Skip main navigation

We See Only What We Look For

Imaging Cardiac Inflammation
Originally publishedhttps://doi.org/10.1161/CIRCIMAGING.113.000166Circulation: Cardiovascular Imaging. 2013;6:165–166

    Myocarditis is responsible for a substantial minority of cases of sudden death and nonischemic dilated cardiomyopathy. In clinical case series of sudden death, myocarditis is often the third leading cause after hypertrophic cardiomyopathy and congenital and atherosclerotic coronary artery disease.1 In autopsy studies of young adults, myocarditis is responsible for 5% to 20% of sudden deaths.2 Nine percent to 40% of acute nonischemic cardiomyopathy cases are a result of myocarditis, a rate that varies depending on the histological or clinical diagnostic criteria used.3 A recent analysis based on hospital dismissal International Classification of Diseases, Ninth Revision, codes estimated that between 0.5% and 4% of prevalent heart failure is caused by myocarditis.4 This value is likely an underestimate because the diagnostic test, endomyocardial biopsy, is not widely performed outside of tertiary care medical centers.

    Article see p 277

    Timely diagnosis of myocarditis can refine estimates of cardiac risk and permit tailored treatment for heart failure. Several studies suggest that more severe myocardial inflammation predicts progression to dilated cardiomyopathy, increased risk of arrhythmias, and chronic heart failure.57 However, the cost, risks, and lack of availability of endomyocardial biopsy limit its widespread clinical use. For epidemiological studies, diagnostic criteria that rely on clinical syndromes, biomarkers, and imaging abnormalities have been proposed, but these criteria sacrifice diagnostic certainty.8 There are no clinically available and highly specific biomarkers or imaging tests that identify the histological type or quantify the severity of myocardial inflammation.

    Past efforts to diagnose myocarditis noninvasively used various features of myocardial damage related to the inflammatory response. In the late 1980s, antimyosin scintigraphy to detect myocyte necrosis had a high sensitivity but low specificity for myocarditis.9 Echocardiography established the prognostic significance of left and right ventricular failure but never challenged the histological gold standard. Imaging has been limited in part because most cases of clinical myocarditis have a short duration or minor degree of myocyte damage. Cardiac magnetic resonance imaging using a combination of T2, early T1, and late T1 sequences achieved a sensitivity and specificity of ≈80%, but only in recent-onset cases.10 Although epicardial and midmyocardial patterns of signal abnormality suggest a postviral myocarditis or nonischemic scar, specific causes and cellular types such as giant-cell or eosinophilic myocarditis cannot be identified by magnetic resonance imaging or even fluoro-D-glucose (18FDG)–positron emission tomography.

    In this issue of Circulation: Cardiovascular Imaging, the study by van Heeswijk et al11 overcomes previous challenges by demonstrating that intravenous injection of perfluorocarbons (PFCs) is taken up by inflammatory cells in the heart and can be detected with 9.4-T cardiac magnetic resonance imaging in a male mouse model of autoimmune myocarditis. If this method can be applied successfully to clinical disease, it could allow widespread and earlier detection and a better understanding of the role of inflammation in the development of chronic dilated cardiomyopathy. Interestingly, PFCs were detected only in macrophages, granulocytes, and dendritic cells, not in lymphocytes. Yet, the signal from the PFCs correlated with the severity of myocardial inflammation. Although studies of myocarditis have focused on the role of lymphocytes in disease pathogenesis, and specifically on T-helper responses, the majority of cells present during acute myocarditis in autoimmune and viral animal models of myocarditis are monocyte/macrophages and granulocytes.12,13 For this reason, the technique presented by van Heeswijk et al holds particular promise for clinical diagnosis.

    Quite a few issues remain to be addressed before 19F imaging will prove clinically useful. In an experimental setting, can PFCs detect inflammation in female mice, which have more lymphocytes, as well as it does in males, which have far more severe inflammation composed primarily of macrophages and granulocytes?12,14 Will this method work as well in other strains of mice or differ between patients susceptible or resistant to chronic dilated cardiomyopathy? The BALB/c mice used in these studies are susceptible to develop dilated cardiomyopathy but may accurately model only one type of myocarditis patient. Timing for the injection of PFCs may also be important. Some mouse models of myocarditis are biphasic, with inflammation disappearing quickly after peak inflammation. If this also occurs in patients with myocarditis, the timing of PCF injection may be critical. Ligands that can be detected with cardiac magnetic resonance imaging or positron emission tomography imaging will need to detect inflammation in >1 phase of the disease to be most useful as a diagnostic tool.

    Questions that will arise early in the clinical arena include the impact of coexisting conditions such as pericarditis and prior ischemic myocardial damage that may lower the specificity of 19F imaging for myocarditis. Common clinical 1.5-T or 3-T magnets may have lower spatial resolution for 19F than the 9.4-T research magnet used by van Heeswijk et al. The risks of 19F-PFC exposure in humans are not fully known. Recognizing these hurdles, the promise of a widely applicable and sensitive noninvasive test for myocardial inflammation is exciting and worth the attention of the entire myocarditis research community. We hope that the foundation provided by Heeswijk et al will open the door to an enduring stream of translational and clinical investigations.

    Footnotes

    The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.

    Correspondence to Leslie T. Cooper, Jr, MD, Consultant, Division of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905. E-mail

    References

    • 1. Maron BJ, Doerer JJ, Haas TS, Tierney DM, Mueller FO . Sudden deaths in young competitive athletes: analysis of 1866 deaths in the United States, 1980-2006.Circulation. 2009; 119:1085–1092.LinkGoogle Scholar
    • 2. Doolan A, Langlois N, Semsarian C . Causes of sudden cardiac death in young Australians.Med J Aust. 2004; 180:110–112.CrossrefMedlineGoogle Scholar
    • 3. Cooper LT . Myocarditis.N Engl J Med. 2009; 360:1526–1538.CrossrefMedlineGoogle Scholar
    • 4. Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, Shibuya K, Salomon JA, Abdalla S, Aboyans V, Abraham J, Ackerman I, Aggarwal R, Ahn SY, Ali MK, Alvarado M, Anderson HR, Anderson LM, Andrews KG, Atkinson C, Baddour LM, Bahalim AN, Barker-Collo S, Barrero LH, Bartels DH, Basáñez MG, Baxter A, Bell ML, Benjamin EJ, Bennett D, Bernabé E, Bhalla K, Bhandari B, Bikbov B, Bin Abdulhak A, Birbeck G, Black JA, Blencowe H, Blore JD, Blyth F, Bolliger I, Bonaventure A, Boufous S, Bourne R, Boussinesq M, Braithwaite T, Brayne C, Bridgett L, Brooker S, Brooks P, Brugha TS, Bryan-Hancock C, Bucello C, Buchbinder R, Buckle G, Budke CM, Burch M, Burney P, Burstein R, Calabria B, Campbell B, Canter CE, Carabin H, Carapetis J, Carmona L, Cella C, Charlson F, Chen H, Cheng AT, Chou D, Chugh SS, Coffeng LE, Colan SD, Colquhoun S, Colson KE, Condon J, Connor MD, Cooper LT, Corriere M, Cortinovis M, de Vaccaro KC, Couser W, Cowie BC, Criqui MH, Cross M, Dabhadkar KC, Dahiya M, Dahodwala N, Damsere-Derry J, Danaei G, Davis A, De Leo D, Degenhardt L, Dellavalle R, Delossantos A, Denenberg J, Derrett S, Des Jarlais DC, Dharmaratne SD, Dherani M, Diaz-Torne C, Dolk H, Dorsey ER, Driscoll T, Duber H, Ebel B, Edmond K, Elbaz A, Ali SE, Erskine H, Erwin PJ, Espindola P, Ewoigbokhan SE, Farzadfar F, Feigin V, Felson DT, Ferrari A, Ferri CP, Fèvre EM, Finucane MM, Flaxman S, Flood L, Foreman K, Forouzanfar MH, Fowkes FG, Franklin R, Fransen M, Freeman MK, Gabbe BJ, Gabriel SE, Gakidou E, Ganatra HA, Garcia B, Gaspari F, Gillum RF, Gmel G, Gosselin R, Grainger R, Groeger J, Guillemin F, Gunnell D, Gupta R, Haagsma J, Hagan H, Halasa YA, Hall W, Haring D, Haro JM, Harrison JE, Havmoeller R, Hay RJ, Higashi H, Hill C, Hoen B, Hoffman H, Hotez PJ, Hoy D, Huang JJ, Ibeanusi SE, Jacobsen KH, James SL, Jarvis D, Jasrasaria R, Jayaraman S, Johns N, Jonas JB, Karthikeyan G, Kassebaum N, Kawakami N, Keren A, Khoo JP, King CH, Knowlton LM, Kobusingye O, Koranteng A, Krishnamurthi R, Lalloo R, Laslett LL, Lathlean T, Leasher JL, Lee YY, Leigh J, Lim SS, Limb E, Lin JK, Lipnick M, Lipshultz SE, Liu W, Loane M, Ohno SL, Lyons R, Ma J, Mabweijano J, MacIntyre MF, Malekzadeh R, Mallinger L, Manivannan S, Marcenes W, March L, Margolis DJ, Marks GB, Marks R, Matsumori A, Matzopoulos R, Mayosi BM, McAnulty JH, McDermott MM, McGill N, McGrath J, Medina-Mora ME, Meltzer M, Mensah GA, Merriman TR, Meyer AC, Miglioli V, Miller M, Miller TR, Mitchell PB, Mocumbi AO, Moffitt TE, Mokdad AA, Monasta L, Montico M, Moradi-Lakeh M, Moran A, Morawska L, Mori R, Murdoch ME, Mwaniki MK, Naidoo K, Nair MN, Naldi L, Narayan KM, Nelson PK, Nelson RG, Nevitt MC, Newton CR, Nolte S, Norman P, Norman R, O’Donnell M, O’Hanlon S, Olives C, Omer SB, Ortblad K, Osborne R, Ozgediz D, Page A, Pahari B, Pandian JD, Rivero AP, Patten SB, Pearce N, Padilla RP, Perez-Ruiz F, Perico N, Pesudovs K, Phillips D, Phillips MR, Pierce K, Pion S, Polanczyk GV, Polinder S, Pope CA, Popova S, Porrini E, Pourmalek F, Prince M, Pullan RL, Ramaiah KD, Ranganathan D, Razavi H, Regan M, Rehm JT, Rein DB, Remuzzi G, Richardson K, Rivara FP, Roberts T, Robinson C, De Leòn FR, Ronfani L, Room R, Rosenfeld LC, Rushton L, Sacco RL, Saha S, Sampson U, Sanchez-Riera L, Sanman E, Schwebel DC, Scott JG, Segui-Gomez M, Shahraz S, Shepard DS, Shin H, Shivakoti R, Singh D, Singh GM, Singh JA, Singleton J, Sleet DA, Sliwa K, Smith E, Smith JL, Stapelberg NJ, Steer A, Steiner T, Stolk WA, Stovner LJ, Sudfeld C, Syed S, Tamburlini G, Tavakkoli M, Taylor HR, Taylor JA, Taylor WJ, Thomas B, Thomson WM, Thurston GD, Tleyjeh IM, Tonelli M, Towbin JA, Truelsen T, Tsilimbaris MK, Ubeda C, Undurraga EA, van der Werf MJ, van Os J, Vavilala MS, Venketasubramanian N, Wang M, Wang W, Watt K, Weatherall DJ, Weinstock MA, Weintraub R, Weisskopf MG, Weissman MM, White RA, Whiteford H, Wiersma ST, Wilkinson JD, Williams HC, Williams SR, Witt E, Wolfe F, Woolf AD, Wulf S, Yeh PH, Zaidi AK, Zheng ZJ, Zonies D, Lopez AD, Murray CJ . Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010.Lancet. 2012; 380:2163–2196.CrossrefMedlineGoogle Scholar
    • 5. Kindermann I, Kindermann M, Kandolf R, Klingel K, Bültmann B, Müller T, Lindinger A, Böhm M . Predictors of outcome in patients with suspected myocarditis.Circulation. 2008; 118:639–648.LinkGoogle Scholar
    • 6. Coronado MJ, Brandt JE, Kim E, Bucek A, Bedja D, Abston ED, Shin J, Gabrielson KL, Mitzner W, Fairweather D . Testosterone and interleukin-1β increase cardiac remodeling during coxsackievirus B3 myocarditis via serpin A 3n.Am J Physiol Heart Circ Physiol. 2012; 302:H1726–H1736.CrossrefMedlineGoogle Scholar
    • 7. Kühl U, Schultheiss HP . Myocarditis: early biopsy allows for tailored regenerative treatment.Dtsch Arztebl Int. 2012; 109:361–368.MedlineGoogle Scholar
    • 8. Sagar S, Liu PP, Cooper LT . Myocarditis.Lancet. 2012; 379:738–747.CrossrefMedlineGoogle Scholar
    • 9. Skouri HN, Dec GW, Friedrich MG, Cooper LT . Noninvasive imaging in myocarditis.J Am Coll Cardiol. 2006; 48:2085–2093.CrossrefMedlineGoogle Scholar
    • 10. 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 ; International Consensus Group on Cardiovascular Magnetic Resonance in Myocarditis. Cardiovascular magnetic resonance in myocarditis: a JACC White Paper.J Am Coll Cardiol. 2009; 53:1475–1487.CrossrefMedlineGoogle Scholar
    • 11. van Heeswijk RB, De Blois J, Kania G, Gonzales C, Blyszczuk P, Stuber M, Eriksson U, Schwitter J . Selective in vivo visualization of immune-cell infiltration in a mouse model of autoimmune myocarditis by fluorine-19 cardiac magnetic resonance.Circ Cardiovasc Imaging. 2013; 6:277–284.LinkGoogle Scholar
    • 12. Frisancho-Kiss S, Davis SE, Nyland JF, Frisancho JA, Cihakova D, Barrett MA, Rose NR, Fairweather D . Cutting edge: cross-regulation by TLR4 and T cell Ig mucin-3 determines sex differences in inflammatory heart disease.J Immunol. 2007; 178:6710–6714.CrossrefMedlineGoogle Scholar
    • 13. Cihakova D, Barin JG, Afanasyeva M, Kimura M, Fairweather D, Berg M, Talor MV, Baldeviano GC, Frisancho S, Gabrielson K, Bedja D, Rose NR . Interleukin-13 protects against experimental autoimmune myocarditis by regulating macrophage differentiation.Am J Pathol. 2008; 172:1195–1208.CrossrefMedlineGoogle Scholar
    • 14. Frisancho-Kiss S, Coronado MJ, Frisancho JA, Lau VM, Rose NR, Klein SL, Fairweather D . Gonadectomy of male BALB/c mice increases Tim-3(+) alternatively activated M2 macrophages, Tim-3(+) T cells, Th2 cells and Treg in the heart during acute coxsackievirus-induced myocarditis.Brain Behav Immun. 2009; 23:649–657.CrossrefMedlineGoogle Scholar