Rationale and design of a randomized, controlled multicentre clinical trial to evaluate the effect of bromocriptine on left ventricular function in women with peripartum cardiomyopathy

Arash Haghikia, Edith Podewski, Dominik Berliner, Kristina Sonnenschein, Dieter Fischer, Christiane E Angermann, Michael Böhm, Philipp Röntgen, Johann Bauersachs, Denise Hilfiker-Kleiner, Arash Haghikia, Edith Podewski, Dominik Berliner, Kristina Sonnenschein, Dieter Fischer, Christiane E Angermann, Michael Böhm, Philipp Röntgen, Johann Bauersachs, Denise Hilfiker-Kleiner

Abstract

Background: Peripartum cardiomyopathy (PPCM) is an idiopathic heart disease that develops in the last month of pregnancy and/or the first months following delivery in previously healthy women and may lead to acute heart failure. A cleaved fragment of the nursing hormone prolactin is considered essential in the pathophysiology of PPCM. To date, no specific therapy has been tested for PPCM in a randomized controlled trial of adequate size.

Aims: The purpose of this trial is to investigate the safety of the dopamin-D2-receptor agonist bromocriptine and its effects on left ventricular (LV) function in women with PPCM.

Methods: This is an 11 center German trial with a prospective randomized controlled open-label design. The trial enrolls females with newly diagnosed PPCM according to European Society of Cardiology criteria with a LV ejection fraction (LVEF) <35 %. Patients are randomized 1:1 to either best supportive care (BSC) including standard heart failure therapy plus 8 weeks of bromocriptine therapy (2.5 mg b.i.d. for 14 days and 2.5 mg q.d. from day 15 to 56) or to BSC plus 1 week of low-dose bromocriptine (2.5 mg q.d.) with anticoagulant therapy at a prophylactic dose administered during the period of bromocriptine treatment in both groups. The primary endpoint is change in LVEF from baseline to 6 months follow-up as assessed by cardiac magnetic resonance imaging (or echocardiography if CMR is not tolerated). The secondary endpoints are hospitalization for worsening heart failure, heart transplantation, and all-cause mortality during follow-up or a combination of these endpoints. A total of 60 patients will be recruited (including 6 potential dropouts) giving a power of 0.9 for an expected LVEF change of 10.8 % between treatment groups at 6 months.

Perspective: This trial will provide important knowledge on potential benefits and safety of prolonged inhibition of prolactin release with bromocriptine in addition to standard heart failure therapy in newly diagnosed PPCM.

Trial registration: ClinicalTrials.gov Identifier: NCT00998556.

Keywords: Bromocriptine; Heart failure; Peripartum cardiomyopathy; Prolactin.

Figures

Fig. 1
Fig. 1
Study design. Asterisks indicates anticoagulation therapy at prophylactic dose is administered during treatment with bromocriptine.

References

    1. Sliwa K, Hilfiker-Kleiner D, Petrie MC, et al. Current state of knowledge on aetiology, diagnosis, management, and therapy of peripartum cardiomyopathy: a position statement from the Heart Failure Association of the European Society of Cardiology Working Group on peripartum cardiomyopathy. Eur J Heart Fail. 2010;12:767–778. doi: 10.1093/eurjhf/hfq120.
    1. Sliwa K, Fett J, Elkayam U. Peripartum cardiomyopathy. Lancet. 2006;368:687–693. doi: 10.1016/S0140-6736(06)69253-2.
    1. Kolte D, Khera S, Aronow WS, et al. Temporal trends in incidence and outcomes of peripartum cardiomyopathy in the United States: a nationwide population-based study. J Am Heart Assoc. 2014;3:e001056. doi: 10.1161/JAHA.114.001056.
    1. Felker GM, Thompson RE, Hare JM, et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. N Engl J Med. 2000;342:1077–1084. doi: 10.1056/NEJM200004133421502.
    1. Haghikia A, Podewski E, Libhaber E, et al. Phenotyping and outcome on contemporary management in a German cohort of patients with peripartum cardiomyopathy. Basic Res Cardiol. 2013;108:366. doi: 10.1007/s00395-013-0366-9.
    1. Pearson GD, Veille JC, Rahimtoola S, et al. Peripartum cardiomyopathy: National Heart, Lung, and Blood Institute and Office of rare diseases (National Institutes of Health) workshop recommendations and review. JAMA. 2000;283:1183–1188. doi: 10.1001/jama.283.9.1183.
    1. Witlin AG, Mabie WC, Sibai BM. Peripartum cardiomyopathy: a longitudinal echocardiographic study. Am J Obstet Gynecol. 1997;177:1129–1132. doi: 10.1016/S0002-9378(97)70028-0.
    1. Forster O, Hilfiker-Kleiner D, Ansari AA, et al. Reversal of IFN-gamma, oxLDL and prolactin serum levels correlate with clinical improvement in patients with peripartum cardiomyopathy. Eur J Heart Fail. 2008;10:861–868. doi: 10.1016/j.ejheart.2008.07.005.
    1. Hilfiker-Kleiner D, Kaminski K, Podewski E, et al. A cathepsin D-cleaved 16 kDa form of prolactin mediates postpartum cardiomyopathy. Cell. 2007;128:589–600. doi: 10.1016/j.cell.2006.12.036.
    1. Halkein J, Tabruyn SP, Ricke-Hoch M, et al. MicroRNA-146a is a therapeutic target and biomarker for peripartum cardiomyopathy. J Clin Invest. 2013;123:2143–2154. doi: 10.1172/JCI64365.
    1. Hilfiker-Kleiner D, Meyer GP, Schieffer E, et al. Recovery from postpartum cardiomyopathy in 2 patients by blocking prolactin release with bromocriptine. J Am Coll Cardiol. 2007;50:2354–2355. doi: 10.1016/j.jacc.2007.10.006.
    1. Sliwa K, Blauwet L, Tibazarwa K, et al. Evaluation of bromocriptine in the treatment of acute severe peripartum cardiomyopathy: a proof-of-concept pilot study. Circulation. 2010;121:1465–1473. doi: 10.1161/CIRCULATIONAHA.109.901496.
    1. Sliwa K, Hilfiker-Kleiner D, Mebazaa A, et al. EURObservational Research Programme: a worldwide registry on peripartum cardiomyopathy (PPCM) in conjunction with the Heart Failure Association of the European Society of Cardiology Working Group on PPCM. Eur J Heart Fail. 2014;16:583–591. doi: 10.1002/ejhf.68.
    1. McMurray JJ, Adamopoulos S, Anker SD, et al. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The task force for the diagnosis and treatment of acute and chronic heart failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail. 2012;14:803–869. doi: 10.1093/eurjhf/hfs033.
    1. Franke J, Zugck C, Wolter JS, et al. A decade of developments in chronic heart failure treatment: a comparison of therapy and outcome in a secondary and tertiary hospital setting. Clin Res Cardiol. 2012;101:1–10. doi: 10.1007/s00392-011-0348-6.
    1. Böhm M, Borer J, Ford I, et al. Heart rate at baseline influences the effect of ivabradine on cardiovascular outcomes in chronic heart failure: analysis from the SHIFT study. Clin Res Cardiol. 2013;102:11–22. doi: 10.1007/s00392-012-0467-8.
    1. Ferreira JP, Santos M, Almeida S, Marques I, Bettencourt P, Carvalho H, et al. Tailoring diuretic therapy in acute heart failure: insight into early diuretic response predictors. Clin Res Cardiol. 2013;102:745–753. doi: 10.1007/s00392-013-0588-8.
    1. Weil C. The safety of bromocriptine in long-term use: a review of the literature. Curr Med Res Opin. 1986;10:25–51. doi: 10.1185/03007998609111089.
    1. Iffy L, Lindenthal J, McArdle JJ, Ganesh V, et al. Severe cerebral accidents postpartum in patients taking bromocriptine for milk suppression. Isr J Med Sci. 1996;32:309–312.
    1. Hopp L, Weisse AB, Iffy L, et al. Acute myocardial infarction in a healthy mother using bromocriptine for milk suppression. Can J Cardiol. 1996;12:415–418.
    1. Brenner B. Haemostatic changes in pregnancy. Thromb Res. 2014;114:409–414. doi: 10.1016/j.thromres.2004.08.004.
    1. Hilfiker-Kleiner D, Haghikia A, Nonhoff J, et al. Peripartum cardiomyopathy: current management and future perspectives. Eur Heart J. 2015;36:1090–1097. doi: 10.1093/eurheartj/ehv009.
    1. Ricke-Hoch M, Bultmann I, Stapel B, et al. Opposing roles of Akt and STAT3 in the protection of the maternal heart from peripartum stress. Cardiovasc Res. 2014;101:587–596. doi: 10.1093/cvr/cvu010.

Source: PubMed

Подписаться