Randomised controlled trial into the role of ramipril in fibrosis reduction in rheumatic heart disease: the RamiRHeD trial protocol

Ade Meidian Ambari, Budhi Setianto, Anwar Santoso, Basuni Radi, Bambang Dwiputra, Eliana Susilowati, Fadilla Tulrahmi, Annemiek Wind, Maarten Jan Maria Cramer, Pieter Doevendans, Ade Meidian Ambari, Budhi Setianto, Anwar Santoso, Basuni Radi, Bambang Dwiputra, Eliana Susilowati, Fadilla Tulrahmi, Annemiek Wind, Maarten Jan Maria Cramer, Pieter Doevendans

Abstract

Introduction: Rheumatic heart disease (RHD) is a major burden in developing countries and accounts for 80% of all people living with the disease, where it causes most cardiovascular morbidity and mortality in children and young adults. Chronic inflammation and fibrosis of heart valve tissue due to chronic inflammation in RHD will cause calcification and thickening of the impacted heart valves, especially the mitral valve. This fibrogenesis is enhanced by the production of angiotensin II by increased transforming growth factor β expression and later by the binding of interleukin-33, which is known to have antihypertrophic and antifibrotic effects, to soluble sST2. sST2 binding to this non-natural ligand worsens fibrosis. Therefore, we hypothesise that ACE inhibitors (ACEIs) would improve rheumatic mitral valve stenosis.

Methods and analysis: This is a single-centre, double-blind, placebo-controlled, randomised clinical trial with a pre-post test design. Patients with rheumatic mitral stenosis and valve dysfunction will be planned for cardiac valve replacement operation and will be given ramipril 5 mg or placebo for a minimum of 12 weeks before the surgery. The expression of ST2 in the mitral valve is considered to be representative of cardiac fibrosis. Mitral valve tissue will be stained by immunohistochemistry to ST2. Plasma ST2 will be measured by ELISA. This study is conducted in the Department of Cardiology and Vascular Medicine, Universitas Indonesia, National Cardiac Center Harapan Kita Hospital, Jakarta, Indonesia, starting on 27 June 2019.

Ethics and dissemination: The performance and dissemination of this study were approved by the ethics committee of National Cardiovascular Center Harapan Kita with ethical code LB.02.01/VII/286/KEP.009/2018.

Trial registration number: NCT03991910.

Keywords: cardiology; cardiothoracic surgery; valvular heart disease.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Hypothesis. Molecular mimicry is a defence mechanism of group A Streptococcus to avoid immune cells. This mechanism allows immune cells to generate autoimmunity against protein the lining of endothelial cells and causing chronic inflammation and valvular damage. Continuous process of chronic inflammation leads to valvular thickening and fibrosis, which is mediated by the angiotensin II. Angiotensin II increase TGF-β expression and cause IL-33 to bind with sST2, and subsequently cause damage and fibrosis to the valvular tissue even more, which later will end with rheumatic heart failure. ACEI is hypothesised to counteract these processes by decreasing angiotensin II conversion from angiotensin I. ACEI, ACE inhibitor; IL, interleukin; TGF-β, transforming growth factor β.
Figure 2
Figure 2
Research flowchart. TGF-β, transforming growth factor β.

References

    1. Rodriguez-Fernandez R, Amiya R, Wyber R, et al. . Rheumatic heart disease among adults in a mining community of Papua, Indonesia: findings from an occupational cohort. Heart Asia 2015;7:44–8. 10.1136/heartasia-2015-010641
    1. Ambari AM, Setianto B, Santoso A, et al. . Survival analysis of patients with rheumatic MS after PBMV compared with MVS in a low-to-middle-income country. Neth Heart J 2019;27:559–64. 10.1007/s12471-019-01315-x
    1. Sharma N, Toor D. Interleukin-10: role in increasing susceptibility and pathogenesis of rheumatic fever/rheumatic heart disease. Cytokine 2017;90:169–76. 10.1016/j.cyto.2016.11.010
    1. Bilik MZ, Kaplan İbrahim, Polat N, et al. . Serum levels of IL-17 and IL-23 in patients with rheumatic mitral stenosis. Medicine 2016;95:e3562. 10.1097/MD.0000000000003562
    1. Ramona J, Alexander von M, Martin F, et al. . Soluble ST2 - A Potential Biomarker of Rheumatic Heart Disease. Clin Med Rev Case Rep 2019;6. 10.23937/2378-3656/1410255
    1. Gardezi SK, Coffey S, Prendergast BD, et al. . Serum biomarkers in valvular heart disease. Heart 2018;104:349–58. 10.1136/heartjnl-2016-310482
    1. Januzzi JL. St2 as a cardiovascular risk biomarker: from the bench to the bedside. J Cardiovasc Transl Res 2013;6:493–500. 10.1007/s12265-013-9459-y
    1. Ayoub S, Ferrari G, Gorman RC, et al. . Heart valve biomechanics and underlying mechanobiology. Compr Physiol 2016;6:1743–80. 10.1002/cphy.c150048
    1. Hus-Citharel A, Bouby N, Iturrioz X, et al. . Multiple cross talk between angiotensin II, bradykinin, and insulin signaling in the cortical thick ascending limb of rat kidney. Endocrinology 2010;151:3181–94. 10.1210/en.2009-1237
    1. Ambari AM, Setianto B, Santoso A, et al. . Angiotensin converting enzyme inhibitors (ACEIs) decrease the progression of cardiac fibrosis in rheumatic heart disease through the inhibition of IL-33/sST2. Front Cardiovasc Med 2020;7:115. 10.3389/fcvm.2020.00115
    1. Chockalingam A, Venkatesan S, Dorairajan S, et al. . Safety and efficacy of enalapril in multivalvular heart disease with significant mitral stenosis--SCOPE-MS. Angiology 2005;56:151–8. 10.1177/000331970505600205
    1. Marzullo A, Ambrosi F, Inchingolo M, et al. . ST2L transmembrane receptor expression: an immunochemical study on endarterectomy samples. PLoS One 2016;11:e0156315–12. 10.1371/journal.pone.0156315
    1. Louangrath PI. Sample size determination for Non-Finite population. SSRN Electron J 2017:1–12. 10.13140/2.1.4785.7286
    1. Reményi B, Wilson N, Steer A, et al. . World Heart Federation criteria for echocardiographic diagnosis of rheumatic heart disease--an evidence-based guideline. Nat Rev Cardiol 2012;9:297–309. 10.1038/nrcardio.2012.7
    1. Kakkar R, Lee RT. The IL-33/ST2 pathway: therapeutic target and novel biomarker. Nat Rev Drug Discov 2008;7:827–40. 10.1038/nrd2660
    1. Miller AM. Role of IL-33 in inflammation and disease. J Inflamm 2011;8:22. 10.1186/1476-9255-8-22
    1. Schmitz J, Owyang A, Oldham E, et al. . Il-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005;23:479–90. 10.1016/j.immuni.2005.09.015
    1. Sanada S, Hakuno D, Higgins LJ, et al. . Il-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest 2007;117:1538–49. 10.1172/JCI30634
    1. Hayakawa H, Hayakawa M, Kume A, et al. . Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation. J Biol Chem 2007;282:26369–80. 10.1074/jbc.M704916200
    1. Martinez-Rumayor A, Camargo CA, Green SM, et al. . Soluble ST2 plasma concentrations predict 1-year mortality in acutely dyspneic emergency department patients with pulmonary disease. Am J Clin Pathol 2008;130:578–84. 10.1309/WMG2BFRC97MKKQKP
    1. Becerra A, Warke RV, Xhaja K, et al. . Increased activity of indoleamine 2,3-dioxygenase in serum from acutely infected dengue patients linked to gamma interferon antiviral function. J Gen Virol 2009;90:810–7. 10.1099/vir.0.004416-0
    1. Brunner M, Krenn C, Roth G, et al. . Increased levels of soluble ST2 protein and IgG1 production in patients with sepsis and trauma. Intensive Care Med 2004;30:1468–73. 10.1007/s00134-004-2184-x
    1. Bartunek J, Delrue L, Van Durme F, et al. . Nonmyocardial production of ST2 protein in human hypertrophy and failure is related to diastolic load. J Am Coll Cardiol 2008;52:2166–74. 10.1016/j.jacc.2008.09.027
    1. Szerafin T, Niederpold T, Mangold A, et al. . Secretion of soluble ST2 - possible explanation for systemic immunosuppression after heart surgery. Thorac Cardiovasc Surg 2009;57:25–9. 10.1055/s-2008-1039044
    1. Sabatine MS, Morrow DA, Higgins LJ, et al. . Complementary roles for biomarkers of biomechanical strain ST2 and N-terminal prohormone B-type natriuretic peptide in patients with ST-elevation myocardial infarction. Circulation 2008;117:1936–44. 10.1161/CIRCULATIONAHA.107.728022
    1. Mueller T, Dieplinger B, Gegenhuber A, et al. . Increased plasma concentrations of soluble ST2 are predictive for 1-year mortality in patients with acute destabilized heart failure. Clin Chem 2008;54:752–6. 10.1373/clinchem.2007.096560
    1. Shimpo M, Morrow DA, Weinberg EO, et al. . Serum levels of the interleukin-1 receptor family member ST2 predict mortality and clinical outcome in acute myocardial infarction. Circulation 2004;109:2186–90. 10.1161/01.CIR.0000127958.21003.5A
    1. Jao G, Lystash J, Sane D. Angiotensin-Converting enzyme inhibitors can increase the transvalvular gradient among patients with aortic stenosis. J Am Coll Cardiol 2012;59:777. 10.1016/j.jacc.2011.10.892
    1. Arat-Ozkan A, Kaya A, Yigit Z, et al. . Serum N-terminal Pro-BNP levels correlate with symptoms and echocardiographic findings in patients with mitral stenosis. Echocardiography 2005;22:473–8. 10.1111/j.1540-8175.2005.04085.x
    1. Iltumur K, Karabulut A, Yokus B, et al. . N-Terminal proBNP plasma levels correlate with severity of mitral stenosis. J Heart Valve Dis 2005;14:735–41.

Source: PubMed

3
订阅