Rationale and design of the PHOspholamban RElated CArdiomyopathy intervention STudy (i-PHORECAST)

W P Te Rijdt, E T Hoorntje, R de Brouwer, A Oomen, A Amin, J F van der Heijden, J C Karper, B D Westenbrink, H H W Silljé, A S J M Te Riele, A C P Wiesfeld, I C van Gelder, T P Willems, P A van der Zwaag, J P van Tintelen, J H Hillege, H L Tan, D J van Veldhuisen, F W Asselbergs, R A de Boer, A A M Wilde, M P van den Berg, W P Te Rijdt, E T Hoorntje, R de Brouwer, A Oomen, A Amin, J F van der Heijden, J C Karper, B D Westenbrink, H H W Silljé, A S J M Te Riele, A C P Wiesfeld, I C van Gelder, T P Willems, P A van der Zwaag, J P van Tintelen, J H Hillege, H L Tan, D J van Veldhuisen, F W Asselbergs, R A de Boer, A A M Wilde, M P van den Berg

Abstract

Background: The p.Arg14del (c.40_42delAGA) phospholamban (PLN) pathogenic variant is a founder mutation that causes dilated cardiomyopathy (DCM) and arrhythmogenic cardiomyopathy (ACM). Carriers are at increased risk of malignant ventricular arrhythmias and heart failure, which has been ascribed to cardiac fibrosis. Importantly, cardiac fibrosis appears to be an early feature of the disease, occurring in many presymptomatic carriers before the onset of overt disease. As with most monogenic cardiomyopathies, no evidence-based treatment is available for presymptomatic carriers.

Aims: The PHOspholamban RElated CArdiomyopathy intervention STudy (iPHORECAST) is designed to demonstrate that pre-emptive treatment of presymptomatic PLN p.Arg14del carriers using eplerenone, a mineralocorticoid receptor antagonist with established antifibrotic effects, can reduce disease progression and postpone the onset of overt disease.

Methods: iPHORECAST has a multicentre, prospective, randomised, open-label, blinded endpoint (PROBE) design. Presymptomatic PLN p.Arg14del carriers are randomised to receive either 50 mg eplerenone once daily or no treatment. The primary endpoint of the study is a multiparametric assessment of disease progression including cardiac magnetic resonance parameters (left and right ventricular volumes, systolic function and fibrosis), electrocardiographic parameters (QRS voltage, ventricular ectopy), signs and/or symptoms related to DCM and ACM, and cardiovascular death. The follow-up duration is set at 3 years.

Baseline results: A total of 84 presymptomatic PLN p.Arg14del carriers (n = 42 per group) were included. By design, at baseline, all participants were in New York Heart Association (NHYA) class I and had a left ventricular ejection fraction > 45% and < 2500 ventricular premature contractions during 24-hour Holter monitoring. There were no statistically significant differences between the two groups in any of the baseline characteristics. The study is currently well underway, with the last participants expected to finish in 2021.

Conclusion: iPHORECAST is a multicentre, prospective randomised controlled trial designed to address whether pre-emptive treatment of PLN p.Arg14del carriers with eplerenone can prevent or delay the onset of cardiomyopathy. iPHORECAST has been registered in the clinicaltrials.gov-register (number: NCT01857856).

Keywords: Cardiomyopathy; Design; Intervention study; Phospholamban; Pre-emptive treatment; Presymptomatic carriers.

Conflict of interest statement

W.P. te Rijdt, E.T. Hoorntje, R. de Brouwer, A. Oomen, A. Amin, J.F. van der Heijden, J.C. Karper, B.D. Westenbrink, H.H.W. Silljé, A.S.J.M. te Riele, A.C.P. Wiesfeld, I.C. van Gelder, T.P. Willems, P.A. van der Zwaag, J.P. van Tintelen, J.H. Hillege, H.L. Tan, D.J. van Veldhuisen, F.W. Asselbergs, R.A. de Boer, A.A.M. Wilde and M.P. van den Berg declare that they have no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Function of phospholamban. Phospholamban (PLN; blue arrow) is a reversibly phosphorylated transmembrane protein that binds to and regulates the activity of the sarcoplasmic reticulum Ca2+ ATPase (SERCA2a) pump. From: MacLennan et al. Nat Rev Mol Cell Biol. 2003 [27]. Printed with permission from Springer Nature
Fig. 2
Fig. 2
Left ventricular delayed contrast enhancement in PLN p.Arg14del carrier. Example of left lateral delayed contrast enhancement (arrow; short axis CMR image) in the left ventricle of PLN p.Arg14del carrier. CMR cardiac magnetic resonance, PLN phospholamban

References

    1. van der Zwaag PA, van Rijsingen IA, Asimaki A, et al. Phospholamban R14del mutation in patients diagnosed with dilated cardiomyopathy or arrhythmogenic right ventricular cardiomyopathy: evidence supporting the concept of arrhythmogenic cardiomyopathy. Eur J Heart Fail. 2012;14:1199–1207. doi: 10.1093/eurjhf/hfs119.
    1. van der Zwaag PA, Cox MG, van der Werf C, et al. Recurrent and founder mutations in the Netherlands : plakophilin-2 p.Arg79X mutation causing arrhythmogenic right ventricular cardiomyopathy/dysplasia. Neth Heart J. 2010;18:583–591. doi: 10.1007/s12471-010-0839-5.
    1. van Rijsingen IA, van der Zwaag PA, Groeneweg JA, et al. Outcome in phospholamban R14del carriers: results of a large multicentre cohort study. Circ Cardiovasc Genet. 2014;7:455–465. doi: 10.1161/CIRCGENETICS.113.000374.
    1. Milano A, Blom MT, Lodder EM, et al. Sudden cardiac arrest and rare genetic variants in the community. Circ Cardiovasc Genet. 2016;9:147–153. doi: 10.1161/CIRCGENETICS.115.001263.
    1. Hof IE, van der Heijden JF, Kranias EG, et al. Prevalence and cardiac phenotype of patients with a phospholamban mutation. Neth Heart J. 2019;27:64–69. doi: 10.1007/s12471-018-1211-4.
    1. Haghighi K, Kolokathis F, Gramolini AO, et al. A mutation in the human phospholamban gene, deleting arginine 14, results in lethal, hereditary cardiomyopathy. Proc Natl Acad Sci U S A. 2006;103:1388–1393. doi: 10.1073/pnas.0510519103.
    1. Eijgenraam TR, Boukens BJ, Boogerd CJ, et al. The phospholamban p.(Arg14del) pathogenic variant leads to cardiomyopathy with heart failure and is unreponsive to standard heart failure therapy. Sci Rep. 2020;10:9819. doi: 10.1038/s41598-020-66656-9.
    1. Posch MG, Perrot A, Geier C, et al. Genetic deletion of arginine 14 in phospholamban causes dilated cardiomyopathy with attenuated electrocardiographic R amplitudes. Heart Rhythm. 2009;6:480–486. doi: 10.1016/j.hrthm.2009.01.016.
    1. Te Rijdt WP, Ten Sande JN, Gorter TM, et al. Myocardial fibrosis as an early feature in phospholamban p.Arg14del mutation carriers: phenotypic insights from cardiovascular magnetic resonance imaging. Eur Heart J Cardiovasc Imaging. 2018;20:92–100. doi: 10.1093/ehjci/jey047.
    1. Chai W, Danser AH. Why are mineralocorticoid receptor antagonists cardioprotective? Naunyn Schmiedebergs Arch Pharmacol. 2006;374:153–162. doi: 10.1007/s00210-006-0107-9.
    1. Iraqi W, Rossignol P, Angioi M, et al. Extracellular cardiac matrix biomarkers in patients with acute myocardial infarction complicated by left ventricular dysfunction and heart failure: insights from the Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study (EPHESUS) study. Circulation. 2009;119:2471–2479. doi: 10.1161/CIRCULATIONAHA.108.809194.
    1. Nehme J, Mercier N, Labat C, et al. Differences between cardiac and arterial fibrosis and stiffness in aldosterone-salt rats: effect of eplerenone. J Renin Angiotensin Aldosterone Syst. 2006;7:31–39. doi: 10.3317/jraas.2006.004.
    1. Nishioka T, Suzuki M, Onishi K, et al. Eplerenone attenuates myocardial fibrosis in the angiotensin II-induced hypertensive mouse: involvement of tenascin-C induced by aldosterone-mediated inflammation. J Cardiovasc Pharmacol. 2007;49:261–268. doi: 10.1097/FJC.0b013e318033dfd4.
    1. Susic D, Varagic J, Ahn J, et al. Long-term mineralocorticoid receptor blockade reduces fibrosis and improves cardiac performance and coronary hemodynamics in elderly SHR. Am J Physiol Heart Circ Physiol. 2007;292:175–179. doi: 10.1152/ajpheart.00660.2006.
    1. Zannad F, Alla F, Dousset B, et al. Limitation of excessive extracellular matrix turnover may contribute to survival benefit of spironolactone therapy in patients with congestive heart failure: insights from the randomized aldactone evaluation study (RALES). Rales Investigators. Circulation. 2000;102:2700–2706. doi: 10.1161/01.CIR.102.22.2700.
    1. Zannad F, Radauceanu A. Effect of MR blockade on collagen formation and cardiovascular disease with a specific emphasis on heart failure. Heart Fail Rev. 2005;10:71–78. doi: 10.1007/s10741-005-2351-3.
    1. Bosman LP, Verstraelen TE, van Lint FHM, et al. The Netherlands arrhythmogenic cardiomyopathy registry: design and status update. Neth Heart J. 2019;27:480–486. doi: 10.1007/s12471-019-1270-1.
    1. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the Task Force Criteria. Eur Heart J. 2010;31:806–814. doi: 10.1093/eurheartj/ehq025.
    1. Mestroni L, Maisch B, McKenna WJ, et al. Guidelines for the study of familial dilated cardiomyopathies. Collaborative Research Group of the European Human and Capital Mobility Project on Familial Dilated Cardiomyopathy. Eur Heart J. 1999;20:93–102. doi: 10.1053/euhj.1998.1145.
    1. Hansson L, Hedner T, Dahlof B. Prospective randomized open blinded end-point (PROBE) study. A novel design for intervention trials. Prospective Randomized Open Blinded End-Point. Blood Press. 1992;1:113–119. doi: 10.3109/08037059209077502.
    1. Zannad F, Rossignol P, Iraqi W. Extracellular matrix fibrotic markers in heart failure. Heart Fail Rev. 2010;15:319–329. doi: 10.1007/s10741-009-9143-0.
    1. Maron BJ, Towbin JA, Thiene G, et al. Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention. Circulation. 2006;113:1807–1816. doi: 10.1161/CIRCULATIONAHA.106.174287.
    1. Elliott P, Andersson B, Arbustini E, et al. Classification of the cardiomyopathies: a position statement from the European Society Of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J. 2008;29:270–276. doi: 10.1093/eurheartj/ehm342.
    1. Corrado D, Basso C, Thiene G, et al. Spectrum of clinicopathologic manifestations of arrhythmogenic right ventricular cardiomyopathy/dysplasia: a multicenter study. J Am Coll Cardiol. 1997;30:1512–1520. doi: 10.1016/S0735-1097(97)00332-X.
    1. Sen-Chowdhry S, Syrris P, Ward D, et al. Clinical and genetic characterization of families with arrhythmogenic right ventricular dysplasia/cardiomyopathy provides novel insights into patterns of disease expression. Circulation. 2007;115:1710–1720. doi: 10.1161/CIRCULATIONAHA.106.660241.
    1. Marcus FI, Zareba W, Calkins H, et al. Arrhythmogenic right ventricular cardiomyopathy/dysplasia clinical presentation and diagnostic evaluation: results from the North American Multidisciplinary Study. Heart Rhythm. 2009;6:984–992. doi: 10.1016/j.hrthm.2009.03.013.
    1. MacLennan DH, Kranias EG. Phospholamban: a crucial regulator of cardiac contractility. Nat Rev Mol Cell Biol. 2003;4:566–577. doi: 10.1038/nrm1151.

Source: PubMed

3
Se inscrever