cardiac Emergency Center, Beth Israel Deaconess Medical Center
Shamai Grossman, MD, MS is a member of the following medical societies: American College of Emergency
Physicians
Disclosure: Nothing to disclose.

John D Newell Jr, MD Professor of Radiology, Head, Division of Radiology, National Jewish Health; Professor,
Department of Radiology, University of Colorado School of Medicine
John D Newell Jr, MD is a member of the following medical societies: American College of Chest Physicians,
American College of Radiology, American Roentgen Ray Society, American Thoracic Society, Association of
University Radiologists, Radiological Society of North America, and Society of Thoracic Radiology

Disclosure: Siemens Medical Grant/research funds Consulting; Vida Corporation Ownership interest Board
membership; TeraRecon Grant/research funds Consulting; Medscape Reference Honoraria Consulting; Humana
Press Honoraria Other

Craig H Selzman, MD, FACS Associate Professor of Surgery, Surgical Director, Cardiac Mechanical Support and
Heart Transplant, Division of Cardiothoracic Surgery, University of Utah School of Medicine
Craig H Selzman, MD, FACS is a member of the following medical societies: Alpha Omega Alpha, American
Association for Thoracic Surgery, American College of Surgeons, American Physiological Society, Association for
Academic Surgery, International Society for Heart and Lung Transplantation, Society of Thoracic Surgeons,
Southern Thoracic Surgical Association, and Western Thoracic Surgical Association
Disclosure: Nothing to disclose.

Gary Setnik, MD Chair, Department of Emergency Medicine, Mount Auburn Hospital; Assistant Professor,
Division of Emergency Medicine, Harvard Medical School

Gary Setnik, MD is a member of the following medical societies: American College of Emergency Physicians,
National Association of EMS Physicians, and Society for Academic Emergency Medicine

Disclosure: SironaHealth Salary Management position; South Middlesex EMS Consortium Salary Management
position; ProceduresConsult.com Royalty Other

Brett C Sheridan, MD, FACS Associate Professor of Surgery, University of North Carolina at Chapel Hill School
of Medicine
Disclosure: Nothing to disclose.

George A Stouffer III, MD Henry A Foscue Distinguished Professor of Medicine and Cardiology, Director of
Interventional Cardiology, Cardiac Catheterization Laboratory, Chief of Clinical Cardiology, Division of Cardiology,
University of North Carolina Medical Center
George A Stouffer III, MD is a member of the following medical societies: Alpha Omega Alpha, American College
of Cardiology, American College of Physicians, American Heart Association, Phi Beta Kappa, and Society for
Cardiac Angiography and Interventions
Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College
of Pharmacy; Editor-in-Chief, Medscape Drug Reference
Disclosure: Medscape Salary Employment

References
1. Framingham Classification: Ho KK, Pinsky JL, Kannel WB, Levy D. The epidemiology of heart failure: the
Framingham Study. J Am Coll Cardiol. 1993 Oct. 22(4 Suppl A):6A-13A. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

13 of 23

http://emedicine.medscape.com/article/163062-overview

2. American Heart Association. Classes of heart failure. Available at http://www.heart.org/HEARTORG


/Conditions/HeartFailure/AboutHeartFailure/Classes-of-Heart-Failure_UCM_306328_Article.jsp. Accessed:
September 6, 2011.
3. [Guideline] Hunt SA, Abraham WT, Chin MH, et al, and the American College of Cardiology Foundation;
American Heart Association. 2009 Focused update incorporated into the ACC/AHA 2005 guidelines for the
diagnosis and management of heart failure in adults: a report of the American College of Cardiology
Foundation/American Heart Association Task Force on practice guidelines developed in collaboration with
the International Society for Heart and Lung Transplantation. J Am Coll Cardiol. 2009 Apr 14.
53(15):e1-e90. [Medline].

4. [Guideline] Hunt SA, for the Task Force on Practice Guidelines (Writing Committee to Update the 2001
Guidelines for the Evaluation and Management of Heart Failure). ACC/AHA 2005 guideline update for the
diagnosis and management of chronic heart failure in the adult: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2005 Sep 20.
46(6):e1-82. [Medline].
5. [Guideline] Dickstein K, Cohen-Solal A, Filippatos G, et al. for the Task Force for the Diagnosis and
Treatment of Acute and Chronic Heart Failure 2008 of the European Society of Cardiology. ESC Guidelines
for the diagnosis and treatment of acute and chronic heart failure 2008: the Task Force for the Diagnosis
and Treatment of Acute and Chronic Heart Failure 2008 of the European Society of Cardiology. Developed
in collaboration with the Heart Failure Association of the ESC (HFA) and endorsed by the European Society
of Intensive Care Medicine (ESICM). Eur Heart J. 2008 Oct. 29(19):2388-442. [Medline].
6. [Guideline] Lindenfeld J, Albert NM, Boehmer JP, et al, for the Heart Failure Society of America. Executive
summary: HFSA 2010 comprehensive heart failure practice guideline. J Card Fail. 2010 Jun. 16(6):e1-194.
[Medline].
7. Stiles S. FDA Approves Heart Sympathetic Activity Imaging Agent for HF Evaluation. Available at
http://www.medscape.com/viewarticle/781309. Accessed: April 5, 2013.
8. Braunwald E. The pathogenesis of heart failure: Then and now. Medicine. 1991. 70:68.

9. Braunwald E, Ross J Jr, Sonnenblick EH. 2 ed. Mechanisms of Contraction of the Normal and Failing Heart.
Boston: Little Brown & Co; 1976. 417.

10. Clifford R Greyson, MD. Pathophysiology of right ventricular failure. Crit Care Med. 2008. 36(suppl):S57-65.
11. Haddad F, Doyle R, Murphy DJ, Hunt SA. Right ventricular function in cardiovascular disease, part II:
pathophysiology, clinical importance, and management of right ventricular failure. Circulation. 2008 Apr 1.
117(13):1717-31. [Medline].

12. Onwuanyi A, Taylor M. Acute decompensated heart failure: pathophysiology and treatment. Am J Cardiol.
2007 Mar 26. 99(6B):25D-30D. [Medline].
13. Ross J Jr, Braunwald E. Studies on Starling's law of the heart. The effects of impeding venous return on
performance of the normal and failing human left ventricle. Circulation. 1954. 30:719.

14. Gheorghiade M, Pang PS. Acute heart failure syndromes. J Am Coll Cardiol. 2009 Feb 17. 53(7):557-73.
[Medline].

15. Kajstura J, Leri A, Finato N, Di Loreto C, Beltrami CA, Anversa P. Myocyte proliferation in end-stage cardiac
failure in humans. Proc Natl Acad Sci U S A. 1998 Jul 21. 95(15):8801-5. [Medline]. [Full Text].
16. Cohn JN. Structural basis for heart failure. Ventricular remodeling and its pharmacological inhibition.
Circulation. 1995 May 15. 91(10):2504-7. [Medline].

17. Cody RJ. Hormonal alterations in heart failure. Hosenpud JB, Greenberg BH, eds. Congestive Heart
Failure: Pathophysiology, Diagnosis and Comprehensive Approach to Management. Philadelphia: Lippincott
Williams & Wilkins; 2000. 199-212.
18. Anversa P, Nadal-Ginard B. Myocyte renewal and ventricular remodelling. Nature. 2002 Jan 10.
415(6868):240-3. [Medline].

19. Leri A, Claudio PP, Li Q, Wang X, Reiss K, Wang S, et al. Stretch-mediated release of angiotensin II
induces myocyte apoptosis by activating p53 that enhances the local renin-angiotensin system and
decreases the Bcl-2-to-Bax protein ratio in the cell. J Clin Invest. 1998 Apr 1. 101(7):1326-42. [Medline].
[Full Text].

20. Kajstura J, Leri A, Castaldo C, Nadal-Ginard B, Anversa P. Myocyte growth in the failing heart. Surg Clin
North Am. 2004 Feb. 84(1):161-77. [Medline].

21. Feldman AM, Combes A, Wagner D, Kadakomi T, Kubota T, Li YY, et al. The role of tumor necrosis factor in
the pathophysiology of heart failure. J Am Coll Cardiol. 2000 Mar 1. 35(3):537-44. [Medline].
22. Rebecca Gary, RN, PhD, Leslie Davis, MSN, RN, et al. Diastolic heart failure. Heart & Lung. 2007.
37(6):405-16.

23. Morris DA, Gailani M, Vaz Perez A, et al. Right ventricular myocardial systolic and diastolic dysfunction in
heart failure with normal left ventricular ejection fraction. J Am Soc Echocardiogr. 2011 Aug. 24(8):886-97.
[Medline].

24. Lam CS, Lyass A, Kraigher-Krainer E, et al. Cardiac dysfunction and noncardiac dysfunction as precursors
of heart failure with reduced and preserved ejection fraction in the community. Circulation. 2011 Jul 5.
124(1):24-30. [Medline].

25. Ho KK, Anderson KM, Kannel WB, Grossman W, Levy D. Survival after the onset of congestive heart failure

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

14 of 23

http://emedicine.medscape.com/article/163062-overview

in Framingham Heart Study subjects. Circulation. 1993 Jul. 88(1):107-15. [Medline].

26. Halley CM, Houghtaling PL, Khalil MK, Thomas JD, Jaber WA. Mortality rate in patients with diastolic
dysfunction and normal systolic function. Arch Intern Med. 2011 Jun 27. 171(12):1082-7. [Medline].

27. Murphy RT, Starling RC. Genetics and cardiomyopathy: where are we now?. Cleve Clin J Med. 2005 Jun.
72(6):465-6, 469-70, 472-3 passim. [Medline].
28. Roger VL, Go AS, Lloyd-Jones DM, et al, for the American Heart Association Statistics Committee and
Stroke Statistics Subcommittee. Heart disease and stroke statistics--2011 update: a report from the
American Heart Association. Circulation. 2011 Feb 1. 123(4):e18-e209. [Medline].
29. Fonarow GC. Epidemiology and risk stratification in acute heart failure. Am Heart J. 2008 Feb.
155(2):200-7. [Medline].

30. Gottlieb SS, Khatta M, Friedmann E, Einbinder L, Katzen S, Baker B, et al. The influence of age, gender,
and race on the prevalence of depression in heart failure patients. J Am Coll Cardiol. 2004 May 5.
43(9):1542-9. [Medline].

31. He J, Ogden LG, Bazzano LA, Vupputuri S, Loria C, Whelton PK. Risk factors for congestive heart failure in
US men and women: NHANES I epidemiologic follow-up study. Arch Intern Med. 2001 Apr 9.
161(7):996-1002. [Medline].
32. Masoudi FA, Havranek EP, Smith G, Fish RH, Steiner JF, Ordin DL, et al. Gender, age, and heart failure
with preserved left ventricular systolic function. J Am Coll Cardiol. 2003 Jan 15. 41(2):217-23. [Medline].

33. Chen J, Normand SL, Wang Y, Krumholz HM. National and regional trends in heart failure hospitalization
and mortality rates for Medicare beneficiaries, 1998-2008. JAMA. 2011 Oct 19. 306(15):1669-78. [Medline].
34. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service
program. N Engl J Med. 2009 Apr 2. 360(14):1418-28. [Medline].

35. Lloyd-Jones D, Adams RJ, Brown TM, Carnethon M, Dai S, De Simone G, et al. Executive summary: heart
disease and stroke statistics--2010 update: a report from the American Heart Association. Circulation. 2010
Feb 23. 121(7):948-54. [Medline].
36. Stewart S, Wilkinson D, Hansen C, Vaghela V, Mvungi R, McMurray J, et al. Predominance of heart failure
in the Heart of Soweto Study cohort: emerging challenges for urban African communities. Circulation. 2008
Dec 2. 118(23):2360-7. [Medline].

37. Damasceno A, Cotter G, Dzudie A, Sliwa K, Mayosi BM. Heart failure in sub-saharan Africa: time for action.
J Am Coll Cardiol. 2007 Oct 23. 50(17):1688-93. [Medline].

38. Cardiovascular Diseases. Dean T. Jamison, Richard G. Feachem, Eduard R. Bos, Malegapuru W.
Makgoba, Florence K. Baingana, Karen J. Hofman, Kahma O. Rogo. Disease and mortality in Sub-Saharan
Africa. Washington D.C: World Bank Publications; 2006. 2: 21.
39. Dries DL, Exner DV, Domanski MJ, Greenberg B, Stevenson LW. The prognostic implications of renal
insufficiency in asymptomatic and symptomatic patients with left ventricular systolic dysfunction. J Am Coll
Cardiol. 2000 Mar 1. 35(3):681-9. [Medline].
40. Fonarow GC, Adams KF Jr, Abraham WT, Yancy CW, Boscardin WJ. Risk stratification for in-hospital
mortality in acutely decompensated heart failure: classification and regression tree analysis. JAMA. 2005
Feb 2. 293(5):572-80. [Medline].

41. Levy D, Kenchaiah S, Larson MG, Benjamin EJ, Kupka MJ, Ho KK, et al. Long-term trends in the incidence
of and survival with heart failure. N Engl J Med. 2002 Oct 31. 347(18):1397-402. [Medline].
42. Lucas C, Johnson W, Hamilton MA, Fonarow GC, Woo MA, Flavell CM, et al. Freedom from congestion
predicts good survival despite previous class IV symptoms of heart failure. Am Heart J. 2000 Dec.
140(6):840-7. [Medline].

43. MacIntyre K, Capewell S, Stewart S, Chalmers JW, Boyd J, Finlayson A, et al. Evidence of improving
prognosis in heart failure: trends in case fatality in 66 547 patients hospitalized between 1986 and 1995.
Circulation. 2000 Sep 5. 102(10):1126-31. [Medline].

44. Ketchum ES, Levy WC. Establishing prognosis in heart failure: a multimarker approach. Prog Cardiovasc
Dis. 2011 Sep-Oct. 54(2):86-96. [Medline].
45. van Diepen S, Bakal JA, McAlister FA, Ezekowitz JA. Mortality and readmission of patients with heart
failure, atrial fibrillation, or coronary artery disease undergoing noncardiac surgery: an analysis of 38 047
patients. Circulation. 2011 Jul 19. 124(3):289-96. [Medline].
46. Bursi F, McNallan SM, Redfield MM, et al. Pulmonary pressures and death in heart failure a community
study. J Am Coll Cardiol. 2012 Jan 17. 59(3):222-31. [Medline]. [Full Text].

47. Ho JE, Liu C, Lyass A, Courchesne P, Pencina MJ, Vasan RS, et al. Galectin-3, a marker of cardiac fibrosis,
predicts incident heart failure in the community. J Am Coll Cardiol. 2012 Oct 2. 60(14):1249-56. [Medline].
48. Dunlay SM, Eveleth JM, Shah ND, McNallan SM, Roger VL. Medication adherence among communitydwelling patients with heart failure. Mayo Clin Proc. 2011 Apr. 86(4):273-81. [Medline]. [Full Text].

49. Dewalt DA, Schillinger D, Ruo B, Bibbins-Domingo K, Baker DW, Holmes GM, et al. A Multisite Randomized
Trial of a Single- versus Multi-Session Literacy Sensitive Self-Care Intervention for Patients with Heart
Failure. Circulation. 2012 May 9. [Medline].
50. Lindenfeld J, Albert NM, Boehmer JP, Collins SP, Ezekowitz JA, Givertz MM, et al. HFSA 2010
Comprehensive Heart Failure Practice Guideline. J Card Fail. 2010 Jun. 16(6):e1-194. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

15 of 23

http://emedicine.medscape.com/article/163062-overview

51. Lainscak M, Cleland JG, Lenzen MJ, Follath F, Komajda M, Swedberg K. International variations in the
treatment and co-morbidity of left ventricular systolic dysfunction: data from the EuroHeart Failure Survey.
Eur J Heart Fail. 2007 Mar. 9(3):292-9. [Medline].
52. [Guideline] Peacock WF, Fonarow GC, Ander DS, Maisel A, Hollander JE, Januzzi JL Jr, et al. Society of
Chest Pain Centers Recommendations for the evaluation and management of the observation stay acute
heart failure patient: a report from the Society of Chest Pain Centers Acute Heart Failure Committee. Crit
Pathw Cardiol. 2008 Jun. 7(2):83-6. [Medline].

53. Steinhart B, Thorpe KE, Bayoumi AM, Moe G, Januzzi JL Jr, Mazer CD. Improving the diagnosis of acute
heart failure using a validated prediction model. J Am Coll Cardiol. 2009 Oct 13. 54(16):1515-21. [Medline].

54. Jessup M, Abraham WT, Casey DE, Feldman AM, Francis GS, Ganiats TG, et al. 2009 focused update:
ACCF/AHA Guidelines for the Diagnosis and Management of Heart Failure in Adults: a report of the
American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines:
developed in collaboration with the International Society for Heart and Lung Transplantation. Circulation.
2009 Apr 14. 119(14):1977-2016. [Medline].
55. Rich MW, McSherry F, Williford WO, Yusuf S. Effect of age on mortality, hospitalizations and response to
digoxin in patients with heart failure: the DIG study. J Am Coll Cardiol. 2001 Sep. 38(3):806-13. [Medline].
56. Badve SV, Roberts MA, Hawley CM, et al. Effects of Beta-adrenergic antagonists in patients with chronic
kidney disease a systematic review and meta-analysis. J Am Coll Cardiol. 2011 Sep 6. 58(11):1152-61.
[Medline].
57. Maisel AS, Krishnaswamy P, Nowak RM, et al. Rapid measurement of B-type natriuretic peptide in the
emergency diagnosis of heart failure. N Engl J Med. 2002 Jul 18. 347(3):161-7. [Medline].

58. Januzzi JL Jr, Camargo CA, Anwaruddin S, et al. The N-terminal Pro-BNP investigation of dyspnea in the
emergency department (PRIDE) study. Am J Cardiol. 2005 Apr 15. 95(8):948-54. [Medline].
59. Wang CS, FitzGerald JM, Schulzer M, Mak E, Ayas NT. Does this dyspneic patient in the emergency
department have congestive heart failure?. JAMA. 2005 Oct 19. 294(15):1944-56. [Medline].

60. Stevenson LW, Perloff JK. The limited reliability of physical signs for estimating hemodynamics in chronic
heart failure. JAMA. 1989 Feb 10. 261(6):884-8. [Medline].

61. Pinamonti B, Di Lenarda A, Sinagra G, Camerini F. Restrictive left ventricular filling pattern in dilated
cardiomyopathy assessed by Doppler echocardiography: clinical, echocardiographic and hemodynamic
correlations and prognostic implications. Heart Muscle Disease Study Group. J Am Coll Cardiol. 1993 Sep.
22(3):808-15. [Medline].
62. Temporelli PL, Scapellato F, Eleuteri E, Imparato A, Giannuzzi P. Doppler echocardiography in advanced
systolic heart failure: a noninvasive alternative to Swan-Ganz catheter. Circ Heart Fail. 2010 May.
3(3):387-94. [Medline].

63. Maisel AS, McCord J, Nowak RM, et al. Bedside B-Type natriuretic peptide in the emergency diagnosis of
heart failure with reduced or preserved ejection fraction. Results from the Breathing Not Properly
Multinational Study. J Am Coll Cardiol. 2003 Jun 4. 41(11):2010-7. [Medline].
64. Januzzi JL, van Kimmenade R, Lainchbury J, et al. NT-proBNP testing for diagnosis and short-term
prognosis in acute destabilized heart failure: an international pooled analysis of 1256 patients: the
International Collaborative of NT-proBNP Study. Eur Heart J. 2006 Feb. 27(3):330-7. [Medline].

65. Maeda K, Tsutamoto T, Wada A, Hisanaga T, Kinoshita M. Plasma brain natriuretic peptide as a biochemical
marker of high left ventricular end-diastolic pressure in patients with symptomatic left ventricular
dysfunction. Am Heart J. 1998 May. 135(5 Pt 1):825-32. [Medline].
66. Fisher C, Berry C, Blue L, Morton JJ, McMurray J. N-terminal pro B type natriuretic peptide, but not the new
putative cardiac hormone relaxin, predicts prognosis in patients with chronic heart failure. Heart. 2003 Aug.
89(8):879-81. [Medline]. [Full Text].
67. Hall C, Rouleau JL, Moye L, et al. N-terminal proatrial natriuretic factor. An independent predictor of
long-term prognosis after myocardial infarction. Circulation. 1994 May. 89(5):1934-42. [Medline].

68. Andersson B, Hall C. N-terminal proatrial natriuretic peptide and prognosis in patients with heart failure and
preserved systolic function. J Card Fail. 2000 Sep. 6(3):208-13. [Medline].
69. Chen HH, Burnett JC. Natriuretic peptides in the pathophysiology of congestive heart failure. Curr Cardiol
Rep. 2000 May. 2(3):198-205. [Medline].
70. Cheng V, Kazanagra R, Garcia A, Lenert L, Krishnaswamy P, Gardetto N, et al. A rapid bedside test for
B-type peptide predicts treatment outcomes in patients admitted for decompensated heart failure: a pilot
study. J Am Coll Cardiol. 2001 Feb. 37(2):386-91. [Medline].

71. Cowie MR, Struthers AD, Wood DA, Coats AJ, Thompson SG, Poole-Wilson PA, et al. Value of natriuretic
peptides in assessment of patients with possible new heart failure in primary care. Lancet. 1997 Nov 8.
350(9088):1349-53. [Medline].

72. Dao Q, Krishnaswamy P, Kazanegra R, Harrison A, Amirnovin R, Lenert L, et al. Utility of B-type natriuretic
peptide in the diagnosis of congestive heart failure in an urgent-care setting. J Am Coll Cardiol. 2001 Feb.
37(2):379-85. [Medline].

73. Maeda K, Tsutamoto T, Wada A, Hisanaga T, Kinoshita M. Plasma brain natriuretic peptide as a biochemical
marker of high left ventricular end-diastolic pressure in patients with symptomatic left ventricular
dysfunction. Am Heart J. 1998 May. 135(5 Pt 1):825-32. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

16 of 23

http://emedicine.medscape.com/article/163062-overview

74. Maisel AS, Koon J, Hope J, et al. A rapid bedside test for brain natriuretic peptide accurately predicts
cardiac function in patients referred for echocardiography. Am Heart J. 2001. 141:374-9.

75. Masson S, Vago T, Baldi G, et al. Comparative measurement of N-terminal pro-brain natriuretic peptide and
brain natriuretic peptide in ambulatory patients with heart failure. Clin Chem Lab Med. 2002 Aug.
40(8):761-3. [Medline].
76. Song BG, Jeon ES, Kim YH, et al. Correlation between levels of N-terminal pro-B-type natriuretic peptide
and degrees of heart failure. Korean J Intern Med. 2005 Mar. 20(1):26-32. [Medline].
77. Hobbs FD, Davis RC, Roalfe AK, et al. Reliability of N-terminal pro-brain natriuretic peptide assay in
diagnosis of heart failure: cohort study in representative and high risk community populations. [abstract].
BMJ. 2002 Jun 22. 324(7352):1498.

78. Redfield MM, Rodeheffer RJ, Jacobsen SJ, Mahoney DW, Bailey KR, Burnett JC Jr. Plasma brain
natriuretic peptide concentration: impact of age and gender. J Am Coll Cardiol. 2002 Sep 4. 40(5):976-82.
[Medline].

79. St Peter JV, Hartley GG, Murakami MM, Apple FS. B-type natriuretic peptide (BNP) and N-terminal pro-BNP
in obese patients without heart failure: relationship to body mass index and gastric bypass surgery. Clin
Chem. 2006 Apr. 52(4):680-5. [Medline].
80. Rivera M, Cortes R, Salvador A, ET AL. Obese subjects with heart failure have lower N-terminal pro-brain
natriuretic peptide plasma levels irrespective of aetiology. Eur J Heart Fail. 2005 Dec. 7(7):1168-70.
[Medline].

81. Hermann-Arnhof KM, Hanusch-Enserer U, Kaestenbauer T, et al. N-terminal pro-B-type natriuretic peptide
as an indicator of possible cardiovascular disease in severely obese individuals: comparison with patients in
different stages of heart failure. Clin Chem. 2005 Jan. 51(1):138-43. [Medline].
82. Seino Y, Ogawa A, Yamashita T, et al. Application of NT-proBNP and BNP measurements in cardiac care: a
more discerning marker for the detection and evaluation of heart failure. Eur J Heart Fail. 2004 Mar 15.
6(3):295-300. [Medline].
83. Colucci WS, Elkayam U, Horton DP, Abraham WT, Bourge RC, Johnson AD, et al. Intravenous nesiritide, a
natriuretic peptide, in the treatment of decompensated congestive heart failure. Nesiritide Study Group. N
Engl J Med. 2000 Jul 27. 343(4):246-53. [Medline].

84. Ezekowitz JA, Hernandez AF, Starling RC, Yancy CW, Massie B, Hill JA, et al. Standardizing care for acute
decompensated heart failure in a large megatrial: the approach for the Acute Studies of Clinical
Effectiveness of Nesiritide in Subjects with Decompensated Heart Failure (ASCEND-HF). Am Heart J. 2009
Feb. 157(2):219-28. [Medline].

85. Mills RM, LeJemtel TH, Horton DP, Liang C, Lang R, Silver MA, et al. Sustained hemodynamic effects of an
infusion of nesiritide (human b-type natriuretic peptide) in heart failure: a randomized, double-blind, placebocontrolled clinical trial. Natrecor Study Group. J Am Coll Cardiol. 1999 Jul. 34(1):155-62. [Medline].
86. Silver MA, Horton DP, Ghali JK, Elkayam U. Effect of nesiritide versus dobutamine on short-term outcomes
in the treatment of patients with acutely decompensated heart failure. J Am Coll Cardiol. 2002 Mar 6.
39(5):798-803. [Medline].

87. Miller WL, Hartman KA, Burritt MF, Borgeson DD, Burnett JC Jr, Jaffe AS. Biomarker responses during and
after treatment with nesiritide infusion in patients with decompensated chronic heart failure. Clin Chem.
2005 Mar. 51(3):569-77. [Medline].
88. Fitzgerald RL, Cremo R, Gardetto N, et al. Effect of nesiritide in combination with standard therapy on
serum concentrations of natriuretic peptides in patients admitted for decompensated congestive heart
failure. Am Heart J. 2005 Sep. 150(3):471-7. [Medline].

89. Michels VV, Moll PP, Miller FA, et al. The frequency of familial dilated cardiomyopathy in a series of patients
with idiopathic dilated cardiomyopathy. N Engl J Med. 1992 Jan 9. 326(2):77-82. [Medline].
90. Baig MK, Goldman JH, Caforio AL, Coonar AS, Keeling PJ, McKenna WJ. Familial dilated cardiomyopathy:
cardiac abnormalities are common in asymptomatic relatives and may represent early disease. J Am Coll
Cardiol. 1998 Jan. 31(1):195-201. [Medline].
91. Grunig E, Tasman JA, Kucherer H, Franz W, Kubler W, Katus HA. Frequency and phenotypes of familial
dilated cardiomyopathy. J Am Coll Cardiol. 1998 Jan. 31(1):186-94. [Medline].

92. McNally E, MacLeod H, Dellefave L. Arrhythmogenic right ventricular dysplasia/cardiomyopathy, autosomal


dominant. 2005. [Medline].

93. Cheitlin MD, Alpert JS, Armstrong WF, Aurigemma GP, Beller GA, Bierman FZ, et al. ACC/AHA Guidelines
for the Clinical Application of Echocardiography. A report of the American College of Cardiology/American
Heart Association Task Force on Practice Guidelines (Committee on Clinical Application of
Echocardiography). Developed in collaboration with the American Society of Echocardiography. Circulation.
1997 Mar 18. 95(6):1686-744. [Medline].
94. Patel AR, Alsheikh-Ali AA, Mukherjee J, Evangelista A, Quraini D, Ordway LJ, et al. 3D Echocardiography
to Evaluate Right Atrial Pressure in Acutely Decompensated Heart Failure Correlation With Invasive
Hemodynamics. JACC Cardiovasc Imaging. 2011 Sep. 4(9):938-45. [Medline].

95. Prior D, Coller J. Echocardiography in heart failure - a guide for general practice. Aust Fam Physician. 2010
Dec. 39(12):904-9. [Medline].
96. Kirkpatrick JN, Wiegers SE, Lang RM. Left ventricular assist devices and other devices for end-stage heart
failure: utility of echocardiography. Curr Cardiol Rep. 2010 May. 12(3):257-64. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

17 of 23

http://emedicine.medscape.com/article/163062-overview

97. Abraham J, Abraham TP. The role of echocardiography in hemodynamic assessment in heart failure. Heart
Fail Clin. 2009 Apr. 5(2):191-208. [Medline].
98. Kim RJ, Wu E, Rafael A, Chen EL, Parker MA, Simonetti O, et al. The use of contrast-enhanced magnetic
resonance imaging to identify reversible myocardial dysfunction. N Engl J Med. 2000 Nov 16.
343(20):1445-53. [Medline].

99. Ritchie JL, Bateman TM, Bonow RO, Crawford MH, Gibbons RJ, Hall RJ, et al. Guidelines for clinical use of
cardiac radionuclide imaging. Report of the American College of Cardiology/American Heart Association
Task Force on Assessment of Diagnostic and Therapeutic Cardiovascular Procedures (Committee on
Radionuclide Imaging), developed in collaboration with the American Society of Nuclear Cardiology. J Am
Coll Cardiol. 1995 Feb. 25(2):521-47. [Medline].

100. Taillefer R, DePuey EG, Udelson JE, Beller GA, Latour Y, Reeves F. Comparative diagnostic accuracy of
Tl-201 and Tc-99m sestamibi SPECT imaging (perfusion and ECG-gated SPECT) in detecting coronary
artery disease in women. J Am Coll Cardiol. 1997 Jan. 29(1):69-77. [Medline].
101. Bonow RO, Maurer G, Lee KL, et al. Myocardial Viability and Survival in Ischemic Left Ventricular
Dysfunction. N Engl J Med. 2011 Apr 4. [Medline].

102. Binanay C, Califf RM, Hasselblad V, et al. Evaluation study of congestive heart failure and pulmonary artery
catheterization effectiveness: the ESCAPE trial. JAMA. 2005 Oct 5. 294(13):1625-33. [Medline].
103. Bitter T, Westerheide N, Prinz C, Hossain MS, Vogt J, Langer C, et al. Cheyne-Stokes respiration and
obstructive sleep apnoea are independent risk factors for malignant ventricular arrhythmias requiring
appropriate cardioverter-defibrillator therapies in patients with congestive heart failure. Eur Heart J. 2011
Jan. 32(1):61-74. [Medline].
104. Groenveld HF, Januzzi JL, Damman K, et al. Anemia and mortality in heart failure patients a systematic
review and meta-analysis. J Am Coll Cardiol. 2008 Sep 2. 52(10):818-27. [Medline].

105. Ronco C, Haapio M, House AA, Anavekar N, Bellomo R. Cardiorenal syndrome. J Am Coll Cardiol. 2008
Nov 4. 52(19):1527-39. [Medline].

106. House AA, Haapio M, Lassus J, Bellomo R, Ronco C. Therapeutic strategies for heart failure in cardiorenal
syndromes. Am J Kidney Dis. 2010 Oct. 56(4):759-73. [Medline].
107. Giamouzis G, Butler J, Starling RC, Karayannis G, Nastas J, Parisis C, et al. Impact of dopamine infusion
on renal function in hospitalized heart failure patients: results of the Dopamine in Acute Decompensated
Heart Failure (DAD-HF) Trial. J Card Fail. 2010 Dec. 16(12):922-30. [Medline].

108. Pleister AP, Baliga RR, Haas GJ. Acute study of clinical effectiveness of nesiritide in decompensated heart
failure: nesiritide redux. Curr Heart Fail Rep. 2011 Sep. 8(3):226-32. [Medline].

109. Konstam MA, Gheorghiade M, Burnett JC Jr, Grinfeld L, Maggioni AP, Swedberg K, et al. Effects of oral
tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial. JAMA. 2007 Mar
28. 297(12):1319-31. [Medline].
110. Massie BM, O'Connor CM, Metra M, Ponikowski P, Teerlink JR, Cotter G, et al. Rolofylline, an adenosine
A1-receptor antagonist, in acute heart failure. N Engl J Med. 2010 Oct 7. 363(15):1419-28. [Medline].
111. Badve SV, Roberts MA, Hawley CM, et al. Effects of Beta-adrenergic antagonists in patients with chronic
kidney disease a systematic review and meta-analysis. J Am Coll Cardiol. 2011 Sep 6. 58(11):1152-61.
[Medline].

112. Roy D, Talajic M, Nattel S, et al, for the Atrial Fibrillation and Congestive Heart Failure Investigators.
Rhythm control versus rate control for atrial fibrillation and heart failure. N Engl J Med. 2008 Jun 19.
358(25):2667-77. [Medline].

113. Hsu LF, Jas P, Sanders P, Garrigue S, Hocini M, Sacher F, et al. Catheter ablation for atrial fibrillation in
congestive heart failure. N Engl J Med. 2004 Dec 2. 351(23):2373-83. [Medline].

114. Wilton SB, Fundytus A, Ghali WA, Veenhuyzen GD, Quinn FR, Mitchell LB, et al. Meta-analysis of the
effectiveness and safety of catheter ablation of atrial fibrillation in patients with versus without left ventricular
systolic dysfunction. Am J Cardiol. 2010 Nov 1. 106(9):1284-91. [Medline].
115. MacDonald MR, Connelly DT, Hawkins NM, Steedman T, Payne J, Shaw M, et al. Radiofrequency ablation
for persistent atrial fibrillation in patients with advanced heart failure and severe left ventricular systolic
dysfunction: a randomised controlled trial. Heart. 2011 May. 97(9):740-7. [Medline].
116. Chen YM, Li ZB, Zhu M, Cao YM. Effects of exercise training on left ventricular remodelling in heart failure
patients: an updated meta-analysis of randomised controlled trials. Int J Clin Pract. 2012 Aug.
66(8):782-791. [Medline].

117. Mozaffarian D, Lemaitre RN, King IB, et al. Circulating Long-Chain {omega}-3 Fatty Acids and Incidence of
Congestive Heart Failure in Older Adults: The Cardiovascular Health Study: A Cohort Study. Ann Intern
Med. 2011 Aug 2. 155(3):160-70. [Medline].
118. Marchioli R, Levantesi G, Silletta MG, et al, for the GISSI-HF investigators. Effect of n-3 polyunsaturated
fatty acids and rosuvastatin in patients with heart failure: results of the GISSI-HF trial. Expert Rev
Cardiovasc Ther. 2009 Jul. 7(7):735-48. [Medline].

119. Yancy CW, Jessup M, Bozkurt B, Masoudi FA, Butler J, McBride PE, et al. 2013 ACCF/AHA Guideline for
the Management of Heart Failure: A Report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013 Jun 5. [Medline].

120. Swedberg K, Komajda M, Bohm M, et al. Ivabradine and outcomes in chronic heart failure (SHIFT): a

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

18 of 23

http://emedicine.medscape.com/article/163062-overview

randomised placebo-controlled study. Lancet. 2010 Sep 11. 376(9744):875-85. [Medline]. [Full Text].

121. Borer JS, Bohm M, Ford I, et al. Effect of ivabradine on recurrent hospitalization for worsening heart failure
in patients with chronic systolic heart failure: the SHIFT Study. Eur Heart J. 2012 Nov. 33(22):2813-20.
[Medline]. [Full Text].

122. US Food and Drug Administration. FDA approves new drug to treat heart failure. Available at
http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm453845.htm. July 7, 2015; Accessed:
July 8, 2015.
123. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, et al. Angiotensin-neprilysin
inhibition versus enalapril in heart failure. N Engl J Med. 2014 Sep 11. 371 (11):993-1004. [Medline]. [Full
Text].
124. Solomon SD, Zile M, Pieske B, Voors A, Shah A, Kraigher-Krainer E, et al. The angiotensin receptor
neprilysin inhibitor LCZ696 in heart failure with preserved ejection fraction: a phase 2 double-blind
randomised controlled trial. Lancet. 2012 Oct 20. 380 (9851):1387-95. [Medline].

125. Massie BM, Collins JF, Ammon SE, Armstrong PW, Cleland JG, Ezekowitz M, et al. Randomized trial of
warfarin, aspirin, and clopidogrel in patients with chronic heart failure: the Warfarin and Antiplatelet Therapy
in Chronic Heart Failure (WATCH) trial. Circulation. 2009 Mar 31. 119(12):1616-24. [Medline].
126. Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL, Redfield MM. Trends in prevalence and
outcome of heart failure with preserved ejection fraction. N Engl J Med. 2006 Jul 20. 355(3):251-9.
[Medline].
127. Hogg K, Swedberg K, McMurray J. Heart failure with preserved left ventricular systolic function;
epidemiology, clinical characteristics, and prognosis. J Am Coll Cardiol. 2004 Feb 4. 43(3):317-27.
[Medline].

128. Gheorghiade M, Abraham WT, Albert NM, et al, for the OPTIMIZE-HF Investigators and Coordinators.
Systolic blood pressure at admission, clinical characteristics, and outcomes in patients hospitalized with
acute heart failure. JAMA. 2006 Nov 8. 296(18):2217-26. [Medline].

129. Masip J, Roque M, Sanchez B, Fernandez R, Subirana M, Exposito JA. Noninvasive ventilation in acute
cardiogenic pulmonary edema: systematic review and meta-analysis. JAMA. 2005 Dec 28.
294(24):3124-30. [Medline].
130. Peter JV, Moran JL, Phillips-Hughes J, Graham P, Bersten AD. Effect of non-invasive positive pressure
ventilation (NIPPV) on mortality in patients with acute cardiogenic pulmonary oedema: a meta-analysis.
Lancet. 2006 Apr 8. 367(9517):1155-63. [Medline].

131. Winck JC, Azevedo LF, Costa-Pereira A, Antonelli M, Wyatt JC. Efficacy and safety of non-invasive
ventilation in the treatment of acute cardiogenic pulmonary edema--a systematic review and meta-analysis.
Crit Care. 2006. 10(2):R69. [Medline]. [Full Text].
132. Vital FM, Saconato H, Ladeira MT, et al. Non-invasive positive pressure ventilation (CPAP or bilevel NPPV)
for cardiogenic pulmonary edema. Cochrane Database Syst Rev. 2008 Jul 16. CD005351. [Medline].
133. Maeder MT, Kaye DM. Heart failure with normal left ventricular ejection fraction. J Am Coll Cardiol. 2009
Mar 17. 53(11):905-18. [Medline].

134. Gray A, Goodacre S, Newby DE, Masson M, Sampson F, Nicholl J, et al. Noninvasive ventilation in acute
cardiogenic pulmonary edema. N Engl J Med. 2008 Jul 10. 359(2):142-51. [Medline].
135. The CONSENSUS Trial Study Group. Effects of enalapril on mortality in severe congestive heart failure.
Results of the Cooperative North Scandinavian Enalapril Survival Study (CONSENSUS). The
CONSENSUS Trial Study Group. N Engl J Med. 1987 Jun 4. 316(23):1429-35. [Medline].
136. The SOLVD Investigators. Effect of enalapril on survival in patients with reduced left ventricular ejection
fractions and congestive heart failure. N Engl J Med. 1991 Aug 1. 325(5):293-302. [Medline].

137. Zannad F, Alla F, Dousset B, Perez A, Pitt B. Limitation of excessive extracellular matrix turnover may
contribute to survival benefit of spironolactone therapy in patients with congestive heart failure: insights from
the randomized aldactone evaluation study (RALES). Rales Investigators. Circulation. 2000 Nov 28.
102(22):2700-6. [Medline].
138. Busko M. Clevidipine Shows Promise for Acute HF with High BP. Medscape Medical News. Available at
http://www.medscape.com/viewarticle/820377. Accessed: February 18, 2014.
139. Felker GM, Lee KL, Bull DA, Redfield MM, Stevenson LW, Goldsmith SR, et al. Diuretic strategies in
patients with acute decompensated heart failure. N Engl J Med. 2011 Mar 3. 364(9):797-805. [Medline].

140. Liu PP. Cardiorenal syndrome in heart failure: a cardiologist's perspective. Can J Cardiol. 2008 Jul. 24 suppl
B:25B-9B. [Medline]. [Full Text].
141. Kramer BK, Schweda F, Riegger GA. Diuretic treatment and diuretic resistance in heart failure. Am J Med.
1999 Jan. 106(1):90-6. [Medline].
142. Nieminen MS, Bohm M, Cowie MR, et al for the ESC Committe for Practice Guideline (CPG). Executive
summary of the guidelines on the diagnosis and treatment of acute heart failure: the Task Force on Acute
Heart Failure of the European Society of Cardiology. Eur Heart J. 2005 Feb. 26(4):384-416. [Medline].

143. Neuberg GW, Miller AB, O'Connor CM, et al. Diuretic resistance predicts mortality in patients with advanced
heart failure. Am Heart J. 2002 Jul. 144(1):31-8. [Medline].
144. Costanzo MR, Saltzberg M, O'Sullivan J, Sobotka P. Early ultrafiltration in patients with decompensated
heart failure and diuretic resistance. J Am Coll Cardiol. 2005 Dec 6. 46(11):2047-51. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

19 of 23

http://emedicine.medscape.com/article/163062-overview

145. Young JB, Abraham WT, Stevenson LW, et al. Results of the VMAC Trial: vasodilation in the management of
acute congestive heart failure. N Engl J Med. 2000. 102:a2794.
146. Publication Committee for the VMAC Investigators (Vasodilatation in the Management of Acute CHF).
Intravenous nesiritide vs nitroglycerin for treatment of decompensated congestive heart failure: a
randomized controlled trial. JAMA. 2002 Mar 27. 287(12):1531-40. [Medline].

147. Costanzo MR, Jessup M. Treatment of congestion in heart failure with diuretics and extracorporeal
therapies: effects on symptoms, renal function, and prognosis. Heart Fail Rev. 2011 May 11. [Medline].

148. Costanzo MR, Guglin ME, Saltzberg MT, Jessup ML, Bart BA, Teerlink JR, et al. Ultrafiltration versus
intravenous diuretics for patients hospitalized for acute decompensated heart failure. J Am Coll Cardiol.
2007 Feb 13. 49(6):675-83. [Medline].
149. CIBIS Investigators and Committees. A randomized trial of beta-blockade in heart failure. The Cardiac
Insufficiency Bisoprolol Study (CIBIS). Circulation. 1994 Oct. 90(4):1765-73. [Medline].

150. Chaudhry SI, Mattera JA, Curtis JP, Spertus JA, Herrin J, Lin Z, et al. Telemonitoring in patients with heart
failure. N Engl J Med. 2010 Dec 9. 363(24):2301-9. [Medline].
151. Koehler F, Winkler S, Schieber M, et al. Impact of Remote Telemedical Management on Mortality and
Hospitalizations in Ambulatory Patients With Chronic Heart Failure: The Telemedical Interventional
Monitoring in Heart Failure Study. Circulation. 2011 May 3. 123(17):1873-1880. [Medline].

152. US Food and Drug Administration. FDA approves first implantable wireless device with remote monitoring to
measure pulmonary artery pressure in certain heart failure patients (news release). Available at
http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm399024.htm. Accessed: June 2,
2014.
153. O'Riordan M. FDA approves first implantable device for remotely monitoring HF patients. Heartwire. May
28, 2014. [Full Text].

154. Abraham WT, Adamson PB, Bourge RC, Aaron MF, Costanzo MR, Stevenson LW, et al. Wireless
pulmonary artery haemodynamic monitoring in chronic heart failure: a randomised controlled trial. Lancet.
2011 Feb 19. 377(9766):658-66. [Medline].

155. Eisen HJ, Kobashigawa J, Keogh A, Bourge R, Renlund D, Mentzer R, et al. Three-year results of a
randomized, double-blind, controlled trial of mycophenolate mofetil versus azathioprine in cardiac transplant
recipients. J Heart Lung Transplant. 2005 May. 24(5):517-25. [Medline].
156. Yancy CW, Lopatin M, Stevenson LW, De Marco T, Fonarow GC. Clinical presentation, management, and
in-hospital outcomes of patients admitted with acute decompensated heart failure with preserved systolic
function: a report from the Acute Decompensated Heart Failure National Registry (ADHERE) Database. J
Am Coll Cardiol. 2006 Jan 3. 47(1):76-84. [Medline].

157. Dickstein K, Vardas PE, Auricchio A, et al al for the Task Force on Acute Heart Failure of the European
Society of Cardiology. 2010 Focused Update of ESC Guidelines on device therapy in heart failure: an
update of the 2008 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure and
the 2007 ESC guidelines for cardiac and resynchronization therapy. Developed with the special contribution
of the Heart Failure Association and the European Heart Rhythm Association. Eur Heart J. 2010 Nov.
31(21):2677-87. [Medline].
158. Tracy CM, Epstein AE, Darbar D, Dimarco JP, Dunbar SB, Estes NA 3rd, et al. 2012 ACCF/AHA/HRS
Focused Update of the 2008 Guidelines for Device-Based Therapy of Cardiac Rhythm Abnormalities: A
Report of the American College of Cardiology Foundation/American Heart Association Task Force on
Practice Guidelines. Circulation. 2012 Sep 10. [Medline].

159. Miller R. AHA issues mechanical circulatory-support guidance for referring docs. Medscape Medical News.
November 1, 2012. [Full Text].

160. Peura JL, Colvin-Adams M, Francis GS, et al, on behalf of the American Heart Association Heart Failure
and Transplantation Committee of the Council on Clinical Cardiology, Council on Cardiopulmonary, et al.
Recommendations for the Use of Mechanical Circulatory Support: Device Strategies and Patient Selection:
A Scientific Statement From the American Heart Association. Circulation. 2012 Oct 29. [Medline].
161. FDA news release. FDA approves expanded indication for certain pacemakers and defibrillators used to
treat heart failure. US Food and Drug Administration. Available at http://www.fda.gov/NewsEvents
/Newsroom/PressAnnouncements/ucm392861.htm. Accessed: April 14, 2014.
162. Stiles S. FDA Approves Medtronic CRT Devices for Mild HF With AV Block. Medscape [serial online].
Available at http://www.medscape.com/viewarticle/823485. Accessed: April 14, 2014.

163. Levy WC, Lee KL, Hellkamp AS, Poole JE, Mozaffarian D, Linker DT, et al. Maximizing survival benefit with
primary prevention implantable cardioverter-defibrillator therapy in a heart failure population. Circulation.
2009 Sep 8. 120(10):835-42. [Medline].
164. Rosanio S, Schwarz ER, Ahmad M, Jammula P, Vitarelli A, Uretsky BF, et al. Benefits, unresolved
questions, and technical issues of cardiac resynchronization therapy for heart failure. Am J Cardiol. 2005
Sep 1. 96(5):710-7. [Medline].

165. Tang AS, Wells GA, Talajic M, Arnold MO, Sheldon R, Connolly S, et al. Cardiac-resynchronization therapy
for mild-to-moderate heart failure. N Engl J Med. 2010 Dec 16. 363(25):2385-95. [Medline].
166. Giraldi F, Cattadori G, Roberto M, et al. Long-term effectiveness of cardiac resynchronization therapy in
heart failure patients with unfavorable cardiac veins anatomy comparison of surgical versus hemodynamic
procedure. J Am Coll Cardiol. 2011 Jul 26. 58(5):483-90. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

20 of 23

http://emedicine.medscape.com/article/163062-overview

167. Abraham WT, Fisher WG, Smith AL, Delurgio DB, Leon AR, Loh E, et al. Cardiac resynchronization in
chronic heart failure. N Engl J Med. 2002 Jun 13. 346(24):1845-53. [Medline].

168. Moss AJ, Hall WJ, Cannom DS, Klein H, Brown MW, Daubert JP, et al. Cardiac-resynchronization therapy
for the prevention of heart-failure events. N Engl J Med. 2009 Oct 1. 361(14):1329-38. [Medline].
169. Arshad A, Moss AJ, Foster E, Padeletti L, Barsheshet A, Goldenberg I, et al. Cardiac resynchronization
therapy is more effective in women than in men: the MADIT-CRT (Multicenter Automatic Defibrillator
Implantation Trial with Cardiac Resynchronization Therapy) trial. J Am Coll Cardiol. 2011 Feb 15.
57(7):813-20. [Medline].

170. Ruwald MH, Ruwald AC, Jons C, Alexis J, McNitt S, Zareba W, et al. Effect of Metoprolol Versus Carvedilol
on Outcomes in MADIT-CRT (Multicenter Automatic Defibrillator Implantation Trial With Cardiac
Resynchronization Therapy). J Am Coll Cardiol. 2013 Apr 9. 61(14):1518-26. [Medline].
171. Cleland JG, Daubert JC, Erdmann E, Freemantle N, Gras D, Kappenberger L, et al. The effect of cardiac
resynchronization on morbidity and mortality in heart failure. N Engl J Med. 2005 Apr 14. 352(15):1539-49.
[Medline].
172. Bristow MR, Saxon LA, Boehmer J, Krueger S, Kass DA, De Marco T, et al. Cardiac-resynchronization
therapy with or without an implantable defibrillator in advanced chronic heart failure. N Engl J Med. 2004
May 20. 350(21):2140-50. [Medline].
173. Curtis AB, Worley SJ, Adamson PB, Chung ES, Niazi I, Sherfesee L, et al. Biventricular pacing for
atrioventricular block and systolic dysfunction. N Engl J Med. 2013 Apr 25. 368(17):1585-93. [Medline].

174. Emery G. Biventricular pacing better for heart failure patients. Medscape Medical News. April 24, 2013. [Full
Text].
175. Caracciolo EA, Davis KB, Sopko G, Kaiser GC, Corley SD, Schaff H, et al. Comparison of surgical and
medical group survival in patients with left main equivalent coronary artery disease. Long-term CASS
experience. Circulation. 1995 May 1. 91(9):2335-44. [Medline].

176. Eleven-year survival in the Veterans Administration randomized trial of coronary bypass surgery for stable
angina. The Veterans Administration Coronary Artery Bypass Surgery Cooperative Study Group. N Engl J
Med. 1984 Nov 22. 311(21):1333-9. [Medline].

177. Robinson TN, Morrell TD, Pomerantz BJ, Heimbach JK, Cairns CB, Harken AH. Therapeutically accessible
clinical cardiac states. J Am Coll Surg. 2000 Oct. 191(4):452-63. [Medline].
178. Senior R, Lahiri A, Kaul S. Effect of revascularization on left ventricular remodeling in patients with heart
failure from severe chronic ischemic left ventricular dysfunction. Am J Cardiol. 2001 Sep 15. 88(6):624-9.
[Medline].

179. Velazquez EJ, Lee KL, Deja MA, Jain A, Sopko G, Marchenko A, et al. Coronary-Artery Bypass Surgery in
Patients with Left Ventricular Dysfunction. N Engl J Med. 2011 Apr 4. [Medline]. [Full Text].

180. Elefteriades JA, Morales DL, Gradel C, Tollis G Jr, Levi E, Zaret BL. Results of coronary artery bypass
grafting by a single surgeon in patients with left ventricular ejection fractions < or = 30%. Am J Cardiol. 1997
Jun 15. 79(12):1573-8. [Medline].
181. Kron IL, Flanagan TL, Blackbourne LH, Schroeder RA, Nolan SP. Coronary revascularization rather than
cardiac transplantation for chronic ischemic cardiomyopathy. Ann Surg. 1989 Sep. 210(3):348-52;
discussion 352-4. [Medline]. [Full Text].

182. Doenst T, Velazquez EJ, Beyersdorf F, Michler R, Menicanti L, Di Donato M, et al. To STICH or not to
STICH: we know the answer, but do we understand the question?. J Thorac Cardiovasc Surg. 2005 Feb.
129(2):246-9. [Medline].

183. Joyce D, Loebe M, Noon GP, McRee S, Southard R, Thompson L, et al. Revascularization and ventricular
restoration in patients with ischemic heart failure: the STICH trial. Curr Opin Cardiol. 2003 Nov. 18(6):454-7.
[Medline].
184. Sharoni E, Song HK, Peterson RJ, Guyton RA, Puskas JD. Off pump coronary artery bypass surgery for
significant left ventricular dysfunction: safety, feasibility, and trends in methodology over time--an early
experience. Heart. 2006 Apr. 92(4):499-502. [Medline]. [Full Text].

185. Calafiore AM, Di Giammarco G, Teodori G, et al. Late results of first myocardial revascularization in multiple
vessel disease: single versus bilateral internal mammary artery with or without saphenous vein grafts. Eur J
Cardiothorac Surg. 2004 Sep. 26(3):542-8. [Medline].
186. Nishimura RA, Grantham JA, Connolly HM, Schaff HV, Higano ST, Holmes DR Jr. Low-output, low-gradient
aortic stenosis in patients with depressed left ventricular systolic function: the clinical utility of the
dobutamine challenge in the catheterization laboratory. Circulation. 2002 Aug 13. 106(7):809-13. [Medline].
187. Carabello BA. Clinical practice. Aortic stenosis. N Engl J Med. 2002 Feb 28. 346(9):677-82. [Medline].
188. Lindblom D, Lindblom U, Qvist J, Lundstrm H. Long-term relative survival rates after heart valve
replacement. J Am Coll Cardiol. 1990 Mar 1. 15(3):566-73. [Medline].

189. Connolly HM, Oh JK, Schaff HV, Roger VL, Osborn SL, Hodge DO, et al. Severe aortic stenosis with low
transvalvular gradient and severe left ventricular dysfunction:result of aortic valve replacement in 52
patients. Circulation. 2000 Apr 25. 101(16):1940-6. [Medline].

190. deFilippi CR, Willett DL, Brickner ME, Appleton CP, Yancy CW, Eichhorn EJ, et al. Usefulness of
dobutamine echocardiography in distinguishing severe from nonsevere valvular aortic stenosis in patients
with depressed left ventricular function and low transvalvular gradients. Am J Cardiol. 1995 Jan 15.

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

21 of 23

http://emedicine.medscape.com/article/163062-overview

75(2):191-4. [Medline].

191. Vaquette B, Corbineau H, Laurent M, Lelong B, Langanay T, de Place C, et al. Valve replacement in
patients with critical aortic stenosis and depressed left ventricular function: predictors of operative risk, left
ventricular function recovery, and long term outcome. Heart. 2005 Oct. 91(10):1324-9. [Medline]. [Full Text].

192. Dujardin KS, Enriquez-Sarano M, Schaff HV, Bailey KR, Seward JB, Tajik AJ. Mortality and morbidity of
aortic regurgitation in clinical practice. A long-term follow-up study. Circulation. 1999 Apr 13. 99(14):1851-7.
[Medline].
193. Chaliki HP, Mohty D, Avierinos JF, Scott CG, Schaff HV, Tajik AJ, et al. Outcomes after aortic valve
replacement in patients with severe aortic regurgitation and markedly reduced left ventricular function.
Circulation. 2002 Nov 19. 106(21):2687-93. [Medline].
194. Enriquez-Sarano M, Tajik AJ. Clinical practice. Aortic regurgitation. N Engl J Med. 2004 Oct 7.
351(15):1539-46. [Medline].

195. Lancellotti P, Grard PL, Pirard LA. Long-term outcome of patients with heart failure and dynamic
functional mitral regurgitation. Eur Heart J. 2005 Aug. 26(15):1528-32. [Medline].

196. Patel JB, Borgeson DD, Barnes ME, Rihal CS, Daly RC, Redfield MM. Mitral regurgitation in patients with
advanced systolic heart failure. J Card Fail. 2004 Aug. 10(4):285-91. [Medline].
197. Bolling SF, Pagani FD, Deeb GM, Bach DS. Intermediate-term outcome of mitral reconstruction in
cardiomyopathy. J Thorac Cardiovasc Surg. 1998 Feb. 115(2):381-6; discussion 387-8. [Medline].

198. Akasaka T, Yoshida K, Hozumi T, Takagi T, Kaji S, Kawamoto T, et al. Restricted coronary flow reserve in
patients with mitral regurgitation improves after mitral reconstructive surgery. J Am Coll Cardiol. 1998 Dec.
32(7):1923-30. [Medline].

199. Wu AH, Aaronson KD, Bolling SF, Pagani FD, Welch K, Koelling TM. Impact of mitral valve annuloplasty on
mortality risk in patients with mitral regurgitation and left ventricular systolic dysfunction. J Am Coll Cardiol.
2005 Feb 1. 45(3):381-7. [Medline].

200. McGee EC, Gillinov AM, Blackstone EH, Rajeswaran J, Cohen G, Najam F, et al. Recurrent mitral
regurgitation after annuloplasty for functional ischemic mitral regurgitation. J Thorac Cardiovasc Surg. 2004
Dec. 128(6):916-24. [Medline].
201. Srichai MB, Grimm RA, Stillman AE, Gillinov AM, Rodriguez LL, Lieber ML, et al. Ischemic mitral
regurgitation: impact of the left ventricle and mitral valve in patients with left ventricular systolic dysfunction.
Ann Thorac Surg. 2005 Jul. 80(1):170-8. [Medline].
202. Morishita A, Shimakura T, Nonoyama M, Takasaki T. Mitral valve replacement in ischemic mitral
regurgitation. Preservation of both anterior and posterior mitral leaflets. J Cardiovasc Surg (Torino). 2002
Apr. 43(2):147-52. [Medline].
203. Yun KL, Sintek CF, Miller DC, Pfeffer TA, Kochamba GS, Khonsari S, et al. Randomized trial comparing
partial versus complete chordal-sparing mitral valve replacement: effects on left ventricular volume and
function. J Thorac Cardiovasc Surg. 2002 Apr. 123(4):707-14. [Medline].

204. Gillinov AM, Wierup PN, Blackstone EH, Bishay ES, Cosgrove DM, White J, et al. Is repair preferable to
replacement for ischemic mitral regurgitation?. J Thorac Cardiovasc Surg. 2001 Dec. 122(6):1125-41.
[Medline].
205. Miller DC. Ischemic mitral regurgitation redux--to repair or to replace?. J Thorac Cardiovasc Surg. 2001
Dec. 122(6):1059-62. [Medline].

206. Feldman T, Kar S, Rinaldi M, Fail P, Hermiller J, Smalling R, et al. Percutaneous mitral repair with the
MitraClip system: safety and midterm durability in the initial EVEREST (Endovascular Valve Edge-to-Edge
REpair Study) cohort. J Am Coll Cardiol. 2009 Aug 18. 54(8):686-94. [Medline].

207. Buckberg GD. Ventricular restoration--a surgical approach to reverse ventricular remodeling. Heart Fail Rev.
2004 Oct. 9(4):233-9; discussion 347-51. [Medline].
208. Yamaguchi A, Ino T, Adachi H, Murata S, Kamio H, Okada M, et al. Left ventricular volume predicts
postoperative course in patients with ischemic cardiomyopathy. Ann Thorac Surg. 1998 Feb. 65(2):434-8.
[Medline].
209. Mickleborough LL, Carson S, Ivanov J. Repair of dyskinetic or akinetic left ventricular aneurysm: results
obtained with a modified linear closure. J Thorac Cardiovasc Surg. 2001 Apr. 121(4):675-82. [Medline].

210. Ott DA, Parravacini R, Cooley DA, DePuey EG, Reul GJ, Duncan JM, et al. Improved cardiac function
following left ventricular aneurysm resection: pre- and postoperative performance studies in 150 patients.
Tex Heart Inst J. 1982 Sep. 9(3):267-73. [Medline]. [Full Text].

211. Athanasuleas CL, Buckberg GD, Stanley AW, Siler W, Dor V, Di Donato M, et al. Surgical ventricular
restoration in the treatment of congestive heart failure due to post-infarction ventricular dilation. J Am Coll
Cardiol. 2004 Oct 6. 44(7):1439-45. [Medline].

212. Jones RH, Velazquez EJ, Michler RE, et al. Coronary bypass surgery with or without surgical ventricular
reconstruction. N Engl J Med. 2009 Apr 23. 360(17):1705-17. [Medline]. [Full Text].
213. Timms D. A review of clinical ventricular assist devices. Med Eng Phys. 2011 Jun 10. [Medline].

214. Pamboukian SV, Tallaj JA, Brown RN, Holman WL, Blood M, George JF, et al. Improvement in 2-year
survival for ventricular assist device patients after implementation of an intensive surveillance protocol. J
Heart Lung Transplant. 2011 Aug. 30(8):879-87. [Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

22 of 23

http://emedicine.medscape.com/article/163062-overview

215. Lietz K, Long JW, Kfoury AG, Slaughter MS, Silver MA, Milano CA, et al. Outcomes of left ventricular assist
device implantation as destination therapy in the post-REMATCH era: implications for patient selection.
Circulation. 2007 Jul 31. 116(5):497-505. [Medline].
216. Daneshmand MA, Rajagopal K, Lima B, Khorram N, Blue LJ, Lodge AJ, et al. Left ventricular assist device
destination therapy versus extended criteria cardiac transplant. Ann Thorac Surg. 2010 Apr. 89(4):1205-9;
discussion 1210. [Medline].

217. Rose EA, Gelijns AC, Moskowitz AJ, Heitjan DF, Stevenson LW, Dembitsky W, et al. Long-term use of a left
ventricular assist device for end-stage heart failure. N Engl J Med. 2001 Nov 15. 345(20):1435-43.
[Medline].
218. Park SJ, Tector A, Piccioni W, Raines E, Gelijns A, Moskowitz A, et al. Left ventricular assist devices as
destination therapy: a new look at survival. J Thorac Cardiovasc Surg. 2005 Jan. 129(1):9-17. [Medline].

219. Starling RC, Naka Y, Boyle AJ, Gonzalez-Stawinski G, John R, Jorde U, et al. Results of the post-U.S. Food
and Drug Administration-approval study with a continuous flow left ventricular assist device as a bridge to
heart transplantation: a prospective study using the INTERMACS (Interagency Registry for Mechanically
Assisted Circulatory Support). J Am Coll Cardiol. 2011 May 10. 57(19):1890-8. [Medline].
220. Ventura PA, Alharethi R, Budge D, Reid BB, Horne BD, Mason NO, et al. Differential impact on
post-transplant outcomes between pulsatile- and continuous-flow left ventricular assist devices. Clin
Transplant. 2011 Jul. 25(4):E390-5. [Medline].

221. Slaughter MS, Pagani FD, et al, for the HeartMate II Clinical Investigators. Clinical management of
continuous-flow left ventricular assist devices in advanced heart failure. J Heart Lung Transplant. 2010 Apr.
29(4 suppl):S1-39. [Medline].
222. Kirklin JK, Naftel DC, Kormos RL, et al. Third INTERMACS Annual Report: the evolution of destination
therapy in the United States. J Heart Lung Transplant. 2011 Feb. 30(2):115-23. [Medline].

223. Taylor DO, Edwards LB, Boucek MM, Trulock EP, Deng MC, Keck BM, et al. Registry of the International
Society for Heart and Lung Transplantation: twenty-second official adult heart transplant report--2005. J
Heart Lung Transplant. 2005 Aug. 24(8):945-55. [Medline].

224. Boucek MM, Edwards LB, Keck BM, Trulock EP, Taylor DO, Hertz MI. Registry of the International Society
for Heart and Lung Transplantation: eighth official pediatric report--2005. J Heart Lung Transplant. 2005
Aug. 24(8):968-82. [Medline].

225. Stevenson LW, Kormos RL, Bourge RC, Gelijns A, Griffith BP, Hershberger RE, et al. Mechanical cardiac
support 2000: current applications and future trial design. June 15-16, 2000 Bethesda, Maryland. J Am Coll
Cardiol. 2001 Jan. 37(1):340-70. [Medline].
226. Gray NA Jr, Selzman CH. Current status of the total artificial heart. Am Heart J. 2006 Jul. 152(1):4-10.
[Medline].

227. Cooley DA, Liotta D, Hallman GL, Bloodwell RD, Leachman RD, Milam JD. Orthotopic cardiac prosthesis
for two-staged cardiac replacement. Am J Cardiol. 1969 Nov. 24(5):723-30. [Medline].

228. Platis A, Larson DF. CardioWest temporary total artificial heart. Perfusion. 2009 Sep. 24(5):341-6. [Medline].
229. Roussel JC, Senage T, Baron O, et al. CardioWest (Jarvik) total artificial heart: a single-center experience
with 42 patients. Ann Thorac Surg. 2009 Jan. 87(1):124-9; discussion 130. [Medline].

230. Anderson E, Jaroszewski D, Pierce C, DeValeria P, Arabia F. Parallel application of extracorporeal


membrane oxygenation and the CardioWest total artificial heart as a bridge to transplant. Ann Thorac Surg.
2009 Nov. 88(5):1676-8. [Medline].
231. Morris RJ. Total artificial heart--concepts and clinical use. Semin Thorac Cardiovasc Surg. 2008 Fall.
20(3):247-54. [Medline].

232. Meyer A, Slaughter M. The total artificial heart. Panminerva Med. 2011 Sep. 53(3):141-54. [Medline].

233. Anand IS, Carson P, Galle E, Song R, Boehmer J, Ghali JK, et al. Cardiac resynchronization therapy
reduces the risk of hospitalizations in patients with advanced heart failure: results from the Comparison of
Medical Therapy, Pacing and Defibrillation in Heart Failure (COMPANION) trial. Circulation. 2009 Feb 24.
119(7):969-77. [Medline].
234. [Guideline] Bangalore S, Kumar S, Messerli FH. When conventional heart failure therapy is not enough:
angiotensin receptor blocker, direct renin inhibitor, or aldosterone antagonist?. Congest Heart Fail. 2012
Dec 12. [Medline].

235. Libby, Bonow, Mann, Zipes. Braunwald's Heart Disease a Textbook of Cardiovascular Medicine. 8th edition.
2008. 509-727.
236. Cleland JG, Teerlink JR, Senior R, Nifontov EM, Mc Murray JJ, Lang CC, et al. The effects of the cardiac
myosin activator, omecamtiv mecarbil, on cardiac function in systolic heart failure: a double-blind, placebocontrolled, crossover, dose-ranging phase 2 trial. Lancet. 2011 Aug 20. 378(9792):676-83. [Medline].
237. DeBakey ME. Development of mechanical heart devices. Ann Thorac Surg. 2005 Jun. 79(6):S2228-31.
[Medline].

238. [Guideline] DiDomenico RJ, Park HY, Southworth MR, Eyrich HM, Lewis RK, Finley JM, et al. Guidelines for
acute decompensated heart failure treatment. Ann Pharmacother. 2004 Apr. 38(4):649-60. [Medline].
239. Eklind-Cervenka M, Benson L, Dahlstrom U, Edner M, Rosenqvist M, Lund LH. Association of candesartan
vs losartan with all-cause mortality in patients with heart failure. JAMA. 2011 Jan 12. 305(2):175-82.
[Medline].

14-Oct-15 11:14 AM

Heart Failure: Practice Essentials, Background, Pathophysiology

23 of 23

http://emedicine.medscape.com/article/163062-overview

240. [Guideline] Feldman D, Pamboukian SV, Teuteberg JJ, et al. The 2013 International Society for Heart and
Lung Transplantation Guidelines for Mechanical Circulatory Support: Executive Summary. J Heart Lung
Transplant. 2013. 32:157-187.
241. Freeman JV, Yang J, Sung SH, Hlatky MA, Go AS. Effectiveness and safety of digoxin among
contemporary adults with incident systolic heart failure. Circ Cardiovasc Qual Outcomes. 2013 Sep 1.
6(5):525-33. [Medline].

242. Fumoto H, Horvath DJ, Rao S, Massiello AL, Horai T, Takaseya T, et al. In vivo acute performance of the
Cleveland Clinic self-regulating, continuous-flow total artificial heart. J Heart Lung Transplant. 2010 Jan.
29(1):21-6. [Medline]. [Full Text].
243. Jarcho JA. Biventricular pacing. N Engl J Med. 2006 Jul 20. 355(3):288-94. [Medline].

244. Jennings DL, Chambers RM, Schillig JM. The pharmacotherapy of the HeartMate II, a continuous flow left
ventricular assist device, in patients with advanced heart failure: integration of disease, device, and drug.
Ann Pharmacother. 2010 Oct. 44(10):1647-50. [Medline].
245. Opie LH. Digitalis, yesterday and today, but not forever. Circ Cardiovasc Qual Outcomes. 2013 Sep 1.
6(5):511-3. [Medline].
246. Plank B, Kutyifa V, Moss AJ, Huang DT, Ruwald AC, McNitt S, et al. Smoking is Associated with an
Increased Risk of First and Recurrent Ventricular Tachyarrhythmias in Mild Heart Failure Patients with
Ischemic and Non-Ischemic Cardiomyopathy - A MADIT-CRT Sub-Study. Heart Rhythm. 2014 Feb 6.
[Medline].

247. Stiles S. Doubling down on RAAS blockade in HF? Aldosterone antagonists, not ARBs, says meta-analysis.
Medscape. Dec 26 2012. [Full Text].
248. Stiles S. Smoking Can Up Risk of Delivered ICD Therapy in Mild HF. Medscape Medical News. Available at
http://www.medscape.com/viewarticle/820637. Accessed: March 2, 2014.
249. Stiles S. A Walk Through Long-Term Mechanical Circulatory Support: New ISHLT Guidelines. Available at
http://www.medscape.com/viewarticle/777744. Accessed: January 23, 2013.
250. Stiles S. Friend or Foe? Digoxin for Systolic HF Ups Mortality in Cohort Study. Medscape Medical News.
Available at http://www.medscape.com/viewarticle/811298. Accessed: September 24, 2013.
251. Topilsky Y, Oh JK, Atchison FW, Shah DK, Bichara VM, Schirger JA, et al. Echocardiographic findings in
stable outpatients with properly functioning HeartMate II left ventricular assist devices. J Am Soc
Echocardiogr. 2011 Feb. 24(2):157-69. [Medline].

252. Yancy CW, Jessup M, Bozkurt B, Masoudi FA, Butler J, McBride PE, et al. 2013 ACCF/AHA Guideline for
the Management of Heart Failure: A Report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013 Jun 5. [Medline].

Medscape Reference 2011 WebMD, LLC

14-Oct-15 11:14 AM

You might also like