Skip to Main Content
Book cover for The ESC Textbook of Cardiovascular Medicine (2 edn) The ESC Textbook of Cardiovascular Medicine (2 edn)

A newer edition of this book is available.

Close

Contents

Book cover for The ESC Textbook of Cardiovascular Medicine (2 edn) The ESC Textbook of Cardiovascular Medicine (2 edn)
Disclaimer
Oxford University Press makes no representation, express or implied, that the drug dosages in this book are correct. Readers must therefore always … More Oxford University Press makes no representation, express or implied, that the drug dosages in this book are correct. Readers must therefore always check the product information and clinical procedures with the most up to date published product information and data sheets provided by the manufacturers and the most recent codes of conduct and safety regulations. The authors and the publishers do not accept responsibility or legal liability for any errors in the text or for the misuse or misapplication of material in this work. Except where otherwise stated, drug dosages and recommendations are for the non-pregnant adult who is not breastfeeding.

Although morbidity and mortality are improving, heart failure continues to present major challenges to healthcare systems. This affliction of mainly the elderly may be falling in incidence but is probably growing in prevalence, in part due to greater longevity resulting from evidence-based drug and device therapy for patients with a low ejection fraction (EF). New guidelines emphasize the combination of an angiotensin-converting enzyme inhibitor and beta-blocker as the cornerstone of therapy, with the addition of either an angiotensin receptor blocker or aldosterone antagonist as the third disease-modifying agent in patients who remain symptomatic. An implanted cardioverter defibrillator should also be added in patients with a persistently low EF and life expectancy of reasonable quality of ≥12 months. In patients in sinus rhythm, New York Heart Association class III–IV, and a QRS duration ≥120ms, cardiac resynchronization therapy has substantial additional morbidity and mortality benefits. This evidence-based care should be delivered through an organized and seamless multidisciplinary framework with a focus on the patients’ and carers’ needs. Apart from transplantation, the place of any other surgical intervention remains uncertain. Unfortunately the success of treatment in low EF heart failure has not been replicated in patients with heart failure and a preserved EF, and treatment of these patients remains empirical. The same is largely true for the treatment of patients with acute heart failure, with no treatment yet shown to be superior to empirical therapy with diuretic, oxygen, and nitrates. In particular the role of inotropes remains uncertain.

Heart failure is the term used to describe a common clinical syndrome arising, in ways that are incompletely understood, as a consequence of reduced cardiac pump function. The term ‘syndrome’ merely describes a constellation of symptoms and signs and, therefore, heart failure is not a diagnosis as such. Unfortunately, the typical symptoms (breathlessness and fatigue) and signs (e.g. oedema) of heart failure are relatively non-specific, making clinical confirmation of the syndrome difficult. The syndrome of heart failure itself can arise as a result of almost any abnormality of the structure, mechanical function, or electrical activity of the heart, each of which may require quite different treatments, emphasizing the importance of appropriate investigation of patients with suspected heart failure.

Many of the typical clinical symptoms and signs of heart failure do not arise directly as a result of the cardiac abnormality but rather from secondary dysfunction of other organs and tissues, e.g. the kidneys, bone marrow, and muscles. These secondary consequences of pump failure are myriad and their causes are not fully elucidated. Dysfunction of tissues and organs remote from the heart cannot, however, be explained solely by reduced perfusion and it is generally believed that other systemic processes (e.g. neurohumoral activation) are involved. In other words, the pathophysiology of heart failure is complex and incompletely understood and, consequently, so is the pathophysiological basis of treatment. Although it has proved difficult to agree a simple definition of heart failure, a pragmatic approach has been advocated (graphic Table 23.1) [1]. The terms used to describe different types of heart failure can also be confusing. Generally the term ‘heart failure’ is used to describe the symptomatic syndrome (graded according to the

Table 23.1
Definition of heart failure

Heart failure is a clinical syndrome in which patients have the following features:

u Symptoms typical of heart failure

 

(breathlessness at rest or on exercise, fatigue, tiredness, ankle swelling)

and

u Signs typical of heart failure

 

(tachycardia, tachypnoea, pulmonary rales, pleural effusion, raised jugular venous pressure, peripheral oedema, hepatomegaly)

and

u Objective evidence of a structural or functional abnormality of the heart at rest

 

(cardiomegaly, third heart sound, cardiac murmurs, abnormality on the echocardiogram, raised natriuretic peptide concentration)

Heart failure is a clinical syndrome in which patients have the following features:

u Symptoms typical of heart failure

 

(breathlessness at rest or on exercise, fatigue, tiredness, ankle swelling)

and

u Signs typical of heart failure

 

(tachycardia, tachypnoea, pulmonary rales, pleural effusion, raised jugular venous pressure, peripheral oedema, hepatomegaly)

and

u Objective evidence of a structural or functional abnormality of the heart at rest

 

(cardiomegaly, third heart sound, cardiac murmurs, abnormality on the echocardiogram, raised natriuretic peptide concentration)

New York Heart Association (NYHA) functional classification), although a patient can be rendered asymptomatic with treatment (graphic Table 23.2). In our view, a patient who has never exhibited the typical signs or symptoms of heart failure is better described as having asymptomatic left ventricular systolic dysfunction (or whatever the underlying cardiac abnormality is). Recently, however, the American College of Cardiology and American Heart Association have adopted a classification of heart failure that includes asymptomatic patients (graphic Table 23.2) [2]. Patients who have had heart failure for some time are often said to have ‘chronic heart failure’. If chronic heart failure deteriorates the patient may be described as ‘decompensated’ and this may happen suddenly, i.e. ‘acutely’, usually leading to hospital admission, an event of considerable prognostic importance [3]. New (‘de novo’) heart failure may also present acutely, for example as a consequence of acute myocardial infarction (or in a subacute or acute on chronic fashion, for example in a patient who has had asymptomatic cardiac dysfunction for an often indeterminate period) and may resolve (the patient may become 'compensated') or persist [1, 4]. ‘Congestive heart failure’ is a term still used commonly in the USA and may describe acute or chronic heart failure with evidence of congestion, i.e. sodium and water retention. Congestion, though not some symptoms of heart failure (e.g. fatigue), may resolve with diuretic treatment. Many or all of these terms may be accurately applied to the same patient at different times, depending on what stage of their illness they are in.

Table 23.2
Classification of heart failure by symptoms relating to structural capacity (NYHA) or by structural abnormality (ACC/AHA)
NYHA functional classification ACC/AHA stages of heart failure
Severity based on symptoms and physical activity Stage of heart failure based on structure and damage to heart muscle

Class I No limitation of physical activity. Ordinary physical activity doses not cause undue fatigue, palpitation, or dyspnoea.

Stage A At high risk for developing heart failure. No identified structural or functional abnormality; no signs or symptoms.

Class II Slight limitation of physical activity. Comfortable at rest, but ordinary physical activity results in fatigue, palpitation, or dyspnoea.

Stage B Developed structural heart disease that is strongly associated with the development of heart failure, but without signs or symptoms.

Class III Marked limitation of physical activity. Comfortable at rest, but less than ordinary activity results in fatigue, palpitation, or dyspnoea.

Stage C Symptomatic heart failure associated with underlying structural heart disease

Class IV Unable to carry on any physical activity without discomfort. Symptoms at rest. If any physical activity is undertaken, discomfort is increased.

Stage D Advanced structural heart disease and marked symptoms of heart failure at rest despite maximal medical therapy.

NYHA functional classification ACC/AHA stages of heart failure
Severity based on symptoms and physical activity Stage of heart failure based on structure and damage to heart muscle

Class I No limitation of physical activity. Ordinary physical activity doses not cause undue fatigue, palpitation, or dyspnoea.

Stage A At high risk for developing heart failure. No identified structural or functional abnormality; no signs or symptoms.

Class II Slight limitation of physical activity. Comfortable at rest, but ordinary physical activity results in fatigue, palpitation, or dyspnoea.

Stage B Developed structural heart disease that is strongly associated with the development of heart failure, but without signs or symptoms.

Class III Marked limitation of physical activity. Comfortable at rest, but less than ordinary activity results in fatigue, palpitation, or dyspnoea.

Stage C Symptomatic heart failure associated with underlying structural heart disease

Class IV Unable to carry on any physical activity without discomfort. Symptoms at rest. If any physical activity is undertaken, discomfort is increased.

Stage D Advanced structural heart disease and marked symptoms of heart failure at rest despite maximal medical therapy.

ACC, American College of cardiology; AHA, American Heart Association; NYHA, New York Heart Association.

Adapted with permission from Dickstein K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442.

The epidemiology of symptomatic heart failure in developed countries is well understood, especially in Europe (graphic Fig. 23.1) [5–14]. Approximately 2% of the adult population has heart failure, although the syndrome mainly afflicts the elderly, affecting 6–10% of people over the age of 65 years [5–15]. In Europe and North America, the lifetime risk of developing heart failure is approximately one in five for a 40-year-old [16, 17]. The age-adjusted incidence of heart failure appears to have remained stable or even decreased over the past 20 years [1820]. Prevalence is thought to be increasing, partly because survival is increasing [20, 21]. Approximately two in 1000 of the adult population are discharged from hospital with heart failure each year and heart failure accounts for about 5% of all medical and geriatric admissions and is the single most common cause of such admissions in those >65 years [22–31]. Age at admission (and at death) seems to be increasing, suggesting that preventive treatments, such as antihypertensives, and secondary prevention after myocardial infarction are delaying the development of heart failure [22–31]. Hospital discharges include patients developing heart failure suddenly, de novo, as a consequence of another cardiac event (usually myocardial infarction); patients presenting for the first time with decompensation of previously unrecognized cardiac dysfunction; and patients with established, chronic heart failure who have suffered worsening sufficiently severe to lead to hospital admission (though it is recognized that the ‘threshold’ for admission to hospital may vary substantially between countries). Some of these admissions are unavoidable, reflecting the progressive natural history of heart failure whereas others may be avoidable (e.g. as a result of non-adherence to treatment, failure of prompt recognition, and treatment of early decompensation) [32]. After years of steady increase, age-adjusted rates of admission for heart failure seem to have reached a plateau, or even decreased, in Europe and North America (graphic Fig. 23.2) though absolute numbers of admissions continue to increase and heart failure is still an enormous burden on health services and a cost to society, accounting for approximately 2% of all healthcare spending [22–31, 33]. Hospital admissions account for the main part of this expenditure, typically about 70%. Even in primary care, heart failure accounts for more consultations than angina (graphic Fig. 23.3), reflecting the limiting symptoms and reduction in well-being experienced by patients with heart failure [34]. Indeed, quality of life has, consistently, been shown to be reduced more by heart failure than by other chronic illnesses (graphic Fig. 23.4) [35].

 Prevalence of heart failure in
cross-sectional population echocardiographic studies: proportion of subjects
with preserved left ventricular systolic function. Adapted with permission
from McMurray JJ, Pfeffer MA. Heart failure. Lancet 2005; 365: 1877–89.
Figure 23.1

Prevalence of heart failure in cross-sectional population echocardiographic studies: proportion of subjects with preserved left ventricular systolic function. Adapted with permission from McMurray JJ, Pfeffer MA. Heart failure. Lancet 2005; 365: 1877–89.

 Trends in hospital admissions for heart
failure demonstrating recent plateau or decline. (A) Age-adjusted discharge
rate for heart failure in men and women. Reproduced with permission from
Mosterd A, Reitsma JB, and Grobbee DE. Angiotensin converting enzyme
inhibition and hospitalisation rates for heart failure in the Netherlands,
1980 to 1999: the end of an epidemic? Heart 2002; 87: 75–6.
(B) Age-adjusted trends in discharges for a first hospitalization for heart
failure according to sex. Reproduced with permission from Jhund PS,
Macintyre K, Simpson CR, et al. Long-term trends in first
hospitalization for heart failure and subsequent survival between 1986 and
2003: a population study of 5.1 million people. Circulation 2009; 119: 515–23. (C) Trends of age-adjusted hospitalization rates for
heart failure (per 100,000) among patients with heart failure as the first
listed or additional (second to seventh) diagnosis for men and women. Blue
squares, men first; dotted blue squares, men second; red triangles, women
first; dotted red triangles, women second. National Hospital Discharge
Survey 1979–2004. Adapted with permission from Fang J, Mensah GA, Croft JB, et al. Heart failure-related hospitalization in the U.S., 1979 to
2004.J Am Coll Cardiol 2008; 52: 428–34. (D) Age-adjusted
annual incidence of first-ever hospitalization for heart failure as the
principal diagnosis. Reproduced with permission from Schaufelberger M. et
al. Decreasing one-year mortality and hospitalization rates for heart
failure in Sweden. Eur Heart J 2004; 25: 300–7.
Figure 23.2

Trends in hospital admissions for heart failure demonstrating recent plateau or decline. (A) Age-adjusted discharge rate for heart failure in men and women. Reproduced with permission from Mosterd A, Reitsma JB, and Grobbee DE. Angiotensin converting enzyme inhibition and hospitalisation rates for heart failure in the Netherlands, 1980 to 1999: the end of an epidemic? Heart 2002; 87: 75–6. (B) Age-adjusted trends in discharges for a first hospitalization for heart failure according to sex. Reproduced with permission from Jhund PS, Macintyre K, Simpson CR, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people. Circulation 2009; 119: 515–23. (C) Trends of age-adjusted hospitalization rates for heart failure (per 100,000) among patients with heart failure as the first listed or additional (second to seventh) diagnosis for men and women. Blue squares, men first; dotted blue squares, men second; red triangles, women first; dotted red triangles, women second. National Hospital Discharge Survey 1979–2004. Adapted with permission from Fang J, Mensah GA, Croft JB, et al. Heart failure-related hospitalization in the U.S., 1979 to 2004.J Am Coll Cardiol 2008; 52: 428–34. (D) Age-adjusted annual incidence of first-ever hospitalization for heart failure as the principal diagnosis. Reproduced with permission from Schaufelberger M. et al. Decreasing one-year mortality and hospitalization rates for heart failure in Sweden. Eur Heart J 2004; 25: 300–7.

 Age-stratified primary-care
consultation rates per 1000 population for heart failure, angina and
hypertension in men. Reproduced from Murphy NF, Simpson CR, McAlister FA, et al. National survey of the prevalence, incidence, primary care
burden, and treatment of heart failure in Scotland. Heart 2004; 90: 1129–36, with permission from BMJ Publishing Group Ltd.
Figure 23.3

Age-stratified primary-care consultation rates per 1000 population for heart failure, angina and hypertension in men. Reproduced from Murphy NF, Simpson CR, McAlister FA, et al. National survey of the prevalence, incidence, primary care burden, and treatment of heart failure in Scotland. Heart 2004; 90: 1129–36, with permission from BMJ Publishing Group Ltd.

 Quality of life in patients with
congestive heart failure compared to other chronic illnesses and the normal
population. The eight scales of the SF-36 short-form health survey
instrument are physical functioning (PF), role limitations due to physical
limitations (RP), bodily pain (BP), general health perceptions (GH),
vitality (VT), social functioning (SF), role limitations caused by emotional
problems (RE), and mental health (MH). A lower score equates to worse
quality of life. Reproduced with permission from Juenger J, Schellberg D,
Kraemer S, et al. Health related quality of life in patients with
congestive heart failure: comparison with other chronic diseases and
relation to functional variables. Heart 2002, 87: 235–41.
Figure 23.4

Quality of life in patients with congestive heart failure compared to other chronic illnesses and the normal population. The eight scales of the SF-36 short-form health survey instrument are physical functioning (PF), role limitations due to physical limitations (RP), bodily pain (BP), general health perceptions (GH), vitality (VT), social functioning (SF), role limitations caused by emotional problems (RE), and mental health (MH). A lower score equates to worse quality of life. Reproduced with permission from Juenger J, Schellberg D, Kraemer S, et al. Health related quality of life in patients with congestive heart failure: comparison with other chronic diseases and relation to functional variables. Heart 2002, 87: 235–41.

Heart failure is deadly as well as disabling. Community-based surveys indicate that 30–40% of patients die within 1 year of diagnosis and 60–70% die within 5 years, mainly from worsening heart failure or suddenly (probably because of a ventricular arrhythmia) [15, 17, 19, 36, 37]. Thus an adult living to age 40 has a one in five risk of developing heart failure and, once apparent, a one in three chance of dying within a year of diagnosis. Mortality is even higher in patients requiring hospital admission, exceeding that of most cancers (graphic Fig. 23.5), though a number of recent studies indicate that prognosis may be improving (graphic Fig. 23.6) [23–31, 38–43].

 Five-year survival following a first
admission to any Scottish hospital in 1991 for heart failure, myocardial
infarction, and the four most common sites of cancer specific to men and
women. Reproduced with permission from Stewart S, MacIntyre K, Hole DJ. More
'malignant' than cancer? Five-year survival following a first admission for
heart failure. Eur J Heart Fail 2001; 3: 315–22.
Figure 23.5

Five-year survival following a first admission to any Scottish hospital in 1991 for heart failure, myocardial infarction, and the four most common sites of cancer specific to men and women. Reproduced with permission from Stewart S, MacIntyre K, Hole DJ. More 'malignant' than cancer? Five-year survival following a first admission for heart failure. Eur J Heart Fail 2001; 3: 315–22.

 Evidence of improving survival from
heart failure in the general population. (A) Hazard ratio and 95% confidence
intervals for all-cause mortality in patients having a first admission for
heart failure, according to year of admission (adjusted for age, gender,
comorbidity, and social deprivation). Hazard ratio for first year of study
(1993/1994) set at 1. Reproduced with permission from Blackledge HM,
Tomlinson J, and Squire IB. Prognosis for patients newly admitted to
hospital with heart failure: survival trends in 12,220 index admissions in
Leicestershire 1993–2001. Heart 2003; 89: 615–20. (B) Adjusted
30-day mortality according to sex and year of admission. Error bars
represent 95% CIs. Reproduced with permission from Jhund PS, Macintyre K,
Simpson CR, et al. Long-term trends in first hospitalization for
heart failure and subsequent survival between 1986 and 2003: a population
study of 5.1 million people. Circulation 2009; 119: 515–23.
Odds ratio for first year of period of study (1986) set at 1. (C)
Age-adjusted survival after the onset of heart failure in men. Values were
adjusted for age (< 55, 55 to 64, 65 to 74, 75 to 84, and ≥ 85
years). Estimates are shown for men who were 65 to 74 years of age. Similar
trends were observed in women. Reproduced with permission from Levy, D et
al. Long-term trends in the incidence of and survival with heart
failure. NEJM 2002; 347: 1397–1402. (D) Standardized 30-day
and 1-year case fatality rates (%) for women with a first admission to
hospital for heart failure. Reproduced with permission from Schaufelberger
M, et al. Decreasing one-year mortality and hospitalization rates for
heart failure in Sweden. Eur Heart J 2004; 25: 300–7.
Figure 23.6

Evidence of improving survival from heart failure in the general population. (A) Hazard ratio and 95% confidence intervals for all-cause mortality in patients having a first admission for heart failure, according to year of admission (adjusted for age, gender, comorbidity, and social deprivation). Hazard ratio for first year of study (1993/1994) set at 1. Reproduced with permission from Blackledge HM, Tomlinson J, and Squire IB. Prognosis for patients newly admitted to hospital with heart failure: survival trends in 12,220 index admissions in Leicestershire 1993–2001. Heart 2003; 89: 615–20. (B) Adjusted 30-day mortality according to sex and year of admission. Error bars represent 95% CIs. Reproduced with permission from Jhund PS, Macintyre K, Simpson CR, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people. Circulation 2009; 119: 515–23. Odds ratio for first year of period of study (1986) set at 1. (C) Age-adjusted survival after the onset of heart failure in men. Values were adjusted for age (< 55, 55 to 64, 65 to 74, 75 to 84, and ≥ 85 years). Estimates are shown for men who were 65 to 74 years of age. Similar trends were observed in women. Reproduced with permission from Levy, D et al. Long-term trends in the incidence of and survival with heart failure. NEJM 2002; 347: 1397–1402. (D) Standardized 30-day and 1-year case fatality rates (%) for women with a first admission to hospital for heart failure. Reproduced with permission from Schaufelberger M, et al. Decreasing one-year mortality and hospitalization rates for heart failure in Sweden. Eur Heart J 2004; 25: 300–7.

Left ventricular function has also been measured in a number of population-based echocardiographic studies, notably in Europe, enabling estimation of the prevalence of heart failure with a low and preserved EF, as well as the prevalence of asymptomatic left ventricular systolic and diastolic dysfunction (graphic Fig. 23.1) [44–54]. Synthesis of these epidemiological surveys suggests that approximately half of patients with symptomatic heart failure in the community have a low EF (and half have a preserved EF) [44]. The epidemiology of symptomatic heart failure with reduced EF differs from that of heart failure with preserved EF in that patients with preserved EF are, on average, older, are more often women, have more comorbidity, and have a better age-adjusted survival (graphic Fig. 23.7) [31–55]. The causes of heart failure in patients with preserved EF also differ from those with a low EF [31–55].

 Unadjusted Kaplan–Meier survival curves
for participants with heart failure (HF) based on left ventricular function
from The Cardiovascular Health Study. Preserved systolic function (PSF),
Systolic function (SF). Reproduced with permission from Gottdiener JS,
McClelland RL, Marshall R, et al. Outcome of congestive heart failure
in elderly persons: influence of left ventricular systolic function. The
Cardiovascular Health Study. Ann Intern Med 2002; 137:
631–9.
Figure 23.7

Unadjusted Kaplan–Meier survival curves for participants with heart failure (HF) based on left ventricular function from The Cardiovascular Health Study. Preserved systolic function (PSF), Systolic function (SF). Reproduced with permission from Gottdiener JS, McClelland RL, Marshall R, et al. Outcome of congestive heart failure in elderly persons: influence of left ventricular systolic function. The Cardiovascular Health Study. Ann Intern Med 2002; 137: 631–9.

Because about half of patients with a low left ventricular EF are asymptomatic the argument has been made for screening for symptomless cases although no consensus has been reached on this point [56, 57]. Recent studies have reported very disparate prevalence rates of diastolic dysfunction and proportions of symptomatic and asymptomatic individuals and no conclusions can yet be drawn about the epidemiology of asymptomatic diastolic dysfunction [44–52, 54, 58].

As already mentioned, any structural, mechanical, or electrical abnormality of the heart can cause it to fail (graphic Table 23.3). Similarly, heart failure can be caused by ischaemic, metabolic, endocrine, immune, inflammatory, infective, genetic, and neoplastic processes, by failure of the heart to develop properly, and even by pregnancy. The potential causes of heart failure are, therefore, legion, vary geographically, and have changed over time. Rheumatic valvular disease remains a common cause in many developing countries whereas this diagnosis is now uncommon in developed countries; in the latter, degenerative valvular disease in the elderly is now more common [42, 59–63] (see graphic Chapter 17). Valve disease may lead to volume and pressure overload of the heart, as described in more detail below. Endocardial disease is very rare in Europe but much less so in parts of Africa, where it can cause what is referred to as a restrictive cardiomyopathy, as described further in the rest of this section [59–63].

Table 23.3
Aetiology of heart failure

There is no agreed or satisfactory classification for the causes of heart failure with much overlap between potential categories, e.g. dilated cardiomyopathy may be variously regarded as idiopathic, genetic, caused by a remote virus infection or the result of current or previous excessive alcohol consumption.

Myocardial disease

 

coronary artery disease

hypertension

immune/inflammatory

viral myocarditis

Chagas’ disease

metabolic/infiltrative

thiamine deficiency

haemochromatosis

amyloidosis

sarcoidosis

endocrine

hypothyroidism

phaeochromocytoma

thyrotoxicosis

toxic

alcohol

cytotoxics (e.g. trastuzumab)

negatively inotropic drugs (e.g. calcium-channel blockers)

idiopathic

cardiomyopathy (dilated, hypertrophic, restrictive, peri-partum)

Valvular disease

 

mitral stenosis/regurgitation

aortic stenosis/regurgitation

pulmonary stenosis/regurgitation

tricuspid stenosis/regurgitation

Pericardial disease

 

effusion

constriction

Endocardial/endomyocardial disease

 

Löffler endocarditis

endomyocardial fibrosis

Congenital heart disease

 

e.g. atrial or ventricular septal defect

Genetic

 

e.g. familial dilated cardiomyopathy

Arrhythmias (brady- or tachy-)

 

atrial

ventricular

Conduction disorders

 

sinus node dysfunction

second-degree atrioventricular block

third-degree atrioventricular block

High output states

 

anaemia

sepsis

thyrotoxicosis

Paget’s disease

arteriovenous fistula

Volume overload

 

renal failure

iatrogenic

There is no agreed or satisfactory classification for the causes of heart failure with much overlap between potential categories, e.g. dilated cardiomyopathy may be variously regarded as idiopathic, genetic, caused by a remote virus infection or the result of current or previous excessive alcohol consumption.

Myocardial disease

 

coronary artery disease

hypertension

immune/inflammatory

viral myocarditis

Chagas’ disease

metabolic/infiltrative

thiamine deficiency

haemochromatosis

amyloidosis

sarcoidosis

endocrine

hypothyroidism

phaeochromocytoma

thyrotoxicosis

toxic

alcohol

cytotoxics (e.g. trastuzumab)

negatively inotropic drugs (e.g. calcium-channel blockers)

idiopathic

cardiomyopathy (dilated, hypertrophic, restrictive, peri-partum)

Valvular disease

 

mitral stenosis/regurgitation

aortic stenosis/regurgitation

pulmonary stenosis/regurgitation

tricuspid stenosis/regurgitation

Pericardial disease

 

effusion

constriction

Endocardial/endomyocardial disease

 

Löffler endocarditis

endomyocardial fibrosis

Congenital heart disease

 

e.g. atrial or ventricular septal defect

Genetic

 

e.g. familial dilated cardiomyopathy

Arrhythmias (brady- or tachy-)

 

atrial

ventricular

Conduction disorders

 

sinus node dysfunction

second-degree atrioventricular block

third-degree atrioventricular block

High output states

 

anaemia

sepsis

thyrotoxicosis

Paget’s disease

arteriovenous fistula

Volume overload

 

renal failure

iatrogenic

In the developed world, ventricular dysfunction is the commonest underlying problem and is caused, mainly, by myocardial infarction (leading to systolic ventricular dysfunction, i.e. failure of normal contraction and emptying of the heart), hypertension (causing systolic dysfunction, diastolic dysfunction, i.e. failure of normal relaxation and filling of the heart or both) or, often, both infarction and hypertension (see graphic Chapter 17). These causes are becoming more important in some parts of the developing world [63]. Whether persisting systolic dysfunction is caused by coronary artery disease in the absence of infarction is uncertain. The converse, i.e. whether treatment of ischaemia and a state known as ‘hibernation’ (where chronic poor perfusion results in non-contracting but viable myocardium) improves systolic function, is also a question of great current interest, addressed by ongoing studies of coronary ‘revascularization’ [64].

In Europe, North America, and Australasia, hypertension was once the principal cause of heart failure whereas now that position is filled by coronary heart disease (or more exactly, myocardial infarction); this is also increasingly the case in many developing countries [63].

While myocardial infarction is a much more important individual risk factor than hypertension, the population-attributable risk due to hypertension is probably still more important [65, 66]. Both causal factors also interact to augment the risk of heart failure, with concomitant hypertension greatly increasing the risk of failure after myocardial infarction [65, 66]. By the time heart failure presents, prior hypertension may no longer be present. Both of these factors probably result in underestimation of the role of hypertension in causing heart failure. Hypertension is a more common aetiology in women than men.

‘Idiopathic’ dilated cardiomyopathy remains the only other cause of systolic dysfunction commonly encountered, perhaps accounting for 15–20% of cases of heart failure with reduced systolic function. These cases probably have multiple causes and an increasing number of genetic causes are being identified [67]. Prior viral infection is a recognized cause and recent research studies suggest that persisting virus infection may be identified (by endomyocardial biopsy) in a high proportion of patients with dilated cardiomyopathy. Whether this is of prognostic significance or requires therapeutic intervention is currently uncertain [68]. Current or previous excessive alcohol consumption may also cause a dilated cardiomyopathy, as can exposure to other toxins, including chemotherapeutic agents used in cancer treatment such as anthracyclines and trastuzumab [69]. These must always be considered when a patient presents with an unexplained dilated cardiomyopathy. If angiography is not carried out to exclude coronary disease, it may also be wrongly concluded that the patient has an ‘idiopathic’ dilated cardiomyopathy. Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, can also cause systolic dysfunction. Though rarely encountered in Europe it is a relatively common cause of heart failure in South America and is now recognized in Central and North America [59, 61].

It is not clear whether diabetes mellitus should be considered an aetiological factor or a comorbidity in heart failure. Its potential role in causation of systolic and diastolic dysfunction is uncertain [70]. Diabetics have a higher prevalence of heart failure. Diabetes accelerates the development of coronary atherosclerosis and is often associated with hypertension. Whether it directly causes a specific cardiomyopathy is, however, uncertain. Diabetes is also associated with a higher risk of developing heart failure in patients with other causes, e.g. acute myocardial infarction. It is believed that diabetes promotes the development of myocardial fibrosis and diastolic dysfunction. Diabetes is also associated with more autonomic dysfunction and worse renal, pulmonary, and endothelial function, as well as worse functional status and a worse prognosis. Conversely, heart failure increases the risk of developing diabetes [70].

Although it is associated with diabetes, hypertension, and coronary artery disease, obesity also seems to be an independent risk factor for developing heart failure [71].

Atrial fibrillation is both an aetiological factor and comorbidity. It can cause heart failure directly as a consequence of the loss of the atrial contribution to cardiac output and reduced diastolic filling as a result of tachycardia [72, 73]. Patients with underlying structural or functional cardiac disease are more likely to develop failure as a consequence of these effects with the onset of atrial fibrillation. There is, however, a growing belief that atrial fibrillation can cause a dilated cardiomyopathy the exact mechanism of which is uncertain, though persistent tachycardia may play a role (i.e. atrial fibrillation may cause a ‘rate-related cardiomyopathy’) [74]. Heart failure also increases the risk of developing atrial fibrillation and this risk increases with the severity of heart failure. Consequently, when a patient presents with left ventricular dilatation, systolic dysfunction, and atrial fibrillation it can be difficult to determine which came first.

It is important to appreciate that heart failure does not occur in isolation. It is caused by an underlying cardiac defect in, usually, elderly individuals frequently treated for other medical problems with multiple medications. Consequently, the patient with heart failure often has comorbidity related to the underlying cardiac problem or its cause (e.g. angina, hypertension, diabetes, smoking-related lung disease) and age (e.g. osteoarthritis), as well as a consequence of heart failure (e.g. arrhythmias) and its treatment (e.g. gout from diuretics) [75]. Some common comorbidities have multiple causes (e.g. renal dysfunction, see graphic Cardiorenal syndrome, p.842), whereas others are not fully explained (e.g. anaemia, depression, disorders of breathing, and cachexia) [75–80]. The existence of multiple comorbidities creates the potential for drug intolerance (e.g. angiotensin-converting enzyme (ACE) inhibitor and renal dysfunction), drug interactions (e.g. non-steroidal anti-inflammatory drugs (NSAIDs) and ACE inhibitors), worsening of heart failure as a specific adverse effect (e.g. thiazolidinediones), and makes the management of heart failure very complex [1, 70, 81–83]. This is especially true of renal dysfunction, the importance of which is increasingly recognized by a growing use of the term ‘cardiorenal syndrome’ [84, 85] to describe concurrent heart and renal failure.

This syndrome arises from multiple interactions between the age-related decline in glomerular filtration, the effects of treatment for heart failure (diuretics, ACE inhibitors, angiotensin receptor blockers (ARBs), and aldosterone antagonists) and other conditions (e.g. NSAIDs for arthritis), comorbidity (e.g. hypertension, diabetes, atherosclerosis), reduced renal blood flow and the actions on the kidneys of the array of neurohumoral pathways activated in heart failure [84, 85]. The prevalence of severe renal dysfunction in heart failure is often underestimated because serum creatinine concentration may not be greatly elevated because of the reduction in skeletal muscle mass in advanced heart failure. Renal dysfunction may contribute to the high prevalence of anaemia in patients with heart failure. The blood concentrations of creatinine, urea (blood urea nitrogen), and estimated glomerular filtration rate are powerful independent predictors of prognosis and deterioration in renal function during an episode of worsening heart failure (e.g. an increase in creatinine concentration of ≥0.3mg/dL or 27µmol/L) is associated with higher morbidity and mortality [86].

Anaemia is another important comorbidity and can be both the cause and, it seems, consequence of heart failure [76, 87–89]. Anaemia is common (especially in more severe heart failure) and is associated with worse symptoms, increased risk of hospital admission, and reduced survival. The causes are unknown but may include haemodilution, renal dysfunction, poor nutrition, inflammation, blood loss related to medication, and reduced production of (or response to) erythropoietin.

Patients with heart failure often exhibit some degree of muscle wasting which is restricted to the lower limbs (disuse atrophy) [77, 90, 91]. This loss of tissue may become more extensive in some patients, usually when their heart failure is more advanced, and may affect all body compartments (muscle, fat, and bone tissue). This general wasting is referred to as cardiac cachexia. The underlying metabolic causes are complex and differ from patient to patient. Three important contributors are, probably, dietary deficiency (exacerbated by anorexia), loss of nutrients through the urinary or digestive tracts (malabsorption), and metabolic dysfunction (including an imbalance of anabolic and catabolic factors and inflammation). The development of cachexia is an ominous sign [77, 90].

It is important to appreciate that it is comorbidity, along with the key pathophysiological processes in heart failure, i.e. left ventricular remodelling and activation of systemic pathways (and age), that are the principal determinants of prognosis.

We have only limited knowledge of the pathophysiology of heart failure, and that of left ventricular systolic dysfunction is best understood. Much of our understanding comes from studies of myocardial infarction (graphic Fig. 23.8) [92, 93]. Following an initial injury to the myocytes and cytoskeleton, heart failure may develop immediately, in the short term (over days or weeks), over a longer time period (months to years), or not at all. The factors leading to the development of heart failure acutely after myocardial infarction (e.g. size of infarction, concomitant hypertension [66], etc.), the important pathophysiological mechanisms operating (e.g. cardiac remodelling), and the time-course of this complication of infarction are fairly well established. On the other hand, the natural history of asymptomatic left ventricular systolic dysfunction is less well understood, as are the pathophysiological mechanisms causing progression from the asymptomatic to the symptomatic state. Once symptomatic heart failure has developed we believe that the pathophysiology is again better understood, at least in patients with systolic dysfunction. One thing is certain: the syndrome is characterized by progressive worsening of the patient’s symptoms, of cardiac function, and of the function of other tissues (e.g. skeletal muscle, bone marrow) and organs (e.g. the kidneys).

 Gross pathological appearance of (A) an
anteroseptal myocardial infarction, with wall thinning and endocardial
fibrosis, in a patient with a background of left ventricular hypertrophy due
to hypertension, and (B) a heart in idiopathic dilated cardiomyopathy
characterized by four chamber enlargement.
Figure 23.8

Gross pathological appearance of (A) an anteroseptal myocardial infarction, with wall thinning and endocardial fibrosis, in a patient with a background of left ventricular hypertrophy due to hypertension, and (B) a heart in idiopathic dilated cardiomyopathy characterized by four chamber enlargement.

The progression of left ventricular systolic dysfunction (and the heart failure syndrome), because of ‘remodelling’ of the left (and right) ventricle (as a result of the loss of myocytes and maladaptive changes in the surviving myocytes and extracellular matrix), probably occurs in two main ways [92–97]. One is because of intercurrent cardiac events (e.g. myocardial infarction [98]) and the other is as a consequence of the local processes (e.g. the autocrine pathway and molecular adaptations, including, perhaps, apoptosis) and systemic processes (e.g. neurohumoral pathways) that are activated as a result of reduced systolic function (graphic Fig. 23.9) [92–97]. These systemic processes, which are discussed in detail in later sections, also have detrimental effects on the functioning of the lungs, blood vessels, kidneys, bone marrow, muscles, and probably other organs (e.g. the liver), and contribute to a pathophysiological vicious cycle (graphic Fig. 23.10). The molecular, structural, and functional changes in the heart and these systemic processes, coupled with electrolyte imbalances, result in electrical as well as mechanical dysfunction of the heart. In addition, cardiac metabolism is altered in the failing heart and some believe that a state of relative energy starvation exists [93, 99].

 A partial wiring diagram of biological
circuits for heart failure. Impaired pump function after myocyte death from
myocardial infarction or abnormal loading conditions such as found in
hypertension (white) activate a biomechanical stress-dependent signalling
cascade (purple). The responsible targets of altered signal transduction
cascades in heart failure include transcription factors, coactivators and
co-repressors for cardiac gene expression (green) as well as the effector
mechanisms like calcium cycling, metabolism, growth and apoptosis (yellow)
that culminate in ventricular dysfunction (orange) and secondary
neurohumoral responses (grey) such as adrenergic drive and intramyocardial
growth factors. Inherited mutations for cardiomyopathy (blue) affect
proteins at many of these points and are thought to engage a similar cascade
of events to elicit the full myopathic phenotype. Cell-based therapies
(red), although often envisioned working chiefly or wholly by replacing dead
myocytes, probably improve ventricular performance through a combination of
mechanisms, including angiogenesis, paracrine signals for myocyte
protection, and conceivably augmenting host self-repair. Reproduced with
permission from Benjamin, IJ, Schneider, MD. Learning from failure:
congestive heart failure in the postgenomic age. J Clin Invest 2005; 115: 495–9.
Figure 23.9

A partial wiring diagram of biological circuits for heart failure. Impaired pump function after myocyte death from myocardial infarction or abnormal loading conditions such as found in hypertension (white) activate a biomechanical stress-dependent signalling cascade (purple). The responsible targets of altered signal transduction cascades in heart failure include transcription factors, coactivators and co-repressors for cardiac gene expression (green) as well as the effector mechanisms like calcium cycling, metabolism, growth and apoptosis (yellow) that culminate in ventricular dysfunction (orange) and secondary neurohumoral responses (grey) such as adrenergic drive and intramyocardial growth factors. Inherited mutations for cardiomyopathy (blue) affect proteins at many of these points and are thought to engage a similar cascade of events to elicit the full myopathic phenotype. Cell-based therapies (red), although often envisioned working chiefly or wholly by replacing dead myocytes, probably improve ventricular performance through a combination of mechanisms, including angiogenesis, paracrine signals for myocyte protection, and conceivably augmenting host self-repair. Reproduced with permission from Benjamin, IJ, Schneider, MD. Learning from failure: congestive heart failure in the postgenomic age. J Clin Invest 2005; 115: 495–9.

 Pathophysiology of heart failure as a
result of left-ventricular systolic dysfunction. Damage to the myocytes and
extracellular matrix leads to changes in the size, shape, and function of
the left ventricle and heart more generally (‘remodelling’). These changes,
in turn, lead to electrical instability, systemic processes resulting in
many effects on other organs and tissues, and further damage to the heart.
These vicious cycles, along with intercurrent events, such as myocardial
infarction, are believed to cause progressive worsening of the heart-failure
syndrome over time. Adapted with permission from McMurray JJ, Pfeffer MA.
Heart failure. Lancet 2005; 365: 1877–89.
Figure 23.10

Pathophysiology of heart failure as a result of left-ventricular systolic dysfunction. Damage to the myocytes and extracellular matrix leads to changes in the size, shape, and function of the left ventricle and heart more generally (‘remodelling’). These changes, in turn, lead to electrical instability, systemic processes resulting in many effects on other organs and tissues, and further damage to the heart. These vicious cycles, along with intercurrent events, such as myocardial infarction, are believed to cause progressive worsening of the heart-failure syndrome over time. Adapted with permission from McMurray JJ, Pfeffer MA. Heart failure. Lancet 2005; 365: 1877–89.

It is important to remember that atrial function, synchronized contraction of the left ventricle, and normal interaction between the right and left ventricles are also important in preserving stroke volume [93, 100, 101]. Loss of these key mechanical interactions is often secondary to disturbances of conduction arising as a consequence of cardiac fibrosis.

The term ‘cardiomyopathy’ is used to define a myocardial disorder in which the heart muscle is structurally and functionally abnormal, in the absence of coronary artery disease (hence the term ‘ischaemic cardiomyopathy’ should be avoided), hypertension, valvular disease, and congenital heart disease sufficient to cause the observed myocardial abnormality [67, 102]. Myocyte necrosis whether caused by infarction or by other injury has a common consequence. Whether diffuse or focal, myocyte loss leads to replacement fibrosis, hypertrophy of the remaining myocytes and dilatation of the affected cardiac chamber [67, 93, 102–104]. How these molecular and cellular changes affect the left ventricle macroscopically (by causing remodelling) is discussed in more detail in later sections. The resulting anatomical and pathophysiological picture is, however, identified by clinicians as a dilated cardiomyopathy. Certain inherited cardiomyopathies result in a similar phenotype. Poor contraction and emptying of the ventricle are usually referred to as systolic dysfunction. Other cardiomyopathic phenotypes occur (graphic Fig. 23.11).

 Summary of proposed classification
system. ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated
cardiomyopathy; HCM, hypertrophic cardiomyopathy; RCM restrictive
cardiomyopathy. Reproduced from Elliott P, Andersson B, Arbustini E, et
al. Classification of the cardiomyopathies: a position statement from
the European Society Of Cardiology Working Group on Myocardial and
Pericardial Diseases. Eur Heart J 2008; 29: 270–6, with
permission from Oxford University Press.
Figure 23.11

Summary of proposed classification system. ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; RCM restrictive cardiomyopathy. Reproduced from Elliott P, Andersson B, Arbustini E, et al. Classification of the cardiomyopathies: a position statement from the European Society Of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2008; 29: 270–6, with permission from Oxford University Press.

Systolic and diastolic dysfunction are terms used to describe whether the principal abnormality of the myocardium is an inability of the ventricle to contract and expel blood or to relax and fill normally, respectively (though in reality these two abnormalities frequently coexist). Systolic dysfunction

is the result of reduced shortening of sarcomeres, which is a consequence of a global or regional reduction of contractility or greatly increased impedance to left ventricular ejection. An increase in preload can provide short-term compensation (via the Frank–Starling mechanism, see p.847) for a reduction in contractility or increases in impedance. However, long-term compensation usually involves myocardial hypertrophy, which is the result of laying down new sarcomeres that increase the width (concentric) or the length (eccentric) of myocytes [93, 105–110]. Remodelling also contributes to reduced sarcomere shortening. All these factors causing reduced fibre shortening and eventually lead to a decrease in the left ventricular ejection fraction (LVEF). Hence, end-systolic volume increases.

Rapid filling during systole is assisted by active, energy-dependent, relaxation of the ventricle [93, 105–110]. Primary myocardial diseases may affect this process. Ventricular relaxation also depends on myocardial mass, collagen content, and extrinsic forces (e.g. the pericardium) [93, 105–110]. The hallmark of diastolic dysfunction is elevation in left ventricular end-diastolic pressure or left arterial pressure in the absence of systolic dysfunction [93, 105, 110].

Infiltrative processes in the myocardium may cause a restrictive cardiomyopathy (graphic Fig. 23.12) [111, 112] (see graphic Chapter 17). The principal consequence is impaired ventricular filling with normal or decreased diastolic volume of either or both ventricles, akin to the diastolic dysfunction described earlier. Systolic function is typically maintained during the early stages of the disease and wall thickness is normal or increased. The main problem is that increased stiffness of the myocardium causes pressure within the ventricle to rise precipitously with only small increases in volume. Restrictive cardiomyopathy may affect either or both ventricles. Elevated jugular venous pressure, peripheral oedema, and ascites are often prominent features. Pericardial constriction has similar pathophysiological effects and causes a similar clinical picture but can be hard to diagnose. Clinical suspicion may be raised by a history of prior tuberculosis, radiotherapy, cardiothoracic surgery, etc. Special investigations, including cardiac computed tomography (CT) scanning and simultaneous right and left heart pressure measurements at catheterization are often necessary [113, 114].

 Transthoracic four-chamber
echocardiogram of patient with cardiac amyloidosis.
Figure 23.12

Transthoracic four-chamber echocardiogram of patient with cardiac amyloidosis.

Arterial hypertension and aortic stenosis cause a sustained increase in systolic wall stress during left ventricular ejection leading to concentric hypertrophy of the left ventricle because of myocyte hypertrophy and extracellular matrix overgrowth [115].

Conversely, mitral and aortic regurgitation result in an increased volume load on the ventricle. The resultant ventricular remodelling is characterized by dilatation, representing, at least in part, lengthening of the cardiac myocytes [116, 117].

These terms are not useful and are reminiscent of the now discarded terminology of ‘backwards’ and ‘forwards’ heart failure. The term right heart failure is often used to describe patients in whom there are prominent signs of peripheral ‘congestion’, e.g. a raised jugular venous pressure, hepatomegaly, and peripheral oedema (graphic Fig. 23.13), on the basis that these findings reflect right ventricular failure; in fact all of these signs are also found in patients with predominantly left ventricular involvement. The description pulmonary heart disease is used to depict patients who do have isolated right heart failure as a result of primary lung disease and has generally replaced the term ‘cor pulmonale’.

 (A) A raised jugular venous pressure
( JVP) reflects an elevation in right atrial pressure as occurs in heart
failure. However this can also be seen in pericardial disease, tricuspid
stenosis, superior vena cava obstruction, reduced compliance of the right
ventricle, and hypervolaemia. (B) Pitting pedal oedema as seen in a patient
with heart failure. This can also be seen in hypoalbuminaemia, nephrotic
syndrome, chronic venous insufficiency and myxoedema.
Figure 23.13

(A) A raised jugular venous pressure ( JVP) reflects an elevation in right atrial pressure as occurs in heart failure. However this can also be seen in pericardial disease, tricuspid stenosis, superior vena cava obstruction, reduced compliance of the right ventricle, and hypervolaemia. (B) Pitting pedal oedema as seen in a patient with heart failure. This can also be seen in hypoalbuminaemia, nephrotic syndrome, chronic venous insufficiency and myxoedema.

A more useful pathophysiological classification is to distinguish between high- and low-output heart failure, although the former is uncommonly encountered in Western clinical practice. Cardiac index is normally 2.2–3.5L/min/m2. Low-output cardiac failure implies that cardiac output fails to rise adequately during exercise or that it is inadequate even at rest. This prototypical form of heart failure is seen in cases of heart failure as a result of left ventricular systolic dysfunction. High-output cardiac failure, on the other hand, implies that although the pumping action of the heart is intact other factors make it difficult for the heart to deliver oxygen commensurate with the needs of the metabolizing tissues, either because of increased tissue demand (e.g. as a result of anaemia, hyperthyroidism, or pregnancy) or reduced oxygen-carrying content of the blood (e.g. anaemia). This type of heart failure can also be caused by arteriovenous shunting.

The Frank–Starling law describes an intrinsic mechanism that helps maintain stroke volume when the heart is acutely injured and may also play a compensatory role in chronic heart failure, though this is less certain [118]. Together with neurohumoral activation (an extrinsic mechanism), the Frank–Starling law is an adaptive phenomenon that comes into play within minutes of cardiac injury. The resultant acute fall in the volume of blood ejected by the ventricle (the stroke volume) leads to a rise in left ventricular end-diastolic volume (and pressure). Through the Frank–Starling mechanism this rise in preload increases the force of contraction, thereby helping restore stroke volume. The mechanism whereby increased stretch of the myocyte causes increased force of contraction is also referred to as the law of heterometric autoregulation. In the chronic setting, sodium retention, water retention, and venoconstriction may represent continuing attempts by the body to use the Frank–Starling mechanism by increasing left ventricular filling pressure and preload.

These adaptations may, however, lead to abnormally high pulmonary capillary and artery pressures, probably contributing to the shortness of breath experienced by patients with heart failure. Furthermore, arterial constriction and stiffening (as a result of sodium and water retention in the vascular wall) increase afterload and will eventually cause the injured left ventricle to fail further because it is especially sensitive to increases in afterload (law of homeometric regulation).

The heart attempts to compensate for increased preload (e.g. because of increased extracellular fluid volume and venous return) and afterload (e.g. because of systemic arterial constriction) in several ways. One is the development of ventricular hypertrophy in an attempt to maintain systolic wall stress within normal limits [92–97, 119, 120]. Pressure overload tends to lead to concentric hypertrophy, whereas volume overload tends to lead to ventricular dilatation [92–97, 119, 120]. Both entities are distinct at the molecular level. Pressure overload is associated with parallel replication of myofibrils and thickening of individual myocytes. Volume overload, on the other hand, leads to replication of sarcomeres in series and elongation of myocytes. The two types of haemodynamic overload are presumed to activate distinct signalling pathways.

Compensated remodelling results in relatively little change in ventricular dimensions, shape, function, and wall thickness. However, these compensatory adaptations only seem to be capable of maintaining pump function over a limited period of time and a ventricle subjected to an elevated load for a prolonged period will ultimately fail. Ventricular dilatation may lead to stretching of the mitral valve ring and cause valvular incompetence (graphic Fig. 23.14). This may further increase the load on the failing ventricle; this is an example of another ‘vicious cycle’ that develops and which may drive progression of heart failure.

 Colour-flow Doppler study of a patient
with mitral regurgitation as a result of left ventricular dilatation seen in
the apical four-chamber view.
Figure 23.14

Colour-flow Doppler study of a patient with mitral regurgitation as a result of left ventricular dilatation seen in the apical four-chamber view.

An initial stress-induced increase in sarcomere length yields an optimal overlap between myofilaments [92–97, 119–121]. Severe haemodynamic overload eventually yields depression of myocardial contractility. In patients with mild disease, this depression is manifested by reduced velocity of shortening of the myocardium or by a reduction in the rate of force development during isometric contraction. More severe stages are accompanied by a decline in isometric force development and shortening as well. EF and cardiac output during exercise decline [92-97, 119–121].

Our understanding of the molecular mechanisms behind these changes is still limited and can only be touched upon briefly (graphic Fig. 23.9). They comprise myocyte loss by necrosis and apoptosis, alterations in excitation–contraction coupling, and alterations in composition of the extracellular matrix [119–120]. Myocyte loss as a result of necrosis is a well-understood process which is localized after myocardial infarction but more diffuse in patients with dilated cardiomyopathy or myocarditis. Apoptosis, or programmed cell death, on the other hand, results from the induction of a genetic programme that leads to degradation of nuclear DNA (graphic Fig. 23.9) [92–97, 119–122]. Several recent reports have described apoptotic cells in the failing myocardium. It may be relevant that several substances, such as angiotensin II, reactive oxygen species, nitric oxide (NO), and pro-inflammatory cytokines may induce apoptosis experimentally in cardiac myocytes. However, the precise frequency of occurrence and role of apoptosis in the failing myocardium remains unclear [122]. Changes in the extracellular matrix are usually manifested by an increase in collagen content though both degradation and synthesis (and the activity of the enzymes controlling these processes) may be increased [93, 95]. While this change in collagen content may contribute to impaired systolic contraction it may be even more important in reducing ventricular compliance and impaired ventricular filling.

The human body responds to the haemodynamic changes in patients with heart failure in a highly complex way. Many neurohumoral systems appear to be involved to varying degrees and at different stages. It has been suggested that these are initially activated in a manner appropriate to haemorrhage or some other crisis threatening vital organ perfusion [93, 123, 124]. Their sustained activation in heart failure, however, is not only inappropriate but probably detrimental (graphic Fig. 23.10). Moreover, at some stage during the progression of heart failure, haemodynamic abnormalities may cease to be the main trigger of neurohumoral activation. Instead, a number of other self-sustaining pathophysiological vicious cycles may develop.

The predominant effects of the neurohumoral systems activated in heart failure are to cause vasoconstriction, sodium and water retention, and abnormal cell growth. Two exceptions are the natriuretic peptides [93, 125] (secreted mainly by the failing heart, graphic Fig. 23.15) and adrenomedullin (secreted mainly from blood vessels) [126].

 Physiological effects of the
natriuretic peptides in heart failure. Increased secretion of the
natriuretic peptides reduces blood pressure and plasma volume through
coordinated actions in the brain, adrenal gland, kidney, and vasculature.
Urodilatin (URO); neutral endopeptidase (NEP); C-type natriuretic peptide
(CNP); natriuretic peptide receptors A, B and C (NPR-A, NPR-B, and NPR-C
respectively); arginine vasopressin (AVP); atrial and brain natriuretic
peptides (ANP and BNP); glomerular filtration rate (GFR); urinary sodium
excretion (UNaV); urinary volume (UV); blood pressure (BP).
Reproduced with permission from Levin, ER, Gardner, DG, Samson WK.
Mechanisms of disease: Natriuretic peptides. N Engl J Med 1998; 339: 321–8.
Figure 23.15

Physiological effects of the natriuretic peptides in heart failure. Increased secretion of the natriuretic peptides reduces blood pressure and plasma volume through coordinated actions in the brain, adrenal gland, kidney, and vasculature. Urodilatin (URO); neutral endopeptidase (NEP); C-type natriuretic peptide (CNP); natriuretic peptide receptors A, B and C (NPR-A, NPR-B, and NPR-C respectively); arginine vasopressin (AVP); atrial and brain natriuretic peptides (ANP and BNP); glomerular filtration rate (GFR); urinary sodium excretion (UNaV); urinary volume (UV); blood pressure (BP). Reproduced with permission from Levin, ER, Gardner, DG, Samson WK. Mechanisms of disease: Natriuretic peptides. N Engl J Med 1998; 339: 321–8.

The neurohumoral pathways activated in heart failure are thought to be particularly important because they appear to explain how many of the successful pharmacological treatments for heart failure work (and offer the potential for more such therapeutic interventions). Neurohumoral activation seems to be much less marked in patients with heart failure and preserved EF, compared to those with a low EF [127].

It has long been recognized that an increased activity of the sympathetic nervous system and parasympathetic withdrawal are prototypical characteristics of heart failure [93, 128–130]. Elevated levels of plasma norepinephrine (noradrenaline) are a common finding in patients with heart failure. Increased sympathetic nerve traffic and enhanced spill-over of norepinephrine from the synaptic cleft account for this, and are particularly pronounced in the heart, kidney, and skeletal muscle [93, 128–131]. Raised plasma norepinephrine concentrations predict higher mortality rates [130, 131]. Heart failure is also characterized by reductions in myocardial norepinephrine stores and in myocardial beta-receptor density. These again reflect generalized adrenergic activation [93, 128–131].

It is believed that enhanced sympathetic activity initially increases myocardial contractility and heart rate (leading to an increase in cardiac output). Sympathetic activation also promotes renin release, sodium retention, and vasoconstriction thereby increasing preload and activating the Frank–Starling mechanism (see graphic Frank–Starling mechanism, p.847). These responses are capable of maintaining ventricular performance and cardiac output for a limited period of time. In part this may be because the increase in afterload caused by arterial constriction (to which the failing ventricle is particularly sensitive) leads to a further fall in stroke volume. Sympathetic overdrive probably alters myocardial metabolism and catecholamines may also even be directly toxic to cardiomyocytes [93, 130]. Excessive adrenergic activity (and reduced vagal activity) also increases the electrical instability of the heart. As well as having complex and changing effects on myocardial contractility and structure, activation of the sympathetic nervous system leads to redistribution of regional blood flow and even to changes in the structure of the vasculature [129].

The precise cause of sympathetic activation in heart failure is unknown. Reduced stimulation of stretch-activated baroreceptors in the carotid arteries and the aorta from decreased arterial pressure and stroke volume may contribute (in an analogous way to haemorrhage). Another factor suggested is structural and functional abnormalities of afferent receptors. Other neurohumoral systems may also activate the sympathetic nervous system and augment its actions, i.e. many neurohumoral systems act in concert, synergistically reinforcing each other.

Increased activity of the renin–angiotensin–aldosterone system (RAAS) also produces deleterious effects on the cardiovascular system (and other organs and tissues) and contributes to the poor prognosis in heart failure [93, 123, 124, 132–134]. The plasma components of this system are usually increased in patients with heart failure and a low serum sodium concentration is a marker for particularly excessive activation of the RAAS [134]. The increase in renin release is mediated by decreased stretch of the glomerular afferent arteriole and reduced delivery of chloride to the macula densa. Although the sympathetic nervous system also stimulates renin release, the two systems are independently regulated. One puzzle about heart failure is why the sodium and volume overload that characterizes the syndrome does not suppress renin release, as would normally occur.

The increased secretion of renin leads to an augmented production of angiotensin II, which, it is believed, has mostly deleterious effects in heart failure (although its efferent glomerular arteriolar action may help maintain glomerular filtration). Angiotensin II not only induces vasoconstriction, but also salt and water retention directly and via aldosterone. Moreover, it mediates myocardial cell hypertrophy and fibrosis and these effects may contribute to the maladaptive remodelling and progressive loss of myocardial function in heart failure. Angiotensin II may also cause activation of the sympathetic nervous system, prothrombotic actions, and augment the release of arginine vasopressin.

Plasma aldosterone levels are also increased in heart failure, and release of this hormone is influenced by angiotensin and other stimuli such as potassium and corticotropin. Aldosterone is an independent and harmful component of the RAAS [135]. It causes sodium and water retention and, importantly, potassium wastage. Potassium loss, along with autonomic dysfunction and myocardial fibrosis (both of which are also thought to be caused by aldosterone), may increase the risk of ventricular arrhythmias. Aldosterone may also contribute to vascular fibrosis in heart failure.

There is recent interest in the possibility that renin itself, acting through a specific renin/pro-renin receptor, may have detrimental effects in heart failure [136].

Vasopressin (also known as antidiuretic hormone) is a neurohypophysial peptide involved in the regulation of free water reabsorption, body fluid osmolality, blood volume, blood pressure, cell contraction, cell proliferation, and adrenocorticotropin secretion [137, 138]. Vasopressin binds to three different specific G protein-coupled receptors. These are currently classified as V1 (or V1A)-vascular, V2-renal, and V3 (or V1B)-pituitary subtypes. All subtypes have distinct pharmacological profiles and intracellular second messengers. As well as reducing renal water excretion, vasopressin is one of the most powerful vasoconstricting substances known. It also stimulates blood platelet aggregation, coagulation factor release, and cellular proliferation. This profile of action is clearly unattractive in heart failure.

Elevated circulating levels of vasopressin are often, but not invariably, found in patients with chronic heart failure. It appears that vasopressin release from the posterior pituitary in heart failure is largely non-osmotic, although, normally, increased serum osmolality is the major physiological stimulus for its secretion.

A-type (atrial) natriuretic peptide (ANP) and B-type (brain) natriuretic peptide (BNP) are released in response to atrial and ventricular wall stretch and, as their names suggest, serve to maintain sodium homeostasis by enhancing renal sodium and water excretion (graphic Fig. 23.15) [93, 125, 139–142]. These peptides also have haemodynamic effects, dilating arteries and, especially, veins. They also suppress the RAAS and, possibly, the sympathetic nervous system. There is some evidence that natriuretic peptides inhibit arginine vasopressin and endothelin-1 release and the biological actions of these peptides. Consequently, the natriuretic peptides are thought to play an important protective role in heart failure, countering the actions of the other vasoconstricting and anti-natriuretic neurohumoral systems which are activated in heart failure.

Circulating levels of both ANP and BNP are greatly increased in heart failure. This is the consequence of an increased synthesis and release of these hormones. In humans, BNP is mostly secreted from the ventricles in both healthy individuals and patients with heart failure. ANP secretion, on the other hand, is mainly from the atria in healthy individuals but from both the atria and the ventricles in patients with heart failure. Therefore, it appears that BNP is the only natriuretic peptide that is specific to the ventricles. Pro-BNP, the precursor of BNP, is stored in granules in myocytes. Pro-BNP is activated by a protease to form its biologically active form, BNP, and N-terminal (NT)-proBNP.

BNP levels vary according to sex and age in healthy subjects [141]. Female patients display higher plasma concentrations than male patients with heart failure. Advancing age and declining renal function are associated with increases in BNP levels.

C-type natriuretic peptide (CNP), which was originally believed to be of endothelial origin, may also be produced by the failing heart. A D-type (Dendroaspis) natriuretic peptide and a renal specific peptide (urodilatin) has also been described recently, though the origin and actions of each in humans are not yet well defined [142].

As well as having important physiological effects, the various A-type and B-type natriuretic peptides and related fragments can be used to aid the diagnosis of heart failure and to provide prognostic information.

Relaxin is a pregnancy-related hormone with vasodilator activity which has been shown to improve haemodynamic indices in heart failure [145, 146].

These three more recently described neurohumoral factors have vasodilator and inotropic properties, as well as other actions [126, 143, 144]. Apelin is unusual in that blood and tissue concentrations are reduced in heart failure [143]. The pathophysiological role, if any, that these factors play in heart failure is presently uncertain as is any therapeutic potential that might exist. [126, 143, 144].

The principal product of the endothelium, NO, plays a central role in vascular homeostasis. Endothelial dysfunction, which is characterized by reduced production and action of NO, occurs as a result of ageing, as well as in a number of chronic conditions related to heart failure, such as hypercholesterolaemia, atherosclerosis, as well as in heart failure itself [147]. Endothelium-dependent dilatation of coronary and peripheral resistance vessels is blunted in patients with heart failure. This probably contributes to the impaired reactive hyperaemia in various vascular beds, an impairment in tissue perfusion and, perhaps, reduced muscular function [148]. There has even been speculation that NO might be a key regulator of lung function and that exercise-induced dyspnoea may be related to impaired pulmonary vasodilatation resulting from reduced NO production compared to healthy subjects. Endothelial dysfunction in heart failure may be partly the result of increased oxidative stress.

Although lack of NO is associated with the development of endothelial dysfunction, its overproduction by the inducible isoform of NO synthase (iNOS) may also be detrimental [148]. Increased iNOS activity is thought to lead to increased free radical formation and to depression of myocardial activity [93]. Increased iNOS expression may result from the actions of inflammatory cytokines.

The endothelins are another important product of the endothelium and endothelin-1 is one of the most powerful vasoconstrictor peptides known [149]. It is also a mitogen and generally shares the potentially detrimental properties of angiotensin II and vasopressin in heart failure. Plasma concentrations of endothelin-1 are increased in heart failure, probably because of increased secretion, both by blood vessels and the failing myocardium. The importance of this is, however, uncertain because specific antagonists have not improved outcome in heart failure.

Oxygen free radicals have a number of potentially detrimental actions in heart failure [93, 150, 151]. They inactivate NO, depress myocardial contractility, and may induce apoptosis [93, 152]. One source of the superoxide anion radical is NADPH oxidase which is activated by angiotensin II and aldosterone [93, 153]. Xanthine oxidase may be another source of increased free oxygen radical load in heart failure and is normally involved in the last step of purine breakdown which yields uric acid. Hyperuricaemia is a consistent finding in patients with heart failure, may reflect impairment of oxidative metabolism, and is a predictor of worse outcome [152]. Oxidative stress may be part of the generalized inflammatory state that characterizes at least some patients with heart failure. The importance of oxidative stress is, however, uncertain because specific antagonists (oxypurinol and vitamin E) did not improve pathophysiological measures or clinical outcomes in heart failure [155, 156].

Inflammation may also be a factor contributing to the progression of heart failure although its role has not been ‘confirmed’ in the same way as neurohumoral activation, i.e. by the demonstration of improved outcomes with blocking agents [93, 157]. Several cytokines have been studied in detail. Different cell types secrete cytokines for the purpose of altering either their own function (autocrine) or that of adjacent cells (paracrine). Some cytokines also act as circulating hormones (i.e. have an endocrine action). Tumour necrosis factor-α (TNF-α), interleukin-1, and interleukin-6 are thought to be the most important pro-inflammatory cytokines that may be implicated in heart failure progression. The cause of cytokine activation in heart failure remains unclear. Pro-inflammatory cytokines may be secreted by mononuclear cells, hypoxic peripheral tissue, or even by the myocardium itself. Catecholamines may augment myocardial cytokine production, one of several possible links between neurohumoral activity and inflammation. It has also been hypothesized that increased bowel wall oedema may lead to translocation of bacterial endotoxin or lipopolysaccharide from the gut which may cause pro-inflammatory cytokine production from blood monocytes and possibly other tissues [157, 158].

Plasma TNF-α and TNF receptor (TNFR) concentrations are increased in some patients with heart failure, especially in those whose disease is severe, and they are independent predictors of poor prognosis. Although TNF-α has several potentially untoward effects that could contribute to the progression of heart failure, studies of TNF antagonists to date have not shown benefit in this syndrome [159, 160].

It is symptoms and signs that usually alert the patient (and physician) to the presence of a cardiac disorder. However, neither the symptoms nor signs commonly recognized as suggesting the presence of heart failure are specific for this syndrome. Therefore confirmation of heart failure requires objective tests to confirm that the patient’s symptoms and the physical findings are the result of abnormal cardiac function and not of another cause (see graphic Simple investigations, p.854) [1, 2].

Fatigue is a key symptom reported by patients with heart failure. Its origins are not clearly understood but probably include low cardiac output and skeletal muscle abnormalities (see graphic Pathophysiology, p.843). Fatigue is very non-specific and is found in the population at large, as well as in many non-cardiovascular disorders.

Dyspnoea or breathlessness is another cardinal symptom of heart failure. Dyspnoea is usually first manifested on exertion and the level of exertion which causes breathlessness is useful in gauging heart failure severity and monitoring the patient’s progress. Though it is more specific than fatigue, dyspnoea is still caused by many other disorders such as pulmonary disease, obesity, and anaemia, which are common in the elderly population and may coexist with heart failure. Even ageing itself is associated with dyspnoea on exercise. The origin of dyspnoea in heart failure is also probably multifactorial. It may be related to elevated pulmonary pressures, abnormalities in pulmonary compliance, respiratory dysfunction, accentuated respiratory drive, increased airway resistance, abnormal chemical and mechanical muscle reflexes [161–166], and even low haemoglobin. It is notable that there is a poor correlation between dyspnoea and left ventricular function at rest [161–166].

Orthopnoea is defined as dyspnoea which occurs in the recumbent position and is usually relieved by sitting upright or by the addition of pillows. In extreme cases, the patient is unable to lie down and may spend the night in the sitting position. Orthopnoea results from the return of venous blood which has pooled in the lower extremities while the patient is ambulatory. The failing heart may be unable to cope with return of this blood from the legs on adoption of the recumbent position and pulmonary oedema may occur.

Paroxysmal nocturnal dyspnoea is characterized by acute episodes of suffocation usually occurring while recumbent at night. It has the same causes as orthopnoea. Paroxysmal nocturnal dyspnoea may manifest as cough or wheezing, possibly because increased pressure in the bronchial arteries (and resultant increase in their diameter), along with interstitial pulmonary oedema, leads to increased airways resistance. Sometimes these patients are described as having ‘cardiac asthma’, which must be differentiated from primary asthma and pulmonary causes of wheezing.

Both orthopnoea and paroxysmal nocturnal dyspnoea are relatively specific for heart failure but are usually only encountered in untreated or advanced heart failure and are uncommon in most patients with mild to moderate heart failure taking diuretics [161–166]. Treated patients developing either symptom should be advised to report this to their physician/nurse as soon as possible. Paroxysmal nocturnal dyspnoea requires urgent treatment.

Cerebral symptoms such as confusion, disorientation, sleep or mood disturbances may be observed in advanced heart failure, particularly in the presence of hyponatraemia. These symptoms can be the first manifestation of heart failure in elderly patients. Sometimes sleep disturbances can be associated with ventilatory abnormalities (including central and obstructive sleep apnoea and Cheyne–Stokes respiration) which occur in advanced heart failure and may be reported by the patient’s spouse or partner [167].

Nausea and abdominal discomfort may occur when there is marked congestion of the liver and gastrointestinal tract (although the former may also be caused by digitalis glycosides). Congestion of the liver and stretching of its capsule may cause pain in the right upper quadrant of the abdomen.

Oliguria is usually present in advanced heart failure as the result of reduced renal perfusion and avid sodium and water retention.

This has been alluded to earlier (graphic Table 23.2). We prefer to use the NYHA functional classification [168]. Although it is subjective and has a large interobserver variation, it is used worldwide and, most importantly, has been employed as an entry criterion in almost all important clinical trials in heart failure.

At the individual patient level it is useful to measure functional limitation (and monitor progress) by way of the distance the patient can walk on the level, the number of steps that can be climbed, and ordinary activities that can (or cannot) be carried out, e.g. washing, bed-making, vacuum-cleaning, sweeping, shopping, etc. Because there is a poor correlation between symptoms and cardiac dysfunction, it must be emphasized that mild symptoms do not imply minor cardiac dysfunction or a good prognosis. Patients with very different EFs can experience quite similar degrees of functional limitation and treatment with a diuretic may lead to a marked improvement in symptoms without having any effect on cardiac function [169].

The Killip classification may be used to assess the severity of heart failure in the acute context, e.g. in myocardial infarction [170].

‘Quality of life’ assessments have been used to assess the impact of heart failure on patient well-being in a more complete way than just measuring specific symptoms or functional limitations. Various dimensions of quality of life including those reflecting physical, social, sexual, and professional activities can be measured, along with indices of mood, emotions, and mental health. Various questionnaires or visual scales have been proposed but none has been universally accepted. One of the most widely used is the Minnesota Living with Heart Failure questionnaire which includes a list of 21 questions, each answered on a scale of 0 to –5 [171]. More recently, the Kansas City Cardiomyopathy Questionnaire has been used [172]. These measures are more often used in clinical trials than in clinical practice.

Clinical examination, including observation of the patient and palpation and auscultation of the heart, is essential in the assessment of an individual with suspected heart failure. Percussion of the heart is seldom performed although it can provide an accurate assessment of cardiac size; percussion of the lung fields is valuable [165, 166, 173].

Patients with advanced heart failure are sometimes severely dyspnoeic even when speaking and have peripheral oedema, cachexia, or cyanosis. Conversely, the general appearance of a patient presenting with mild to moderate heart failure is often normal.

Systolic blood pressure is usually reduced in heart failure because of left ventricular systolic dysfunction, especially if severe or treated. Sometimes blood pressure may be elevated, especially if hypertension is the cause of heart failure and particularly if systolic function is preserved. Blood pressure can be markedly increased during an episode of acute pulmonary oedema. It is important to distinguish between low blood pressure (hypotension) which may be unimportant per se and hypoperfusion of the vital organs, i.e. where there are symptoms such as dizziness or confusion, renal dysfunction, or myocardial ischaemia, which is always important and requires attention.

Sinus tachycardia is a non-specific sign which is caused by increased sympathetic activity and can be absent in the presence of conduction disturbances (or if the patient is taking beta-adrenergic blocker therapy). Some patients may also have tachycardia because of atrial fibrillation (or another supraventricular arrhythmia) or, rarely, ventricular tachycardia.

Peripheral vasoconstriction with coldness, cyanosis, and pallor of extremities is also caused by increased sympathetic activity.

Peripheral oedema is a key manifestation of heart failure but is non-specific and usually absent in patients already treated with diuretics (graphic Fig. 23.13). It is related to extracellular volume expansion, is accompanied (and even preceded) by weight gain, and is progressive. It is usually bilateral and symmetrical, painless, pitting, and occurs first in the lower extremities in ambulatory patients, namely the feet and the ankles. In bedridden patients, oedema may instead be found over the sacrum and scrotum. Even oedema to mid-calf may reflect an increase of ≥2L in extracellular fluid volume. Long-standing leg oedema may be associated with indurated and pigmented skin. If untreated, oedema may become generalized (anasarca), with the development of hepatic congestion, ascites, and hydrothorax (pleural effusions). At this stage there is usually clear jugular venous distension (graphic Fig. 23.13—see graphic Cardiac signs, p.853). Generalized oedema is often accompanied by resistance to oral diuretic treatment. Patients should be warned to be observant for progressive increases in weight, accompanied by ankle swelling and, especially, increasing dyspnoea. Daily weight monitoring is important to identify sodium and water retention episodes and initiate early therapy. A prompt (and often temporary) increase in diuretic therapy may resolve worsening congestion in this situation.

Hepatomegaly is an important but uncommon sign in patients with heart failure. The liver is usually tender except in long-standing heart failure and can pulsate during systole in the presence of tricuspid regurgitation. Firm and continuous compression of the right upper abdominal quadrant for 30s to 1min may exhibit hepato-jugular reflux, i.e. an increase in jugular distension that is sustained during and after compression (see graphic jugular venous distension in Cardiac signs, p.853). Examination should be made on a patient lying comfortably with their head resting on a pillow.

Jugular venous distension, detected by inspection of the internal jugular veins, may identify an elevated right atrial pressure and by inference (and in the absence of tricuspid and pulmonary valve disease) left atrial pressure (graphic Fig. 23.13). Although, estimates of jugular venous pressure by physical examination correlate poorly with invasive measurement of right atrial pressure and interobserver reproducibility is low among non-specialists, elevation of jugular venous pressure is of prognostic importance [165, 166, 173–177]. Giant ‘V waves’ indicate the presence of tricuspid incompetence.

A third heart sound is usually only heard when there is left ventricular dilatation and systolic dysfunction, but interobserver agreement on this sign is low. A third heart sound is more common in severe heart failure and is associated with poor prognosis [165, 166, 173, 175].

A systolic murmur as because mitral or tricuspid regurgitation can be present, even in the absence of primary valve disease, when the left or right ventricle is markedly enlarged, leading to a dilatation of the mitral or the tricuspid annulus. In the latter case, the tricuspid murmur is selectively increased in loudness following inspiration (Carvallo’s sign). The degree of mitral regurgitation can be dynamic and increase on exertion.

Pulmonary crackles (crepitations or rales) result from the transudation of fluid from the intravascular space into the alveoli. The presence of crackles at the lung bases is suggestive of pulmonary congestion but the positive predictive value of this sign is low and the interobserver variability is high [165, 166, 173]. Moreover, the origin of crackles can be difficult to assess in smokers who may also have chronic pulmonary disease (or in patients at risk of pulmonary disease for some other reason). In acute pulmonary oedema, bubbling crackles may be accompanied by expectoration of frothy sputum which is blood stained. Patients with long-standing heart failure may become resistant to developing pulmonary oedema and only do so at very high left atrial pressures. Pleural effusions can also be detected in patients with heart failure; they are normally bilateral and usually associated with marked dyspnoea and generalized congestion.

Overall, the presence of several of the aforementioned symptoms and signs, particularly in the context of a history of previous cardiac disease, is suggestive of heart failure, if notits precise cause. The declining skill of physicians in clinical examination, the lack of sensitivity and specificity of most signs (and the large interobserver variability in their detection), and the subjective nature of clinical assessment highlight the need for objective assessment of cardiac function. This objective assessment is also essential for diagnosis of the cause of heart failure and, therefore, treatment tailored to aetiology.

A resting 12-lead electrocardiogram (ECG) is one of the most useful investigations in a patient with suspected heart failure and is recommended as a first-line diagnostic test in the European Society of Cardiology (ESC) guidelines (graphic Fig. 23.16) [1]. The ECG provides diagnostic and prognostic information and helps in choosing treatment. ECG changes are frequent in heart failure and a normal ECG virtually excludes left ventricular systolic dysfunction [1,177]. Various abnormalities may be present, such as abnormal Q waves (graphic Fig. 23.17), left bundle branch block (graphic Fig. 23.18) and other conduction disturbances, left atrial or left ventricular hypertrophy (graphic Fig. 23.19), atrial or ventricular arrhythmias may suggest a specific aetiology or precipitating factor. Some abnormalities provide prognostic information and help in choosing treatment, e.g. bundle branch block is predictive of a worse prognosis (178, 179) in patients with left ventricular systolic dysfunction and may also identify patients who benefit from a specific treatment (cardiac resynchronization therapy). The same is true of atrial fibrillation where there may be an indication for warfarin.

 Flow chart for the diagnosis of heart
failure with natriuretic peptides in untreated patients with symptoms
suggestive of heart failure. Adapted with permission from Dickstein K,
Cohen-Solal A, Filippatos G, et al. 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; 29:
2388–442.
Figure 23.16

Flow chart for the diagnosis of heart failure with natriuretic peptides in untreated patients with symptoms suggestive of heart failure. Adapted with permission from Dickstein K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442.

 Twelve-lead ECG depicting an
established transmural inferior myocardial infarction—there are Q-waves and
T-wave inversion seen in leads II, III, and aVF.
Figure 23.17

Twelve-lead ECG depicting an established transmural inferior myocardial infarction—there are Q-waves and T-wave inversion seen in leads II, III, and aVF.

 Twelve-lead electrocardiogram
depicting left bundle branch block (LBBB).
Figure 23.18

Twelve-lead electrocardiogram depicting left bundle branch block (LBBB).

 Twelve-lead electrocardiogram
depicting left ventricular hypertrophy.
Figure 23.19

Twelve-lead electrocardiogram depicting left ventricular hypertrophy.

The chest X-ray (or radiograph) is also recommended as a first-line investigation (graphic Fig. 23.16). It may identify a non-cardiac cause for the patient’s symptoms. It also provides information on the size and shape of the cardiac silhouette and the state of the pulmonary vasculature [180]. This information is, however, of limited value [181]. The absence of cardiomegaly (graphic Fig. 23.20) does not exclude valve disease or left ventricular systolic dysfunction (heart size is more often normal in patients with left ventricular diastolic dysfunction and in acute compared to chronic heart failure). Even if cardiomegaly is present (graphic Fig. 23.20), the chest X-ray does not identify the cause of cardiac enlargement. The relationship between central haemodynamic and pulmonary vascular radiological abnormalities is also variable and some patients with long-standing severe heart failure may not show pulmonary venous congestion or oedema (graphic Fig. 23.20) despite a very high pulmonary capillary pressure [182].

 Posteroanterior chest X-rays of (A) a
patient with left ventricular systolic dysfunction showing marked
cardiomegaly; (B) a patient in acute pulmonary oedema. There is evidence of
‘bat’s wings pulmonary oedema’, upper lobe venous diversion, as well as
fluid in the horizontal fissure.
Figure 23.20

Posteroanterior chest X-rays of (A) a patient with left ventricular systolic dysfunction showing marked cardiomegaly; (B) a patient in acute pulmonary oedema. There is evidence of ‘bat’s wings pulmonary oedema’, upper lobe venous diversion, as well as fluid in the horizontal fissure.

Several laboratory investigations are recommended in the ESC guidelines as part of the routine diagnostic evaluation of patients with suspected heart failure: complete blood count, electrolytes, glucose, urea, creatinine, hepatic enzymes, and urinalysis [1]. Myocardial biomarkers such as troponin T or I are important during an acute episode to rule out myocardial infarction, although some degree of troponin elevation occurs in around a third of patients hospitalized with acute heart failure and is a powerful predictor of outcome [183, 184]. Other tests including serum uric acid, C-reactive protein, and thyroid stimulating hormone are optional. It is important to repeat some of these tests during follow-up and after the initiation of certain treatments (or dose changes), e.g. urea, creatinine, and potassium.

Electrolyte disturbances are uncommon in untreated mild to moderate heart failure.

Hyponatraemia is sometimes found in severe heart failure and its cause is complex and probably multifactorial. Impaired free water excretion, sodium restriction, and diuretic therapy (especially thiazide diuretic treatment or excessive use of loop diuretics) are thought to be important. Hyponatraemia may identify patients with an elevated plasma concentration of arginine vasopressin (AVP) and a particularly activated RAAS [134, 137]. AVP antagonists may have a therapeutic role in patients with hyponatraemia [185].

Potassium concentration is usually normal but may be reduced by the use of kaliuretic agents such as loop diuretics, or increased in patients with end-stage heart failure with markedly reduced glomerular filtration rate, particularly in the presence of concomitant renal disease, treatment with an inhibitor of the RAAS (e.g. some combination of an ACE inhibitor, angiotensin II receptor blocker, or spironolactone) [185]. Nephrotoxic drugs such as NSAIDs may also precipitate hyperkalaemia.

Elevation of creatinine and urea is common in treated heart failure, especially if severe. A significant reduction in glomerular filtration rate may be present despite a normal urea and creatinine, especially in patients with reduced muscle mass. Several causes are recognized: (1) reduced glomerular filtration rate in advanced heart failure; (2) excessive treatment with diuretics alone or in combination with inhibitors of the RAAS (e.g. some combination of an ACE inhibitor, angiotensin II receptor blockerl or spironolactone); (3) ageing; and (4) primary renal disease, including renal artery stenosis.

Impaired renal function and worsening renal function are associated with a poor outcome in chronic heart failure though, paradoxically, drugs which improve prognosis, particularly inhibitors of the RAAS, may cause some deterioration in renal function (although this is usually mild).

Elevation of liver enzymes and other markers, including aspartate aminotransferase (AST) and alanine aminotransferase (ALT) and of serum bilirubin is often observed in heart failure. These changes may be caused by reduced hepatic blood flow as much as by liver congestion and are prognostically important [187, 188].

Anaemia may be the cause or consequence of heart failure. It is common, especially in advanced heart failure, and is associated with a worse prognosis.

Urinalysis is important for the detection of proteinuria or glycosuria, indications of underlying renal disease and diabetes mellitus, respectively.

Thyroid function tests are indicated if either hyper- or hypothyroidism is suspected.

The use of plasma natriuretic peptides as a tool for the diagnosis of heart failure has developed in recent years, particularly in primary care and emergency units (graphic Fig. 23.21) [189]. Both BNP and N-terminal pro-BNP are available as commercial assays. In clinical practice, both are used as ‘rule out’ tests, i.e. a normal concentration of either peptide in an untreated patient means that it is very unlikely that heart failure is present [189]. Conversely, an elevated concentration identifies a patient who merits comprehensive cardiac examination including echocardiography. Natriuretic peptides may, therefore, offer a cost-effective means of ensuring the efficient use of echocardiography. One important caveat is that prior treatment may reduce natriuretic peptide concentrations to within the normal range.

 Box plots showing median levels of
B-type natriuretic peptide (BNP) measured in men and women over 70 years of
age with dyspnoea not caused by heart failure, and those with an adjudicated
final diagnosis of heart failure, subdivided into those with systolic and
those with non-systolic congestive heart failure. CHF, congestive heart
failure. Reproduced with permission from 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; 41: 2010–17.
Figure 23.21

Box plots showing median levels of B-type natriuretic peptide (BNP) measured in men and women over 70 years of age with dyspnoea not caused by heart failure, and those with an adjudicated final diagnosis of heart failure, subdivided into those with systolic and those with non-systolic congestive heart failure. CHF, congestive heart failure. Reproduced with permission from 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; 41: 2010–17.

Age and female gender both increase plasma concentration of natriuretic peptides and must be considered when defining ‘normal’ cut-off values which are also assay-specific [189]. Plasma levels are also increased in other conditions including renal dysfunction, pulmonary embolism, left ventricular hypertrophy, acute ischaemia, and hypertension (but reduced in obesity). Because these various cardiac and non-cardiac disorders lead to a moderate increase in plasma concentrations of natriuretic peptides, a ‘grey zone’ exists and is the reason why natriuretic peptides are used as a ‘rule out’ (rather than a ‘rule in’) test.

In heart failure with preserved EF, plasma levels of BNP, although generally higher than in patients without heart failure, are significantly lower than in patients with left ventricular systolic dysfunction (graphic Fig. 23.21) [127]. In a general population, the ability of plasma natriuretic peptides to detect left ventricular hypertrophy appears limited [190]. Patients with relaxation abnormalities and mild symptoms or who are asymptomatic may have normal levels of natriuretic peptides. Thus low levels may not exclude a diagnosis of heart failure with preserved EF although those with more severe diastolic dysfunction seem to have higher natriuretic peptide concentrations [106, 127].

Transthoracic Doppler echocardiography (or ultrasound) is recognized by the ESC guidelines as the most important investigation for the patient with suspected heart failure. Echocardiography is a widely available, rapid, non-invasive, and safe technique which provides extensive information on chamber dimensions, wall thickness, and measures of systolic and diastolic function. Disappointingly, despite the value of Doppler echocardiography, surveys in Europe commonly show that cardiac function is only evaluated in approximately 50% of patients treated for suspected heart failure [191, 192]. The recent development of portable echocardiography machines, together with the potential of remote analysis of echocardiograms, may help improve this situation.

Determination of the LVEF is a key measure of left ventricular systolic function and has been used to enrol patients in almost all important clinical trials. Systolic function is usually considered to be reduced when the LVEF is <0.40 and ‘preserved’ if > 0.50. This clearly leaves a ‘grey area’ in the range 0.40–0.50. Furthermore, LVEF is a crude method for the evaluation of systolic function and is dependent not only on the intrinsic inotropic state of the myocardium, but also on the loading conditions of the heart.

The most widely recommended approach to the accurate measurement of LVEF is the modified Simpson’s method, using apical biplane summation of discs (graphic Fig. 23.22) [193]. It is, however, dependent on reliable endocardial detection. Other methods are less accurate, particularly in the presence of regional hypokinesia or akinesia. Other measures are, however, used including fractional shortening, sphericity index, and left ventricular wall motion index.

 Measurement of ejection fraction using
Simpson’s biplane method. Only the apical four-chamber view at end-diastole
is shown here. This young man has a dilated cardiomyopathy, with a left
ventricular end-diastolic diameter of 9.7cm.
Figure 23.22

Measurement of ejection fraction using Simpson’s biplane method. Only the apical four-chamber view at end-diastole is shown here. This young man has a dilated cardiomyopathy, with a left ventricular end-diastolic diameter of 9.7cm.

Identification of diastolic dysfunction is more difficult and requires evidence of abnormal left ventricular relaxation (graphic Fig. 23.23) or diastolic distensibility or diastolic stiffness. Which measures to use in daily practice are still debated [106, 194].

The finding of morphological changes such as left atrial dilatation or left ventricular hypertrophy, as well as abnormal measures of left ventricular diastolic function, is particularly helpful in determining whether diastolic dysfunction is clinically important. Abnormal diastolic function has prognostic as well as diagnostic significance [106, 195].

It is important to appreciate that the terms heart failure with preserved EF and heart failure as a result of diastolic dysfunction (‘diastolic heart failure’) describe overlapping but not identical syndromes.

Valve function can also be assessed by Doppler echocardiography, e.g. semi-quantitative evaluation of mitral regurgitation is possible (graphic Fig. 23.14), as is calculation of pulmonary artery systolic pressure (based on the velocity of tricuspid regurgitation). Doppler echocardiography may also be repeated during the follow-up of patients receiving treatment, to assess changes in cardiac structure and function.

Other new ultrasound techniques, such as three-dimensional echocardiography, are still being evaluated clinically.

Dobutamine (or exercise) stress echocardiography (graphic Chapter 4) may be used to detect ischaemia as a cause of cardiac dysfunction and to assess myocardial viability in the presence of marked hypokinesia or akinesia. It may be used to identify myocardial stunning and hibernating myocardium [196].

This technique provides information on left and right ventricular volumes and EF but is not widely available. It does not provide information on valve function but is generally more accurate than echocardiography for measuring right ventricular function. The reproducibility of this technique is also better than that of echocardiography.

Cardiac magnetic resonance (CMR) allows comprehensive and reproducible analysis of cardiac anatomy and function, including cardiac volumes and mass, global and regional function, and wall thickening [197]. When combined with contrast agents such as gadolinium, CMR also provides information on myocardial perfusion at rest (graphic Fig. 23.24) or following pharmacological intervention. CMR is now the gold standard for the assessment of mass volumes and wall motion. It also can give clues to the aetiology of specific cardiomyopathies (e.g. arrhythmogenic right ventricular cardiomyopathy) and specific heart muscle diseases. CMR is expensive and is not as widely available as echocardiography. In addition, CMR cannot be performed in patients with certain metallic implants, including cardiac devices, and patient refusal is relatively common because of claustrophobia and other factors.

 (A)Short-axis contrast-enhanced CMR
demonstrating extensive inferior myocardial infarction indicated by late
gadolinium enhancement. There is also a smaller anterior infarct. (B)
Non-contrast still end-diastolic cine image showing marked wall thinning in
the inferior wall corresponding with the area of infarction. There is marked
ventricular dilatation and systolic dysfunction. (C) Vertical long-axis view
of the same patient demonstrating both a large inferior infarct and a much
smaller apical anterior infarct. (Courtesy of Dr Patrick Mark, Western
Infirmary, Glasgow.)
Figure 23.24

(A)Short-axis contrast-enhanced CMR demonstrating extensive inferior myocardial infarction indicated by late gadolinium enhancement. There is also a smaller anterior infarct. (B) Non-contrast still end-diastolic cine image showing marked wall thinning in the inferior wall corresponding with the area of infarction. There is marked ventricular dilatation and systolic dysfunction. (C) Vertical long-axis view of the same patient demonstrating both a large inferior infarct and a much smaller apical anterior infarct. (Courtesy of Dr Patrick Mark, Western Infirmary, Glasgow.)

Treadmill or bicycle testing can be used to determine the maximum level of exercise which can be achieved. Recent recommendations on testing have been published [198]. Small increments in workload are recommended. As well as exercise duration, gas exchange is often measured. Peak oxygen uptake (vo  2) and the anaerobic threshold are useful indicators of the patient’s capacity to exercise. There is a poor correlation between exercise capacity and EF. Peak vo  2 has been used also for prognostication and selection of patients for heart transplantation. A peak vo  2 >14mL/kg/min is associated with a relatively good prognosis unlikely to be improved by heart transplantation. Patients with a vo  2 max of <14mL/kg/min have been shown to have a better survival if transplanted than if treated medically. This latter threshold is part of the criteria used for listing patients for heart transplantation [195]. It should be noted that the survival studies on which this threshold is based were carried out before the advent of modern treatments for heart failure, such as beta-blockers and cardiac resynchronization therapy.

 Patterns of left ventricular diastolic
filling as shown by standard Doppler echocardiography. The abnormal
relaxation pattern (mild diastolic dysfunction) is brought on by abnormally
slow left ventricular relaxation, a reduced velocity of early filling (E
wave), an increase in the velocity associated with atrial contraction (A
wave), and a ratio of E to A that is lower than normal. In more advanced
heart disease, when left atrial pressure has risen, the E-wave velocity and
E: A ratio is similar to that in normal subjects (the pseudonormal pattern).
In advanced disease, abnormalities in left ventricular compliance may
supervene (called the restrictive pattern because it was originally
described in patients with restrictive cardiomyopathy). Reprinted with
permission from Aurigemma GP, Gaasch WH. Diastolic Heart Failure. N Engl
J Med 2004; 351: 1097–105.
Figure 23.23

Patterns of left ventricular diastolic filling as shown by standard Doppler echocardiography. The abnormal relaxation pattern (mild diastolic dysfunction) is brought on by abnormally slow left ventricular relaxation, a reduced velocity of early filling (E wave), an increase in the velocity associated with atrial contraction (A wave), and a ratio of E to A that is lower than normal. In more advanced heart disease, when left atrial pressure has risen, the E-wave velocity and E: A ratio is similar to that in normal subjects (the pseudonormal pattern). In advanced disease, abnormalities in left ventricular compliance may supervene (called the restrictive pattern because it was originally described in patients with restrictive cardiomyopathy). Reprinted with permission from Aurigemma GP, Gaasch WH. Diastolic Heart Failure. N Engl J Med 2004; 351: 1097–105.

Pulmonary function testing is indicated in patients in whom the origin of dyspnoea is unclear, to determine whether it is of cardiac or pulmonary origin or both. Heart failure itself is associated with abnormalities of pulmonary function [200]. These include reductions in vital capacity, pulmonary diffusion at rest (and on exercise), and pulmonary compliance. Conversely, airway resistance is usually increased.

Pulmonary arterial catheterization is usually only used in acute or emergency situations, especially in patients not responding to appropriate medical therapy and is not routinely indicated [201]. This procedure allows close monitoring of haemodynamic changes following medical intervention. It is also indicated in patients with valvular heart disease who are candidates for valve replacement or repair and in the assessment of patients for transplantation.

Coronary angiography is indicated in patients with heart failure and angina or evidence of myocardial ischaemia, if revascularization is being considered. However, the role of revascularization in the treatment of heart failure, including in patients with hibernating myocardium, remains to be determined [64, 202].

Coronary angiography may also be indicated in acute heart failure with shock that is not responding to initial therapy.

Many also believe that coronary angiography is indicated as a diagnostic test in patients with heart failure or left ventricular systolic dysfunction of unknown origin.

Endomyocardial biopsy of the left or the right ventricle is only indicated when a specific myocarditis or a specific myocardial disease is suspected and during the follow-up of patients after cardiac transplantation to detect graft rejection [203].

Overall, the indication for invasive procedures in heart failure has declined. They are not necessary to establish the presence of heart failure but may be helpful on an individual basis to determine the aetiology and for the monitoring of patients in acute or difficult situations.

Ambulatory monitoring is valuable in the assessment of patients with symptoms suggestive of an arrhythmia (e.g. palpitations and syncope) and in monitoring ventricular rate control in patients with atrial fibrillation.

Asymptomatic ventricular premature beats and non-sustained ventricular tachycardia are frequent in heart failure but do not appear to be predictive of sudden death or of selecting treatment to reduce sudden death.

The following describes a stepwise approach to the diagnosis of the patient with suspected heart failure (graphic Table 23.4).

Table 23.4
Stepwise approach to the diagnosis of heart failure

Step 1: Diagnosis

Signs and symptoms of heart failure

History of cardiac disease

First-line tests: ECG, X-ray, BNP or NT-proBNP

Documentation of cardiac dysfunction: Doppler echocardiography

Nuclear angiography/Nuclear magnetic resonance

Step 2: Clinical profile

Clinical presentation: acute de novo/decompensated/chronic heart failure

Left/right side heart failure

Comorbidities

Age and severity

Step 3: Aetiology

Consider other diagnostic tests (coronary angiography, central haemodynamics, etc.)

Step 4: Precipitating factors

Anaemia

Infection (pulmonary infection)

Tachycardia (atrial fibrillation)

Bradycardia

Pulmonary embolism

Hypertensive crisis

Acute myocardial ischaemia

Poor compliance (diet and/or drugs)

Thyroid disorders

Medications (NSAIDs, cyclooxygenase-2 inhibitors, glitazones, class I antiarrhythmics, corticosteroids, tricyclic antidepressants)

Valve disorders

Acute myocardial ischaemia

Step 5: Prognostic evaluation

Clinical factors (e.g. age, sex)

Biological factors (e.g. blood pressure, ejection fraction)

Neurohumoral factors and cytokines

Electrical variables (e.g. bundle branch block, arrhythmias)

Imaging variables

Exercise testing

Central haemodynamic indices

Genetic factors

Comorbidities (e.g. diabetes mellitus)

Step 6: Treatment and follow-up

Step 1: Diagnosis

Signs and symptoms of heart failure

History of cardiac disease

First-line tests: ECG, X-ray, BNP or NT-proBNP

Documentation of cardiac dysfunction: Doppler echocardiography

Nuclear angiography/Nuclear magnetic resonance

Step 2: Clinical profile

Clinical presentation: acute de novo/decompensated/chronic heart failure

Left/right side heart failure

Comorbidities

Age and severity

Step 3: Aetiology

Consider other diagnostic tests (coronary angiography, central haemodynamics, etc.)

Step 4: Precipitating factors

Anaemia

Infection (pulmonary infection)

Tachycardia (atrial fibrillation)

Bradycardia

Pulmonary embolism

Hypertensive crisis

Acute myocardial ischaemia

Poor compliance (diet and/or drugs)

Thyroid disorders

Medications (NSAIDs, cyclooxygenase-2 inhibitors, glitazones, class I antiarrhythmics, corticosteroids, tricyclic antidepressants)

Valve disorders

Acute myocardial ischaemia

Step 5: Prognostic evaluation

Clinical factors (e.g. age, sex)

Biological factors (e.g. blood pressure, ejection fraction)

Neurohumoral factors and cytokines

Electrical variables (e.g. bundle branch block, arrhythmias)

Imaging variables

Exercise testing

Central haemodynamic indices

Genetic factors

Comorbidities (e.g. diabetes mellitus)

Step 6: Treatment and follow-up

This requires the presence of signs and symptoms suggestive of heart failure at rest or on exercise and also objective evidence of a cardiac abnormality, preferably by Doppler echocardiography. Doppler echocardiography may not be necessary in previously untreated patients with a normal plasma natriuretic peptide concentration and these patients should be investigated for another cause of their symptoms or signs (graphic Fig. 23.16). Other essential first-line diagnostic tests should be performed, including an ECG, chest X-ray, and blood tests. Pulmonary function and exercise testing may help when the diagnosis remains doubtful.

The diagnosis of heart failure with preserved EF can be particularly difficult. Although often described as a diagnosis of exclusion, i.e. other possible causes of the patient’s symptoms and signs must be ruled out, this type of heart failure may co-exist with other relevant comorbidities, e.g. chronic lung disease and anaemia. graphic Figs. 23.25 and 23.26 show an approach to this problem.

 Diagnostic flowchart on ‘How to
diagnose HF-PEF’ in a patient suspected of HF-PEF. LVEDVI, left ventricular
end-diastolic volume index; mPCW, mean pulmonary capillary wedge pressure;
LVEDP, left ventricular end-diastolic pressure; τ, time constant of left
ventricular relaxation; b, constant of left ventricular chamber stiffness;
TD, tissue Doppler; E, early mitral valve flow velocity; E’, early TD
lengthening velocity; NT-proBNP, N-terminal-pro brain natriuretric peptide;
BNP, brain natriuretic peptide; E/A, ratio of early (E) to
late (A) mitral flow velocity; DT, deceleration time; LVMI, left
ventricular mass index; LAVI, left atrial volume index; Ard, duration of
reverse pulmonary vein atrial systole flow; Ad, duration of mitral valve
atrial wave flow. Reproduced with permission from Paulus WJ, Tschope C,
Sanderson JE, et al. How to diagnose diastolic heart failure: a
consensus statement on the diagnosis of heart failure with normal left
ventricular ejection fraction by the Heart Failure and Echocardiography
Associations of the European Society of Cardiology. Eur Heart J 2007; 28: 2539–50.
Figure 23.25

Diagnostic flowchart on ‘How to diagnose HF-PEF’ in a patient suspected of HF-PEF. LVEDVI, left ventricular end-diastolic volume index; mPCW, mean pulmonary capillary wedge pressure; LVEDP, left ventricular end-diastolic pressure; τ, time constant of left ventricular relaxation; b, constant of left ventricular chamber stiffness; TD, tissue Doppler; E, early mitral valve flow velocity; E’, early TD lengthening velocity; NT-proBNP, N-terminal-pro brain natriuretric peptide; BNP, brain natriuretic peptide; E/A, ratio of early (E) to late (A) mitral flow velocity; DT, deceleration time; LVMI, left ventricular mass index; LAVI, left atrial volume index; Ard, duration of reverse pulmonary vein atrial systole flow; Ad, duration of mitral valve atrial wave flow. Reproduced with permission from Paulus WJ, Tschope C, Sanderson JE, et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology. Eur Heart J 2007; 28: 2539–50.

 Diagnostic flow chart on ‘How to
exclude HF-PEF in a patient presenting with breathlessness and no signs of
fluid overload. S, TD shortening velocity; TD, tissue Doppler. Reproduced
with permission from Paulus WJ, Tschope C, Sanderson JE, et al.. How
to diagnose diastolic heart failure: a consensus statement on the diagnosis
of heart failure with normal left ventricular ejection fraction by the Heart
Failure and Echocardiography Associations of the European Society of
Cardiology. Eur Heart J 2007; 28: 2539–50.
Figure 23.26

Diagnostic flow chart on ‘How to exclude HF-PEF in a patient presenting with breathlessness and no signs of fluid overload. S, TD shortening velocity; TD, tissue Doppler. Reproduced with permission from Paulus WJ, Tschope C, Sanderson JE, et al.. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology. Eur Heart J 2007; 28: 2539–50.

This step includes:

The assessment of clinical profile: patients may present with acute new-onset heart failure, e.g. after acute myocardial infarction, acute or subacute decompensation of chronic heart failure, or acute or subacute onset of heart failure in a patient with previously asymptomatic cardiac dysfunction. The clinical presentation may be in primary care or to hospital. The patient may have breathlessness, fatigue, or both with few clinical signs. Alternatively, the patient may have these symptoms and peripheral oedema or may present as an emergency with frank pulmonary oedema. The clinical profile of patients with preserved systolic function may be different from that of patients with systolic dysfunction: on average, the former patients are older and are more likely to be women. They are also more likely to have hypertension but are less likely to have a third heart sound [106]. It is important to assess the severity of symptoms, with the Killip and NYHA classifications being the most widely used in the acute and chronic setting, respectively. In acute new-onset heart failure, the patient may be hypotensive if there has been a haemodynamic catastrophe, e.g. rupture of a mitral valve papillary muscle or interventricular septum (graphic Fig. 23.27).

In advanced heart failure, another system of classification has been advocated. This is based on the presence/absence of signs of congestion and adequate/inadequate perfusion. Four categories have been proposed: dry–warm; wet–warm; dry–cold; and wet–cold [1, 204]. Wet patients with or without hypoperfusion are at higher risk than dry patients.

It is also important to identify comorbidities such as stroke, chronic obstructive pulmonary disease, asthma, diabetes mellitus, and renal failure, which can complicate patient management and modify outcome. The proportion of patients presenting with multiple comorbidities increases with age. In the Euro Heart Failure Survey, 9% had had a previous stroke, 10% a previous transient ischaemic attack, 27% were reported to have diabetes mellitus, 12% had dementia, 17% a renal dysfunction, and 32% a pulmonary disease [191].

 Colour flow Doppler of mitral
regurgitation and a ventricular septal rupture post-myocardial infarction in
the parasternal long-axis view.
Figure 23.27

Colour flow Doppler of mitral regurgitation and a ventricular septal rupture post-myocardial infarction in the parasternal long-axis view.

Ischaemic heart disease and hypertension are the commonest causes of heart failure in Western countries and their contribution to heart failure is also rapidly expanding in the developing countries. In the Euro Heart Failure Survey, ischaemic heart disease was the commonest cause of heart failure, 40% of patients having myocardial infarction and 51% a history of angina; 53% of the patients also had hypertension [191]. The extent of investigation to identify the underlying cardiac disease should be decided on an individual basis taking into consideration the patient profile (age, severity of heart failure and comorbidities) and the potential reversibility of cardiac dysfunction (such as valvular heart disease and reversible ischaemia).

It is also important to identify precipitating factors: decompensation of heart failure is frequently associated with comorbid factors which:

increase body metabolic requirements such as fever, infection (hence the recommendation that influenza and pneumococcal immunization/vaccination is offered to patients with heart failure) [1] or hyperthyroidism;

decrease cardiac output due to a rapid tachycardia or marked bradycardia;

reduce the oxygen transport capacity (anaemia) or oxygen exchange (pulmonary infection);

induce a sudden haemodynamic overload (pulmonary embolism or hypertensive crisis) or water and sodium overload (excessive sodium intake, poor compliance with diet, and heart failure medications)

ischaemic episodes (graphic Table 23.4).

Air pollution and seasonal temperature changes are more recently described factors associated with risk of decompensation and hospitalization [200, 201].

Among arrhythmias, a new episode of atrial fibrillation can be particularly harmful because it combines both an increase in ventricular rate (with a risk of myocardial ischaemia and reduced time for diastolic filling) and loss of atrial contraction [205, 206].

Prognostic assessment in heart failure remains difficult. Indeed the number of clinical, aetiological, comorbid, biological, haemodynamic, structural, functional, electrical, and neurohumoral variables independently associated with poor outcome is high and suggests that there is no simple method to assess the risk of death or rehospitalization in patients with this syndrome (graphic Table 23.5) [209, 210]. Most studies have been performed in populations of patients with systolic dysfunction and little is known about prognostic evaluation in patients with preserved systolic function.

Table 23.5
Prognostic factors
Clinical factors

Age, ethnicity, NYHA class

Signs of congestion, jugular vein pressure, third heart sound, low systolic blood pressure

Diabetes mellitus, renal dysfunction, depression

Ischaemic aetiology

Clinical factors

Age, ethnicity, NYHA class

Signs of congestion, jugular vein pressure, third heart sound, low systolic blood pressure

Diabetes mellitus, renal dysfunction, depression

Ischaemic aetiology

Biochemical factors

Serum sodium

Serum creatinine/creatinine clearance

Haemoglobin

Biochemical factors

Serum sodium

Serum creatinine/creatinine clearance

Haemoglobin

Neurohormones and cytokines

Plasma renin activity

Angiotensin II

Aldosterone

Galectin-3

Norepinephrine

Endothelin-1

Adrenomedullin/mid-regional pro-adrenomedullin

B type natriuretic peptide/N-terminal pro-BNP

Tumour necrosis factor-α

Vasopressin

Neurohormones and cytokines

Plasma renin activity

Angiotensin II

Aldosterone

Galectin-3

Norepinephrine

Endothelin-1

Adrenomedullin/mid-regional pro-adrenomedullin

B type natriuretic peptide/N-terminal pro-BNP

Tumour necrosis factor-α

Vasopressin

Electrical variables

QRS width

Left ventricular hypertrophy

Atrial fibrillation

Complex ventricular arrhythmia

Heart rate variability

Electrical variables

QRS width

Left ventricular hypertrophy

Atrial fibrillation

Complex ventricular arrhythmia

Heart rate variability

Imaging variables

Left ventricular internal dimensions and fractional shortening

Cardiothoracic ratio X-ray (normal <0.55)

Wall motion index (various*)

Ejection fraction (normal >0.40)

Restrictive filling pattern/short deceleration time (various*)

Right ventricular function (various*)

Mitral regurgitation

Estimated pulmonary artery pressure

Imaging variables

Left ventricular internal dimensions and fractional shortening

Cardiothoracic ratio X-ray (normal <0.55)

Wall motion index (various*)

Ejection fraction (normal >0.40)

Restrictive filling pattern/short deceleration time (various*)

Right ventricular function (various*)

Mitral regurgitation

Estimated pulmonary artery pressure

Exercise test/haemodynamic variables (rest/exercise)

Vo2 max/peak (normal >20mL/kg/min)

6-minute walk distance (normal >600 m)

Cardiac index (normal >2.5 L/min/m2)

Left ventricular end-diastolic pressure/pulmonary artery wedge pressure (normal <12mmHg)

Exercise test/haemodynamic variables (rest/exercise)

Vo2 max/peak (normal >20mL/kg/min)

6-minute walk distance (normal >600 m)

Cardiac index (normal >2.5 L/min/m2)

Left ventricular end-diastolic pressure/pulmonary artery wedge pressure (normal <12mmHg)

*

Various measures/classifications can be used and no single threshold for normal/abnormal can be given; † functional capacity varies greatly according to prior fitness, age and sex; values given are a guideline for older (>65 years) adults.

The assessment of prognosis has also been conducted differently in acute and in chronic heart failure: in acute heart failure, in-hospital and short-term (3–6 months) mortality or readmission have usually been evaluated whereas in chronic heart failure, long-term (> 1 year) prognosis and rehospitalization rates have normally been considered.

Furthermore, factors which predispose to overall mortality or pump failure mortality do not necessarily apply to sudden death.

An additional problem is that extrapolation of risk assessment based on a small series of selected patients exposed to conventional therapy (including low rate of prescription of ACE inhibitors and beta-blockers) to the overall current heart failure population is difficult. The changing background therapy of heart failure also makes it difficult to provide simple prognostic algorithms. For instance, beta-blockers have more influence on the remodelling process than exercise capacity, so that the relative role of these two independent predictors of mortality may be different in patients treated with a beta-blocker and those not so treated.

The temporal role of the various prognostic factors can be variable: the time course of the activation of the neurohumoral systems after myocardial injury is different. Therefore, the relative weight of elevated plasma levels of neurohumoral factors may be different in the short term compared to the longer term. Moreover, little is known about the relation between the change in plasma concentrations of biochemical markers as a result of treatment and long-term prognosis [211, 212].

The multivariable analyses reported so far usually include only a limited number of parameters and these may lose their predictive power in more comprehensive analyses.

Finally, some factors can provide prognostic information in advanced heart failure but not in mild to moderate heart failure: severely reduced functional capacity, measured by VO2, is a recognized index for the selection of patients who might benefit from heart transplantation whereas elevated, non-reversible, pulmonary resistance is an index of poor outcome after heart transplantation or implantation of a ventricular assist device [199].

Full diagnosis is a prerequisite for optimal treatment [1]. Often, however, a diuretic may be required before a full diagnostic work-up is completed. The further treatment of heart failure depends on the underlying aetiology (e.g. valve replacement for aortic stenosis), functional status (e.g. spironolactone for severely symptomatic patients), comorbidity (e.g. warfarin if atrial fibrillation), and the results of investigations (e.g. cardiac resynchronization therapy if a broad QRS on the ECG). Each of these may contraindicate treatments as well as indicate them (e.g. caution with ACE inhibitors in aortic stenosis, beta-blockers if atrioventricular block, spironolactone if renal dysfunction etc.).

The goals of treatment for patients with heart failure are relief of symptoms, avoidance of hospital admission, and prevention of premature death.

Drugs are the mainstay of the treatment of patients with heart failure based upon a series of key randomized controlled trials (graphic Table 23.6). However, devices and surgery have an important and increasing role, particularly in patients with more severe heart failure (graphic Fig. 23.28). How care is structured and delivered is also important. Although lifestyle measures are also considered important, the evidence base for these interventions is less robust.

Table 23.6
Positive randomized controlled trials* in patients with symptomatic heart failure and a low left ventricular ejection fraction
Treatment, trial, year published N Severity of heart failure Estimated first year placebo/control group mortality Background treatment*  * Treatment added Trial duration (years) Primary endpoint RRR (%)*  *  * Events prevented per 1000 patients treated††
Death HF Hosp. Death or HF Hosp.

ACE inhibitors

CONSENSUS 1987

 

SOLVD-T 1991

253

 

2569

End-stage

 

Mild–severe

52

 

15.7

Spiro

 

Enalapril 20mg BID

 

Enalapril 20mg BID

0.54

 

3.5

Death

 

Death

40

 

16

146

 

45

 

96

 

108

Beta-blockers

CIBIS-2 1999

 

MERIT-HF 1999

 

COPERNICUS 2001

 

SENIORS 2005

2647

 

3991

 

2289

 

2128

Moderate–severe

 

Mild–serve

 

Severe

 

Mild–severe

13.2

 

11.0

 

19.7

 

8.5

ACE-I

 

ACE-I

 

ACE-I

 

ACE-I + Spiro

Bisoprolol 10mg QD

 

Metoprolol CR/XL, 200mg QD

 

Carvedilol 25mg BID

 

Nebivolol 10mg QD

1.3

 

1.0

 

0.87

 

1.75

Death

 

Death

 

Death

 

Death or CV Hosp.

34

 

34

 

35

 

14

55

 

36

 

55

 

23

56

 

46

 

65

 

0

 

63

 

81

 

0

ARBs

Val-HeFT 2001

 

CHARM-Alternative 2003

 

CHARM-Added 2003

5010

 

2028

 

2548

Mild–severe

 

Mild–severe

 

Moderate–severe

∼8.0

 

12.6

 

10.6

ACE-I

 

BB

 

ACE-I +BB

Valsartan 160mg BID

 

Candesartan 32mg QD

 

Candesartan 32mg QD

1.9

 

2.8

 

3.4

CV Death or Morbidity

 

CV Death or HF Hosp.

 

CV Death or HF Hosp.

13

 

23

 

15

0

 

30

 

28

35

 

31

 

47

33†††

 

60

 

39

Aldosterone blockade

RALES 1999

1663

Severe

∼25

ACE-I

Spirolactone 25–50mg QD

2.0

Death

30

113

95

H-ISDN

V-HeFT-1 1986

 

A-HeFT 2004

459

 

1050

Mild–severe

 

Moderate–severe

26.4

 

∼9.0

 

ACE-I+BB + Spiro

Hydralazine 75mg TID–QID

 

ISDN 40mg QID

 

Hydralazine 75mg TID

 

ISDN 40mg TID

2.3

 

0.83

Death

 

Composite

34

 

52

 

40

0

 

80

 

n-3 PUFA

GISSI-HF 2008

6975

Mild–severe

∼9.0

ACE-I + BB + Spiro

n-3 PUFA 1g QD

3.9

Death

 

Death or CV Hosp.

9

 

8

18

0

Digitalis glycosides

 

DIG 1997

6800

Mild–severe

∼11.0

ACE-I

Digoxin

3.1

Death

0

0

79

73

Exercise

HF-ACTION 2009

2331

Mild–severe

∼6.0

ACE-I + BB + Spiro

Exercise

2.5

Death or CV Hosp.

11

0

CRT

COMPANION 2004

 

CARE-HF 2005

925

 

813

Moderate–severe

 

Moderate–severe

19.0

 

12.6

ACE-I + BB + Spiro

 

ACE-I + BB + Spiro

CRT

 

CRT

1.35

 

2.45

Death or Any Hosp.

 

Death or CV Hosp.

19

 

37

38

 

97

 

151

87

 

184

CRT-D

COMPANION 2004

903

Moderate–severe

19.0

ACE-I + BB + Spiro

CRT-ICD

1.35

Death or Any Hosp.

20

74

114

ICD

SCD-HeFT 2005

1676

Mild–severe

∼7.0

ACE-I + BB

ICD

3.8

Death

23

VAD

REMATCH 2001

129

End-stage

75

ACE-I + Spiro

LVAD

1.8

Death

48

282

Treatment, trial, year published N Severity of heart failure Estimated first year placebo/control group mortality Background treatment*  * Treatment added Trial duration (years) Primary endpoint RRR (%)*  *  * Events prevented per 1000 patients treated††
Death HF Hosp. Death or HF Hosp.

ACE inhibitors

CONSENSUS 1987

 

SOLVD-T 1991

253

 

2569

End-stage

 

Mild–severe

52

 

15.7

Spiro

 

Enalapril 20mg BID

 

Enalapril 20mg BID

0.54

 

3.5

Death

 

Death

40

 

16

146

 

45

 

96

 

108

Beta-blockers

CIBIS-2 1999

 

MERIT-HF 1999

 

COPERNICUS 2001

 

SENIORS 2005

2647

 

3991

 

2289

 

2128

Moderate–severe

 

Mild–serve

 

Severe

 

Mild–severe

13.2

 

11.0

 

19.7

 

8.5

ACE-I

 

ACE-I

 

ACE-I

 

ACE-I + Spiro

Bisoprolol 10mg QD

 

Metoprolol CR/XL, 200mg QD

 

Carvedilol 25mg BID

 

Nebivolol 10mg QD

1.3

 

1.0

 

0.87

 

1.75

Death

 

Death

 

Death

 

Death or CV Hosp.

34

 

34

 

35

 

14

55

 

36

 

55

 

23

56

 

46

 

65

 

0

 

63

 

81

 

0

ARBs

Val-HeFT 2001

 

CHARM-Alternative 2003

 

CHARM-Added 2003

5010

 

2028

 

2548

Mild–severe

 

Mild–severe

 

Moderate–severe

∼8.0

 

12.6

 

10.6

ACE-I

 

BB

 

ACE-I +BB

Valsartan 160mg BID

 

Candesartan 32mg QD

 

Candesartan 32mg QD

1.9

 

2.8

 

3.4

CV Death or Morbidity

 

CV Death or HF Hosp.

 

CV Death or HF Hosp.

13

 

23

 

15

0

 

30

 

28

35

 

31

 

47

33†††

 

60

 

39

Aldosterone blockade

RALES 1999

1663

Severe

∼25

ACE-I

Spirolactone 25–50mg QD

2.0

Death

30

113

95

H-ISDN

V-HeFT-1 1986

 

A-HeFT 2004

459

 

1050

Mild–severe

 

Moderate–severe

26.4

 

∼9.0

 

ACE-I+BB + Spiro

Hydralazine 75mg TID–QID

 

ISDN 40mg QID

 

Hydralazine 75mg TID

 

ISDN 40mg TID

2.3

 

0.83

Death

 

Composite

34

 

52

 

40

0

 

80

 

n-3 PUFA

GISSI-HF 2008

6975

Mild–severe

∼9.0

ACE-I + BB + Spiro

n-3 PUFA 1g QD

3.9

Death

 

Death or CV Hosp.

9

 

8

18

0

Digitalis glycosides

 

DIG 1997

6800

Mild–severe

∼11.0

ACE-I

Digoxin

3.1

Death

0

0

79

73

Exercise

HF-ACTION 2009

2331

Mild–severe

∼6.0

ACE-I + BB + Spiro

Exercise

2.5

Death or CV Hosp.

11

0

CRT

COMPANION 2004

 

CARE-HF 2005

925

 

813

Moderate–severe

 

Moderate–severe

19.0

 

12.6

ACE-I + BB + Spiro

 

ACE-I + BB + Spiro

CRT

 

CRT

1.35

 

2.45

Death or Any Hosp.

 

Death or CV Hosp.

19

 

37

38

 

97

 

151

87

 

184

CRT-D

COMPANION 2004

903

Moderate–severe

19.0

ACE-I + BB + Spiro

CRT-ICD

1.35

Death or Any Hosp.

20

74

114

ICD

SCD-HeFT 2005

1676

Mild–severe

∼7.0

ACE-I + BB

ICD

3.8

Death

23

VAD

REMATCH 2001

129

End-stage

75

ACE-I + Spiro

LVAD

1.8

Death

48

282

*

excluding active–controlled trials (patients with preserved LVEF as well as low LVEF were included in CONSNSUS and SENIORS).; ** in >1/3 of patients: ACE-I + BB means ACE-Is used in almost all patients and BB in the majority. Most patients also taking diuretics and many digoxin (except in DIG). Spironolactone was used at baseline in 5% Val-HeFT, 8% MERIT-HF, 17% CHARM-Added, 19% SCD-HeFT, 20% COPERNICUS, and 24% in CHARM Alternative.

***

relative risk reduction in primary endpoint. HF hosp., patients with at least one hospital admission for worsening HF; some patients had multiple admissions.; † stopped early for benefit.; †† individual trials may not have been designed or powered to evaluate effect of treatment on these outcomes.; ††† primary endpoint which also included treatment of HF with intravenous drugs for 4 hours or more without admission and resuscitated cardiac arrest (both added small numbers).

ACE-I, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; BB, beta-blocker; CRT, cardiac resynchronisation therapy (biventricular pacing); CRT-D, CRT device that also defibrillates; CV,cardiovascular; hosp., hospital admission; ICD, implantable cardioverter defibrillator; ISDN, isosorbide dinitrate; LVAD, left ventricular assist device; pub., published; RRR, relative risk reduction; spiro, spironolactone; VAD, ventricular assist device.

A-HeFT, African-American Heart Failure Trial; CARE HF, Cardiac Resynchronization-Heart Failure; COPERNICUS, Carvedilol Prospective Randomized Cumulative Survival; CIBIS, Cardiac Insufficiency Bisoprolol Study COMPANION, Comparison of Medical Therapy, Pacing, and Defibrillation in Heart Failure; CONSENSUS, Cooperative North Scandinavian Enalapril Survival Study; DIG, Digitalis Investigation Group; GISSI-HF, Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico -Heart Failure; HF-ACTION, Heart Failure- A Controlled Trial Investigating Outcomes Exercise TraiNing; MERIT-HF, Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure; RALES, Randomized Aldactone Evaluation Study; REMATCH, Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure; SENIORS, Study of Effects of Nebivolol Intervention on Outcomes and Rehospitalization in Seniors with Heart Failure; SOLVD-T, Studies of Left Ventricular Dysfunction Treatment; V-HeFT, Vasodilator Heart Failure Trial; Val-HeFT, Valsartan Heart Failure Trial. Adapted with permission from McMurray JJ, Pfeffer MA. Heart failure. Lancet 2005; 365: 1877–89.

 A treatment algorithm for patients
with symptomatic heart failure and reduced ejection fraction. ACE-I,
angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker;
CAD, coronary artery disease; CRT, cardiac resynchronization therapy (D,
with defibrillator function; P, pacing only); ICD, implantable cardioverter
defibrillator; LVAD, left ventricular assist device; LVEF, left ventricular
ejection fraction. Dickstein K, Cohen-Solal A, Filippatos G, et al.
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; 29: 2388–422. Copyright ©ESC, 2008.
Figure 23.28

A treatment algorithm for patients with symptomatic heart failure and reduced ejection fraction. ACE-I, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; CAD, coronary artery disease; CRT, cardiac resynchronization therapy (D, with defibrillator function; P, pacing only); ICD, implantable cardioverter defibrillator; LVAD, left ventricular assist device; LVEF, left ventricular ejection fraction. Dickstein K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–422. Copyright ©ESC, 2008.

Diuretics act by blocking sodium reabsorption at specific sites in the renal tubule, thereby enhancing urinary excretion of sodium and water.

Although not proven to improve mortality and morbidity in large trials, diuretics are required in nearly all patients with symptomatic heart failure to relieve dyspnoea and the signs of sodium and water retention (‘congestion’), i.e. peripheral and pulmonary oedema [213, 214]. No other treatment relieves symptoms and the signs of sodium and water overload as rapidly and effectively. Once a patient needs a diuretic, treatment is usually necessary for the rest of the patient’s life, though the dose and type of diuretic may vary.

The key principle is to prescribe the minimum dose of diuretic needed to maintain an oedema-free state (‘dry weight’) [213]. Excessive use can lead to electrolyte imbalances, such as hyponatraemia, hypokalaemia (with the risk of digitalis toxicity), hyperuricaemia (with the risk of gout), and uraemia. The risk of renal dysfunction is increased by concomitant use of NSAIDs. Diuretic-induced hypovolaemia may also cause symptomatic hypotension and pre-renal uraemia. Restriction of dietary sodium intake may help reduce, but not eliminate, the requirement for diuretics. Diuretic dosing should be flexible with temporary increases for evidence of fluid retention (e.g. increasing symptoms, weight gain, oedema) and decreases for evidence of hypovolaemia (e.g. as consequence of increased electrolyte loss owing to gastroenteritis, decreased fluid intake, or both).

In some patients with milder symptoms of heart failure (NYHA class II), a thiazide diuretic such as bendroflumethiazide may suffice. In more severe heart failure or in patients with concomitant renal dysfunction, a loop diuretic such as furosemide is often needed. Loop diuretics cause a rapid onset of an intense but relatively short-lived diuresis, compared with the longer-lasting but gentler effect of a thiazide diuretic. The timing of administration of a loop diuretic, which need not be taken first thing every morning, can be adjusted according to the patient’s social activities. The dose may be postponed or even temporarily omitted if the patient has to travel or has another activity that might be compromised by the prompt action of the diuretic. In severe heart failure, the effects of long-term administration of a loop diuretic may be diminished by increased sodium reabsorption at the distal tubule. This problem can be offset by use of the combination of a loop diuretic and a thiazide or thiazide-like diuretic (e.g. metolazone), which acts in synergy with a loop diuretic by blocking sodium reabsorption in different segments of the nephron [215]. This combination requires more frequent monitoring of electrolytes and renal function for diuretic-induced hyponatraemia, abnormalities of the serum potassium level, and prerenal uraemia.

A period of intravenous loop diuretic, given either as bolus injections or by continuous infusion, may be required in patients who become resistant to the action of oral diuretics [216]. Why this resistance develops is uncertain, but factors thought to be important include impaired absorption of oral diuretics owing to gut oedema, hypotension, reduced renal blood flow, and adaptive changes in the nephron. In cases of severe resistant volume overload, mechanical removal of fluid using ultra-filtration may be considered [217].

Patients with severe failure (NYHA class III–IV) should usually also be treated with an aldosterone antagonist, such as spironolactone, which increases excretion of sodium but not potassium [218]. Patients receiving a combination of diuretics require careful monitoring of blood chemistry and clinical status. The use of a potassium sparing diuretic or aldosterone antagonist along with an ACE inhibitor or ARB (treatment with all three is not recommended) requires particular care and surveillance for hyperkalaemia [219].

Although highly effective in relieving symptoms and signs, diuretics alone are not a sufficient treatment for heart failure. The addition of other disease-modifying treatments is essential to slow pathophysiological progression, better maintain clinical stability, and reduce the risk of hospital admission and premature death.

These drugs act by inhibiting the enzyme that converts the inactive decapeptide angiotensin I to the active octapeptide angiotensin II. Excessive angiotensin II is thought to exert the myriad of harmful actions described earlier by stimulating the angiotensin II type 1 receptor subtype (AT1R)—see graphic Pathophysiology, p.843. ACE inhibitors also reduce the breakdown of bradykinin (as ACE is identical to kininase II) and the resultant accumulation of bradykinin is directly or indirectly responsible for two of the specific adverse effects of ACE inhibitors: cough and angio-oedema. Bradykinin may, however, also have beneficial effects (vasodilation, inhibition of adverse cardiovascular remodelling, and antithrombotic actions), though the importance of these bradykinin-mediated actions to the clinical benefits of ACE inhibition is uncertain.

When added to diuretics and digoxin, treatment with an ACE inhibitor decreases left ventricular size, improves systolic function, reduces symptoms and hospital admissions, and prolongs survival (graphic Table 23.6; Fig. 23.29) [220, 221]. These agents also reduce the risk of developing myocardial infarction and possibly atrial fibrillation [222]. Consequently, treatment with an ACE inhibitor is recommended for all patients with systolic dysfunction, irrespective of symptom severity or aetiology. ACE inhibitors are not a substitute for a diuretic but mitigate diuretic-induced hypokalaemia.

 Meta-analysis of long-term (>
1-year duration) placebo-controlled trials (> 1000 patients) of
angiotensin-converting enzyme (ACE) inhibitors in chronic heart failure or
left ventricular dysfunction after a recent myocardial infarction. RR,
relative risk. Reproduced with permission from Flather, MD, Yusuf, Køber L, et al. Long-term ACE-inhibitor therapy in patients with heart
failure or left-ventricular dysfunction: a systematic overview of data from
individual patients. ACE-Inhibitor Myocardial Infarction Collaborative
Group. Lancet 2000; 355: 1575–81.
Figure 23.29

Meta-analysis of long-term (> 1-year duration) placebo-controlled trials (> 1000 patients) of angiotensin-converting enzyme (ACE) inhibitors in chronic heart failure or left ventricular dysfunction after a recent myocardial infarction. RR, relative risk. Reproduced with permission from Flather, MD, Yusuf, Køber L, et al. Long-term ACE-inhibitor therapy in patients with heart failure or left-ventricular dysfunction: a systematic overview of data from individual patients. ACE-Inhibitor Myocardial Infarction Collaborative Group. Lancet 2000; 355: 1575–81.

ACE inhibitors should be introduced as early as possible in a patient’s treatment [219]. The only contraindications are current symptomatic hypotension, severe aortic stenosis, and bilateral renal artery stenosis; the latter is often associated with a prompt and marked increase in serum levels of blood urea and creatinine when renal perfusion is reduced precipitously by inhibiting the production and actions of angiotensin. Treatment should be started in a low dose (graphic Table 23.7) and gradually increased toward that proven to be of benefit in a clinical trial (graphic Table 23.8). The patient should be evaluated for symptomatic hypotension, uraemia, and hyperkalaemia after each dose increment; these adverse effects are uncommon and can usually be resolved by reduction in the dose of diuretic (if the patient is oedema free) or concomitant hypotensive or nephrotoxic medications (e.g. nitrates, calcium-channel blockers, or NSAIDs). A dry, non-productive cough occurs in approximately 15% of patients treated with an ACE inhibitor and, if troublesome, substitution of an ARB is recommended [223]. In the rare cases of angio-oedema, the ACE inhibitor should be stopped and not used again; an ARB can be cautiously substituted.

Table 23.7
Dosages of commonly used drugs in heart failure
Starting dose (mg)Target dose (mg) ACE-I

Captopril

6.25 TID

50–100 TID

Enalapril

2.5 BID

10–20 BID

Lisinopril

2.5–5.0 OD

20–35 OD

Ramipril

2.5 OD

5 BID

Trandolapril

0.5 OD

4 OD

ARB

Candesartan

4 or 8 OD

32 OD

Valsartan

40 BID

160 BID

Aldosterone antagonist

Eplerenone

25 OD

50 OD

Spironolactone

25 OD

25–50 OD

Beta-blocker

Bisoprolol

1.25 OD

10 OD

Carvedilol

3.125 BID

25–50 BID

Metoprolol succinate

12.2/25 OD

200 OD

Nebivolol

1.25 OD

10 OD

Starting dose (mg)Target dose (mg) ACE-I

Captopril

6.25 TID

50–100 TID

Enalapril

2.5 BID

10–20 BID

Lisinopril

2.5–5.0 OD

20–35 OD

Ramipril

2.5 OD

5 BID

Trandolapril

0.5 OD

4 OD

ARB

Candesartan

4 or 8 OD

32 OD

Valsartan

40 BID

160 BID

Aldosterone antagonist

Eplerenone

25 OD

50 OD

Spironolactone

25 OD

25–50 OD

Beta-blocker

Bisoprolol

1.25 OD

10 OD

Carvedilol

3.125 BID

25–50 BID

Metoprolol succinate

12.2/25 OD

200 OD

Nebivolol

1.25 OD

10 OD

ACE-I, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; BID, twice daily; OD, once daily;TID, three times daily.

Adapted with permission from Dickstein K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442. Copyright © ESC 2008.

Table 23.8
Practical guidance on the use of ACE inhibitors in patients with heart failure due to left ventricular systolic dysfunction

Why?

 

Two major randomized trials (CONSENSUS I and SOLVD-T) and a meta-analysis of smaller trials have conclusively shown that ACE inhibitors increase survival, reduce hospital admissions, and improve NYHA class and quality of life in patients with all grades of symptomatic heart failure. Other major randomized trials in patients with systolic dysfunction after acute myocardial infarction (SAVE, AIRE, TRACE) have shown that ACE inhibitors increase survival. In patients with heart failure (ATLAS), the composite endpoint of death or hospital admission was reduced by higher doses of ACE inhibitor compared to lower doses. ACE inhibitors have also been shown to delay or prevent the development of symptomatic heart failure in patients with asymptomatic LVSD

In whom and when?

Indications:

Potentially all patients with heart failure

First-line treatment (along with beta-blockers) in patients with NYHA Class II–IV HF; start as early as possible in course of disease. ACE inhibitors are also of benefit in patients with asymptomatic LVSD (NYHA class I)

 

Contraindications:

History of angioneurotic oedema

Severe aortic stenosis

Known bilateral renal artery stensis

 

Cautions/seek specialist advice:

Significant hyperkalaemia (K+ >5.0mmol/L)

Significant renal dysfunction (creatinine 221µmol/L or >2.5mg/dL)

Symptomatic or severe asymptomatic hypotension (systolic BP <90mmHg)

 

Drug interactions to look out for:

K+ supplements/K+ sparing diuretics e.g. amiloride and triamterene (beware combination preparations with furosemide).

Aldosterone antagonists (spironolactone and eplerenone)

Aldosterone antagonists (spironolactone, eplerenone), angiotensin receptor blockers, NSAIDS*

‘Low salt’ substitutes with a high K+ content

Where?

 

In the community for most patients

 

Exceptions—see ‘Cautions/specialist advice’

Which ace inhibitor and what dose?

Starting dose (mg)

Target dose (mg)

Captopril

6.25 TID

50 TID

Enalapril

2.5 BID

10–20 BID

Lisinopril

2.5–5.0 OD

20–35 OD

Ramipril

2.5 OD

5 BID or 10 OD

Trandolapril

0.5 OD

4 OD

How to use?

 

Start with a low dose (see above)

 

Double dose at not less than 2-weekly intervals

 

Aim for target dose (see above) or, failing that, the highest tolerated dose

 

Remember some ACE inhibitor is better than no ACE inhibitor

 

Monitor blood pressure and blood chemistry (urea/BUN, creatinine, K+)

 

Check blood chemistry 1–2 weeks after initiation and 1–2 weeks after final dose titration

 

When to stop up-titration/reduce dose/stop treatment—see ‘Problem solving’

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), biochemical monitoring, and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, to prevent worsening of heart failure leading to hospital admission, and to increase survival

Advice to patient?

 

Symptoms improve within a few weeks to a few months of starting treatment

 

Advise patients to report principal adverse effects i.e. dizziness/symptomatic hypotension, cough—see ‘Problem solving’

 

Advise patients to avoid NSAIDs* not prescribed by a physician (self-purchased ‘over-the counter’) and salt substitutes high in K+— see ‘Problem solving’

Problem solving

Asymptomatic low blood pressure:

Does not usually require any change in therapy

 

Symptomatic hypotension:

If dizziness, light-headedness and/or confusion, and a low blood pressure reconsider need for nitrates, calcium-channel blockers**, and other vasodilators

If no signs/symptoms of congestion consider reducing diuretic dose

If these measures do not solve problem seek specialist advice

 

Cough:

Cough is common in patients with heart failure, many of whom have smoking-related lung disease

Cough is also a symptom of pulmonary oedema which should be excluded when a new or worsening cough develops

ACE inhibitor-induced cough rarely requires treatment discontinuation

When a very troublesome cough does develop (e.g. one stopping the patient sleeping) and can be proven to be due to ACE inhibition (i.e. recurs after ACE inhibitor withdrawal and rechallenge) substitution of an ARB should be made (graphic Table 23.9)

 

Worsening renal function:

Some rise in urea (BUN), creatinine, and potassium is to be expected after initiation of an ACE inhibitor; if an increase is small and asymptomatic no action is necessary

An increase in creatinine of up to 50% above baseline, or 266µmol/L (3mg/dL), which ever is the smaller, is acceptable

An increase in potassium to <5.5mmol/L is acceptable

If urea, creatinine, or potassium do rise excessively consider stopping concomitant nephrotoxic drugs (e.g. NSAIDs*), other potassium supplements/retaining agents (triamterene, amiloride, spironolactone/eplerenone***) and, if no signs of congestion, reducing the dose of diuretic

If greater rises in creatinine or potassium than those outlined above persist despite adjustment of concomitant medications the dose of the ACE inhibitor should be halved and blood chemistry rechecked within 1–2 weeks; if there is still an unsatisfactory response specialist advice should be sought

If potassium rises to >5.5mmol/L or creatinine increases by >100% or to above 310µmol/L (3.5mg/dL) the ACE inhibitor should be stopped and specialist advice sought

Blood chemistry should be monitored frequently and serially until potassium and creatinine have plateaued

Why?

 

Two major randomized trials (CONSENSUS I and SOLVD-T) and a meta-analysis of smaller trials have conclusively shown that ACE inhibitors increase survival, reduce hospital admissions, and improve NYHA class and quality of life in patients with all grades of symptomatic heart failure. Other major randomized trials in patients with systolic dysfunction after acute myocardial infarction (SAVE, AIRE, TRACE) have shown that ACE inhibitors increase survival. In patients with heart failure (ATLAS), the composite endpoint of death or hospital admission was reduced by higher doses of ACE inhibitor compared to lower doses. ACE inhibitors have also been shown to delay or prevent the development of symptomatic heart failure in patients with asymptomatic LVSD

In whom and when?

Indications:

Potentially all patients with heart failure

First-line treatment (along with beta-blockers) in patients with NYHA Class II–IV HF; start as early as possible in course of disease. ACE inhibitors are also of benefit in patients with asymptomatic LVSD (NYHA class I)

 

Contraindications:

History of angioneurotic oedema

Severe aortic stenosis

Known bilateral renal artery stensis

 

Cautions/seek specialist advice:

Significant hyperkalaemia (K+ >5.0mmol/L)

Significant renal dysfunction (creatinine 221µmol/L or >2.5mg/dL)

Symptomatic or severe asymptomatic hypotension (systolic BP <90mmHg)

 

Drug interactions to look out for:

K+ supplements/K+ sparing diuretics e.g. amiloride and triamterene (beware combination preparations with furosemide).

Aldosterone antagonists (spironolactone and eplerenone)

Aldosterone antagonists (spironolactone, eplerenone), angiotensin receptor blockers, NSAIDS*

‘Low salt’ substitutes with a high K+ content

Where?

 

In the community for most patients

 

Exceptions—see ‘Cautions/specialist advice’

Which ace inhibitor and what dose?

Starting dose (mg)

Target dose (mg)

Captopril

6.25 TID

50 TID

Enalapril

2.5 BID

10–20 BID

Lisinopril

2.5–5.0 OD

20–35 OD

Ramipril

2.5 OD

5 BID or 10 OD

Trandolapril

0.5 OD

4 OD

How to use?

 

Start with a low dose (see above)

 

Double dose at not less than 2-weekly intervals

 

Aim for target dose (see above) or, failing that, the highest tolerated dose

 

Remember some ACE inhibitor is better than no ACE inhibitor

 

Monitor blood pressure and blood chemistry (urea/BUN, creatinine, K+)

 

Check blood chemistry 1–2 weeks after initiation and 1–2 weeks after final dose titration

 

When to stop up-titration/reduce dose/stop treatment—see ‘Problem solving’

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), biochemical monitoring, and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, to prevent worsening of heart failure leading to hospital admission, and to increase survival

Advice to patient?

 

Symptoms improve within a few weeks to a few months of starting treatment

 

Advise patients to report principal adverse effects i.e. dizziness/symptomatic hypotension, cough—see ‘Problem solving’

 

Advise patients to avoid NSAIDs* not prescribed by a physician (self-purchased ‘over-the counter’) and salt substitutes high in K+— see ‘Problem solving’

Problem solving

Asymptomatic low blood pressure:

Does not usually require any change in therapy

 

Symptomatic hypotension:

If dizziness, light-headedness and/or confusion, and a low blood pressure reconsider need for nitrates, calcium-channel blockers**, and other vasodilators

If no signs/symptoms of congestion consider reducing diuretic dose

If these measures do not solve problem seek specialist advice

 

Cough:

Cough is common in patients with heart failure, many of whom have smoking-related lung disease

Cough is also a symptom of pulmonary oedema which should be excluded when a new or worsening cough develops

ACE inhibitor-induced cough rarely requires treatment discontinuation

When a very troublesome cough does develop (e.g. one stopping the patient sleeping) and can be proven to be due to ACE inhibition (i.e. recurs after ACE inhibitor withdrawal and rechallenge) substitution of an ARB should be made (graphic Table 23.9)

 

Worsening renal function:

Some rise in urea (BUN), creatinine, and potassium is to be expected after initiation of an ACE inhibitor; if an increase is small and asymptomatic no action is necessary

An increase in creatinine of up to 50% above baseline, or 266µmol/L (3mg/dL), which ever is the smaller, is acceptable

An increase in potassium to <5.5mmol/L is acceptable

If urea, creatinine, or potassium do rise excessively consider stopping concomitant nephrotoxic drugs (e.g. NSAIDs*), other potassium supplements/retaining agents (triamterene, amiloride, spironolactone/eplerenone***) and, if no signs of congestion, reducing the dose of diuretic

If greater rises in creatinine or potassium than those outlined above persist despite adjustment of concomitant medications the dose of the ACE inhibitor should be halved and blood chemistry rechecked within 1–2 weeks; if there is still an unsatisfactory response specialist advice should be sought

If potassium rises to >5.5mmol/L or creatinine increases by >100% or to above 310µmol/L (3.5mg/dL) the ACE inhibitor should be stopped and specialist advice sought

Blood chemistry should be monitored frequently and serially until potassium and creatinine have plateaued

NB it is very rarely necessary to stop an ACE inhibitor and clinical deterioration is likely if treatment is withdrawn—ideally, specialist advice should be sought before treatment discontinuation.

*

avoid unless essential.; ** calcium-channel blockers should be discontinued unless absolutely essential (e.g. for angina or hypertension).; *** The safety and efficacy of an ACE inhibitor used with an ARB and spironolactone (as well as beta-blocker) is uncertain and the use of all 3 inhibitors of the renin-angiotensin-aldosterone system together is not recommended. ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker; AIRE, Acute Infarction Ramipril Efficacy; ATLAS, Assessment of Treatment with Lisinopril And Survival; BID, twice a day; BUN, blood urea nitrogen; CONSENSUS, Cooperative North Scandinavian Enalapril Survival Study; LVSD, left ventricular systolic dysfunction; OD, once a day; SOLVD-T, Studies of Left Ventricular Dysfunction Treatment; SAVE, Survival and Ventricular Enlargement; TID, three times a day; TRACE, TRAndolapril Cardiac Evaluation.

Adapted with permission from McMurray J, Cohen-Solal A, Dietz R, et al. Practical recommendations for the use of ACE inhibitors, beta-blockers, aldosterone antagonists and angiotensin receptor blockers in heart failure: putting guidelines into practice. Eur J Heart Fail 2005; 7: 710–21.

Instead of inhibiting the production of angiotensin II through ACE, ARBs block the binding of angiotensin II to the AT1R. This distinct mechanism of action may be important because angiotensin II is also believed to be produced by other enzymes such as chymase. ARBs do not inhibit kininase II or the breakdown of bradykinin, so they do not cause cough or angio-oedema.

When used as the sole RAAS blocking agent in addition to a diuretic and digoxin, ARBs produce similar benefits to ACE inhibitors and can be substituted for them in patients who have cough or angio-oedema with ACE inhibitors [223, 224]. When used in clinically-effective doses, other adverse effects such as hypotension, renal dysfunction, and hyperkalaemia are encountered as frequently as with an ACE inhibitor. As with an ACE inhibitor, the specific agents, dosing regimens, and target doses that were of demonstrable benefit in clinical trials are recommended (graphic Table 23.7).

An ARB used in combination with an ACE inhibitor (and beta-blocker) further improves LVEF, relieves symptoms, reduces the risk of hospital admission for worsening heart failure, and can also reduce the risk of cardiovascular death (graphic Table 23.6) [225–228]. Consequently, the addition of an ARB to both an ACE inhibitor and a beta-blocker should be considered in any patient with persisting symptoms (NYHA class II–IV). There is, however, also strong evidence that adding an aldosterone antagonist to an ACE inhibitor is of benefit in patients with advanced (NYHA class III–IV) heart failure (see graphic Aldosterone antagonists, Clinical benefits, p.874), but the efficacy and safety of the four-drug combination of an ACE inhibitor, beta-blocker, ARB, and aldosterone antagonist are uncertain [218, 229]. Consequently, either an ARB or an aldosterone antagonist, but not both, should be added to an ACE inhibitor and a beta-blocker in such patients.

The approach to initiation, titration, and monitoring of an ARB is similar to an ACE inhibitor (graphic Table 23.9). The adverse effects, with the exception of cough and angio-oedema, are similar. Use of multiple inhibitors of the RAAS requires even more diligent monitoring, especially in patients at higher risk of uraemia, hypotension, or hyperkalaemia (i.e. patients ≥75 years of age or with a systolic blood pressure <100mmHg, diabetes, or renal impairment) [219, 230, 231].

Table 23.9
Practical guidance on the use of ARBs in patients with heart failure due to left ventricular systolic dysfunction

Why?

 

When added to standard therapy, including an ACE inhibitor, in patients with all grades of symptomatic heart failure, the ARBs valsartan and candesartan have been shown, in two major randomized trials (Val-HeFT and CHARM), to reduce heart failure hospital admissions, improve NYHA class, and maintain quality of life. The two CHARM low LVEF trials (CHARM Alternative and CHARM-Added) also showed that candesartan reduced all-cause mortality. In patients previously intolerant of an ACE inhibitor, candesartan has been shown to reduce the risk of the composite outcome of cardiovascular death or heart failure hospitalization, the risk of heart failure hospital admission, and to improve NYHA class. These findings in heart failure are supported by another randomized trial in patients with left ventricular systolic dysfunction, heart failure, or both complicating acute myocardial infarction (VALIANT) in which valsartan was as effective as the ACE inhibitor captopril in reducing mortality and cardiovascular morbidity.

In whom and when?

Indications:

Potentially all patients with heart failure

First-line treatment (along with beta-blockers) in patients with NYHA class II–IV HF intolerant of an ACE inhibitor

Second-line treatment (after optimization of ACE inhibitor and beta-blocker*) in patients with NYHA class II–IV heart failure

 

Contraindications:

Known bilateral renal artery stenosis

Severe aortic stenosis

Cautions/seek specialist advice:

Significant hyperkalemia (K+ >5.0mmol/L)

Significant renal dysfunction (creatinine 221µmol/L or >2.5mg/dL)

Symptomatic or severe asymptomatic hypotension (systolic BP <90mmHg)

 

Drug interactions to look out for:

K+ supplements/K+-sparing diuretics, e.g. amiloride and triamterene (beware combination preparations with furosemide)

Aldosterone antagonists (spironolactone and eplerenone), ACE inhibitors, NSAIDS**

‘Low salt’ substitutes with a high K+ content

Where?

 

In the community for most patients

 

Exceptions—see ‘Cautions/specialist advice’

Which ARB and what dose?

Starting dose (mg)

Target dose (mg)

Candesartan

4 or 8mg OD

32mg OD

Valsartan

40mg BID

160mg BID

How to use?

 

Start with a low dose (see above)

 

Double dose at not less than 2-weekly intervals

 

Aim for target dose (see above) or, failing that, the highest tolerated dose

 

Remember some ARB is better than no ARB

 

Monitor blood pressure and blood chemistry (urea/BUN, creatinine, K+)

 

Check blood chemistry 1–2 weeks after initiation and 1–2 weeks after final dose titration

 

When to stop up-titration/reduce dose/stop treatment—see ‘Problem solving’

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), biochemical monitoring, and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, prevent worsening of heart failure leading to hospital admission, and to increase survival

 

Symptoms improve within a few weeks to a few months of starting treatment

 

Advise patients to principal adverse effect, i.e. report dizziness/ symptomatic hypotension—see ‘Problem solving’

 

Advise patients to avoid NSAIDs** not prescribed by a physician (self-purchased ‘over-the-counter’) and salt substitutes high in K+—see ‘Problem solving’

Problem solving

Asymptomatic low blood pressure:

Does not usually require any change in therapy

 

Symptomatic hypotension:

If dizziness, light-headedness and/or confusion, and a low blood pressure reconsider need for nitrates, calcium-channel blockers***, and other vasodilators

If no signs/symptoms of congestion consider reducing diuretic dose

If these measures do not solve problem, seek specialist advice

 

Worsening renal function:

Some rise in urea (BUN), creatinine and potassium is to be expected after initiation of an ARB; if the increase is small

and asymptomatic no action is necessary

An increase in creatinine of up to 50% above baseline, or 266µmol/L (3mg/dL), which ever is the smaller, is acceptable

An increase in potassium to < 5.5mmol/L is acceptable

If urea, creatinine, or potassium do rise excessively consider stopping concomitant nephrotoxic drugs (e.g. NSAIDs**), potassium supplements/retaining agents (triamterene, amiloride, spironolactone/eplerenone*), and, if no signs of congestion, reducing the dose of diuretic

If greater rises in creatinine or potassium than those outlined above persist despite adjustment of concomitant medications, the dose of the ARB should be halved and blood chemistry rechecked within 1–2 weeks; if there is still an unsatisfactory response specialist advice should be sought.

If potassium rises to >5.5mmol/L or creatinine increases by >100% or to above 310µmol/L (3.5mg/dL) the ARB should be stopped and specialist advice sought

Blood chemistry should be monitored frequently and serially until potassium and creatinine have plateaued

Why?

 

When added to standard therapy, including an ACE inhibitor, in patients with all grades of symptomatic heart failure, the ARBs valsartan and candesartan have been shown, in two major randomized trials (Val-HeFT and CHARM), to reduce heart failure hospital admissions, improve NYHA class, and maintain quality of life. The two CHARM low LVEF trials (CHARM Alternative and CHARM-Added) also showed that candesartan reduced all-cause mortality. In patients previously intolerant of an ACE inhibitor, candesartan has been shown to reduce the risk of the composite outcome of cardiovascular death or heart failure hospitalization, the risk of heart failure hospital admission, and to improve NYHA class. These findings in heart failure are supported by another randomized trial in patients with left ventricular systolic dysfunction, heart failure, or both complicating acute myocardial infarction (VALIANT) in which valsartan was as effective as the ACE inhibitor captopril in reducing mortality and cardiovascular morbidity.

In whom and when?

Indications:

Potentially all patients with heart failure

First-line treatment (along with beta-blockers) in patients with NYHA class II–IV HF intolerant of an ACE inhibitor

Second-line treatment (after optimization of ACE inhibitor and beta-blocker*) in patients with NYHA class II–IV heart failure

 

Contraindications:

Known bilateral renal artery stenosis

Severe aortic stenosis

Cautions/seek specialist advice:

Significant hyperkalemia (K+ >5.0mmol/L)

Significant renal dysfunction (creatinine 221µmol/L or >2.5mg/dL)

Symptomatic or severe asymptomatic hypotension (systolic BP <90mmHg)

 

Drug interactions to look out for:

K+ supplements/K+-sparing diuretics, e.g. amiloride and triamterene (beware combination preparations with furosemide)

Aldosterone antagonists (spironolactone and eplerenone), ACE inhibitors, NSAIDS**

‘Low salt’ substitutes with a high K+ content

Where?

 

In the community for most patients

 

Exceptions—see ‘Cautions/specialist advice’

Which ARB and what dose?

Starting dose (mg)

Target dose (mg)

Candesartan

4 or 8mg OD

32mg OD

Valsartan

40mg BID

160mg BID

How to use?

 

Start with a low dose (see above)

 

Double dose at not less than 2-weekly intervals

 

Aim for target dose (see above) or, failing that, the highest tolerated dose

 

Remember some ARB is better than no ARB

 

Monitor blood pressure and blood chemistry (urea/BUN, creatinine, K+)

 

Check blood chemistry 1–2 weeks after initiation and 1–2 weeks after final dose titration

 

When to stop up-titration/reduce dose/stop treatment—see ‘Problem solving’

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), biochemical monitoring, and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, prevent worsening of heart failure leading to hospital admission, and to increase survival

 

Symptoms improve within a few weeks to a few months of starting treatment

 

Advise patients to principal adverse effect, i.e. report dizziness/ symptomatic hypotension—see ‘Problem solving’

 

Advise patients to avoid NSAIDs** not prescribed by a physician (self-purchased ‘over-the-counter’) and salt substitutes high in K+—see ‘Problem solving’

Problem solving

Asymptomatic low blood pressure:

Does not usually require any change in therapy

 

Symptomatic hypotension:

If dizziness, light-headedness and/or confusion, and a low blood pressure reconsider need for nitrates, calcium-channel blockers***, and other vasodilators

If no signs/symptoms of congestion consider reducing diuretic dose

If these measures do not solve problem, seek specialist advice

 

Worsening renal function:

Some rise in urea (BUN), creatinine and potassium is to be expected after initiation of an ARB; if the increase is small

and asymptomatic no action is necessary

An increase in creatinine of up to 50% above baseline, or 266µmol/L (3mg/dL), which ever is the smaller, is acceptable

An increase in potassium to < 5.5mmol/L is acceptable

If urea, creatinine, or potassium do rise excessively consider stopping concomitant nephrotoxic drugs (e.g. NSAIDs**), potassium supplements/retaining agents (triamterene, amiloride, spironolactone/eplerenone*), and, if no signs of congestion, reducing the dose of diuretic

If greater rises in creatinine or potassium than those outlined above persist despite adjustment of concomitant medications, the dose of the ARB should be halved and blood chemistry rechecked within 1–2 weeks; if there is still an unsatisfactory response specialist advice should be sought.

If potassium rises to >5.5mmol/L or creatinine increases by >100% or to above 310µmol/L (3.5mg/dL) the ARB should be stopped and specialist advice sought

Blood chemistry should be monitored frequently and serially until potassium and creatinine have plateaued

NB it is very rarely necessary to stop an ARB and clinical deterioration is likely if treatment is withdrawn—ideally, specialist advice should be sought before treatment discontinuation.

*

The safety and efficacy of an ARB used with an ACE inhibitor and spironolactone (as well as beta-blocker) is uncertain and the use of all three inhibitors of the RAAS together is not recommended.; ** avoid unless essential.; *** calcium-channel blockers should be discontinued unless absolutely essential (e.g. for angina or hypertension). ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker; BID, twice a day; BUN, blood urea nitrogen; CHARM, Candesartan in Heart failure: Assessment of Reduction in Mortality and Morbidity; LVEF, left ventricular ejection fraction; OD, once a day; Val-HeFT, Valsartan Heart Failure Trial; VALIANT, VALsartan In Acute myocardial iNfarcTion.

Adapted with permission from McMurray J, Cohen-Solal A, Dietz R, et al. Practical recommendations for the use of ACE inhibitors, beta-blockers, aldosterone antagonists and angiotensin receptor blockers in heart failure: putting guidelines into practice. Eur J Heart Fail 2005; 7: 710–21.

As with ACE inhibitors, beta-blockers and aldosterone antagonists, treatment with ARBs should be indefinite unless there is intolerance.

Beta-blockers are believed to counteract the many harmful effects of sympathetic nervous system hyperactivity described earlier—see graphic Pathophysiology, p.843.

The long-term addition of a beta-blocker to an ACE inhibitor (and diuretic and digoxin) further improves left ventricular function and symptoms, reduces hospital admissions, and improves survival, strikingly (graphic Table 23.6; Fig. 23.30). Consequently, a beta-blocker is recommended for all patients with symptomatic systolic dysfunction, irrespective of aetiology and severity. The combination of a beta-blocker with an ACE inhibitor is now the cornerstone of the treatment of symptomatic heart failure (graphic Fig. 23.28) [232–239].

 Kaplan–Meier curves of the three major
survival studies with beta-blocker therapy: (A) Metoprolol CR/XL Randomized
Intervention Trial (MERIT-HF); (B) Cardiac Insufficiency Bisoprolol Study II
(CIBIS-II); (C) Carvedilol Prospective Randomized Cumulative Survival
(COPERNICUS) study. (D) SENIORS (Study of the Effects of Nebivolol on
Outcomes and Rehospitalisation in Seniors with heart failure). Reproduced
with permission from MERIT-HF Study Group Effect of metoprolol CR/XL in
chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in
Congestive Heart Failure (MERIT-HF). Lancet 1999; 353: 2001–7;
CIBIS-II Investigators and Committees. The Cardiac Insufficiency Bisoprolol
Study II (CIBIS-II): a randomised trial. Lancet 1999; 353:
9–13; Packer,M, Coats AJ, Fowler MB et al.; Carvedilol Prospective
Randomized Cumulative Survival Study Group. Effect of carvedilol on survival
in severe chronic heart failure. N Engl J Med 2001; 344:
1651–8.
Figure 23.30

Kaplan–Meier curves of the three major survival studies with beta-blocker therapy: (A) Metoprolol CR/XL Randomized Intervention Trial (MERIT-HF); (B) Cardiac Insufficiency Bisoprolol Study II (CIBIS-II); (C) Carvedilol Prospective Randomized Cumulative Survival (COPERNICUS) study. (D) SENIORS (Study of the Effects of Nebivolol on Outcomes and Rehospitalisation in Seniors with heart failure). Reproduced with permission from MERIT-HF Study Group Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 1999; 353: 2001–7; CIBIS-II Investigators and Committees. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet 1999; 353: 9–13; Packer,M, Coats AJ, Fowler MB et al.; Carvedilol Prospective Randomized Cumulative Survival Study Group. Effect of carvedilol on survival in severe chronic heart failure. N Engl J Med 2001; 344: 1651–8.

The major contraindications to using a beta-blocker in heart failure are asthma (though it is important to note that the dyspnoea caused by pulmonary congestion can be confused with reactive airway disease) and second- or third-degree atrioventricular block [219]. Initiation of treatment during an episode of acute decompensated heart failure should also generally be deferred until the patient is stabilized and recovering (but, ideally, treatment should be commenced before discharge). In addition, caution is advised in patients with a heart rate <60 beats per minute (bpm) or a systolic blood pressure <100mmHg. It is recommended that a beta-blocker shown to produce benefits in a randomized trial be used (graphic Table 23.7).

As with ACE inhibitors, beta-blockers should be introduced as early as possible in a patient’s treatment, started in a low dose (graphic Table 23.7) and increased gradually toward the target dose used in a clinical trial (the ‘start low–go slow’ approach). The patient should be checked for symptomatic hypotension and excessive bradycardia after each dose increment, but both of these side effects are uncommon, and hypotension can often be resolved by reduction in the dose of other non-essential blood pressure-lowering medications, e.g. nitrates and calcium-channel blockers (graphic Table 23.10). Bradycardia is more likely in patients who are also taking digoxin, ivabradine, or amiodarone and the necessity for the simultaneous use of these agents should be reviewed if excessive bradycardia occurs. Occasionally, symptomatic worsening and fluid retention (i.e. weight gain or oedema) may occur after initiation of a beta-blocker or during dose up-titration; these side effects usually can be resolved by a temporary increase in the diuretic dose without necessitating discontinuation of the beta-blocker.

Table 23.10
Practical guidance on the use of beta-blockers in patients with heart failure due to left ventricular systolic dysfunction

Why?

 

Several major randomized controlled trials (i.e. USCP, CIBIS II, MERIT-HF, COPERNICUS) have shown, conclusively, that certain beta-blockers increase survival, reduce hospital admissions, and improve NYHA class and quality of life when added to standard therapy (diuretics, digoxin, and ACE inhibitors) in patients with stable mild and moderate heart failure and in some patients with severe heart failure. In the SENIORS trial which differed substantially in design from the aforementioned studies (older patients, some patients with preserved left ventricular systolic function, longer follow-up), nebivolol appeared to have a smaller treatment effect, though direct comparison is difficult. One other trial (BEST) did not show a reduction in all cause mortality but did report a reduction in cardiovascular mortality and is otherwise broadly consistent with the aforementioned studies. The COMET trial showed that carvedilol was substantially more effective than a low dose of short-acting metoprolol tartrate* (long acting metoprolol succinate was used in MERIT-HF).

In whom and when?

 

Indications:

Potentially all patients with stable mild and moderate heart failure; patients with severe heart failure should be referred for specialist advice

First-line treatment (along with ACE inhibitors) in patients with stable NYHA class II–III heart failure; start as early as possible in course of disease

 

Contraindications:

Asthma

Second- or third-degree atrioventricular block

 

Cautions/seek specialist advice:

Severe (NYHA class IV) heart failure

Current or recent (<4 weeks) exacerbation of heart failure, e.g. hospital admission with worsening heart failure

Heart block or heart rate <60bpm.

Persisting signs of congestion, hypotension/low blood pressure (systolic <90mmHg), raised jugular venous pressure, ascites, marked peripheral oedema

 

Drug interactions to look out for:

Verapamil/diltiazem (should be discontinued)**

Digoxin, amiodarone

Where?

 

In the community in stable patients (NYHA class IV/severe heart failure patients should be referred for specialist advice)

 

Not in unstable patients hospitalised with worsening heart failure

 

Other exceptions—see ‘Cautions/seek specialist advice’

Which beta-blocker and what dose?

Starting dose (mg)

Target dose (mg)

Bisoprolol

1.25 OD

10 OD

Carvedilol

3.125 BID

12.5–25 OD

Metoprolol CR/XL

25–50 BID

200 OD*

Nebivolol

1.25 OD

10 OD

How to use?

 

Start with a low dose (see above)

 

Double dose at not less than 2-weekly intervals

 

Aim for target dose (see above) or, failing that, the highest tolerated dose

 

Remember some beta-blocker is better than no beta-blocker

 

Monitor heart rate, blood pressure, clinical status (symptoms, signs—especially signs of congestion, body weight)

 

Check blood chemistry 1–2 weeks after initiation and 1–2 weeks after final dose titration

 

When to stop up-titration/reduce dose/stop treatment—see ‘Problem solving’

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, prevent worsening of heart failure leading to hospital admission, and to increase survival

 

Symptomatic improvement may develop slowly after starting treatment, taking 3–6 months or longer

 

Temporary symptomatic deterioration may occur during initiation/up-titration phase; in long-term beta-blockers improve well-being

 

Advise patient to report deterioration (see ‘Problem solving’) and that deterioration (tiredness, fatigue, breathlessness) can usually be easily managed by adjustment of other medication; patients should be advised not to stop beta-blocker therapy without consulting their physician

 

To detect and treat deterioration early, patients should be encouraged to weigh themselves daily (after waking, before dressing, after voiding, before eating) and to increase their diuretic dose should their weight increase, persistently (>2 days), by >1.5–2.0kg***

Problem solving

  

Worsening symptoms/signs (e.g. increasing dyspnoea, fatigue, oedema, weight gain):

If increasing congestion increase dose of diuretic and/or halve dose of beta-blocker (if increasing diuretic doesn’t work)

If marked fatigue (and/or bradycardia—see below) halve dose of beta-blocker (rarely necessary)

Review patient in 1–2 weeks; if not improved seek specialist advice

If serious deterioration halve dose of beta-blocker or stop this treatment (rarely necessary); seek specialist advice

 

Low heart rate:

If <50bpm and worsening symptoms—halve dose beta-blocker or, if severe deterioration, stop beta-blocker (rarely necessary)

Review need for other heart rate slowing drugs, e.g. digoxin, amiodarone, diltiazem/verapamil**

Arrange ECG to exclude heart block

Seek specialist advice

  

Asymptomatic low blood pressure:

Does not usually require any change in therapy

 

Symptomatic hypotension:

If dizziness, light-headedness and/or confusion, and a low blood pressure reconsider need for nitrates, calcium-channel blockers** and other vasodilators

If no signs/symptoms of congestion consider reducing diuretic dose or ACE inhibitor

If these measures do not solve problem seek specialist advice

  

Why?

 

Several major randomized controlled trials (i.e. USCP, CIBIS II, MERIT-HF, COPERNICUS) have shown, conclusively, that certain beta-blockers increase survival, reduce hospital admissions, and improve NYHA class and quality of life when added to standard therapy (diuretics, digoxin, and ACE inhibitors) in patients with stable mild and moderate heart failure and in some patients with severe heart failure. In the SENIORS trial which differed substantially in design from the aforementioned studies (older patients, some patients with preserved left ventricular systolic function, longer follow-up), nebivolol appeared to have a smaller treatment effect, though direct comparison is difficult. One other trial (BEST) did not show a reduction in all cause mortality but did report a reduction in cardiovascular mortality and is otherwise broadly consistent with the aforementioned studies. The COMET trial showed that carvedilol was substantially more effective than a low dose of short-acting metoprolol tartrate* (long acting metoprolol succinate was used in MERIT-HF).

In whom and when?

 

Indications:

Potentially all patients with stable mild and moderate heart failure; patients with severe heart failure should be referred for specialist advice

First-line treatment (along with ACE inhibitors) in patients with stable NYHA class II–III heart failure; start as early as possible in course of disease

 

Contraindications:

Asthma

Second- or third-degree atrioventricular block

 

Cautions/seek specialist advice:

Severe (NYHA class IV) heart failure

Current or recent (<4 weeks) exacerbation of heart failure, e.g. hospital admission with worsening heart failure

Heart block or heart rate <60bpm.

Persisting signs of congestion, hypotension/low blood pressure (systolic <90mmHg), raised jugular venous pressure, ascites, marked peripheral oedema

 

Drug interactions to look out for:

Verapamil/diltiazem (should be discontinued)**

Digoxin, amiodarone

Where?

 

In the community in stable patients (NYHA class IV/severe heart failure patients should be referred for specialist advice)

 

Not in unstable patients hospitalised with worsening heart failure

 

Other exceptions—see ‘Cautions/seek specialist advice’

Which beta-blocker and what dose?

Starting dose (mg)

Target dose (mg)

Bisoprolol

1.25 OD

10 OD

Carvedilol

3.125 BID

12.5–25 OD

Metoprolol CR/XL

25–50 BID

200 OD*

Nebivolol

1.25 OD

10 OD

How to use?

 

Start with a low dose (see above)

 

Double dose at not less than 2-weekly intervals

 

Aim for target dose (see above) or, failing that, the highest tolerated dose

 

Remember some beta-blocker is better than no beta-blocker

 

Monitor heart rate, blood pressure, clinical status (symptoms, signs—especially signs of congestion, body weight)

 

Check blood chemistry 1–2 weeks after initiation and 1–2 weeks after final dose titration

 

When to stop up-titration/reduce dose/stop treatment—see ‘Problem solving’

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, prevent worsening of heart failure leading to hospital admission, and to increase survival

 

Symptomatic improvement may develop slowly after starting treatment, taking 3–6 months or longer

 

Temporary symptomatic deterioration may occur during initiation/up-titration phase; in long-term beta-blockers improve well-being

 

Advise patient to report deterioration (see ‘Problem solving’) and that deterioration (tiredness, fatigue, breathlessness) can usually be easily managed by adjustment of other medication; patients should be advised not to stop beta-blocker therapy without consulting their physician

 

To detect and treat deterioration early, patients should be encouraged to weigh themselves daily (after waking, before dressing, after voiding, before eating) and to increase their diuretic dose should their weight increase, persistently (>2 days), by >1.5–2.0kg***

Problem solving

  

Worsening symptoms/signs (e.g. increasing dyspnoea, fatigue, oedema, weight gain):

If increasing congestion increase dose of diuretic and/or halve dose of beta-blocker (if increasing diuretic doesn’t work)

If marked fatigue (and/or bradycardia—see below) halve dose of beta-blocker (rarely necessary)

Review patient in 1–2 weeks; if not improved seek specialist advice

If serious deterioration halve dose of beta-blocker or stop this treatment (rarely necessary); seek specialist advice

 

Low heart rate:

If <50bpm and worsening symptoms—halve dose beta-blocker or, if severe deterioration, stop beta-blocker (rarely necessary)

Review need for other heart rate slowing drugs, e.g. digoxin, amiodarone, diltiazem/verapamil**

Arrange ECG to exclude heart block

Seek specialist advice

  

Asymptomatic low blood pressure:

Does not usually require any change in therapy

 

Symptomatic hypotension:

If dizziness, light-headedness and/or confusion, and a low blood pressure reconsider need for nitrates, calcium-channel blockers** and other vasodilators

If no signs/symptoms of congestion consider reducing diuretic dose or ACE inhibitor

If these measures do not solve problem seek specialist advice

  

NB beta-blockers should not be stopped suddenly unless absolutely necessary (there is a risk of a ‘rebound’ increase in myocardial ischaemia/infarction and arrhythmias)—ideally specialist advice should be sought before treatment discontinuation.

*

metoprolol tartrate should not be used in preference to an evidence-based beta-blocker in heart failure.; ** calcium-channel blockers should be discontinued unless absolutely necessary and diltiazem and verapamil are generally contraindicated in heart failure.; *** this is generally good advice for all patients with heart failure. ACE, angiotensin-converting enzyme; BD, twice a day; BEST, Beta-Blocker Evaluation Survival Trial; CIBIS, Cardiac Insufficiency Bisoprolol Study; COMET, Carvedilol or Metoprolol European Trial; COPERNICUS, Carvedilol Prospective Randomized Cumulative Survival; MERIT-HF, Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure; OD, once a day; SENIORS, Study of the Effects of Nebivolol on Outcomes and Rehospitalisation in Seniors with heart failure; USCP, US Carvedilol heart failure Program.

Adapted with permission from McMurray J, Cohen-Solal A, Dietz R, et al. Practical recommendations for the use of ACE inhibitors, beta-blockers, aldosterone antagonists and angiotensin receptor blockers in heart failure: putting guidelines into practice. Eur J Heart Fail 2005; 7: 710–21.

Treatment with a beta-blocker should be given for life, though the dose may need to be decreased or temporarily discontinued during episodes of acute decompensation if the patient shows signs of circulatory hypoperfusion or refractory congestion.

These agents block the undesirable actions of aldosterone described earlier and act as potassium sparing diuretics.

The aldosterone antagonist spironolactone (graphic Table 23.6) improves symptoms, reduces hospital admissions, and increases survival when added to an ACE inhibitor (and diuretics and digoxin) in patients with a reduced LVEF and severely symptomatic heart failure (graphic Fig. 23.31) [218]. Eplerenone, another aldosterone antagonist, reduces mortality and morbidity when added to both an ACE inhibitor and beta-blocker in patients with a reduced LVEF and heart failure or diabetes after a recent myocardial infarction (graphic Table 23.7). Consequently, an aldosterone antagonist should be considered in patients who remain in severe heart failure (NYHA class III or IV) despite treatment with a diuretic, ACE inhibitor (or ARB), and beta-blocker. When begun, it should be given indefinitely. The value of an aldosterone antagonist in patients with milder heart failure is uncertain but is under investigation. The combination of an ACE inhibitor, an ARB, and an aldosterone antagonist has not been adequately evaluated and is not recommended  .

 Effect of spironolactone in severe
chronic heart failure: findings from RALES (Randomized Aldactone Evaluation
Study). Risk reduction 0.70; 95% CI 0.62–0.80; P < 0.001.
Reproduced with permission from Pitt B, Zannad F, Remme WJ, et al.
The effect of spironolactone on morbidity and mortality in patients with
severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 1999; 341: 709–17.
Figure 23.31

Effect of spironolactone in severe chronic heart failure: findings from RALES (Randomized Aldactone Evaluation Study). Risk reduction 0.70; 95% CI 0.62–0.80; P < 0.001. Reproduced with permission from Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 1999; 341: 709–17.

Treatment with an aldosterone antagonist should be initiated with a low dose (graphic Table 23.7) with careful monitoring of serum electrolytes and renal function (graphic Table 23.11). Hyperkalaemia and uraemia are the adverse effects of greatest concern (as with ACE inhibitors and ARBs) and an aldosterone antagonist should not be given to patients with a serum potassium concentration of >5mmol/L, serum creatinine >221µmol/L (>2.5mg/dL) or other evidence of markedly impaired renal function. The importance of patient selection and dose are underscored by reports of a worrisome incidence of serious hyperkalaemia in community practice settings. Spironolactone can have antiandrogenic effects, especially painful gynaecomastia, in men. Since eplerenone does not block the androgen receptor, it is a reasonable substitute in patients who experience this adverse effect [240].

Table 23.11
Practical guidance on the use of spironolactone in patients with heart failure due to left ventricular systolic dysfunction

Why?

 

The RALES study showed that low-dose spironolactone increased survival, reduced hospital admissions, and improved NYHA class when added to standard therapy (diuretic, digoxin, ACE inhibitor, and, in a minority of cases, a beta-blocker) in patients with severe (NYHA class III or IV) heart failure. These findings in heart failure are supported by another randomized trial in patients with left ventricular systolic dysfunction and heart failure (or diabetes) complicating acute myocardial infarction (EPHESUS) in which another aldosterone antagonist, eplerenone, increased survival and reduced hospital admissions for cardiac causes.

In whom and when?

Indications:

Potentially all patients with symptomatically moderately severe or severe HF (NYHA class III/IV)

Second-line therapy (after ACE inhibitors and beta-blockers*) in patients with NYHA class III–IV heart failure; there is no evidence of benefit in patients with milder heart failure

 

Cautions/seek specialist advice:

Significant hyperkalaemia (K+ >5.0mmol/L)**

Significant renal dysfunction (creatinine >221µmol/L or 2.5mg/dL)**

 

Drug interactions to look out for:

K+ supplements/K+-sparing diuretics, e.g. amiloride and triamterene (beware combination preparations with furosemide)

Aldosterone antagonists (spironolactone and eplerenone), ACE inhibitors, ARBs, NSAIDS***

‘Low salt’ substitutes with a high K+ content

Where?

 

In the community or in hospital

 

Exceptions—see ‘Cautions/seek specialist advice’

Which dose?**

Starting dose (mg)

Target dose (mg)

Spironolactone

25 OD or on alternate days

25–50 OD

Eplerenone

25 OD

50 OD

How to use?

 

Start with a low dose (see above)

 

Check blood chemistry at 1, 4, 8, and 12 weeks; 6, 9, and 12 months; 6-monthly thereafter

 

If K+ rises above 5.5mmol/L or creatinine rises to 221µmol/L (2.5mg/dL) reduce dose to 25mg on alternate days and monitor blood chemistry closely

 

If K+ rises to >6.0mmol/L or creatinine to > 310μmol/L (3.5mg/dL) stop spironolactone immediately and seek specialist advice

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), biochemical monitoring, and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, prevent worsening of heart failure leading to hospital admission and to increase survival

 

Symptom improvement occurs within a few weeks to a few months of starting treatment

 

Avoid NSAIDs***not prescribed by a physician (self-purchased ‘over-the-counter’) and salt substitutes high in K+

 

If diarrhoea and/or vomiting occurs patients should stop spironolactone and contact their physician

Problem solving

 

Worsening renal function/hyperkalaemia:

 

See ‘How to use?’

 

Major concern is hyperkalaemia (>6.0mmol/L); although this was uncommon in RALES it has been seen more commonly in clinical practice: conversely, a high normal potassium may be desirable in heart failure patients, especially if taking digoxin

 

It is important to avoid other K+-retaining drugs (e.g. K+-sparing diuretics such as amiloride and triamterene) and nephrotoxic agents (e.g. NSAIDs***)

 

The risk of hyperkalaemia and renal dysfunction when an aldosterone antagonist is given to patients already taking an ACE inhibitor and ARB is higher than when an aldosterone is added to just an ACE inhibitor or ARB given singly; close and careful monitoring is mandatory*

Some ‘low salt’ substitutes have a high K+ content

 

Male patients treated with spironolactone may develop breast discomfort and/or gynaecomastia (these problems are significantly less common with eplerenone)

Why?

 

The RALES study showed that low-dose spironolactone increased survival, reduced hospital admissions, and improved NYHA class when added to standard therapy (diuretic, digoxin, ACE inhibitor, and, in a minority of cases, a beta-blocker) in patients with severe (NYHA class III or IV) heart failure. These findings in heart failure are supported by another randomized trial in patients with left ventricular systolic dysfunction and heart failure (or diabetes) complicating acute myocardial infarction (EPHESUS) in which another aldosterone antagonist, eplerenone, increased survival and reduced hospital admissions for cardiac causes.

In whom and when?

Indications:

Potentially all patients with symptomatically moderately severe or severe HF (NYHA class III/IV)

Second-line therapy (after ACE inhibitors and beta-blockers*) in patients with NYHA class III–IV heart failure; there is no evidence of benefit in patients with milder heart failure

 

Cautions/seek specialist advice:

Significant hyperkalaemia (K+ >5.0mmol/L)**

Significant renal dysfunction (creatinine >221µmol/L or 2.5mg/dL)**

 

Drug interactions to look out for:

K+ supplements/K+-sparing diuretics, e.g. amiloride and triamterene (beware combination preparations with furosemide)

Aldosterone antagonists (spironolactone and eplerenone), ACE inhibitors, ARBs, NSAIDS***

‘Low salt’ substitutes with a high K+ content

Where?

 

In the community or in hospital

 

Exceptions—see ‘Cautions/seek specialist advice’

Which dose?**

Starting dose (mg)

Target dose (mg)

Spironolactone

25 OD or on alternate days

25–50 OD

Eplerenone

25 OD

50 OD

How to use?

 

Start with a low dose (see above)

 

Check blood chemistry at 1, 4, 8, and 12 weeks; 6, 9, and 12 months; 6-monthly thereafter

 

If K+ rises above 5.5mmol/L or creatinine rises to 221µmol/L (2.5mg/dL) reduce dose to 25mg on alternate days and monitor blood chemistry closely

 

If K+ rises to >6.0mmol/L or creatinine to > 310μmol/L (3.5mg/dL) stop spironolactone immediately and seek specialist advice

 

A specialist heart failure nurse may assist with patient education, follow-up (in person/by telephone), biochemical monitoring, and dose up-titration

Advice to patient?

 

Explain expected benefits (see ‘Why?’)

 

Treatment is given to improve symptoms, prevent worsening of heart failure leading to hospital admission and to increase survival

 

Symptom improvement occurs within a few weeks to a few months of starting treatment

 

Avoid NSAIDs***not prescribed by a physician (self-purchased ‘over-the-counter’) and salt substitutes high in K+

 

If diarrhoea and/or vomiting occurs patients should stop spironolactone and contact their physician

Problem solving

 

Worsening renal function/hyperkalaemia:

 

See ‘How to use?’

 

Major concern is hyperkalaemia (>6.0mmol/L); although this was uncommon in RALES it has been seen more commonly in clinical practice: conversely, a high normal potassium may be desirable in heart failure patients, especially if taking digoxin

 

It is important to avoid other K+-retaining drugs (e.g. K+-sparing diuretics such as amiloride and triamterene) and nephrotoxic agents (e.g. NSAIDs***)

 

The risk of hyperkalaemia and renal dysfunction when an aldosterone antagonist is given to patients already taking an ACE inhibitor and ARB is higher than when an aldosterone is added to just an ACE inhibitor or ARB given singly; close and careful monitoring is mandatory*

Some ‘low salt’ substitutes have a high K+ content

 

Male patients treated with spironolactone may develop breast discomfort and/or gynaecomastia (these problems are significantly less common with eplerenone)

*

the safety and efficacy of spironolactone used with an ACE inhibitor and an ARB (as well as beta-blocker) is uncertain and the use of all 3 inhibitors of the RAAS together is not recommended.; ** it is extremely important that these cautions and doses are adhered to in the light of recent evidence of serious hyperkalaemia with spironolactone in usual clinical practice in Ontario, Canada.; *** avoid unless essential. ACE, angiotensin-converting enzyme; ARB, angiotensin receptor blocker; EPHESUS, Epleronone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study; OD, once a day; RALES, Randomized Aldactone Evaluation Study.

Adapted with permission from McMurray J, Cohen-Solal A, Dietz R, et al. Practical recommendations for the use of ACE inhibitors, beta-blockers, aldosterone antagonists and angiotensin receptor blockers in heart failure: putting guidelines into practice. Eur J Heart Fail 2005; 7: 710–21.

Digitalis glycosides inhibit the cell membrane sodium–potassium adenosine triphosphatase (ATP) pump, thereby increasing intracellular calcium and myocardial contractility. In addition, digoxin is thought to enhance parasympathetic and reduce sympathetic nervous activity, as well as inhibit renin release [241].

Only one large randomized placebo-controlled trial examined the effects of starting (as opposed to withdrawing) digoxin on mortality and morbidity in patients with heart failure in sinus rhythm [241, 242]. In that trial, digoxin did not reduce mortality but did decrease the risk of admission to hospital for worsening heart failure when added to a diuretic and an ACE inhibitor (graphic Table 23.6). In patients in sinus rhythm, addition of digoxin is recommended only for patients whose heart failure remains symptomatic despite standard three-drug treatment with a diuretic, ACE inhibitor, beta-blocker, and either an ARB or aldosterone antagonist. In patients with atrial fibrillation, digoxin may be used at an earlier stage if a beta-blocker fails to control the ventricular rate (ideally <80bpm at rest and <110–120bpm during exercise; see graphic Chapter 29). Digoxin can also be used to control the ventricular rate when beta-blocker treatment is being initiated or up-titrated.

Digoxin should be avoided in patients with second-degree or greater atrioventricular block and pre-excitation syndromes; it should be used with caution in patients with sick-sinus syndrome. Hypokalaemia should be corrected before digoxin is administered. A loading dose of digoxin is generally not needed in stable patients in sinus rhythm. A single daily oral maintenance dose of 0.25mg is commonly used in adults with normal renal function. In the elderly and in those with renal impairment, a dose of 0.125mg or 0.0625mg may suffice. If the effect of digoxin is needed urgently, loading with 10–15mcg/kg lean body weight, given in three divided doses, 6 hours apart, may be used. The maintenance dose should be one-third of the loading dose. Smaller maintenance doses (e.g. one-quarter the loading dose and not more than 62.5mcg/day) should be used in the elderly and in patients with reduced renal function, as well as in patients with a low body mass. Monitoring of the serum digoxin concentration is recommended because of the narrow therapeutic window. A steady state is reached 7–10 days after starting treatment; blood should be collected at least 6 hours (and ideally 8–24 hours) after the last dose. The currently recommended therapeutic range is 0.6–1.2ng/mL (approximately 0.77–1.54nmol/L) [1, 2, 243].

Digoxin can cause anorexia, nausea, sinoatrial and atrioventricular block, arrhythmias, confusion, and visual disturbances (including xanthopsia), especially if the serum concentration is >2.0ng/mL. Hypokalaemia increases susceptibility to the adverse effects. The dose of digoxin should be reduced in the elderly and patients with renal dysfunction. Certain drugs increase serum digoxin concentration, including amiodarone, verapamil, and diltiazem.

Hydralazine is a powerful direct acting, arterial vasodilator. Its mechanism of action is not understood, though it may inhibit enzymatic production of superoxide, which neutralizes NO and may induce nitrate tolerance. Nitrates dilate both veins and arteries, thereby reducing preload and afterload by stimulating the NO pathway and increasing cyclic GMP in vascular smooth muscle. Neither drug on its own nor any other direct acting vasodilator has been demonstrated to be beneficial in heart failure.

Although this combination has been known for some time to improve systolic function and probably reduce death in NYHA class II–IV heart failure compared with placebo, a head-to-head comparison showed an ACE inhibitor was superior for improving survival (graphic Table 23.6) [244, 245]. Nevertheless, based on subgroup analyses suggesting that African Americans responded better to hydralazine and isosorbide dinitrate, a subsequent randomized controlled trial showed that the addition of hydralazine and isosorbide dinitrate in African Americans, most of whom were receiving an ACE inhibitor and beta-blocker and many of whom were on spironolactone, further reduced mortality and hospital admissions for heart failure and improved quality of life [246]. A fixed combination of 37.5mg of hydralazine and 20mg of isosorbide dinitrate was used in the trial; one tablet was given and if tolerated a second was given 12 hours later. One tablet was then prescribed three times daily for 3–5 days, at which point the dose was increased to the target maintenance of two tablets three times daily, i.e. a daily dose of 225mg hydralazine and 120mg isosorbide dinitrate. Because of the limited inclusion criteria of this trial, however, it is uncertain whether this combination of vasodilators is an effective addition in other patient populations.

Other than for African Americans, the main indication for hydralazine and isosorbide dinitrate is as a substitute in patients with intolerance to an ACE inhibitor and an ARB. Hydralazine and isosorbide dinitrate should be used as additional treatment in African Americans and considered, on an empirical basis, for other patients who remain symptomatic on other proven therapies. The main dose-limiting adverse effects with hydralazine and isosorbide dinitrate are headache and dizziness. A rare adverse effect of higher doses of hydralazine, especially in slow acetylators is a systemic lupus erythematosus-like syndrome.

A recent study showed that treatment with 1g of omega-3 polyunsaturated fatty acids (n-3 PUFA) (850–852mg eicosapentaenoic acid and docosahexaenoic acid as ethyl esters in the average ratio of 1:1.2) per day led to a small reduction in cardiovascular morbidity and mortality in patients with heart failure (graphic Table 23.6) [247]. The exact mechanism of action of this treatment is uncertain, although it may have beneficial, anti-inflammatory and electrophysiological effects (the latter reducing the risk of arrhythmias).

It is important to note that the aforementioned treatments are the only pharmacological agents shown to be of benefit in patients with heart failure and a low LVEF. Many other treatments have been tested in randomized trials and shown to have a neutral (e.g. amlodipine) [248] or uncertain (e.g. alpha-adrenoceptor blockers, bosentan, and etanercept) [249–252] effect on mortality and morbidity. Some also increased mortality and have either been withdrawn from the market or should be avoided in heart failure (e.g. dronedarone, milrinone, flosequinan, vesnarinone, and moxonidine) [253–257].

Other therapies of proven value for cardiovascular conditions underlying or associated with heart failure have not been specifically tested in heart failure (e.g. antiplatelet treatment in patients with coronary heart disease, see graphic Chapter 16) or may not be as clearly beneficial in heart failure (e.g. statins) [258, 259]. In patients with advanced systolic heart failure of ischaemic aetiology, statins did not reduce the primary outcome of cardiovascular death, myocardial infarction, or stroke (or all-cause mortality) but did reduce the number of cardiovascular hospitalizations [258]. A vitamin K antagonist, e.g. warfarin, is indicated in patients with atrial fibrillation provided there is no contraindication to its use (graphic Chapters 11 and 29). Warfarin may also be used in patients with evidence of intra-cardiac thrombus (e.g. detected during echocardiographic examination) or systemic thromboembolism. Warfarin’s many interactions with other drugs, including with some statins and amiodarone (graphic Chapter 11), must always be considered when initiating warfarin or another drug in a patient taking warfarin. Low-molecular-weight heparin prophylaxis (graphic Chapter 37) against deep venous thrombosis is indicated when patients with heart failure are bed-bound, for example during hospital admission, although heparin can cause hyperkalaemia and the dose must be reduced in patients with renal impairment.

Vaccination against influenza and pneumococcal infection is advised in all patients with heart failure because the stress of infection can lead to clinical deterioration [1].

Patients with heart failure, especially if severe, often have renal and hepatic dysfunction, so any drug excreted predominantly by the kidneys or metabolized by the liver may accumulate [75, 83–85, 187, 188]. Similarly, because of their extensive comorbidity, patients with heart failure are inevitably treated with multiple drugs, thereby increasing the risk of drug interactions.

Drugs that should be avoided, if possible, in heart failure include most antiarrhythmic drugs including dronedarone [257] (with the exception of amiodarone and dofetilide), most calcium channel blockers (with the exception of amlodipine), corticosteriods, NSAIDs, COX-2 inhibitors, and many antipsychotics (e.g. clozapine) and antihistamines. Thiazolidinediones (because of the risk of fluid retention) should be used with caution [70, 81–83, 260]. The use of metformin is usually not recommended because of the risk of lactic acidosis although this may be overstated [70]. Some salt substitutes contain substantial amounts of potassium and must be used cautiously. Other dietary constituents (e.g. grapefruit and cranberry juice) and supplements such as St. John’s Wort can interact with drugs taken by patients with heart failure, especially warfarin and digoxin [1, 2].

Several studies have shown that organized, nurse-led, multidisciplinary care can improve outcomes in patients with heart failure, particularly by reducing recurrent hospital admissions [261–263]. The most successful approach seems to involve education of the patients, their families, and caregivers about heart failure and its treatment (including flexible diuretic dosing and reinforcing the importance of adherence), recognizing (and acting upon) early deterioration (dyspnoea, weight gain, oedema), and optimizing proven pharmacological treatments (see graphic General measures, p.913) (graphic Table 23.12). A home-based rather than clinic-based approach may be best, though trials are needed to compare these types of interventions directly. Even telephone follow-up is of value. New technology enabling non-invasive home telemonitoring of physiological measures (e.g. heart rate and rhythm, blood pressure, temperature, respiratory rate, weight, and estimated body water content) and implanted devices, which collect similar data and may be interrogated remotely, are also being tested as aids to monitoring and management [264, 265].

Table 23.12
Essential topics in patient education with associated skills and appropriate self-care behaviours
Educational topics Skills and self-care behaviours

Definition and aetiology of heart failure

Understand the cause of heart failure and why symptoms occur

Symptoms and signs of heart failure

Monitor and recognize signs and symptoms

Record daily weight and recognize rapid weight gain

Know how and when to notify healthcare provider

Use flexible diuretic therapy if appropriate and recommended

Pharmacological treatment

Understand indications, dosing, and effects of drugs

Recognize the common side effects of each drug prescribed

Risk factor modification

Understand the importance of smoking cessation

Monitor blood pressure if hypertensive

Maintain good glucose control if diabetic

Avoid obesity

Diet recommendation

Sodium restriction if prescribed

Avoid of excessive fluid intake

Modest intake of alcohol

Monitor and prevent malnutrition

Exercise recommendations

Be reassured and comfortable about physical activity

Understand the benefits of exercise

Perform exercise training regularly

Sexual activity

Be reassured about engaging in sex and discuss problems with healthcare professionals

Understand specific sexual problems and various coping strategies

Immunization

Receive immunization against infections such as influenza and pneumococcal disease

Sleep and breathing disorders

Recognize preventive behaviour such as reducing weight of obese, smoking cession, and abstinence from alcohol

Learn about treatment options if appropriate

Adherence

Understand the importance of following treatment recommendations and maintaining motivation to follow treatment plan

Psychosocial aspects

Understand that depressive symptoms and cognitive dysfunction are common in patients with heart failure and the importance of social support

Learn about treatment options in appropriate

Prognosis

Understand important prognostic factors and make realistic decisions

Seek psychosocial support if appropriate

Educational topics Skills and self-care behaviours

Definition and aetiology of heart failure

Understand the cause of heart failure and why symptoms occur

Symptoms and signs of heart failure

Monitor and recognize signs and symptoms

Record daily weight and recognize rapid weight gain

Know how and when to notify healthcare provider

Use flexible diuretic therapy if appropriate and recommended

Pharmacological treatment

Understand indications, dosing, and effects of drugs

Recognize the common side effects of each drug prescribed

Risk factor modification

Understand the importance of smoking cessation

Monitor blood pressure if hypertensive

Maintain good glucose control if diabetic

Avoid obesity

Diet recommendation

Sodium restriction if prescribed

Avoid of excessive fluid intake

Modest intake of alcohol

Monitor and prevent malnutrition

Exercise recommendations

Be reassured and comfortable about physical activity

Understand the benefits of exercise

Perform exercise training regularly

Sexual activity

Be reassured about engaging in sex and discuss problems with healthcare professionals

Understand specific sexual problems and various coping strategies

Immunization

Receive immunization against infections such as influenza and pneumococcal disease

Sleep and breathing disorders

Recognize preventive behaviour such as reducing weight of obese, smoking cession, and abstinence from alcohol

Learn about treatment options if appropriate

Adherence

Understand the importance of following treatment recommendations and maintaining motivation to follow treatment plan

Psychosocial aspects

Understand that depressive symptoms and cognitive dysfunction are common in patients with heart failure and the importance of social support

Learn about treatment options in appropriate

Prognosis

Understand important prognostic factors and make realistic decisions

Seek psychosocial support if appropriate

Adapted with permission from Dickstein K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442. Copyright © ESC 2008.

Education of the patient, family, and caregivers is invaluable (graphic Table 23.12) [1, 2, 266]. Detection of early signs and symptoms of deterioration provides for earlier intervention. Counselling on the proper use of therapies, with an emphasis on adherence, is critical.

Useful patient and carer orientated material is available from the Heart Failure Association of the European Society of Cardiology in several languages (currently English, French, German, and Spanish) and from the Heart Failure Society of America and other organizations (see graphic Online resources, p.892).

When appropriate, a patient should be taught how to adjust the dose of diuretic within individualized limit—see graphic Diuretics, p.865. The dose should be increased (or a supplementary diuretic added) if there is evidence of fluid retention (symptoms of congestion), and decreased if evidence of hypovolaemia (e.g. increased thirst associated with weight loss or postural dizziness, especially during hot weather or an illness causing decreased fluid intake or sodium and water loss). If hypovolaemia is more marked, the doses of other medications (e.g. ACE inhibitors, spironolactone) also will have to be reduced.

The expected effects, beneficial and adverse, of other drugs should also be explained in detail (e.g. possible association of cough with ACE inhibitor). It is useful to inform patients that improvement with many drugs is gradual and may become fully apparent only after several weeks or even months of treatment. It is also important to explain the need for gradual titration with ACE-inhibitors, ARBs, and beta-blocking drugs to a desired dose level, which again may take weeks or even months to achieve. Patients should be advised not to use NSAIDs without consultation and to be cautious about using herbal or other non-proprietary preparations.

Education and counselling of the patient, caregiver, and family promotes adherence, which is associated with better outcomes [267]. Drug adherence can also be helped by certain pharmacy aids such as dose allocation (‘Dosette’) boxes.

The recent Heart Failure: A Controlled Trial Investigating Outcomes of Exercise TraiNing (HF-ACTION) trial also showed that a programme of tailored, structured, aerobic exercise is safe, improves functional capacity and quality of life, and may also reduce cardiovascular mortality and heart failure hospitalization [268, 269]. The regimen used was, however, labour intensive (graphic Table 23.13) and, on average, patients did not attain the target level of exercise, especially during the home-maintenance phase.

Table 23.13
HF-ACTION trial exercise training programme
Training phase Location Week* Weekly sessions Aerobic duration (min) Intensity (percentage of HRR) Mode of exercise

Initial, supervised

Clinic

1–2

3

15–30

60

Walk or cycle

Supervised

Clinic

3–6

3

30–35

70

Walk or cycle

Clinic/Home

7–12

3/2

30–35

70

Walk or cycle

Maintenance

Home

13 to end of treatment

5

40

60–70

Walk or cycle

Training phase Location Week* Weekly sessions Aerobic duration (min) Intensity (percentage of HRR) Mode of exercise

Initial, supervised

Clinic

1–2

3

15–30

60

Walk or cycle

Supervised

Clinic

3–6

3

30–35

70

Walk or cycle

Clinic/Home

7–12

3/2

30–35

70

Walk or cycle

Maintenance

Home

13 to end of treatment

5

40

60–70

Walk or cycle

HRR, heart rate recovery.

*

Week intervals shown are goals and may vary for individual patients.

Reproduced with permission from Whellan, DJ, O’Connor, CM, Lee, KL et al; HF-ACTION Trial Investigators. Heart failure and a controlled trial investigating outcomes of exercise training (HF-ACTION): design and rationale. Am Heart J 2007; 153: 201–11.

Most guidelines advocate avoidance of foods containing relatively high salt content in the belief that this may reduce the need for diuretic therapy, although there is little evidence from clinical trials to support this recommendation [1]. Some also believe that excess sodium intake can be a precipitant of clinical decompensation. Certain salt substitutes have a high potassium content, which can lead to hyperkalaemia.

Restriction of fluid intake is indicated only during episodes of decompensation associated with peripheral oedema or hyponatraemia. In these situations, daily intake should be restricted to 1.5–2L to help facilitate elimination of excess extracellular fluid volume and avoid hyponatraemia.

Reducing excessive weight will reduce the work of the heart and may lower blood pressure and is recommended in those who are obese (body mass index >30kg/m2). Conversely, malnutrition is common in severe heart failure, and the development of cardiac cachexia is an ominous sign [77, 90, 91]. Sometimes reduced food intake is caused by nausea (e.g. related to digoxin use or hepatosplenic congestion) or abdominal bloating (e.g. due to ascites). In these cases, small frequent meals and high protein and calorie liquids may be helpful. In severe decompensated heart failure, eating may be difficult because of dyspnoea.

Moderate alcohol intake (up to 10–20g/day, e.g. 1–2 standard glasses of wine is permissible) is not thought to be harmful in heart failure, although excessive intake can cause cardiomyopathy and atrial arrhythmias in susceptible individuals. In patients with suspected alcoholic cardiomyopathy, abstinence from alcohol may improve cardiac function and is recommended.

Smoking causes peripheral vasoconstriction, which is detrimental in heart failure [1, 2]. Nicotine replacement therapy is believed to be safe in heart failure. The safety of bupropion and varenicline in heart failure are uncertain.

Sexual activity need not be restricted in patients with compensated heart failure, though dyspnoea may be limiting [1, 2]. Sublingual nitrate may be used as prophylaxis against chest pain and dyspnoea caused by sexual activity. In men with erectile dysfunction, treatment with a cyclic guanine monophosphate phosphodiesterase type V inhibitor, such as sildenafil, can be useful, but these drugs must not be taken within 24 hours of prior nitrate use, and nitrates must not be restarted for at least 24 hours afterwards [270].

Patients with heart failure can continue to drive provided their condition does not induce undue dyspnoea, fatigue, or other incapacitating symptoms [1, 2]. Patients with recent syncope, cardiac surgery, percutaneous coronary intervention, or device placement may be restricted from driving, at least temporarily, according to local regulations. Patients holding an occupational or commercial licence may be subject to additional restrictions.

Short flights are unlikely to cause problems for a patient with compensated heart failure [1, 2]. Cabin pressure is generally maintained to provide an oxygen level no lower than equivalent to 6000 feet above sea level, which should be well tolerated in patients without severe pulmonary disease or pulmonary hypertension. Longer journeys may cause limb oedema and dehydration, thereby predisposing to venous thrombosis. Adjustment of the dose of diuretics and other treatments should be discussed with the patient wishing to travel to a warm climate or a country where the risk of gastroenteritis is high. It is also advisable for heart failure patients to carry a list of medications and contact information for their healthcare provider.

Beta-blockers are of benefit in both angina and heart failure. Nitrates relieve angina but, on their own, are not of proven value in chronic heart failure. Calcium-channel blockers should generally be avoided in heart failure as they have a negative inotropic action and cause peripheral oedema; only amlodipine has been shown to have no adverse effect on survival [248]. Trimetazine, ranolazine, and nicorandil are available in certain countries but their safety in heart failure is uncertain. Ivabradine is an inhibitor of the If current in the sinoatrial node, reduces heart rate, and is an effective antianginal agent. It has been evaluated in patients with coronary heart disease and an EF <40% many of which had heart failure and most of which were treated with an ACE inhibitor/ARB and beta-blocker. In the morBidity-mortality EvAlUaTion of the If inhibitor ivabradine in patients with coronary disease and left-ventricULar dysfunction (BEAUTIFUL) trial, ivabradine did not reduce the primary composite endpoint of cardiovascular death, myocardial infarction, or heart failure hospitalisation. However, no safety concerns were identified [271]. Percutaneous and surgical revascularization are also of value in relieving angina in selected patients with heart failure.

The recent Atrial Fibrillation and Congestive Heart Failure (AF-CHF) trial showed that there is no evidence to support a strategy of restoring sinus rhythm over one of controlling the ventricular rate (coupled with thromboembolism prophylaxis) in most patients with heart failure [272]. Exceptions include patients in which new-onset atrial fibrillation has caused myocardial ischaemia, hypotension, or pulmonary oedema and where pharmacological rate control is not rapidly achieved; in these patients, prompt electrical or pharmacological cardioversion may be indicated. In the routine setting, a beta-blocker alone or in combination with digoxin should be used to control the ventricular rate. The patient should be supervised closely after the initiation of these treatments because underlying sinus node dysfunction may raise the risk of bradycardia. Atrioventricular node ablation and pacing may be required to control ventricular rate in resistant cases. There is current interest in catheter ablation to cure atrial fibrillation in patients with heart failure, though this approach remains experimental [273]. There is a strong indication for thromboembolism prophylaxis with warfarin in patients with heart failure and atrial fibrillation.

Sustained ventricular tachycardia causing syncope or haemodynamic instability and ventricular fibrillation is an indication for an implantable cardioverter defibrillator (ICD) for ‘secondary prevention’ of sudden arrhythmic death. Amiodarone can be used to suppress recurrent arrhythmias and reduce ICD firing. General measures to reduce the substrate for arrhythmias are also important, including institution of an optimal combination of evidence-based pharmacological treatment (ACE inhibitor, beta-blocker, and either an ARB and aldosterone antagonist) used in optimal doses, correction of electrolyte imbalances, amelioration of myocardial ischaemia, cessation of pro-arrhythmic drugs etc. Catheter ablation may also be useful in selected cases [274].

Asthma is a contraindication for use of a beta-blocker but most patients with COPD can tolerate a beta-blocker. Pulmonary congestion can mimic COPD. Systemic administration of a corticosteroid to treat reversible airways obstruction may cause sodium and water retention and exacerbate heart failure, whereas inhalation therapy is better tolerated [200].

Beta-blocker treatment is not contraindicated and is of benefit in patients with diabetes and heart failure. Thiazolidinediones cause sodium and water retention and may lead to decompensation [70, 81, 260]. Metformin may cause lactic acidosis [70]. As a result, neither drug is recommended in patients with severe heart failure.

Amiodarone can also induce both hypothyroidism and hyperthyroidism, the latter being particularly difficult to diagnose [275].

Hyperuricaemia and gout are common in heart failure and, in part, are caused by diuretic treatment. Allopurinol may prevent gout. Acute attacks are better treated with colchicine or intra-articular steroids, rather than NSAIDs, COX-2 inhibitors, or oral steroids.

Most patients with heart failure have a reduced glomerular filtration rate (GFR) [83–86]. ACE inhibitors, ARBs, and aldosterone antagonists often cause a further small reduction in GFR and rise in serum urea/blood urea nitrogen (BUN) and creatinine levels, which, if limited, should not lead to discontinuation of treatment (graphic Tables 23.8, 23.9, and 23.11). Marked increases in BUN and creatinine, however, should prompt consideration of underlying renal artery stenosis. Renal dysfunction may also be caused by sodium and water depletion leading to relative hypovolaemia (e.g. due to excessive diuresis, diarrhoea and vomiting) or hypotension [83–86]. Nephrotoxic agents such as NSAIDs and certain antibiotics such as trimethoprim are also a common cause of renal dysfunction in heart failure [83–86].

For prostatic disease, a 5-alpha-reductase inhibitor may be preferable to an alpha-adrenoceptor antagonist, which can cause hypotension and salt and water retention [276]. Prostatic obstruction should also be considered in male patients with deteriorating renal function.

A normocytic, normochromic anaemia is common in heart failure, in part because of the high prevalence of renal dysfunction. Malnutrition and blood loss may also contribute. The roles of iron replacement and erythropoietic-stimulating substances in treating the anaemia of heart failure are under investigation and these are not currently recommended treatments [76, 87–89, 277].

Depression is common in patients with heart failure, perhaps partly owing to disturbance of the hypothalamic pituitary axis and other neurochemical pathways, but also as a result of social isolation and the adjustment to chronic disease. Depression is associated with worse functional status, reduced adherence to treatment, and poor clinical outcomes [78, 278]. Both psychosocial interventions and pharmacological treatment are helpful. Selective serotonin reuptake inhibitors are believed to be the best tolerated pharmacological agents, whereas tricyclic antidepressants should be avoided because of their anticholinergic actions and potential to cause arrhythmias [78].

Many anticancer drugs, particularly anthracyclines, cyclophosphamide, and trastuzumab (Herceptin®) can cause myocardial damage and heart failure, as can mediastinal radiotherapy [69, 279]. Pericardial constriction can be a result of previous radiotherapy, and malignant pericardial involvement can cause effusion and tamponade.

About half of patients with heart failure die suddenly, mainly as the result of a ventricular arrhythmia. The relative risk of sudden death, as opposed to death from progressive heart failure, is greatest in patients with milder heart failure. In patients with more advanced heart failure, progressive pump failure deaths are relatively more common. Antiarrhythmic drugs have not been shown to improve survival in heart failure, but ICDs (graphic Fig. 23.32) reduce the risk of death in selected patients after myocardial infarction (graphic Chapter 30) and improve survival in patients with NYHA class II–III heart failure due to systolic dysfunction of both ischaemic and non-ischaemic aetiology who were otherwise treated with optimal medical therapy (graphic Fig. 23.33) [280–282]. As a result, all patients with NYHA class II and III heart failure, irrespective of aetiology, and LVEF ≤35% without other conditions greatly limiting life expectancy (i.e. anticipated survival ≥1 year) or the quality of life should be considered for an ICD (graphic Fig. 23.28). The guidelines recognize that there is more evidence of benefit from ICDs in patients with heart failure of an ischaemic [274, 280, 281] compared to non-ischaemic aetiology [283] (graphic Table 23.14).

 Chest radiograph showing an ICD in
situ.
Figure 23.32

Chest radiograph showing an ICD in situ.

 Sudden Cardiac Death in Heart Failure
Trial (SCD-HeFT): Implantable cardioverter defibrillator vs. amiodarone vs.
placebo. Reproduced with permission from Bardy GH, Lee KL, Mark DB, et
al. Amiodarone or an implantable cardioverter-defibrillator for
congestive heart failure. N Engl J Med 2005; 352: 225–37.
Figure 23.33

Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT): Implantable cardioverter defibrillator vs. amiodarone vs. placebo. Reproduced with permission from Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med 2005; 352: 225–37.

Table 23.14
Class I recommendations for devices in patients with left ventricular systolic dysfunction
ICD

Prior resuscitated cardiac arrest

 

Ischaemic aetiology and >40 days of myocardial infarction

 

Non-ischaemic aetiology

Class I, Level A

 

Class I, Level A

 

Class I, Level B

ICD

Prior resuscitated cardiac arrest

 

Ischaemic aetiology and >40 days of myocardial infarction

 

Non-ischaemic aetiology

Class I, Level A

 

Class I, Level A

 

Class I, Level B

CRT

NYHA class III/IV and QRS >120ms

 

To improve symptoms/reduce hospitalization

 

To reduce mortality

Class I, Level A

 

Class I, Level A

 

Class I, Level A

CRT

NYHA class III/IV and QRS >120ms

 

To improve symptoms/reduce hospitalization

 

To reduce mortality

Class I, Level A

 

Class I, Level A

 

Class I, Level A

ICD, implantable cardioverter defibrillator; CRT, cardiac resynchronization therapy.

Adapted with permission from Dickstein K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442. © ESC 2008.

Between a quarter and a third of patients with heart failure have substantial prolongation of the QRS duration on the surface ECG, which is a marker of abnormal electrical activation of the left ventricle causing dyssynchronous contraction, less efficient ventricular emptying, and, often, mitral regurgitation [276]. Atrioventricular coupling may also be abnormal, as reflected by a prolonged PR interval, as may interventricular synchrony. One recent study showed that 10% of patients develop substantial new widening of the QRS each year, suggesting regular ECG review is worthwhile [283]. Cardiac resynchronization therapy (CRT) with

atriobiventricular or multisite pacing optimizes atrioventricular timing and improves synchronization of cardiac contraction. In selected patients with severe heart failure, CRT improves pump function, reduces mitral regurgitation, relieves symptoms, and significantly prolongs exercise capacity. In two major trials, CRT reduced the composite of death or hospital admission in patients with severe heart failure (graphic Table 23.6) by >35%, and in one trial it also reduced the relative risk of death from any cause by 36% (and absolute risk by 10%) (graphic Fig. 23.34) [284, 285]. Many other outcome measures, including quality of life, were also improved. The current debate focuses on how best to select patients who will benefit from CRT. The key trials to date selected patients on the basis of a markedly prolonged QRS duration, usually manifest as left bundle branch block and a QRS duration of >120ms (graphic Table 23.14). Although tissue Doppler echocardiography and other imaging techniques have been advocated as tools to identify patients likely to benefit from CRT, the evidence does not support such approaches [286, 287]. Whether patients with right bundle branch block, atrial fibrillation, milder heart failure, or with dyssychrony without marked QRS prolongation are helped by CRT is uncertain [288, 289]. There is no consensus yet about whether (or in whom) CRT pacing alone, i.e. CRT-P (graphic Fig. 23.35) or a CRT device with an ICD function i.e. CRT-D (graphic Fig. 23.36) should be used.

 Cardiac resynchronization therapy for
severe heart failure: two pivotal trials. Reproduced with permission from
Bristow MR, Saxon LA, Boehmer J, et al.; Comparison of Medical
Therapy, Pacing, and Defibrillation in Heart Failure (COMPANION)
Investigators. Cardiac-resynchronization therapy with or without an
implantable defibrillator in advanced chronic heart failure. N Engl J Med
2004; 350: 2140–50; and Cleland JG, Daubert JC, Erdmann E, et al.;
Cardiac Resynchronization-Heart Failure (CARE-HF) Study Investigators. The
effect of cardiac resynchronization on morbidity and mortality in heart
failure. N Engl J Med 2005; 352: 1539–49.
Figure 23.34

Cardiac resynchronization therapy for severe heart failure: two pivotal trials. Reproduced with permission from Bristow MR, Saxon LA, Boehmer J, et al.; Comparison of Medical Therapy, Pacing, and Defibrillation in Heart Failure (COMPANION) Investigators. Cardiac-resynchronization therapy with or without an implantable defibrillator in advanced chronic heart failure. N Engl J Med 2004; 350: 2140–50; and Cleland JG, Daubert JC, Erdmann E, et al.; Cardiac Resynchronization-Heart Failure (CARE-HF) Study Investigators. The effect of cardiac resynchronization on morbidity and mortality in heart failure. N Engl J Med 2005; 352: 1539–49.

 CRT-P device in situ showing
right and left ventricular pacing leads.
Figure 23.35

CRT-P device in situ showing right and left ventricular pacing leads.

 CRT-D device in situ.
Figure 23.36

CRT-D device in situ.

There is also uncertainty about how CRT devices should be ‘optimized’ [290].

The combination of multiple neurhohumoral inhibitors (in patients with mild–moderate symptoms) and CRT (in patients with severe symptoms) has led to stepwise improvements in survival over the past two decades (graphic Figs. 23.37 and 23.38).

With the exception of cardiac transplantation (and possibly ventricular assist devices), there are no generally accepted criteria for surgical intervention [199, 291]. There are very few sizeable clinical trials of surgical strategies in patients with heart failure. The largest to date did not show any benefit of surgical ventricular reconstruction [292]. Use of operative procedures is, therefore, very variable among centres and greatly dependent on local experience and expertise. Expert imaging and detailed haemodynamic and functional assessments are usually required when any patient with heart failure is considered for surgery, and close liaison between the relevant experts in these fields is essential. The collective expertise in surgical centres is often used to make highly individualized decisions about whether to operate and what procedures will be attempted. ‘Established’ operative treatments for patients with heart failure include coronary artery bypass grafting (CABG), surgery for valvular disease, left ventricular remodelling surgery (including aneurysmectomy), implantation of ventricular assist devices, and heart transplantation. ‘Experimental’ approaches include ventricular constraint devices and intramyocardial cell transplantation.

PCI or CABG (see graphic Chapter 17), when appropriate, is indicated for relief of angina or reversible ischaemia that contributes to the heart failure syndrome. The extent of ischaemia and myocardial viability can be assessed using non-invasive assessments such as dobutamine echocardiography (see graphic Chapter 4), cardiac MRI (see graphic Chapter 5), or PET scanning (see graphic Chapter 7) in patients with impaired LVEF. Whether CABG is beneficial in patients with coronary artery disease but without angina is uncertain, but it is postulated that improvement of coronary blood flow to viable but non-contracting (‘hibernating’) myocardium may improve ventricular function and clinical outcomes even in patients without inducible ischaemia. A large clinical trial comparing surgery (CABG and/or surgical ventricular reconstruction) and medical therapy has completed enrolment and will report initial findings in 2009/2010 [64].

One randomized controlled trial showed that long-term use of a left ventricular assist device (LVAD) led to a short but significant prolongation of survival in patients which had end-stage heart failure and were ineligible for transplantation (graphic Table 23.6) [291]. After 2 years of follow up, all patients in both the LVAD and medical therapy arms were dead. Device complications (e.g. due to infection and thromboembolism) were frequent. Such devices are also very expensive, as is the healthcare infrastructure required to support their use [293]. Despite this, in some centres, patients with severe heart failure are receiving these devices both as a ‘bridge to transplantation’ and as ‘destination therapy’, i.e. as the permanent, definitive, procedure [294, 295]. Short-term LVADs are also used, increasingly, as a ‘bridge to decision’ in the management of patients with acute heart failure who cannot be stabilized on inotropes and intra-aortic balloon pumps. Patients can be supported to cardiac transplantation, ‘upgraded’ to long-term ventricular assist devices or sometimes (e.g. in those with myocarditis) weaned to medical therapy. There is large variation in these practices between and within countries. There has also been some progress in LVAD design that might warrant further controlled trials.

Cardiac transplantation (graphic Chapter 18) remains the most accepted surgical intervention in heart failure. Most patients undergoing cardiac transplantation in the modern era are those presenting with acute severe heart failure [199, 296]. Some ambulatory patients with severe cardiac dysfunction and unacceptable symptoms also warrant consideration of listing. Conventional selection criteria for this group were developed in the era before beta-blockers, spironolactone, ICDs, and CRT, and up-to-date risk stratification tools are needed.

Most of the randomized controlled trials underpinning the evidence-based treatment of heart failure included only patients with a low LVEF (graphic Table 23.6). Treatment of this heart failure syndrome is, therefore, mainly empirical. Treatment of the underlying cardiovascular and other disorders thought to contribute to the development of heart failure with preserved LVEF (HF-PEF), such as hypertension, myocardial ischaemia, and diabetes, should be given as usual [297]. In patients with atrial fibrillation, control of the ventricular rate with a beta-blocker or a rate-limiting calcium-channel blocker (i.e. verapamil or diltiazem) (or restoration of sinus rhythm) is important (graphic Chapter 29). Diuretics are used, empirically, to treat sodium and water retention, according to the same principles as in heart failure with a low LVEF. Two small studies in patients in sinus rhythm showed that the alcium-channel blocker verapamil can improve symptoms and exercise capacity in patients with heart failure and preserved LVEF, possibly by reducing heart rate, and thereby increasing the duration of diastolic left ventricular filling, as well as by directly enhancing myocardial relaxation [298]. There are, however, no prospective randomized controlled mortality–morbidity trials with this drug in patients with heart failure and preserved LVEF. One medium-sized trial with an ACE inhibitor and two large trials with ARBs failed to show a clear-cut benefit on NYHA class, quality of life, or morbidity/mortality with drugs that block the RAAS [299–301].

Heart failure also can arise as a result of regurgitant and stenotic valve disease. The objective of treatment of primary valve disease is the prevention of heart failure by surgical repair or replacement of the diseased valve or valves (graphic Chapter 21). The development of overt heart failure is an ominous sign, sometimes requiring urgent valve replacement (e.g. aortic stenosis) but occasionally indicating that valve replacement may no longer be possible (e.g. because of severe pulmonary hypertension).

Evaluation of the aortic valve can be difficult in patients with poor left ventricular systolic function. Such patients may have insufficient cardiac output to generate a gradient across even a severely stenotic valve. Conversely, a calcified and degenerate but non-stenotic aortic valve may appear stenosed simply because it does not open normally in patients with very low cardiac output. Valve area provides a better assessment of whether there is significant aortic stenosis in these patients. Stress echocardiography may help assess the potential for ventricular recovery following relief of aortic stenosis (and the patient’s operative risk). Consideration should also be given to the occurrence of reversible depression of systolic function due to concomitant myocardial ischaemia resulting from coronary artery disease. There is a great deal of current interest in the potential of transcatheter valve replacement for aortic stenosis.

Sometimes it can be difficult to determine whether mitral regurgitation is primary or secondary in a patient with heart failure and left ventricular dilatation, though a prior history of known valve disease or rheumatic fever may suggest a primary valve problem. Surgery sometimes will result in clinical improvement but some patients will have such advanced left ventricular dysfunction that they will not achieve substantial benefit (e.g. mitral valve surgery in a patient with long-standing severe mitral regurgitation). Valve repair or annuloplasty may, however, have a role in the treatment of some carefully selected patients with secondary mitral regurgitation caused, or exacerbated, by left ventricular dilatation. Valve repair is generally preferable to valve replacement. As with aortic stenosis, surgical treatment is largely empirical and there is a need for randomized clinical trials to provide evidence-based management.

 Cumulative benefit of polypharmacy in
mild–moderate heart failure
Figure 23.37

Cumulative benefit of polypharmacy in mild–moderate heart failure

Patients with heart failure and normal coronary arteries should be evaluated for possible reversible causes. Untreated hypertension is now an unusual cause of dilated cardiomyopathy in developed countries but hypertension was once a leading cause in Europe and the USA and still remains a major consideration in many parts of the world. Infiltrative cardiomyopathies (e.g. haemochromatosis, amyloidosis, sarcoidosis) sometimes have specific recommended therapies. Chagas disease must be considered in patients from endemic areas. However, most cases of dilated cardiomyopathy will be ‘idiopathic’ (i.e. no specific aetiology is apparent). Possible inherited causes should be considered (graphic Chapter 18) and these patients should otherwise be treated in the same way as patients whose dilated, poorly contracting, left ventricle is a result of coronary artery disease [67, 102–104].

Heart failure can arise in patients with hypertrophic cardiomyopathy because of predominant diastolic dysfunction, left ventricular outflow tract obstruction (by either the septum or anterior mitral valve leaflet), associated mitral incompetence, or the development of systolic dysfunction. The management of hypertrophic cardiomyopathy and its complications is quite different than that of dilated cardiomyopathies and is discussed in graphic Chapter 18 [67, 102–104].

 Cumulative benefit of polypharmacy
(and cardiac resynchronization therapy) in severe heart failure.
Figure 23.38

Cumulative benefit of polypharmacy (and cardiac resynchronization therapy) in severe heart failure.

Patients presenting with acute heart failure include those who develop heart failure ‘de novo’ as a consequence of another cardiac event, usually a myocardial infarction, and those who present for the first time with decompensation of previously asymptomatic and often unrecognized cardiac dysfunction (patients previously in NYHA class I). However, due to frequent recurrences, most episodes of acute decompensation occur in patients with established, chronic heart failure that has worsened because of the unavoidable natural progression of the syndrome, an intercurrent cardiac (e.g. arrhythmia) or non-cardiac (e.g. pneumonia) event, or as a consequence of an avoidable reason such as non-adherence with treatment or use of an agent that can alter renal function. Although not always identified, searching for a reversible precipitant is an important aspect of the initial therapy plan (graphic Table 23.4).

Most patients with acute heart failure require admission to the hospital, especially if pulmonary oedema is present. In contrast to chronic heart failure, randomized controlled trials showing benefits of therapy are generally not available in acute heart failure. The principal goals of management are to relieve symptoms, the most important of which is extreme dyspnoea, and maintain or restore vital organ perfusion [1, 2, 303]. An intravenous bolus or infusion of a loop diuretic, and, in hypoxaemic patients, oxygen are the key first-line treatments. Non-invasive ventilation improves symptoms but does not reduce mortality [302]. Intravenous infusion of a nitrate is also valuable in patients with a systolic blood pressure ≥100mmHg (graphic Fig. 23.39) Intravenous nesiritide (human BNP) which is available in some countries may reduce the pulmonary capillary wedge pressure more promptly than intravenous glyceryltrinitrate, but the effect of this short-term therapy on other clinical outcomes is controversial. Levosimendan has both vasodilator and inotropic actions. An intravenous opiate is also valuable in excessively anxious or distressed patients and those in pain (graphic Fig. 23.39). In volume overloaded patients with severe heart failure unresponsive to diuretics, ultrafiltration is an option at specialized centres [304].

 Treatment algorithm for suspected
acute pulmonary oedema.
Figure 23.39

Treatment algorithm for suspected acute pulmonary oedema.

In patients with marked hypotension or other evidence of organ hypoperfusion, an inotropic agent such as dobutamine a phosphodiesterase inhibitor (e.g. milrinone), or levosimendan should be considered, although none of these treatments has been shown to reduce in-hospital deaths or readmission [1]. In general, potent inotropic agents should be used at the lowest clinically effective dose and for the shortest duration possible in a setting with close cardiac monitoring. Low-dose dopamine may be administered in an attempt to improve renal function, although there are limited data in support of this benefit [305].

In more critically ill patients, mechanical support, for example with an intra-aortic balloon pump, may also be considered. As alluded to earlier, short-term use of a mechanical assist device is sometimes also used in this setting. The aim of treatment is to support the patient’s circulation and vital organ function until either their own heart recovers or a definitive operative procedure can be performed (e.g. transplantation or long-term implantation of a ventricular assist device) [291–295].

In patients admitted to hospital, discharge planning and subsequent management to reduce the risk of readmission is important. Ideally, an effective oral diuretic regimen should have been identified, and fluid-volume and biochemical stability should have achieved. This optimization of volume status and development of a stable oral regimen prior to discharge is thought to reduce the risk of early readmission. Treatment with an ACE inhibitor, beta-blocker, and ARB or aldosterone antagonist, as appropriate, should also be started and titrated in the stabilized patient prior to discharge [306, 307]. Outpatient follow-up should be arranged to ensure that any of those treatments that have not been started, prior to discharge are initiated after discharge, and that the dose of each drug is increased, as tolerated, to the appropriate target [308].

The key to successful follow-up is the careful tracking of clinical symptoms and patient weights, which often involves interviewing not only the patient, but also family members, who may be more aware of changes in status than the patient. Continuity of care and seamless transitions from the inpatient to outpatient setting are crucial aspects of optimal management. Patients with severe heart failure and patients requiring frequent hospitalization require special care. Programmes that provide telephone-based tracking of daily weights and symptoms may detect deterioration in time to intervene before the need for hospitalization [261, 264]. Although these programmes may be costly, several evaluations have found them to be cost-effective [309]. Because the management of these patients requires considerable experience and expertise, specialized heart failure disease-management programmes and clinics have been developed and may provide additional benefit compared with traditional care [263].

Though predicting the trajectory of illness in patients with advanced heart failure is notoriously difficult, it is often apparent when a patient has progressed to end-stage heart failure, commonly associated with renal failure [310–312]. In these circumstances, the expertise of the palliative care team may be especially helpful. Useful websites providing information on palliative care relevant to heart failure are available (see graphic Online resources, p.892). Medications such as parenteral opiates (with an antiemetic) and benzodiazepines may be particularly helpful in relieving dyspnoea, anxiety, and pain that arises from ascites, hepatic congestion, lower limb oedema, and pressure points. At this stage in the patient’s illness, it may be appropriate to discuss withdrawal of conventional treatment, deactivation of an ICD to avoid undesired and unpleasant electrical discharges, and a ‘do not resuscitate order’ if the patient and others involved in the patient’s care agree that comfort-care is appropriate. Hospice care may be chosen by some at this point.

The authors would like to thank Dr Davide Castagno for his valuable contribution to this chapter.

Personal perspective

The past two decades have seen almost unimaginable improvements in morbidity and, especially, mortality in patients with heart failure and a low EF. Remarkably, these have been brought about by only a handful of drugs (from among the many tested) and two devices. These treatments (ACE inhibitors, beta-blockers, ARBs, aldosterone antagonists, ICDs, and CRT) have changed the natural history of heart failure and created new problems bringing new challenges. This is no more apparent than in the emergence of the ‘cardiorenal anaemia’ syndrome in elderly patients surviving with advanced heart failure. New treatments targeting renal dysfunction (adenosine antagonists) and new applications of a treatment used for anaemia in chronic kidney disease (erythropoiesis-stimulating agents) are currently being evaluated in these patients. But this is even more the era of devices, with unresolved questions (CRT-P or CRT-D?) and potentially broader application (e.g. CRT in less symptomatic patients or those with narrower QRS) being evaluated. While the prospect of new treatments is always exciting, we can still do better in applying the very effective treatments currently available to us and this may be aided by better information technology and perhaps by using biochemical markers such as natriuretic peptides to encouraging physicians to optimize therapy. Assessing the role of monitoring (either biochemically or technologically) to detect earlier deterioration (or pre-empt it) continues to be a priority as does the development of affordable and effective ventricular assist or even replacement devices. Transcatheter valve replacement and ablation therapy may yet improve outlook for patients with aortic stenosis and atrial fibrillation.

We also have to recognize, however, that the success of treatments for low EF heart failure has led to a growing population of very elderly patients with end-stage heart failure and that we need to help these patients die better having allowed them live better. We will need to apply the skills and resources of palliative care services to this new population.

Finding an effective treatment for HF-PEF has proved remarkably difficult and only spironolactone is presently being tested in a large-scale study in this type of heart failure. Similarly, bettering diuretics and nitrates has not been possible in acute heart failure although a very large trial is currently evaluating nesiritide in these patients. Although impressive recovery of myocardial function is now rou-tinely seen in patients treated with multiple neurohumoral antagonists (and in some receiving ventricular assist devices), this is not universal and the dream of replacing scar tissue with new myocytes continues to drive research with cell therapies although when, if ever, a therapeutic breakthrough will occur is uncertain. To repeat the therapeutic triumphs of the past two decades in the next two is hugely challenging but equally rewarding if we can do it.

Arnold
JM, Liu P, Demers C, et al. Canadian Cardiovascular Society consensus conference recommendations on heart failure
2006
: diagnosis and management.
Can J Cardiol
2006; 22: 23–45. Erratum in: Can J Cardiol 2006; 22: 271.

Arnold
JM, Howlett JG, Dorian P, et al. Canadian Cardiovascular Society Consensus Conference recommendations on heart failure update
2007
: Prevention, management during intercurrent illness or acute decompensation, and use of biomarkers. Can J Cardiol 2007; 23: 21–45.

Braunwald
E.
Biomarkers in heart failure.
 N Engl J Med  
2008
; 358: 2148–59.

Dickstein
K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442.

Hunt
SA, Abraham WT, Chin MH, et al. 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 (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure): developed in collaboration with the American College of Chest Physicians and the International Society for Heart and Lung Transplantation: endorsed by the Heart Rhythm Society.
Circulation
2005; 112: e154–e1235.

Jessup
M, Brozena S. Heart failure.
N Engl J Med
 
2003
; 348: 2007–18.

Krum
H, Jelinek MV, Stewart S, et al. Guidelines for the prevention, detection and management of people with chronic heart failure in Australia
2006
.
Med J Aust
2006; 185: 549–57.

Liew
CC, Dzau VJ. Molecular genetics and genomics of heart failure.
Nat Rev Genet
 
2004
; 5: 811–25.

Maisel
A, Mueller C, Adams K, Jr., et al. State of the art: using natriuretic peptide levels in clinical practice.
Eur J Heart Fail
 
2008
; 10: 824–39.

Malcom
J, Arnold O, Howlett JG, et al. Canadian Cardiovascular Society Consensus Conference guidelines on heart failure –
2008
update: best practices for the transition of care of heart failure patients, and the recognition, investigation and treatment of cardiomyopathies.
Can J Cardiol
2008; 24: 21–40.

McAlister
FA, Ezekowitz J, Hooton N, et al. Cardiac resynchronization therapy for patients with left ventricular systolicdysfunction: a systematic review.
JAMA
 
2007
; 297: 2502–14.

McMurray
JJ, Pfeffer MA.
Heart failure.
 
Lancet
 
2005
; 365: 1877–89.

Mosterd
A, Hoes AW.
Clinical epidemiology of heart failure.
 
Heart
 
2007
; 93: 1137–46.

Paulus
WJ, Tschöpe C, Sanderson JE, et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology.
Eur Heart J
 
2007
; 28: 2539–50.

Stewart
S, McAlister FA, McMurray JJ. Heart failure management programs reduce readmissions and prolong survival.
Arch Intern Med
 
2005
; 165: 1311; author reply 1311–12.

Zipes
DP, Camm AJ, Borggrefe M, et al. ACC/AHA/ESC
2006
guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death – executive summary: A report of the American College of Cardiology/American Heart Association Task Force and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop Guidelines for Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death) Developed in collaboration with the European Heart Rhythm Association and the Heart Rhythm Society. Eur Heart J 2006; 27: 2099–140.

1  

Dickstein
K, Cohen-Solal A, Filippatos G, et al. 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; 29: 2388–442.

2  

Hunt
SA, Abraham WT, Chin MH, et al. 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 (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure): developed in collaboration with the American College of Chest Physicians and the International Society for Heart and Lung Transplantation: endorsed by the Heart Rhythm Society.
Circulation
2005; 112: e154–e235.

3  

Solomon
SD, Dobson J, Pocock S, et al. Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity (CHARM) Investigators. Influence of nonfatal hospitalization for heart failure on subsequent mortality in patients with chronic heart failure.
Circulation
 
2007
; 116: 1482–7.

4  

Fonarow
GC. Epidemiology and risk stratification in acute heart failure.
Am Heart J
 
2008
; 155: 200–7.

5  

Gardin
JM, Siscovick D, Anton Culver H, et al. Sex, age, and disease affect echocardiographic left ventricular mass and systolic function in the free-living elderly: The Cardiovascular Health Study.
Circulation
 
1995
; 91: 1739–48.

6  

Kupari
M, Lindroos M, Livanainen AM, et al.
Congestive heart failure in old age: prevalence,
mechanisms and 4-year prognosis in the Helsinki Ageing Study.
J Intern Med
 
1997
; 241: 387–94.

7  

Morgan
S, Smith H, Simpson I, et al. Prevalence and clinical characteristics of left ventricular dysfunction among elderly patients in general practice setting: cross sectional survey.
BMJ
 
1999
; 318: 368–72.

8  

Hedberg
P, Lonnberg I, Jonasson T, et al. Left ventricular systolic dysfunction in 75-year-old men and women: a population-based study.
Eur Heart J
 
2001
; 22: 676–83.

9  

Nielsen
OW, Hilden J, Larsen CT, et al. Cross sectional study estimating prevalence of heart failure and left ventricular systolic dysfunction in community patients at risk.
Heart
 
2001
; 86: 172–8.

10  

Cortina
A, Reguero J, Segovia E, et al.
Prevalence of heart failure in Asturias (a region in the north of Spain).
 Am J Cardiol  
2001
; 87: 1417–19.

11  

Ceia
F, Fonseca C, Mota T, et al. Prevalence of chronic heart failure in South Western Europe: EPICA study. Eur J Heart Fail  
2002
; 4: 531–9.

12  

Redfield
MM, Jacobsen SJ, Burnett JC, Jr., et al. Burden of systolic and diastolic ventricular dysfunction in the community: appreciating the scope of the heart failure epidemic.
JAMA
 
2003
; 289: 194–202.

13  

Mosterd
A, Hoes AW, Bruyne MC, et al. Prevalence of heart failure and left ventricular dysfunction in the general population; the Rotterdam Study.
Eur Heart J
 
1999
; 20: 447–55.

14  

Anguita
Sánchez M, Crespo Leiro MG, de Teresa Galván E, et al. PRICE Study Investigators. Prevalence of heart failure in the Spanish general population aged over 45 years. The PRICE Study. Rev Esp Cardiol  
2008
; 61: 1041–9.

15  

Ho
KK, Pinsky JL, Kannel WB, et al. The epidemiology of heart failure: the Framingham Study.
J Am Coll Cardiol
 
1993
; 22: 6A–13A.

16  

Lloyd-Jones
DM, Larson MG, Leip EP, et al. Framingham Heart Study. Lifetime risk for developing congestive heart failure: the Framingham Heart Study.
Circulation
 
2002
; 106: 3068–72.

17  

Bleumink
GS, Knetsch AM, Sturkenboom MC, et al. Quantifying the heart failure epidemic: prevalence, incidence rate, lifetime risk and prognosis of heart failure. The Rotterdam Study. Eur Heart J  
2004
; 25: 1614–19.

18  

Levy
D, Kenchaiah S, Larson MG, et al.
Long-term trends in the incidence of and survival with heart failure.
 N Engl J Med  
2002
; 347: 1397–402.

19  

Roger
VL, Weston SA, Redfield MM, et al.
Trends in heart failure incidence and survival in a community-based population.
 
JAMA
 
2004
; 292: 344–50.

20  

Curtis
LH, Whellan DJ, Hammill BG, et al.
Incidence and prevalence of heart failure in elderly persons,
1994–2003.
Arch Intern Med
 
2008
; 168: 418–24.

21  

Stewart
S, MacIntyre K, Capewell S, et al.
Heart
failure and the aging population: an increasing burden in the 21st century? Heart  
2003
; 89: 49–53.

22  

Stewart
S, MacIntyre K, MacLeod MM, et al.
Trends in hospitalization for heart failure in Scotland,
1990–1996. An epidemic that has reached its peak?
Eur Heart J
 
2001
; 22: 209–17.

23  

Schaufelberger
M, Swedberg K, Koster M, et al. Decreasing one-year mortality and hospitalization rates for heart failure in Sweden; Data from the Swedish Hospital Discharge Registry 1988 to 2000.
Eur Heart J
 
2004
; 25: 300–7.

24  

Mosterd
A, Reitsma JB, Grobbee DE. Angiotensin converting enzyme inhibition and hospitalisation rates for heart failure in The Netherlands, 1980 to 1999: the end of an epidemic?
Heart
 
2002
; 87: 75–6.

25  

Cowie
MR, Fox KF, Wood DA, et al.
Hospitalization of patients with heart failure: a population-based study.
 
Eur Heart J
 
2002
; 23: 877–85.

26  

Blackledge
HM, Tomlinson J, Squire IB. Prognosis for patients newly admitted to hospital with heart failure: survival trends in 12 220 index admissions in Leicestershire 1993–2001.
Heart
 
2003
; 89: 615–20.

27  

Lee
DS, Mamdani MM, Austin PC, et al. Trends in heart failure outcomes and pharmacotherapy: 1992 to 2000.
Am J Med
 
2004
; 116: 581–9.

28  

Baker
DW, Einstadter D, Thomas C, et al. Mortality trends for 23 505 Medicare patients hospitalized with heart failure in Northeast Ohio, 1991 to 1997.
Am Heart J
 
2003
; 146: 258–64.

29  

Fang
J, Mensah GA, Croft JB, et al. Heart failure-related Hospitalization in the U.S., 1979 to 2004. J Am Coll Cardiol  
2008
; 52: 428–34.

30  

Guijarro
R, San Román CM, Perelló JI, et al. Efficiency Group of the Internal Medicine Services of Andalusia; Strategic Plan of the SADEMI (Andalusia Society of Internal Medicine). A study of hospital discharges for venous thromboembolism in the south of Spain. An analysis of 19,170 cases from a regional database from 1998 to 2001. Eur J Intern Med  
2005
; 16: 279–86.

31  

Mosterd
A, Hoes AW. Clinical epidemiology of heart failure.
Heart
 
2007
; 93: 1137–46.

32  

Stewart
S, Horowitz JD. Detecting early clinical deterioration in chronic heart failure patients post-acute hospitalisation – a critical component of multidisciplinary, home-based intervention? Eur J Heart Fail  
2002
; 4: 345–51.

33  

Stewart
S, Jenkins A, Buchan S, et al. The current cost of heart failure to the National Health Service in the UK. Eur J Heart Fail  
2002
; 4: 361–71.

34  

Murphy
NF, Simpson CR, McAlister FA, et al.
National survey of the prevalence,
incidence, primary care burden, and treatment of heart failure in Scotland.
Heart
 
2004
; 90: 1129–36.

35  

Hobbs
FD, Kenkre JE, Roalfe AK, et al. Impact of heart failure and left ventricular systolic dysfunction on quality of life: a cross-sectional study comparing common chronic cardiac and medical disorders and a representative adult population.
Eur Heart J
 
2002
; 23: 1867–76.

36  

Cowie
MR, Wood DA, Coats AJ, et al. Survival of patients with a new diagnosis of heart failure: a population based study.
Heart
 
2000
; 83: 505–10.

37  

Loehr
LR, Rosamond WD, Chang PP, et al. Heart failure incidence and survival (from the Atherosclerosis Risk in Communities study).
Am J Cardiol
 
2008
; 101: 1016–22.

38  

MacIntyre
K, Capewell S, Stewart S, et al. Evidence of improving prognosis in heart failure: trends in case fatality in 66 547 patients hospitalised between 1986 and 1995.
Circulation
 
2000
; 102: 1126–31.

39  

Stewart
S, MacIntyre K, Hole DJ, et al. More 'malignant' than cancer? Five-year survival following a first admission for heart failure.
Eur J Heart Fail
 
2001
; 3: 315–22.

40  

Jhund
PS, MacIntyre K, Simpson CR, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people.
Circulation
 
2009
; 119: 515–23.

41  

Ko
DT, Alter DA, Austin PC, et al. Life expectancy after an index hospitalization for patients with heart failure: a population-based study.
Am Heart J
 
2008
; 155: 324–31.

42  

Miyagishima
K, Hiramitsu S, Kimura H, et al.
Long term prognosis of chronic heart failure.
 
Circulation
 
2009
; 73: 92–9.

43  

Mehta
PA, Dubrey SW, McIntyre HF, et al. Mode of death in patients with newly diagnosed heart failure in the general population.
Eur J Heart Fail
 
2008
; 10: 1108–16.

44  

Hogg
K, Swedberg K, McMurray J. Heart failure with preserved left ventricular systolic function: epidemiology, clinical characteristics, and prognosis.
J Am Coll Cardiol
 
2004
; 43: 317–27.

45  

Thomas
MD, Fox KF, Coats AJ, et al. The epidemiological enigma of heart failure with preserved systolic function.
Eur J Heart Fail
 
2004
; 6: 125–36.

46  

Senni
M, Redfield MM. Heart failure with preserved systolic function. A different natural history?
J Am Coll Cardiol
 
2001
; 38: 1277–82.

47  

Banerjee
P, Banerjee T, Khand A, et al.
Diastolic heart failure: neglected or misdiagnosed?
 
J Am Coll Cardiol
 
2002
; 39: 138–41.

48  

Gottdiener
JS, McClelland RL, Marshall R, et al. Outcome of congestive heart failure in elderly persons: influence of left ventricular systolic function. The Cardiovascular Health Study. Ann Intern Med  
2002
; 137: 631–9.

49  

Lenzen
MJ, Scholte op Reimer WJ, Boersma E, et al. Differences between patients with a preserved and a depressed left ventricular function: a report from the EuroHeart Failure Survey. Eur Heart J  
2004
; 25: 1214–20.

50  

Fischer
M, Baessler A, Hense HW, et al. Prevalence of left ventricular diastolic dysfunction in the community. Results from a Doppler echocardiographic-based survey of a population sample. Eur Heart J  
2003
; 24: 320–8.

51  

Pedersen
F, Raymond I, Mehlsen J, et al. Prevalence of diastolic dysfunction as a possible cause of dyspnea in the elderly.
Am J Med
 
2005
; 118: 25–31.

52  

Badano
LP, Albanese MC, Biaggio P, et al. Prevalence, clinical characteristics, quality of life, and prognosis of patients with congestive heart failure and isolated left ventricular diastolic dysfunction.
J Am Soc Echocardiogr
 
2004
; 17: 253–61.

53  

Abhayaratna
WP, Smith WT, Becker NG, et al. Prevalence of heart failure and systolic ventricular dysfunction in older Australians: the Canberra Heart Study.
Med J Aust
 
2006
; 184: 151–4.

54  

Bursi
F, Weston SA, Redfield MM, et al.
Systolic and diastolic heart failure in the community.
 
JAMA
 
2006
; 296: 2209–16.

55  

Massie
BM, Carson PE, McMurray JJ, et al. the I-PRESERVE Investigators. Irbesartan in Patients with Heart Failure and Preserved Ejection Fraction.
N Engl J Med
 
2008
; 359: 2456–67.

56  

Wang
TJ, Levy D, Benjamin EJ, et al. The epidemiology of ‘asymptomatic’ left ventricular systolic dysfunction: implications for screening.
Ann Intern Med
 
2003
; 138: 907–16.

57  

McDonagh
TA, McDonald K, Maisel AS. Screening for asymptomatic left ventricular dysfunction using B-type natriuretic Peptide.
Congest Heart Fail
 
2008
; 14(4 Suppl.1): 5–8.

58  

Pedersen
F, Raymond I, Madsen LH, et al. Echocardiographic indices of left ventricular diastolic dysfunction in 647 individuals with preserved left ventricular systolic function. Eur J Heart Fail  
2004
; 6: 439–47.

59  

Mendez
GF, Cowie MR. The epidemiological features of heart failure in developing countries: a review of the literature. Int J Cardiol  
2001
; 80: 213–19.

60  

Agarwal
AK, Venugopalan P, Bono D. Prevalence and aetiology of heart failure in an Arab population.
Eur J Heart Fail
 
2001
; 3: 301–305.

61  

Bocchi
E, Guimarães G, Tarasoutshi F, et al.
Cardiomyopathy,
adult valve disease, and heart failure in South America.
Heart
 
2000
; 95: 181–9.

62  

Marijon
E, Ou P, Celermajers DS, et al.
Prevalence of rheumatic heart disease detected by echocardiographic screening.
 
N Engl J Med
 
2007
; 357: 470–6.

63  

Stewart
S, Wilkinson D, Hansen C, et al. A predominance of heart failure in the Heart of Soweto Study cohort: emerging challenges for urban African communities.
Circulation
 
2008
; 118: 2360–7.

64  

Velazquez
EJ, Lee KL, O’Connor CM, et al. STICH Investigator. The rationale and design of the Surgical Treatment for Ischemic Heart Failure (STICH) trial.
J Thorac Cardiovasc Surg
 
2007
; 134: 1540–7.

65  

Levy
D, Larson MG, Vasan RS, et al. The progression from hypertension to congestive heart failure.
JAMA
 
1996
; 275: 1557–62.

66  

Kostis
JB, Davis BR, Cutler J et al. Prevention of heart failure by antihypertensive drug treatment in older persons with isolated systolic hypertension. SHEP Cooperative Research Group.
JAMA
 
1997
; 278: 212–16.

67  

Lehnart
SE, Ackerman MJ, Benson DW, Jr., et al. Inherited arrhythmias: a National Heart, Lung, and Blood Institute and Office of Rare Diseases workshop consensus report about the diagnosis, phenotyping, molecular mechanisms, and therapeutic approaches for primary cardiomyopathies of gene mutations affecting ion channel function.
Circulation
 
2007
; 116: 2325–45.

68  

Kuethe
F, Sigusch HH, Hilbig K, et al. Detection of viral genome in the myocardium: lack of prognostic and functional relevance in patients with acute dilated cardiomyopathy.
Am Heart J
 
2007
; 153: 850–8.

69  

Broder
H, Gottlieb RA, Lepor NE. Chemotherapy and cardiotoxicity.
Rev Cardiovasc Med
 
2008
; 9: 75–83.

70  

MacDonald
MR, Petrie MC, Hawkins NM, et al.
Diabetes, left ventricular systolic dysfunction, and chronic heart failure.
 
Eur Heart J
 
2008
; 29: 1224–40.

71  

Murphy
NF, MacIntyre K, Stewart S, et al. Long-term cardiovascular consequences of obesity: 20-year follow-up of more than 15 000 middle-aged men and women (the Renfrew-Paisley study). Eur Heart J  
2006
; 27: 96–106.

72  

Stewart
S, Hart CL, Hole DJ, et al. A population-based study of the long-term risks associated with atrial fibrillation: 20-year follow-up of the Renfrew/Paisley study.
Am J Med
 
2002
; 113: 359–64.

73  

Jeong
YH, Choi KJ, Song JM, et al. Diagnostic approach and treatment strategy in tachycardia-induced cardiomyopathy.
Clin Cardiol
 
2008
; 31: 172–8.

74  

Shinbane
JS, Wood MA, Jensen DN, et al. Tachycardia-induced cardiomyopathy: a review of animal models and clinical studies.
J Am Coll Cardiol
 
1997
; 29: 709–15.

75  

Braunstein
JB, Anderson GF, Gerstenblith G, et al. Noncardiac comorbidity increases preventable hospitalizations and mortality among Medicare beneficiaries with chronic heart failure. J Am Coll Cardiol  
2003
; 42: 1226–33.

76  

Tang
YD, Katz SD. The prevalence of anemia in chronic heart failure and its impact on the clinical outcomes. Heart Fail Rev  
2008
; 13: 387–92.

77  

Pocock
SJ, McMurray JJ, Dobson J, et al. Weight loss and mortality risk in patients with chronic heart failure in the candesartan in heart failure: assessment of reduction in mortality and morbidity (CHARM) programme.
Eur Heart J
 
2008
; 29: 2641–50.

78  

Macchia
A, Monte S, Pellegrini F, et al. Depression worsens outcomes in elderly patients with heart failure: an analysis of 48,117 patients in a community setting.
Eur J Heart Fail
 
2008
; 10: 714–21.

79  

Mookadam
F, Calvin AD, Somers VK. Prevalence and management of central sleep apnea in heart failure patients. Curr Heart Fail Rep  
2008
; 5: 233–7.

80  

Petrie
CJ, Mark PB, Weir RA. Broken pump or leaky filter? Renal dysfunction in heart failure a contemporary review. Int J Cardiol  
2008
; 128: 154–65.

81  

Lago
RM, Singh PP, Nesto RW. Congestive heart failure and cardiovascular death in patients with prediabetes and type 2 diabetes given thiazolidinediones: a meta-analysis of randomised clinical trials.
Lancet
 
2007
; 370: 1129–36.

82  

Hudson
M, Rahme E, Richard H, et al. Risk of congestive heart failure with nonsteroidal antiinflammatory drugs and selective Cyclooxygenase 2 inhibitors: a class effect?
Arthritis Rheum
 
2007
; 57: 516–23.

83  

Shlipak
MG. Pharmacotherapy for heart failure in patients with renal insufficiency.
Ann Intern Med
 
2003
; 138: 917–24.

84  

Bongartz
LG, Cramer MJ, Doevendans PA, et al.
The severe cardiorenal syndrome: ‘Guyton revisited’.
 
Eur Heart J
 
2005
; 26: 11–17.

85  

Ronco
C, Haapio M, House AA, et al.
Cardiorenal syndrome.
 
J Am Coll Cardiol
 
2008
; 52: 1527–39.

86  

Damman
K, Navis G, Voors AA, et al. Worsening renal function and prognosis in heart failure: systematic review and meta-analysis.
J Card Fail
 
2007
; 13: 599–608.

87  

Levy
WC. Anemia in heart failure: marker or mediator of adverse prognosis?
J Am Coll Cardiol
 
2008
; 51: 577–8.

88  

Murphy
NF, McDonald K. Treatment of anaemia in chronic heart failure – optimal approach still unclear.
Eur Heart J
 
2007
; 28: 2185–7.

89  

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
; 52: 818–27.

90  

Anker
SD, Negassa A, Coats AJ, et al. Prognostic importance of weight loss in chronic heart failure and the effect of treatment with angiotensin-converting-enzyme inhibitors: an observational study.
Lancet
 
2003
; 361: 1077–83.

91  

von
Haehling S, Lainscak M, Springer J, et al.
Cardiac cachexia: A systematic overview.
 
Pharmacol Ther
 
2009
; 121: 227–52.

92  

Cohn
JN, Ferrari R, Sharpe N. Cardiac remodelling – concepts and clinical implications: a consensus paper from an international forum on cardiac remodeling. Behalf of an International Forum on Cardiac Remodeling.
J Am Coll Cardiol
 
2000
; 35: 569–82.

93  

Mudd
JO, Kass DA. Tackling heart failure in the twenty first century.
Nature
 
2008
; 451: 919–27.

94  

Divakaran
V, Mann DL. The emerging role of microRNAs in cardiac remodeling and heart failure.
Circ Res
 
2008
; 103: 1072–83.

95  

Levick
SP, Brower GL. Regulation of matrix metalloproteinases is at the heart of myocardial remodeling. Am J Physiol Heart Circ Physiol  
2008
; 295: H1375–H1376.

96  

Latini
R, Masson S, Staszewsky L, et al. Neurohormonal modulation in heart failure of ischemic etiology: correlates with left ventricular remodeling.
Curr Heart Fail Rep
 
2006
; 3: 157–63.

97  

Mudd
JO, Kass DA. Reversing chronic remodeling in heart failure.
Expert Rev Cardiovasc Ther
 
2007
; 5: 585–98.

98  

Abrahamsson
P, Dobson J, Granger CB, et al. for the CHARM Investigators. Impact of hospitalization for acute coronary events on subsequent mortality in patients with chronic heart failure. Heart J  
2009
; 30: 338–45.

99  

Neubauer
S. The failing heart – an engine out of fuel.
N Engl J Med
 
2007
; 356: 1140–51.

100  

Bleasdale
RA, Frenneaux MP. Cardiac resynchronisation therapy: when the drugs don't work.
Heart
 
2004
; 90: vi2–4.

101  

Ashrafian
H, Williams L, Frenneaux MP. The pathophysiology of heart failure: a tale of two old paradigms revisited.
Clin Med
 
2008
; 8: 192–7.

102  

Elliott
P, Andersson B, Arbustini E, et al. Classification of the cardiomyopathies: a position statement from the European Society Of Cardiology Working Group on Myocardial and Pericardial Diseases.
Eur Heart J
 
2008
; 29: 270–6.

103  

Deo
R, MacRae CA. The genetics of cardiomyopathies: what clinicians should know.
Curr Heart Fail Rep
 
2007
; 4: 229–35.

104  

Malcom
J, Arnold O, Howlett JG, et al. Canadian Cardiovascular Society. Canadian Cardiovascular Society Consensus Conference guidelines on heart failure –
2008
update: best practices for the transition of care of heart failure patients, and the recognition, investigation and treatment of cardiomyopathies.
Can J Cardiol
2008; 24: 21–40.

105  

Kitzman
DW. Diastolic dysfunction: one piece of the heart failure with normal ejection fraction puzzle.
Circulation
 
2008
; 117: 2044–6.

106  

Paulus
WJ, Tschöpe C, Sanderson JE, et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology.
Eur Heart J
 
2007
; 28: 2539–50.

107  

Martos
R, Baugh J, Ledwidge M, et al. Diastolic heart failure: evidence of increased myocardial collagen turnover linked to diastolic dysfunction.
Circulation
 
2007
; 115: 888–95.

108  

Lam
CS, Roger VL, Rodeheffer RJ, et al. Cardiac structure and ventricular-vascular function in persons with heart failure and preserved ejection fraction from Olmsted County, Minnesota.
Circulation
 
2007
; 115: 1982–90.

109  

Cohen-Solal
A, Logeart D, Tartiere JM. ‘Diastolic’ heart failure, overlooked systolic dysfunction, altered ventriculo-arterial coupling or limitation of cardiac reserve?
Int J Cardiol
 
2008
; 128: 299–303.

110  

Zile
MR, Baicu CF, Gaasch WH. Diastolic heart failure – abnormalities in active relaxation and passive stiffness of the left ventricle.
N Engl J Med
 
2004
; 350: 1953–9.

111  

Kushwaha
SS, Fallon JT, Fuster V. Restrictive cardiomyopathy.
N Engl J Med
 
1997
; 336: 267–76.

112  

Eshaghian
S, Kaul S, Shah PK. Cardiac amyloidosis: new insights into diagnosis and management.
Rev Cardiovasc Med
 
2007
; 8: 189–99.

113  

Kim
JS, Kim HH, Yoon Y. Imaging of pericardial diseases.
Clin Radiol
 
2007
; 62: 626–31.

114  

Goldstein
JA.
Cardiac tamponade,
constrictive pericarditis, and restrictive cardiomyopathy.
Curr Probl Cardiol
 
2004
; 29: 503–67.

115  

Zile
MR, Gaasch WH. Heart failure in aortic stenosis – improving diagnosis and treatment.
N Engl J Med
 
2003
; 348: 1735–6.

116  

Borer
JS, Truter SL, Gupta A, et al. Heart failure in aortic regurgitation: the role of primary fibrosis and its cellular and molecular pathophysiology.
Adv Cardiol
 
2004
; 41: 16–24.

117  

Mehra
MR, Gheorghiade M, Bonow RO. Mitral regurgitation in chronic heart failure: more questions than answers? Curr Cardiol Rep  
2004
; 6: 96–9.

118  

Weil
J, Eschenhagen T, Hirt S, et al.
Preserved Frank-Starling mechanism in human end stage heart failure.
 
Cardiovasc Res
 
1998
; 37: 541–8.

119  

Sandler
H, Dodge HT. Left ventricular tension and stress in man.
Circ Res
 
1963
; 13: 91.

120  

Opie
LH, Commerford PJ, Gersh BJ, et al.
Controversies in ventricular remodelling.
 
Lancet
 
2006
; 367: 356–67.

121  

Spann
JF, Buccino RA, Sonnenblick EH, et al. Contractile state of cardiac muscle obtained from cats with experimentally produced ventricular hypertrophy and heart failure. Circ Res  
1967
; 21: 341–54.

122  

Nishida
K, Yamaguchi Otsu K, et al.
Crosstalk between autophagy and apoptosis in heart disease.
 
Circ Res
 
2008
; 103: 343–51.

123  

Francis
GS, Cohn JN. Heart failure: mechanisms of cardiac and vascular dysfunction and the rationale for pharmacologic intervention.
FASEB J
 
1990
; 4: 3068–75.

124  

Harris
P.
Biology of cardiac failure.
 Eur Heart J  
1982
; 3(Suppl.D): 5–10.

125  

de
Sa DD, Chen HH. The role of natriuretic peptides in heart failure.
Curr Cardiol Rep
 
2008
; 10: 182–9.

126  

Bunton
DC, Petrie MC, Hillier C, et al. The clinical relevance of adrenomedullin: a promising profile? Pharmacol Ther  
2004
; 103: 179–201.

127  

Hogg
K, McMurray J. Neurohumoral pathways in heart failure with preserved systolic function.
Prog Cardiovasc Dis
 
2005
; 47: 357–66.

128  

Braunwald
E, Harrison DC, Chidsey CA. The heart as an endocrine organ.
Am J Med
 
1964
; 36: 1–4.

129  

Francis
GS, Cohn JN. The autonomic nervous system in congestive heart failure.
Annu Rev Med
 
1986
; 37: 235–47.

130  

Tsutamoto
T, Nishiyama K, Sakai H, et al. Transcardiac increase in norepinephrine and prognosis in patients with chronic heart failure.
Eur J Heart Fail
 
2008
; 10: 1208–14.

131  

Cohn
JN, Levine TB, Olivari MT, et al. Plasma norepinephrine as a guide to prognosis in patients with chronic congestive heart failure.
N Engl J Med
 
1984
; 311: 819–23.

132  

Unger
T, Li J. The role of the renin-angiotensin-aldosterone system in heart failure. J Renin Angiotensin Aldosterone Syst  
2004
; 5(Suppl.1): S7–S10.

133  

Epelman
S, Tang WH, Chen SY, et al. Detection of soluble angiotensin-converting enzyme 2 in heart failure: insights into the endogenous counter-regulatory pathway of the renin-angiotensin-aldosterone system.
J Am Coll Cardiol
 
2008
; 52: 750–4.

134  

Levine
TB, Cohn JN, Vrobel T, Franciosa JA. High renin in heart failure: a manifestation of hyponatremia. Trans Assoc Am Physicians  
1979
; 92: 203–7.

135  

Pitt
B. Aldosterone blockade in patients with chronic heart failure.
Cardiol Clin
 
2008
; 26: 15–21.

136  

Batenburg
WW, Jan Danser AH. The (pro)renin receptor: a new addition to the renin-angiotensin system? Eur J Pharmacol  
2008
; 585: 320–4.

137  

Goldsmith
SR, Gheorghiade M. Vasopressin antagonism in heart failure.
J Am Coll Cardiol
 
2005
; 46: 1785–91.

138  

Finley
JJ, 4th, Konstam MA, Udelson JE. Arginine vasopressin antagonists for the treatment of heart failure and hyponatremia.
Circulation
 
2008
; 118: 410–21.

139  

Daniels
LB, Maisel AS.
Natriuretic peptides.
 J Am Coll Cardiol  
2007
; 50: 2357–68.

140  

Richards
AM. Natriuretic peptides: update on Peptide release, bioactivity, and clinical use.
Hypertension
 
2007
; 50: 25–30.

141  

Lee
CY, Burnett JC, Jr. Natriuretic peptides and therapeutic applications.
Heart Fail Rev
 
2007
; 12: 131–42.

142  

Akashi
YJ, Springer J, Lainscak M, et al.
Atrial natriuretic peptide and related peptides.
 
Clin Chem Lab Med
 
2007
; 45: 1259–67.

143  

Chandrasekaran
B, Dar O, McDonagh T. The role of apelin in cardiovascular function and heart failure.
Eur J Heart Fail
 
2008
; 10: 725–32.

144  

Tang
WH, Francis GS. Exploring new drugs for heart failure: the case of urocortin.
Eur Heart J
 
2007
; 28: 2561–2.

145  

Fisher
C, MacLean M, Morecroft I, et al. Is the pregnancy hormone relaxin also a vasodilator peptide secreted by the heart?
Circulation
 
2002
; 106: 292–5.

146  

Teerlink
JR, Metra M, Felker GM, et al. Relaxin for the treatment of patients with acute heart failure (Pre-RELAX-AHF): a multicentre, randomised, placebo-controlled, parallel-group, dose-finding phase IIb study.
Lancet
 
2009
; 373: 1429–39.

147  

Drexler
H, Hayoz D, Munzel T, et al.
Endothelial function in chronic congestive heart failure.
 
Am J Cardiol
 
1992
; 69: 1596–601.

148  

Hambrecht
R, Adams V, Gielen S, et al. Exercise intolerance in patients with chronic heart failure and increased expression of inducible nitric oxide synthase in the skeletal muscle. J Am Coll Cardiol  
1999
; 33: 174–9.

149  

Cowburn
PJ, Cleland JG. Endothelin antagonists for chronic heart failure: do they have a role?
Eur Heart J
 
2001
; 22: 1772–84.

150  

McMurray
J, Chopra M, Abdullah I, et al.
Evidence of oxidative stress in chronic heart failure in humans.
 Eur Heart J  
1993
; 14: 1493–8.

151  

Grieve
DJ, Shah AM. Oxidative stress in heart failure. More than just damage.
Eur Heart J
 
2003
; 24: 2161–3.

152  

Landmesser
U, Spiekermann S, Dikalov S, et al. Vascular oxidative stress and endothelial dysfunction in patients with chronic heart failure: role of xanthine-oxidase and extracellular superoxide dismutase.
Circulation
 
2002
; 106: 3073–8.

153  

Berry
C, Hamilton CA, Brosnan MJ, et al. Investigation into the sources of superoxide in human blood vessels: angiotensin II increases superoxide production in human internal mammary arteries.
Circulation
 
2000
; 101: 2206–12.

154  

Doehner
W, Springer J, Landmesser U, et al.
Uric acid in chronic heart failure – current pathophysiological concepts.
 
Eur J Heart Fail
 
2008
; 10: 1269–70.

155  

Hare
JM, Mangal B, Brown J, et al. OPT-CHF Investigators. Impact of oxypurinol in patients with symptomatic heart failure. Results of theOPT-CHF study.
J Am Coll Cardiol
 
2008
; 51: 2301–9.

156  

Keith
ME, Jeejeebhoy KN, Langer A, et al. A controlled clinical trial of vitamin E supplementation in patients with congestive heart failure.
Am J Clin Nutr
 
2001
; 73: 219–24.

157  

Heymans
S, Hirsch E, Anker S, et al. Inflammation as a therapeutic target in heart failure? A Scientific Statement From the Translational Research Committee of the Heart Failure Association of the European Society of Cardiology.
Eur J Heart Failure
 
2009
; 11: 119–29.

158  

Rauchhaus
M, Coats AJ, Anker SD. The endotoxin–lipoprotein hypothesis.
Lancet
 
2000
; 356: 930–3.

159  

Mann
DL, McMurray JJ, Packer M, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL).
Circulation
 
2004
; 109: 1594–602.

160  

Chung
ES, Packer M, Lo KH, et al. Anti-TNF Therapy Against Congestive Heart Failure Investigators. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial.
Circulation
 
2003
; 107: 3133–40.

161  

Mancini
DM. Pulmonary factors limiting exercise capacity in patients with heart failure.
Prog Cardiovasc Dis
 
1995
; 37: 347.

162  

Piepoli
M, Ponikowski P, Clark AL. A neural link to explain the ‘muscle hypothesis’ of exercise intolerance in chronic heart failure.
Am Heart J
 
1999
; 137: 1050–6.

163  

Geltman
EM. Mild heart failure: diagnosis and treatment.
Am Heart J
 
1989
; 118: 1277.

164  

Clark
AL. Origin of symptoms in chronic heart failure.
Heart
 
2006
; 92: 12–16.

165  

Davie
AP, Francis CM, Caruana L, et al.
Assessing diagnosis in heart failure: which features are any use?
 Q J Med  
1997
; 90: 335–9.

166  

Madhok
V, Falk G, Rogers A, et al. The accuracy of symptoms, signs and diagnostic tests in the diagnosis of left ventricular dysfunction in primary care: a diagnostic accuracy systematic review.
BMC Fam Pract
 
2008
; 9: 56.

167  

Anselm
AH, Gauthier N, Beanlands RS, et al.
Sleep apnea in chronic heart failure.
 
Curr Opin Cardiol
 
2008
; 23: 121–6.

168  

Criteria
Committee, New-York Association, Inc. Diseases of the Heart and Blood Vessels. Nomenclature and Criteria for Diagnosis, 6th edn.,
1964
. Boston: Little Brown & Co., p. 114.

169  

Marantz
PR, Tobin JN, Wassertheil-Smoller S, et al.
The relationship by clinical criteria.
 
Circulation
 
1988
; 77: 607–12.

170  

Khot
UN, Jia G, Moliterno DJ, et al. Prognostic importance of physical examination for heart failure in non-ST-elevation acute coronary syndromes: the enduring value of Killip classification.
JAMA
 
2003
; 290: 2174–81.

171  

Rector
TS, Cohn JN. Assessment of patient outcome with the Minnesota Living with Heart Failure questionnaire: reliability and validity during a randomised, double-blind, placebo-controlled trial of pimobendan. Pimobendan Multicenter Research Group.
Am Heart J
 
1992
; 124: 1017–25.

172  

Spertus
JA, Jones PG, Kim J, et al.
Validity,
reliability, and responsiveness of the Kansas City Cardiomyopathy Questionnaire in anemic heart failure patients.
Qual Life Res
 
2008
; 17: 291–8.

173  

Spiteri
MA, Cook DG, Clarke SW. Reliability of eliciting physical signs in examination of the chest.
Lancet
 
1988
; 1: 873–5.

174  

Butman
SM, Ewy GA, Standen JR, et al. Bedside cardiovascular examination in patients with severe chronic heart failure: importance of rest or inducible jugular venous distension. J Am Coll Cardiol  
1993
; 22: 968–74.

175  

Lok
CE, Morgan CD, Ranganathan N. The accuracy and interobserver agreement in detecting the ‘gallop sounds’ by cardiac auscultation.
Chest
 
1998
; 114: 1283–8.

176  

Drazner
MH, Rame JE, Stevenson LW, Dries DL. Prognostic importance of elevated jugular venous pressure and a third heart sound in patients with heart failure.
N Engl J Med
 
2001
; 345: 574–81.

177  

Rihal
CS, Davis KB, Kennedy JW, et al. The utility of clinical, electrocardiographic and roentgenographic variables in the prediction of left ventricular function.
Am J Cardiol
 
1995
; 75: 220–3.

178  

Clark
AL, Goode K, Cleland JG. The prevalence and incidence of left bundle branch block in ambulant patients with chronic heart failure.
Eur J Heart Fail
 
2008
; 10: 696–702.

179  

Hawkins
NM, Wang D, McMurray JJ, et al. CHARM Investigators and Committees. Prevalence and prognostic impact of bundle branch block in patients with heart failure: evidence from the CHARM programme.
Eur J Heart Fail
 
2007
; 9: 510–17.

180  

Gillespie
ND, McNeill G, Pringle T, et al. Cross sectional study of contribution of clinical assessment and simple cardiac investigations to diagnosis of left ventricular systolic dysfunction in patients admitted with acute dyspnoea.
BMJ
 
1997
; 314: 936–40.

181  

Petrie
MC, McMurray JJV. It cannot be cardiac failure because the heart is not enlarged on the chest X-ray. Eur J Heart Fail  
2003
; 5: 117–19.

182  

Chakko
S, Woska D, Martinez H, et al. Clinical, radiographic and hemodynamic correlations in chronic congestive heart failure: conflicting results may lead to inappropriate care.
Am J Med
 
1991
; 90: 353–9.

183  

Peacock
WF 4th, De Marco T, Fonarow GC, et al. ADHERE Investigators. Cardiac troponin and outcome in acute heart failure.
N Engl J Med
 
2008
; 358: 2117–26.

184  

Tang
WH, Francis GS, Morrow DA, et al. National Academy of Clinical Biochemistry Laboratory Medicine. National Academy of Clinical Biochemistry Laboratory Medicine practice guidelines: Clinical utilization of cardiac biomarker testing in heart failure.
Circulation
 
2007
; 116: e99–e109.

185  

Gheorghiade
M, Konstam MA, Burnett JC Jr., et al. Efficacy of Vasopressin Antagonism in Heart Failure Outcome Study With Tolvaptan(EVEREST) Investigators. Short-term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status Trials.
JAMA
 
2007
; 297: 1332–43.

186  

Juurlink
DN, Mamdani MM, Lee DS, et al. Rates of hyperkalaemia after publication of the Randomized Aldactone Evaluation Study.
N Engl J Med
 
2004
; 351: 543–551.

187 Allen LA, Felker GM, Pocock S, et al. Liver function abnormalities and outcome in patients with chronic heart failure: data from the Candesartan in Heart Failure: Assessment of Reduction in Mortality and Morbidity (CHARM) program. Eur J Heart Fail 2009; 11: 170–7.

188  

Felker
GM, Allen LA, Pocock SJ, et al. CHARM Investigators.Red cell distribution width as a novel prognostic marker in heart failure:data from the CHARM Program and the Duke Databank. J Am Coll Cardiol  
2007
; 50: 40–7.

189  

Maisel
A, Mueller C, Adams K, Jr., et al.
State of the art: using natriuretic peptide levels in clinical practice.
 Eur J Heart Fail  
2008
; 10: 824–39.

190  

Vajan
RS, Benjamin EJ, Larson MG, et al. Plasma natriuretic peptides for community screening for left ventricular hypertrophy and systolic dysfunction. The Framingham Heart Study.
JAMA
 
2002
; 288: 1252–9.

191  

Cleland,
JG, Swedberg, K, Follath, F et al. The Euro Heart Failure Survey Progamme: a survey on the quality of care among patients with heart failure in Europe. Part I: patient characteristics and diagnosis.
Eur Heart J
 
2003
; 24: 442–63.

192  

Remme
WJ, McMurray JJ, Hobbs FD, et al. SHAPE Study Group. Awareness and perception of heart failure among European cardiologists, internists, geriatricians, and primary care physicians. Eur Heart J  
2008
; 29: 1739–52.

193  

Otterstad
JE, Froeland G, Sutton STM, et al. Accuracy and reproducibility of biplane two-dimensional echocardiographic measurements of left ventricular dimensions and function.
Eur Heart J
 
1997
; 18: 507–13.

194 Nagueh SF, Appleton CP, Gillebert TC, et al. Recommendations for the evaluation of left ventricular diastolic function by echocardiography. J Am Soc Echocardiogr 2009; 22: 107–33.

195  

Whalley
GA, Gamble GD, Dini FL, et al. Meta-Analysis Research Group in Echocardiography (MeRGE) Collaborators. Individual patient meta-analyses of restrictive diastolic filling pattern and mortality in patients post acute myocardial infarction and in patients with chronic heart failure.
Int J Cardiol
 
2007
; 122: 207–15.

196 Sicari R, Nihoyannopoulos P, Evangelista A, et al. Stress Echocardiography Expert Consensus Statement – Executive Summary: European Association of Echocardiography (EAE) (a registered branch of the ESC). Eur Heart J 2009; 30: 278–89.

197  

Assomull
RG, Pennell DJ, Prasad SK. Cardiovascular magnetic resonance in the evaluation of heart failure.
Heart
 
2007
; 93: 985–92.

198 Task Force of the Italian Working Group on Cardiac Rehabilitation and Prevention (Gruppo Italiano di Cardiologia Riabilitativa e Prevenzione,GICR); Working Group on Cardiac Rehabilitation and Exercise Physiology of the European Society of Cardiology,

Piepoli
MF, Corrà U, et al. Statement on cardiopulmonary exercise testing in chronic heart failure due toleft ventricular dysfunction: recommendations for performance andinterpretation Part II: How to perform cardiopulmonary exercise testing in chronic heart failure.
Eur J Cardiovasc Prev Rehabil
 
2006
; 13: 300–11.

199  

Gardner
RS, McDonagh TA, MacDonald M, et al.
Who needs a heart transplant?
 
Eur Heart J
 
2006
; 27: 770–2.

200 Hawkins NM, Petrie MC, Jhund PS, et al. Heart failure and chronic obstructive pumonary disease: diagnostic pitfalls and epidemiology. Eur J Heart Fail 2009; 11: 130–9.

201  

Binanay
C, Califf RM, Hasselblad V, et al. ESCAPE Investigators and ESCAPE Study Coordinators. Evaluation study of congestive heart failure and pulmonary artery catheterization effectiveness: the ESCAPE trial.
JAMA
 
2005
; 294: 1625–33.

202  

Cleland
JC, Pennell DJ, Ray SG, et al. Myocardial viability as a determinant of the ejection fraction response to carvedilol in patients with heart failure (CHRISTMAS trial): randomised controlled trial.
Lancet
 
2003
; 362: 14–21.

203  

Cooper
LT, Baughman KL, Feldman AM, et al. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology Endorsed by the Heart Failure Society of America and the Heart Failure Association of the European Society of Cardiology.
Eur Heart J
 
2007
; 28: 3076–93.

204  

Nohria
A, Tsang SW, Fang JC, et al. Clinical assessment identifies hemodynamic profiles that predict outcomes in patients admitted with heart failure.
J Am Coll Cardiol
 
2003
; 41: 1797–804.

205  

Pope
CA 3rd, Renlund DG, Kfoury AG, et al. Relation of heart failure hospitalization to exposure to fine particulate air pollution.
Am J Cardiol
 
2008
; 102: 1230–4.

206  

Stewart
S, McMurray JJ. Chilling findings: the need for winter vigilance in heart failure.
J Card Fail
 
2006
; 12: 120–1.

207  

Olsson
LG, Swedberg K, Ducharme A, et al. Atrial fibrillation and risk of clinical events in chronic heart failure with and without left ventricular systolic dysfunction: results from the Candesartan in Heart failure – Assessment of Reduction in Mortality and morbidity (CHARM) program. J Am Coll Cardiol
2006
; 47: 1997–2004.

208  

Rivero-Ayerza
M, Scholte Op Reimer W, Lenzen M, et al. New-onset atrial fibrillation is an independent predictor of in-hospital mortality in hospitalized heart failure patients: results of the EuroHeart Failure Survey.
Eur Heart J
 
2008
; 29: 1618–24.

209  

Levy
WC, Mozaffarian D, Linker DT, et al.
The Seattle Heart Failure Model: prediction of survival in heart failure.
 
Circulation
 
2006
; 113: 1424–33.

210  

Pocock
SJ, Wang D, Pfeffer MA, et al.
Predictors of mortality and morbidity in patients with chronic heart failure.
 
Eur Heart J
 
2006
; 27: 65–75.

211  

Latini
R, Masson S, Anand I, et al. The comparative prognostic value of plasma neurohormones at baseline in patients with heart failure enrolled in Val-HeFT.
Eur Heart J
 
2004
; 25: 292–9.

212  

Masson
S, Latini R, Anand IS; on behalf of the Val-HeFT Investigators. Prognostic value of changes in N-terminal pro-brain natriuretic peptide in Val-HeFT (Valsartan Heart Failure Trial). J Am Coll Cardiol  
2008
; 52: 997–1003.

213  

Goebel
JA, Van Bakel AB. Rational use of diuretics in acute decompensated heart failure.
Curr Heart Fail Rep
 
2008
; 5: 153–62.

214  

Faris
R, Flather M, Purcell H, et al. Current evidence supporting the role of diuretics in heart failure: a meta analysis of randomised controlled trials.
Int J Cardiol
 
2002
; 82: 149–58.

215  

Rosenberg
J, Gustafsson F, Galatius S, et al. Combination therapy with metolazone and loop diuretics in outpatients with refractory heart failure: an observational study and review of the literature.
Cardiovasc Drugs Ther
 
2005
; 19: 301–6.

216  

Iyengar
S, Abraham WT. Diuretic resistance in heart failure.
Curr Heart Fail Rep
 
2006
; 3: 41–5.

217 Costanzo MR. Ultrafiltration in the management of heart failure. Curr Opin Crit Care 2008; 14: 524–30.

218  

Pitt
B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med  
1999
; 341: 709–17.

219  

McMurray
J, Cohen-Solal A, Dietz R, et al. Practical recommendations for the use of ACE inhibitors, beta-blockers, aldosterone antagonists and angiotensin receptor blockers in heart failure: putting guidelines into practice.
Eur J Heart Fail
 
2005
; 7: 710–21.

220 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
; 316: 1429–35.

221 Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The SOLVD Investigators.

N Engl J Med
 
1991
; 325: 293–302.

222  

Flather
MD, Yusuf S, Køber L, et al. Long-term ACE-inhibitor therapy in patients with heart failure or left-ventricular dysfunction: a systematic overview of data from individual patients. ACE-Inhibitor Myocardial Infarction Collaborative Group.
Lancet
 
2000
; 355: 1575–81.

223  

Granger
CB, McMurray JJ, Yusuf S, et al. CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function intolerant to angiotensin-converting-enzyme inhibitors: the CHARM-Alternative trial.
Lancet.
 
2003
; 362: 772–6.

224  

Maggioni
AP, Anand I, Gottlieb SO, et al. Val-HeFT Investigators (Valsartan Heart Failure Trial). Effects of valsartan on morbidity and mortality in patients with heart failure not receiving angiotensin-converting enzyme inhibitors.
J Am Coll Cardiol
 
2002
; 40: 1414–21.

225  

Cohn
JN, Tognoni G; Valsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure.
N Engl J Med
 
2001
; 345: 1667–75.

226  

McMurray
JJ, Ostergren J, Swedberg K, et al. CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial.
Lancet
 
2003
; 362: 767–71.

227  

McKelvie
RS, Yusuf S, Pericak D, et al.
Comparison of candesartan,
enalapril, and their combination in congestive heart failure: randomized evaluation of strategies for left ventricular dysfunction (RESOLVD) pilot study. The RESOLVD Pilot Study Investigators.
Circulation
 
1999
; 100: 1056–64.

228  

McKelvie
RS, Rouleau JL, White M, et al.
Comparative impact of enalapril,
candesartan or metoprolol alone or in combination on ventricular remodelling in patients with congestive heart failure.
Eur Heart J
 
2003
; 24: 1727–34.

229  

Weir
RA, McMurray JJ, Puu M, et al. CHARM Investigators. Efficacy and tolerability of adding an angiotensin receptor blocker in patients with heart failure already receiving an angiotensin-converting inhibitor plus aldosterone antagonist, with or without a beta blocker. Findings from the Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity (CHARM)-Added trial. Eur J Heart Fail  
2008
; 10: 157–63.

230  

Desai
AS, Swedberg K, McMurray JJ, et al. CHARM Program Investigators. Incidence and predictors of hyperkalemia in patients with heart failure: an analysis of the CHARM Program. J Am Coll Cardiol  
2007
; 50: 1959–66.

231  

Juurlink
DN, Mamdani MM, Lee DS, et al. Rates of hyperkalemia after publication of the Randomized Aldactone Evaluation Study.
N Engl J Med
 
2004
; 351: 543–51.

232 The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial.

Lancet
 
1999
; 353: 9–13.

233 Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF).

Lancet
 
1999
; 353: 2001–7.

234  

Hjalmarson
A, Goldstein S, Fagerberg B, et al. Effects of controlled-release metoprolol on total mortality, hospitalizations, and well-being in patients with heart failure: the Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure (MERIT-HF). MERIT-HF Study Group.
JAMA
 
2000
; 283: 1295–302.

235  

Packer
M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group.
N Engl J Med
 
1996
; 334: 1349–55.

236  

Packer
M, Coats AJ, Fowler MB, et al. Carvedilol Prospective Randomized Cumulative Survival Study Group. Effect of carvedilol on survival in severe chronic heart failure.
N Engl J Med
 
2001
; 344: 1651–8.

237  

Packer
M, Fowler MB, Roecker EB, et al. Carvedilol Prospective Randomized Cumulative Survival (COPERNICUS) Study Group. Effect of carvedilol on the morbidity of patients with severe chronic heart failure: results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study.
Circulation
 
2002
; 106: 2194–9.

238  

Flather
MD, Shibata MC, Coats AJ, et al. SENIORS Investigators. Randomized trial to determine the effect of nebivolol on mortality and cardiovascular hospital admission in elderly patients with heart failure (SENIORS).
Eur Heart J
 
2005
; 26: 215–25.

239  

Poole-Wilson
PA, Swedberg K, Cleland JG, et al. Carvedilol Or Metoprolol European Trial Investigators. Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol Or Metoprolol European Trial (COMET): randomised controlled trial.
Lancet
 
2003
; 362: 7–13.

240  

Pitt
B, Remme W, Zannad F, et al.
Eplerenone
Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study Investigators. Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med  
2003
; 348: 1309–21.

241  

Hood
WB, Jr., Dans AL, Guyatt GH, et al. Digitalis for treatment of congestive heart failure in patients in sinus rhythm: a systematic review and meta-analysis.
J Card Fail
 
2004
; 10: 155–64.

242 The effect of digoxin on mortality and morbidity in patients with heart failure. The Digitalis Investigation Group. N Engl J Med  

1997
; 336: 525–33.

243  

Rathore
SS, Curtis JP, Wang Y, et al. Association of serum digoxin concentration and outcomes in patients with heart failure.
JAMA
 
2003
; 289: 871–8.

244  

Cohn
JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study.
N Engl J Med
 
1986
; 314: 1547–52.

245  

Cohn
JN, Johnson G, Ziesche S, et al. A comparison of enalapril with hydralazine-isosorbide dinitrate in the treatment of chronic congestive heart failure.
N Engl J Med
 
1991
; 325: 303–10.

246  

Taylor
AL, Ziesche S, Yancy C, et al. African-American Heart Failure Trial Investigators. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure.
N Engl J Med
 
2004
; 351: 2049–57.

247  

Gissi-Hf
Investigators. Effect of n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial.
Lancet
 
2008
; 372: 1223–30.

248  

Packer
M, O’Connor CM, Ghali JK, et al. Effect of amlodipine on morbidity and mortality in severe chronic heart failure. Prospective Randomized Amlodipine Survival Evaluation Study Group.
N Engl J Med
 
1996
; 335: 1107–14.

249  

Dorszewski
A, Göhmann E, Dorszewski B, et al. Vasodilation by urapidil in the treatment of chronic congestive heart failure in addition to angiotensin-converting enzyme inhibitors is not beneficial: results of a placebo-controlled, double-blind study. J Card Fail  
1997
; 3: 91–6.

250  

Einhorn
PT, Davis BR, Massie BM, et al. ALLHAT Collaborative Research Group. The Antihypertensive and Lipid Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) Heart Failure Validation Study: diagnosis and prognosis.
Am Heart J
 
2007
; 153: 42–53.

251  

Packer
M, McMurray J, Massie BM, et al. Clinical effects of endothelin receptor antagonism with bosentan in patients with severe chronic heart failure: results of a pilot study.
J Card Fail
 
2005
; 11: 12–20.

252  

Mann
DL, McMurray JJ, Packer M, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL).
Circulation
 
2004
; 109: 1594–602.

253  

Cohn
JN, Goldstein SO, Greenberg BH, et al. A dose-dependent increase in mortality with vesnarinone among patients with severe heart failure. Vesnarinone Trial Investigators.
N Engl J Med
 
1998
; 339: 1810–16.

254  

Cohn
JN, Pfeffer MA, Rouleau J, et al. MOXCON Investigators. Adverse mortality effect of central sympathetic inhibition with sustained-release moxonidine in patients with heart failure (MOXCON).
Eur J Heart Fail
 
2003
; 5: 659–67.

255  

van
Veldhuisen DJ, Poole-Wilson PA. The underreporting of results and possible mechanisms of ‘negative’ drug trials in patients with chronic heart failure.
Int J Cardiol
 
2001
; 80: 19–27.

256  

Packer
M, Carver JR, Rodeheffe RJ, et al. Effect of oral milrinone on mortality in severe chronic heart failure. The PROMISE Study Research Group.
N Engl J Med
 
1991
; 325: 1468–75.

257  

Køber
L, Torp-Pedersen C, McMurray JJ, et al. Dronedarone Study Group. Increased mortality after dronedarone therapy for severe heart failure.
N Engl J Med
 
2008
; 358: 2678–87.

258  

Kjekshus
J, Apetrei E, Barrios V, et al. CORONA Group.
Rosuvastatin in older patients with systolic heart failure.
 N Engl J Med  
2007
; 357: 2248–61.

259  

Gissi-Hf
Investigators. Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial.
Lancet
 
2008
; 372: 1231–9.

260 Home PD, Pocock SJ, Beck-Nielsen H, et al. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet  

2009
; Jun 5. [Epub ahead of print.]

261 Sochalski J, Jaarsma T, Krumholz HM, et al. What works in chronic care management: the case of heart failure. Health Aff (Millwood) 2009; 28: 179–89.

262  

Jaarsma
T, van der Wal MH, Lesman-Leegte I, et al. Coordinating Study Evaluating Outcomes of Advising and Counseling in Heart Failure (COACH) Investigators. Effect of moderate or intensive disease management program on outcome in patients with heart failure: Coordinating Study Evaluating Outcomes of Advising and Counseling in Heart Failure (COACH).
Arch Intern Med
 
2008
; 168: 316–24.

263  

Hauptman
PJ, Rich MW, Heidenreich PA, et al. Heart Failure Society of America. The heart failure clinic: a consensus statement of the Heart Failure Society of America.
J Card Fail
 
2008
; 14: 801–15.

264  

Clark
RA, Inglis SC, McAlister FA, et al. Telemonitoring or structured telephone support programmes for patients with chronic heart failure: systematic review and meta-analysis.
BMJ
 
2007
; 334: 942.

265  

Bourge
RC, Abraham WT, Adamson PB, et al. COMPASS-HF Study Group. Randomized controlled trial of an implantable continuous hemodynamic monitor in patients with advanced heart failure: the COMPASS-HF study.
J Am Coll Cardiol
 
2008
; 51: 1073–9.

266  

Edwardson
SR.
Patient education in heart failure.
 
Heart Lung
 
2007
; 36: 244–52.

267  

Granger
BB, Swedberg K, Ekman I, et al. CHARM investigators. Adherence to candesartan and placebo and outcomes in chronic heart failure in the CHARM programme: double-blind, randomised, controlled clinical trial.
Lancet
 
2005
; 366: 2005–11.

268 O’Connor CM,

Whellan
DJ, Lee KL, et al. Efficacy and safety of exercise training in patients with chronic heart failure: HF-ACTION randomized controlled trail. JAMA 2009; 301: 1439–50.

269 Flynn KE, Piña IL,

Whellan
DJ, et al. Effects of exercise training on health status in patients with chronic heart failure: HF-ACTION randomized controlled trail. JAMA 2009; 301: 1451–9.

270  

Guazzi
M. Sildenafil and phosphodiesterase-5 inhibitors for heart failure.
Curr Heart Fail Rep
 
2008
; 5: 110–14.

271  

Fox
K, Ford I, Steg PG, et al. on behalf of the BEAUTIFUL Investigators. Ivabradine for patients with stable coronary artery disease and left-ventricular systolic dysfunction (BEAUTIFUL): a randomised, double-blind, placebo-controlled trial.
Lancet
 
2008
; 372: 807–16.

272  

Roy
D, Talajic M, Nattel S, et al. Atrial Fibrillation and Congestive Heart Failure Investigators. Rhythm control versus rate control for atrial fibrillation and heart failure.
N Engl J Med
 
2008
; 358: 2667–77.

273  

Khan
MN, Jaïs P, Cummings J, et al. PABA-CHF Investigators. Pulmonary-vein isolation for atrial fibrillation in patients with heart failure.
N Engl J Med
 
2008
; 359: 1778–85.

274  

Zipes
DP, Camm AJ, Borggrefe M, et al. ACC/AHA/ESC
2006
guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: a report of the American College of Cardiology/American Heart Association Task Force and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop Guidelines for Management of Patients With Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death). Developed in collaboration with the European Heart Rhythm Association and the Heart Rhythm Society.
Europace
2006; 8: 746–837.

275  

Newman
CM, Price A, Davies DW. Amiodarone and the thyroid: a practical guide to the management of thyroid dysfunction induced by amiodarone therapy.
Heart
 
1998
; 79: 121–7.

276  

Riegger
GA, Haeske W, Kraus C, et al. Contribution of the renin-angiotensin-aldosterone system to development of tolerance and fluid retention in chronic congestive heart failure during prazosin treatment.
Am J Cardiol
 
1987
; 59: 906–10.

277  

van
Veldhuisen DJ, McMurray JJ; RED-HF Executive Committee. Are erythropoietin stimulating proteins safe and efficacious in heart failure? Why we need an adequately powered randomised outcome trial.
Eur J Heart Fail
 
2007
; 9: 110–12.

278  

O’Connor
CM, Jiang W, Kuchibhatla M, et al.
Antidepressant use,
depression, and survival in patients with heart failure.
Arch Intern Med
 
2008
; 168: 2232–7.

279  

Cardinale
D, Colombo A, Cipolla CM. Prevention and treatment of cardiomyopathy and heart failure in patients receiving cancer chemotherapy.
Curr Treat Options Cardiovasc Med
 
2008
; 10: 486–95.

280  

Moss
AJ, Zareba W, Hall WJ, et al. Multicenter Automatic Defibrillator Implantation Trial II Investigators. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction.
N Engl J Med
 
2002
; 346: 877–83.

281  

Bardy
GH, Lee KL, Mark DB, et al. Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT) Investigators. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med  
2005
; 352: 225–37.

282  

Kadish
A, Dyer A, Daubert JP, et al. Defibrillators in Non-Ischemic Cardiomyopathy Treatment Evaluation (DEFINITE) Investigators. Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy.
N Engl J Med
 
2004
; 350: 2151–8.

283  

Clark
AL, Goode K, Cleland JG. The prevalence and incidence of left bundle branch block in ambulant patients with chronic heart failure.
Eur J Heart Fail
 
2008
; 10: 696–702.

284  

Bristow
MR, Saxon LA, Boehmer J, et al. Comparison of Medical Therapy, Pacing, and Defibrillation in Heart Failure (COMPANION) Investigators. Cardiac-resynchronization therapy with or without an implantable defibrillator in advanced chronic heart failure. N Engl J Med  
2004
; 350: 2140–50.

285  

Cleland
JG, Daubert JC, Erdmann E, et al. Cardiac Resynchronization-Heart Failure (CARE-HF) Study Investigators. The effect of cardiac resynchronization on morbidity and mortality in heart failure.
N Engl J Med
 
2005
; 352: 1539–49.

286  

Hawkins
NM, Petrie MC, Burgess MI, et al. Selecting patients for cardiac resynchronization therapy: the fallacy of echocardiographic dyssynchrony. J Am Coll Cardiol 2009; 53: 1944–59.

287  

Chung
ES, Leon AR, Tavazzi L, et al.
Results of the Predictors of Response to CRT (PROSPECT) trial.
 
Circulation
 
2008
; 117: 2608–16.

288  

Linde
C, Abraham WT, Gold MR, et al. REVERSE (REsynchronization reVErses Remodeling in Systolic left vEntricular dysfunction) Study Group. Randomized trial of cardiac resynchronization in mildly symptomatic heart failure patients and in symptomatic patients with left ventricular dysfunction and previous heart failure symptoms. J Am Coll Cardiol  
2008
; 52: 1834–43.

289  

Beshai
JF, Grimm RA, Nagueh SF, et al. RethinQ Study Investigators. Cardiac-resynchronization therapy in heart failure with narrow QRS complexes.
N Engl J Med
 
2007
; 357: 461–71.

290  

Stanton
T, Hawkins NM, Hogg KJ, et al. How should we optimize cardiac resynchronization therapy?
Eur Heart J
 
2008
; 29: 2458–72.

291  

Rose
EA, Gelijns AC, Moskowitz AJ, et al. Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure (REMATCH) Study Group. Long-term mechanical left ventricular assistance for end-stage heart failure.
N Engl J Med
 
2001
; 345: 1435–43.

292 Jones RH, Velazquez EJ, Michler RE, et al. Coronary bypass surgery with or without surgical ventricular reconstruction.

N Engl J Med
2009; 360: 1705–17.

293  

Hernandez
AF, Shea AM, Milano CA, et al. Long-term outcomes and costs of ventricular assist devices among Medicare beneficiaries.
JAMA
 
2008
; 300: 2398–406.

294  

Lietz
K, Miller LW. Destination therapy: current results and future promise.
Semin Thorac Cardiovasc Surg
 
2008
; 20: 225–33.

295  

Hiestand
BC. Circulatory assist devices in heart failure patients.
Heart Fail Clin
 
2009
; 5: 55–62, vi.

296  

Hunt
SA, Haddad F. The changing face of heart transplantation.
J Am Coll Cardiol
 
2008
; 52: 587–98.

297  

Shah
SJ, Gheorghiade M. Heart failure with preserved ejection fraction: treat now by treating comorbidities.
JAMA
 
2008
; 300: 431–3.

298  

Hogg
K, McMurray J. The treatment of heart failure with preserved ejection fraction (‘diastolic heart failure’). Heart Fail Rev  
2006
; 11: 141–6.

299  

Yusuf
S, Pfeffer MA, Swedberg K, et al. CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and preserved left-ventricular ejection fraction: the CHARM-Preserved Trial.
Lancet
 
2003
; 362: 777–81.

300  

Cleland
JG, Tendera M, Adamus J, et al. PEP-CHF Investigators. The perindopril in elderly people with chronic heart failure (PEP-CHF) study.
Eur Heart J
 
2006
; 27: 2338–45.

301  

Massie
BM, Carson PE, McMurray JJ, et al. I-PRESERVE Investigators. Irbesartan in patients with heart failure and preserved ejection fraction.
N Engl J Med
 
2008
; 359: 2456–67.

302  

Gray
A, Goodacre S, Newby DE, et al. 3CPO Trialists.
Noninvasive ventilation in acute cardiogenic pulmonary edema.
 N Engl J Med  
2008
; 359: 142–51.

303  

Ezekowitz
JA, Hernandez AF, Starling RC, 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
; 157: 219–28.

304  

Wertman
BM, Gura V, Schwarz ER. Ultrafiltration for the management of acute decompensated heart failure.
J Card Fail
 
2008
; 14: 754–9.

305  

Elkayam
U, Ng TM, Hatamizadeh P, et al. Renal Vasodilatory Action of Dopamine in Patients With Heart Failure: Magnitude of Effect and Site of Action.
Circulation
 
2008
; 117: 200–5.

306  

Fonarow
GC, Abraham WT, Albert NM, et al. OPTIMIZE-HF Investigators and Coordinators. Carvedilol use at discharge in patients hospitalized for heart failure is associated with improved survival: an analysis from Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart Failure (OPTIMIZE-HF).
Am Heart J
 
2007
; 153: 82.e1–e11.

307  

Fonarow
GC, Abraham WT, Albert NM, et al.
Dosing of beta-blocker therapy before,
during, and after hospitalization for heart failure (from Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart Failure).
Am J Cardiol
 
2008
; 102: 1524–9.

308  

Phillips
CO, Wright SM, Kern DE, et al. Comprehensive discharge planning with post discharge support for older patients with congestive heart failure: a meta-analysis.
JAMA
 
2004
; 291: 1358–67.

309  

Stewart
S, Blue L, Walker A, et al. An economic analysis of specialist heart failure nurse management in the UK; can we afford not to implement it?
Eur Heart
 
2002
; 23: 1369–78.

310  

Stewart
S, McMurray JJ. Palliative care for heart failure.
BMJ
 
2002
; 325: 915–16.

311  

Allen
LA, Yager JE, Funk MJ, et al. Discordance between patient-predicted and model-predicted life expectancy among ambulatory patients with heart failure.
JAMA
 
2008
; 299: 2533–42.

312 Jaarsma T, Beattie JM, Ryder M, et al. Palliative care in heart failure: a position statement from the palliative care workshop of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2009; 11: 433–43.

Useful patient and carer orientated material is available from the following, and other, organizations:

graphic Heart Failure Association of the European Society of Cardiology in several languages: http://www.heartfailurematters.org/English_Lang/Pages/index.aspx

graphic Heart Failure Society of America: http://www.hfsa.org/hf_modules.asp

graphic National Gold Standards Framework (GSF) Centre England: http://www.goldstandardsframework.nhs.uk/index.php

graphic For full references and multimedia materials please visit the online version of the book (http://esctextbook.oxfordonline.com).

Close
This Feature Is Available To Subscribers Only

Sign In or Create an Account

Close

This PDF is available to Subscribers Only

View Article Abstract & Purchase Options

For full access to this pdf, sign in to an existing account, or purchase an annual subscription.

Close