426
Views
2
CrossRef citations to date
0
Altmetric
Editorial

What’s next in peripartum cardiomyopathy investigation?

Pages 743-746 | Published online: 10 Jan 2014

Peripartum cardiomyopathy (PPCM) is a devastating illness afflicting new mothers worldwide. Despite being relatively uncommon in many areas of the world, PPCM is nonetheless an important cause of morbidity and mortality in new mothers. For this reason, it has received welcome attention by investigators in the past decade, with numerous reports and recent general reviewsCitation[1–6]. PPCM represents a syndrome of pregnancy-associated cardiomyopathy (PACM) Citation[7] with multiple antecedents and complex biopathological processes. Whatever the precipitating factor(s) may be, immune system dysfunction with evolution to an autoimmune cardiomyopathy seems highly likely Citation[5,6,8–11]. At the present time, two hypotheses of etiology are foremost: pregnancy-associated hormonal changes, specifically the role of prolactin (PRL); and viral infection.

Pregnancy-associated hormonal changes

In the PRL theory, proposed and advanced by Sliwa and Hilfiker-Kleiner Citation[12,13], activated cathepsin-D cleaves normal 23-kDa PRL, resulting in a purported cardiotoxic 16-kDa PRL. In their pilot study, impressive gains in systolic function at 6 months were reported for many of the ten women receiving bromocriptine and conventional heart failure treatment compared with ten control mothers receiving conventional treatment alone. The appropriate balance and caution in exploring this important hypothesis are provided in the recent editorial by Elkayam and Goland, emphasizing the need for “a large, well-designed, prospective study” Citation[14]. This editorial also reports a trial of bromocriptine in two PPCM patients without benefit.

Given the number of PPCM patients who recover to left ventricular ejection fraction (LVEF) more than 0.50 with contemporary evidence-based treatment without bromocriptine, the challenge will be to devise a study with sufficient power to give valid results Citation[15–17]. It is also difficult to devise randomly assigned, double-blinded trials owing to the need to assure adequate newborn nutrition for those to whom breast milk will not be available as a result of PRL inhibition.

Analysis of individual patients from these investigator reports with limited testing to date has demonstrated that not all PPCM mothers test positive for cathepsin-D activation and detectable 16-kDa PRL Citation[12–14]; not all mothers tested have shown elevated plasma levels of PRL at the time of diagnosis (four out of ten in each group had time-of-diagnosis of PPCM plasma levels of PRL less than 25 µg/l, levels seen in nonpregnant, nonlactating women Citation[18]); not all mothers tested have documented suppression of PRL; and not all mothers treated with bromocriptine have shown improvement in LVEF in the first 6 months postdiagnosis. These are essential details that must be explored in this challenging hypothesis that Sliwa et al. have advanced Citation[13].

In subsequent research, it may be best to conduct trials with bromocriptine treatment in PPCM mothers who:

  • • Demonstrate serum cathepsin-D activation

  • • Test positive for the presence of serum 16-kDa PRL

  • • Accept lactation suppression and the inability to breastfeed

  • • Have assured alternative nutrition for their newborns

In addition, because almost all PPCM patients with diagnostic LVEF of 0.35 or more go on to recovery with evidence-based conventional treatment of diuretics, ACE inhibitors and b-blockade Citation[17,19], one may wish to limit the use of PRL inhibition treatment to those with a LVEF of less than 0.35 at diagnosis. Arbitrary use of bromocriptine outside of a research protocol will not answer the lingering questions and may subject patients to as yet unidentified risks. Additional research also needs to continue on the evidence that ACE-inhibitors and β-blockers help to correct the immune system dysfunction that occurs with PPCM/PACM, quite apart from their hemodynamic benefits Citation[20–23].

Viral infection

It is clear in both humans and experimental animals that infection with cardiotropic viruses can result in dilated cardiomyopathy and heart failure Citation[11,24–26]. Evidence supporting a role for viral infection in PPCM stems from case reports documenting viral positivity by IgM serum antibodies and PCR viral genomes in serum and/or cardiomyocytes Citation[27–30]. Viruses involved include adenovirus, coxsackievirus B (enterovirus), cytomegalovirus, E-B virus, hepatitis C virus, human herpesvirus 6, influenza virus A/B and parvovirus B19 Citation[24–42]. Further documentation will require systematic testing of peripheral blood in a large series of prospectively identified PPCM mothers, as suggested in an earlier publication Citation[29].

It is this group of mothers with PACM in whom intravenous immunoglobulin may still hold some promise for effective treatment and warrant additional trials; most particularly, the nonresponders to conventional evidence-based treatments, including those who have fulminant disease with imminent mortality Citation[30,43–46].

For at least some cardiotropic viruses, presence in the myocardium does not appear to be sufficient to confirm a causal role. This may particularly be the case for parvovirus B19 Citation[47]. Additional evidence of replication and myocardial viral load may be necessary to confirm a pathogenic role Citation[48]. It also remains to be seen if the additional presence of cellular infiltrate with a lymphocytic myocarditis or inflammatory cardiomyopathy could help to differentiate pathogenesis from the nonpathogenic coincidental presence of viral particles.

Antecedent disease

There appears to be a group of PPCM mothers who may have had asymptomatic systolic dysfunction (ASD) before the index pregnancy in which PPCM was diagnosed Citation[49,50]. Unless an echocardiogram had been carried out previously there would be no way of knowing this. Aggravation of antecedent ASD with current pregnancy-associated heart failure could occur from unrecognized preceding pregnancy PPCM, unrecognized familial dilated cardiomyopathy or other forms of cardiomyopathy, or unrecognized nonpregnancy-associated antecedent viral cardiomyopathy.

Other cardiac stressors

There are multiple cardiac stressors that may or may not play a causative role, but may tip the balance into overt heart failure. These include: pregnancy-induced hypertension, chronic hypertension, toxemia of pregnancy, anemia, multiple gestations, prolonged tocolytic therapy, hormonal manipulations with assisted reproductive technologies and oxidative stress. Each merits additional investigation.

Profiling PPCM patients

Whatever the role of the aforementioned factors may be, it may be helpful to attempt to understand and classify PPCM patients on the basis of their:

  • • Autoimmune antibody profiles (particularly cardiac β-1-adrenoreceptor and cardiac myosin heavy-chain autoantibodies)

  • • Pro- and anti-inflammatory cytokine and chemokine profiles

  • • C-reactive protein levels

  • • B-type natriuretic peptide levels Citation[9,10,51–58].

Genetics may play a very important role in determining cytokine and chemokine imbalances, and thus subsequent susceptibilities to immune system dysfunction Citation[9]. It should not be overlooked that two African-origin PPCM populations (Soweto [South Africa] and Haiti) have demonstrated significantly reduced plasma levels of the cytokine TGF-b, important in fibrosis, inflammation and combating viral infections Citation[56,59–61]. This could factor into the increased incidence of PPCM in African-origin populations Citation[62,63].

Post-PPCM pregnancies

After concern for survival and recovery, one of the most important issues facing PPCM mothers is the question of the safety of a post-PPCM pregnancy. Studies concerning this issue are in agreement that the risk of relapse of heart failure is high for those who have not reached apparent recovery levels of systolic heart function Citation[64–67]. Only one study has prospectively followed PPCM mothers through a subsequent pregnancy, suggesting that the lowest risk for relapse attends those who have recovered to a LVEF of at least 0.55, maintain that recovery level after safe discontinuation of heart failure medications and demonstrate adequate contractile reserve on stress testing Citation[68].

The Peripartum Cardiomyopathy Network, a NIH-funded North American study of PPCM linking more than 30 medical centers, is now poised to address these issues [McNamara D, Pers. Comm.]. Focus on PPCM issues in the past decade have brought us to the position of potentially making breakthrough discoveries that will markedly improve prospects for PPCM mothers all over the world.

Financial & competing interests disclosure

The author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Tibazarwa K, Sliwa K. Peripartum cardiomyopathy in Africa: challenges in diagnosis, prognosis and therapy. Prog. Cardiovasc. Dis.52(4), 317–325 (2010).
  • Ramaraj R, Sorrell VL. Peripartum cardiomyopathy: causes, diagnosis and treatment. Cleve. Clin. J. Med.76(5), 289–296 (2009).
  • Selle T, Renger I, Labidi S, Bultmann I, Hilfiker-Kleiner D. Reviewing peripartum cardiomyopathy: current state of knowledge. Future Cardiol.5(2), 175–189 (2009).
  • Ntusi NB, Mayosi BM. Aetiology and risk factors of peripartum cardiomyopathy: a systematic review. Int. J. Cardiol.131, 168–179 (2009).
  • Sliwa K, Fett J, Elkayam U. Peripartum cardiomyopathy. Lancet368, 687–693 (2006).
  • Pearson GD, Veille JC, Rahiimtoola S et al. Peripartum cardiomyopathy: National Heart, Lung and Blood Institute and Offices of Rare Diseases (National Institutes of Health) Workshop recommendations and review. JAMA283, 1183–1188 (2000).
  • Elkayam U, Akhter MW, Singh H et al. Pregnancy-associated cardiomyopathy: clinical characteristics and a comparison between early and late presentation. Circulation111, 2050–2055 (2005).
  • Sundstrom JB, Fett JD, Carraway RD, Ansari AA. Is peripartum cardiomyopathy an organ specific autoimmune disease? Autoimmun. Rev.1, 73–77 (2002).
  • Ansari AA, Fett JD, Caraway RD, Mayne AE, Onlamoon M, Sundstrom JB. Autoimmune mechanisms as the basis for human peripartum cardiomyopathy. Clin. Rev. Allergy Immunol.23, 301–324 (2002).
  • Warraich RS, Fett JD, Damasceno A et al. Impact of pregnancy related heart failure on humoral immunity: clinical relevance of G3-subclass immunoglobulins in peripartum cardiomyopathy. Am. Heart. J.150, 263–269 (2005).
  • Hill SL, Rose NR. The transition from viral to autoimmune myocarditis. Autoimmunity34, 169–176 (2001).
  • Hilfiker-Kleiner D, Kaminski K, Podewski E et al. A cathepsin D-cleaved 16 kDa form of prolactin mediates postpartum cardiomyopathy. Cell128, 589–600 (2007).
  • 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. Circulation121(13), 1465–1473 (2010).
  • Elkayam U, Goland S. Bromocriptine for the treatment of peripartum cardiomyopathy. Circulation121, 1463–1464 (2010).
  • Amos AM, Jaber WA, Russell SD. Improved outcomes in peripartum cardiomyopathy with contemporary. Am. Heart. J.152, 509–513 (2006).
  • Fett JD, Sannon H, Thelisma E, Sprunger T, Suresh V. Recovery from severe heart failure following peripartum cardiomyopathy. Int. J. Gynaecol. Obstet.104, 125–127 (2009).
  • American Heart Association. The AHA Guidelines and Scientific Statements Handbook. Fuster V (Ed.). Wiley-Blackwell, Oxford, UK (2009).
  • Riordan J. Breastfeeding and Human Lactation (3rd Edition). Jones and Bartlett, MA, USA, 75–77 (2005).
  • McMurray J, Swedberg K. Treatment of chronic heart failure: a comparison between the major guidelines. Eur. Heart J.27(15), 1773–1777 (2006).
  • Godsel LM, Leon JS, Engman DM. Angiotensin converting enzyme inhibitors and angiotensin II receptor antagonists in experimental myocarditis. Curr. Pharm. Des.9, 723–735 (2003).
  • Ohtsuka T, Hamada M, Hiasa G et al. Effect of β-blockers on circulating levels of inflammatory and anti-inflammatory cytokines in patients with dilated cardiomyopathy. J. Am. Coll. Cardiol.37, 412–417 (2001).
  • Yuan Z, Shioji K, Kihara Y, Takenaka H, Onozawa Y, Kishimoto C. Cardioprotective effects of carvedilol on acute autoimmune myocarditis: anti-inflammatory effects associated with antioxidant property. Am. J. Physiol. Heart Circ. Physiol.286(1), H83–H90 (2004).
  • Platten M, Youssef S, Hur EM et al. Blocking angiotensin-converting enzyme induces potent regulatory T cells and modulates Th1 and Th17-mediated autoimmunity. Proc. Natl Acad. Sci. USA106, 14948–14953 (2009).
  • Kawai C. From myocarditis to cardiomyopathy: mechanisms of inflammation and cell death. Circulation99, 1091–1100 (1999).
  • Mason JW. Viral latency: a link between myocarditis and dilated cardiomyopathy? J. Mol. Cell Cardiol.34, 695–698 (2002).
  • Fujioka S, Kitaura Y, Deguchi H et al. Evidence of viral infection in the myocardium of American and Japanese patients with idiopathic dilated cardiomyopathy. Am. J. Cardiol.94, 602–605 (2004).
  • Bultmann BD, Klingel K, Nabauer M, Wallwiener D, Kandolf R. High prevalence of viral genomes and inflammation in peripartum cardiomyopathy. Am. J. Obstet. Gynecol.193, 363–365 (2005).
  • Ambrosini G, Nanhornbque K, Pascoli I, Cester M, Cosmi E. Mirror syndrome due to coxsackie B virus associated to maternal peripartum cardiomyopathy. J. Perinat. Med.36, 453–454 (2008).
  • Fett JD. Diagnosis of viral cardiomyopathy by analysis of peripheral blood? Expert Opin. Ther. Targets12, 1073–1075 (2008).
  • Muroya T, Ikeda S, Yamasa T et al. High dose immune globulin therapy ameliorates peripartum cardiomyopathy with elevated serum antibody titer to influenza virus: case report of two patients. Med. Sci. Monit.16(2), CS11–CS14 (2010).
  • Halapas A, Pissimississ N, Lembessis P et al. Molecular diagnosis of the viral component in cardiomyopathies: pathophysiological, clinical and therapeutic implications. Expert Opin. Ther. Targets12, 821–836 (2008).
  • Dennert R, Crijns HJ, Heymans S. Acute viral myocarditis. Eur. Heart J. DOI: 10.1093/eurheartj/ehn296 (2008) (Epub ahead of print).
  • Mahrholdt H, Wagner A, Deluigi CC et al. Presentation, patterns of myocardial damage and clinical course of viral myocarditis. Circulation114, 1581–1590 (2006).
  • Mahrholdt H, Goedecke C, Wagner A et al. Cardiovascular magnetic resonance assessment of human myocarditis: a comparison to histology and molecular pathology. Circulation109, 1250–1258 (2004).
  • Tavora F, Gonzalez-Cuyar LF, Dalal JS et al. Fatal parvoviral myocarditis: a case report and review of literature. Diagn. Pathol.3, 21 (2008).
  • Tam PY, Butler K, McMahon CJ. Incidental detection of parvovirus myocarditis at time of resection of discrete subaortic stenosis. Cardiol. Young16, 596–598 (2006).
  • Lamparter S, Schoppet M, Pankuweit S, Maisch B. Acute parvovirus B19 infection associated with myocarditis in an immunocompetent adult. Hum. Pathol.34, 725–728 (2003).
  • Murry CE, Jerome KR, Reichenbach DD. Fatal parvovirus myocarditis in a 5-year-old girl. Hum. Pathol.32, 342–345 (2001).
  • Letsas KP, Filippatos GS, Delimpasi S et al. Enterovirus-induced fulminant myocarditis and hemophagocytic syndrome. J. Infect.54, e75–e77 (2007).
  • Ishikawa T, Zhu BL, Li DR et al. Epstein–Barr virus myocarditis as a cause of sudden death: two autopsy cases. Int. J. Legal Med.119, 231–235 (2005).
  • Frustaci A, Calabrese F, Chimenti C et al. Lone hepatitis C virus myocarditis responsive to immunosuppressive therapy. Chest122, 1348–1356 (2002).
  • Harder TC, Hufnagel M, Zahn K et al. New light cycler PCR for rapid and sensitive quantification of parvovirus B19 DNA guides therapeutic decision-making in relapsing infections. J. Clin. Microbiol.39, 4413–4419 (2001).
  • Goland S, Czer LS, Siegel RJ et al. Intravenous immunoglobulin in treatment for acute fulminant inflammatory cardiomyopathy: series of six patients and review of literature. Can. J. Cardiol.24, 571–574 (2008).
  • Bozkurt B, Villaneuva S, Holubkov R et al. Intravenous immune globulin in the therapy of peripartum cardiomyopathy. J. Am. Coll. Cardiol.34, 177–180 (1999).
  • Cooper LT, Virmani R, Chapman NM et al. National Institutes of Health-Sponsored Workshop on Inflammation and Immunity in Dilated Cardiomyopathy. Mayo Clin. Proc.81, 199–204 (2006).
  • Kaveri SV, Lacroix-Desmazes S, Bayry J. Clinical implications of basic research: the antinflammatory IgG. N. Engl. J. Med.359, 307–309 (2008).
  • Lindner J, Noutsias M, Lassner D et al. Adaptive immune responses against parvovirus B19 in patients with myocardial disease. J. Clin. Virol.44, 27–32 (2009).
  • Bock CT, Klingel K, Kanddolf R. Human parvovirus B19-associated myocarditis. N. Engl. J. Med.362, 1248–1249 (2010).
  • Fett JD, Christie LG, Carraway RD, Ansari AA, Sundstrom JB, Murphy JG. Unrecognized peripartum cardiomyopathy in Haitian women. Int. J. Gynaecol. Obstet.90, 161–166 (2005).
  • Vettori DV, Rohde LE, Clauself N. Asymptomatic left ventricular dysfunction in puerperal women: an echocardiographic-based study. Int. J. Cardiol. DOI: 10.1016/j.ijcard.2010.01.015 (2010) (Epub ahead of print).
  • Riad A, Westermann D, Escher F et al. Myeloid differentiation factor-88 contributes to TLR9-mediated modulation of acute coxsackievirus B3-induced myocarditis in vivo. Am. J. Physiol. Heart Circ. Physiol. DOI: 10.1152/ajpheart.01188.2009 (2010) (Epub ahead of print).
  • Jahns R, Boivin V, Hein L et al. Direct evidence for a β1-adrenergic receptor-directed autoimmune attack as a cause of idiopathic dilated cardiomyopathy. J. Clin. Invest.113, 1419–1429 (2004).
  • Freedmann NJ, Lefkowitz RJ. Anti-β1-adrenergic receptor antibodies and heart failure: causation, not just correlation. J. Clin. Invest.113, 1379–1382 (2004).
  • Fett JD, Ansari AA, Sundstrom JB, Combs GR Jr. Peripartum cardiomyopathy: a selenium disconnection and an autoimmune connection. Int. J. Cardiol.86, 311–316 (2002).
  • Muscaro-Blanco A, Alvarez K, Yu X et al. Consequences of unlocking the cardiac myosin molecule in human myocarditis and cardiomyopathies. Autoimmunity41, 442–453 (2008).
  • Ellis JE, Ansari AA, Fett JD et al. Inhibition of progenitor dendritic cell maturation by plasma from patients with peripartum cardiomyopathy: role in pregnancy-associated heart disease. Clin. Devel. Immunol.12, 265–273 (2005).
  • Satoh M, Nakamura M, Akatsu T, Shimoda Y, Segawa I, Hiramori K. C-reactive protein co-expresses with tumor necrosis factor-a in the myocardium in human dilated cardiomyopathy. Eur. J. Heart Fail.7, 748–754 (2005).
  • Lindberg E, Magnusson Y, Karason K, Andersson B. Lower levels of the host protective IL-10 in DCM – a feature of autoimmune pathogenesis? Autoimmunity41, 478–483 (2008).
  • Sliwa K, Forster O, Libhaber E et al. Peripartum dardiomyopathy: inflammatory markers as predictors of outcome in 100 propspectively studied patients. Eur. Heart J.27, 441–446 (2006).
  • Horwitz MS, Knudsen M, Ilic A, Fine C, Sarvetnick N. Transforming growth factor-β inhibits coxsackievirus-mediated autoimmune myocarditis. Viral Immunol.19, 722–733 (2006).
  • Lucas JA, Zhang Y, Li P et al. Inhibition of transforming growth factor-β signaling induces left ventricular dilation and dysfunction in the pressure-overloaded heart. Am. J. Physiol. Heart Circ. Physiol.298, H424–H432 (2010).
  • Gentry MB, Dias JK, Luis A, Patel R, Thornton J, Reed GL. African–American women have a higher risk for developing peripartum cardiomyopathy. J. Am. Coll. Cardiol.55(7), 654–659 (2010).
  • Brar SS, Khan SS, Sandhu GK et al. Incidence, mortality and racial difference in peripartum cardiomyopathy. Am. J. Cardiol.100, 302–304 (2007).
  • Fett JD, Christie LG, Murphy JG. Outcomes of subsequent pregnancy after peripartum cardiomyopathy: a case series from Haiti. Ann. Int. Med.145, 30 –34 (2006).
  • Elkayam U, Tummala PP, Rao K et al. Maternal and fetal outcomes of subsequent pregnancies in women with peripartum cardiomyopathy. N. Engl. J. Med.344(21), 1567–1571 (2001).
  • Habli M, O’Brien T, Nowack E, Khoury S, Barton JR, Sibai B. Peripartum cardiomyopathy: prognostic factors for long-term maternal outcome. Am. J. Obstet. Gynecol.199(4), 415.e1–5 (2008).
  • Modi KA, Illum S, Jariatul K, Caldito G, Reddy PC. Poor outcome of indigent patients with peripartum cardiomyopathy in the United States. Am. J. Obstet. Gynecol.201(2), 171.e1–e5 (2009).
  • Fett JD, Fristoe KL, Welsh SN. Risk of heart failure relapse in subsequent pregnancy among peripartum cardiomyopathy mothers. Int. J. Gynecol. Obstet.109(1), 34–36 (2010).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.