Heart Failure as a Consequence of Viral and Nonviral Myocarditis


In the attempt to diagnose heart disease more accurately, the term myocarditis is wisely being abandoned in large part; we must remember, nevertheless, that there does exist such a condition as myocarditis . . . . Paul Dudley White

Dr. White’s concerns about the “abandonment” of the term myocarditis were unfounded, and over half a century after his initial observations, myocarditis remains an important pathologic term defining of diverse set of cardiac disorders involving primary myocardial inflammation. For his readership in the 1950s, the primary infectious causes of myocardial inflammation were rheumatic fever and diphtheroids, for which effective antibiotic therapies were later developed. In the present day, an ever-changing group of viral etiologies has proven much more difficult to eradicate. Acute myocardial inflammation plays a critical role in viral clearing, but in chronic pathologic states inflammation may play a role in the pathogenesis of nonischemic cardiomyopathy. While immune suppression plays a therapeutic role in specific subsets of myocarditis, for the majority of cases there is no proven benefit.

Although infectious causes remain important, the category of myocarditis includes a diverse set of disorders for which there is no discernible infectious cause, from transient myocardial dysfunction with allergic eosinophilic myocarditis to the progressive myocyte destruction that characterizes giant cell myocarditis (GCM). This chapter will discuss the pathogenesis of heart failure from viral and nonviral forms of myocarditis, the current practice of supportive heart failure treatment, the hope for future targeted therapeutics, and in the case of fulminant myocarditis, the role of mechanical therapy as a bridge to recovery. Despite decades of clinical and basic investigation since Paul Dudley White’s initial observations, the diagnosis and treatment of myocarditis remain extremely challenging.

History

The French pathologist Corvisart described “carditis” in 1806 as an important clinical syndrome, which most commonly was acute and fatal, but could develop into more “chronic organic disease.” The term carditis was later refined to myocarditis, first introduced by Sobernheim in 1837, to describe the myocardial inflammation presumed to be the cause of most nonvalvular cardiac dysfunction. By the latter part of the nineteenth century it was increasingly recognized that primary myocardial inflammation was responsible for only a small subset of nonvalvular cardiac dysfunction, because coronary disease and hypertensive heart disease were far more common causes. In contrast to the previous nondiscriminant use of the term, attempts at more accurate cardiac diagnosis of myocarditis in the twentieth century led to a marked diminishment in its recognition.

The emergence of endomyocardial biopsy (EMB) in the 1960s as a diagnostic tool allowed clinicians to delineate cellular inflammation of the myocardium, and fueled the hope that this heterogeneous group of disorders could now be classified into histopathologic subsets with distinct therapies and outcomes. The Dallas criteria were developed in 1986 by leading cardiac pathologists to standardize the histologic definition of lymphocytic myocarditis (LM), the most commonly observed form of cellular inflammation. They defined borderline myocarditis as mononuclear cell infiltrates without myocyte necrosis ( Fig. 28.1 ) and myocarditis as cellular infiltration with myocyte necrosis ( Fig. 28.2 ). Despite the wide acceptance of these histologic criteria for the pathologic assessment of myocarditis, significant variation in interpretation remained in the practical application. A decade later, the World Health Organization task force defined inflammatory cardiomyopathy as “myocarditis in association with myocardial dysfunction. Myocarditis is an inflammatory disease of the myocardium and is diagnosed by established histologic, immunologic, and immunohistochemical criteria.” This task force expanded the definition by adding both quantitative and immunohistochemical criteria for detection of cellular infiltrates and expression of human leukocyte antigen (HLA) class II molecules.

Fig. 28.1, Histopathologic appearance of borderline myocarditis by Dallas criteria (lymphocytic infiltrates without myocyte necrosis) with routine staining with hematoxylin and eosin (H&E) under (A) low power (100×) and (B) high power (350×).

Fig. 28.2, Histopathologic appearance of myocarditis by Dallas criteria (lymphocytic infiltrates with myocyte necrosis) with routine staining with hematoxylin and eosin under (A) low power (100×) and (B) high power (350×).

The observed histologic similarity of LM to cardiac allograft rejection led to the hypothesis that a therapeutic strategy of immunosuppression would improve clinical outcomes. When the Myocarditis Treatment Trial (MTT) demonstrated no benefit, the absence of a histologically guided therapy led to a diminished role for EMB, and native cardiac biopsy is not commonly performed in the United States. Efforts continue to improve the diagnostic utility of EMB with the addition of molecular diagnostics to detect viral nucleic acids and routine immunohistochemistry to improve the specificity, but this is only performed at selected centers. The histologic and clinical diversity of myocarditis remains a challenge in terms of the development of targeted therapeutics.

Viral Etiologies

The most common infectious agents initiating myocarditis in North America are viral pathogens ( Table 28.1 ), although the dominant viral species continues to evolve over time. Enteroviruses were first described in clinical and serologic studies decades ago and remain a major cause of myocarditis in infants and children. In adults, adenovirus, influenza A and B, and herpesviruses also have been implicated as important viral pathogens, whereas hepatitis C has been implicated in Asia, but less commonly reported in case series in the United States. Over the past decade, influenza and erythroviruses, such as parvovirus B19, emerged as two important pathogens associated with myocarditis. The changing viral milieu represents a considerable challenge for developing therapeutic efforts targeting specific viral pathogens. Effective viral therapies can diminish viral pathogenesis. For example, human immunodeficiency virus (HIV) infection has been associated with myocarditis and dilated cardiomyopathy (DCM) ; however, the incidence of HIV-associated cardiomyopathy has been diminished with more aggressive antiviral therapies against HIV.

TABLE 28.1
Viral Causes of Inflammatory Cardiomyopathy and Myocarditis
Parvovirus (Parvovirus B19)
Adenoviruses
Influenza A and B
Enteroviruses (coxsackie A and B)
Herpesvirus (human herpesvirus-6)
Varicella-zoster
Cytomegalovirus
Epstein-Barr virus
Hepatitis B and C virus
Human immunodeficiency virus
Poliovirus
Variola virus (smallpox)
Rubella virus
Echovirus
Polio

In the developing world, bacterial pathogens remain important causes as myocarditis as a complication of rheumatic fever and diphtheroids is much more prevalent. In Central and South America, the most common infectious agent is the protozoa Trypanosoma cruzi, the causative agent for Chagas’ disease, which is endemic in certain areas and may affect 15 to 20 million people. This disorder is not seen in North America without a travel history to endemic areas. In immunocompromised hosts, other pathogens, such as toxoplasmosis and aspergillus, can cause myocardial inflammation but generally in the setting of a systemic infection and not as isolated myocarditis.

Autoimmune (Nonviral) Etiologies

There are several autoimmune forms of myocardial inflammation for which no infectious agent can be identified. For certain diagnoses, histopathology combined with the clinical setting can point toward targeted treatments. Multinucleated giant cells in the myocardium in the setting of fulminant myocarditis suggests the diagnosis of GCM ( Fig. 28.3 ), a progressive and destructive form of myocarditis with a high mortality rate on conventional therapy but responsive to immunosuppressive therapy. Finding similar multinucleated granulomas in the myocardium, but with few other inflammatory cells in a more compensated patient, may suggest systemic sarcoidosis, a disorder responsive to treatment with corticosteroids.

Fig. 28.3, Histopathologic appearance of giant cell myocarditis (multinucleated giant cell) with routine staining with hematoxylin and eosin under (A) low power (100×) and (B) high power (350×).

Eosinophilic predominance of inflammatory cells in the myocardium with new myocardial dysfunction may suggest an allergic hypersensitivity myocarditis. Several pharmacologic agents, including tricyclic antidepressants, antipsychotics, and cephalosporin, have been implicated as triggering eosinophilic myocarditis. This disorder is generally self-limited, and the myocardium will recover with removal of the triggering agent. For persistent myocarditis in the setting of peripheral eosinophilia, a short course of corticosteroids can be considered. Chronic peripheral eosinophilia without a clear initiating agent should merit consideration of a systemic syndrome such as Churg–Strauss vasculitis or hypereosinophilic syndrome (HES), for which immunosuppressive therapy is generally indicated.

Myocarditis may also be a clinical feature of several systemic autoimmune disorders. Systemic lupus erythematosus (SLE), dermatopolymyositis, and rheumatoid arthritis also have been associated with inflammatory myocarditis. In general, immunosuppressive therapy is directed toward the systemic autoimmune disorder and will result in the recovery of myocardial function as well. EMB is rarely performed, except in cases where the systemic disorder is quiescent and the finding of myocardial inflammation may be the only indication for immunosuppressive therapy.

Pathogenesis in Murine Models

Much of what is known of the basic pathogenesis of viral myocarditis is derived from murine models of inoculation with the cardiotropic coxsackie group B virus or encephalomyocarditis virus into susceptible strains. The viral particles are taken up by myocytes via receptor-mediated endocytosis. Translation of viral proteins and replication of viral particles can result in cell lysis within 3 days ( Fig. 28.4 ) before the initiation of myocardial inflammation. Macrophage activation results in cytokine expression, including interferon-γ and the activation of natural killer (NK) cells, both of which limit viral replication. Infiltrates of antigen-specific T lymphocytes including T-helper (CD4+) cells and cytotoxic T lymphocyte (CD8+) cells are seen within 7 days of infection. Recognition of viral peptides by cytotoxic T cells results in cellular toxicity of virally infected cells. Up to 20% of infiltrating lymphocytes are B cells, and neutralizing antibodies are important in viral clearing although not in cytotoxicity. Viral particles are no longer evident within 15 days of the initial inoculation, although inflammatory infiltrates may persist for 90 days. Viral nucleic acid can be detected after 90 days in only a small percentage of virally infected mice; however, long-term ventricular dilation and remodeling may develop and are associated with myocardial fibrosis. Myocardial inflammation followed by ventricular dilation and fibrosis occurs in transgenic mice overexpressing TNF-α, and in murine models using antimyosin antibodies, demonstrating the role of chronic inflammation in the pathogenesis of nonischemic DCM.

Fig. 28.4, Timeline of progression in murine models from initial viral inoculation to acute myocarditis to subacute myocarditis to chronic myocarditis.

Clinical Presentation

Acute myocarditis will typically present with a variety of symptoms, including dyspnea, chest pain, palpitations, syncope, and near syncope, and when associated with left ventricular (LV) dysfunction, most commonly presents with signs of heart failure. A presentation with dyspnea or chest pain associated with electrocardiogram abnormalities or an elevation of cardiac enzymes may be the first sign of a possible myocarditis or myopericarditis. The cardiac examination may be unremarkable or may reveal a pericardial rub. Tachycardia, relative hypotension, jugular venous distention, and peripheral edema may suggest more significant hemodynamic compromise. Chest examination may reveal congestion, and decreased breath sounds are suggestive of pleural effusions.

Electrocardiographic findings are generally diffuse and nonspecific. PR depression and global ST elevation may be seen in cases with an associated pericarditis. Low voltage may be evident, particularly in subjects with an associated pericardial effusion. Heart block, ventricular tachycardia, or a new bundle branch block are all suggestive of a more fulminant disorder. Evaluation of biomarkers may reveal an elevation in cardiac troponin and brain natriuretic peptide. More fulminant myocarditis may be associated with elevated liver function tests and a compromise of renal function. Chest x-ray may be unremarkable or show evidence of heart failure and cardiomegaly.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here