* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project
Download Dilated Cardiomyopathy
Electrocardiography wikipedia , lookup
Heart failure wikipedia , lookup
Cardiac surgery wikipedia , lookup
Antihypertensive drug wikipedia , lookup
Remote ischemic conditioning wikipedia , lookup
Coronary artery disease wikipedia , lookup
Hypertrophic cardiomyopathy wikipedia , lookup
Cardiac contractility modulation wikipedia , lookup
Arrhythmogenic right ventricular dysplasia wikipedia , lookup
5 5 Dilated Cardiomyopathy Biykem Bozkurt and Douglas L. Mann Definition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Epidemiology of Dilated Cardiomyopathy . . . . . . . . . . Natural History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1233 1234 1235 1236 Key Points • Dilated cardiomyopathy (DCM) refers to a spectrum of heterogenous myocardial disorders that are characterized by ventricular dilation and depressed myocardial contractility in the absence of abnormal loading conditions (such as hypertension or valvular disease) or ischemic heart disease sufficient to cause global systolic impairment. • The dilated cardiomyopathies constitute the largest group of myopathic disorders that are responsible for systolic heart failure. The reported incidence of DCM varies annually from about five to eight cases per 100,000 of the population. In most multicenter, randomized trials in heart failure, approximately one third of the enrolled patients have nonischemic dilated cardiomyopathy. • Once DCM patients become symptomatic, the available evidence suggests that the prognosis is relatively poor, with 25% mortality at 1 year and 50% mortality at 5 years. More recent studies suggest that the prognosis for patients with DCM and mild left ventricular dilation may be more favorable, perhaps reflecting earlier diagnosis and better treatment. • Ventricular enlargement, ejection fraction, persistent S3 gallop, right-sided heart failure, elevated left ventricle (LV) filling pressures, mitral regurgitation, pulmonary hypertension, electrocardiogram (ECG) fi ndings of atrioventricular block or intraventricular conduction delay, recurrent ventricular tachycardia, elevated levels of neurohormones, cytokines or markers of myocardial cell death, impaired peak oxygen consumption, hyponatremia, New York Heart Association (NYHA) class, age >64 years are among a number of parameters that predict a poor prognosis in patients with DCM. • The most important point in the treatment of DCM patients would be to determine the underlying etiology for which there may be a specific form of treatment. The differential treatment benefit seen in DCM patients compared with patients with ischemic cardiomyopathy that Myocardial Diseases Presenting as Dilated Cardiomyopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1237 Clinical Recognition . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1247 Treatment Strategies for Dilated Cardiomyopathy . . . 1249 has been observed in several randomized clinical trials such as with digoxin or amiodarone suggests that there may be therapeutic differences between ischemic and nonischemic heart failure (HF). • Existing studies suggest that both patients with ischemic cardiomyopathy or DCM have significant survival benefit from the use of angiotensin-converting enzyme (ACE) inhibitors and beta-blockers. • Similarly, in patients with advanced heart failure, aldosterone antagonism with spironolactone or eplerenone has been reported to reduce mortality; and angiotensin receptor blockade, when added to background therapy with ACE-inhibitors, have been shown to reduce combined morbidity and mortality, both in dilated or ischemic cardiomyopathy patients. • The role of implantable defibrillators for secondary prevention of sudden cardiac death or the use of resynchronization therapy with biventricular pacing to improve outcomes in a subgroup of patients is clearly established both for ischemic and nonischemic cardiomyopathy patients; however, similar evidence supporting the role of prophylactic implantable cardiodefibrillator (ICD) implantation in patients with DCM is controversial. • Treatment modalities using growth hormone, gene therapy, angiogenesis or cell therapy, immunosuppression, or immunomodulation are promising but remain investigational at the present time. Definition The term dilated cardiomyopathy (DCM) refers to a spectrum of heterogeneous myocardial disorders (Table 55.1) that are characterized by ventricular dilation and depressed myocardial contractility in the absence of abnormal loading conditions (such as hypertension or valvular disease) or ischemic heart disease sufficient to cause global systolic impairment.1,2 The dilated cardiomyopathies constitute the largest group of myopathic disorders that are responsible for systolic heart 12 3 3 CAR055.indd 1233 11/24/2006 1:28:32 PM 12 3 4 chapter TABLE 55.1. Etiologies of dilated cardiomyopathy Idiopathic Idiopathic dilated cardiomyopathy Idiopathic arrhythmogenic right ventricular dysplasia Familial (hereditary) Autosomal dominant X-chromosomal Polymorphism Toxic Ethanol Cocaine Adriamycin Catecholamine excess Phenothiazines, antidepressants Cobalt Carbon monoxide Lead Lithium Cyclophosphamide Methysergide Inflammatory: infectious etiology Viral (Coxsackie virus, parvovirus, adenovirus, echovirus, influenza virus, HIV) Spirochete (leptospirosis, syphilis) Protozoal (Chagas’ disease, toxoplasmosis, trichinosis) Inflammatory: noninfectious etiology Collagen vascular disease (scleroderma, lupus erythematosus, dermatomyositis, rheumatoid arthritis) Kawasaki disease Hypersensitivity myocarditis Miscellaneous acquired cardiomyopathy Postpartum cardiomyopathy Obesity Metabolic/nutritional Thiamine Kwashiorkor pellagra Scurvy Hypervitaminosis D Selenium deficiency Carnitine deficiency Endocrine Diabetes mellitus Acromegaly Thyrotoxicosis Myxedema Uremia Cushing’s disease Pheochromocytoma Electrolyte imbalance Hypophosphatemia Hypocalcemia Physiologic agents Tachycardia Heat stroke Hypothermia Radiation Autoimmune disorders failure. Indeed, more than 75 specific diseases can produce a dilated cardiomyopathic phenotype. Thus, DCM can be envisioned as the final common pathway for a myriad of cardiac disorders that either damage the heart muscle or, alternatively, disrupt the ability of the myocardium to generate force and subsequently cause chamber dilation. In clinical practice and recent multicenter trials in heart failure (HF), the etiology of HF has often been categorized as CAR055.indd 1234 55 either ischemic or nonischemic cardiomyopathy, and the term dilated cardiomyopathy has been interchangeably used with nonischemic cardiomyopathy. Though this approach may be practical, it has certain drawbacks.3,4 It fails to recognize that the term nonischemic cardiomyopathy may include cardiomyopathies due to volume or pressure overload—such as hypertension or valvular heart disease—that are not conventionally accepted under the definition of DCM.2 Epidemiology of Dilated Cardiomyopathy The reported incidence of DCM varies annually from about five to eight cases per 100,000 of the population. The true incidence may be underestimated due to underreporting or underdetection of asymptomatic cases of DCM, which may occur in as many as 50% to 60% of patients. In most multicenter randomized trials in HF, approximately one third of the enrolled patients have nonischemic DCM.5–7 The ageadjusted prevalence of DCM in the United States averages 36 cases per 100,000 population and DCM accounts for 10,000 deaths annually.8 Compared with whites, African Americans have almost a threefold increase in risk for developing DCM. This increased risk for developing DCM in African Americans is not explained by differences in hypertension, cigarette smoking, alcohol use, or socioeconomic factors.9,10 Moreover, African Americans have approximately a 1.5- to 2.0-fold higher risk of dying from DCM when compared to age-matched whites with DCM.11–13 Although the reason(s) for these differences are not known, there are several potential explanations including differences in the number of risk factors for development of HF, in the rate of progression of HF, the etiology of HF, access to medical care, and response to therapy. Data from the second Vasodilator–Heart Failure Trial (V-HeFT II),14 Beta-Blocker Evaluation of Survival Trial,15,16 and Studies of Left Ventricular Dysfunction (SOLVD)17 suggest that AfricanAmerican patients with HF may not benefit from commonly used doses of currently recommended therapies to the same extent as white patients, although unanimity of opinion does not exist with regard to this issue.18 In general, HF is more common in men than women. Epidemiologic data suggesting gender-related differences in the occurrence and prognosis of HF are conflicting and may be confounded both by differing etiologies of HF. The overall effect of female gender on the prognosis of HF is not clear at this time, in large measure because many of the early clinical trials in HF have been comprised predominantly of male subjects. Women with HF in the Framingham Heart Study (even after controlling for age and etiology of HF)19 as well as women in National Health and Nutrition Examination Survey-I (NHANES-I)20 had improved survival compared with men. Studies of Left Ventricular Dysfunction, in which only 15% of the patients were women, reported no difference in gender related survival.21 However, the SOLVD registry, which included approximately 20% female patients, suggested that women had significantly higher annual risk of HF-related mortality and higher rates of hospitalization than age-matched male subjects.22 In the Italian Multicenter Cardiomyopathy Registry, women with idiopathic DCM presented with more advanced HF; and had a trend toward worse 11/24/2006 1:28:32 PM 12 3 5 di l at e d c a r diom yopat h y Natural History The natural history of DCM is not well established for two reasons: first, as noted at the outset, DCM represents a heterogeneous spectrum of myocardial disorders that may each progress at different rates; second; the onset of the disease may be insidious, particularly in the case of familial or idiopathic dilated cardiomyopathies. Indeed, approximately 4% to 13% of patients with DCM present with asymptomatic left ventricular dysfunction and left ventricular dilatation. For these patients, the overall prognosis is unclear. However, once DCM patients become symptomatic, the available evidence suggests that the prognosis is relatively poor, with 25% mortality at 1 year and 50% mortality at 5 years (Fig. 55.1).26 The cause of death appears to be primarily pump failure in approximately 70% of patients with DCM, whereas sudden cardiac death accounts for approximately 30% of all deaths in patients with DCM.33,34 However, it should be recognized that many of the natural history studies of DCM were performed before angiotensin-converting enzyme (ACE) inhibitors and beta-blockers were routinely used. More recent studies suggest that the prognosis for patients with DCM and mild left ventricular dilation may be more favorable, perhaps reflecting earlier diagnosis and better treatment.28,34 Furthermore, it should be recognized that approximately 25% of CAR055.indd 1235 100 80 Survival (%) survival.23 Analyses from Metoprolol Extended-Release Randomized Intervention Trial in Heart Failure (MERIT-HF)24 and Cardiac Insufficiency Bisoprolol Study (CIBIS II)25 betablocker treatment trials in HF suggest that female gender may be a significant independent predictor of survival in patients with HF, independent of ischemic or nonischemic etiology. Thus, further studies will be necessary to defi ne the role of gender in the prognosis of DCM. Advancing age is a risk factor for mortality in HF. As reported in the SOLVD registry, the risk of death at 1 year for a person over the age of 64 years with HF was 1.5 times greater than for subjects who were ≥64 years of age.22 Advancing age has been reported as an independent risk factor for mortality in DCM in several studies.26,27 Indeed, Sugrue et al.28 reported the relative risk of dying from HF increases by 0.51 with every 10 years of increase in age. The prevalence, etiology, and prognosis of DCM can differ according to the geographic location and sociodemographics of the affected populations. For example, in Western developed countries, dilated nonischemic cardiomyopathy accounts for approximately 30% to 40% of the HF population, of which idiopathic cardiomyopathy is the most frequent etiology. The prevalence of DCM in Japan appears to be about half,29 and in Africa and Latin America approximately double, that of the Western populations.30 As populations go through epidemiologic, socioeconomic transitions and health care modifications, the features of DCM will continue to change.31 Failure of treatment and eradication of AIDS globally and especially in Africa, the rising prevalence of ethanol or substance abuse in Western countries, the development of new metabolic and dietary trends in North America,32 and the success in treatment of protozoal diseases in Latin America will continue to play a dynamic role in the epidemiology of DCM. A (n = 50) B (n = 40) 60 C (n = 201) 40 D (n = 169) E (n = 104) F (n = 77) G (n = 152) 20 0 1 2 3 4 5 6 7 8 9 10 Years after presentation FIGURE 55.1. Survival of patients with idiopathic dilated cardiomyopathy in seven published series (A to G). n, number of patients enrolled. To identify each specific series, see Dec and Fuster.26 DCM patients with the recent onset of symptoms of HF will improve spontaneously, including patients who have been referred for cardiac transplantation.35 This statement notwithstanding, patients with symptoms lasting more than 3 months who present with severe clinical decompensation generally have less chance of recovery.35 As shown in Table 55.2 there are a number of other parameters that predict a poor prognosis in patients with TABLE 55.2. Factors predicting a poor prognosis in patients with dilated cardiomyopathy Left ventricular enlargement Right ventricular enlargement Left and right ventricular ejection fraction Elevated LV fi lling pressures Persistent S3 gallop Right-sided heart failure Pulmonary hypertension Moderate to severe mitral regurgitation ECG fi ndings: fi rst- or second-degree atrioventricular block, or LBBB Recurrent ventricular tachycardia Reduced heart rate variability Late potentials of QRS in signal average ECG Myocytolysis on endomyocardial biopsy Elevated levels of neurohormones [brain natriuretic peptide (BNP), norepinephrine (NE), plasma renin activity (PRA) and endothelin 1 (ET-1)] Elevated levels of cytokines (TNF-α and IL-6) Elevation of serum CK/MB, troponin T, troponin I levels Peak oxygen consumption <10 to 12 mL/kg/min Reduced contractile response with dobutamine Serum sodium <137 mmol/L Advanced New York Heart Association class Advanced age (>64 years) 11/24/2006 1:28:32 PM 12 3 6 chapter DCM, including left ventricular (LV) and right ventricular (RV) enlargement,26,27,36,37 LV and RV ejection fraction,38 persistent S3 gallop, right-sided HF, elevated LV filling pressures,39 moderate to severe mitral regurgitation,40 pulmonary hypertension, electrocardiogram (ECG) fi ndings of first- or second-degree atrioventricular block, left bundle branch block (LBBB) or marked intraventricular conduction delay, recurrent ventricular tachycardia,41 reduced heart rate variability,42 late potentials of QRS in signal average ECG,43 myocytolysis on endomyocardial biopsy,44 elevated levels of neurohormones (norepinephrine, plasma renin activity, atrial natriuretic peptide, brain natriuretic peptide and endothelin1),45,46 elevated levels of cytokines (tumor necrosis factor-α and interleukin-6),47,48 persistently elevated markers of myocardial cell death (elevation of serum troponin T levels),49 an increased ratio of creatine kinase MB2/MB1,50 peak oxygen consumption <10 to 12 mL/kg/min,51–53 serum sodium <137 mmol/L,54 New York Heart Association class rating,55 age >64 years,22 and reduced contractile response with dobutamine.56–60 Concomitant renal or hepatic dysfunction may limit the use of optimal diuretics or ACE inhibitors and therefore may have an impact on prognosis that is not easily appreciated. Finally, it should be emphasized that comorbid conditions such as diabetes and hypertension increase the risk of developing HF approximately fivefold. Ischemic vs. Dilated Cardiomyopathy In general practice and clinical research trials, the term ischemic cardiomyopathy is defined as cardiomyopathy due to ischemic heart disease. The classification of cardiomyopathies proposed by the World Health Organization/International Society and Federation of Cardiology (WHO/ISFC) Task Force in 1995,2 however, defi nes ischemic cardiomyopathy as “a dilated cardiomyopathy with impaired contractile performance not explained by the extent of coronary disease or ischemic damage” reserved for the remodeling process of the noninfarcted myocardium. In this chapter, we use the term ischemic cardiomyopathy defined as cardiomyopathy due to ischemic heart disease rather than the WHO/ISFC task force definition. The existing clinical studies suggest that patients with idiopathic DCM have a lower total mortality.61 The risk of sudden cardiac death, however, appears to be relatively higher in patients with DCM in some studies.61 The differential treatment benefit seen in DCM patients compared with patients with ischemic cardiomyopathy that has been observed in several randomized clinical trials such as with digoxin62,63 or amiodarone64 suggest that there may be therapeutic differences between ischemic and nonischemic HF. Similar earlier reports of survival difference with betablockers7 or amlodipine65 in patients with dilated but not ischemic cardiomyopathy have not been reproduced in subsequent large-scale randomized trials,5,7,66 in which the benefit was similar in both the ischemic and nonischemic HF patients. This has raised the question of whether there truly is a difference in response to treatment according to etiology of HF. On the other hand, the absence of a rigorous definition of nonischemic HF in many studies may account for this discrepancy and make interpretation of the results CAR055.indd 1236 55 difficult. Further studies to clarify the effects of the etiology of HF could be particularly important to achieve further treatment benefit over and above the conventional treatment strategies that target HF as a single disease entity. Pathophysiology Dilated cardiomyopathy may be viewed as a progressive disorder that is initiated after an “index event” either damages the heart muscle, with a resultant loss of functioning cardiac myocytes, or alternatively disrupts the ability of the myocardium to generate force, thereby preventing the heart from contracting normally. This index event may have an abrupt onset, as in the case of acute exposure to toxins, or it may have a gradual or insidious onset, as in the case of hemodynamic pressure or volume overloading, or it may be hereditary, as in the case of many of the familial cardiomyopathies. Regardless of the nature of the inciting event, the feature that is common to each of these index events is that they all, in some manner, produce a decline in pumping capacity of the heart. The anatomic and pathophysiologic abnormalities that occur in LV remodeling are discussed in another chapter. Patients with DCM generally present with dilation of all four chambers of the heart (Fig. 55.2). Despite the fact that there is thinning of the left ventricular wall in patients with DCM, there is massive hypertrophy at the level of the intact heart, as well as at the level of the cardiac myocyte, which has a characteristic elongated appearance that is observed in myocytes obtained from hearts subjected to chronic volume overload (Fig. 55.3). The coronary arteries are usually normal in DCM, although it should be emphasized that the end-stage FIGURE 55.2. Pathology of normal heart (left) and a dilated cardiomyopathic ventricle (right). The dilated cardiomyopathic ventricle is characterized by enlargement of all four cardiac chambers, and a more spherical shape, in comparison to the normal ventricle. 11/24/2006 1:28:32 PM di l at e d c a r diom yopat h y 12 37 Idiopathic Dilated Cardiomyopathy A B FIGURE 55.3. Cardiac myocyte structure (A) in normal myocardium and in dilated cardiomyopathy (B). Cardiac myocytes isolated from myocardium from patients with DCM have an elongated shaped as the result of the sarcomeres being formed in series. Although the term idiopathic DCM has become synonymous with that of DCM in some HF parlance, the word idiopathic was originally intended to characterize the subset of DCM patients in whom no known etiologic cause for ventricular dilation and depressed myocardial contractility was apparent. However, with increasing sophistication in diagnostic testing, clinicians have become aware that many cases of so-called idiopathic DCM may occur as the result of inherited or spontaneous mutations of genes that regulate cardiac structure or function, such as the genes for cytoskeletal proteins, or as a consequence of undiagnosed hypertension, autoimmune diseases, viral illness, or toxin exposure. Nonetheless, in the context of this chapter, we use the term idiopathic DCM to refer to those patients with DCM whose etiologic cause remains unknown. As shown in Figure 55.4, idiopathic DCM represents the largest subset of patients with DCM; however, as alluded to immediately above, it is likely that the proportion of patients with idiopathic DCM will diminish with increased sophistication in diagnostic testing. Familial Cardiomyopathy “ischemic cardiomyopathies” may also present with a dilated phenotype. The cardiac valves are anatomically normal; however, there is usually tricuspid and mitral annular dilatation due to cavity enlargement, distortion of subvalvular apparatus, and stretching of the papillary muscles giving rise to valvular regurgitation. Intracavitary thrombi are common, usually in the ventricular apex. Myocardial Diseases Presenting as Dilated Cardiomyopathy Alcoholic/Toxic Cardiomyopathy As shown in Figure 55.4, the most common causes of DCM are idiopathic, alcoholic/toxic, inflammatory, familial, and miscellaneous (inflammatory noninfections, acquired, metabolic, and nutritional) causes. However, it should be recognized that the exact prevalence of the various forms of DCM varies based on the demographics of the patient population. In the subsections that follow, we review that various specific etiologies that lead to the development of DCM. Idiopathic Viral Alcoholic/toxic Miscellaneous Familial FIGURE 55.4. Etiologies of dilated cardiomyopathy. As shown, the most frequent causes of DCM are idiopathic dilated cardiomyopathy, familial cardiomyopathy, alcoholic/toxic cardiomyopathy, viral cardiomyopathy, and miscellaneous (inflammatory noninfections, acquired, metabolic, and nutritional causes). CAR055.indd 1237 There is growing evidence that many cases of previously diagnosed “idiopathic” DCM have a genetic basis. Indeed, although the familial occurrence of idiopathic DCM was first recognized by Battersby and Glenner in 1961,67 it has not been until recently that investigators have been able to delineate the molecular basis for genetic cardiomyopathies. It is estimated that at least 20% to 30% of DCM cases are familial.68 Alcoholic Cardiomyopathy Chronic alcoholism is one of the most important causes of DCM in the Western world.69 It is estimated that two thirds of the adult population use alcohol to some extent and more than 10% are heavy users.70 In the United States, in both sexes and all races, long-term heavy alcohol consumption (of any beverage type) has been cited by some investigators as the leading cause of a nonischemic DCM.69 A large proportion of chronic alcoholics demonstrate impairment of cardiac function. The clinical diagnosis of alcoholic cardiomyopathy can be made when biventricular dysfunction and dilation are persistently observed in a heavy drinker, in the absence of other known causes for myocardial disease.71 Thus, the diagnosis of alcoholic cardiomyopathy remains a diagnosis of exclusion. The prevalence of alcoholic cardiomyopathy is variable and ranges from 23% to 40%. In some series, as many as 21% of subjects with excessive alcohol intake were reported to have clinical evidence of HF.72 Alcoholic cardiomyopathy most commonly occurs in men 30 to 55 years of age who have been heavy consumers of alcohol for more than 10 years.73 Women represent approximately 14% of the alcoholic cardiomyopathy cases, but women may be more vulnerable to the development of alcohol-related cardiomyopathy, since it has been reported that alcoholic cardiomyopathy develops in 11/24/2006 1:28:33 PM 12 3 8 chapter women with a lower total lifetime exposure to alcohol compared to men.69 In all races, death rates due to alcoholic heart muscle disease are greater in men compared to women, and are greater in African-American men and women compared to white men and women with alcoholic cardiomyopathy.69 Even before the clinically overt HF, LV contractile dysfunction can be demonstrated in alcoholics. Although chronic alcoholic liver disease and HF usually are not observed clinically in the same patient, cirrhotic patients may present with asymptomatic LV dysfunction, in which case atrial fibrillation is usually the initial presenting manifestation of cardiac dysfunction.74 The point at which these abnormalities appear during the course of an individual’s lifetime of drinking, such that the abnormalities can be called a DCM, is not well established and is highly individualized. The risk of developing alcoholic cardiomyopathy is related to both the mean daily alcohol intake and the duration of drinking. In general, alcoholic patients consuming >90 g of alcohol a day (approximately seven to eight standard drinks per day) for more than 5 years are at risk for the development of asymptomatic alcoholic cardiomyopathy. Those who continue to drink may become symptomatic and develop signs and symptoms of HF. However, it bears emphasis that there is significant individual susceptibility to the toxic effects of alcohol. For example, it is not known whether the persistent DCM that develops in only 1% to 2% of chronic drinkers occurs because of genetic predisposition or because of the presence of synergistic cardiovascular risk factors such as hypertension or arrhythmias. Studies in experimental animals have demonstrated that both acute and chronic ethanol administration impairs cardiac contractility. Alcohol results in acute as well as chronic depression of myocardial contractility even when ingested by normal individuals in quantities consumed during social drinking.75 Compensatory mechanisms such as vasodilation or sympathetic stimulation may mask the direct acute myocardial depressant effects of alcohol. Despite the known deleterious effects of alcohol, it has been difficult to produce HF in animal models in which ethanol has been administered. Thus, the direct causal relationship between alcohol consumption and the development of cardiomyopathy has not been rigorously demonstrated in experimental models, despite the long-recognized clinical relationship between alcohol consumption and the development of DCM. The pathophysiology and progression of alcoholic cardiomyopathy is complex and involves changes in many aspects of myocyte function. The potential mechanisms that have been invoked to explain the depressed myocardial function include the direct toxic effects of alcohol on striated muscle, since most alcoholics have manifestations of skeletal myopathy and cardiomyopathy.76,77 Shifts in the relative expression of the contractile proteins β-myosin heavy chain (β-MHC) to αMHC have been reported in animal models after exposure to ethanol.69 Alcohol and its metabolite acetaldehyde can also cause alterations in cellular calcium, magnesium, or phosphate homeostasis. The toxic effects of acetaldehyde or the formation of fatty acid ethyl esters may also impair mitochondrial oxidative phosphorylation. In acute ethanol toxicity, free radical damage or ischemia may occur, possibly due to increased xanthine oxidase activity or β-adrenergic stimu- CAR055.indd 1238 55 lation, respectively.78 In addition, ethanol and its metabolites interfere with numerous membrane and cellular functions such as transport and binding of calcium, mitochondrial respiration, lipid metabolism, myocardial protein synthesis, signal transduction, and excitation contraction coupling.70,76,79 Both autopsy and endomyocardial biopsy specimens from alcoholic cardiomyopathy patients reveal marked mitochondrial swelling, with fragmentation of cristae, swelling of endoplasmic reticulum, cytoskeletal disorganization, and destruction of myofibrils.78 Several studies suggest that heavy drinking alters both lymphocyte and granulocyte production and function, raising the possibility that myocardial damage secondary to prolonged alcohol consumption might initiate autoreactive mechanisms comparable to those observed in viral or idiopathic myocarditis. The point at which the changes in mitochondrial, sarcoplasmic reticulum, contractile protein, and calcium homeostasis culminate in intrinsic cell dysfunction is incompletely understood. There are several early reports that support the role for myocyte loss as a final mechanism underlying alcohol-induced cardiac dysfunction.80 Apoptosis has been proposed as a critical mechanism for the development of fetal alcohol syndrome.81 Application of insulin-like growth factor-1 (IGF-1) has been reported to attenuate the apoptotic effects of ethanol in primary neonatal myocyte cell cultures.69 Recent reports suggest that certain genetic traits influence the occurrence, pathogenesis, and progression of alcoholic cardiomyopathy, which may explain interindividual variations in the sensitivity of the myocardium to alcoholinduced myocardial damage. These include multiple point mutations in the mitochondrial DNA, which have been associated with the occurrence of other cardiomyopathies.82 In addition, nutritional deficiencies, commonly thiamine deficiency, may play an additive role to the direct myocardial damage of ethanol. Thus, the cardiomyopathy that develops following chronic alcohol consumption may be multifactorial in origin. The management of patients with alcohol cardiomyopathy begins with total abstinence from alcohol, in addition to the conventional management of HF, as described below. There are currently no studies of specific pharmacotherapies in patients with alcoholic cardiomyopathy other than the standard therapy for HF; however, there are numerous reports that detail the reversibility of depressed left ventricular dysfunction after the cessation of drinking.83,84 Even if the depressed left ventricular function does not normalize completely, the symptoms and signs of congestive HF improve after abstinence.84 However, the overall prognosis remains poor, with a mortality of 40% to 50% within 3 to 6 years, if the patient is not abstinent.69 Survival is significantly lower for patients who continue to drink compared to patients with idiopathic DCM or alcoholic cardiomyopathy patients who abstain.69,85 Cocaine Cardiomyopathy Long-term abuse of cocaine, a drug that causes postsynaptic nervous system norepinephrine reuptake blockade and possible presynaptic release of dopamine and norepinephrine, may eventuate in DCM even without the presence of coronary artery disease, vasculitis, or regional myocardial injury. 11/24/2006 1:28:33 PM di l at e d c a r diom yopat h y This has been termed “cocaine-related cardiomyopathy” and perhaps reflects the direct toxicity of the cocaine on the myocardium. In 84 asymptomatic cocaine abusers studied at least 2 weeks after abstinence from the drug, Bertolet and colleagues86 found that 7% had left ventricular dysfunction. These patients were young and did not have evidence of myocardial infarction or coronary arterial disease. Depressed LV function (LV ejection fraction <45%) has been reported in 4% to 9% of asymptomatic cocaine abusers.87,88 Similarly, Moliterno et al.89 reported 18% of cocaine abusers who underwent cardiac catheterization had normal coronary arteries with low ejection fraction and global hypokinesia. The cardiovascular evaluation of a patient with cocaine abuse usually reflects the following. The ECG may reveal increased QRS voltage, early repolarization, ischemic or nonspecific ST-T changes, or pathologic Q waves. Episodes of ST elevation may be seen during Holter monitoring. The echocardiogram usually reveals left ventricular hypertrophy. Segmental wall motion abnormalities usually suggest myocardial injury; however, as mentioned above, approximately 18% of patients with cocaine abuse manifest global hypokinesia and depressed myocardial function. Cardiac catheterization in these patients may reveal normal coronaries or mild coronary artery disease not significant enough to explain the extent of myocardial dysfunction. However, accelerated coronary atherosclerosis, coronary vasculitis, coronary spasm, or coronary thrombosis are perhaps more common fi ndings in cocaine-related heart disease. Cocaine may produce left ventricular dysfunction through its direct toxic effects on the myocardium, by provoking coronary arterial spasm (and hence myocardial ischemia), and by causing increased release of catecholamines, which may be directly toxic to cardiac myocytes.90 These effects decrease myocardial oxygen supply and may increase demand if heart rate and blood pressure rise. The vasoactive effects of cocaine are further complicated with enhanced platelet aggregation, anticardiolipin antibody formation, and endothelial release of potent vasoconstrictors such as endothelin-1. Upregulation of tissue plasminogen activator inhibitors, increased platelet aggregation, and decreased fibrinolysis by cocaine predispose to coronary thrombosis and/or microvascular disease.87,91 Myocarditis with inflammatory lymphocyte and eosinophils has also been reported in 20% of patients dying of natural or homicidal causes in whom cocaine was detected, raising the possibility of hypersensitivity myocarditis due to cocaine or associated contaminants.92 Alternatively, the myocarditis may occur directly in response to the necrosis that is caused by cocaine. Scattered foci of myocyte necrosis, contraction band necrosis, and foci of myocyte fibrosis have been reported in patients with cocaine abuse. Additionally, experimental studies and clinical case reports suggest that cocaine may also cause lethal arrhythmias. Cocaine prolongs repolarization by a depressant effect on potassium current and may generate early afterdepolarizations.87 Other than abstinence, very little is known about the treatment of cocaine-induced cardiac dysfunction. Indeed, there are case reports of reversibility of cardiac function after cessation of drug use.93 In patients who develop cardiomyopathy, the traditional therapy for LV dysfunction is appropriate. Given that some of the toxicity of cocaine is caused CAR055.indd 1239 12 3 9 by catecholamine excess or myocardial ischemia, the use of β-adrenergic blocking agents appeared to be a logical treatment, both in terms of preventing further disease progression as well as treating the ventricular arrhythmias that are prone to develop in this setting. Two decades ago, the treatment of cocaine-induced cardiovascular effects favored the use of beta-blockers, especially propranolol. As the clinical use of propranolol increased, reports of accentuation of cocaineinduced hypertension and myocardial ischemia began to surface, blaming the unopposed alpha effects of the betablockers. Although these reports were isolated, the routine use of propranolol and subsequently all β-adrenergic blockers decreased to the point that beta-blockers were considered relatively contraindicated in treating cocaine-induced cardiovascular emergencies. The end result is that an entire generation of potent and selective β-adrenergic blocking agents have been overlooked, for both acute and chronic treatment of cocaine-related cardiac disease, all in the name of “unopposed alpha effects.” The focus of treatment shifted from the cardiovascular effects to combating central nervous stimulation. As a result, benzodiazepines have been the drug of choice in treating the cerebrovascular and subsequent systemic hyperadrenergic complications of cocaine, and nitroprusside or phentolamine has been advocated for peripheral vasodilatory effects. It is now becoming apparent that the treatment of cardiovascular effects of cocaine should involve a multifactorial approach to combat both central nervous system and peripheral vasospastic effects of cocaine. In this regard, β-adrenergic blocking agents, especially betablocking agents with both alpha- and beta-blocking properties such as labetalol or carvedilol, may play an important role in treating cocaine-related cardiomyopathies.94 Anthracycline Anthracyclines, such as doxorubicin (Adriamycin) and daunorubicin, produce cardiac toxicity, possibly by increasing oxygen free radical generation, platelet-activating factor, prostaglandins, histamine, calcium, and C-13 hydroxy metabolites, or by interfering with sarcolemmal sodium potassium pump and mitochondrial electron transport chain.95 The formation of oxygen free-radicals that are generated by iron-catalyzed pathways appears to be the most important pathway in the pathogenesis of anthracyclineinduced cardiomyopathy, as it has been noted that ironchelating agents that prevent generation of oxygen free-radicals, such as dexrazoxane, are cardioprotective. Due to their high antineoplastic activity, anthracycline antibiotics are the most widely used agents in oncology practice. During the recent clinical trials, the prevalence of anthracycline-induced cardiomyopathy has been noted to increase.96 This may be due to mainly the improvement of diagnostic methods, the tendency to use high doses of anthracyclines, limited cardioprotection, and prolonged survival time of patients with malignancies, as well as combined treatment methods characterized by synergistic cardiotoxicity. The toxic effect of anthracyclines on the heart may occur at any stage of treatment. Three major types of anthracycline cardiotoxicity are distinguished—acute, chronic, and late onset—which differ considerably as to clinical picture and prognosis.96,97 Acute complications are observed in 0.4% to 11/24/2006 1:28:33 PM 12 4 0 chapter 41% of patients, and their occurrence is directly connected with the administration of these agents. Arrhythmias occur during or several hours after the infusion and most frequently manifest as repolarization abnormalities, low-voltage QRS complex, sinus tachycardia, accessory ventricular and supraventricular beats, or QT prolongation. Acute myocardial ischemia is rarely observed. These electrocardiographic disturbances are usually asymptomatic and reversible, withdrawing spontaneously several hours (arrhythmia) or several weeks (changes in ST-T) after the completion of chemotherapy. In some cases, the cardiotoxicity may progress into fulminant cardiac failure, which may be fatal in some cases. Chronic cardiotoxicity is a result of repeated exposure of cardiomyocytes to anthracycline antibiotics; it is observed in 0.4% to 23% of the patients treated.96,97 Several weeks or months after chemotherapy, severe congestive HF develops (it usually affects the left ventricle, or rarely both ventricles), with features of biventricular overload and low QRS voltage. The clinical picture of post-anthracycline cardiomyopathy is not distinct; the most common symptoms include decreased exercise tolerance, effort dyspnea, cardiomegaly on chest xray, and lower left ventricular ejection fraction (LVEF) on echocardiography. The pharmacologic treatment of HF can improve the quality of life and reduce the frequency of HF complications; however, cardiomyopathy due to anthracycline still carries a very poor outcome, with mortality rates ranging from 27% to 61% despite aggressive medical treatment. A less favorable prognosis is associated with early-onset HF developing within the first 4 weeks after chemotherapy. Late cardiotoxicity is diagnosed within several years after chemotherapy. It may occur in children or adults who received relatively low total doses of doxorubicin (<480 mg/m2) and may clinically manifest as congestive HF, arrhythmia, or conduction abnormalities, such as atrioventricular block, ventricular, tachycardia, or fibrillation. Approximately 5% of the patients treated with doxorubicin may develop HF or symptomatic arrhythmia in 10 years.97 The prognosis of anthracycline-induced cardiomyopathy relates to the time course of treatment and preexisting additional risk factors for myocardial injury such as radiation, coexisting coronary artery disease, and preexisting cardiac dysfunction. In general, patients with anthracycline-induced cardiomyopathy generally have a worse survival than that seen with idiopathic DCM. Cardiomyopathy develops more frequently in the patients under age 15 or over age 65 during doxorubicin treatment. Among children treated with anthracyclines, girls are more sensitive to cardiotoxic effects of this agent than boys, although the reason for this is unknown.98 The pharmacokinetic data demonstrate that cardiotoxicity increases with higher drug concentrations in the serum. Thus, high single doses or short-term intravenous infusions of doxorubicin may be associated with more cardiotoxicity than continuous low-dose infusions. Prior irradiation of mediastinum is also associated with increased cardiotoxicity. It was estimated that risk of postanthracycline cardiomyopathy increased by 1.6 when there was a history of radiation of the heart. Improper nutrition, diabetes, or simultaneous administration of other antineoplastic agents damaging the myocardium such as cyclophosphamide, actinomycin D, mitomycin C, mithramycin, dacarbazine, bleomycin, etoposide, cisplatin, busulphan, CAR055.indd 1240 55 methotrexate, melphalan, and vincristine have been associated with increased cardiotoxicity. The most powerful predictor of anthracycline toxicity is the total cumulative dose administered. Clinically apparent cardiotoxicity is rare (less than 3%) below cumulative doses of 400 mg/m2, and toxicity becomes more frequent at higher doses. Nonetheless, it bears emphasis that subclinical cardiac toxicity or myocardial damage can be demonstrated even at lower doses. For example, Bristow et al.99 demonstrated evidence of myocyte damage on endomyocardial biopsy in 93% of the patients who received greater than 272 mg/m2 and in 3% of the patients who received as little as 45 mg/m2. Available data suggest that circulating markers such as cardiac troponins and brain natriuretic peptide (BNP) could potentially be useful for predicting cardiac toxicity with anthracycline. Elevated levels of cardiac troponin have been positively correlated with the dose of anthracycline administered. Cardiac troponin T (cTnT) levels were reported to be elevated in about 30% of children treated with doxorubicin, and cardiac troponin I (cTnI) in about one third of adults 12 to 72 hours after high-dose chemotherapy. Raised concentrations of cardiac troponins sometimes persist for months, suggesting long-term myocardial injury.100 Moreover, elevated levels of troponins correlated also with anthracyclineinduced cardiac damage; a raised cTnI indicated a significantly greater decrease in LVEF (by an absolute 16% on average compared with only 5% when the cTnI was not raised).100 Clinical trials are currently evaluating the role of these markers in predicting both early and late clinical and subclinical damage associated with anthracyclines. The current guidelines for administration and monitoring of patients receiving doxorubicin is as follows: doxorubicin should not be administered to patients with a baseline LVEF of ≤30%. If the baseline LVEF is between ≥31% and <50%, LV function should be assessed prior to each subsequent dose, and doxorubicin therapy should be discontinued if the LVEF declines by ≥10% or to a value <30%. If, on the other hand, the patient has a baseline LVEF of ≥50%, an assessment of the LVEF should be repeated after the patient has received a total dose of 300 to 350 mg/m2, and then again after each subsequent dose of doxorubicin. Therapy should be discontinued if the LVEF declines by ≥10% or to a value <50%. In addition to the guidelines for administration of doxorubicin outlined above, patients who develop HF should be managed with conventional HF management, which may lead to symptomatic improvement in 65% to 87% of patients.101,102 Indeed, complete reversal of left ventricular dysfunction has been reported in anecdotal cases.101 Prevention of anthracycline-induced myocardial damage by use of free radical “scavengers” and antioxidants may reduce cardiotoxicity in some patients. The investigations of vitamin E, ascorbic acid, Q-10 coenzyme and N-acetylcysteine have not proved them to be cardioprotective.102 At present, the most effective protection of cardiomyocytes during chemotherapy with anthracyclines is with dexrazoxane (ICRF187-cardioxan).103 This preparation chelates intracellular iron in vitro and in vivo and it decreases the level of free radicals; additionally, it regulates topoisomerase II activity and has an immunomodulating effect on myocarditis secondary to the administration of anthracycline. Randomized clinical trials showed a cardioprotective effect of dexrazoxane reflected in, 11/24/2006 1:28:33 PM di l at e d c a r diom yopat h y for example, a two- to threefold decrease in the risk of anthracycline-induced cardiomyopathy as well as a significantly lower percentage of severe damage of cardiomyocytes in heart biopsies. However, further studies are necessary before introducing dexrazoxane into wide clinical practice. Although the drug proved efficient in the prevention of early anthracycline cardiotoxicity, its role in the prophylaxis of late myocardial damage remains unknown. Another antioxidant raising hope for selective cardioprotection during chemotherapy with anthracyclines is probucol, a vitamin E derivative used in the treatment of lipid disorders.104 Preclinical studies revealed that it increased possible neutralization of free radicals in cardiocytes by stimulating the activity of glutathione peroxidase and superoxide dismutase. Although the results of preclinical studies are promising, no randomized trials were conducted with probucol in humans.104 The cardioprotective activity of the compounds without antioxidant preparations, such as β-adrenergic receptor inhibitors, calcium channel antagonists, antihistamine and antiinflammatory drugs, digitalis glycosides, prednisone, bipyridine derivatives (amrinone, milrinone), and carnitine have not demonstrated efficacy for prevention of anthracyclineinduced cardiomyopathy.97,104 Synthesis of doxorubicin analogues with lesser cardiotoxic characteristics have not been very promising thus far either.96 Other chemotherapeutic agents in cancer associated with cardiac toxicity complication are the monoclonal antibody trastuzumab (Herceptin), high-dose cyclophosphamide, mitomycin-C, 5-fluorouracil, and the interferons. These agents can cause decreased left ventricular function, which in some cases may result in clinical manifestations of congestive HF. Trastuzumab has proved to be a useful palliative treatment for metastatic breast cancers that overexpress HER2/neu protein (about 30% of all breast cancers), and it is the first new agent to improve survival for patients with metastatic breast cancer in the past 25 years.105 The occurrence of cardiac toxicity was unexpectedly high (28%) in patients treated with trastuzumab (Herceptin), either as a single agent or in combination with chemotherapy, especially in patients previously or concomitantly treated with anthracyclines.105 This was a surprising finding as cardiac toxicity could not be predicted on the basis of the putative mechanism of action of the antibody. The mechanism of trastuzumab-associated cardiac dysfunction is unknown. Although the family of human epidermal growth factors, of which HER2/neu is a member, has an important role in cardiac embryogenesis, there is little evidence of localization of anti-HER2/neu to cardiac tissue. Importantly, this toxic effect seems to respond better to medical therapy for congestive HF than does anthracycline-induced dysfunction.100 Other Myocardial Toxins In addition to the classic toxins described above, as shown in Table 55.1, there are a number of other toxic agents that may lead to left ventricular dysfunction and HF, including cobalt and catecholamines. Inflammation-Induced Cardiomyopathy Over the past 10 to 15 years, there has been increasing evidence that suggests that inflammation or inflammatory pro- CAR055.indd 1241 12 41 cesses may contribute to the overall pathogenesis of DCM. Moreover, there is increasing evidence that biologic properties of inflammatory mediators, such as proinflammatory cytokines, are also sufficient to produce a dilated cardiac phenotype.106,107 As will be discussed below, both infectious and noninfectious inflammatory processes may lead to the development of DCM. Although a great many infections and noninfectious processes may impact the myocardium, and may transiently lead to systolic dysfunction and congestive symptomatology, the great majority of these infections and noninfectious processes do not lead to the development of DCM. Therefore, in the section that follows, we focus primarily on those disease states that are considered to lead to DCM. Infectious Causes VIRAL CARDIOMYOPATHY The subject of viral myocarditis is covered in detail in another chapter. ACQUIRED IMMUNODEFICIENCY SYNDROME (AIDS) Several investigators have reported that there is an association between AIDS and DCM.108,109 Reviews and studies published before the introduction of highly active antiretroviral therapy (HAART) regimens have correlated the incidence and course of human immunodeficiency virus (HIV) infection in relation to DCM in both children and adults. In a long-term echocardiographic follow-up by Barbaro and coworkers,110 8% of initially asymptomatic HIV-positive patients were diagnosed with DCM during the 60-month follow-up. All patients with DCM were in NYHA functional class III (84%) or IV (16%). The mean annual incidence rate was 15.9 cases per 1000 patients. The extent of immunodeficiency of the patients, as assessed by the CD4 count, influenced the incidence of DCM; specifically, there was a higher incidence among patients with a CD4 count of less than 400 cells per cubic millimeter. Current hypotheses concerning the pathogenesis of cardiomyopathy associated with infection with HIV include infection of myocardial cells with HIV type 1 (HIV-1) or coinfection with other cardiotropic viruses, postviral cardiac autoimmunity, autonomic dysfunction, cardiotoxicity from illicit drugs and pharmacologic agents (such as nucleoside analogues and pentamidine), nutritional deficiencies, and prolonged immunosuppression. It has been demonstrated that targeted myocardial expression of HIV transactivator in transgenic mice results in cardiomyopathy and mitochondrial damage,111,112 emphasizing the role of the HIV infection itself in AIDS cardiomyopathy. The role of myocarditis in the development of DCM in HIV has not been fully characterized.113 Lymphocytic myocarditis can be found at autopsy in 46% to 52% of patients with AIDS, as reported in a review of the literature.114 Autopsy series of persons dying from AIDS-related illnesses demonstrate histologic evidence of myocarditis in approximately 50% of the patients. Symptomatic HF is seen in approximately half of these patients with myocardial involvement. Whether early treatment with ACE inhibitors or beta-blockers will prevent or retard disease progression in these patients is unknown at this time. The treatment of patients with symptomatic HIV cardiomyopa- 11/24/2006 1:28:34 PM 12 4 2 chapter thy is the same as the conventional treatment for patients with DCM. CHAGAS’ DISEASE Although Chagas’ disease is a relatively uncommon cause of DCM in North America, it remains a leading cause of death in many areas of Central and South America. Indeed, 50,000 people die of Chagas’ disease each year.115 Trypanosoma cruzi, the causative organism for Chagas’ disease, is found only in the Western Hemisphere, where it primarily infects wild and domestic mammals and insects. Humans become involved when infected vectors infest the simple houses that are common in Latin America. It is estimated that 16 to 18 million people have chronic T. cruzi infection. Chagas control programs in certain South American countries have demonstrated significant decline in the seropositive subjects from 47.8% to 17.1%, which is most marked among children and teenagers, declining from 29.9% to 1.9%, suggesting reduction in transmission of the disease.116 There are three different clinical and pathophysiologic phases of disease: acute, indeterminate, and chronic. Sudden cardiac death can occur during each phase; however, DCM is a late manifestation of the disease and is generally seen during the chronic phase. Acute Chagas’ disease is usually a mild illness with a case fatality rate of less than 5%. The systemic spread of the parasites from the site of entry and their initial multiplication may be accompanied by fever, malaise, and edema of the face and lower extremities, as well as generalized lymphadenopathy and hepatosplenomegaly. Muscles, including the heart, are often heavily parasitized, and severe myocarditis develops in a small proportion of patients. The acute illness resolves spontaneously over a period of 4 to 6 weeks in most patients, who then enter the indeterminate phase of T. cruzi infection. In this phase, there are no symptoms, but there are lifelong, low-grade parasitemias in association with antibodies to many T. cruzi antigens. Many people in this phase have subtle signs of cardiac or gastrointestinal involvement long before the disease becomes symptomatic. Most infected people remain in the indeterminate phase for life. However, this carrier state can be a major cause of transfusion-associated transmission of the parasite.117 Symptomatic chronic Chagas’ disease develops in an estimated 10% to 30% of infected persons, years or even decades after the T. cruzi infection is acquired. The heart is most commonly affected, and the pathologic changes may include biventricular enlargement, thinning of ventricular walls, apical aneurysms, and mural thrombi. Widespread lymphocytic infiltration is often seen in stained specimens of cardiac tissue, as well as diffuse interstitial fibrosis and atrophy of myocardial cells. The conduction system is often affected, typically resulting in right bundle-branch block, left anterior fascicular block, or complete atrioventricular block. The symptoms reflect the dysrhythmias, cardiomyopathy, and thromboembolism that develop over time. Death usually results from rhythm disturbances (∼40% of patients) or progressive HF (∼60% of patients). The overall prognosis for patients with Chagas cardiomyopathy and HF is poor, with 50% of patients dying within a period of 47 months. The presence of complete heart block, atrial fibrillation, LBBB, and complex ventricular ectopy augurs a poor prognosis. CAR055.indd 1242 55 The leading hypothesis with respect to the pathogenesis of Chagas’ cardiomyopathy is that patients develop progressive myocardial damage due to parasite persistence and autoimmune responses.115 Individuals with parasite persistence and no myocardial damage or very limited damage on endomyocardial biopsies (indeterminate phase) have a nearly normal life expectancy. In contrast, those patients, who have parasite persistence and diffuse myocardial damage (arrhythmic phase) or end-stage heart disease, have dramatically diminished survival at 12 years of follow-up.116 The neurogenic hypothesis of Chagas’ cardiomyopathy suggests that the cardiac parasympathetic neurons are irreversibly damaged by the parasite during the acute phase of the disease. As a consequence, the cardiac sympathetic nervous system is unopposed, and the cardiotoxic effects of a permanent and excessive sympathetic activation are responsible for the relentless progression of myocardial damage. However, activation of the sympathetic nervous system and of the reninangiotensin-aldosterone system is a late event in the natural history of Chagas’ disease, and patients with parasite persistence but no segmental myocardial damage do not have neurohormonal activation.115 Myocardial ischemia and coronary microcirculation abnormalities have also been demonstrated in animal models and in humans with Chagas’ disease. The pharmacologic treatment of T. cruzi infection remains unsatisfactory. Extensive clinical experience has been accumulated with two drugs, benznidazole and nifurtimox. Both shorten the acute phase of T. cruzi infection and decrease mortality, but they achieve parasitologic cures in only about 50% of treated patients; moreover, these drugs cause substantial toxicity. There is no evidence that drug treatment of persons with chronic T. cruzi infection alters the natural course of the disease, and a sizable proportion of such patients remain positive for parasites indefinitely.118 Treatment of Chagas cardiomyopathy is the same as the conventional treatment for patients with DCM. Placebo-controlled clinical investigations and short-term pharmacologic studies have consistently shown that ACE inhibitors improve and prolong the life of Chagas’ patients with end-stage heart disease.119 Noninfectious Causes HYPERSENSITIVITY MYOCARDITIS Hypersensitivity to a variety of agents may result in allergic reactions that involve the myocardium, characterized by peripheral eosinophilia, and a perivascular infiltration of the myocardium by eosinophils, lymphocytes, and histiocytes. These infiltrates may be occasionally associated with necrosis. A variety of drugs, most commonly the sulfonamides, penicillins, methyldopa, and other agents such as amphotericin B, streptomycin, phenytoin, isoniazid, tetanus toxoid, hydrochlorothiazide, and chlorthalidone, have been reported to cause allergic hypersensitivity myocarditis. Most patients are not clinically ill, but may die suddenly, presumably secondary to an arrhythmia. Hypersensitivity myocarditis is recognized only rarely clinically, but may be sufficient to produce global or regional myocardial dysfunction detected by noninvasive methods. This entity is often first diagnosed on postmortem examination, and occasionally on endomyocardial biopsy.120 11/24/2006 1:28:34 PM di l at e d c a r diom yopat h y SYSTEMIC LUPUS ERYTHEMATOSUS (SLE) Although a number of cardiac abnormalities have been reported in patients with SLE, the development of DCM is not a prominent manifestation of this disease process. Global LV dysfunction has been reported in 5%, segmental LV wall motion abnormalities in 4%, and RV enlargement in 4% of patients with SLE. In general the abnormalities in cardiac function usually correlate with disease activity. The myocardial involvement is frequently found at autopsy or at endomyocardial biopsy and is less easily detected clinically. The myocardial lesions are characterized by an increase in interstitial connective tissue and myocardial scarring. Recent studies suggest that depolarization abnormalities on signal average ECG accompanied with echocardiographic evidence of abnormal LV filling may reflect the presence of myocardial fibrosis and could be a marker of subclinical myocardial involvement in SLE patients.121 Cardiac involvement may manifest itself by conduction system abnormalities such as complete atrioventricular heart block. Especially neonatal lupus is characterized by congenital heart block, as well as cardiomyopathy, cutaneous lupus lesions, hepatobiliary disease, and thrombocytopenia. Accumulating evidence indicates that the disease is probably caused by maternal autoantibodies.122 SCLERODERMA The development of DCM is rare in patients with scleroderma. A recent echocardiographic study showed that although there was no difference in LV dimensions or fractional shortening in patients with scleroderma, there was indication of systolic impairment by systolic time intervals with an increased preejection period to LV ejection time (LVET) ratio and also an increased isovolumic contraction time to LVET ratio in the majority of patients.123 A distinctive focal myocardial lesion ranging from contraction band necrosis to replacement fibrosis without morphologic abnormalities of the coronary arteries is noted in approximately half of the patients with scleroderma. This is postulated to be due to intermittent vascular spasm with intramyocardial Raynaud’s phenomenon.124 Thus, progressive systemic sclerosis can lead to conduction abnormalities, arrhythmias, HF, angina pectoris with normal coronary arteries, myocardial fibrosis, pericarditis, and sudden death. Late contrast enhancement with gadolinium may be used to characterize patchy fibrosis and myocardial edema interspersed with normal myocardium in scleroderma. Cardiac involvement in systemic sclerosis portends an ominous prognosis, and is probably most directly related to the extent of myocardial fibrosis. R HEUMATOID A RTHRITIS Cardiac involvement in rheumatoid arthritis generally results from the development of myocarditis or pericarditis. However, the development of DCM is rare in these patients. In a retrospective study of 172 patients with juvenile rheumatoid arthritis, symptomatic cardiac involvement occurred in 7.6% of patients, including pericarditis, perimyocarditis, and myocarditis. Both myocarditis and pericarditis are regarded as poor prognostic factors in rheumatoid arthritis.125 Myocardial involvement in rheumatoid arthritis is thought to be secondary to disturbances in the microcirculation sec- CAR055.indd 1243 12 4 3 ondary to microvasculitis, and occurs in the absence of any clinical symptoms of ECG changes. K AWASAKI DISEASE Kawasaki disease is an acute febrile illness associated with mucosal inflammation, skin rash, and cervical lymphadenopathy. This disease is recognized most often in children younger than 4 years of age. Kawasaki disease represents an acute vasculitic syndrome of unknown etiology that primarily affects small and medium-sized arteries, including coronary arteries. Coronary arterial aneurysms are seen in 25% to 55% of the acute Kawasaki cases. Of these, 4.7% progress to premature atherosclerotic ischemic heart disease. Myocardial infarction is noted in approximately 2% of patients, and cardiovascular death is reported in 0.8%. Although the development of DCM is not typical for Kawasaki disease, repetitive infarctions secondary to coronary arterial aneurysms may lead to a dilated cardiomyopathic phenotype. PERIPARTUM CARDIOMYOPATHY Peripartum cardiomyopathy is a disease of unknown cause in which severe LV dysfunction occurs during the last trimester of pregnancy or the early puerperium. It is reported in one in 1300 to one in 4000 live births. In the past, the diagnosis of this entity was made on clinical grounds; however, modern echocardiographic techniques have allowed more accurate diagnoses by excluding cases of diseases that mimic the clinical symptoms and signs of HF. Risk factors for peripartum cardiomyopathy include advanced maternal age, multiparity, African descent, twinning, and long-term tocolysis. Anticoagulation is strongly recommended, especially if ventricular function is persistent. Although its etiology remains unknown, most theories have focused on the hemodynamic126 and immunologic stresses of pregnancy.127 An immune pathogenesis is supported by the frequent finding of lymphocytic myocarditis on myocardial biopsy128 and the fact that multiparity or previous exposure to fetal antigens is a significant risk factor.129 The prognosis of peripartum cardiomyopathy is related to the recovery of ventricular function. In contrast to patients with idiopathic DCM, significant improvement in myocardial function is seen in 30% to 50% of patients in the first 6 months after presentation.129 However, for those patients who do not recover to normal or near-normal function, the prognosis is similar to other forms of DCM.130 Cardiomegaly that persists for more than 4 months after diagnosis indicates a poor prognosis, with a 50% mortality at 6 years. Caution is advised in recommending subsequent pregnancy, especially if LV dysfunction is persistent. Heart failure recurs during subsequent pregnancies in more than 50% of the patients.127 Endocrine and Metabolic Causes of Cardiomyopathy Obesity Heart failure in the markedly obese usually develops over a long period of time, and can be directly related to the duration of obesity. Initially, the dyspnea and edema in these 11/24/2006 1:28:34 PM 12 4 4 chapter patients is simply related to alterations in LV compliance with resultant elevated filling pressures. However, with chronicity, these patients develop a DCM phenotype, with further worsening of the patient’s volume overload status. Although the precise reasons for obesity-related HF are not known, it is thought that the chronic increase in cardiac work and chronic increase in systemic blood pressure ultimately lead to myocardial failure.131 In addition, there is an increased prevalence of hypertension and coronary artery disease in obese patients, which may also contribute to the development of DCM in these patients. Obesity-related hypoventilation and sleep apnea may also contribute to the pathophysiology. Although there are anecdotal reports regarding symptomatic improvement following weight reduction in obesity-induced HF, large-scale clinical trials on the role of weight loss in obesity have not yet been performed. A study examining the relationship between obesity and HF in participants in the Framingham Heart Study132 reported that after adjustment for established risk factors, there was an increase in the risk of HF of 5% for men and 7% for women for each increment of 1 in body mass index. When compared with subjects with a normal body mass index, obese subjects had a doubling of the risk of HF. Obesity alone was estimated to account for 11% of cases of HF in men and 14% of those in women.132 Nearly 60% of the adult population of the United States is overweight or obese, resulting in high morbidity and mortality.133 Given the high prevalence of obesity in the United States, strategies to promote optimal body weight may reduce the population burden of HF. It is recognized that obesity, particularly truncal obesity, has been associated with the metabolic syndrome, which is characterized by insulin resistance and particularly atherogenic dyslipidemia; with high levels of low-density lipoprotein cholesterol, very-low-density lipoprotein cholesterol, and triglycerides; and low levels of high-density lipoprotein cholesterol.134 The metabolic syndrome has emerged as a major risk factor for cardiovascular events and may be the precursor of undetected atherosclerotic cardiovascular disease ultimately resulting in HF. Diabetic Cardiomyopathy Since its first description in 1972,135 a considerable amount of experimental, pathologic, epidemiologic, and clinical data have accumulated to support the existence of diabetic cardiomyopathy. So far, a large body of literature suggests that the occurrence of diabetic cardiomyopathy is an independent phenomenon from macroangiographic changes in coronary arteries and hypertension. Diabetes is now well recognized as an independent risk factor for the development of HF despite correcting for age, hypertension, obesity, hypercholesterolemia, and coronary artery disease. Diabetic men have more than twice the frequency of HF compared with nondiabetic cohorts, whereas diabetic women have a fivefold increased risk of developing HF.135,136 Possible mechanisms that have been implicated in the pathogenesis of diabetic cardiomyopathy include microangiopathic changes in the small vessels of the heart, such as microvascular constriction or microaneurysms; changes in fatty-acid metabolism; unavailability of glucose as an energy substrate, and shifting the intracellular metabolism from glycolysis to free fatty acid CAR055.indd 1244 55 oxidation, thus resulting in inadequate adenosine triphosphate (ATP) generation and increased production of oxygen free radicals; alterations in calcium and potassium transport and calcium handling; cardiac hypertrophy and increase in LV mass independent of loading conditions; defects in collagen structure resulting in myocardial, perivascular, and interstitial fibrosis; abnormalities in the conducting system; and a decrease in the function of autonomic nerves and reduced sympathetic innervation.135 The frequency and pace by which these preclinical abnormalities progress to clinically evident myocardial dysfunction in patients with diabetes is not well established. Moreover, the role of metabolic control of diabetes mellitus in the prevention or the reversal of myocardial dysfunction is unclear. It is interesting to note, however, that diabetic patients have broader indications for initiation of ACE inhibitors other than LV dysfunction, such as diabetic nephropathy or proteinuria. Perhaps the more widespread and earlier use of ACE inhibitors in diabetic patients, before the appearance of the cardiomyopathy phenotype, will reduce the propensity to develop clinical diabetic cardiomyopathy in this high-risk population. Hyperthyroidism Hyperthyroidism has been implicated in causing DCM; however, in view of the increased cardiac contractile function of patients with hyperthyroidism, the development of HF is unexpected and raises the question of whether there truly is a direct causal association between hyperthyroidism and cardiomyopathy.137 Patients with hyperthyroidism may occasionally have exertional dyspnea or other symptoms and signs of HF. Many of these clinical manifestations may be attributed to the direct effects of thyroid hormone on cardiovascular hemodynamics. In most patients with hyperthyroidism, cardiac output is high, and the subnormal response to exercise may be the result of an inability to increase heart rate maximally or to lower vascular resistance further, as normally occurs with exercise.138 The term high-output failure is not appropriate for all cases of cardiomyopathy related to hyperthyroidism, because the ability of the heart to maintain increased cardiac output at rest and with exercise is usually preserved.137 Occasional patients with severe, longstanding hyperthyroidism have poor cardiac contractility, low cardiac output, and symptoms and signs of HF, including a third heart sound and pulmonary congestion.139 This complex of findings most commonly occurs with persistent sinus tachycardia or atrial fibrillation and is the result of socalled rate-related HF. In older patients with heart disease, the increased workload that results from hyperthyroidism may further impair cardiac function and thus result in DCM. The presence of ischemic or hypertensive heart disease may compromise the ability of the myocardium to respond to the metabolic demands of hyperthyroidism.137 Histologic examination usually reveals a nonspecific pattern, including foci of lymphocytic and eosinophilic infiltration, fibrosis, fatty infiltration, and myofibril hypertrophy. Although enhanced adrenergic activity contributes to the hyperdynamic state in hyperthyroidism, β-adrenergic blockade does not fully return the heart rate or contractility to normal, which may suggest a primary cardiac effect of thyroid hormone with contributory effects of cathecolamines.137 11/24/2006 1:28:34 PM di l at e d c a r diom yopat h y Hypothyroidism Abnormalities in cardiac systolic and diastolic performance have been reported both in experimental and in clinical studies of hypothyroidism; however, the classic findings of myxedema does not usually indicate cardiomyopathy.137 The most common signs are bradycardia, mild hypertension, a narrowed pulse pressure, and attenuated activity on the precordial examination. Pericardial effusions and nonpitting edema (myxedema) can occur in patients with severe, longstanding hypothyroidism. The low cardiac output is caused by bradycardia, a decrease in ventricular filling, and a decrease in cardiac contractility. Systemic vascular resistance may increase by as much as 50%, and diastolic relaxation and filling are slowed.137 However, HF is rare, because the cardiac output is usually sufficient to meet the lowered demand for peripheral oxygen delivery. Positron emission tomographic studies of oxygen consumption in patients with hypothyroidism have revealed that myocardial work efficiency is lower than in normal subjects. From 10% to 25% of patients have diastolic hypertension, which, combined with the increase in vascular resistance, raises cardiac afterload and cardiac work.137 Abnormal systolic force may improve with thyroid hormone replacement but does not return to normal levels, suggesting the possibility of persistent myocardial dysfunction.140 Thyroid hormone replacement should be initiated at low doses and titrated slowly as LV failure may be precipitated. Interestingly, patients with HF also have low serum triiodothyronine concentrations, and the decrease is proportional to the degree of HF, as assessed by the NYHA functional classification. Whether such changes in thyroid hormone metabolism contribute to further impairment of cardiovascular function in patients with HF is not known.137 Acromegaly and Growth Hormone Deficiency Impaired cardiovascular function has recently been demonstrated to potentially reduce life expectancy both in patients with growth hormone deficiency and in those with a growth hormone excess. Experimental and clinical studies have supported the evidence that growth hormone and IGF-1 are implicated in cardiac development.141 In most patients with acromegaly, a specific cardiomyopathy, characterized by myocardial hypertrophy with interstitial fibrosis, lymphomononuclear infiltration, and areas of monocyte necrosis, results in biventricular concentric hypertrophy.141 Myocardial dysfunction is a major cause of morbidity and mortality in acromegaly and appears to be related to both the severity and duration of growth hormone excess. In contrast, patients with childhood- or adulthood-onset growth hormone deficiency may suffer from both structural cardiac abnormalities, such as narrowing of cardiac walls, and functional impairment, which combine to reduce diastolic filling and impair LV response to peak exercise.141 In addition, growth hormone deficiency patients may have an increase in vascular intima-media thickness and a higher occurrence of atheromatous plaques, which can further aggravate the hemodynamic conditions and contribute to increased cardiovascular and cerebrovascular risk. Several studies have suggested that the cardiovascular abnormalities can be partially reversed by suppressing growth hormone and IGF-1 levels in acromegaly or after growth hormone replacement therapy in CAR055.indd 1245 12 4 5 growth hormone deficiency patients.141 Receptors for both growth hormone and IGF-1 are expressed by cardiac myocytes; therefore, growth hormone may act directly on the heart or via the induction of local or systemic IGF-1, whereas IGF-1 may act by endocrine, paracrine, or autocrine mechanisms. Animal models of pressure and volume overload have demonstrated upregulation of cardiac IGF-1 production and expression of growth hormone and IGF-1 receptors, implying that the local regulation of these factors is influenced by hemodynamic changes.142 Moreover, experimental studies suggest that growth hormone and IGF-1 have stimulatory effects on myocardial contractility, possibly mediated by changes in intracellular calcium handling. Thus recently, much attention has been focused on the ability of growth hormone to increase cardiac mass, suggesting its possible use in the treatment of chronic HF. Initial studies demonstrated that treatment with growth hormone may result in improvement in hemodynamic and clinical status of patients with HF; however, recently, two randomized, placebo-controlled studies did not show any significant growth hormone–mediated improvement in cardiac performance in patients with DCM, despite significant increases in IGF-1. Although these data need to be confirmed in more extensive studies, such promising results seem to open new perspectives for growth hormone treatment in humans.143 Nutritional Causes of Cardiomyopathy Thiamine Deficiency Thiamine serves as a cofactor for several enzymes involved primarily in the carbohydrate catabolism and is found in high concentrations in the heart, skeletal muscle, liver, kidneys, and brain.144 The most common cause of thiamine deficiency in Western countries is chronic alcoholism and anorexia nervosa. AIDS and pregnancy can account for other rare causes of thiamine deficiency.145 A state of severe depletion can develop in patients on a strict thiamine-deficient diet in approximately 18 to 21 days. The major manifestations of thiamine deficiency in humans involve the cardiovascular (wet beriberi) and nervous (dry beriberi, or neuropathy, or Wernicke-Korsakoff syndrome) systems. The cardiovascular signs and symptoms include dyspnea, fatigue, leg edema, and palpitations. Tachycardia is common and there is usually increased jugular venous pressure along warm extremities. Biventricular HF is present and the circulation is usually hyperkinetic. Ultimately, circulatory collapse, metabolic acidosis, or shock can develop, at which time, the disease has advanced from chronic beriberi to fulminating beriberi HF (Shoshin beriberi). Severe lactic acidemia in the presence of a high cardiac output and extremely low oxygen consumption are the classic features of acute fulminant cardiovascular beriberi, which, if unrecognized and untreated, can lead to high cardiac output failure and death. Treatment for Beriberi should consist of administration of thiamine along with other conventional treatments of circulatory support and HF.146 Carnitine Deficiency l-carnitine and its derivative, propionyl-l-carnitine, are organic amines necessary for oxidation of fatty acids, the 11/24/2006 1:28:34 PM 12 4 6 chapter deficiency of which may be associated with a syndrome of progressive skeletal myopathy and lipid vacuoles on muscle biopsy.147 They have also been shown to reduce intracellular accumulation of toxic metabolites during ischemia, demonstrate protective effect on muscle metabolism injuries, inhibit caspases, and decrease the levels of TNF-α and sphingosine, as well as reduce apoptosis of skeletal muscles cells and thus have been impliated in the treatment of congestive HF.148 It has been shown that several metabolic genes, including the muscle carnitine palmitoyltransferase-I (CPT-I), are downregulated in the failing human heart.149 Inhibitors of CPT-I, the key enzyme for the transport of long-chain acylcoenzyme A (acyl-CoA) compounds into mitochondria, have been developed as agents for treating diabetes mellitus. Interestingly, a CPT inhibitor, etomoxir, has been reported to enhance levels of the sarcoplasmic reticulum Ca(2+)– adenosine triphosphatase (ATPase), a key protein in calcium cycling, and of the α-MHC in normal and pressureoverloaded rat myocardium.150 This finding can be attributed to selective changes in the dysregulated gene expression of hypertrophied cardiomyocytes. Selenium Deficiency Selenium deficiency is associated with cardiomyopathy and congestive HF in geographic areas where dietary selenium intake is low.151 This has been named the Keshan disease, due to the geographic prevalence of a specific form of DCM in northeast China, where the soil has a low selenium content. Chronic selenium deficiency may also occur in individuals with malabsorption and long-term seleniumdeficient parenteral nutrition.152 Selenium deficiency is implicated in causing cardiomyopathy as a result of the depletion of selenium-associated antioxidant enzymes, selenoenzymes, which protect cell membranes from damage by free radicals. The cardiomyopathy is manifested by the insidious onset of congestive HF or a complication of sudden death or thromboembolic phenomena. The heart shows biventricular enlargement and histologically exhibits edema, mitochondrial swelling, hypercontraction bands, widespread myocytolysis, and extensive fibrosis.151 Hematologic Causes of Cardiomyopathy Cardiomyopathy Due to Iron Overload: Hemochromatosis and Thalassemia Iron-overload cardiomyopathy manifests itself as systolic or diastolic dysfunction secondary to increased deposition of iron in the heart and occurs with common genetic disorders such as primary hemochromatosis and β-thalassemia major. Hereditary hemochromatosis is an inherited disorder of iron metabolism and is the most common hereditary disease of Northern Europeans, with a prevalence of approximately 5 per 1000. It is an autosomal recessive disorder of iron metabolism characterized by increased iron absorption and deposition in the liver, pancreas, heart, joints, and pituitary gland. Without treatment, death may occur from cirrhosis, primary liver cancer, diabetes, or cardiomyopathy. In 1996, HFE, the gene for hereditary hemochromatosis, was mapped on the short arm of chromosome 6.154 The H63D mutation is significantly increased in patients with idiopathic DCM.153 CAR055.indd 1246 55 The high correlation of the HFE to hemochromatosis has caused it to be considered as a candidate gene for populationbased genetic testing for diagnosis and detection of predisposition to hemochromatosis. Although the exact mechanism of iron-induced HF remains to be elucidated, the toxicity of iron in biologic systems is attributed to its ability to catalyze the generation of oxygen free radicals. It was shown that chronic iron-overload results in dose-dependent increases in myocardial iron burden, decreases in the protective antioxidant enzyme activity, increased free-radical production, and increased mortality.154 These findings suggest that the mechanism of iron-induced heart dysfunction involves in part free radical-mediated processes.154 This also implies that the disease process is reversible if the tissue iron concentration can be controlled. Myocardial iron deposits do not occur until other organs such as liver, pancreas, and connective tissues are saturated with iron. Thus, the extracardiac manifestations are present before the cardiomyopathy develops.154 During the initial phases of the cardiomyopathy, the hemodynamic profile represents a restrictive pattern. As the severity of cardiomyopathy advances, DCM with biventricular enlargement and HF ensue. The spectrum of arrhythmia ranges from minor abnormalities on the electrocardiogram to supraventricular arrhythmia, atrioventricular conduction block, and ventricular tachyarrhythmia, presumably due to myocardial dysfunction and iron deposition in the conduction system and AV node.155 The ECG most commonly shows decreased voltage and nonspecific ST and T wave changes; Q waves are uncommon. The diagnosis of hemochromatosis is suggested by elevated serum ferritin and an increased ratio of iron to total iron binding capacity (TIBC). The most sensitive screening test for hemochromatosis is saturation of the transferrin with iron; a fasting value greater than 50% is strongly suggestive of the disease. The most definitive test for calculation of iron stores in the body is measurement of iron concentration by liver biopsy. Magnetic resonance imaging (MRI) can also be very useful in identifying the iron-laden organs and cardiac involvement.156 Though not required to demonstrate cardiac involvement in every case, endomyocardial biopsy can be useful in assessment of cardiac iron deposition.157 Before phlebotomy and chelation therapy, survival among patients with hemochromatosis and HF was less than 20% in 5 years. In patients with thalassemia major, cardiac failure is one of the most frequent causes of death.158 Chelation therapy, including newer forms of oral chelators, such as deferoxamine, and phlebotomy have dramatically improved the outcome of hemochromatosis. It is important to start therapy early, as treatment may prevent or reverse cardiac involvement. Early diagnosis and treatment by phlebotomy before tissue damage has occurred is essential, because life span seems to be normal in treated patients but markedly shortened in those who are not. Additionally, genetic counseling with evaluation of firstdegree relatives is mandatory.154 Physical Agents Tachycardia-Induced Dilated Cardiomyopathy The concept that incessant or chronic tachycardia can lead to reversible LV dysfunction is supported both by animal 11/24/2006 1:28:34 PM di l at e d c a r diom yopat h y models of chronic pacing as well as by human studies documenting improvement in ventricular function with tachycardia rate or rhythm control. Sustained rapid pacing in experimental animal models can produce severe biventricular systolic dysfunction.159,160 In humans, descriptions of reversal of cardiomyopathy with rate or rhythm control of incessant or chronic tachycardias have been reported with atrial tachycardias, accessory pathway reciprocating tachycardias, atrioventricular node reentry, and atrial fibrillation with rapid ventricular responses.161 Control of the rapid ventricular responses in atrial fibrillation has been shown to improve ventricular function, following conversion to sinus rhythm, pharmacologic ventricular rate control, and AV junction ablation and permanent ventricular pacing.162,163 The investigation of potential tachycardia-induced cardiomyopathy in patients with HF requires further prospective confirmation in larger numbers of patients, with study of mechanisms, patient groups affected, and optimal therapies. Tachycardia-induced cardiomyopathy may be a more common mechanism of LV dysfunction than recognized, and aggressive treatment of the arrhythmia should be considered. 12 47 inferences regarding cause and effect are, invariably, indirect and circumstantial. Clinical Recognition The most common initial presenting manifestations of DCM are related to the presence of left HF, and include progressive exertional dyspnea, fatigue, weakness, diminished exercise capacity, orthopnea, paroxysmal nocturnal dyspnea, and nocturnal cough. With the development of right HF, abdominal distention, right upper quadrant pain, early satiety, postprandial fullness, and nausea appear. Some cases will eventually develop cardiac cachexia and wasting from advanced HF. Systemic embolization, pulmonary emboli can be seen in 1% to 4% of cases and are more commonly seen with cardiomegaly. Palpitations can occur with atrial fibrillation and ventricular arrhythmias. Ventricular arrhythmias are common toward advanced stages of cardiomyopathy; however, syncope and sudden cardiac death are rare initial presentations. Autoimmune Mechanisms Diagnostic Evaluation There has been increasing evidence suggesting that abnormalities in cellular and humoral immunity may contribute to the overall pathogenesis of DCM. Circulating autoantibodies to a variety of cardiac antigens including G-protein– linked receptors (such as those to β1-adrenoreceptors and muscarinic receptors), mitochondrial antigens, adenosine diphosphate, adenosine triphosphate carrier proteins, and cardiac MHC have been identified in patients with DCM.164–167 In this regard, it is interesting to note that immunization with certain cardiac muscle (but not skeletal) antigens such as α-MHC can result in the development of a dilated cardiac phenotype in certain susceptible strains of mice. Moreover, a recent meta-analysis has shown that there is increased expression of the antigens of genes located at the major histocompatibility complex on chromosome 6, which is the locus that is responsible for regulating immune responses. This study showed that human leukocyte antigens (HLA) class II such as DR4 or DQw4 were present in 63% of the patients with cardiomyopathy as compared to 26% in the control subjects.168 Features that support an autoimmune etiology in patients who present with myocarditis and DCM include familial aggregation, a weak association with HLA-DR4 haplotype, abnormal expression of HLA class II in cardiac tissue, and detection of organ- and diseasespecific cardiac autoantibodies by immunofluorescence and immunoabsorption techniques. Nonetheless, a precise interpretation of the above fi ndings has been complicated by the knowledge that low titers of autoantibodies, which can be part of the normal immunologic repertoire, are not always pathogenic. For example, tissue injury secondary to ischemia or infection may lead to autoantibody production because of alterations of self-antigens or exposure of antigens that are normally sequestered from the immune system. In such situations, the generation of autoantibodies is the result, and not the cause, of the tissue injury. Furthermore, observations about autoimmune responses are generally made in patients with established disease; accordingly, any The diagnostic evaluation of patients with DCM, in general, is similar to that for patients with ischemic heart disease and is discussed in another chapter. CAR055.indd 1247 Laboratory Testing Initial Laboratory Tests Initial screening laboratory studies for DCM patients should include a routine assessment of serum electrolytes, liver function tests, white blood cell count, and a hemoglobin and hematocrit. Beyond these routine tests, the positive predictive value or utility of additional laboratory studies remains low unless supported by specific elements of the history and physical exam. One possible exception to this statement is the use of the BNP as a biochemical marker for both its diagnostic and prognostic properties in HF patients. Brain Natriuretic Peptide B-type BNP is a neurohormone secreted mainly in the cardiac ventricles in response to volume expansion and pressure overload. It can be used for identifying patients with asymptomatic LV systolic dysfunction169–171 or for patients with symptomatic HF, as a marker for prognosis and risk stratification of patients with HF, and as a tool for tailoring inpatient and outpatient therapy in HF.172,173 In conjunction with other clinical information, rapid measurement of B-type natriuretic peptide is very useful in establishing or excluding the diagnosis of congestive HF in patients with acute dyspnea with excellent specificity and high negative predictive values.173 Brain natriuretic peptide has been shown to be a powerful marker for prognosis and risk stratification in the setting of HF.45 Elevated levels have been correlated with increased HF mortality and sudden cardiac death.172 Because BNP is a volume-sensitive hormone with a short half-life, BNP levels can be used in guiding 11/24/2006 1:28:35 PM 12 4 8 chapter 55 myocarditis or when there is extensive LV fibrosis even without a discrete myocardial scar.178 A variety of atrial and ventricular tachyarrhythmias and atrioventricular conduction disturbances may also be seen. Atrial fibrillation develops in approximately 20% of the patients. Premature ventricular contractions (PVCs) are not an uncommon finding on routine ECGs in patients with DCM. diuretic and vasodilator therapy on presentation with decompensated HF. Falling BNP levels during hospitalization correlate with clinical improvement and predict lower readmission rates.172 Brain natriuretic peptide can also be used in the outpatient setting to guide “tailored therapy” such as titration of beta-blockers. This has been demonstrated by the Australia–New Zealand Heart Failure Group, which reported that BNP was the best prognostic predictor of the success or failure of carvedilol.174 Troughton et al.175 randomized 69 patients to N-terminal BNP (N-BNP)–guided treatment versus symptom-guided therapy. Patients receiving N-BNP–guided therapy had lower N-BNP levels, along with reduced incidence of cardiovascular death, readmission, and new episodes of decompensated HF. Studies have shown that elevated levels of these markers have a sensitivity of ≥80% to 90% and a specificity of ≥70% to 90% for detecting patients with ejection fractions <30%.171 The negative predictive value of BNP levels under 100 pg/mL is the strongest feature of this peptide. Although the positive predictive value in a given patient at a cutoff of 100 pg/mL is 80%, most patients with significant congestive heart failure (CHF) as a cause of their dyspnea will have levels of >400 pg/mL.172 Brain natriuretic peptide is elevated probably in all forms of cardiomyopathy including DCM. One study reported higher BNP levels in patients with hypertrophic cardiomyopathy than patients with idiopathic DCM.176 This may be related to the difference in proportional production of BNP by markedly hypertrophied and pressure-loaded ventricles. There is an inverse relationship between the severity of ventricular arrhythmia and LV ejection fraction.178,179 Thus, it is perhaps not surprising that the majority of DCM patients have PVCs when monitored over a 24-hour period; moreover, approximately half of the patients with DCM have nonsustained ventricular tachycardia on routine ambulatory 24hour Holter monitoring. However, the predictive value of ambulatory Holter monitoring as a routine screening tool in asymptomatic patients for identification of risk for sudden cardiac death has not been validated. There are conflicting data from studies involving small groups of patients regarding the role of routine ambulatory ECG monitoring as a significant independent risk factor for sudden cardiac death.180–182 Thus, at the present time, routine screening with Holter monitoring in asymptomatic HF patients is not recommended as a reliable means to predict those patients who will develop sudden cardiac death. Chest Radiography Signal Averaged ECG In the early phases of DCM, cardiac enlargement may be minimal and may not be detected by chest radiography. However, in general, the chest x-ray usually reveals LV enlargement or generalized cardiomegaly that involves all four cardiac chambers. Depending on the patient’s volume status, there may or may not be findings of pulmonary congestion. Cephalization of blood flow or pulmonary vascular redistribution are early signs of volume overload, followed by the development of interstitial edema with appearance of Kerley B lines and fluid in the interlobar fissures, followed by frank alveolar edema in advanced volume overload. Pleural effusions may be present and the azygos vein and superior vena cava may be dilated, especially with RV failure. In patients with syncope or coronary artery disease, the presence of late potentials on a signal average ECG identifies those patients at high risk for developing ventricular tachycardia and sudden death. Although late potentials are less frequent in patients with DCM, the current evidence suggests that an abnormal signal average ECG can predict ventricular arrhythmias and sudden cardiac death also in patients with DCM. However, studies evaluating the utility of a positive signal averaged ECG to identify a subgroup of dilated HF patients at risk for sudden death have yielded disparate results.183–185 Those studies that have demonstrated an increased risk of sudden death in subjects in the presence of late potentials on signal averaged ECG have used different signal averaging methods than those that have not shown the signal averaged ECG to have predictive value. Indeed, the sensitivity of the signal average ECG varies between 22% and 100% and the specificity ranges from 45% to 96% in various studies.182,183,186–188 Thus, although the signal average ECG may be useful for predicting future events in some patients with DCM, the inherent problems with the sensitivity, specificity, and predictive value suggest that the signal average ECG may not be a reliable tool for screening asymptomatic patients who are at a high risk for sudden cardiac death. Electrocardiography If the patient with DCM presents with signs or symptoms referable to HF, the ECG usually reveals sinus tachycardia. However, it is important to note that sinus bradycardia may also be present in many patients with end-stage DCM. The ECG morphology is seldom normal, and often shows nonspecific repolarization or ST segment abnormalities. Conduction abnormalities, especially LBBB, left anterior hemiblock, and nonspecific intraventricular conduction delays, and occasionally first-degree atrioventricular block are common in patients with long-standing symptoms, and they may be markers of increasing interstitial fibrosis or myocyte hypertrophy. Right bundle branch block is rare.177 Left atrial or biatrial enlargement may be present. Pathologic anterior, anterolateral, or diffuse Q waves mimicking myocardial infarction, or poor R wave progression may be seen with viral CAR055.indd 1248 Ambulatory Electrocardiographic (Holter) Monitoring Two-Dimensional and Doppler Echocardiography Two-dimensional (2D) and Doppler echocardiography are extremely useful techniques for assessing LV function, LV dimensions, and valvular structures in DCM patients. Generally, patients will have global LV dilation, LV wall thin- 11/24/2006 1:28:35 PM di l at e d c a r diom yopat h y ning, global hypokinesis, and an LV ejection fraction of less than 35% to 40%. Segmental wall motion abnormalities may be seen with altered regional wall stress due to elevated filling pressures and depressed contractility, or with acute myocarditis due to segmental myocardial injury. Interestingly, the presence of segmental wall motion abnormalities suggests a more favorable outcome than if global hypokinesis is present.189 Left ventricular apical thrombi can be identified in as many as 40% of the patients with DCM.190 Small pericardial effusion can sometimes be demonstrated, especially if an infectious or noninfectious inflammatory etiology is responsible for producing the DCM. Doppler echocardiography usually reveals mild degrees of mitral and tricuspid regurgitation and sometimes trace pulmonary insufficiency. The mechanisms of mitral and tricuspid regurgitation have been attributed to ventricular enlargement, annular dilatation, lengthening of chordae tendinea, abnormalities in contractility of the papillary muscles, and displacement of the coaptation point of the mitral leaflets. The Doppler mitral inflow patterns can also provide useful information on elevated LV filling pressures, which are suggested by the appearance of a high E/A ratio (“pseudonormalization”), and a short isovolumic relaxation time and deceleration time.191,192 The elevated early filling wave results from elevated left atrial pressures, whereas the rapid deceleration time reflects further decrease in LV compliance. These Doppler findings have been associated with a poor prognosis in DCM. Radionuclide Techniques Multigated radionuclide angiocardiography (MUGA) is a very useful test for the initial assessment of LVEF in patients with DCM. Both the right and left ventricular ejection fractions can be quantified reliably with this technique; moreover, the interobserver and inter-test variability appear to be less than with echocardiography.193 Thallium-201 myocardial scintigraphy is not a reliable technique for differentiating patients with ischemic heart disease from those with DCM, insofar as patients with DCM may have both reversible and fi xed perfusion abnormalities that are related to the presence of myocardial fibrosis.194 In addition to the above methodology, radionuclide techniques have also been used to detect the presence of myocardial inflammation. Both gallium-67– (a marker for inflammation) and indium-III–labeled antimyosin antibodies (a marker for myocyte necrosis) have been used to detect myocarditis in small uncontrolled studies.195,196 These techniques are supportive of the diagnosis of myocarditis when clinically suspected, but do not have the specificity or the sensitivity to be used reliably as screening tools for patients with myocarditis.196 Cardiac Catheterization Coronary arteriography is primarily used as a basis for planning further treatment options in patients who have ischemic heart disease. In the majority of patients with DCM, the coronary arteries are normal with no atherosclerotic lesions; however, in a small portion, there may be evidence of mild, nonobstructive, isolated atherosclerotic lesions that may not be sufficient to explain the extent of cardiomyopathy. Therefore, in these cases, other etiologies for the cardio- CAR055.indd 1249 12 4 9 myopathy should be sought. The ventriculogram generally reveals a dilated ventricle with depressed contractility. The LV end-diastolic pressures are usually elevated. Right heart catheterization is a useful means for adjusting medical therapy in patients with advanced symptoms,197 especially when they are considered for vasodilator, inotropic, or mechanical support, or for assessing the suitability of certain patients for cardiac transplantation. Endomyocardial Biopsy The routine use of RV endomyocardial biopsy as a diagnostic tool for evaluating patients with newly diagnosed DCM has fallen out of favor in many laboratories because of the apparent lack of benefit of immunosuppressive therapy in patients with myocarditis.198 Moreover, the current criteria for diagnosing myocarditis by myocardial biopsy may be overly specific and lack sensitivity. Indeed, the reported incidence of biopsy-verified myocarditis ranges from 1% to 70%, with an average detection rate of 10%. The countervailing argument that has been raised in support of the use of endomyocardial biopsy in DCM is that this technique may yield a new diagnosis (i.e., different from idiopathic cardiomyopathy) in up to 20% of patients, and may provide a potentially new or beneficial form of therapy in 5% of the patients.197,199 Thus, myocardial biopsy should be considered in patients with suspected treatable systemic or infiltrative myocardial diseases, such as sarcoidosis, hypereosinophilic syndrome, hypersensitivity myocarditis, amyloidosis, hemochromatosis, doxorubicininduced cardiomyopathy, or fulminant myocarditis. Moreover, as noted above, in certain types of fulminant myocarditis, especially giant cell myocarditis, there is the suggestion that aggressive immunosuppression may attenuate disease progression.200 Therefore, endomyocardial biopsies may be extremely useful in diagnosing patients who present with a fulminant course, or who progressively decompensate despite optimal medical therapy. In experienced laboratories, the risk of perforation of the heart from the biopsy is approximately 1/200 (0.5%) and the risk of death from the procedure is 3 in 10,000 (0.03%).199 Therefore, in electing to perform a myocardial biopsy, the clinician must weigh the potential benefits of rendering a new diagnosis for which a specific form of therapy exists against the risks associated with performing myocardial biopsy.199 Treatment Strategies for Dilated Cardiomyopathy The first priority in implementing treatment strategies for patients with DCM is to determine if the condition has an etiology for which there is a specific form of treatment. Table 55.3 lists a number of “etiology specific” treatment strategies designed to treat the underlying disease process that is responsible for causing the DCM. The second priority in implementing treatment strategies for DCM is to initiate supportive HF therapy, the goals of which should be to (1) improve the quality of life, (2) avoid the need for future hospitalizations, (3) prolong life, and (4) to prevent HF progression. In this section, we discuss only those treatment 11/24/2006 1:28:35 PM 12 5 0 chapter 55 TABLE 55.3. Primary treatment approaches to dilated cardiomyopathies Etiology of DCM Primary treatment Alcoholic Cocaine Anthracycline Systemic lupus erythematosus Viral myocarditis Abstinence Abstinence, beta-blockers? Cessation of anthracycline therapy Steroids, cytotoxic agents Prednisone and immunosuppressant therapy for fulminant course Benzonidazole, nifurtimox Steroids, Ca channel blockers for Raynaud's phenomenon IV immunoglobulin Replacement Achieve euthyroid state Increase CD4 count Dialysis Removal of tumor AV nodal ablation, beta-blockers Chagas’ disease Scleroderma Kawasaki disease Thiamine, selenium, or carnitine deficiency Hyperthyroidism/hypothyroidism AIDS Uremia Pheochromocytoma Tachycardia induced strategies that are unique to the treatment of HF for patients with DCM. Standard Medical Therapy Existing studies suggest that patients with ischemic cardiomyopathy and patients with DCM have significant survival benefit from the use of ACE inhibitors and betablockers.5,6,15,16,21,201–203 In addition to the ACE inhibitors and beta-blockers, the four-drug approach including digoxin and diuretics are advocated for all suitable patients with LV dysfunction and symptomatic HF regardless of the etiology (Fig. 55.5). There is no information on the effects of diuretics on Favors treatment medication Favors placebo SOLVD-T (Enalapril) V-HeFT-II (Enalapril) MERIT HF (Metoprolol-XL) CIBISII (Bisoprolol) BEST (Bucindolol) US Carvedilol (Carvedilol) PRAISE I (Amlodipine) PRAISE II (Amlodipine) 0.2 0.4 0.6 0.8 1.0 1.2 Hazard ratio Val-HeFT (Valsartan) RALES (Spironolactone) 1.4 1.6 1.8 Ischemic Nonischemic FIGURE 55.5. Hazard ratio and 95% confidence intervals for overall mortality or combined end point of overall and cardiovascular mortality for ischemic cardiomyopathy and nonischemic cardiomyopathy from recent multicenter randomized trials with different treatment strategies for heart failure. Medications used are shown in parentheses next to the trial names. Ischemic cardiomyopathy is represented as the black square with solid line, the nonischemic cardiomyopathy as the white square with dashed line. CAR055.indd 1250 morbidity or mortality in ischemic versus nonischemic HF. Pooled results from the Prospective Randomized Study of Ventricular Function and Efficacy of Digoxin (PROVED) and the Randomized Assessment of Digoxin and Inhibitors of Angiotensin-Converting Enzyme (RADIANCE) trials indicate that digoxin increases LVEF more in patients with DCM than in patients with ischemic heart disease, and that withdrawal of digoxin leads to significantly greater likelihood of clinical deterioration in the DCM patients.204 In the Digitalis Investigators’ Group trial,63 digoxin was associated with a significant reduction in a combined end point of death or hospitalization as a result of worsening HF in patients in sinus rhythm. This benefit was somewhat greater in patients with nonischemic etiology than in those with ischemic etiology. That is, patients with DCM were noted to have a 33% risk reduction in the combined end point of death and HF hospitalization compared to a 21% risk reduction in patients with ischemic cardiomyopathy. The cause of this therapeutic difference is not clear at this point. Aldosterone antagonism with spironolactone has been reported to reduce mortality in patients with advanced HF, both in dilated and ischemic cardiomyopathy.205 According to the results of the Randomized Aldactone Evaluation Study (RALES) trial,205 spironolactone (25 to 50 mg/day) is recommended for patients with recent or current symptoms of HF at rest despite the use of ACE inhibitors, diuretics, digoxin, and a beta-blocker, regardless of the etiology of HF, in patients with NYHA class III to IV HF and LVEF <35%, with adjustment of potassium supplements and close laboratory follow-up. In the last decade, several studies have shown that angiotensin receptor blockers (ARBs), when added to background therapy with ACE inhibitors, can result in improvement in combined morbidity and mortality in patients with chronic HF6,206 in both ischemic and nonischemic DCM patients, similar to the benefit shown with spironolactone. It is important to recognize that in chronic HF, the ARB trials6,206,207 were conducted predominantly in patients with mild to moderate HF (NYHA class II to III), whereas the RALES trial205 11/24/2006 1:28:35 PM di l at e d c a r diom yopat h y (the only aldosterone receptor antagonist trial in chronic HF including patients with nonischemic etiology), was conducted in patients with severe HF (NYHA class III to IV). At the present time, there are no large-scale, prospective, randomized clinical trials with aldosterone receptor blockers in mild to moderate chronic HF patients, and there are no headto-head trials comparing ARBs and aldosterone receptor blockers in the treatment of HF, either for ischemic or nonischemic patients. Thus, it is not feasible to compare the efficacy or safety of these two treatment strategies in mild to moderate HF at the present time. The use of first-generation calcium antagonists (diltiazem, verapamil, or nifedipine) is potentially harmful in all patients with HF.208 Recent studies with second-generation calcium channel blockers, such as amlodipine and felodipine, suggest that these agents may be safer but do not confer survival benefit in patients with HF. Initial reports of survival benefit with amlodipine [Prospective Randomized Amlodipine Survival Evaluation Study (PRAISE-I)]65 in patients with dilated but not ischemic cardiomyopathy have failed to reproduce the subsequent large-scale randomized trial (PRAISE-II),66 in which the survival was similar in both the ischemic and nonischemic HF patients. Studies with felodipine showed similar results. In the V-HeFT III trial,209 felodipine, when used as supplementary vasodilator therapy in patients with chronic HF, was safe but had no additional benefit. Recently, the addition of a fi xed dose of isosorbide dinitrate plus hydralazine to standard therapy for HF including ACE inhibitors and beta-blockers was shown to be efficacious in increasing survival among black patients with advanced (NYHA class III to IV) HF.210 Interestingly, in 77% to 78% of the patients in this trial, the etiology of HF was nonischemic (40% due to hypertension), but the subgroup analysis of outcomes according to the etiology of HF has not been reported.210 Antiarrhythmic Therapy: Implantable Devices The role of implantable defibrillators for secondary prevention of sudden cardiac death is clearly established for both ischemic and nonischemic cardiomyopathy patients. Implantable defibrillators decrease sudden death risk in patients with LV dysfunction who have survived sudden cardiac death, or sustained ventricular tachycardia or fibrillation.211 For primary prevention, there is a large body of literature supporting the prophylactic use of implantable defibrillators to prevent sudden cardiac death in ischemic cardiomyopathy patients with reduced LVEF and post–myocardial infarction.212,213 Similar evidence supporting the role of prophylactic ICD implantation in patients with DCM, however, is controversial. Two earlier studies have examined the role of ICD therapy in patients with nonischemic cardiomyopathy and HF, the Amiodarone Versus Implantable Cardioverter– Defibrillator Trial (AMIOVIRT)214 and the DEFINITE trial215 failed to show a significant survival benefit, whereas the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT)216 provided the first large-scale evidence of survival benefit with ICDs for primary prevention of sudden cardiac death in patients with DCM. In this trial, in patients with NYHA class II or III HF and a LVEF of 35%, the single-lead, shock-only ICD therapy reduced overall mortality by 23%, CAR055.indd 1251 12 51 whereas amiodarone had no favorable effect on survival. The cause of HF was ischemic in 52% and nonischemic in 48%, and the results did not vary according to either ischemic or nonischemic causes of HF. Subsequently, the Center for Medicare and Medicaid Services (CMS) expanded its ICD coverage substantially to reflect the results of this trial, including coverage for patients with nonischemic DCM with duration over 9 months and measured LVEF ≤30%, if the patient is enrolled in either a clinical trial or a qualifying national database.217 Medical Antiarrhythmic Therapy Studies addressing the role of antiarrhythmic therapy in prolonging survival or preventing sudden death in HF have yielded disparate results. The Survival Trial of Antiarrhythmic Therapy in Congestive Heart Failure with Amiodarone (CHF-STAT)64 revealed that amiodarone treatment was effective in suppressing ventricular arrhythmias and improving ventricular function; however, it did not reduce the incidence of sudden death or prolong survival among patients with HF, except for a trend toward reduced mortality among those with nonischemic cardiomyopathy. In the trial by Grupo de Estudio de la Sobrevida en la Insuficiencia Cardiaca en Argentina (GESICA),218 low-dose amiodarone resulted in reduction in total mortality, sudden cardiac death, and HF death in both the dilated nonischemic and ischemic patients. As mentioned above, in the SCD-HeFT trial,216 amiodarone failed to have any beneficial effect on survival when compared with placebo in HF patients regardless of etiology. Presently, antiarrhythmic medical therapy is reserved for individualized treatment of symptomatic arrhythmias, especially for suppression of ventricular arrhythmias after ICD implantation or supraventricular arrhythmias such as atrial fibrillation. It is important to emphasize that beta-blockers have also been shown to reduce sudden cardiac death risk, by approximately 30%, in patients with ischemic or nonischemic HF in addition to their overall benefit in reducing mortality and HF hospitalizations.5,7,15,203 Cardiac Resynchronization Therapy Many patients with DCM have cardiac conduction abnormalities, such as LBBB or intraventricular conduction delays (IVCDs) that can lead to ventricular dyssynchrony and reduction in stroke volume as a consequence. In patients with HF, the presence of LBBB or IVCD further degrades ventricular function, contributing directly to the severity of their HF symptoms. Biventricular pacing with transvenous leads in the right atrium, right ventricle, and left ventricle via the coronary sinus can restore cardiac resynchronization. Several recent studies—the Multicenter InSync Randomized Clinical Evaluation (MIRACLE),219 Multisite Stimulation in Cardiomyopathy (MUSTIC),220 Comparison of Medical Therapy, Pacing, and Defibrillation in Heart Failure (COMPANION),221 and the Cardiac Resynchronization–Heart Failure (CARE-HF)222 trials—validated the safety and efficacy of cardiac resynchronization therapy in advanced HF. Data from these studies have shown statistically significant improvements in LVEF, NYHA class, exercise 11/24/2006 1:28:35 PM 12 5 2 chapter tolerance, and quality of life. Furthermore, the COMPANION trial demonstrated that in patients with advanced HF and a prolonged QRS interval, cardiac-resynchronization therapy decreased the combined risk of death from any cause or first hospitalization, and when combined with an implantable defibrillator, significantly reduced mortality.221 However, in the COMPANION trial, the decrease in the risk of death was not significant with cardiac resynchronization therapy alone. The more recent CARE-HF trial222 demonstrated that in patients with HF, class III or IV HF due to left ventricular systolic dysfunction (LVEF <35%), and cardiac dyssynchrony (QRS >120 ms and/or other evidence of dyssynchrony), cardiac resynchronization improved the symptoms and the quality of life, and reduced the risk of death. In these trials, the benefit was present in both ischemic and nonischemic HF patients. The proposed mechanisms involved in improving ventricular function with biventricular pacing include improved septal contribution to ventricular ejection, increased diastolic filling times, reduced mitral regurgitation, and prevention of progressive remodeling. Resynchronization therapy may also reduce arrhythmias by stabilization of the arrhythmia substrate and by improving the HF status. Anticoagulants Patients with DCM have multiple factors that predispose them to thromboembolic events. However, the appropriateness of chronic anticoagulant therapy in DCM has been controversial.223 The Warfarin and Antiplatelet Therapy in Chronic Heart failure (WATCH) trial, a multicenter, randomized, international trial comparing the efficacy of aspirin, clopidogrel, and warfarin in HF patients, was the largest trial of antithrombotic therapies conducted in chronic HF patients. It reportedly failed to show major differences among the three medications to prevent the combined end point of nonfatal myocardial infarction, stroke, and mortality.224 In the absence of controlled trials, the available data support the use of anticoagulants in the presence of atrial fibrillation, previous stroke or other thromboembolic events, or visible protruding or mobile thrombus on echocardiography. Immunosuppression and Immunomodulation Although an autoimmune pathogenesis has been postulated for DCM, immunosuppressive therapy has not been shown to be effective in clinical trials. The failure of the Myocarditis Treatment Trial225 to show a significant benefit for immunosuppressive therapy is discussed in another chapter. Based on its possible immunomodulatory effects, intravenous immunoglobulin has been studied as a potential beneficial agent in patients with DCM. Although the exact mechanism of action of intravenous immunoglobulin is not known, a number of different mechanisms have been proposed, including Fc receptor blockade, neutralization of autoantibodies, modulation of cytokine activity, and activation of an inhibitory Fc receptor. Based on an initial report that intravenous immunoglobulin was beneficial in acute cardiomyopathy, Gullestad and colleagues226 conducted a double-blind trial with intravenous immunoglobulin for 26 weeks in 47 patients with moderate HF who were receiving conventional therapy CAR055.indd 1252 55 for HF, including ACE inhibitors and beta-blockers. They observed that, in comparison to placebo, intravenous immunoglobulin induced a marked rise in plasma levels of the antiinflammatory mediators (interleukin-10, and interleukin-1 receptor antagonist), and that these changes were accompanied by a significant increase in LV ejection performance by 10%, and a decrease in N-terminal pro-atrial natriuretic peptide levels. Thus, in this small study, immunomodulatory therapy with intravenous immunoglobulin was effective in patients with HF. Immune modulatory therapy with immune globulin is an effective therapy for Kawasaki disease in children, and it has been reported to improve ventricular function in children with new-onset DCM,227 in adults228 with recent onset of symptoms of HF, and in women with postpartum cardiomyopathy.229 However, the initial promising results observed in the adult population were not confirmed by the randomized, placebo-controlled, multicenter Intervention in Myocarditis and Acute Cardiomyopathy (IMAC) trial.230 In this trial, no significant treatment effect could be seen with intravenous immune globulin over placebo. Both groups had a 14% increase in LVEF. Whether the observed differences represent differences in patient selection or dosing is not known at this time. The technique of immunoadsorption, removal of autoantibodies with immunoadsorption by passing a patient’s plasma over columns that contain immobilized antibodies against immunoglobulin (IgG) kappa and lambda light chains and IgG heavy chains, has been reported to be beneficial in DCM. Wallukat et al.231 were the first to show that this technique efficiently removed circulating antibodies directed against the β1-adrenoreceptor and were associated with an improvement in NYHA functional class in patients with DCM. This study was soon followed by another pilot study that reported an improvement in short-term hemodynamic effects in patients with severe HF who were refractory to conventional medical therapy.232 However, these early studies lacked a concurrent control group. Felix and coworkers233 showed that immunoadsorption and intravenous IgG administration resulted in a significant improvement in cardiac hemodynamics in 18 patients with DCM who were randomized for 3 months to immunoadsorption and subsequent IgG replacement or conventional medical therapy. Most recently, Muller and coworkers234 reported that within 1 year of treatment, immunoadsorption therapy resulted in a significant lowering of anti–β1-adrenoreceptor antibodies, a significant (70%) increase in ejection fraction, and a significant (15%) decrease in LV end-diastolic dimension in the treatment group. Taken together, the above studies suggest that immunoadsorption leads to improvements not only in LV structure and function but also in functional status in selected patients with DCM. However, it is important to identify the existing gaps in our knowledge with respect to immunoadsorption in patients with DCM. First, at the present time, it is unclear whether all patients with DCM will benefit from immunoadsorption therapy, or whether patients with elevated levels of circulating autoantibodies should be treated. Second, the mechanism(s) of action of immunoadsorption is not known. Although studies have demonstrated decreases in circulating autoantibodies, it is not at all clear that a cause-and-effect relationship has been established. 11/24/2006 1:28:35 PM di l at e d c a r diom yopat h y Immunoadsorption can lead to a decline in systemic vascular resistance independent of the removal of circulating autoantibodies.167 Thus, part of the hemodynamic benefit that has been observed may be related to vasodilation and not an improvement in LV function. Third, the optimal strategy for immunoadsorption has yet to be determined. Different investigators use different protocols and different immunoadsorbent devices. It is not clear from existing studies whether immunoadsorption alone, which would be expected to modulate humoral immunity, will be sufficient in the long term, or whether it may be necessary to incorporate strategies that lead to suppression of cellular-mediated immunity as well.167 Growth Hormone There has been growing enthusiasm for the use of growth hormone in patients with DCM, based on the rationale that there may be inadequate compensatory cardiac hypertrophy in this disorder. Growth hormone and IGF-1 are involved in several physiologic processes such as the control of muscle mass and function, body composition, and regulation of nutrient metabolism. Earlier reports suggested that growth hormone therapy could increase myocardial mass and reduce the size of the LV chamber, resulting in improvement in hemodynamics, myocardial energy metabolism, and the clinical status of the patients with HF.235 More recently, two randomized, placebo-controlled studies of recombinant human growth hormone did not show significant improvement in cardiac performance or clinical status of patients with DCM, despite significant increases in LV mass related to changes in serum IGF-1 concentrations.143,236 Whether longer treatment with recombinant growth hormone would result in an improved clinical outcome in the patients with DCM is unknown. Gene Therapy After a decade of preclinical and early phase one clinical investigations, gene therapy is proving likely to be a viable alternative to conventional therapies for HF. So far in preclinical studies, three types of gene transfer studies have been investigated to enhance myocardial contractility: modulating calcium homeostasis (overexpression of SERCA2a by rat or transgenic mice DCM models), manipulating β-adrenergic receptor signaling (transfection or overexpression of βadrenergic receptor gene in rat or rabbit DCM models), and augmenting cardiomyocyte resistance to apoptosis (adenoviral gene transfer of antiapoptotic Bcl-2 and Akt in gp130 knockout mice, with DCM associated with massive cardiomyocyte apoptosis).237 However, at present, it has been difficult to develop highly effective gene delivery techniques in the myocardium. Since the identification of the gene for Duchenne muscular dystrophy in 1987, many groups have reported successful approaches for the adenoviral vector delivery of dystrophin, the Duchenne muscular dystrophy gene product, resulting in restoration of dystrophin-associated glycoprotein complex proteins to the sarcolemma and amelioration of muscular dystrophy symptoms in animal models.238 Although dystrophin is the most extensively studied gene that is implicated CAR055.indd 1253 12 5 3 in DCM, correction of the dystrophin defects, associated with X-linked cardiomyopathy, has not been studied in humans yet.237,238 Angiogenesis and Cell Therapy These modalities are discussed in another chapter. Acknowledgments The authors would like to thank Dr. Andrew I. Schafer for his past and present guidance and support. The research reviewed herein was supported, in part, by research funds from the Department of Veterans Affairs and the National Institutes of Health (P50 HL-O6H, RO1 HL58081-01A1, and RO1 HL61543-01). References 1. Report of the WHO/ISFC task force on the defi nition and classification of cardiomyopathies. Br Heart J 1980;44(6):672–673. 2. Richardson P, McKenna W, Bristow M, et al. Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Defi nition and Classification of cardiomyopathies. Circulation 1996;93(5):841–842. 3. Maisch B. [Classification of cardiomyopathies according to the WHO/ISFC Task Force—more questions than answers?] Med Klin 1998;93(4):199–209. 4. Boffa GM, Tarantini G, Abbasciano A, Razzolini R, Chioin R, Thiene G. Ischemic cardiomyopathy: lack of clinical applicability of the WHO/ISFC classification of cardiomyopathies. Ital Heart J 2001;2(10):778–781. 5. Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet 1999;353(9169):2001–2007. 6. Cohn JN, Tognoni G. A randomized trial of the angiotensinreceptor blocker valsartan in chronic heart failure. N Engl J Med 2001;345(23):1667–1675. 7. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet 1999;353(9146):9–13. 8. Manolio TA, Baughman KL, Rodeheffer R, et al. Prevalence and etiology of idiopathic dilated cardiomyopathy (summary of a National Heart, Lung, and Blood Institute workshop. Am J Cardiol 1992;69(17):1458–1466. 9. Coughlin SS, Gottdiener JS, Baughman KL, et al. Black-white differences in mortality in idiopathic dilated cardiomyopathy: the Washington, DC, Dilated Cardiomyopathy Study. J Natl Med Assoc 1994;86(8):583–591. 10. Dries DL, Exner DV, Gersh BJ, Cooper HA, Carson PE, Domanski MJ. Racial differences in the outcome of left ventricular dysfunction. N Engl J Med 1999;340(8):609–616. 11. Coughlin SS, Myers L, Michaels RK. What explains blackwhite differences in survival in idiopathic dilated cardiomyopathy? The Washington, DC, Dilated Cardiomyopathy Study. J Natl Med Assoc 1997;89(4):277–282. 12. Watkins LO, Williams RA. Cardiomyopathy in blacks. Cardiovasc Clin 1991;21(3):279–296. 13. Yancy CW, Fowler MB, Colucci WS, et al. Race and the response to adrenergic blockade with carvedilol in patients with chronic heart failure. N Engl J Med 2001;344(18):1358–1365. 14. Carson P, Ziesche S, Johnson G, Cohn JN. Racial differences in response to therapy for heart failure: analysis of the vasodilator-heart failure trials. Vasodilator-Heart Failure Trial Study Group. J Card Fail 1999;5(3):178–187. 15. A trial of the beta-blocker bucindolol in patients with advanced chronic heart failure. N Engl J Med 2001;344(22):1659–1667. 11/24/2006 1:28:36 PM 12 5 4 chapter 16. Braunwald E. Expanding indications for beta-blockers in heart failure. N Engl J Med 2001;344(22):1711–1712. 17. Exner DV, Dries DL, Domanski MJ, Cohn JN. Lesser response to angiotensin-converting-enzyme inhibitor therapy in black as compared with white patients with left ventricular dysfunction. N Engl J Med 2001;344(18):1351–1357. 18. Dries DL, Strong MH, Cooper RS, Drazner MH. Efficacy of angiotensin-converting enzyme inhibition in reducing progression from asymptomatic left ventricular dysfunction to symptomatic heart failure in black and white patients. J Am Coll Cardiol 2002;40(2):311–317. 19. Ho KK, Anderson KM, Kannel WB, Grossman W, Levy D. Survival after the onset of congestive heart failure in Framingham Heart Study subjects. Circulation 1993;88(1):107–115. 20. Schocken DD, Arrieta MI, Leaverton PE, Ross EA. Prevalence and mortality rate of congestive heart failure in the United States. J Am Coll Cardiol 1992;20(2):301–306. 21. 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(5): 293–302. 22. Bourassa MG, Gurne O, Bangdiwala SI, et al. Natural history and patterns of current practice in heart failure. The Studies of Left Ventricular Dysfunction (SOLVD) Investigators. J Am Coll Cardiol 1993;22(4 Suppl A):14A–19A. 23. De Maria R, Gavazzi A, Recalcati F, Baroldi G, De Vita C, Camerini F. Comparison of clinical fi ndings in idiopathic dilated cardiomyopathy in women versus men. The Italian Multicenter Cardiomyopathy Study Group (SPIC). Am J Cardiol 1993;72(7):580–585. 24. Ghali JK, Pina IL, Gottlieb SS, Deedwania PC, Wikstrand JC. Metoprolol CR/XL in female patients with heart failure: analysis of the experience in Metoprolol Extended-Release Randomized Intervention Trial in Heart Failure (MERIT-HF). Circulation 2002;105(13):1585–1591. 25. Simon T, Mary-Krause M, Funck-Brentano C, Jaillon P. Sex differences in the prognosis of congestive heart failure: results from the Cardiac Insufficiency Bisoprolol Study (CIBIS II). Circulation 2001;103(3):375–380. 26. Dec GW, Fuster V. Idiopathic dilated cardiomyopathy. N Engl J Med 1994;331(23):1564–1575. 27. Fuster V, Gersh BJ, Giuliani ER, Tajik AJ, Brandenburg RO, Frye RL. The natural history of idiopathic dilated cardiomyopathy. Am J Cardiol 1981;47(3):525–531. 28. Sugrue DD, Rodeheffer RJ, Codd MB, Ballard DJ, Fuster V, Gersh BJ. The clinical course of idiopathic dilated cardiomyopathy. A population-based study. Ann Intern Med 1992;117(2): 117–123. 29. Miura K, Nakagawa H, Morikawa Y, et al. Epidemiology of idiopathic cardiomyopathy in Japan: results from a nationwide survey. Heart 2002;87(2):126–130. 30. Amoah AG, Kallen C. Aetiology of heart failure as seen from a National Cardiac Referral Centre in Africa. Cardiology 2000; 93(1–2):11–18. 31. Mendez GF, Cowie MR. The epidemiological features of heart failure in developing countries: a review of the literature. Int J Cardiol 2001;80(2–3):213–219. 32. Nelson DE, Bland S, Powell-Griner E, et al. State trends in health risk factors and receipt of clinical preventive services among US adults during the 1990s. JAMA 2002;287(20): 2659–2667. 33. Saxon LA, De Marco T. Arrhythmias associated with dilated cardiomyopathy. Card Electrophysiol Rev 2002;6(1–2):18–25. 34. Saxon LA, Stevenson WG, Middlekauff HR, et al. Predicting death from progressive heart failure secondary to ischemic or idiopathic dilated cardiomyopathy. Am J Cardiol 1993;72(1): 62–65. CAR055.indd 1254 55 35. Steimle AE, Stevenson LW, Fonarow GC, Hamilton MA, Moriguchi JD. Prediction of improvement in recent onset cardiomyopathy after referral for heart transplantation. J Am Coll Cardiol 1994;23(3):553–559. 36. Romeo F, Pelliccia F, Cianfrocca C, et al. Determinants of endstage idiopathic dilated cardiomyopathy: a multivariate analysis of 104 patients. Clin Cardiol 1989;12(7):387–392. 37. Lewis JF, Webber JD, Sutton LL, Chesoni S, Curry CL. Discordance in degree of right and left ventricular dilation in patients with dilated cardiomyopathy: recognition and clinical implications. J Am Coll Cardiol 1993;21(3):649–654. 38. Juilliere Y, Barbier G, Feldmann L, Grentzinger A, Danchin N, Cherrier F. Additional predictive value of both left and right ventricular ejection fractions on long-term survival in idiopathic dilated cardiomyopathy. Eur Heart J 1997;18(2): 276–280. 39. Werner GS, Schaefer C, Dirks R, Figulla HR, Kreuzer H. Prognostic value of Doppler echocardiographic assessment of left ventricular fi lling in idiopathic dilated cardiomyopathy. Am J Cardiol 1994;73(11):792–798. 40. Junker A, Thayssen P, Nielsen B, Andersen PE. The hemodynamic and prognostic significance of echo-Doppler-proven mitral regurgitation in patients with dilated cardiomyopathy. Cardiology 1993;83(1–2):14–20. 41. De Maria R, Gavazzi A, Caroli A, Ometto R, Biagini A, Camerini F. Ventricular arrhythmias in dilated cardiomyopathy as an independent prognostic hallmark. Italian Multicenter Cardiomyopathy Study (SPIC) Group. Am J Cardiol 1992;69(17): 1451–1457. 42. Yi G, Goldman JH, Keeling PJ, Reardon M, McKenna WJ, Malik M. Heart rate variability in idiopathic dilated cardiomyopathy: relation to disease severity and prognosis. Heart 1997;77(2): 108–114. 43. Fauchier L, Babuty D, Cosnay P, Poret P, Rouesnel P, Fauchier JP. Long-term prognostic value of time domain analysis of signal-averaged electrocardiography in idiopathic dilated cardiomyopathy. Am J Cardiol 2000;85(5):618–623. 44. Hammond EH, Menlove RL, Anderson JL. Predictive value of immunofluorescence and electron microscopic evaluation of endomyocardial biopsies in the diagnosis and prognosis of myocarditis and idiopathic dilated cardiomyopathy. Am Heart J 1987;114(5):1055–1065. 45. Koglin J, Pehlivanli S, Schwaiblmair M, Vogeser M, Cremer P, vonScheidt W. Role of brain natriuretic peptide in risk stratification of patients with congestive heart failure. J Am Coll Cardiol 2001;38(7):1934–1941. 46. Bahler RC. Assessment of prognosis in idiopathic dilated cardiomyopathy. Chest 2002;121(4):1016–1019. 47. Deswal A, Petersen NJ, Feldman AM, Young JB, White BG, Mann DL. Cytokines and cytokine receptors in advanced heart failure: an analysis of the cytokine database from the Vesnarinone Trial (VEST). Circulation 2001;103(16):2055– 2059. 48. Seta Y, Shan K, Bozkurt B, Oral H, Mann DL. Basic mechanisms in heart failure: the cytokine hypothesis. J Card Fail 1996;2(3):243–249. 49. Sato Y, Yamada T, Taniguchi R, et al. Persistently increased serum concentrations of cardiac troponin T in patients with idiopathic dilated cardiomyopathy are predictive of adverse outcomes. Circulation 2001;103(3):369–374. 50. Hossein-Nia M, Baig K, Goldman JH, et al. Creatine kinase isoforms as circulating markers of deterioration in idiopathic dilated cardiomyopathy. Clin Cardiol 1997;20(1):55–60. 51. Likoff MJ, Chandler SL, Kay HR. Clinical determinants of mortality in chronic congestive heart failure secondary to idiopathic dilated or to ischemic cardiomyopathy. Am J Cardiol 1987;59(6):634–638. 11/24/2006 1:28:36 PM di l at e d c a r diom yopat h y 52. Cohn JN, Johnson GR, Shabetai R, et al. Ejection fraction, peak exercise oxygen consumption, cardiothoracic ratio, ventricular arrhythmias, and plasma norepinephrine as determinants of prognosis in heart failure. The V-HeFT VA Cooperative Studies Group. Circulation 1993;87(6 Suppl):VI5–16. 53. Mancini DM, Eisen H, Kussmaul W, Mull R, Edmunds LH Jr, Wilson JR. Value of peak exercise oxygen consumption for optimal timing of cardiac transplantation in ambulatory patients with heart failure. Circulation 1991;83(3):778–786. 54. Lee WH, Packer M. Prognostic importance of serum sodium concentration and its modification by converting-enzyme inhibition in patients with severe chronic heart failure. Circulation 1986;73(2):257–267. 55. van den Broek SA, van Veldhuisen DJ, de Graeff PA, Landsman ML, Hillege H, Lie KI. Comparison between New York Heart Association classification and peak oxygen consumption in the assessment of functional status and prognosis in patients with mild to moderate chronic congestive heart failure secondary to either ischemic or idiopathic dilated cardiomyopathy. Am J Cardiol 1992;70(3):359–363. 56. Naqvi TZ, Goel RK, Forrester JS, Siegel RJ. Myocardial contractile reserve on dobutamine echocardiography predicts late spontaneous improvement in cardiac function in patients with recent onset idiopathic dilated cardiomyopathy. J Am Coll Cardiol 1999;34(5):1537–1544. 57. Marmor A, Schneeweiss A. Prognostic value of noninvasively obtained left ventricular contractile reserve in patients with severe heart failure. J Am Coll Cardiol 1997;29(2):422–428. 58. Nagaoka H, Isobe N, Kubota S, et al. Myocardial contractile reserve as prognostic determinant in patients with idiopathic dilated cardiomyopathy without overt heart failure. Chest 1997;111(2):344–350. 59. Scrutinio D, Napoli V, Passantino A, Ricci A, Lagioia R, Rizzon P. Low-dose dobutamine responsiveness in idiopathic dilated cardiomyopathy: relation to exercise capacity and clinical outcome. Eur Heart J 2000;21(11):927–934. 60. Drozdz J, Krzeminska-Pakula M, Plewka M, Ciesielczyk M, Kasprzak JD. Prognostic value of low-dose dobutamine echocardiography in patients with idiopathic dilated cardiomyopathy. Chest 2002;121(4):1216–1222. 61. Ehlert FA, Cannom DS, Renfroe EG, et al. Comparison of dilated cardiomyopathy and coronary artery disease in patients with life-threatening ventricular arrhythmias: Differences in presentation and outcome in the AVID registry. Am Heart J 2001;142(5):816–822. 62. Follath F, Cleland JG, Klein W, Murphy R. Etiology and response to drug treatment in heart failure. J Am Coll Cardiol 1998; 32(5):1167–1172. 63. The effect of digoxin on mortality and morbidity in patients with heart failure. The Digitalis Investigation Group. N Engl J Med 1997;336(8):525–533. 64. Singh SN, Fletcher RD, Fisher SG, et al. Amiodarone in patients with congestive heart failure and asymptomatic ventricular arrhythmia. Survival Trial of Antiarrhythmic Therapy in Congestive Heart Failure. N Engl J Med 1995;333(2): 77–82. 65. 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(15):1107–1114. 66. Thackray S, Witte K, Clark AL, Cleland JG. Clinical trials update: OPTIME-CHF, PRAISE-2, ALL-HAT. Eur J Heart Fail 2000;2(2):209–212. 67. Michels VV, Moll PP, Miller FA, et al. The frequency of familial dilated cardiomyopathy in a series of patients with idiopathic dilated cardiomyopathy. N Engl J Med 1992;326(2): 77–82. CAR055.indd 1255 12 5 5 68. Grunig E, Tasman JA, Kucherer H, Franz W, Kubler W, Katus HA. Frequency and phenotypes of familial dilated cardiomyopathy. J Am Coll Cardiol 1998;31(1):186–194. 69. Piano MR. Alcoholic cardiomyopathy: incidence, clinical characteristics, and pathophysiology. Chest 2002;121(5):1638–1650. 70. Preedy VR, Atkinson LM, Richardson PJ, Peters TJ. Mechanisms of ethanol-induced cardiac damage. Br Heart J 1993; 69(3):197–200. 71. Wilke A, Kaiser A, Ferency I, Maisch B. [Alcohol and myocarditis]. Herz 1996;21(4):248–257. 72. Schenk EA, Cohen J. The heart in chronic alcoholism. Clinical and pathologic fi ndings. Pathol Microbiol (Basel) 1970;35(1): 96–104. 73. Cerqueira MD, Harp GD, Ritchie JL, Stratton JR, Walker RD. Rarity of preclinical alcoholic cardiomyopathy in chronic alcoholics less than 40 years of age. Am J Cardiol 1991;67(2): 183–187. 74. Regan TJ. Alcohol and the cardiovascular system. JAMA 1990; 264(3):377–381. 75. Lang RM, Borow KM, Neumann A, Feldman T. Adverse cardiac effects of acute alcohol ingestion in young adults. Ann Intern Med 1985;102(6):742–747. 76. Diamond I. Alcoholic myopathy and cardiomyopathy. N Engl J Med 1989;320(7):458–460. 77. Fernandez-Sola J, Estruch R, Grau JM, Pare JC, Rubin E, UrbanoMarquez A. The relation of alcoholic myopathy to cardiomyopathy. Ann Intern Med 1994;120:529–536. 78. Patel VB, Why HJ, Richardson PJ, Preedy VR. The effects of alcohol on the heart. Adverse Drug React Toxicol Rev 1997; 16:15–43. 79. Guarnieri T, Lakatta EG. Mechanism of myocardial contractile depression by clinical concentrations of ethanol: a study in ferret papillary muscles. J Clin Invest 1990;85:1462–1467. 80. Capasso JM, Li P, Guideri G, Malhotra A, Cortese R, Anversa P. Myocardial mechanical, biochemical, and structural alterations induced by chronic ethanol ingestion in rats. Circ Res 1992;71(2):346–356. 81. Cartwright MM, Smith SM. Increased cell death and reduced neural crest cell numbers in ethanol-exposed embryos: partial basis for the fetal alcohol syndrome phenotype. Alcohol Clin Exp Res 1995;19(2):378–386. 82. Teragaki M, Takeuchi K, Toda I, et al. Point mutations in mitochondrial DNA of patients with alcoholic cardiomyopathy. Heart Vessels 2000;15(4):172–175. 83. Pavan D, Nicolosi GL, Lestuzzi C, Burelli C, Zardo F, Zanuttini D. Normalization of variables of left ventricular function in patients with alcoholic cardiomyopathy after cessation of excessive alcohol intake: an echocardiographic study. Eur Heart J 1987;85:535–540. 84. Nethala V, Brown EJ Jr, Timson CR, Patcha R. Reversal of alcoholic cardiomyopathy in a patient with severe coronary artery disease. Chest 1993;104:626. 85. Fauchier L, Babuty D, Poret P, et al. Comparison of long-term outcome of alcoholic and idiopathic dilated cardiomyopathy. Eur Heart J 2000;21(4):306–314. 86. Bertolet BD, Freund G, Martin CA, Perchalski DL, Williams CM, Pepine CJ. Unrecognized left ventricular dysfunction in an apparently healthy cocaine abuse population. Clin Cardiol 1990;13:323–328. 87. Chakko S, Myerburg RJ. Cardiac complications of cocaine abuse. Clin Cardiol 1995;18(2):67–72. 88. Roldan CA, Aliabadi D, Crawford MH. Prevalence of heart disease in asymptomatic chronic cocaine users. Cardiology 2001;95(1):25–30. 89. Moliterno DJ, Willard JE, Lange RA, et al. Coronary-artery vasoconstriction induced by cocaine, cigarette smoking, or both. N Engl J Med 1994;330(7):454–459. 11/24/2006 1:28:36 PM 12 5 6 chapter 90. Mann DL, Kent RL, Parsons B, Cooper G. Adrenergic effects on the biology of the adult mammalian cardiocyte. Circulation 1992;85(2):790–804. 91. Shin GY, Rice P. Cocaine use and acute coronary syndromes. Lancet 2001;358(9290):1367–1368. 92. Virmani R, Robinowitz M, Smialek JE, Smyth DF. Cardiovascular effects of cocaine: an autopsy study of 40 patients. Am Heart J 1988;115:1068–1076. 93. Henzlova MJ, Smith SH, Prchal VM, Helmcke FR. Apparent reversibility of cocaine-induced congestive cardiomyopathy. Am Heart J 1991;122:577–579. 94. Leikin JB. Cocaine and beta-adrenergic blockers: a remarriage after a decade-long divorce? Crit Care Med 1999;27(4): 688–689. 95. Olson RD, Mushlin PS. Doxorubicin cardiotoxicity: analysis of prevailing hypotheses. FASEB J 1990;4:3076–3086. 96. Shan K, Lincoff AM, Young JB. Anthracycline-induced cardiotoxicity. Ann Intern Med 1996;125(1):47–58. 97. Allen A. The cardiotoxicity of chemotherapeutic drugs. Semin Oncol 1992;19(5):529–542. 98. Lipshultz SE, Lipsitz SR, Mone SM, et al. Female sex and drug dose as risk factors for late cardiotoxic effects of doxorubicin therapy for childhood cancer. N Engl J Med 1995;332:1738– 1743. 99. Bristow MR, Mason JW, Billingham ME, Daniels JR. Doxorubicin cardiomyopathy: evaluation by phonocardiography, endomyocardial biopsy, and cardiac catheterization. Ann Intern Med 1978;88:168–175. 100. Sparano JA, Wolff AC, Brown D. Troponins for predicting cardiotoxicity from cancer therapy. Lancet 2000;356(9246): 1947–1948. 101. Saini J, Rich MW, Lyss AP. Reversibility of severe left ventricular dysfunction due to doxorubicin cardiotoxicity. Report of three cases. Ann Intern Med 1987;106:814–816. 102. Doroshow JH. Doxorubicin-induced cardiac toxicity. N Engl J Med 1991;324:843–845. 103. Hellmann K. Anthracycline cardiotoxicity prevention by dexrazoxane: breakthrough of a barrier—sharpens antitumor profi le and therapeutic index. J Clin Oncol 1996;14(2):332–333. 104. Nelson MA, Frishman WH, Seiter K, Keefe D, Dutcher J. Cardiovascular considerations with anthracycline use in patients with cancer. Heart Dis 2001;3(3):157–168. 105. Schneider JW, Chang AY, Garratt A. Trastuzumab cardiotoxicity: Speculations regarding pathophysiology and targets for further study. Semin Oncol 2002;29(3 suppl 11):22–28. 106. Bozkurt B, Kribbs S, Clubb FJ Jr, et al. Pathophysiologically relevant concentrations of tumor necrosis factor-α promote progressive left ventricular dysfunction and remodeling in rats. Circulation 1998;97:1382–1391. 107. Kubota T, McTiernan CF, Frye CS, et al. Dilated cardiomyopathy in transgenic mice with cardiac specific overexpression of tumor necrosis factor-alpha. Circ Res 1997;81:627–635. 108. Rerkpattanapipat P, Wongpraparut N, Jacobs LE, Kotler MN. Cardiac manifestations of acquired immunodeficiency syndrome. Arch Intern Med 2000;160(5):602–608. 109. Panther LA. How HIV infection and its treatment affects the cardiovascular system: what is known, what is needed. Am J Physiol Heart Circ Physiol 2002;283(1):H1–H4. 110. Barbaro G, Di Lorenzo G, Grisorio B, Barbarini G. Incidence of dilated cardiomyopathy and detection of HIV in myocardial cells of HIV-positive patients. Gruppo Italiano per lo Studio Cardiologico dei Pazienti Affetti da AIDS. N Engl J Med 1998; 339(16):1093–1099. 111. Raidel SM, Haase C, Jansen NR, et al. Targeted myocardial transgenic expression of HIV Tat causes cardiomyopathy and mitochondrial damage. Am J Physiol Heart Circ Physiol 2002; 282(5):H1672–H1678. CAR055.indd 1256 55 112. Lewis W. AIDS cardiomyopathy: physiological, molecular, and biochemical studies in the transgenic mouse. Ann N Y Acad Sci 2001;946:46–56. 113. Herskowitz A, Wu T-C, Willoughby SB, et al. Myocarditis and cardiotropic viral infection associated with severe left ventricular dysfunction in late-stage infection with human immunodeficiency virus. J Am Coll Cardiol 1994;24:1025– 1032. 114. Kaul S, Fishbein MC, Siegel RJ. Cardiac manifestations of acquired immune deficiency syndrome: a 1991 update. Am Heart J 1991;122:535–544. 115. Rossi MA, Bestetti RB. The challenge of chagasic cardiomyopathy. The pathologic roles of autonomic abnormalities, autoimmune mechanisms and microvascular changes, and therapeutic implications. Cardiology 1995;86(1):1–7. 116. Carrasco HA, Parada H, Guerrero L, Duque M, Duran D, Molina C. Prognostic implications of clinical, electrocardiographic and hemodynamic fi ndings in chronic Chagas’ disease. Int J Cardiol 1994;43(1):27–38. 117. Kirchhoff LV. Current concepts: American trypanosomiasis (Chagas’ Disease)—a tropical disease now in the United States. N Engl J Med 1993;329:639–644. 118. Marr JJ, Docampo R. Chemotherapy for Chagas’ disease: a perspective on current therapy and considerations for future research. Rev Infect Dis 1986;8:884–903. 119. Roberti RR, Martinez EE, Andrade JL, et al. Chagas cardiomyopathy and captopril. Eur Heart J 1992;13(7):966–970. 120. Burke AP, Saenger J, Mullick F, Virmani R. Hypersensitivity myocarditis. Arch Pathol Lab Med 1991;115(8):764–769. 121. Paradiso M, Gabrielli F, Masala C, et al. Evaluation of myocardial involvement in systemic lupus erythematosus by signalaveraged electrocardiography and echocardiography. Acta Cardiol 2001;56(6):381–386. 122. Lee LA. Neonatal lupus: clinical features, therapy, and pathogenesis. Curr Rheumatol Rep 2001;3(5):391–395. 123. Kazzam E, Caidahl K, Hallgren R, Gustafsson R, Landelius J, Waldenstrom A. Non-invasive assessment of systolic left ventricular function in systemic sclerosis. Eur Heart J 1991;12: 151–156. 124. Bulkley BH, Ridolfi RL, Salyer WR, Hutchins GM. Myocardial lesions of progressive systemic sclerosis. A cause of cardiac dysfunction. Circulation 1976;53:483–490. 125. Goldenberg J, Ferraz MB, Pessoa AP, et al. Symptomatic cardiac involvement in juvenile rheumatoid arthritis. Int J Cardiol 1992;34:57–62. 126. Marin-Neto JA, Maciel BC, Urbanetz LL, Gallo JL, AlmeidaFilho OC, Amorim DS. High output failure in patients with peripartum cardiomyopathy: a comparative study with dilated cardiomyopathy. Am Heart J 1991;121(1 pt 1): 134–140. 127. Lampert MB, Lang RM. Peripartum cardiomyopathy. Am Heart J 1995;130(4):860–870. 128. Melvin KR, Richardson PJ, Olsen EG, Daly K, Jackson G. Peripartum cardiomyopathy due to myocarditis. N Engl J Med 1982;307(12):731–734. 129. Elkayam U, Tummala PP, Rao K, et al. Maternal and fetal outcomes of subsequent pregnancies in women with peripartum cardiomyopathy. N Engl J Med 2001;344(21):1567–1571. 130. O’Connell JB, Costanzo-Nordin MR, Subramanian R, et al. Peripartum cardiomyopathy: clinical, hemodynamic, histologic and prognostic characteristics. J Am Coll Cardiol 1986; 8(1):52–56. 131. Alpert MA. Obesity cardiomyopathy: pathophysiology and evolution of the clinical syndrome. Am J Med Sci 2001;321(4): 225–236. 132. Kenchaiah S, Evans JC, Levy D, et al. Obesity and the risk of heart failure. N Engl J Med 2002;347(5):305–313. 11/24/2006 1:28:36 PM di l at e d c a r diom yopat h y 133. Must A, Spadano J, Coakley EH, Field AE, Colditz G, Dietz WH. The disease burden associated with overweight and obesity. JAMA 1999;282(16):1523–1529. 134. Abate N. Obesity and cardiovascular disease. Pathogenetic role of the metabolic syndrome and therapeutic implications. J Diabetes Complications 2000;14(3):154–174. 135. Francis GS. Diabetic cardiomyopathy: fact or fiction? Heart 2001;85(3):247–248. 136. Devereux RB, Roman MJ, Paranicas M, et al. Impact of diabetes on cardiac structure and function: the strong heart study. Circulation 2000;101(19):2271–2276. 137. Klein I, Ojamaa K. Thyroid hormone and the cardiovascular system. N Engl J Med 2001;344(7):501–509. 138. Ladenson PW. Recognition and management of cardiovascular disease related to thyroid dysfunction. Am J Med 1990;88(6): 638–641. 139. Klein I, Ojamaa K. Clinical review 36: Cardiovascular manifestations of endocrine disease. J Clin Endocrinol Metab 1992; 75(2):339–342. 140. Lee RT, Plappert M, Sutton MG. Depressed left ventricular systolic ejection force in hypothyroidism. Am J Cardiol 1990;65(7):526–527. 141. Colao A, Marzullo P, Di Somma C, Lombardi G. Growth hormone and the heart. Clin Endocrinol (Oxf) 2001;54(2): 137–154. 142. Volterrani M, Giustina A, Manelli F, Cicoira MA, Lorusso R, Giordano A. Role of growth hormone in chronic heart failure: therapeutic implications. Ital Heart J 2000;1(11):732–738. 143. Osterziel KJ, Strohm O, Schuler J, et al. Randomised, doubleblind, placebo-controlled trial of human recombinant growth hormone in patients with chronic heart failure due to dilated cardiomyopathy. Lancet 1998;351(9111):1233–1237. 144. Singleton CK, Martin PR. Molecular mechanisms of thiamine utilization. Curr Mol Med 2001;1(2):197–207. 145. Djoenaidi W, Notermans SL, Dunda G. Beriberi cardiomyopathy. Eur J Clin Nutr 1992;46(3):227–234. 146. Sole MJ, Jeejeebhoy KN. Conditioned nutritional requirements: therapeutic relevance to heart failure. Herz 2002;27(2): 174–178. 147. Retter AS. Carnitine and its role in cardiovascular disease. Heart Dis 1999;1(2):108–113. 148. Vescovo G, Ravara B, Gobbo V, et al. L-Carnitine: a potential treatment for blocking apoptosis and preventing skeletal muscle myopathy in heart failure. Am J Physiol Cell Physiol 2002;283(3): C802–C810. 149. Razeghi P, Young ME, Ying J, et al. Downregulation of metabolic gene expression in failing human heart before and after mechanical unloading. Cardiology 2002;97(4):203–209. 150. Zarain-Herzberg A, Rupp H. Therapeutic potential of CPT I inhibitors: cardiac gene transcription as a target. Expert Opin Invest Drugs 2002;11(3):345–356. 151. Cheng TO. Selenium deficiency and cardiomyopathy. J R Soc Med 2002;95(4):219–220. 152. Manar MJ, MacPherson GD, Mcardle F, Jackson MJ, Hart CA. Selenium status, kwashiorkor and congestive heart failure. Acta Paediatr 2001;90(8):950–952. 153. Hetet G, Grandchamp B, Bouchier C, et al. Idiopathic dilated cardiomyopathy: lack of association with haemochromatosis gene in the CARDIGENE study. Heart 2001;86(6):702–703. 154. Gochee PA, Powell LW. What’s new in hemochromatosis. Curr Opin Hematol 2001;8(2):98–104. 155. Wang TL, Chen WJ, Liau CS, Lee YT. Sick sinus syndrome as the early manifestation of cardiac hemochromatosis. J Electrocardiol 1994;27(1):91–96. 156. Blankenberg F, Eisenberg S, Scheinman MN, Higgins CB. Use of cine gradient echo (GRE) MR in the imaging of cardiac hemochromatosis. J Comput Assist Tomogr 1994;18(1):136–138. CAR055.indd 1257 12 5 7 157. Lombardo T, Tamburino C, Bartoloni G, et al. Cardiac iron overload in thalassemic patients: an endomyocardial biopsy study. Ann Hematol 1995;71(3):135–141. 158. Mariotti E, Agostini A, Angelucci E, Lucarelli G, Sgarbi E. Echocardiographic study in ex-thalassemic patients with iron overload, preliminary observations during phlebotomy therapy. Bone Marrow Transplant 1993;12(suppl 1):106–107. 159. Spinale FG, Tomita M, Zellner JL, Cook JC, Crawford FA, Zile MR. Collagen remodeling and changes in LV function during development and recovery from supraventricular tachycardia. Am J Physiol 1991;261:H308–H318. 160. Mukherjee R, Crawford FA, Hewett KW, Spinale FG. Cell and sarcomere contractile performance from the same cardiocyte using video microscopy. J Appl Physiol 1993;74:2023– 2033. 161. Shinbane JS, Wood MA, Jensen DN, Ellenbogen KA, Fitzpatrick AP, Scheinman MM. Tachycardia-induced cardiomyopathy: a review of animal models and clinical studies. J Am Coll Cardiol 1997;29:709–715. 162. Schumacher B, Luderitz B. Rate issues in atrial fibrillation: consequences of tachycardia and therapy for rate control. Am J Cardiol 1998;82:29N–36N. 163. Luchsinger JA, Steinberg JS. Resolution of cardiomyopathy after ablation of atrial flutter. J Am Coll Cardiol 1998;32:205– 210. 164. Limas CJ, Goldenberg IF, Limas C. Autoantibodies against β-adrenoceptors in human idiopathic dilated cardiomyopathy. Circ Res 1989;64:97–103. 165. Caforio AL, Grazzini M, Mann JM, et al. Identification of alpha- and beta-cardiac myosin heavy chain isoforms as major autoantigens in dilated cardiomyopathy. Circulation 1992;85: 1734–1742. 166. Ansari AA, Wang YC, Danner DJ, et al. Abnormal expression of histocompatibility and mitochondrial antigens by cardiac tissue from patients with myocarditis and dilated cardiomyopathy. Am J Pathol 1991;139:337–354. 167. Mann DL. Autoimmunity, immunoglobulin adsorption and dilated cardiomyopathy: has the time come for randomized clinical trials? J Am Coll Cardiol 2001;38(1):184–186. 168. Carlquist JF, Menlove RL, Murray MB, O’Connell JB, Anderson JL. HLA class II (DR and DQ) antigen associations in idiopathic dilated cardiomyopathy. Circulation 1991;83:515–522. 169. Cowie MR, Struthers AD, Wood DA, et al. Value of natriuretic peptides in assessment of patients with possible new heart failure in primary care. Lancet 1997;350:1349–1353. 170. McDonagh TA, RObb SD, Murdoch DR, et al. Biochemical detection of left-ventricular systolic dysfunction. Lancet 1998; 351:9–13. 171. Davis M, Espiner E, Richards G, et al. Plasma brain natriuretic peptide in assessment of acute dyspnoea [see comments]. Lancet 1994;343:440–444. 172. Maisel A. B-type natriuretic peptide levels: diagnostic and prognostic in congestive heart failure: what’s next? Circulation 2002;105(20):2328–2331. 173. Maisel AS, Krishnaswamy P, Nowak RM, et al. Rapid measurement of B-type natriuretic peptide in the emergency diagnosis of heart failure. N Engl J Med 2002;347(3):161–167. 174. Richards AM, Doughty R, Nicholls MG, et al. Neurohumoral prediction of benefit from carvedilol in ischemic left ventricular dysfunction. Australia-New Zealand Heart Failure Group. Circulation 1999;99(6):786–792. 175. Troughton RW, Frampton CM, Yandle TG, Espiner EA, Nicholls MG, Richards AM. Treatment of heart failure guided by plasma aminoterminal brain natriuretic peptide (N-BNP) concentrations. Lancet 2000;355(9210):1126–1130. 176. Mizuno Y, Yoshimura M, Harada E, et al. Plasma levels of Aand B-type natriuretic peptides in patients with hypertrophic 11/24/2006 1:28:36 PM 12 5 8 chapter cardiomyopathy or idiopathic dilated cardiomyopathy. Am J Cardiol 2000;86(9):1036–40, A11. 177. Wilensky RL, Yudelman P, Cohen AI, et al. Serial electrocardiographic changes in idiopathic dilated cardiomyopathy confi rmed at necropsy. Am J Cardiol 1988;62:276–283. 178. Meinertz T, Hofmann T, Kasper W, et al. Significance of ventricular arrhythmias in idiopathic dilated cardiomyopathy. Am J Cardiol 1984;53:902–907. 179. Mestroni L, Miani D, Neri R, Di Lenarda A, Camerini F. Ambulatory ECG in cardiomyopathies. G Ital Cardiol 1987;17: 1139–1144. 180. Kron J, Hart M, Schual-Berke S, Niles NR, Hosenpud JD, McAnulty JH. Idiopathic dilated cardiomyopathy. Role of programmed electrical stimulation and Holter monitoring in predicting those at risk of sudden death. Chest 1988;93:85–90. 181. Olshausen KV, Stienen U, Schwarz F, Kubler W, Meyer J. Longterm prognostic significance of ventricular arrhythmias in idiopathic dilated cardiomyopathy. Am J Cardiol 1988;61: 146–151. 182. Ikegawa T, Chino M, Hasegawa H, et al. Prognostic significance of 24–hour ambulatory electrocardiographic monitoring in patients with dilative cardiomyopathy: a prospective study. Clin Cardiol 1987;10:78–82. 183. Mancini DM, Wong KL, Simson MB. Prognostic value of an abnormal signal-averaged electrocardiogram in patients with nonischemic congestive cardiomyopathy [see comments]. Circulation 1993;87(4):1083–1092. 184. Schmitt C, Barthel P, Ndrepepa G, et al. Value of programmed ventricular stimulation for prophylactic internal cardioverterdefibrillator implantation in postinfarction patients preselected by noninvasive risk stratifiers. J Am Coll Cardiol 2001;37(7): 1901–1907. 185. Bigger JT Jr. Prophylactic use of implanted cardiac defibrillators in patients at high risk for ventricular arrhythmias after coronary-artery bypass graft surgery. Coronary Artery Bypass Graft (CABG) Patch Trial Investigators. N Engl J Med 1997;337(22): 1569–1575. 186. Keeling PJ, Kulakowski P, Yi G, Slade AK, Bent SE, McKenna WJ. Usefulness of signal-averaged electrocardiogram in idiopathic dilated cardiomyopathy for identifying patients with ventricular arrhythmias. Am J Cardiol 1993;72:78–84. 187. Middlekauff HR, Stevenson WG, Woo MA, Moser DK, Stevenson LW. Comparison of frequency of late potentials in idiopathic dilated cardiomyopathy and ischemic cardiomyopathy with advanced congestive heart failure and their usefulness in predicting sudden death. Am J Cardiol 1990;66:1113–1117. 188. Meinertz T, Treese N, Kasper W, et al. Determinants of prognosis in idiopathic dilated cardiomyopathy as determined by programmed electrical stimulation. Am J Cardiol 1985;56: 337–341. 189. Wallis DE, O’Connell JB, Henkin RE, Costanzo-Nordin MR, Scanlon PJ. Segmental wall motion abnormalities in dilated cardiomyopathy. J Am Coll Cardiol 1989;13:311–315. 190. Takamoto T, Kim D, Urie PM, et al. Comparative recognition of left ventricular thrombi by echocardiography and cineangiography. Br Heart J 1985;53:36–42. 191. Kono T, Sabbah HN, Rosman H, Alam M, Stein PD, Goldstein S. Left atrial contribution to ventricular fi lling during the course of evolving heart failure. Circulation 1992;86:1317–1322. 192. Nagueh SF, Kopelen HA, Zoghbi WA. Feasibility and accuracy of Doppler echocardiographic estimation of pulmonary artery occlusive pressure in the intensive care unit. Am J Cardiol 1995;75:1256–1262. 193. Pohost GM, Fallon JT, Strauss HW. The role of radionuclide techniques in patients with myocardial disease. Cardiovasc Clin 1979;10:149–163. CAR055.indd 1258 55 194. Glamann DB, Lange RA, Corbett JR, Hillis LD. Utility of various radionuclide techniques for distinguishing ischemic from nonischemic dilated cardiomyopathy. Arch Intern Med 1992;152:769–772. 195. O’Connell JB, Henkin RE, Robinson JA, Subramanian R, Scanlon PJ, Gunnar RM. Gallium-67 imaging in patients with dilated cardiomyopathy and biopsy-proven myocarditis. Circulation 1984;70:58–62. 196. Dec GW, Palacios I, Yasuda T, et al. Antimyosin antibody cardiac imaging: its role in the diagnosis of myocarditis. J Am Coll Cardiol 1990;16:97–104. 197. Stevenson LW. Heart transplant centers: no longer the end of the road for heart failure. J Am Coll Cardiol 1996;27:1198– 1200. 198. Quigley PJ, Richardson PJ, Meany BT. Long term follow up of acute myocarditis. Correlation of ventricular function and outcome. Eur Heart J Suppl J 1987;8:39–42. 199. Mason JW. Clinical merit of endomyocardial biopsy. Circulation 1989;79:971–979. 200. Cooper LT Jr, Berry GJ, Shabetai R. Idiopathic giant-cell myocarditis—natural history and treatment. N Engl J Med 1997; 336:1860–1866. 201. 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(23):1429–1435. 202. Packer M, Coats AJ, Fowler MB, et al. Effect of carvedilol on survival in severe chronic heart failure. N Engl J Med 2001; 344(22):1651–1658. 203. 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(21):1349–1355. 204. Adams KF Jr, Gheorghiade M, Uretsky BF, et al. Patients with mild heart failure worsen during withdrawal from digoxin therapy. J Am Coll Cardiol 1997;30(1):42–48. 205. 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(10):709–717. 206. McMurray JJ, Ostergren J, Swedberg K, et al. 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(9386):767–771. 207. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomised trial–the Losartan Heart Failure Survival Study ELITE II. Lancet 2000;355(9215):1582– 1587. 208. Packer M, Kessler PD, Lee WH. Calcium-channel blockade in the management of severe chronic congestive failure: a bridge too far. Circulation 1987;75(suppl V):V-56–V-64. 209. Cohn JN, Ziesche S, Smith R, et al. Effect of the calcium antagonist felodipine as supplementary vasodilator therapy in patients with chronic heart failure treated with enalapril: VHeFT III. Vasodilator-Heart Failure Trial (V-HeFT) Study Group. Circulation 1997;96(3):856–863. 210. Taylor AL, Ziesche S, Yancy C, et al. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med 2004;351(20):2049–2057. 211. A comparison of antiarrhythmic-drug therapy with implantable defibrillators in patients resuscitated from near-fatal ventricular arrhythmias. The Antiarrhythmics versus Implantable Defibrillators (AVID) Investigators. N Engl J Med 1997;337(22): 1576–1583. 11/24/2006 1:28:36 PM di l at e d c a r diom yopat h y 212. Moss AJ, Zareba W, Hall WJ, et al. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. N Engl J Med 2002;346(12):877– 883. 213. Buxton AE, Lee KL, Fisher JD, Josephson ME, Prystowsky EN, Hafley G. A randomized study of the prevention of sudden death in patients with coronary artery disease. Multicenter Unsustained Tachycardia Trial Investigators. N Engl J Med 1999;341(25):1882–1890. 214. Strickberger SA, Hummel JD, Bartlett TG, et al. Amiodarone versus implantable cardioverter-defibrillator: randomized trial in patients with nonischemic dilated cardiomyopathy and asymptomatic nonsustained ventricular tachycardia–AMIOVIRT. J Am Coll Cardiol 2003;41(10):1707–1712. 215. Kadish A, Dyer A, Daubert JP, et al. Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy. N Engl J Med 2004;350(21):2151–2158. 216. Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med 2005;352(3):225–237. 217. McClellan MB, Tunis SR. Medicare coverage of ICDs. N Engl J Med 2005;352(3):222–224. 218. Doval HC, Nul DR, Grancelli HO, Perrone SV, Bortman GR, Curiel R. Randomised trial of low-dose amiodarone in severe congestive heart failure. Grupo de Estudio de la Sobrevida en la Insuficiencia Cardiaca en Argentina (GESICA). Lancet 1994;344(8921):493–498. 219. Abraham WT, Fisher WG, Smith AL, et al. Cardiac resynchronization in chronic heart failure. N Engl J Med 2002;346(24): 1845–1853. 220. Linde C, Leclercq C, Rex S, et al. Long-term benefits of biventricular pacing in congestive heart failure: results from the MUltisite STimulation in cardiomyopathy (MUSTIC) study. J Am Coll Cardiol 2002;40(1):111–118. 221. Bristow MR, Saxon LA, Boehmer J, et al. Cardiac-resynchronization therapy with or without an implantable defibrillator in advanced chronic heart failure. N Engl J Med 2004;350(21): 2140–2150. 222. Cleland JG, Daubert JC, Erdmann E, et al. The effect of cardiac resynchronization on morbidity and mortality in heart failure. N Engl J Med 2005;352(15):1539–1549. 223. Koniaris LS, Goldhaber SZ. Anticoagulation in dilated cardiomyopathy. J Am Coll Cardiol 1998;31(4):745–748. 224. Massie BM, Krol WF, Ammon SE, et al. The Warfarin and Antiplatelet Therapy in Heart Failure trial (WATCH): rationale, design, and baseline patient characteristics. J Card Fail 2004; 10(2):101–112. CAR055.indd 1259 12 5 9 225. Mason JW, O’Connel JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis. N Engl J Med 1995;333:269–275. 226. Gullestad L, Aass H, Fjeld JG, et al. Immunomodulating therapy with intravenous immunoglobulin in patients with chronic heart failure. Circulation 2001;103(2):220–225. 227. McCarthy RE III, Boehmer JP, Hruban RH, et al. Long-term outcome of fulminant myocarditis as compared with acute (nonfulminant) myocarditis. N Engl J Med 2000;342(10): 690–695. 228. McNamara DM, Holubkov R, Starling RC, et al. Controlled trial of intravenous immune globulin in recent-onset dilated cardiomyopathy. Circulation 2001;103(18):2254–2259. 229. Bozkurt B, Villaneuva FS, Holubkov R, et al. Intravenous immune globulin in the therapy of peripartum cardiomyopathy. J Am Coll Cardiol 1999;34(1):177–180. 230. McNamara DM, Starling RC, Dec GW. Intervention in myocarditis and acute cardiomyopathy with immune globulin: results from the randomized placebo controlled IMAC trial. Circulation 1999;100(suppl I):I-21(abstr). 231. Wallukat G, Reinke P, Dorffel WV, et al. Removal of autoantibodies in dilated cardiomyopathy by immunoadsorption. Int J Cardiol 1996;54(2):191–195. 232. Dorffel WV, Wallukat G, Baumann G, Felix SB. Immunoadsorption in dilated cardiomyopathy. Ther Apher 2000;4(3): 235–238. 233. Felix SB, Staudt A, Dorffel WV, et al. Hemodynamic effects of immunoadsorption and subsequent immunoglobulin substitution in dilated cardiomyopathy: three-month results from a randomized study. J Am Coll Cardiol 2000;35(6):1590–1598. 234. Muller J, Wallukat G, Dandel M, et al. Immunoglobulin adsorption in patients with idiopathic dilated cardiomyopathy. Circulation 2000;101(4):385–391. 235. Fazio S, Sabatini D, Capaldo B, et al. A preliminary study of growth hormone in the treatment of dilated cardiomyopathy. N Engl J Med 1996;334:809–814. 236. Pinamonti B, Zecchin M, Di Lenarda A, Gregori D, Sinagra G, Camerini F. Persistence of restrictive left ventricular filling pattern in dilated cardiomyopathy: an ominous prognostic sign. J Am Coll Cardiol 1997;29:604–612. 237. Isner JM. Myocardial gene therapy. Nature 2002;415(6868): 234–239. 238. Dubowitz V. Special Centennial Workshop—101st ENMC International Workshop: Therapeutic Possibilities in Duchenne Muscular Dystrophy, 30th November–2nd December 2001, Naarden, The Netherlands. Neuromuscul Disord 2002; 12(4):421–431. 11/24/2006 1:28:37 PM CAR055.indd 1260 11/24/2006 1:28:37 PM