The management of myocarditis - European Heart Journal

13
REVIEW Frontiers in cardiovascular medicine The management of myocarditis Heinz-Peter Schultheiss 1,2 * , Uwe Ku ¨hl 1,2 , and Leslie T. Cooper 1,2 1 Charite Hospital, Berlin, Germany; and 2 Mayo Clinic, Rochester, MN, USA Received 2 January 2011; revised 25 March 2011; accepted 28 April 2011; online publish-ahead-of-print 23 June 2011 Despite considerable advances in our understanding of myocarditis pathogenesis, the clinical management of myocarditis has changed relatively little in the last few years. This review aims to help bridge the widening gap between recent mechanistic insights, which are largely derived from animal models, and their potential impact on disease burden. We illustrate the pathogenenic mechanisms that are prime targets for novel therapeutic interventions. Pathway and pathogen-specific molecular diagnostic tests have expanded the role for endomyocardial biopsy. State of the art cardiac magnetic resonance imaging can now provide non-invasive tissue characterization and localize inflammatory infiltrates but imaging techniques are misleading if infectious agents are involved. We emphasize the gaps in our current clinical knowledge, particularly with respect to aetiology-based therapy, and suggest opportunities for high impact, translational investigations. ----------------------------------------------------------------------------------------------------------------------------------------------------------- Keywords Myocarditis pathogenesis Clinical management Aetiology of myocarditis Aetiologies of myocarditis include a number of infectious and non- infectious agents such as viruses, bacteria, protozoa, fungi, toxins, myocardial involvement in systemic diseases, or physical condition, but often the underlying cause cannot be identified (Table 1). Drugs can induce myocardial inflammation by either direct toxic effects on heart tissue or by inducing hypersensitivity reactions, which are often associated with an eosinophilic myocarditis. 1 Eosi- nophils are also observed in myocardial inflammatory processes which are associated with Churg–Strauss or hypereosinophilic syndromes, vaccination against smallpox or caused by helminthic and parasitic infections. Myocardial involvement may be caused by granulomatous and systemic diseases or (auto)immune pro- cesses with often unknown pathogenetic mechanisms, but all these aetiologies are far less common than virus-induced myocar- ditis or post-infectious inflammatory cardiomyopathy. 2 4 Apart from enteroviruses, which traditionally have been considered the most common agent in myocarditis and dilated cardiomyopathy, distinct RNA- and DNA viruses and virus subtypes have been identified with varying degrees of frequency (Table 1). 5 22 Although viral infections can cause serious human diseases, the majority of viral infections are asymptomatic or oligosymptomatic and therefore, such infections are frequently not recognized as possible causes of delayed onset of heart disease. 23 In the past, viral myocarditis and chronic viral heart disease have therefore more often been a clinically derived diagnosis of exclusion, rather than a specifically proven diagnosis. Beside the temporary changes of virus epidemics and the geographical differences in the aetiological profiles of viruses, the presence of myocarditic and non-myocarditic virus variants, the enormous variability of clinical symptoms of viral heart disease, which may range from asymptomatic presentation to manifest heart failure, and last but not least the lack of consequent diagnostic efforts for complete virus analysis have hampered an early identification of afflicted patients and thus have prevented the generation of valid epidemiological data. Pathogenic mechanisms For many viruses, the exact cardiac infection site and the under- lying pathogenic mechanisms are unknown. Most information on the pathophysiology of viral heart disease and post-infectious auto- immune myocarditis in both rodent models and humans is known from enteroviral infections such as coxsackievirus B3. Entero- viruses enter the host through the gastrointestinal or respiratory tract, reside in the reticuloendothelial system as an extracardiac reservoir, and attack heart tissue as a secondary target organ. After enterovirus internalization, the negative strand RNA is reversely transcribed into a positive strand for subsequent virus replication. 22,24 A direct virus-related cytolysis of cardiomyocytes * Corresponding author. Tel: +49 308 445 2344; Fax: +49 308 445 3565; Email: [email protected] Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: [email protected] European Heart Journal (2011) 32, 2616–2625 doi:10.1093/eurheartj/ehr165 Downloaded from https://academic.oup.com/eurheartj/article/32/21/2616/439730 by guest on 18 December 2021

Transcript of The management of myocarditis - European Heart Journal

REVIEW

Frontiers in cardiovascular medicine

The management of myocarditisHeinz-Peter Schultheiss 1,2*, Uwe Kuhl1,2, and Leslie T. Cooper1,2

1Charite Hospital, Berlin, Germany; and 2Mayo Clinic, Rochester, MN, USA

Received 2 January 2011; revised 25 March 2011; accepted 28 April 2011; online publish-ahead-of-print 23 June 2011

Despite considerable advances in our understanding of myocarditis pathogenesis, the clinical management of myocarditis has changedrelatively little in the last few years. This review aims to help bridge the widening gap between recent mechanistic insights, which arelargely derived from animal models, and their potential impact on disease burden. We illustrate the pathogenenic mechanisms that areprime targets for novel therapeutic interventions. Pathway and pathogen-specific molecular diagnostic tests have expanded the role forendomyocardial biopsy. State of the art cardiac magnetic resonance imaging can now provide non-invasive tissue characterization and localizeinflammatory infiltrates but imaging techniques are misleading if infectious agents are involved. We emphasize the gaps in our current clinicalknowledge, particularly with respect to aetiology-based therapy, and suggest opportunities for high impact, translational investigations.- - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -Keywords Myocarditis pathogenesis † Clinical management

Aetiology of myocarditisAetiologies of myocarditis include a number of infectious and non-infectious agents such as viruses, bacteria, protozoa, fungi, toxins,myocardial involvement in systemic diseases, or physical condition,but often the underlying cause cannot be identified (Table 1).Drugs can induce myocardial inflammation by either direct toxiceffects on heart tissue or by inducing hypersensitivity reactions,which are often associated with an eosinophilic myocarditis.1 Eosi-nophils are also observed in myocardial inflammatory processeswhich are associated with Churg–Strauss or hypereosinophilicsyndromes, vaccination against smallpox or caused by helminthicand parasitic infections. Myocardial involvement may be causedby granulomatous and systemic diseases or (auto)immune pro-cesses with often unknown pathogenetic mechanisms, but allthese aetiologies are far less common than virus-induced myocar-ditis or post-infectious inflammatory cardiomyopathy.2– 4 Apartfrom enteroviruses, which traditionally have been considered themost common agent in myocarditis and dilated cardiomyopathy,distinct RNA- and DNA viruses and virus subtypes have beenidentified with varying degrees of frequency (Table 1).5– 22

Although viral infections can cause serious human diseases, themajority of viral infections are asymptomatic or oligosymptomaticand therefore, such infections are frequently not recognized aspossible causes of delayed onset of heart disease.23 In the past,viral myocarditis and chronic viral heart disease have therefore

more often been a clinically derived diagnosis of exclusion,rather than a specifically proven diagnosis. Beside the temporarychanges of virus epidemics and the geographical differences inthe aetiological profiles of viruses, the presence of myocarditicand non-myocarditic virus variants, the enormous variability ofclinical symptoms of viral heart disease, which may range fromasymptomatic presentation to manifest heart failure, and last butnot least the lack of consequent diagnostic efforts for completevirus analysis have hampered an early identification of afflictedpatients and thus have prevented the generation of validepidemiological data.

Pathogenic mechanismsFor many viruses, the exact cardiac infection site and the under-lying pathogenic mechanisms are unknown. Most information onthe pathophysiology of viral heart disease and post-infectious auto-immune myocarditis in both rodent models and humans is knownfrom enteroviral infections such as coxsackievirus B3. Entero-viruses enter the host through the gastrointestinal or respiratorytract, reside in the reticuloendothelial system as an extracardiacreservoir, and attack heart tissue as a secondary target organ.After enterovirus internalization, the negative strand RNA isreversely transcribed into a positive strand for subsequent virusreplication.22,24 A direct virus-related cytolysis of cardiomyocytes

* Corresponding author. Tel: +49 308 445 2344; Fax: +49 308 445 3565; Email: [email protected]

Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: [email protected]

European Heart Journal (2011) 32, 2616–2625doi:10.1093/eurheartj/ehr165

Dow

nloaded from https://academ

ic.oup.com/eurheartj/article/32/21/2616/439730 by guest on 18 D

ecember 2021

is already detected before any inflammatory infiltrate develops andappears to be decisive in fulminant cases of myocarditis.25,26

In contrast to enteroviruses, which primarily infect and injurecardiomyocytes, other frequent cardiotropic viruses such as ery-throviruses or human herpesvirus 6 A/B infect the vascular endo-thelial cells (ECs).27– 30 Following primary infection in childhood,erythroviruses, including its genotype 1 (parvovirus B19, B19V),may reside asymptomatically in the bone marrow of a vast majorityof the adult population. The in vivo tropism of B19V infection isregulated by a number of determinants and persistent infectionand replication is mainly restricted to erythroid progenitor cells,but also EC, by the distribution of the primary erythroviral recep-tor, the P antigen, and reported co-receptors-like integrin a5b1and the KU80 protein.31–33 With respect to the heart tissue, ery-throviruses do not infect cardiomyocytes. In biopsy samples ofpatients with fulminant myocarditis or sudden onset heart failure,B19V genomes have been localized in EC of venuoles, smallarteries, or arterioles.27 In chronic inflammatory cardiomyopathy,B19V infection is predominantly detected in ECs of smallcapillaries.28,29

Endothelial cell infection is associated with endothelial dysfunc-tion which predicts long-term disease progression in chronic heartfailure.34 –39 The pathogenetic mechanisms by which B19 exertsendothelial damage are complex and may involve cytotoxicity ofthe non-structural protein 1, transactivation of interleukin-6(IL-6), and tumour necrosis factor a, as well as induction of apop-tosis as shown in vitro.40 –45. Recently, we could show that B19V isreleased from the bone marrow by infected capillary precursorcells and that interferon-b (IFN-b) improves viability of B19Vinfected human ECs.46 Since endothelial dysfunction and respectivesymptoms improved upon IFN-b treatment while both remained

unchanged in non-treated patients providing evidence thatB19-induced damage of ECs is at least partially mediated viadirect virus–cell interaction.46

HHV-6 is a lymphotropic virus with tropism mainly for CD4+and CD8+ T cells, B cells, and natural killer cells. AlthoughHHV-6 is thought of as a lymphotropic virus, it also can infectvascular endothelium.47,48 Several studies have identifiedHHV-6-specific DNA in the vascular endothelium in vivo, andsuggested EC damage by the virus.48 –51 It has been suggestedthat ECs and cardiac myocytes might be an important reservoirfor viral latency and reactivation.47

Similar to the other herpes viruses, HHV-6 becomes frequentlyreactivated by infections or drugs with subacute clinical presenta-tions, especially in acquired or drug-induced immunodeficiencies(e.g. transplant recipients) or in patients with autoimmune dis-orders. It has been suggested that often HHV-6 rather enhancesthe pathogenicity of other viruses than being a pathogen itself. Ina minority of patients (,1%), HHV-6 is integrated into thechromosome of all nucleated cells resulting in lifelong persistenceof HHV-6 which is then passed to 50% of the progenies.

Since distinct cardiac target structures are affected by differentinfectious agents and post-infectious mechanisms, viral myocarditis(viral heart disease) and post-infectious immune myocarditispresent with heterogenic clinical phenotypes. If the contractileapparatus (cardiomyocytes) or interstitial cells and matrixstructures (e.g. dystrophin) are preferentially involved, ventriculardilatation and systolic heart failure with progression towards post-myocarditic dilated cardiomyopathy (DCM) is directly causedby cytolytic viruses or viral proteases.52 Involvement ofvascular tissue (ECs) by other viruses may affect myocardialcontractility indirectly, e.g. as a consequence of endothelial

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Table 1 Causes of myocarditis

Infectious causes Non-infectious causes

RNA viruses: picornaviruses (coxsackie A + B, echovirus, poliovirus,hepatitis virus), orthomyxovirus (influenza), paramyxoviruses(respiratory syncitial virus, mumps), togaviruses (rubella), flaviviruses(dengue fever, yellow fever)

Autoimmune diseases: dematomyositis, inflammatory bowel disease,rheumatoid arthritis, sjogren syndrome, systemic lupus erythematodes,Wegener’s granulomatosis, giant cell myocarditis

DNA viruses: adenovirus (A 1, 2 ,3, and 5), erythrovirus [1 (B19V) and 2],herpesviruses (human herpes virus 6 A/B, cytomegalievirus, Epstein-Barrvirus, varicella-zoster virus), retrovirus (HIV)

Drugs: aminophyllin, amphetamine, anthracyclin, catecholamines,chloramphenicol, cocain cyclophosphamid, doxorubicin, 5-fluoruracil,mesylate, methylsergit, phenytoin, trastuzumab, zidovudine

Bacteria: chlamydia (C. pneumonia/psittacosis) haemophilus influence,legionella, pneumophilia, brucella clostridium, francisella tularensis,neisseria meningitis, mycobacterium (tuberculosis), salmonella,staphylococcus, streptococcus A, S. pneumonia, tularemia, tetanus,syphilis, Vibrio cholera

Hypersensitivity reactions (drugs): azitromycin, benzodiazepines, clozapine,cephalosporins, dapsone, dobutamin, lithium, diuretics, thiazide,methyldopa, mexiletine, Streptomycin, sulfonamides, non-steroidalanti-inflammational drugs, tetanus toxoid, tetracycline, tricyclicantidepressiva

Spirocheta: Borrelia recurrentis, leptospira, Treponema pallidum Hypersensitivity reactions (venomes): bee, wasp, black widow spider,scorpion, snakes

Reckettsia: Coxiella burnetii, R. rickettsii/prowazekii Systemic diseases: Churg-Strauss syndrome, collagen diseases, sarcoidosis,Kawasaki disease, scleroderma

Fungi: actinomyces, aspergillus, candida, cryptococcus, histoplasma,nocardia

Others: heart stroke, hypothermia, transplant rejection, radiation injury

Protozoa: Entamoeba histolytica, leishmania, Plasmodium falciparum,Trypanosoma cruzi, Trypanosoma brucei, Toxoplasma gondii

Helmintic: ascaris, Echinococcus granulosus, Schistosoma, Trichinella spiralis,Wuchereria bancrofti

The management of myocarditis 2617D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

dysfunction-associated chronic ischaemia with less often early sys-tolic ventricular dysfunction and possibly slower progressiontowards systolic heart failure.36,46,53,54 Often patients with EC

infection complain for symptoms associated with vasospasm andEC-dysfunction despite normal or nearly normal left ventricularfunction. The contribution of the virus loads and effects of different

Figure 1 Distinct phases of myocardial injury in infectious and post-infectious myocarditis. Myocarditis is an inflammatory disease of thecardiac muscle caused by myocardial infiltration of immunocompetent cells following any kind of cardiac injury. Acute myocarditis is often aresult of a viral infection that produces myocardial necrosis and triggers an immune response to eliminate the infectious agent (healed myo-carditis and/or dilated cardiomyopathy). Chronic myocardial injury may be caused by post-infectious immune or autoimmune processes[indlammatory cardiomyopathy (DCMi)], be associated with systemic autoimmune diseases or develop by a persisting virus infection (viralheart disease), which, in the long run, are responsible for persistent or progressive ventricular dysfunction, arrhythmias, and cardiac complaints.The disease often presents as an acute form of dilated cardiomyopathy but due to its broad spectrum of presentation the clinical diagnosis isfrequently misleading. If the underlying infectious or immune-mediated causes of the disease are carefully defined by clinical and biopsy-basedtools, specific immunosuppressive and antiviral treatment options in addition to basic symptomatic therapy may improve prognosis in a numberof patients with acute and chronic disease.

H.-P. Schultheiss et al.2618D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

virus subtypes are still poorly understood.55 Consequently, distinctviruses with different infection sites in cardiac tissue do not onlyexplain the heterogenic and unpredictable course of viral heartdisease with respect to the expression of its phenotype andearly or late clinical presentation but also determine indicationand response to treatment, or prognosis.

Direct cytopathic injury, apoptosis, activation of the innate andadaptive immune system, and cardiac remodelling have all beenimplicated in the pathogenesis of viral myocarditis. It developswith pathologically distinct phases which time depending deter-mine both clinical presentation and indication for symptomaticor specific treatment strategies (Figure 1).3,56

The early phase of viral myocarditis is initiated by infection ofcardiac myocytes, fibroblasts, or ECs through receptor-mediatedendocytosis.57 –59 Acute myocardial injury can result from eitherdirect virus-mediated lytic processes or are caused by the emer-ging antiviral immune response (Figure 1).60,61 In fulminant casesof myocarditis, resulting myocyte necrosis may cause a significantloss of contractile tissue, which is accompanied by rapidly

developing cardiac failure and early death of the host (earlyphase). Cytokines released by macrophages and activation ofnatural killer cells that directly kill virus-infected heart cellsthrough perforin or granzyme-mediated lysis contribute to earlymyocardial lesions and impaired myocardial function.

The activation of antigen-specific immunity mediated by T-cells,B-cells, and antibody production (adaptive immune response)initiates the second phase of virus clearance (Figure 2).62– 64 Therecruitment of leucocytes to sites of infection is crucial to theinflammatory clearance of pathogens. Various molecules maycontrol inflammatory cell trafficking including chemokines, a familyof low-molecular-weight proteins involved in adherence of inflam-matory cells to activated endothelium and leucocyte chemotaxis(Table 2). Chemokines are important for containment of the infec-tious agents but may extend tissue injury, if attracted inflammatorycells produce pathological mediators that injure terminally differen-tiated cardiomyocytes or induce extensive fibrosis (e.g. TGF-b).65

T-regulatory cells are also important in inhibiting the pathogen-esis of acute viral myocarditis and subsequent dilated

Figure 2 Infection of cardiac endothelial cells or cardiac myocytes by virus causes direct cellular damage and subsequently an innate andadaptive immune response, all of which contribute to cardiomyopathy. Cardiomyopathy from viral injury and the subsequent immune reactioncan include diastolic as well as systolic dysfunction.

The management of myocarditis 2619D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

cardiomyopathy. Adoptive transfer of T-regulatory cells protectsmice from coxsackievirus B3 (CVB3)-induced myocarditisthrough the transforming growth factor b-coxsackie-adenovirusreceptor pathway and thus suppresses the immune response tocardiac tissue.66 Coxsackievirus B3 causes severe myocarditis inBALB/c mice. BALB/c mice receiving CD4(+) CD25(+) T regulat-ory cells from gd(+) T-cell-depleted donors developed signifi-cantly less myocarditis and CD4(+) Th1 cell responsescompared with mice receiving equal numbers of CD4(+)CD25(+) cells from infected gd (+) T-cell-sufficient animals.This study showed that gd (+) cells promote CD4(+)IFN-g(+) acute and memory responses by limiting FoxP3(+) Tregulatory cell activation.67 Finally, male BALB/c mice infectedwith CVB3 develop more severe acute inflammation in the heartcompared with females due to fewer T regulatory cells andTim-3(+) M2 macrophages.68

Negative immune modulation, an important property of anintact immune system to prevent excessive tissue damage by anoverwhelming immune response, normally occurs rapidly aftersuccessful elimination of the infectious pathogens. Under certaincircumstances, chronic immune stimulation and autoimmunitymay result from incompletely cleared virus infection or in responseto the virus- and immune-mediated chronic tissue damage,respectively. Both cellular and humeral inflammatory processesmay contribute to the progression of chronic myocardialinjury.69 The underlying pathological processes are not well

understood but in chronic inflammatory cardiomyopathy, sub-groups of patients may benefit from immunosuppression.

If viral infection and autoimmune processes have resolved, themagnitude of the remaining tissue damage determines the furthercourse of the disease.70 This late phase of post-infectious diseaseis distinguished by cardiac remodelling, progressive dilitation, andchronic heart failure. Aetiology-specific treatment is probably notuseful but only standard heart failure medication and/or devicesmay prevent or delay progression and improve prognosis.

Genetic predisposition is a likely factor in some cases of myocar-ditis, although direct evidence in human disease is lacking. Inmurine models of myocarditis, genetic predisposition to Th1,Th2, and Th17 cytokine responses influence the severity andtime course of viral infection (reviewed in Rose 2009).71 Similarly,truncations in the 5′ untranslated region of the Coxsackie B virusgenome can lower viral replication rate and lead to chronic infec-tion.72 Few genomic studies in human disease have been per-formed in part due to relatively small patient cohorts and lack oflarge multi-centre biobanks linked to well-characterized clinicalphenotypes with outcome data.

Autoimmune myocarditis, exemplified by giant cell myocarditis,usually occurs without an identified trigger such as a viral infection;although vial infection can amplify naturally occurring autoantibodiesand autoreactive T cells. The time course of cytokine and chemokineexpression following experimental autoimmune myocarditis in theLewis rat is similar to the pattern observed in models of severe

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Table 2 Potential pathogenesis-directed therapies depend on the phase of myocarditis. At all phases, guideline basedtreatments appropriate to the clinical scenario are indicated

Phase of disease Proposed mechanism and infectious agents Potential therapy

Symptomatic heart failure medication

Acute myocarditis (early phase) Direct cytopathic injury Antiviral agents?Innate immune system activation (macrophages,

NK-cells, and cytokines)Antiviral agents? intravenous immune

globulin?

Post-infectious (auto)immunity Adaptive immune response (T-cells, B-cells,antibody production)

Immune modulationSteroidsImmunoadsorptionIntravenous immune globulinMuronomab-CD3

Chronic viral cardiomyopathy Enterovirus Interferon-bAdenovirus Interferon-bErythro-/parvovirus Intravenous immune globulin (acute infection)

Type I interferons (chronic infection)

Human herpesvirus 6 Val-/GanciclovirCytomegalovirus Val-/ganciclovir

FoscanetCidovir

Ebstein-Barr virus Val-/ganciclovirFoscanetCidovovir

Herpes simplex virus AciclovirVaricella AciclovirRespiratory syntitial virus RibavirinHepatitis C virus Pegylated Interferon-a + ribavirinHIV Anti-retrovirals

H.-P. Schultheiss et al.2620D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

enteroviral myocarditis (Figure 3)73 The early rise in pro-inflammatorycytokines such as Il-2 and IFN-g, is followed by regulatory cytokinessuch as IL-10 and profibrotic cytokines like TGF-b1.65,74–76

However, in other models of autoimmune and viral myocarditisgender, hormone milieu, and the nature of the initial innateimmune response impact the severity and time course of subsequentinflammation and cardiomyopathy.77,78 In naıve BABL/c mice organ-specific, IL-17-mediated autoimmunity and heart failure is inducedby self-antigen loaded activated dendritic cells (DCs) that primeMYHC-a-specific CD4 T-cells if DCs are activated through toll-likereceptors and CD40 costimulation.79,80 In such a model, suppressionof activated autoreactive Th17-cells by IFN-g-producing monocyteslimits cardiac inflammation and prevents further tissue injury.81

Select knowledge gaps and opportunities in the pathogenesis ofmyocarditis† The heterogeneity of clinical presentations and incomplete

understanding of human immunopathology are obstacles tocurrent investigation.

† The major long-term consequence of myocarditis is chronicdilated cardiomyopathy, but the pathways that lead to myo-cardial fibrosis are poorly understood.

† Investigation of the pathways that lead to fibrosis and DCMafter viral injury requires bidirectional collaboration betweeninvestigators with small animal models that recapitulate latecardiac fibrosis and clinicians who take a multi-centre,programmatic approach to patient care.

† The genetic factors contributing to human myocarditis arepoorly understood. Advances in the genetics of myocarditiswill require multi-centre, pathogen-specific registries withlinked biobanks, core facilities with next generation wholegenome sequencing and candidate driven transcriptomic andmetabolomic studies.

Diagnosis and prognosisThe clinical presentation of acute myocarditis ranges broadly fromsubclinical disease to fulminant heart failure, and chest pain, palpi-tations, and syncope are not uncommon. Young children oftenhave a more fulminant presentation than adults.82 Men with pre-sumed viral myocarditis may have more severe damage thanwomen.83 A viral prodrome including fever and respiratory or gas-trointestinal symptoms frequently precedes the onset of myocar-ditis.84 Chest pain in acute myocarditis may resemble angina withischaemic electrocardiographic changes or be more typical forpericarditis, when both the epicardium and adjacent pericardiumare inflamed. In a representative sample of the adult patientsscreened in the European Study of the Epidemiology and Treat-ment of inflammatory Heart Disease, 72% had dyspnoea, 32%had chest pain, and 18% had arrhythmias.85

The sensitivity of cardiac biomarkers of myocardial injury variesdepending on the time from symptom onset to testing and thecut-off values used. For example, in acute paediatric myocarditis,the sensitivity of specificity of Troponin T (TnT) were 75 and75% when the cut-off was set at 0.026 ng/mL and 63 and 89%with a cut-off value of 0.071 ng/mL.86 Elevations of Troponin I(TnI) in patients with myocarditis were significantly correlatedwith ≤1-month duration of heart failure symptoms.87 Troponin Ior TnT are more commonly elevated than creatinine kinase MBin both adults and children with acute myocarditis.87–89 Non-specific serum makers of inflammation, including erythrocytesedimentation rate, C-reactive protein, and leucocyte count maybe elevated.

The most common electrocardiogram (ECG) findings arenon-specific T-wave changes.90 Occasionally, the ECG changesmay mimic acute myocardial infarction or pericarditis with STsegment elevation, ST segment depression, PR segmentdepression, and pathological Q-waves.91,92 Tachyarrhythmias are

Figure 3 Adapted from Kodama M. et al. Animal Models of Autoimmune Myocarditis, in Cooper, LT, ed, Myocarditis from Bench to Bedside.Humana Press, Towtowa, NJ 2003.

The management of myocarditis 2621D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

often non-sustained and rarely cause haemodynamic compromisein adult viral myocarditis. The prognostic significance and optimalmanagement of non-sustained ventricular tachycardia in thesetting of acute myocarditis are not known. However, Q-wavesand a widened QRS complex, including left bundle branch block,are associated with higher rates of death or cardiactransplantation.90,93 –95

Echocardiography is useful to exclude other causes of heartfailure and identify ventricular thrombi. There are no specific echo-cardiographic features of myocarditis.96,97 Indeed, segmental orglobal wall motion abnormalities can mimic myocardial infarction.98

Patients with fulminant myocarditis tend to present with morenormal cardiac chamber dimensions and thickened walls, com-pared with patients with less acute myocarditis who have greaterleft ventricular dilation and normal wall thickness.99 Right ventricu-lar dysfunction is an uncommon but important predictor of deathor cardiac transplantation.100 Newer imaging techniques includingstrain echocardiography may have better specificity and specificityfor myocarditis.

Cardiovascular magnetic resonance (CMR) in suspected myo-carditis can localize and quantitate tissue injury, includingoedema, hyperaemia, and fibrosis.101 In a recent series of 82patients with myocarditis all of whom had biopsy-proven disease,CMR alone made the correct diagnosis in 80% (66 out of 82)cases.102 However, both T2- and T1-weighted imaging areneeded to achieve optimal sensitivity and specificity, and in con-trast to older reports, CMR abnormalities do not correlateclosely with endomyocardial biopsy (EMB) evidence of myocar-ditis.39 When two or more of the three ‘Lake Louise’ criteria arepositive, myocardial inflammation can be predicted with a diagnos-tic accuracy of 78%; if only delayed, post-gadolinium enhancementimaging is performed, the diagnostic accuracy drops to 68%.101

Prospective clinical studies of the prognostic value of CMR areneeded to identify whether tissue characterization adds to themanagement or outcome of patients with myocarditis.

Confirmation of myocarditis still requires histological or immu-nohistological evidence of inflammation in heart tissue. Endomyo-cardial biopsy can be performed with a very low majorcomplication rate when performed by highly experienced oper-ators.103 In experienced hands, left ventricular biopsy is as safeas right ventricular biopsy.39 An AHA/ACCF/ESC joint scientificstatement recommended that EMB should be performed (Class Iindication) in patients with heart failure and (i) a normal sizedor dilated left ventricle, ,2 weeks of symptoms, and haemo-dynamic compromise and also in (ii) patents with a dilated ventri-cle, 2 weeks to 3 months of symptoms, new ventriculararrhythmias or Mobitz type II second degree or third degreeheart block, or who fail to respond to usual care within 1–2weeks.104 More recently, EMB-based criteria (inflammationpresent by immunohistology and viral genomes absent by poly-merase chain reaction) have been used to define a cohort ofpatients with chronic DCM who respond to immunosuppres-sion.105 In clinical practice, EMB should be used in those scenariosin which the incremental prognostic and therapeutic informationgained from biopsy outweighs the risk and cost. This will vary bymedical centre, depending on availability of necessary facilitiesand expertise.

Select knowledge gaps and opportunities in the diagnosis and prog-nosis of myocarditis† The incidence and prevalence of myocarditis are unknown.

There is an immediate need for inexpensive, sensitive, andspecific diagnostic tests that can be used in population-basedstudies in regions without access to advanced imaging orcardiac catheterization laboratories.

† Gender issues: the protective effect of estrogens and the roleof viral myocarditis in peripartum cardiomyopathy areincompletely understood.

† Paediatric cases: the optimal strategy for diagnosingmyocarditis in children is controversial and based largelyon expert opinion.

† Molecular inflammatory markers in peripheral blood com-bined with newer echocardiographic and CMR imaging tech-niques and EMB may lead to more accurate diagnosis andaetiology-specific treatments.

TreatmentCardiomyocytes can be destroyed by direct virus damage, the anti-viral immune response, or a truly autoimmune injury. Since adultcardiomyocytes rarely regenerate, recovery of myocardial functiondepends on the residual myocardial tissue. The treatment responseof acute and chronic myocarditis therefore depends on the specificcauses of the disease, severity of irreversible tissue alterations atthe onset of treatment and consequently on the potential of themyocardium to compensate for such processes. If the pre-treatment damage is severe, aetiology-specific treatment optionsat best can halt rapid progression of the disease but will notachieve significant improvement in ventricular function.

Treatment for heart failureThe mainstay of treatment for myocarditis presenting as dilated cardi-omyopathy is an optimal heart failure medical regimen. If the left ven-tricular ejection fraction (LVEF) is ,40%, we recommend that anangiotensin-converting enzyme-inhibitor/angiotensin receptorblocker and/or a b-adrenergic blocking agent be used according tothe current AHA/ACCF and ESC guidelines for the management ofheart failure.106–108 Supporting this strategy are experimentalstudies that show captopril and candesartan improve myocarditis inmurine myocarditis models.109,110 Non-steroidal anti-inflammatoryagents such as indomethacin should be considered for patients witha myopericarditis-like syndrome of chest pain and normal or near-normal ventricular function because they can worsen cardiomyopathyin experimental models.111 In addition to guideline-based medicalmanagement, we recommend that patients with acute myocarditisrefrain from competitive athletics for a period of months after theacute infection or until ventricular recovery has been documentedby non-invasive imaging.

Antiviral treatmentTreatment of early diseaseElimination of viral translation, transcription, and proliferation withthe use of antiviral medications that target viral attachment tohost-cell receptors, virus entry, or virus uncoating, e.g. Pleconaril,

H.-P. Schultheiss et al.2622D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

WIN 54954, or soluble CAR-Fc, would be effective in the earlystages, but, unfortunately, most adult patients present in thechronic phases of disease (Figure 1).112 These agents, therefore,are of limited use in virus-associated heart disease. The currentchallenge of antiviral therapy in patients with chronic cardiac viralinfections therefore is the timing of treatment that prevents pro-gressive myocardial injury by viral clearance before chronicallyinfected heart tissue has irreversible damage.

Treatment of chronic viral heart diseaseInterferon-betaInterferons serve as a natural defense against many viral infections.Their innate production is associated with clinical recovery fromviral infection and subsequent sequelae, while exogenous adminis-tration is protective. Type I interferons therefore constitute apromising choice for treatment of chronic viral cardiomyopathy.Currently, there is no approved treatment for chronic viral heartdisease, but data from uncontrolled open labelled phase IIstudies have demonstrated that subgroups of patients, who hadnot improved upon regular heart failure medication, may get sig-nificant benefit even years after onset of chronic disease.

In a first study, patients with persistent enterovirus and adenovirusinfections of the myocardium responded well to a 6-monthsinterferon-beta (IFN-b1a) course.113 Complete elimination ofenteroviral and adenoviral genome was proved by follow-up biopsiestaken 3 month after termination of the antiviral therapy. Virus clear-ance was paralleled by an improvement of mean left ventricular func-tion, a decrease in ventricular size, an amelioration of heart failuresymptoms, and a decrease of infiltrating inflammatory cells. Ofnote, no patient deteriorated and patients with severely affectedLV-dysfunction gained most benefit.113

The drug is usually administered subcutaneously every other dayin addition to constant heart failure medication for a 6–months’period. In order to limit IFN-specific side effects, the patientshould enter a run-in period to improve tolerance following astepped regimen, during which the patient receives 2 × 106 IUIFN-b per application every other day for 1 week. Within the fol-lowing 2 weeks, the study medication can be elevated to 4 × 106

and 6 × 106 IU IFN-b, respectively, and continued for the follow-ing 21 weeks. The IFN-b1a medication was well tolerated with nounexpected non-cardiologic or cardiologic side effects. Frequentlyreported IFN-associated side effects were fatigue, influenza-likesymptoms, and injection site erythema but symptoms vanished reg-ularly during the first 4 weeks of treatment. No major clinicalevents occurred during the treatment phase or follow-up.

If patients with a severely depressed cardiac contractility (LVEF,25%) are treated with an immunmodulatory drug such as IFN-b,LV function should be close-mesh monitored by echocardiography.Between Week 4 and 12, patients complain for a mild aggravation ofheart failure symptoms which is often associated with a walloedema, a slight increase of LV-dimensions and a minor deteriorationof LVEF. Complains regularly disappear within 1 or 2 weeks followedby an direct and continuous improvement of heart failure in �40% ofpatients. Improvement may start with a delay of for 2–4 months in25–30% of patients. This outlined course, which is probably causedby an IFN-induced cellular immune response, preferentially concernsviruses that infect cardiomyocytes (e.g. enteroviruses). It is

uncommonly observed in infections such as parvovirus B19 orhuman herpesvirus 6 which according to a distinct pathophysiologydo not directly affect contractile cells of the myocardium.

Parvovirus B19 and human herpes virus 6 respond less well uponIFN-b treatment with respect to virus clearance and haemodynamicchanges, although affected patients too improve clinically despiteincomplete virus clearance following reduction of virus load and/orimprovement of endothelial dysfunction.114,115 Complete clearanceof those viruses may need longer treatment intervals, higher doses,or even a complete change of the antiviral treatment regimens. Cur-rently, effective treatment conditions for viruses other than entero-virus and adenovirus have not yet been tested consequently.

Information taken from uncontrolled cohorts of treated virus-positive patients may be, however, of limited value for generaltreatment recommendations for a number of reasons. Accordingto animal and cell culture studies, different viruses and even differ-ent virus subtypes may respond in distinct and unpredictable waysto immunomodulatory treatment. Furthermore, virus load and thetype of the infected cell, both of which influence course of thedisease and drug response, have to be taken into consideration.Despite such comprehensible reservations, a recent randomized,placebo controlled phase II trial (BICC-Study) has confirmed thateven patients with a long history who had not responded to con-ventional heart failure treatment can get clinical and haemo-dynamic benefit from an IFN therapy.115

Response to the IFN-b1a therapy may be influenced by thevirus-associated local inflammation and the extent of the myocardialinjury at the time when the specific therapy is started. The lack ofhaemodynamic improvement in some of the patients is not unex-pected, if the long history and the above-mentioned pathogenetic sug-gestions are taken into consideration. In view of the informationderived from other virus infections of the heart, one can expect thatthe chronic enteroviral and adenoviral infections or thevirus-associated inflammatory process may already have caused con-siderable myocardial damage and consequently, ventricular dysfunc-tion may be unable to recover completely in those patients who didnot improve despite virus clearance. Nevertheless, the completeclearance of both viruses after treatment suggests that early biopsy-based diagnosis and timely treatment may prevent disease progressionand thereby improve the outcome of chronic viral cardiomyopathy.

Select knowledge gaps and opportunities in the treatment ofmyocarditis† Studies have not been done to determine when and how to

discontinue standard heart failure therapy in patients whorecovery LV function.

† More work is needed to identify patient cohorts who willbenefit from tailored antiviral or immunosuppressivetherapy.

† The optimal time frame to initiate immunosuppressive treat-ment improves cardiac function and health without impair-ment of a healing immune processes is unknown.

† With respect to our limited knowledge of involved patho-logical processes and optimal treatment conditions, preven-tion of disease progression rather than curative treatmentmay turn out to be the most realistic short-term goal.

The management of myocarditis 2623D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

ImmunosuppressionMyocardial inflammatory processes due to pathogenic autoimmu-nity may survive myocardial virus elimination and warrant immuno-suppressive treatment in order to prevent later immune-mediatedmyocardial injury.116 –118 Immunosuppression demands biopsy-based exclusion of virus from treated patients since virus-positivepatients do not improve upon anti-inflammatory treatment, whilevirus-negative patients with post-infectious or auto-immune inflam-matory processes respond well in early clinical trials.105,117,119,120

Frequently administered anti-inflammatory drugs are immuno-globulins, corticosteroids, azathioprine, and cyclosporine, whichare administered on top of regular heart failure medication.a-Methylprednisolone is generally given at Charite Hospital, at adose of 1 mg/kg body weight, initially for 4 weeks. Depending onthe body weight, azathioprine is administered at a dose of 100–150 mg daily in addition to the corticosteroid. The steroiddosage is titrated down every 2 weeks in increments of 10 mguntil a maintenance dose of 10 mg is reached. The treatment dur-ation should last for 3 to 6 months. Actual data of first randomizedtrials confirm efficacy of those treatment regimens in carefullyselected patients.105,118

Sustained beneficial effects of immunosuppression on heartfailure symptoms, left ventricular dimensions, and LVEF in immuno-histologically biopsy-proven inflammatory cardiomyopathy havebeen confirmed in a randomized trial with 41 patients at 2 yearsfollow-up after ≥3 months of treatment with corticosteroidsand azathioprine.118 This trial ultimately validates the diagnosticsensitivity and accuracy of cell adhesion molecule (CAM) abun-dance for indlammatory cardiomyopathy (DCMi) even in theabsence of lymphocytic infiltration, possibly due to the close func-tional association between CAM induction and immunocompetentinfiltration and cytokine induction,121 and thus constitutes animportant criterion for selecting those patients who will likelybenefit from immunosuppression. Furthermore, this studyshowed that a 3-month regimen may equally be effective as pre-vious trials that used 6 months of immunosuppression, and thatbeneficial effects last for an extended period of time (2 years). Inanother recently published randomized trial (TIMIC study), theauthors confirmed a positive treatment response in patients withchronic active myocarditis.105 Thirty-eight out of 43 patients onimmunosuppressive therapy (88%) showed a improvement ofcardiac function and dimensions, defined as an increase of .10percentage points in the absolute EF and a reduction of LV end-diastolic volume (EDV) or LV end-diastolic diameter (EDD) 10%(i.e. LVEF from 26.4+ 6.9 to 48.0+7.3%, LVEDV from 258.0+52.5 to 125.9+ 29.6, and LVEDD from 68.6+7.4 to 52.8+6.3 mm). None of the untreated patients show at 6 monthimprovement of LVEF, that significantly worsened compared withbaseline. In particular, 35 out of 42 patients (83%) showedfurther impairment of cardiac function (LVEF from 27.6+6.6 to19.5+4.8, LVEDV from 244.7+48.0 to 287.3+48.0, andLVEDD from 69.2+ 7.9 to 75.3+ 7.4), while the remaining 7patients remained stationary. Finally, LVEF declined to baseline(27.2+ 5.6%) or lower (19.7+4.4%) values.

Currently available data show that immunosuppressive therapyin patients with biopsy-proven, virus-negative inflammatory

cardiomyopathy is an effective and safe option in addition to sup-portive treatment for recovery of cardiac failure. However, largerstudies powered to detect a difference in clinical endpoints such asheart failure hospitalization, transplantation, and death are stillneeded.

Intravenous immunoglobulinand immunoadsorptionThe first data of acute myocarditis treated with intravenous immu-noglobulin (IVIG) suggested that use of high-dose IVIG for treat-ment of acute myocarditis is associated with improved recoveryof left ventricular function and with a tendency to better survivalduring the first year after presentation.122 Later investigationsand randomized studies which compared IVIG and cortisone treat-ment revealed that the treatment with intravenous immune globu-lin in children was not effective.123 Freedom from death ortransplantation was 81% at 1 year, and 74% at 5 years, with nodifference between the modes of treatments. The median timeto recovery of function was also comparable between thegroups. Thus, treatment with intravenous immune globulinappear to confer no advantage to steroid therapy alone.124

The rationale for immunoadsorption is to lower cardiotoxicantibodies in the patient’s plasma, and with serial treatmentsover 5 or more days, extract antibodies and immune complexesfrom the heart as well. The plasma is separated from cellular com-ponents by a centrifuge or column and passed through an immu-noadsorbtion column. IgG and to a lesser degree IgA and IgMare non-specifically adsorbed during repetitive sessions. PlasmaIgG levels are partially restored by infusion of 0.5 g/kg polyclonalIgG .18 h after the last apheresis treatment. The favourablehaemodynamic results of immunoadsorption in patients withDCMi may be related to removal of functionally active cardiacautoantibodies or other immunologically active compounds, sinceimmunoadsorption leads to biopsy-proven decrease in lymphocy-tic infiltration and CAM expression.

Summary and perspectivesCurrent pharmacological heart failure therapy ofmyocarditis-associated cardiomyopathy mainly focuses on decreas-ing the activity of the neuroendocrine system but does not directlyinfluence virus-induced inflammatory cardiomyopathy.Interferon-b suppresses erythrovirus replication while at thesame time it improves replication and viability of human ECs.46

Accumulating experimental and clinical data indicate that cellulartransplantation may improve myocardial function.26,125,126

Mesenchymal stem cells (MSCs) have anti-apoptotic, anti-fibroticproperties, are non-immunogenic, and possess immunomodulatoryproperties.127 –129 They suppress T-cell responses, induce apopto-sis of activated T cells and increase T regulatory cells.130 –132

Interferon-g primes MSC-mediated immunoregulatory effectsand induces nitric oxide (NO) production in MSCs the latter ofwhich exerts anti-apoptotic effects on cardiomyocytes and hasantiviral properties in a NO-dependent manner.129,133 – 135 Thesedifferent strategies including pharmacological and gene therapeutic

H.-P. Schultheiss et al.2624D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

approaches directed at blocking viral replication or stimulating theantiviral-directed immune response, are under investigation inexperimental and clinical studies.70,114,136,137 Because of the lowrate of diagnosis, multi-centre collaborations with standardizedevaluations and treatment protocols, mechanistically orientedregistries, and core molecular diagnostic facilities will be needed.

FundingBasic experimental diagnostic and clinical works have been supportedby a grant from the German Research Foundation (DFG), Transregio-nal Collaborative Research Centre ‘Inflammatory Cardiomyopathy–Molecular Pathogenesis and Therapy’ (SFB TR 19 04) (HPS, UK,Charite Berlin).

Conflict of interest: none declared.

References1. Taliercio CP, Olney BA, Lie JT. Myocarditis related to drug hypersensitivity. Mayo

Clin Proc 1985;60:463–468.2. Guarner J, Paddock CD, Shieh WJ, Packard MM, Patel M, Montague JL,

Uyeki TM, Bhat N, Balish A, Lindstrom S, Klimov A, Zaki SR. Histopathologicand immunohistochemical features of fatal influenza virus infection in childrenduring the 2003–2004 season. Clin Infect Dis 2006;43:132–140.

3. Liu PP, Schultheiss HP. Myocarditis. In: Baunwald, E. (ed), Heart Disease. 8th ed.Philadelphia: W B Saunders Co; 2008, p1775–1792.

4. Blauwet LA, Cooper LT. Myocarditis. Prog Cardiovasc Dis 2010;52:274–288.5. Calabrese F, Rigo E, Milanesi O, Boffa GM, Angelini A, Valente M, Thiene G. Mol-

ecular diagnosis of myocarditis and dilated cardiomyopathy in children: clinico-pathologic features and prognostic implications. Diagn Mol Pathol 2002;11:212–221.

6. Bowles NE, Kearney DL, Ni J, Perez-Atayde AR, Kline MW, Bricker JT,Ayres NA, Lipshultz SE, Shearer WT, Towbin JA. The detection of viralgenomes by polymerase chain reaction in the myocardium of pediatric patientswith advanced HIV disease. J Am Coll Cardiol 1999;34:857–865.

7. Bowles NE, Ni J, Kearney DL, Pauschinger M, Schultheiss HP, McCarthy R,Hare J, Bricker JT, Bowles KR, Towbin JA. Detection of viruses in myocardialtissues by polymerase chain reaction. Evidence of adenovirus as a commoncause of myocarditis in children and adults. J Am Coll Cardiol 2003;42:466–472.

8. Carturan E, Milanesi O, Kato Y, Giacometti C, Biffanti R, Thiene G, Calabrese F.Viral detection and tumor necrosis factor alpha profile in tracheal aspirates fromchildren with suspicion of myocarditis. Diagn Mol Pathol 2008;17:21–27.

9. Comar M, D’Agaro P, Campello C, Poli A, Breinholt JP III, Towbin JA, Vatta M.Human herpes virus 6 in archival cardiac tissues from children with idiopathicdilated cardiomyopathy or congenital heart disease. J Clin Pathol 2009;62:80–83.

10. Dettmeyer R, Baasner A, Schlamann M, Haag C, Madea B. Coxsackie B3 myo-carditis in 4 cases of suspected sudden infant death syndrome: diagnosis byimmunohistochemical and molecular-pathologic investigations. Pathol Res Pract2002;198:689–696.

11. Gouton M, Di Filippo S, Sassolas F, Stamm D, Bozio A, Floret D. [Acute infec-tious myocarditis in children. Apropos of 2 series from Lyon]. Arch Mal CoeurVaiss 1995;88:753–759.

12. Martin AB, Webber S, Fricker FJ, Jaffe R, Demmler G, Kearney D, Zhang YH,Bodurtha J, Gelb B, Ni J. Acute myocarditis. Rapid diagnosis by PCR in children.Circulation 1994;90:330–339.

13. Munro K, Croxson MC, Thomas S, Wilson NJ. Three cases of myocarditis inchildhood associated with human parvovirus (B19 virus). Pediatr Cardiol 2003;24:473–475.

14. Savon C, Acosta B, Valdes O, Goyenechea A, Gonzalez G, Pinon A, Mas P,Rosario D, Capo V, Kouri V, Martinez PA, Marchena JJ, Rodriguez H,Guzman MG. A myocarditis outbreak with fatal cases associated with adenovirussubgenera C among children from Havana City in 2005. J Clin Virol 2008;43:152–157.

15. Schowengerdt KO, Ni J, Denfield SW, Gajarski RJ, Bowles NE, Rosenthal G,Kearney DL, Price JK, Rogers BB, Schauer GM, Chinnock RE, Towbin JA. Associ-ation of parvovirus B19 genome in children with myocarditis and cardiac allo-graft rejection: diagnosis using the polymerase chain reaction. Circulation 1997;96:3549–3554.

16. Talsma MD, Kroos MA, Visser G, Kimpen JL, Niezen KE. A rare presentation ofchildhood pompe disease: cardiac involvement provoked by Epstein-Barr virusinfection. Pediatrics 2002;109:e65.

17. Vare D, Vare B, Dauphin C, Lafeuille H, Gaulme J, Labbe A, Motreff P, Lusson JR.[Acute myocarditis in children. Study of 11 clinical cases]. Arch Mal Coeur Vaiss2000;93:571–579.

18. Camargo PR, Okay TS, Yamamoto L, Del Negro GM, Lopes AA. Myocarditis inchildren and detection of viruses in myocardial tissue: implications for immuno-suppressive therapy. Int J Cardiol 2011;148:204–248.

19. Kuhl U, Lassner D, Pauschinger M, Gross UM, Seeberg B, Noutsias M, Poller W,Schultheiss HP. Prevalence of erythrovirus genotypes in the myocardium ofpatients with dilated cardiomyopathy. J Med Virol 2008;80:1243–1251.

20. Kuhl U, Pauschinger M, Noutsias M, Seeberg B, Bock T, Lassner D, Poller W,Kandolf R, Schultheiss HP. High prevalence of viral genomes and multiple viralinfections in the myocardium of adults with “idiopathic” left ventricular dysfunc-tion. Circulation 2005;111:887–893.

21. Pauschinger M, Bowles NE, Fuentes-Garcia FJ, Pham V, Kuhl U,Schwimmbeck PL, Schultheiss HP, Towbin JA. Detection of adenoviralgenome in the myocardium of adult patients with idiopathic left ventricular dys-function. Circulation 1999;99:1348–1354.

22. Pauschinger M, Doerner A, Kuehl U, Schwimmbeck PL, Poller W, Kandolf R,Schultheiss HP. Enteroviral RNA replication in the myocardium of patientswith left ventricular dysfunction and clinically suspected myocarditis. Circulation1999;99:889–895.

23. Modlin FJ. Picornaviridae. In: Mandell GL, Douglas RG, Bennett JE Jr (eds), Prin-ciples and Practice of Infectious Diseases. New York: Chrchill Livingstone; 1990,p721–732.

24. Fujioka S, Kitaura Y, Ukimura A, Deguchi H, Kawamura K, Isomura T, Suma H,Shimizu A. Evaluation of viral infection in the myocardium of patients with idio-pathic dilated cardiomyopathy. J Am Coll Cardiol 2000;36:1920–1926.

25. Chow LH, Beisel KW, McManus BM. Enteroviral infection of mice with severecombined immunodeficiency. Evidence for direct viral pathogenesis of myocar-dial injury. Lab Invest 1992;66:24–31.

26. Yuan JP, Zhao W, Wang HT, Wu KY, Li T, Guo XK, Tong SQ. CoxsackievirusB3-induced apoptosis and caspase-3. Cell Res 2003;13:203–209.

27. Bultmann BD, Klingel K, Sotlar K, Bock CT, Baba HA, Sauter M, Kandolf R. Fatalparvovirus B19-associated myocarditis clinically mimicking ischemic heartdisease: an endothelial cell-mediated disease. Hum Pathol 2003;34:92–95.

28. Bock CT, Klingel K, Aberle S, Duechting A, Lupescu A, Lang F, Kandolf R. Humanparvovirus B19: a new emerging pathogen of inflammatory cardiomyopathy. J VetMed 2005;52:340–343.

29. Klingel K, Sauter M, Bock CT, Szalay G, Schnorr JJ, Kandolf R. Molecular pathol-ogy of inflammatory cardiomyopathy. Med Microbiol Immunol (Berl) 2004;193:101–107.

30. Krueger GRF, Rojo J, Buja LM, Lassner D, Kuehl U. Human herpesvirus-6(HHV-6) is a possible cardiac pathogen: an immunihistological and ultrastruc-tural study. Hosp Gen 2008;71:187–191.

31. Brown KE, Anderson SM, Young NS. Erythrocyte P antigen: cellular receptor forB19 parvovirus. Science 1993;262:114–117.

32. Weigel-Kelley KA, Yoder MC, Srivastava A. Alpha5beta1 integrin as a cellularcoreceptor for human parvovirus B19: requirement of functional activation ofbeta1 integrin for viral entry. Blood 2003;102:3927–3933.

33. Munakata Y, Saito-Ito T, Kumura-Ishii K, Huang J, Kodera T, Ishii T, Hirabayashi Y,Koyanagi Y, Sasaki T. Ku80 autoantigen as a cellular coreceptor for human par-vovirus B19 infection. Blood 2005;106:3449–3456.

34. Vallbracht KB, Schwimmbeck PL, Kuhl U, Seeberg B, Schultheiss HP.Endothelium-dependent flow-mediated vasodilation of systemic arteries isimpaired in patients with myocardial virus persistence. Circulation 2004;110:2938–2945.

35. Tschope C, Bock CT, Kasner M, Noutsias M, Westermann D, Schwimmbeck PL,Pauschinger M, Poller WC, Kuhl U, Kandolf R, Schultheiss HP. High prevalenceof cardiac parvovirus B19 infection in patients with isolated left ventricular dias-tolic dysfunction. Circulation 2005;111:879–886.

36. Vallbracht KB, Schwimmbeck PL, Kuhl U, Rauch U, Seeberg B, Schultheiss HP.Differential aspects of endothelial function of the coronary microcirculationconsidering myocardial virus persistence, endothelial activation, and myocardialleukocyte infiltrates. Circulation 2005;111:1784–1791.

37. Lupescu A, Bock C-T, Lang PA, Aberle S, Kaiser H, Kandolf R, Lang F. Phospho-lipase A2 activity-dependent stimulation of Ca2+ entry by human parvovirusB19 capsid protein VP1 J Virol 2006;80:11370–11380 doi:10.1128/JVI.01041-06.

38. Fischer D, Rossa S, Landmesser U, Spiekermann S, Engberding N, Hornig B,Drexler H. Endothelial dysfunction in patients with chronic heart failure is inde-pendently associated with increased incidence of hospitalization, cardiac trans-plantation, or death. Eur Heart J 2005;26:65–69.

39. Yilmaz A, Kindermann I, Kindermann M, Mahfoud F, Ukena C, Athanasiadis A,Hill S, Mahrholdt H, Voehringer M, Schieber M, Klingel K, Kandolf R, Bohm M,Sechtem U. Comparative evaluation of left and right ventricular endomyocardial

The management of myocarditis 2625D

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

biopsy: differences in complication rate and diagnostic performance. Circulation2010;122:900–909.

40. Moffat S, Yaegashi N, Tada K, Tanaka N, Sugamura K. Human parvovirus B19nonstructural (NS1) protein induces apoptosis in erythroid lineage cells. J Virol1998;72:3018–3028.

41. Hsu TC, Tzang BS, Huang CN, Lee YJ, Liu GY, Chen MC, Tsay G. Increasedexpression and secretion of interleukin-6 in human parvovirus B19 non-structural protein (NS1) transfected COS-7 epithelial cells. Clin Exp Immunol2006;144:152–157.

42. Fu Y, Ishii KK, Munakata Y, Saitoh T, Kaku M, Sasaki T. Regulation of tumornecrosis factor alpha promotor by human parvovirus B19 NS1 through acti-vation of AP-1 and AP-2. J Virol 2008;82:7942–7952.

43. Duechting A, Tschope C, Kaiser H, Lamkemeyer T, Tanaka N, Aberle S, Lang F,Torresi J, Kandolf R, Bock C-T. Human parvovirus B19 NS1 protein modulatesinflammatory signaling by activation of STAT3/PIAS3 in human endothelial cells.J Virol 2008;82:7942–7952.

44. Poole BD, Karetny YV, Naides S. Parvovirus B19-induced apoptosis of hepato-cytes. J Virol 2004;78:7775–7783.

45. Poole BD, Zhou J, Grote A, Schiffenbauer A, Naides SJ. Apoptosis of liver-derived cells induced by parvovirus B19 nonstructural protein. J Virol 2006;80:4114–4121.

46. Schmidt-Lucke C, Spillmann F, Bock T, Kuhl U, Van Linthout S, Schultheiss HP,Tschope C. Interferon beta modulates endothelial damage in patients withcardiac persistence of human parvovirus B19 infection. J Infect Dis 2010;201:6.

47. Rotola A, Di Luca D, Cassai E, Ricotta D, Giulio A, Turano A, Caruso A,Muneretto C. Human herpesvirus 6 infects and replicates in aortic endothelium.J Clin Microbiol 2000;38:3135–3136.

48. Wu CA, Shanley JD. Chronic infection of human umbilical vein endothelial cellsby human herpesvirus-6. J Gen Virol 1998;79:1247–1256.

49. Aita K, Jin Y, Irie H, Takahashi I, Kobori K, Nakasato Y, Kodama H, Yanagawa Y,Yoshikawa T, Shiga J. Are there histopathologic characteristics particular to ful-minant hepatic failure caused by human herpesvirus-6 infection? A case reportand discussion. Hum Pathol 2001;32:887–889.

50. Caruso A, Rotola A, Comar M, Favilli F, Galvan M, Tosetti M, Campello C,Caselli E, Alessandri G, Grassi M, Garrafa E, Cassai E, Di Luca D. HHV-6infects human aortic and heart microvascular endothelial cells, increasing theirability to secrete proinflammatory chemokines. J Med Virol 2002;67:528–533.

51. Takatsuka H, Wakae T, Mori A, Okada M, Fujimori Y, Takemoto Y, Okamoto T,Kanamaru A, Kakishita E. Endothelial damage caused by cytomegalovirus andhuman herpesvirus-6. Bone Marrow Transplant 2003;31:475–479.

52. Badorff C, Lee GH, Lamphear BJ, Martone ME, Campbell KP, Rhoads RE,Knowlton KU. Enteroviral protease 2A cleaves dystrophin: evidence of cyto-skeletal disruption in an acquired cardiomyopathy. Nat Med 1999;5:320–326.

53. Kuhl U, Pauschinger M, Bock T, Klingel K, Schwimmbeck CP, Seeberg B,Krautwurm L, Poller W, Schultheiss HP, Kandolf R. Parvovirus B19 infectionmimicking acute myocardial infarction. Circulation 2003;108:945–950.

54. Yilmaz A, Mahrholdt H, Athanasiadis A, Vogelsberg H, Meinhardt G,Voehringer M, Kispert EM, Deluigi C, Baccouche H, Spodarev E, Klingel K,Kandolf R, Sechtem U. Coronary vasospasm as the underlying cause for chestpain in patients with PVB19 myocarditis. Heart 2008;94:1456–1463.

55. Bock CT, Klingel K, Kandolf R. Human parvovirus B19-associated myocarditis. NEngl J Med 2010;362:1248–1249.

56. Mason JW. Myocarditis and dilated cardiomyopathy: an inflammatory link. Cardi-ovasc Res 2003;60:5–10.

57. Bergelson JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithivas A, Hong JS,Horwitz MS, Crowell RL, Finberg RW. Isolation of a common receptor for Cox-sackie B viruses and adenoviruses 2 and 5. Science 1997;275:1320–1323.

58. Martino TA, Petric M, Brown M, Aitken K, Gauntt CJ, Richardson CD, Chow LH,Liu PP. Cardiovirulent coxsackieviruses and the decay-accelerating factor(CD55) receptor. Virology 1998;244:302–314.

59. Liu PP, Opavsky MA. Viral myocarditis: receptors that bridge the cardiovascularwith the immune system?. Circ Res 2000;86:253–254.

60. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell2006;124:783–801.

61. Kawai T, Akira S. TLR signaling. Cell Death Differ 2006;13:816–825.62. Yuan J, Cao AL, Yu M, Lin QW, Yu X, Zhang JH, Wang M, Guo HP, Liao YH.

Th17 cells facilitate the humoral immune response in patients with acute viralmyocarditis. J Clin Immunol 2010;30:226–234.

63. Yuan J, Yu M, Lin QW, Cao AL, Yu X, Dong JH, Wang JP, Zhang JH, Wang M,Guo HP, Cheng X, Liao YH. Th17 cells contribute to viral replication in coxsack-ievirus B3-induced acute viral myocarditis. J Immunol 2010;185:4004–4010.

64. Yuan J, Yu M, Lin QW, Cao AL, Yu X, Dong JH, Wang JP, Zhang JH, Wang M,Guo HP, Liao YH. Neutralization of IL-17 inhibits the production of anti-ANTautoantibodies in CVB3-induced acute viral myocarditis. Int Immunopharmacol2010;10:272–276.

65. Rutschow S, Leschka S, Westermann D, Puhl K, Weitz A, Ladyszenskij L,Jaeger S, Zeichhardt H, Noutsias M, Schultheiss HP, Tschope C,Pauschinger M. Left ventricular enlargement in coxsackievirus-B3 inducedchronic myocarditis—ongoing inflammation and an imbalance of the matrixdegrading system. Eur J Pharmacol 2009;630:145–151.

66. Puglielli MT, Browning JL, Brewer AW, Schreiber RD, Shieh WJ, Altman JD,Oldstone MB, Zaki SR, Ahmed R. Reversal of virus-induced systemic shockand respiratory failure by blockade of the lymphotoxin pathway. Nat Med1999;5:1370–1374.

67. Huber SA. Depletion of gammadelta+ T cells increases CD4+ FoxP3 (T regu-latory) cell response in coxsackievirus B3-induced myocarditis. Immunology 2009;127:567–576.

68. Frisancho-Kiss S, Coronado MJ, Frisancho JA, Lau VM, Rose NR, Klein SL,Fairweather D. Gonadectomy of male BALB/c mice increases Tim-3(+) alterna-tively activated M2 macrophages, Tim-3(+) T cells, Th2 cells and Treg in theheart during acute coxsackievirus-induced myocarditis. Brain Behav Immun2009;23:649–657.

69. Rose NR. Autoimmunity in coxsackievirus infection. Curr Top Microbiol Immunol2008;323:293–314.

70. Kuhl U, Schultheiss HP. Viral myocarditis: diagnosis, aetiology and management.Drugs 2009;69:1287–1302.

71. Rose NR. Myocarditis: infection versus autoimmunity. J Clin Immunol 2009;29:730–737.

72. Kim KS, Tracy S, Tapprich W, Bailey J, Lee CK, Kim K, Barry WH, Chapman NM.5′-Terminal deletions occur in coxsackievirus B3 during replication in murinehearts and cardiac myocyte cultures and correlate with encapsidation ofnegative-strand viral RNA. J Virol 2005;79:7024–7041.

73. Okura Y, Yamamoto T, Goto S, Inomata T, Hirono S, Hanawa H, Feng L,Wilson C, Kihara T, Izumi T, Shibata A, Aizawa Y, Seki S, Abo T. Characterizationof cytokine and iNOS mRNA expression in situ during the course of experimen-tal autoimmune myocarditis in rats. J Mol Cell Cardiol 1997;29:491–502.

74. Kania G, Blyszczuk P, Stein S, Valaperti A, Germano D, Dirnhofer S, Hunziker L,Matter CM, Eriksson U. Heart-infiltrating prominin-1/CD133 progenitor cellsrepresent the cellular source of transforming growth factor b-mediatedcardiac fibrosis in experimental autoimmune myocarditis. Circ Res 2009;105:462–470.

75. Riad A, Westermann D, Escher F, Becher PM, Savvatis K, Lettau O,Heimesaat MM, Bereswill S, Volk HD, Schultheiss HP, Tschope C. Myeloid differ-entiation factor-88 contributes to TLR9-mediated modulation of acute coxsack-ievirus B3-induced myocarditis in vivo. Am J Physiol Heart Circ Physiol 2010;298:H2024–H2031.

76. Rutschow S, Li J, Schultheiss HP, Pauschinger M. Myocardial proteases andmatrix remodeling in inflammatory heart disease. Cardiovasc Res 2006;69:646–656.

77. Frisancho-Kiss S, Coronado M, Frisancho J, Lau V, Rose N, Klein S,Fairweather D. Gonadectomy of male BALB/c mice increases Tim-3(+) alterna-tively activated M2 macrophages, Tim-3(+) T cells, Th2 cells and Treg in theheart during acute coxsackievirus-induced myocarditis. Brain Behav Immun2009;23:649–657.

78. Fairweather D, Frisancho-Kiss S, Yusung S, Barrett M, Davis S, Gatewood S,Njoku D, Rose N. Interferon-gamma protects against chronic viral myocarditisby reducing mast cell degranulation, fibrosis, and the profibrotic cytokines trans-forming growth factor-beta 1, interleukin-1 beta, and interleukin-4 in the heart.Am J Pathol 2004;165:1883–1894.

79. Eriksson U, Ricci R, Hunziker L, Kurrer MO, Oudit GY, Watts TH,Sonderegger I, Bachmaier K, Kopf M, Penninger JM. Dendritic cell-induced auto-immune heart failure requires cooperation between adaptive and innate immu-nity. Nat Med 2003;9:1484–1490.

80. Rangachari M, Mauermann N, Marty RR, Dirnhofer S, Kurrer MO,Komnenovic V, Penninger JM, Eriksson U. T-bet negatively regulates auto-immune myocarditis by suppressing local production of interleukin 17. J ExpMed 2006;205:2009–2019.

81. Valaperti A, Marty RR, Kania G, Germano D, Mauermann N, Dirnhofer S,Leimenstoll B, Blyszczuk P, Dong C, Mueller C, Lukas Hunziker L, Eriksson U.CD11b monocytes abrogate Th17 CD4 T cell-mediated experimental auto-immune myocarditis. J Immunol 2008;180:2686–2695.

82. Amabile N, Fraisse A, Bouvenot J, Chetaille P, Ovaert C. Outcome of acute ful-minant myocarditis in children. Heart 2006;92:1269–1273.

83. Cocker MS, Abdel-Aty H, Strohm O, Friedrich MG. Age and gender effects onthe extent of myocardial involvement in acute myocarditis: a cardiovascular mag-netic resonance study. Heart 2009;95:1925–1930.

84. Mason JW, O’Connell JB, Herskowitz A, Rose NR, McManus BM, Billingham ME,Moon TE. A clinical trial of immunosuppressive therapy for myocarditis. TheMyocarditis Treatment Trial Investigators. N Engl J Med 1995;333:269–275.

H.-P. Schultheiss et al.2625aD

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

85. Grimm W, Glaveris C, Hoffmann J, Menz V, Muller HH, Hufnagel G, Maisch B.Arrhythmia risk stratification in idiopathic dilated cardiomyopathy based onechocardiography and 12-lead, signal-averaged, and 24-hour holter electrocar-diography. Am Heart J 2000;140:43–51.

86. Soongswang J, Durongpisitkul K, Nana A, Laohaprasittiporn D, Kangkagate C,Punlee K, Limpimwong N. Cardiac troponin T: a marker in the diagnosis ofacute myocarditis in children. Pediatr Cardiol 2005;26:45–49.

87. Smith SC, Ladenson JH, Mason JW, Jaffe AS. Elevations of cardiac troponin Iassociated with myocarditis. Experimental and clinical correlates. Circulation1997;95:163–168.

88. Franz WM, Remppis A, Kandolf R, Kubler W, Katus HA. Serum troponin T: diag-nostic marker for acute myocarditis. Clin Chem 1996;42:340–341.

89. Lauer B, Niederau C, Kuhl U, Schannwell M, Pauschinger M, Strauer BE,Schultheiss HP. Cardiac troponin T in patients with clinically suspected myocar-ditis. J Am Coll Cardiol 1997;30:1354–1359.

90. Morgera T, Di Lenarda A, Dreas L, Pinamonti B, Humar F, Bussani R, Silvestri F,Chersevani D, Camerini F. Electrocardiography of myocarditis revisited: clinicaland prognostic significance of electrocardiographic changes. Am Heart J 1992;124:455–467.

91. Vignola PA, Aonuma K, Swaye PS, Rozanski JJ, Blankstein RL, Benson J,Gosselin AJ, Lister JW. Lymphocytic myocarditis presenting as unexplained ven-tricular arrhythmias: diagnosis with endomyocardial biopsy and response toimmunosuppression. J Am Coll Cardiol 1984;4:812–819.

92. Dec GW Jr, Waldman H, Southern J, Fallon JT, Hutter AM Jr, Palacios I. Viralmyocarditis mimicking acute myocardial infarction. J Am Coll Cardiol 1992;20:85–89.

93. Magnani JW, Danik HJ, Dec GW Jr, DiSalvo TG. Survival in biopsy-proven myo-carditis: a long-term retrospective analysis of the histopathologic, clinical, andhemodynamic predictors. Am Heart J 2006;151:463–470.

94. Nakashima H, Katayama T, Ishizaki M, Takeno M, Honda Y, Yano K. Q wave andnon-Q wave myocarditis with special reference to clinical significance. Jpn Heart J1998;39:763–774.

95. Greenwood RD, Nadas AS, Fyler DC. The clinical course of primary myocardialdisease in infants and children. Am Heart J 1976;92:549–560.

96. Pinamonti B, Alberti E, Cigalotto A, Dreas L, Salvi A, Silvestri F, Camerini F.Echocardiographic findings in myocarditis. Am J Cardiol 1988;62:285–291.

97. Escher F, Westermann D, Gaub R, Pronk J, Bock T, Al-Saadi N, Kuhl U,Schultheiss HP, Tschope C. Development of diastolic heart failure in a 6-yearfollow-up study in patients after acute myocarditis. Heart 2011;97:709–714.

98. Angelini A, Calzolari V, Calabrese F, Boffa GM, Maddalena F, Chioin R, Thiene G.Myocarditis mimicking acute myocardial infarction: role of endomyocardialbiopsy in the differential diagnosis. Heart 2000;84:245–250.

99. Felker GM, Boehmer JP, Hruban RH, Hutchins GM, Kasper EK, Baughman KL,Hare JM. Echocardiographic findings in fulminant and acute myocarditis. J AmColl Cardiol 2000;36:227–232.

100. Mendes LA, Dec GW, Picard MH, Palacios IF, Newell J, Davidoff R. Right ventri-cular dysfunction: an independent predictor of adverse outcome in patients withmyocarditis. Am Heart J 1994;128:301–307.

101. Friedrich MG, Sechtem U, Schulz-Menger J, Holmvang G, Alakija P, Cooper LT,White JA, Abdel-Aty H, Gutberlet M, Prasad S, Aletras A, Laissy JP, Paterson I,Filipchuk NG, Kumar A, Pauschinger M, Liu P. Cardiovascular magnetic reson-ance in myocarditis: A JACC White Paper. J Am Coll Cardiol 2009;53:1475–1487.

102. Baccouche H, Mahrholdt H, Meinhardt G, Merher R, Voehringer M, Hill S,Klingel K, Kandolf R, Sechtem U, Yilmaz A. Diagnostic synergy of non-invasivecardiovascular magnetic resonance and invasive endomyocardial biopsy introponin-positive patients without coronary artery disease. Eur Heart J 2009;30:2869–2879.

103. Holzmann M, Nicko A, Kuhl U, Noutsias M, Poller W, Hoffmann W, Morguet A,Witzenbichler B, Tschoepe C, Schultheiss HP, Pauschinger M. Complication ratein right ventricular endomyocardial biopsy—a retro- and prospective study overa 11 year period analyzing 3048 diagnostic procedures. Circulation 2008;118:667–671.

104. Cooper LT, Baughman KL, Feldman AM, Frustaci A, Jessup M, Kuhl U,Levine GN, Narula J, Starling RC, Towbin J, Virmani R. The role of endomyocar-dial biopsy in the management of cardiovascular disease: a scientific statementfrom the American Heart Association, the American College of Cardiology,and the European Society of Cardiology. Endorsed by the Heart FailureSociety of America and the Heart Failure Association of the European Societyof Cardiology. J Am Coll Cardiol 2007;50:1914–1931.

105. Frustaci A, Russo MA, Chimenti C. Randomized study on the efficacy of immu-nosuppressive therapy in patients with virus-negative inflammatory cardiomyo-pathy: the TIMIC study. Circulation 2008;118:667–671.

106. Hunt SA, Abraham WT, Chin MH, Feldman AM, Francis GS, Ganiats TG,Jessup M, Konstam MA, Mancini DM, Michl K, Oates JA, Rahko PS, Silver MA,Stevenson LW, Yancy CW. 2009 Focused update incorporated into the ACC/

AHA 2005 Guidelines for the Diagnosis and Management of Heart Failure inAdults A Report of the American College of Cardiology Foundation/AmericanHeart Association Task Force on Practice Guidelines Developed in Collabor-ation With the International Society for Heart and Lung Transplantation. J AmColl Cardiol 2009;53:e1–e90.

107. Lindenfeld J, Albert NM, Boehmer JP, Collins SP, Ezekowitz JA, Givertz MM,Katz SD, Klapholz M, Moser DK, Rogers JG, Starling RC, Stevenson WG,Tang WH, Teerlink JR, Walsh MN. HFSA 2010 Comprehensive Heart FailurePractice Guideline. J Card Fail 16:e1–194.

108. Dickstein K, Cohen-Solal A, Filippatos G, McMurray JJ, Ponikowski P,Poole-Wilson PA, Stromberg A, van Veldhuisen DJ, Atar D, Hoes AW,Keren A, Mebazaa A, Nieminen M, Priori SG, Swedberg K, Vahanian A,Camm J, De Caterina R, Dean V, Funck-Brentano C, Hellemans I,Kristensen SD, McGregor K, Sechtem U, Silber S, Tendera M, Widimsky P,Zamorano JL. ESC Guidelines for the diagnosis and treatment of acute andchronic heart failure 2008: the Task Force for the Diagnosis and Treatment ofAcute and Chronic Heart Failure 2008 of the European Society of Cardiology.Developed in collaboration with the Heart Failure Association of the ESC(HFA) and endorsed by the European Society of Intensive Care Medicine(ESICM). Eur Heart J 2008;29:2388–2442.

109. Rezkalla SH, Raikar S, Kloner RA. Treatment of viral myocarditis with focus oncaptopril. Am J Cardiol 1996;77:634–637.

110. Saegusa S, Fei Y, Takahashi T, Sumino H, Moriya J, Kawaura K, Yamakawa J,Itoh T, Morimoto S, Nakahashi T, Iwai K, Matsumoto M, Kanda T. Oral admin-istration of candesartan improves the survival of mice with viral myocarditisthrough modification of cardiac adiponectin expression. Cardiovasc Drugs Ther2007;21:155–160.

111. Costanzo-Nordin MR, Reap EA, JB OC, Robinson JA, Scanlon PJ. A nonsteroidanti-inflammatory drug exacerbates Coxsackie B3 murine myocarditis. J Am CollCardiol 1985;6:1078–1082.

112. Schultheiss HP, Kuehl U. Cardiovascular viral infections. In: Lennette EH (ed),Lennette’s Laboratory Diagnosis of Viral Infections, Chapter 18. 4th ed. New York:Informa Healthcare USA Inc; 2010, p304–318.

113. Kuhl U, Pauschinger M, Schwimmbeck PL, Seeberg B, Lober C, Noutsias M,Poller W, Schultheiss HP. Interferon-beta treatment eliminates cardiotropicviruses and improves left ventricular function in patients with myocardial persist-ence of viral genomes and left ventricular dysfunction. Circulation 2003;107:2793–2798.

114. Schmidt-Lucke C, Spillmann F, Bock T, Van Linthout S, Kuhl U, Schultheiss HP,Tschoepe C. Interferon-beta modulates endothelial damage in patients withcardiac persistance of parvovirus B19V. J Infect Dis 2010;201:936–945.

115. Schultheiss HP, Piper C, Sowade K, Karason JF, Kapp G, Groetzbach F,Waagstein E, Arbustini E, Siedentop H, Kuehl U. The effect of subcutaneoustreatment with interferon-beta-1b over 24 weeks on safety, virus eliminationand clinical outcome in patients with chronic viral cardiomyopathy. Eur Heart J2009;30:1995–2002.

116. Frustaci A, Russo MA, Chimenti C. Randomized study on the efficacy of immu-nosuppressive therapy in patients with virus-negative inflammatory cardiomyo-pathy: the TIMIC study. Eur Heart J 2009;30:1995–2002.

117. Kuhl U, Strauer BE, Schultheiss HP. Methylprednisolone in chronic myocarditis.Postgrad Med J 1994;70:S35–S42.

118. Wojnicz R, Nowalany-Kozielska E, Wojciechowska C, Glanowska G,Wilczewski P, Niklewski T, Zembala M, Polonski L, Rozek MM, Wodniecki J.Randomized, placebo-controlled study for immunosuppressive treatment ofinflammatory dilated cardiomyopathy: two-year follow-up results. Circulation2001;104:39–45.

119. Frustaci A, Chimenti C, Calabrese F, Pieroni M, Thiene G, Maseri A. Immunosup-pressive therapy for active lymphocytic myocarditis: virological and immunologicprofile of responders versus nonresponders. Circulation 2003;107:857–863.

120. Stanton C, Mookadam F, Cha S, McNamara D, Aukrust P, Wojnicz R, Bailey KR,Cooper LT. Greater symptom duration predicts response to immunomodula-tory therapy in dilated cardiomyopathy. Int J Cardiol 2008;128:38–41.

121. Noutsias M, Seeberg B, Schultheiss HP, Kuhl U. Expression of cell adhesion mol-ecules in dilated cardiomyopathy: evidence for endothelial activation in inflam-matory cardiomyopathy. Circulation 1999;99:2124–2131.

122. Drucker NA, Colan SD, Lewis AB, Beiser AS, Wessel DL, Takahashi M,Baker AL, Perez-Atayde AR, Newburger JW. Gamma-globulin treatment ofacute myocarditis in the pediatric population. Circulation 1994;89:252–257.

123. English RF, Janosky JE, Ettedgui JA, Webber SA. Outcomes for children withacute myocarditis. Cardiol Young 2004;14:488–493.

124. Klugman D, Berger JT, Sable CA, He J, Khandelwal SG, Slonim AD. Pediatricpatients hospitalized with myocarditis: a multi-institutional analysis. PediatrCardiol 2009;31:222–228.

125. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP,Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB Jr,

The management of myocarditis 2625bD

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021

Reisman MA, Schaer GL, Sherman W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymalstem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol2009;54:2277–2286.

126. Yerebakan C, Kaminski A, Liebold A, Steinhoff G. Safety of intramyocardial stemcell therapy for the ischemic myocardium: results of the Rostock trial after5-year follow-up. Cell Transplant 2007;16:935–940.

127. Li JH, Zhang N, Wang JA. Improved anti-apoptotic and anti-remodeling potencyof bone marrow mesenchymal stem cells by anoxic pre-conditioning in diabeticcardiomyopathy. J Endocrinol Invest 2008;31:103–110.

128. Ohnishi S, Sumiyoshi H, Kitamura S, Nagaya N. Mesenchymal stem cells attenu-ate cardiac fibroblast proliferation and collagen synthesis through paracrineactions. FEBS Lett 2007;581:3961–3966.

129. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, Zhao RC, Shi Y. Mesench-ymal stem cell-mediated immunosuppression occurs via concerted action ofchemokines and nitric oxide. Cell Stem Cell 2008;2:141–150.

130. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P,Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli.Blood 2002;99:3838–3843.

131. Plumas J, Chaperot L, Richard MJ, Molens JP, Bensa JC, Favrot MC. Mesenchymalstem cells induce apoptosis of activated T cells. Leukemia 2005;19:1597–1604.

132. Di Ianni M, Del Papa B, De Ioanni M, Moretti L, Bonifacio E, Cecchini D,Sportoletti P, Falzetti F, Tabilio A. Mesenchymal cells recruit and regulate T regu-latory cells. Exp Hematol 2008;36:309–318.

133. Oh I, Ozaki K, Sato K, Meguro A, Tatara R, Hatanaka K, Nagai T, Muroi K,Ozawa K. Interferon-gamma and NF-kappaB mediate nitric oxide productionby mesenchymal stromal cells. Biochem Biophys Res Commun 2007;355:956–962.

134. Sheng H, Wang Y, Jin Y, Zhang Q, Zhang Y, Wang L, Shen B, Yin S, Liu W, Cui L,Li N. A critical role of IFNgamma in priming MSC-mediated suppression of T cellproliferation through up-regulation of B7-H1. Cell Res 2008;18:846–857.

135. Zell R, Markgraf R, Schmidtke M, Gorlach M, Stelzner A, Henke A, Sigusch HH,Gluck B. Nitric oxide donors inhibit the coxsackievirus B3 proteinases 2A and3C in vitro, virus production in cells, and signs of myocarditis in virus-infectedmice. Med Microbiol Immunol 2004;193:91–100.

136. Kuhl U, Pauschinger M, Schwimmbeck PL, Seeberg B, Lober C, Noutsias M,Poller W, Schultheiss HP. Interferon-beta treatment eliminates cardiotropicviruses and improves left ventricular function in patients with myocardial persist-ence of viral genomes and left ventricular dysfunction. Circulation 2003;107:2793–2798.

137. Pinkert S, Westermann D, Wang X, Klingel K, Dorner A, Savvatis K, Grossl T,Krohn S, Tschope C, Zeichhardt H, Kotsch K, Weitmann K, Hoffmann W,Schultheiss HP, Spiller OB, Poller W, Fechner H. Prevention of cardiac dysfunc-tion in acute coxsackievirus B3 cardiomyopathy by inducible expression of asoluble coxsackievirus-adenovirus receptor. Circulation 2009;120:2358–2366.

H.-P. Schultheiss et al.2625cD

ownloaded from

https://academic.oup.com

/eurheartj/article/32/21/2616/439730 by guest on 18 Decem

ber 2021