ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 1 of 55

Treatment for Pulmonary Arterial Hypertension-Associated Right Ventricular Dysfunction

Jose Gomez-Arroyo MD, PhD1,2, Julio Sandoval MD2, Marc A. Simon MD, MS3, Erick Dominguez-Cano MD, MS2, Norbert F. Voelkel MD4 and Harm J. Bogaard MD, PhD5

1

Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United

States; 2Departamento de Cardioneumologia, Instituto Nacional de Cardiologia “Ignacio Chavez”, Mexico City, Mexico; 3Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States; 4Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States; 5Department of Pulmonary Medicine, VU Medical Center, Amsterdam, Netherlands

Word Count: 6281

Corresponding author: Harm Jan Bogaard, MD PhD Associate Professor of Pulmonary Medicine Department of Pulmonary Medicine VU University Medical Center Amsterdam, the Netherlands [email protected]

1

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 2 of 55

Abstract

Pulmonary arterial hypertension (PAH) includes a heterogeneous group of diseases characterized by pulmonary vasoconstriction and remodeling of the lung circulation. Although PAH is a disease of the lungs, patients with PAH frequently die of right heart failure. Indeed, survival of PAH patients depends on the adaptive response of the right ventricle to the changes in the lung circulation. PAH specific drugs affect the function of right ventricle through afterload reduction and perhaps also through direct effects on the myocardium. Prostacyclins, type 5 phosphodiesterase inhibitors and guanylyl cyclase stimulators may directly enhance myocardial contractility through increased cyclic adenosine and guanosine monophosphate availability. While this may initially improve cardiac performance, the long term effects on myocardial oxygen consumption and function are unclear. Cardiac effects of endothelin receptor antagonists may be opposite, as endothelin-1 is known to suppress cardiac contractility. Because PAH is increasingly considered as a disease with quasi-malignant growth of cells in the pulmonary vascular wall, therapies are being developed which inhibit hypertrophy and angiogenesis, and promote apoptosis. The inherent danger of these therapies is a further compromise to the already ischemic, fibrotic and dysfunctional right ventricle. More recently, the right heart was identified as a direct treatment target in PAH. The effects of well-established therapies for left heart failure, such as β-adrenergic receptor blockers, inhibitors of the renin-angiotensin system, exercise training and assist devices are currently investigated in PAH. Future treatment of PAH patients will likely consist of a multifaceted approach aiming to reduce the pressure in the lung circulation and improving right heart adaptation.

Key words: heart failure; pharmacology; pulmonary heart disease; right ventricle

2

Copyright © 2014 by the American Thoracic Society

Page 3 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Introduction

The primary determinant of survival in pulmonary arterial hypertension (PAH) is the response of the right ventricle (RV) to the functional and structural alterations of the pulmonary circulation1-3. As reviewed recently4, PAH-associated RV failure is thought to result from multiple interactions between an increased afterload and a derailed autocrine, paracrine and neuro-endocrine signaling, ultimately leading to RV-arterial uncoupling, a loss of cardiomyocytes, metabolic remodeling, mitochondrial dysfunction, maladaptive changes of the extracellular matrix, myocardial ischemia and inflammation5 The relative importance of these interacting pathological mechanisms is unclear and the impact of current PAH treatments on the adapting RV are, likewise, obscure. In this review we sought to recapitulate some of the known effects of standard PAH treatments on the heart, as well as to explore potentially new additional therapies to directly treat RV failure. The pharmacological management of acute RV failure (with inotropes or vasopressors) is not a topic included in this review and we refer the interested reader to existing reviews on this matter6.

Current PAH-specific pharmacotherapies and their effect on the right ventricle

Guidelines for the treatment of PAH patients recommend oxygen and diuretics as needed, as well as anticoagulants in patients without contraindications7;8. Calcium channel blockers are used in patients with significant pulmonary vascular reactivity, while other symptomatic patients are treated with prostacyclin (PGI2) analogs, endothelin (ET)-1 receptor antagonists and type 5 phosphodiesterase (PDE5) inhibitors, alone or in combinations7. It is clear that current pharmacotherapies improve pulmonary

3

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 4 of 55

hemodynamics, cardiac performance and functional class of patients with PAH, however, whether any of the clinical benefits from the above mentioned groups of drugs can be explained by direct effects on the RV has not yet been fully investigated.

When it comes to the evaluation of direct RV effects of vasodilator treatment in PAH, two pathophysiological concepts need to be considered. First, RV contractility is greater than normal in virtually all PAH patients and therefore RV dysfunction is best explained by an increase in afterload out of proportion to the increase in RV contractility (RV-arterial uncoupling, assessed using pressure volume analysis)9. RV-arterial coupling can be improved by either an increase in contractility or by a decrease in afterload. Even if a pulmonary vasodilator drug has negative inotropic effects it could still improve RVarterial coupling, as long as the decrease in afterload is larger than the decrease in contractility. Second, while a short term increase in contractility may be beneficial, longterm effects of enhanced contractility (and an accompanying increase in myocardial oxygen consumption) may be detrimental. In other words, increasing contractility might not be the ideal long-term goal of treatment if cardiac cellular homeostasis has not been similarly restored.

Supportive treatment and calcium channel blockers Oxygen is thought to lower pulmonary vascular resistance by releasing hypoxic pulmonary vasoconstriction and decelerating vascular remodeling; a target oxygen saturation of 90% has been recommended10. There is no evidence for direct effects of supplemental oxygen on the RV in PAH, while either a protection against ischemia11 or an increased production of reactive oxygen species and suppression of hypoxia inducible factor (HIF)-1α expression could be hypothesized.

4

Copyright © 2014 by the American Thoracic Society

Page 5 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

The consensus use of anticoagulant therapy is based on indirect evidence12;13, however, a recent analysis from the Comparative, Prospective Registry of Newly Initiated Therapies for Pulmonary Hypertension (COMPERA) reported that – at least in patients with idiopathic PAH – anticoagulant therapy was associated with a significantly better 3year survival when compared to patients who never received anticoagulation13.

Although there is data to suggest that digoxin treatment results in an acute increase in cardiac output along with a reduction in serum nor-epinephrine levels14, the long-term benefit of digoxin in PAH remains unclear. Diuretic treatment leads to symptomatic improvement in the fluid-overloaded patient. The beneficial effect of the aldosterone antagonist spironolactone in heart failure; however, is also associated with immune modulation, reversal of maladaptive remodeling and prevention of hypokalemia and arrhythmia, which are all potentially important but unexplored mechanisms in RV dysfunction associated with PAH15.

Calcium channel blockers are used by a small group of PAH patients with a positive vasoreactivity test in whom a survival benefit has been suggested12. The consequences of potentially negative inotropic effects of calcium channel blockers on RV function in PAH are unknown, although dihydropyridine-type calcium channel blocker (nifedipine and amlodipine) are generally considered safe in PAH.

Prostacyclin analogues For almost twenty years, intravenous administration of epoprostenol (synthetic PGI2) has been the cornerstone of PAH treatment16. It is generally assumed that the therapeutic effect of PGI2 in PAH is explained by an induction of pulmonary vasodilatation and inhibition of vascular remodeling17. However, no evidence exists that chronic

5

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 6 of 55

prostacyclin treatment reverses lung vessel remodeling in PAH18 and there is histological data to suggest that pulmonary vascular remodeling in PAH progresses despite longterm PGI2 treatment19. Rich et al reported that long-term prostacyclin treatment might have an antithrombotic effect but does not prevent or reverse the formation of advanced vascular lesions20.

It has to be recognized, however, that PGI2 has important direct effects on the heart. In patients with severe heart failure, PGI2 treatment results in an immediate and substantial increase in cardiac output and a reduction in cardiac filling pressures21. Recent work shows that unlike other pulmonary vasodilators, PGI2 analogs improve RV stroke work in PAH22, which is probably a better reflection of cardiac function than cardiac output. Although this improvement in cardiac function could be an adaptive response to systemic vasodilation or the result of pulmonary vasodilation with improved right ventriculo-arterial coupling21;23, a direct effect on cardiac cells and cell signaling pathways is another possibility. In an animal model of flow-associated PAH, a PGI2 analog, iloprost, improved RV contractility and capillary-to-myocyte ratio, independently from a change in RV afterload24. Syed et al recently reported that treatment with iloprost does not modify the pulmonary artery pressure, but does improve RV contractility and reduces fibrosis in the sugen (SU5416)/hypoxia rat model of RV failure and severe PAH25. In addition, PGI2 has been reported to suppress pressure overload–induced left ventricular hypertrophy26 and fibrosis27. Both effects are considered to originate from the action of cells other than cardiomyocytes, however, the exact mechanism remains undetermined26.

Potentially beneficial direct effects of PGI2 treatment on the heart were the main reason for initiating large clinical trials with epoprostenol treatment in patients with severe left

6

Copyright © 2014 by the American Thoracic Society

Page 7 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

heart failure. Unexpectedly and despite previous positive results, six months of treatment with epoprostenol was associated with increased mortality28;29. A possible explanation for this adverse outcome was the increase in cardiac output upon initiation of epoprostenol therapy21. As such, increases in myocardial contractility and cardiac output may initially improve exercise capacity, but the accompanying increase in myocardial oxygen consumption could be detrimental in the long-term. Such a sequence of events could be related to the results from the Beraprost Study, which is the only randomized clinical trial with a follow-up time of one year with specific PAH therapy. It was reported that although exercise capacity improved after 12 weeks of treatment with an oral PGI2 analog, this benefit disappeared after one year30.

Indeed, the effects of prostacyclin analogues on the function of the RV are still poorly understood. An increase in myocardial contractility may play a different role in PAH (where improved RV-arterial coupling is desired) when compared to end-stage left heart disease. In the future it will be important to evaluate whether any potential direct cardiac effects of prostacyclin treatment depend on exposure time or are just an idiosyncratic response to therapy, specific for every patient.

Endothelin receptor blockers In PAH and heart failure, ET-1 serum concentrations are elevated due to increased production by endothelial cells and cardiomyocytes in response to various stimuli (e.g. vasoactive hormones, growth factors, shear stress, hypoxia and reactive oxygen species, ROS)31-34. ET-1 not only increases pulmonary vascular tone, but also regulates a variety of biological processes in non-vascular tissues. ET-1 augments cardiomyocyte contractility and plays a role in the development of pressure overload-induced cardiac hypertrophy31;35. In patients with PAH associated heart failure, the direct effects of ET-1

7

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 8 of 55

signaling on the heart are mixed with indirect effects via stimulation of pulmonary vasoconstriction and vascular remodeling. ET-1 exerts its effects through two receptor subtypes, ETA and ETB, the former predominating in the rat myocardium36. Heart failure in rats leads to an increased ETA receptor density33. ET-1 affects cardiomyocyte survival and pressure overload induced hypertrophy by interacting with bcl-237, the epidermal growth factor receptor (EGF-R)38 and mitogen-activated protein kinase (MAPK) cascades35.

Clinical trials in patients with left sided heart failure with orally administered ET-receptor antagonists (REACH-1, ENCOR, ENABLE and EARTH were the largest, although the results have never been fully published) suggest that direct effects of ET-receptor antagonists on the heart are not favorable39-41. It has been postulated that the inotropic actions of ET-1 are in fact beneficial in chronic heart failure, providing partial compensation for a decreased contractility.

Detrimental actions of ET receptor blocker in the RV of patients with PAH have not (yet) been shown. Experimental studies, however, do suggest that endothelin receptor blockade worsens the contractility of the pressure overloaded RV in rats42. Whether this occurs in PAH and would translate into negative effects for individual patients would depend on the relative magnitudes of decreases in afterload and contractility and the resultant effect on RV-arterial coupling.

Phosphodiesterase inhibitors Cyclic guanosine monophosphate (cGMP) is a ubiquitous intracellular secondary messenger. The natriuretic peptides generate cGMP via activation of the particulate guanyl cyclase (pGC), whereas nitric oxide (NO) induces the formation of cGMP through

8

Copyright © 2014 by the American Thoracic Society

Page 9 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

activation of soluble guanylate cyclase (sGC)43. cGMP is degraded by the action of phosphodiesterases (PDEs). Some PDE subtypes hydrolyze only cGMP (PDE5, PDE6, PDE9), whereas others degrade cAMP (PDE3, PDE4, PDE7, PDE8) or both cGMP and cAMP (PDE1, PDE2)44. cGMP lowers Ca2+ sensitivity and intracellular Ca2+ concentration in pulmonary vascular smooth muscle cells. The resulting vasodilating and antiproliferative properties of the molecule explain the therapeutic benefit of the PDE5 inhibitors, sildenafil and tadalafil in PAH45;46. More recently, the therapeutic benefit of increasing cGMP availability with the sGC stimulator Riociguat has been shown in a randomized clinical trial47. cGMP/protein kinase G (PKG) signaling protects the heart from apoptosis48;49 and blunts the hypertrophic response to pressure overload and isoproterenol (an adrenergic agonist)50-52.

Multiple roles for cGMP in cardiac contractility, lusitropy and ion channel responsivity have been well characterized, however the extent to which natriuretic peptides predominate over NO to mediate these effects is less clear53. The effect of cGMP on myocardial contractility depends on its interactions with PDEs and cAMP. Theoretically, cGMP can decrease contractility by decreasing cAMP concentrations through inhibition of adenylate cyclase and induction of PDE254;55. In addition, phosphorylation of troponin I by a cGMP-dependent protein kinase can decrease the sensitivity of the contractile apparatus to Ca2+ and accelerate myocardial relaxation54. On the other hand, it was recently shown in PAH patients and in rats with monocrotaline (MCT) induced RV hypertrophy, that cGMP can in fact increase contractility by increasing cAMP concentrations56. The authors explained this apparent paradox by cGMP related inhibition of the cGMP sensitive PDE3. They also showed that, compared with a normal RV, RV hypertrophy (both in humans and rats) is associated with a considerable

9

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 10 of 55

decrease in PKG activity. At the same time, PDE5 was only expressed in the hypertrophic RV and not in the normal RV56.

Once again, the long-term effects of PDE5 inhibition and sGC stimulation on RV adaptation in PAH are unknown. The initial preclinical evidence seemed to justify a provisional conclusion that these strategies could have positive direct effects on the heart. PDE5 inhibition prevents and reverses pressure overload induced hypertrophy in mice, which was associated with enhanced systolic function52. Sildenafil protects against cardiomyocyte apoptosis48;49, and may decrease myocardial oxygen consumption by inhibiting adenylate cyclase55 and accelerating myocardial relaxation54.

Despite such positive experimental data, clinical studies of drugs that increase cGMP availability in patients with heart failure have yielded mixed results. After first positive results of PDE5 inhibition in patients with non-PAH heart failure with decreased57;58 and preserved ejection fraction (HFPEF)59, recent randomized clinical trials using sildenafil and riociguat in these patient categories failed to meet their primary endpoints60;61. A single dose of sildenafil was shown to improve RV diastolic function in PAH patients62. It remains to be determined whether this is a direct cardiac effect or whether it was mediated by a decrease in RV afterload.

New treatment strategies in PAH: good for the lung, bad for the heart?

One hypothesis that has been advanced to explain the pathobiology of severe PAH is based on the concept of a quasi-malignant nature of endothelial cells in the lung vasculature. This hypothesis postulates that after an initial injury, pulmonary vascular

10

Copyright © 2014 by the American Thoracic Society

Page 11 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

endothelial cells undergo apoptosis, but a group of surviving cells switch their phenotype and become apoptosis-resistant and hyperproliferative to the point of lumen occlusion. In other words, the hypothesis reflects a process of “wound healing gone awry”5. There is now a search for molecular targets and mechanisms which could potentially drive this “quasi-malignant” lung vessel remodeling. However, as the knowledge of the pathobiology of PAH evolves, future therapeutic strategies face one critical paradox: While the remodeled lung vasculature in PAH is characterized by angiogenesis, apoptosis resistance and cell proliferation, the failing RV may suffer from ischemia, capillary rarefaction and cardiomyocyte apoptosis63. Thus, new treatments designed to tackle any quasi-malignant feature in the sick-lung circulation could have detrimental effects in an ischemic, fibrotic and dysfunctional RV.

Experimental models of right ventricular dysfunction As is the case with many other diseases, potential new compounds for the treatment of PAH are generally tested first in animals. Several experimental models have been developed and the interested reader is referred to a recent review on this topic providing a detailed overview64. Rat models have generally provided superior insights into the mechanisms of severe pulmonary vascular disease and associated RV failure, as severe pulmonary hypertension is very difficult to induce in mice65. Pulmonary artery banding, which creates mechanical stress on the RV without pulmonary vascular disease, has been useful to explore side effects of drugs on RV adaptation without the interfering effects from changes in pulmonary vascular resistance63;66.

Tyrosine kinase inhibitors Platelet derived growth factor (PDGF) has been implicated in the pathobiology of pulmonary vascular remodeling in PAH67 and treatment with imatinib, an inhibitor of the

11

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 12 of 55

tyrosine kinase domain of the PDGF receptor has been beneficial in isolated cases68-71. Nonetheless, a phase II trial with PAH patients failed to meet its primary endpoint of improved exercise capacity after 4 months72 and a randomized clinical trial in selected PAH patients (PVR ≥ 800 dyne·s·cm-5; IMPRES study) showed no improvements in functional class, time to clinical worsening or mortality with imatinib, despite a small improvement in exercise capacity73. The trial was discontinued because of severe side effects, in particular subdural hematoma. Another major concern with imatinib treatment in PAH is the fact that the drug may have detrimental effects on the heart74;75. After myocardial infarction, PDGF enhances cardiomyocyte survival, modulates inflammatory responses and activates pro-angiogenic progenitor cells76-78. Because RV capillary rarefaction and ischemia may play a role in the transition from adaptive hypertrophy to RV dilatation and failure4;63, imatinib could potentially worsen maladaptive cardiac remodeling in PAH. Negative direct cardiac effects were not seen in a post-hoc analysis of echocardiographic parameters in patients treated with imatinib in the IMPRES trial, however79.

In contrast to PDGF receptor blockers, EGF-R blockers may have beneficial effects on both pulmonary vascular and RV remodeling. In maladaptive cardiac remodeling, activation of the type 1 angiotensin receptor (AT1R), the mineralocorticoid receptor and the type A endothelin receptor (ETA) results in a transactivation of the EGF-R38;80;81. Moreover, pulmonary hypertension in rats with MCT-induced PAH is ameliorated by EGF-R blockers82. Interestingly, although not mechanistically tested, the reduced RV hypertrophy observed in this study, might have not been necessarily the result of a reduced afterload, but could have also resulted from decreased myocardial fibrosis and inflammation.

12

Copyright © 2014 by the American Thoracic Society

Page 13 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Paradoxically, tyrosine kinase inhibitors are not only potential treatments for PAH, they have also been implicated in the development of the disease. First, the vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor SU5416, in combination with a hypoxic or allergic challenge, has provided a useful tool to study PAH in rats83. Second, Dasatinib, a dual Src/Abl kinase inhibitor used in the treatment of chronic myelogenous leukaemia, was associated with cases of severe PAH, potentially reversible after dasatinib withdrawal84. Possible dual effects of tyrosine kinase inhibitors need to be taken in account if this group of drugs is further developed for PAH treatment.

Rho kinase inhibitors and HMG-CoA reductase inhibitors (statins) Inhibition of Rho kinase (ROCK) has been suggested as a new target for PAH treatment. Acute administration of ROCK inhibitors has resulted in modest pulmonary vasodilation in PAH patients85;86 and chronic administration prevented pulmonary vascular remodeling in experimental pulmonary hypertension87-91. The effects of long-term ROCK inhibition on the RV in PAH are unknown and based on the available literature, both beneficial (inhibition of apoptosis, reduction of inflammatory cell influx) or detrimental effects (contractile depression and excessive reduction of hypertrophy) can be postulated4. Statins may interfere with ROCK activation through inhibition of isoprenoid synthesis and subsequent rho geranylgeranylation, which is the proposed mechanism by which statins reduce cardiac ROS production and hypertrophy after ATII infusion and pressure overload92. Additional beneficial effects of statins in heart failure consist of inhibition of ET-1 and renin-angiotensin signaling, stimulation of NO production, restoration of autonomic imbalance and prevention of matrix metalloproteinase activation93. Statins have been shown to be effective in reversing pulmonary vascular remodeling and RV hypertrophy in the SU5416/hypoxia model94, but the results from the clinical trial evaluating simvastatin in human PAH were disappointing95.

13

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 14 of 55

Control of gene expression by histone acetylation/deacetylation Histone-dependent packaging of genomic DNA is central mechanism for gene regulation in eukaryotes. When there is no transcription, DNA is wrapped around histone octameres in nucleosomes, which are the basic units of chromatin. The highly compact structure that is formed by interacting nucleosomes limits access of transcriptional enzymes to genomic DNA, thereby repressing gene expression96. Acetylation of histones by histone acetylases relaxes the nucleosomal structures, thereby facilitating gene expression. The opposite effect is established by histone deacytelases (HDACs), which repress transcription. Inhibitors of HDACs have been shown to reverse pulmonary artery smooth muscle cell hypertrophy in experimental models of pulmonary hypertension97.

By repressing the transcription of genes encoding proteins involved in signaling that leads to cardiac hypertrophy98;99, HDAC inhibitors could be hypothesized to have positive direct effects on the RV. Surprisingly, RV adaptation to mechanical pressure overload in the pulmonary artery banding animal model, is seriously hampered by the HDAC inhibitors trichostatin A and valproic acid66. The opposite effects of HDAC inhibitors in the two pressure overloaded ventricles is a strong reminder of the fact that success of a drug in the treatment of left sided heart failure, does not guarantee beneficial effects in the context of PAH associated RV failure.

RV-targeted therapies: a new concept

Indeed, patients with PAH die of RV failure and the prognostic role of the RV has recently been revisited. Van de Veerdonk et al. demonstrated that even after decreasing

14

Copyright © 2014 by the American Thoracic Society

Page 15 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

the pulmonary vascular resistance with PAH-specific therapy, those patients that remain with low RV ejection fraction continue to have a poor prognosis3. However, despite its prognostic importance, the cellular and molecular mechanisms that explain RV failure are limited and frequently extrapolated from studies of chronic left heart failure. Nonetheless, there is increasing evidence to support the hypothesis that mechanisms of RV failure may not be identical to those of the left ventricle. Some therapeutic options to treat left heart failure have been successfully applied to treat the RV in experimental PAH, but others have demonstrated contradictory results.

β-adrenoreceptor blockers Similar to patients with left heart failure, PAH is characterized by increased activity of the sympathetic nervous system (which finding has prognostic significance)100 and downregulation of the β-adrenergic receptor (AR)101. Counteracting these mechanisms led to a firm central place of β-AR blocker treatment in patients with left heart failure102. However, direct effects of β-AR blockade such as systemic vasodilatation, reduction of myocardial contractility and decreased heart rate, have prevented the use of this class of drugs in PAH. In portopulmonary hypertension, β-AR blockers have been associated with worsening hemodynamics and a decreased exercise capacity103.

There are, however, a number of reasons why the careful use of β-AR blockers in selected patients may be considered. The associated reduction of myocardial oxygen consumption is probably one of the explanations of the benefit of β-AR blockers in heart failure. In addition, leakiness of the Ryanodine receptor (RyR) is reversed by β-AR blockers, thereby restoring Ca2+ handling and preventing calcineurin/nuclear factor of activated T-cells (NFAT) upregulation104. β-AR blockers are effective in preventing arrhythmias, which is a problem associated with a markedly increased mortality in

15

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 16 of 55

PAH105. Interestingly, in a recent prospective study, So and collaborators reported that the use of β-AR blockers in PAH is not uncommon106 and most importantly, this study reported no statistical differences in PAH-related hospitalization or all-cause mortality between patients with or without β-AR blocker treatment. However, a properly designed clinical trial to assess the safety of this class of drugs is warranted.

Experimentally, carvedilol treatment improves exercise tolerance, induces RV capillarization and improves RV function107;108. In another study in rats with MCT induced pulmonary hypertension, bisoprolol treatment resulted in an improved RV-arterial coupling and improved survival109. In comparison to the more selective β1-AR blockers, carvedilol has a unique mechanism by which it suppresses ventricular arrhythmias through inhibition of the spontaneous release of calcium from the sarcoplasmic reticulum110. In addition, carvedilol has intrinsic antioxidant properties

111;112

by which the

drug is capable to prevent ROS-induced cardiomyocyte apoptosis113. Some of the cardioprotective effects of carvedilol seem to be independent of blockade of the α- or βAR, such as direct transactivation of the EGF-R via beta arrestins stimulation114. and upregulation of the expression of the peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), a master regulator of mitochondrial biology and cardiac metabolism115 Another strategy to restore β-AR function without the negative chronotropic effects of β-AR blockade would be to prevent G protein-coupled receptor kinase-2 (GRK2)-mediated uncoupling of β-AR using the small molecule Gallein.116

ACE inhibitors and AT1R antagonists Although the renin angiotensin system is clearly involved in pressure overload related cardiac remodeling, the role of ACE inhibitors and AT1R antagonists in PAH has not

16

Copyright © 2014 by the American Thoracic Society

Page 17 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

been thoroughly evaluated and its use as a treatment for PAH-associated RV failure remains controversial. In a small-case series study, four-days of treatment with captopril reduced mean pulmonary arterial pressure and increased RV ejection fraction in three out of four patients117. In another study, however, 12-weeks of captopril did not modify pulmonary hemodynamics or exercise capacity118 Experimentally, both the ACE inhibitor ramipril and the AT1R blocker losartan improved RV systolic function in rabbits subjected to pulmonary artery banding. In contrast, no direct effects of ACE inhibitors or AT1R blockers on RV function or hypertrophy were seen in experimental models of pulmonary hypertension94;119;120. The improved RV function after pulmonary artery banding with ACE inhibition seemo be related to a decreased rate of apoptosis but not as a consequence of reduced hypertrophy121.

Metabolic modulators Abnormal mitochondrial metabolism has long been implicated in the development of chronic heart failure and it has been proposed that a switch from aerobic to anaerobic metabolism could contribute to the development of RV failure122;123. Whereas it has been demonstrated that RV failure is characterized by increased expression of glycolysisrelated genes107 and increased glycolysis enzymatic rates124, it is unclear whether this “metabolic remodeling” is an adaptive or a maladaptive response. We have demonstrated that along with increased glycolysis, RV failure is characterized by a downregulated expression of multiple genes required for fatty acids metabolism supporting a switch in substrate utilization125 The mechanisms responsible for the downregulation of fatty acid oxidation in the failing RV are not well defined, but reduced transcriptional activation of genes regulated by the coactivator PGC-1α and its corresponding nuclear receptor PPAR-α appear to be involved125.

17

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 18 of 55

Multiple studies have shown that the rate of fatty acid oxidation is preserved or increased in physiological/adaptive left ventricular hypertrophy, and that it decreases during the progression of heart failure126. In a similar fashion, rats with adaptive RV hypertrophy after pulmonary artery banding have increased rates of fatty acid oxidation127. However, whether or not decreased impaired fatty acid oxidation contributes to the development of RV failure has not been mechanistically evaluated. Metabolic modulators designed block fatty acid oxidation such as trimetazidine or ranolazine, have been used to prevent cardiac output reduction in rats with pulmonary artery banding, however, the effects of partial fatty acid oxidation inhibition is modest when treating established RV dysfunction127.

We have evaluated the effects of etomoxir, a potent fatty acid oxidation blocker in the SU5416/hypoxia PAH model and report that etomoxir treatment neither worsens nor improves RV failure (Figure 1). Multiple clinical trials evaluating the role of fatty acid oxidation blockers in left heart failure have been designed, however, none of these metabolic modulators have become a standard treatment of heart failure128.

It has also been proposed that not only fatty acid oxidation but also glucose oxidation is impaired and that it could be restored using a pyruvate dehydrogenase kinase (PDK) inhibitor: dichloroacetate (DCA)124. In MCT-induced pulmonary hypertension, DCA decreased the severity of pulmonary hypertension and regressed RV hypertrophy, an effect which was perhaps partially related to a reduction in afterload. The effects of DCA therapy were only moderate in pulmonary artery banding -induced RV hypertrophy124. A clinical trial studying DCA in PAH-associated RV failure

is currently underway

(ClinicalTrials.gov Identifier NCT01083524).

18

Copyright © 2014 by the American Thoracic Society

Page 19 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Antioxidants and tetrahydrobiopterin ROS have been implicated in the pathobiology of chronic left heart dysfunction for a long time129 but their role in RV failure remains to be investigated in depth. ROS can reduce myocyte contractility by affecting calcium handling through suppression of L-type Ca2+ channels and SERCA: the Ca2+-ATPase of the sarcoplasmic reticulum130;131. ROS can be generated from many sources (such as NADPH oxidases). In the heart, mitochondria are an important source of ROS, as they are generated as byproducts of an incomplete reduction of oxygen in the electron transport chain. ROS are very unstable, electrophilic and react with macromolecules, such as proteins and nucleic acids, generating adducts and altering function132. Interruption of the ATII-rho-NAD(P)H-ROS pathway with the xanthine oxidase inhibitor allopurinol improves myocardial contractility

133

and

ameliorates chronic hypoxic PAH in rats134. Hydralazine inhibits NADPH oxidase, but it is unclear whether its antioxidant effects can be achieved at concentrations that are employed clinically53. Another source of ROS in chronic pressure overload comes from uncoupled nitric oxide synthetase (NOS)3 and is associated with reduced availability of tetrahydrobiopterin (BH4), a NOS3 reducing cofactor. Supplementing BH4 in a pressure overload mouse model of NOS3 uncoupling has been shown to be sufficient to reduce ROS production and prevent maladaptive remodeling in experimental left heart failure135.

Probably, the strongest evidence for the role of ROS in RV dysfunction comes from the study of hemeoxygenase-1 (HO-1) knock-out mice. HO-1 plays a categorical role in the hearts response to ROS by inducing the expression of genes that codify for antioxidant enzymes. Yet, Lee and collaborators demonstrated that mice lacking HO-1 show severe RV dilatation and infarction after hypoxia-induced pulmonary hypertension136. On the other hand, we have shown that treatment with protrandim, upregulates HO-1 expression and is associated with less RV dysfunction in experimental pulmonary

19

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

hypertension

Page 20 of 55

63

. Importantly, ROS play a dual role in cells. Whereas ROS have a direct

toxic effect, they also serve as signaling mediators to induce an antioxidant response132. This dual role could potentially complicate the therapeutic potential of antioxidant treatment for RV failure.

Non-pharmacological RV-targeted therapies

Exercise It was previously believed that physical exercise had to be avoided by patients with PAH. After it was shown that exercise training corrected endothelial dysfunction and improved exercise capacity in chronic heart failure

137

, a randomized controlled trial was designed

to evaluate the effects of exercise rehabilitation in PAH

138

. An exercise and respiratory

training program of 4 months’ duration was well tolerated and improved scores of quality of life and exercise capacity (peak workload and oxygen uptake). The mean difference in the six-minute walking distance between intervention and control groups (15 patients in both groups) was 111 meters, which is a considerably larger improvement than observed in most PAH drug trials. Since systolic PAP at rest and exercise cardiac output did not change significantly after training, the authors attributed the positive outcome to adaptations in gas exchange and respiratory and peripheral muscle function

138

.

However, hemodynamic data were obtained non-invasively and the increase in peak oxygen uptake could have been due to an improvement in RV performance.

Experimental data has suggested that exercise could have direct positive effects in the heart. For instance, exercise induces the expression of PGC-1α, which is also associated with increased VEGF expression and reduced capillary rarefaction

139

.

20

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 21 of 55

However, whether PGC-1α-induced angiogenesis could explain an improvement in RV function after exercise training has yet to be determined.

Cardiac resynchronization Cardiac dyssynchrony is a common problem in left heart failure and even patients with mild-to-moderate resynchronization

left

ventricular

therapy.140

systolic

Indeed,

the

dysfunction

benefit

from

cardiac-

Resynchronization–Defibrillation

for

Ambulatory Heart Failure Trial (RAFT) showed that among patients with NYHA class II or III heart failure, a wide QRS complex, and left ventricular systolic dysfunction, the addition of cardiac-resynchronization therapy to an implantable cardioverter–defibrillator reduced rates of death and hospitalization for heart failure.140

Recently a post follow-up analysis from the Multicenter Automatic Defibrillator Implantation Trial with Cardiac Resynchronization Therapy (MADIT-CRT) demonstrated that in patients with mild heart failure, left ventricular dysfunction, and left bundle-branch block, early intervention with cardiac-resynchronization therapy with a defibrillator was associated with a significant long-term survival benefit. However resynchronization– defibrillation did not confer any clinical benefit in patients without left bundle-branch block141.

Ventricular dyssynchrony is also common in patients with PAH and often easily recognized by echocardiogram (paradoxical septal movement), however, compared to the left heart failure, ventricular dyssynchrony in PAH is mostly caused by a difference in duration of RV contraction rather than a difference in the onset of contraction142. There is significantly less evidence to support the use of CRT in patients with severe RV failure, but experimental data has demonstrated that RV pacing improves RV systolic function,

21

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 22 of 55

improves adverse diastolic interaction and resynchronized RV and LV peak pressures143. A small study evaluating RV pacing in patients with chronic thromboembolic pulmonary hypertension demonstrated an improvement in diastolic relaxation, LV stroke volume and RV contractility144, but larger studies evaluating safety and efficacy are warranted.

Atrial septostomy It has been shown that some PAH patients can benefit from ‘decompressing’ therapeutic strategies such as atrial septostomy (AS) or Pott’s shunt. Atrial septostomy is a procedure that creates a right-to-left shunt at the inter-atrial septum level with the use of a balloon-catheter in a step-by-step fashion145. This results in an increase in LV preload and systemic cardiac output at the expense of a drop in systemic arterial oxygen saturation (SaO2%). Theoretically, the drop in SaO2% is compensated by the increase in cardiac output and thus systemic oxygen transport is maintained. Atrial septostomy may also decompress the RV by reducing its preload146;147.

The results of this intervention are sometimes spectacular with disappearance of syncope and fluid retention within a matter of days, but not all patients improve after septostomy and the procedure is not risk free. In a recent review of almost 400 procedures performed worldwide148, there was a 24 hour procedure-related mortality of about 7% mainly resulting from refractory hypoxemia, and a one-month mortality rate of 5% due to RV failure progression. It is fair to say, however, that in most circumstances this intervention has been performed in severely ill patients.

There are no current guidelines for the optimal size of the shunt and therefore the appropriate size should be individualized. Massive right-to-left shunting as a result of an excessively large shunt may result in inadequate pulmonary blood flow and severe,

22

Copyright © 2014 by the American Thoracic Society

Page 23 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

refractory hypoxemia and death – the aim is to achieve a fall in SaO2% below 10%. No studies have evaluated the effect of shunting on the RV at the molecular level. It is important to underscore the fact that balloon dilation atrial septostomy should only be performed in centers experienced in both interventional cardiology and pulmonary hypertension. The creation of a post-tricuspid shunt (“Potts’ anastomosis”) instead of an interatrial shunt might be another (surgical or transcatheter) approach to manage refractory RV dysfunction in the setting of PAH, particularly in children149.

Mechanical RV support and transplantation Mechanical support of the failing RV has generally been thought of as a palliative temporary approach, although as newer technologies develop, there continues to be an interest in the potential for longer term support. Extracorporeal membrane oxygenation (ECMO) has been reported as a method to support the RV in patients with PH and massive pulmonary embolism150;151. It can be implanted quickly, making it advantageous during emergent situations. Cannulae are implanted surgically, typically in a venoarterial arrangement, in which venous blood is withdrawn, pumped through an oxygenator and then returned to the arterial system, bypassing the lungs. Risks include bleeding, thromboembolism, and vessel injury. ECMO support is only temporary and requires a plan for removal: i.e. there is a high likelihood of RV recovery, transplant within a short timeframe is likely and possible, or transition to a more permanent support (such as a right ventricular assist device; to date unlikely in PH) is an option.

Mechanical circulatory assist devices, or ventricular assist devices (VADs), have been used in the setting of RV failure after cardiotomy, cardiac transplant, RV infarct, or LV assist device (LVAD) implantation152-155. They have not been used for the treatment of PAH-associated RV failure largely owing to the concern of pulmonary hemorrhage due

23

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 24 of 55

to input of high flows into a diseased vasculature, which is a result of the pumps being designed for support of the LV. This has raised interest in the concept of partial-assist pumps that could provide enough flow to assist circulation without the risk of pulmonary hemorrhage. Additional benefits of a partial assist pump would be a smaller device size that could reduce surgical times and complications. These potential benefits must be weighed against the risk of thrombosis at lower flows, which has been another limiting factor in adapting LV pumps to the RV. In addition to thromboembolism, other risks of mechanical blood pumps to bear in mind are bleeding and infection. For RV support, blood is typically withdrawn from a cannula surgically placed in the RV and returned from the pump via a cannula in the pulmonary artery.

Although originally developed for support of the LV, there are two devices approved by the FDA for support of the RV, the Thoratec PVAD and CentriMag (both manufactured by Thoratec, Pleasanton, CA, USA). The Thoratec PVAD is a pneumatically driven pulsatile pump that can be used for long-term support. The CentriMag is a continuous flow pump used for short-term support (approved for up to 30 days)156. The field of cardiac mechanical support has progressed to preferentially use continuous-flow (axial or centrifugal in design) devices as they are smaller and have improved durability. Two continuous-flow devices have been developed that are placed percutaneously for temporary support: TandemHeart® (CardiacAssist, Inc., Pittsburgh, PA) and Impella® (Abiomed, Danvers, MA)157-159. The Impella has a unique design with its small axial flow pump being located on the end of the catheter where it sits in the ventricle and its adaptation for RV support, the Impella RP, recently received approval in Europe with a trial in the US to start soon160;161.

Lung transplantation

24

Copyright © 2014 by the American Thoracic Society

Page 25 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Lung transplantation should be considered for severe PAH if the RV is not severely dysfunctional (typically defined as the need for inotropic support), although such cases represent only 3% of all lung transplants162. Combined heart-lung transplantation can occasionally be considering in cases of severe PAH with RV failure although less than 100 cases are performed worldwide per year. Median survival after lung transplant is 5 years for PAH, which improves to 10 years for patients that survive the first year after transplant; similar results are seen for heart-lung transplant163. The limited number of organs available requires better treatment strategies for RV failure in PAH.

Conclusions

The first decade of the 21st century finds the community of PAH trialists and researchers in a peculiar situation: The available drugs have some impact on patient survival but do not alter the remodeled lung circulation. Conversely, effective treatments targeting the lung vessels could have detrimental effects on the failing RV. Thus, the development of PAH treatments that both reduce pulmonary vascular resistance and improve RV function is not straightforward.

There are contrasting priorities within the different cell populations of heart and lungs. For example, we look for increased contractility in cardiomyocytes but relaxation in pulmonary vascular smooth muscle cells. On the other hand, we look for apoptosis of the pulmonary endothelial cells in plexiform lesions, whilst we need to promote cardiomyocyte and cardiac endothelial cells survival. It is very well possible to define common goals, however. Mitigating the inflammatory response, preventing ROS/RNS disequilibrium and reversing extracellular matrix remodeling are likely to be beneficial in

25

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 26 of 55

both the heart and the pulmonary circulation. Aldosterone receptor blockers (and other inhibitors of the Renin-Angiotensin-Aldosterone system), BH4, statins and EGF-R blockers could fulfill the task. It may also be possible to specifically target the heart, without affecting pulmonary vasculature. Specific support of RV function can perhaps be accomplished directly with carvedilol or metabolic modulators. Other important effects that ought to be considered for the treatment of RV failure are summarized in Figure 2.

Finally, it is important to remember that PAH is a cardiopulmonary disease. Thus, as new treatments arise, trialists should not only focus on the lung circulation, but also consider every potential positive or negative effect that a drug may have on the RV.

Disclosures: None.

26

Copyright © 2014 by the American Thoracic Society

Page 27 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Reference List 1. D'Alonzo, G. E., R. J. Barst, S. M. Ayres, E. H. Bergofsky, B. H. Brundage, K. M. Detre, A. P. Fishman, R. M. Goldring, B. M. Groves, J. T. Kernis, et al. 1991. Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. Ann.Intern.Med. 115:343-349.

2. Sandoval, J., O. Bauerle, A. Palomar, A. Gomez, M. L. Martinez-Guerra, M. Beltran, and M. L. Guerrero. 1994. Survival in primary pulmonary hypertension. Validation of a prognostic equation. Circulation. 89:1733-1744.

3. van de Veerdonk, T. Kind, J. T. Marcus, G. J. Mauritz, M. W. Heymans, H. J. Bogaard, A. Boonstra, K. M. Marques, N. Westerhof, and A. Vonk-Noordegraaf. 2011. Progressive right ventricular dysfunction in patients with pulmonary arterial hypertension responding to therapy. J.Am.Coll.Cardiol. 58:2511-2519.

4. Bogaard, H. J., K. Abe, A. Vonk-Noordegraaf, and N. F. Voelkel. 2009. The right ventricle under pressure; cellular and molecular mechanisms of right heart failure in pulmonary hypertension. Chest 135:794-804.

5. Voelkel, N. F., J. G. Gomez-Arroyo, A. Abbate, H. J. Bogaard, and M. R. Nicolls. 2012. Pathobiology of pulmonary arterial hypertension and right ventricular failure. Eur Respir J. 40:1555-1565.

6. Haddad, F., R. Doyle, D. J. Murphy, and S. A. Hunt. 2008. Right ventricular function in cardiovascular disease, part II: pathophysiology, clinical importance, and management of right ventricular failure. Circulation 117:1717-1731.

7. The Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS) endorsed by the International Society of Heart and Lung Transplantation (ISHLT), N. Galie, M. M. Hoeper, M. Humbert, A. Torbicki, J. L. Vachiery, J. A. Barbera, M. Beghetti,

27

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 28 of 55

P. Corris, S. Gaine, et al. 2009. Guidelines for the diagnosis and treatment of pulmonary hypertension. European Respiratory Journal 34:1219-1263.

8. Galie, N., P. A. Corris, A. Frost, R. E. Girgis, J. Granton, Z. C. Jing, W. Klepetko, M. D. McGoon, V. V. McLaughlin, I. R. Preston, et al. 2013. Updated treatment algorithm of pulmonary arterial hypertension. J.Am.Coll.Cardiol. 62:D60-D72.

9. Spruijt, O. A., H. J. Bogaard, and A. Vonk-Noordegraaf. 2014. Assessment of right ventricular responses to therapy in pulmonary hypertension. Drug Discov.Today. (in press)

10. LEE, S. H. and L. J. Rubin. 2005. Current treatment strategies for pulmonary arterial hypertension. J Internal Med 258:199-215.

11. Gomez, A., D. Bialostozky, A. Zajarias, E. Santos, A. Palomar, M. L. Martinez, and J. Sandoval. 2001. Right ventricular ischemia in patients with primary pulmonary hypertension. J Am.Coll.Cardiol. 38:1137-1142.

12. Rich, S., E. Kaufmann, and P. S. Levy. 1992. The effect of high doses of calcium-channel blockers on survival in primary pulmonary hypertension. N Engl J Med 327:76-81.

13. Olsson, K. M., M. Delcroix, H. A. Ghofrani, H. Tiede, D. Huscher, R. Speich, E. Grunig, G. Staehler, S. Rosenkranz, M. Halank, et al. 2014. Anticoagulation and survival in pulmonary arterial hypertension: results from the Comparative, Prospective Registry of Newly Initiated Therapies for Pulmonary Hypertension (COMPERA). Circulation 129:5765.

14. Rich, S., M. Seidlitz, E. Dodin, D. Osimani, D. Judd, D. Genthner, V. McLaughlin, and G. Francis. 1998. The short-term effects of digoxin in patients with right ventricular dysfunction from pulmonary hypertension. Chest. 114:787-792.

28

Copyright © 2014 by the American Thoracic Society

Page 29 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

15. Tang, W. H. W., A. C. Parameswaran, A. P. Maroo, and G. S. Francis. 2005. Aldosterone Receptor Antagonists in the Medical Management of Chronic Heart Failure. Mayo Clinic Proceedings 80:1623-1630.

16. Paramothayan, N. S., T. J. Lasserson, A. U. Wells, and E. H. Walters. 2005. Prostacyclin for pulmonary hypertension in adults. Cochrane.Database.Syst.Rev.CD002994.

17. Olschewski, H., F. Rose, R. Schermuly, H. A. Ghofrani, B. Enke, A. Olschewski, and W. Seeger. 2004. Prostacyclin and its analogues in the treatment of pulmonary hypertension. Pharmacol.Ther. 102:139-153.

18. Achcar, R. O., G. L. Yung, H. Saffer, C. D. Cool, N. F. Voelkel, and E. S. Yi. 2006. Morphologic changes in explanted lungs after prostacyclin therapy for pulmonary hypertension. Eur J Med Res. 11:203-207.

19. Stacher, E., B. B. Graham, J. M. Hunt, A. Gandjeva, S. D. Groshong, V. V. McLaughlin, M. Jessup, W. E. Grizzle, M. A. Aldred, C. D. Cool, et al. 2012. Modern Age Pathology of Pulmonary Arterial Hypertension. Am.J Respir Crit Care Med. 186:261-272.

20. Pogoriler, J. E., S. Rich, S. L. Archer, and A. N. Husain. 2012. Persistence of complex vascular lesions despite prolonged prostacyclin therapy of pulmonary arterial hypertension. Histopathology.10-2559.

21. Yui, Y., H. Nakajima, C. Kawai, and T. Murakami. 1982. Prostacyclin therapy in patients with congestive heart failure. Am.J.Cardiol. 50:320-324.

22. Brittain, E. L., M. E. Pugh, L. A. Wheeler, I. M. Robbins, J. E. Loyd, J. H. Newman, E. D. Austin, and A. R. Hemnes. 2013. Prostanoids But Not Oral Therapies Improve Right Ventricular Function in Pulmonary Arterial Hypertension. JACC.Heart Fail. 1:300-307.

29

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 30 of 55

23. Kerbaul, F., S. Brimioulle, B. Rondelet, C. Dewachter, I. Hubloue, and R. Naeije. 2007. How prostacyclin improves cardiac output in right heart failure in conjunction with pulmonary hypertension. Am.J.Respir.Crit Care Med. 175:846-850.

24. van Albada, M. E., R. M. Berger, M. Niggebrugge, V. R. van, A. H. Cromme-Dijkhuis, and R. G. Schoemaker. 2006. Prostacyclin therapy increases right ventricular capillarisation in a model for flow-associated pulmonary hypertension. Eur.J Pharmacol. 549:107-116.

25. Syed, A. A., J. Gomez-Arroyo, M. Sakagami, L. Farkas, D. Farkas, A. A. Al-Hussaini, D. Kraskauskas, P. Byron, and N. F. Voelkel. 2012. Inhaled Iloprost Improves Right Ventricular Function In The Sugen5416/Hypoxia Model Of Severe Pulmonary Hypertension. Am.J.Respir.Crit.Care Med. 185:A3415.

26. Hara, A., K. Yuhki, T. Fujino, T. Yamada, K. Takayama, S. Kuriyama, O. Takahata, H. Karibe, Y. Okada, C. Y. Xiao, et al. 2005. Augmented cardiac hypertrophy in response to pressure overload in mice lacking the prostaglandin I2 receptor. Circulation. 112:84-92.

27. Francois, H., K. Athirakul, D. Howell, R. Dash, L. Mao, H. S. Kim, H. A. Rockman, G. A. Fitzgerald, B. H. Koller, and T. M. Coffman. 2005. Prostacyclin protects against elevated blood pressure and cardiac fibrosis. Cell Metab. 2:201-207.

28. Sueta, C. A., M. Gheorghiade, J. Adams, R. C. Bourge, S. Murali, B. F. Uretsky, M. R. Pritzker, M. D. McGoon, S. M. Butman, S. H. Grossman, et al. 1995. Safety and efficacy of epoprostenol in patients with severe congestive heart failure. Am J Cardiol 75:34A43A.

29. Califf, R. M., K. F. Adams, W. J. McKenna, M. Gheorghiade, B. F. Uretsky, S. E. McNulty, H. Darius, K. Schulman, F. Zannad, E. Handberg-Thurmond, et al. 1997. A randomized controlled trial of epoprostenol therapy for severe congestive heart failure: The Flolan International Randomized Survival Trial (FIRST). Am.Heart J. 134:44-54.

30

Copyright © 2014 by the American Thoracic Society

Page 31 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

30. Barst, R. J., M. McGoon, V. McLaughlin, V. Tapson, R. Oudiz, S. Shapiro, I. M. Robbins, R. Channick, D. Badesch, B. K. Rayburn, et al. 2003. Beraprost therapy for pulmonary arterial hypertension. J Am.Coll.Cardiol. 41:2119-2125.

31. Giannessi, D., R. S. Del, and R. L. Vitale. 2001. The role of endothelins and their receptors in heart failure. Pharmacol.Res. 43:111-126.

32. Cody, R. J., G. J. Haas, P. F. Binkley, Q. Capers, and R. Kelley. 1992. Plasma endothelin correlates with the extent of pulmonary hypertension in patients with chronic congestive heart failure. Circulation. 85:504-509.

33. Sakai, S., T. Miyauchi, T. Sakurai, Y. Kasuya, M. Ihara, I. Yamaguchi, K. Goto, and Y. Sugishita. 1996. Endogenous endothelin-1 participates in the maintenance of cardiac function in rats with congestive heart failure. Marked increase in endothelin-1 production in the failing heart. Circulation. 93:1214-1222.

34. Stewart, D. J., R. D. Levy, P. Cernacek, and D. Langleben. 1991. Increased plasma endothelin-1 in pulmonary hypertension: marker or mediator of disease? Ann.Intern.Med. 114:464-469.

35. Sugden, P. H. 2003. An overview of endothelin signaling in the cardiac myocyte. J Mol.Cell Cardiol. 35:871-886.

36. Hilal-Dandan, R., M. T. Ramirez, S. Villegas, A. Gonzalez, Y. Endo-Mochizuki, J. H. Brown, and L. L. Brunton. 1997. Endothelin ETA receptor regulates signaling and ANF gene expression via multiple G protein-linked pathways. Am.J Physiol. 272:H130-H137.

37. Kawamura, T., K. Ono, T. Morimoto, M. Akao, E. Iwai-Kanai, H. Wada, N. Sowa, T. Kita, and K. Hasegawa. 2004. Endothelin-1-Dependent Nuclear Factor of Activated T Lymphocyte Signaling Associates With Transcriptional Coactivator p300 in the Activation of the B Cell Leukemia-2 Promoter in Cardiac Myocytes. Circ Res 94:1492-1499.

31

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 32 of 55

38. Asakura, M., M. Kitakaze, S. Takashima, Y. Liao, F. Ishikura, T. Yoshinaka, H. Ohmoto, K. Node, K. Yoshino, H. Ishiguro, et al. 2002. Cardiac hypertrophy is inhibited by antagonism of ADAM12 processing of HB-EGF: metalloproteinase inhibitors as a new therapy. Nat.Med. 8:35-40.

39. Anand, I., J. McMurray, J. N. Cohn, M. A. Konstam, T. Notter, K. Quitzau, F. Ruschitzka, and T. F. Luscher. 2004. Long-term effects of darusentan on left-ventricular remodelling and clinical outcomes in the EndothelinA Receptor Antagonist Trial in Heart Failure (EARTH): randomised, double-blind, placebo-controlled trial. Lancet. 364:347-354.

40. Kalra, P. R., J. C. Moon, and A. J. Coats. 2002. Do results of the ENABLE (Endothelin Antagonist Bosentan for Lowering Cardiac Events in Heart Failure) study spell the end for non-selective endothelin antagonism in heart failure? Int.J.Cardiol. 85:195-197.

41. Kaluski, E., G. Cotter, M. Leitman, O. Milo-Cotter, R. Krakover, I. Kobrin, T. Moriconi, M. Rainisio, A. Caspi, L. Reizin, et al. 2008. Clinical and hemodynamic effects of bosentan dose optimization in symptomatic heart failure patients with severe systolic dysfunction, associated with secondary pulmonary hypertension--a multi-center randomized study. Cardiology. 109:273-280.

42. Nagendran, J., G. Sutendra, I. Paterson, H. C. Champion, L. Webster, B. Chiu, A. Haromy, I. M. Rebeyka, D. B. Ross, and E. D. Michelakis. 2013. Endothelin axis is upregulated in human and rat right ventricular hypertrophy. Circ.Res. 112:347-354.

43. McFarlane, S. I., N. Winer, and J. R. Sowers. 2003. Role of the natriuretic peptide system in cardiorenal protection. Arch.Intern.Med. 163:2696-2704.

44. Moreland, R. B., I. Goldstein, I, N. N. Kim, and A. Traish. 1999. Sildenafil Citrate, a Selective Phosphodiesterase Type 5 Inhibitor. Trends Endocrinol.Metab. 10:97-104.

32

Copyright © 2014 by the American Thoracic Society

Page 33 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

45. Galie, N., H. A. Ghofrani, A. Torbicki, R. J. Barst, L. J. Rubin, D. Badesch, T. Fleming, T. Parpia, G. Burgess, A. Branzi, et al. 2005. Sildenafil Citrate Therapy for Pulmonary Arterial Hypertension. N Engl J Med 353:2148-2157.

46. Galie, N., B. H. Brundage, H. A. Ghofrani, R. J. Oudiz, G. Simonneau, Z. Safdar, S. Shapiro, R. J. White, M. Chan, A. Beardsworth, et al. 2009. Tadalafil therapy for pulmonary arterial hypertension. Circulation 119:2894-2903.

47. Ghofrani, H. A., N. Galie, F. Grimminger, E. Grunig, M. Humbert, Z. C. Jing, A. M. Keogh, D. Langleben, M. O. Kilama, A. Fritsch, et al. 2013. Riociguat for the treatment of pulmonary arterial hypertension. N.Engl.J.Med. 369:330-340.

48. Das, A., L. Xi, and R. C. Kukreja. 2005. Phosphodiesterase-5 inhibitor sildenafil preconditions adult cardiac myocytes against necrosis and apoptosis. Essential role of nitric oxide signaling. J Biol.Chem. 280:12944-12955.

49. Fisher, P. W., F. Salloum, A. Das, H. Hyder, and R. C. Kukreja. 2005. Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation. 111:1601-1610.

50. Zahabi, A., S. Picard, N. Fortin, T. L. Reudelhuber, and C. F. Deschepper. 2003. Expression of constitutively active guanylate cyclase in cardiomyocytes inhibits the hypertrophic effects of isoproterenol and aortic constriction on mouse hearts. J Biol.Chem. 278:47694-47699.

51. Fiedler, B., S. M. Lohmann, A. Smolenski, S. Linnemuller, B. Pieske, F. Schroder, J. D. Molkentin, H. Drexler, and K. C. Wollert. 2002. Inhibition of calcineurin-NFAT hypertrophy signaling by cGMP-dependent protein kinase type I in cardiac myocytes. Proc.Natl.Acad.Sci.U.S.A. 99:11363-11368.

33

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 34 of 55

52. Takimoto, E., H. C. Champion, M. Li, D. Belardi, S. Ren, E. R. Rodriguez, D. Bedja, K. L. Gabrielson, Y. Wang, and D. A. Kass. 2005. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat.Med. 11:214-222.

53. Hare, J. M. and J. S. Stamler. 2005. NO/redox disequilibrium in the failing heart and cardiovascular system. J.Clin.Invest. 115:509-517.

54. Semigran, M. J. 2005. Type 5 phosphodiesterase inhibition: the focus shifts to the heart. Circulation. 112:2589-2591.

55. Zaccolo, M. and M. A. Movsesian. 2007. cAMP and cGMP signaling cross-talk: role of phosphodiesterases and implications for cardiac pathophysiology. Circ.Res. 100:15691578.

56. Nagendran, J., S. L. Archer, D. Soliman, V. Gurtu, R. Moudgil, A. Haromy, C. Aubin, L. Webster, I. M. Rebeyka, D. B. Ross, et al. 2007. Phosphodiesterase Type 5 Is Highly Expressed in the Hypertrophied Human Right Ventricle, and Acute Inhibition of Phosphodiesterase Type 5 Improves Contractility. Circulation 116:238-248.

57. Lewis, G. D., J. Lachmann, J. Camuso, J. J. Lepore, J. Shin, M. E. Martinovic, D. M. Systrom, K. D. Bloch, and M. J. Semigran. 2007. Sildenafil Improves Exercise Hemodynamics and Oxygen Uptake in Patients With Systolic Heart Failure. Circulation 115:59-66.

58. Guazzi, M., M. Vicenzi, R. Arena, and M. D. Guazzi. 2011. PDE5 inhibition with sildenafil improves left ventricular diastolic function, cardiac geometry, and clinical status in patients with stable systolic heart failure: results of a 1-year, prospective, randomized, placebo-controlled study. Circ.Heart Fail. 4:8-17.

59. Guazzi, M., M. Vicenzi, R. Arena, and M. D. Guazzi. 2011. Pulmonary hypertension in heart failure with preserved ejection fraction: a target of phosphodiesterase-5 inhibition in a 1-year study. Circulation. 124:164-174.

34

Copyright © 2014 by the American Thoracic Society

Page 35 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

60. Redfield, M. M., H. H. Chen, B. A. Borlaug, M. J. Semigran, K. L. Lee, G. Lewis, M. M. LeWinter, J. L. Rouleau, D. A. Bull, D. L. Mann, et al. 2013. Effect of phosphodiesterase5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA 309:1268-1277.

61. Bonderman, D., S. Ghio, S. B. Felix, H. A. Ghofrani, E. Michelakis, V. Mitrovic, R. J. Oudiz, F. Boateng, A. V. Scalise, L. Roessig, et al. 2013. Riociguat for patients with pulmonary hypertension caused by systolic left ventricular dysfunction: a phase IIb double-blind, randomized, placebo-controlled, dose-ranging hemodynamic study. Circulation 128:502-511.

62. Gan, C. T.-J., S. Holverda, J. T. Marcus, W. J. Paulus, K. M. Marques, J. G. F. Bronzwaer, J. W. Twisk, A. Boonstra, P. E. Postmus, and A. Vonk-Noordegraaf. 2007. Right Ventricular Diastolic Dysfunction and the Acute Effects of Sildenafil in Pulmonary Hypertension Patients. Chest 132:11-17.

63. Bogaard, H. J., R. Natarajan, S. C. Henderson, C. S. Long, D. Kraskauskas, L. Smithson, R. Ockaili, J. M. McCord, and N. F. Voelkel. 2009. Chronic pulmonary artery pressure elevation is insufficient to explain right heart failure. Circulation. 120:1951-1960.

64. Guihaire, J., H. J. Bogaard, E. Flecher, P. E. Noly, O. Mercier, F. Haddad, and E. Fadel. 2013. Experimental models of right heart failure: a window for translational research in pulmonary hypertension. Semin.Respir.Crit Care Med. 34:689-699.

65. Gomez-Arroyo, J., S. J. Saleem, S. Mizuno, A. A. Syed, H. J. Bogaard, A. Abbate, L. Taraseviciene-Stewart, Y. Sung, D. Kraskauskas, D. Farkas, et al. 2012. A brief overview of mouse models of pulmonary arterial hypertension: problems and prospects. Am J Physiol Lung Cell Mol.Physiol. 302:L977-L991.

66. Bogaard, H. J., S. Mizuno, A. A. Al Hussaini, S. Toldo, A. Abbate, D. Kraskauskas, M. Kasper, R. Natarajan, and N. F. Voelkel. 2011. Suppression of Histone Deacetylases

35

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 36 of 55

Worsens Right Ventricular Dysfunction after Pulmonary Artery Banding in Rats. Am.J.Respir.Crit.Care Med. 183:1402-1410.

67. Schermuly, R. T., E. Dony, H. A. Ghofrani, S. Pullamsetti, R. Savai, M. Roth, A. Sydykov, Y. J. Lai, N. Weissmann, W. Seeger, et al. 2005. Reversal of experimental pulmonary hypertension by PDGF inhibition. J.Clin.Invest. 115:2811-2821.

68. Ghofrani, H. A., W. Seeger, and F. Grimminger. 2005. Imatinib for the Treatment of Pulmonary Arterial Hypertension. N Engl J Med 353:1412-1413.

69. Overbeek, M. J., G. P. van Nieuw Amerongen, A. Boonstra, E. F. Smit, and A. VonkNoordegraaf. 2008. Possible role of imatinib in clinical pulmonary veno-occlusive disease. Eur.Respir.J. 32:232-235.

70. Patterson, K. C., A. Weissmann, T. Ahmadi, and H. W. Farber. 2006. Imatinib mesylate in the treatment of refractory idiopathic pulmonary arterial hypertension. Ann.Intern.Med. 145:152-153.

71. Souza, R., O. Sitbon, F. Parent, G. Simonneau, and M. Humbert. 2006. Long term imatinib treatment in pulmonary arterial hypertension. Thorax. 61:736.

72. Ghofrani, H. A., N. W. Morrell, M. M. Hoeper, H. Olschewski, A. J. Peacock, R. J. Barst, S. Shapiro, H. Golpon, M. Toshner, F. Grimminger, et al. 2010. Imatinib in pulmonary arterial hypertension patients with inadequate response to established therapy. Am.J.Respir.Crit Care Med. 182:1171-1177.

73. Hoeper, M. M., R. J. Barst, R. C. Bourge, J. Feldman, A. E. Frost, N. Galie, M. A. GomezSanchez, F. Grimminger, E. Grunig, P. M. Hassoun, et al. 2013. Imatinib mesylate as add-on therapy for pulmonary arterial hypertension: results of the randomized IMPRES study. Circulation 127:1128-1138.

36

Copyright © 2014 by the American Thoracic Society

Page 37 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

74. Edelberg, J. M., S. H. LEE, M. Kaur, L. Tang, N. M. Feirt, S. McCabe, O. Bramwell, S. C. Wong, and M. K. Hong. 2002. Platelet-derived growth factor-AB limits the extent of myocardial infarction in a rat model: feasibility of restoring impaired angiogenic capacity in the aging heart. Circulation. 105:608-613.

75. Hsieh, P. C., C. MacGillivray, J. Gannon, F. U. Cruz, and R. T. Lee. 2006. Local controlled intramyocardial delivery of platelet-derived growth factor improves postinfarction ventricular function without pulmonary toxicity. Circulation. 114:637-644.

76. Tang, J., K. Kozaki, A. G. Farr, P. J. Martin, P. Lindahl, C. Betsholtz, and E. W. Raines. 2005. The absence of platelet-derived growth factor-B in circulating cells promotes immune and inflammatory responses in atherosclerosis-prone ApoE-/- mice. Am.J.Pathol. 167:901-912.

77. Xaymardan, M., L. Tang, L. Zagreda, B. Pallante, J. Zheng, J. L. Chazen, A. Chin, I. Duignan, P. Nahirney, S. Rafii, et al. 2004. Platelet-derived growth factor-AB promotes the generation of adult bone marrow-derived cardiac myocytes. Circ.Res. 94:E39-E45.

78. Beltrami, A. P., L. Barlucchi, D. Torella, M. Baker, F. Limana, S. Chimenti, H. Kasahara, M. Rota, E. Musso, K. Urbanek, et al. 2003. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 114:763-776.

79. Shah, A. M., P. Campbell, G. Q. Rocha, A. Peacock, R. J. Barst, D. Quinn, and S. D. Solomon. 2014. Effect of imatinib as add-on therapy on echocardiographic measures of right ventricular function in patients with significant pulmonary arterial hypertension. Eur.Heart J. (In press)

80. Grossmann, C., A. W. Krug, R. Freudinger, S. Mildenberger, K. Volker, and M. Gekle. 2007. Aldosterone-induced Epidermal Growth Factor Receptor (EGFR) expression: Interaction Between the Human Mineralocorticoid Receptor and the Human EGFR Promoter. Am.J Physiol Endocrinol.Metab. 292:E1790-E1800.

37

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 38 of 55

81. Mazak, I., A. Fiebeler, D. N. Muller, J. K. Park, E. Shagdarsuren, C. Lindschau, R. Dechend, C. Viedt, B. Pilz, H. Haller, et al. 2004. Aldosterone potentiates angiotensin IIinduced signaling in vascular smooth muscle cells. Circulation. 109:2792-2800.

82. Merklinger, S. L., P. L. Jones, E. C. Martinez, and M. Rabinovitch. 2005. Epidermal growth factor receptor blockade mediates smooth muscle cell apoptosis and improves survival in rats with pulmonary hypertension. Circulation. 112:423-431.

83. Nicolls, M. R., S. Mizuno, L. Taraseviciene-Stewart, L. Farkas, J. I. Drake, A. A. Al Hussaini, J. Gomez-Arroyo, N. F. Voelkel, and H. J. Bogaard. 2012. New models of pulmonary hypertension based on VEGF receptor blockade-induced endothelial cell apoptosis. Pulm.Circ. 2:434-442.

84. Montani, D., A. Seferian, L. Savale, G. Simonneau, and M. Humbert. 2013. Drug-induced pulmonary arterial hypertension: a recent outbreak. Eur.Respir.Rev. 22:244-250.

85. Fukumoto, Y., T. Matoba, A. Ito, H. Tanaka, T. Kishi, S. Hayashidani, K. Abe, A. Takeshita, and H. Shimokawa. 2005. Acute vasodilator effects of a Rho-kinase inhibitor, fasudil, in patients with severe pulmonary hypertension. Br.Heart J. 91:391-392.

86. Ishikura, K., N. Yamada, M. Ito, S. Ota, M. Nakamura, N. Isaka, and T. Nakano. 2006. Beneficial Acute Effects of Rho-Kinase Inhibitor in Patients With Pulmonary Arterial Hypertension. Circ J 70:174-178.

87. Abe, K., H. Shimokawa, K. Morikawa, T. Uwatoku, K. Oi, Y. Matsumoto, T. Hattori, Y. Nakashima, K. Kaibuchi, K. Sueishi, et al. 2004. Long-Term Treatment With a RhoKinase Inhibitor Improves Monocrotaline-Induced Fatal Pulmonary Hypertension in Rats. Circ Res 94:385-393.

88. Fagan, K. A., M. Oka, N. R. Bauer, S. A. Gebb, D. D. Ivy, K. G. Morris, and I. F. McMurtry. 2004. Attenuation of acute hypoxic pulmonary vasoconstriction and hypoxic

38

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 39 of 55

pulmonary hypertension in mice by inhibition of Rho-kinase. Am J Physiol Lung Cell Mol Physiol 287:L656-L664.

89. Nagaoka, T., S. A. Gebb, V. Karoor, N. Homma, K. G. Morris, I. F. McMurtry, and M. Oka. 2006. Involvement of RhoA/Rho kinase signaling in pulmonary hypertension of the fawn-hooded rat. J Appl Physiol 100:996-1002.

90. Abe, K., S. Tawara, K. Oi, T. Hizume, T. Uwatoku, Y. Fukumoto, K. Kaibuchi, and H. Shimokawa. 2006. Long-term inhibition of Rho-kinase ameliorates hypoxia-induced pulmonary hypertension in mice. J Cardiovasc Pharmacol 48:280-285.

91. Oka, M., N. Homma, L. Taraseviciene-Stewart, K. G. Morris, D. Kraskauskas, N. Burns, N. F. Voelkel, and I. F. McMurtry. 2007. Rho kinase-mediated vasoconstriction is important in severe occlusive pulmonary arterial hypertension in rats. Circ.Res. 100:923929.

92. Takemoto, M., K. Node, H. Nakagami, Y. Liao, M. Grimm, Y. Takemoto, M. Kitakaze, and J. K. Liao. 2001. Statins as antioxidant therapy for preventing cardiac myocyte hypertrophy. J.Clin.Invest. 108:1429-1437.

93. Lipinski, M. J., A. Abbate, V. Fuster, and G. W. Vetrovec. 2007. Drug insight: statins for nonischemic heart failure--evidence and potential mechanisms. Nat.Clin Pract.Cardiovasc.Med. 4:196-205.

94. Taraseviciene-Stewart, L., R. Scerbavicius, K. H. Choe, C. Cool, K. Wood, R. M. Tuder, N. Burns, M. Kasper, and N. F. Voelkel. 2006. Simvastatin Causes Endothelial Cell Apoptosis and Attenuates Severe Pulmonary Hypertension. Am J Physiol Lung Cell Mol Physiol 291:L668-L676.

95. Kawut, S. M., E. Bagiella, D. J. Lederer, D. Shimbo, E. M. Horn, K. E. Roberts, N. S. Hill, R. G. Barr, E. B. Rosenzweig, W. Post, et al. 2011. Randomized clinical trial of aspirin

39

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 40 of 55

and simvastatin for pulmonary arterial hypertension: ASA-STAT. Circulation. 123:29852993.

96. Fischle, W., Y. Wang, and C. D. Allis. 2003. Histone and chromatin cross-talk. Curr.Opin.Cell Biol. 15:172-183.

97. Zhao, L., C. N. Chen, N. Hajji, E. Oliver, E. Cotroneo, J. Wharton, D. Wang, M. Li, T. A. McKinsey, K. R. Stenmark, et al. 2012. Histone Deacetylation Inhibition in Pulmonary Hypertension: Therapeutic Potential of Valproic Acid (VPA) and Suberoylanilide Hydroxamic Acid (SAHA). Circulation. 126:455-467.

98. Antos, C. L., T. A. McKinsey, M. Dreitz, L. M. Hollingsworth, C. L. Zhang, K. Schreiber, H. Rindt, R. J. Gorczynski, and E. N. Olson. 2003. Dose-dependent blockade to cardiomyocyte hypertrophy by histone deacetylase inhibitors. J.Biol.Chem. 278:2893028937.

99. Trivedi, C. M., Y. Luo, Z. Yin, M. Zhang, W. Zhu, T. Wang, T. Floss, M. Goettlicher, P. R. Noppinger, W. Wurst, et al. 2007. Hdac2 regulates the cardiac hypertrophic response by modulating Gsk3 beta activity. Nat.Med. 13:324-331.

100. Ciarka, A., V. Doan, S. Velez-Roa, R. Naeije, and B. P. van de. 2010. Prognostic Significance of Sympathetic Nervous System Activation in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med. 181:1269-1275.

101. Bristow, M. R., W. Minobe, R. Rasmussen, P. Larrabee, L. Skerl, J. W. Klein, F. L. Anderson, J. Murray, L. Mestroni, and S. V. Karwande. 1992. Beta-adrenergic neuroeffector abnormalities in the failing human heart are produced by local rather than systemic mechanisms. J.Clin.Invest. 89:803-815.

102. Dickstein, K., A. Cohen-Solal, G. Filippatos, J. J. McMurray, P. Ponikowski, P. A. PooleWilson, A. Stromberg, D. J. van Veldhuisen, D. Atar, A. W. Hoes, et al. 2008. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2008: the

40

Copyright © 2014 by the American Thoracic Society

Page 41 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Task Force for the diagnosis and treatment of acute and chronic heart failure 2008 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association of the ESC (HFA) and endorsed by the European Society of Intensive Care Medicine (ESICM). Eur.J.Heart Fail. 10:933-989.

103. Provencher, S., P. Herve, X. Jais, D. Lebrec, M. Humbert, G. Simonneau, and O. Sitbon. 2006. Deleterious effects of beta-blockers on exercise capacity and hemodynamics in patients with portopulmonary hypertension. Gastroenterology. 130:120-126.

104. Reiken, S., X. H. Wehrens, J. A. Vest, A. Barbone, S. Klotz, D. Mancini, D. Burkhoff, and A. R. Marks. 2003. Beta-blockers restore calcium release channel function and improve cardiac muscle performance in human heart failure. Circulation. 107:2459-2466.

105. Tongers, J., B. Schwerdtfeger, G. Klein, T. Kempf, A. Schaefer, J. M. Knapp, M. Niehaus, T. Korte, and M. M. Hoeper. 2007. Incidence and clinical relevance of supraventricular tachyarrhythmias in pulmonary hypertension. Am.Heart J. 153:127-132.

106. So, P. P., R. A. Davies, G. Chandy, D. Stewart, R. S. Beanlands, H. Haddad, C. Pugliese, and L. M. Mielniczuk. 2012. Usefulness of beta-blocker therapy and outcomes in patients with pulmonary arterial hypertension. Am.J Cardiol. 109:1504-1509.

107. Drake, J. I., H. J. Bogaard, S. Mizuno, B. Clifton, B. Xie, Y. Gao, C. I. Dumur, P. Fawcett, N. F. Voelkel, and R. Natarajan. 2011. Molecular Signature of a Right Heart Failure Program in Chronic Severe Pulmonary Hypertension. Am.J.Respir.Cell Mol.Biol. 45:1239-1247.

108. Bogaard, H. J., R. Natarajan, S. Mizuno, A. Abbate, P. Chang, V. Chau, N. Hoke, D. Kraskauskas, M. Kasper, F. Salloum, et al. 2010. Adrenergic receptor blockade reverses right heart remodeling and dysfunction in pulmonary hypertensive rats. Am J Respir Crit Care Med 182:652-660.

41

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 42 of 55

109. de Man, F. S., M. L. Handoko, J. J. van Ballegoij, I. Schalij, S. J. Bogaards, P. E. Postmus, d. van, V, N. Westerhof, W. J. Paulus, and A. Vonk-Noordegraaf. 2012. Bisoprolol delays progression towards right heart failure in experimental pulmonary hypertension. Circ.Heart Fail. 5:97-105.

110. Zhou, Q., J. Xiao, D. Jiang, R. Wang, K. Vembaiyan, A. Wang, C. D. Smith, C. Xie, W. Chen, J. Zhang, et al. 2011. Carvedilol and its new analogs suppress arrhythmogenic store overload-induced Ca2+ release. Nat.Med. 17:1003-1009.

111. Ma, X. L., T. L. Yue, B. L. Lopez, F. C. Barone, T. A. Christopher, R. R. Ruffolo, Jr., and G. Z. Feuerstein. 1996. Carvedilol, a new beta adrenoreceptor blocker and free radical scavenger, attenuates myocardial ischemia-reperfusion injury in hypercholesterolemic rabbits. J Pharmacol.Exp.Ther. 277:128-136.

112. Yue, T. L., H. Y. Cheng, P. G. Lysko, P. J. McKenna, R. Feuerstein, J. L. Gu, K. A. Lysko, L. L. Davis, and G. Feuerstein. 1992. Carvedilol, a new vasodilator and beta adrenoceptor antagonist, is an antioxidant and free radical scavenger. J Pharmacol.Exp.Ther. 263:92-98.

113. Wang, R., T. Miura, N. Harada, R. Kametani, M. Shibuya, Y. Fukagawa, S. Kawamura, Y. Ikeda, M. Hara, and M. Matsuzaki. 2006. Pleiotropic effects of the beta-adrenoceptor blocker carvedilol on calcium regulation during oxidative stress-induced apoptosis in cardiomyocytes. J Pharmacol.Exp.Ther. 318:45-52.

114. Kim, I. M., D. G. Tilley, J. Chen, N. C. Salazar, E. J. Whalen, J. D. Violin, and H. A. Rockman. 2008. Beta-blockers alprenolol and carvedilol stimulate beta-arrestin-mediated EGFR transactivation. Proc.Natl.Acad.Sci.U.S.A. 105:14555-14560.

115. Drake, J. I., J. Gomez-Arroyo, C. I. Dumur, D. Kraskauskas, R. Natarajan, H. J. Bogaard, P. Fawcett, and N. F. Voelkel. 2013. Chronic carvedilol treatment partially reverses the

42

Copyright © 2014 by the American Thoracic Society

Page 43 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

right ventricular failure transcriptional profile in experimental pulmonary hypertension. Physiol Genomics 45:449-461.

116. Piao, L., Y. H. Fang, K. S. Parikh, J. J. Ryan, K. M. D'Souza, T. Theccanat, P. T. Toth, J. Pogoriler, J. Paul, B. C. Blaxall, et al. 2012. GRK2-mediated inhibition of adrenergic and dopaminergic signaling in right ventricular hypertrophy: therapeutic implications in pulmonary hypertension. Circulation 126:2859-2869.

117. Ikram, H., A. H. Maslowski, M. G. Nicholls, E. A. Espiner, and F. T. Hull. 1982. Haemodynamic and hormonal effects of captopril in primary pulmonary hypertension. Br.Heart J. 48:541-545.

118. Leier, C. V., D. Bambach, S. Nelson, J. B. Hermiller, P. Huss, R. D. Magorien, and D. V. Unverferth. 1983. Captopril in primary pulmonary hypertension. Circulation. 67:155-161.

119. Rouleau, J. L., G. Kapuku, S. Pelletier, H. Gosselin, A. Adam, C. Gagnon, C. Lambert, and S. Meloche. 2001. Cardioprotective effects of ramipril and losartan in right ventricular pressure overload in the rabbit: importance of kinins and influence on angiotensin II type 1 receptor signaling pathway. Circulation. 104:939-944.

120. de Man, F. S., L. Tu, M. L. Handoko, S. Rain, G. Ruiter, C. Francois, I. Schalij, P. Dorfmuller, G. Simonneau, E. Fadel, et al. 2012. Dysregulated Renin-AngiotensinAldosterone System Contributes to Pulmonary Arterial Hypertension. Am.J Respir Crit Care Med. 186:780-789.

121. Braun, M. U., P. Szalai, R. H. Strasser, and M. M. Borst. 2003. Right ventricular hypertrophy and apoptosis after pulmonary artery banding: regulation of PKC isozymes. Cardiovasc.Res. 59:658-667.

122. Neubauer, S. 2007. The failing heart--an engine out of fuel. N.Engl.J.Med. 356:11401151.

43

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 44 of 55

123. Piao, L., G. Marsboom, and S. L. Archer. 2010. Mitochondrial metabolic adaptation in right ventricular hypertrophy and failure. J Mol.Med (Berl). 88:1011-1020.

124. Piao, L., Y. H. Fang, V. J. Cadete, C. Wietholt, D. Urboniene, P. T. Toth, G. Marsboom, H. J. Zhang, I. Haber, J. Rehman, et al. 2010. The inhibition of pyruvate dehydrogenase kinase improves impaired cardiac function and electrical remodeling in two models of right ventricular hypertrophy: resuscitating the hibernating right ventricle. J Mol.Med (Berl). 88:47-60.

125. Gomez-Arroyo, J., S. Mizuno, K. Szczepanek, T. B. Van, R. Natarajan, C. G. dos Remedios, J. I. Drake, L. Farkas, D. Kraskauskas, D. S. Wijesinghe, et al. 2013. Metabolic gene remodeling and mitochondrial dysfunction in failing right ventricular hypertrophy secondary to pulmonary arterial hypertension. Circ.Heart Fail. 6:136-144.

126. Abel, E. D. and T. Doenst. 2011. Mitochondrial adaptations to physiological vs. pathological cardiac hypertrophy. Cardiovasc.Res. 90:234-242.

127. Fang, Y. H., L. Piao, Z. Hong, P. T. Toth, G. Marsboom, P. Bache-Wiig, J. Rehman, and S. L. Archer. 2012. Therapeutic inhibition of fatty acid oxidation in right ventricular hypertrophy: exploiting Randle's cycle. J Mol.Med (Berl). 90:31-43.

128. Ashrafian, H., M. P. Frenneaux, and L. H. Opie. 2007. Metabolic mechanisms in heart failure. Circulation. 116:434-448.

129. Giordano, F. J. 2005. Oxygen, oxidative stress, hypoxia, and heart failure. J.Clin.Invest. 115:500-508.

130. Nakaya, H., Y. Takeda, N. Tohse, and M. Kanno. 1992. Mechanism of the membrane depolarization induced by oxidative stress in guinea-pig ventricular cells. J.Mol.Cell Cardiol. 24:523-534.

44

Copyright © 2014 by the American Thoracic Society

Page 45 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

131. Kaplan, P., E. Babusikova, J. Lehotsky, and D. Dobrota. 2003. Free radical-induced protein modification and inhibition of Ca2+-ATPase of cardiac sarcoplasmic reticulum. Mol.Cell Biochem. 248:41-47.

132. Gems, D. and L. Partridge. 2008. Stress-response hormesis and aging: "that which does not kill us makes us stronger". Cell Metab. 7:200-203.

133. Cappola, T. P., D. A. Kass, G. S. Nelson, R. D. Berger, G. O. Rosas, Z. A. Kobeissi, E. Marban, and J. M. Hare. 2001. Allopurinol improves myocardial efficiency in patients with idiopathic dilated cardiomyopathy. Circulation. 104:2407-2411.

134. Hoshikawa, Y., S. Ono, S. Suzuki, T. Tanita, M. Chida, C. Song, M. Noda, T. Tabata, N. F. Voelkel, and S. Fujimura. 2001. Generation of oxidative stress contributes to the development of pulmonary hypertension induced by hypoxia. J Appl.Physiol. 90:12991306.

135. Takimoto, E., H. C. Champion, M. Li, S. Ren, E. R. Rodriguez, B. Tavazzi, G. Lazzarino, N. Paolocci, K. L. Gabrielson, Y. Wang, et al. 2005. Oxidant stress from nitric oxide synthase-3 uncoupling stimulates cardiac pathologic remodeling from chronic pressure load. J.Clin.Invest. 115:1221-1231.

136. Yet, S. F., M. A. Perrella, M. D. Layne, C. M. Hsieh, K. Maemura, L. Kobzik, P. Wiesel, H. Christou, S. Kourembanas, and M. E. Lee. 1999. Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. J.Clin.Invest. 103:R23-R29.

137. Hambrecht, R., E. Fiehn, C. Weigl, S. Gielen, C. Hamann, R. Kaiser, J. Yu, V. Adams, J. Niebauer, and G. Schuler. 1998. Regular physical exercise corrects endothelial dysfunction and improves exercise capacity in patients with chronic heart failure. Circulation 98:2709-2715.

138. Mereles, D., N. Ehlken, S. Kreuscher, S. Ghofrani, M. M. Hoeper, M. Halank, F. J. Meyer, G. Karger, J. Buss, J. Juenger, et al. 2006. Exercise and Respiratory Training Improve

45

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 46 of 55

Exercise Capacity and Quality of Life in Patients With Severe Chronic Pulmonary Hypertension. Circulation 114:1482-1489.

139. Chinsomboon, J., J. Ruas, R. K. Gupta, R. Thom, J. Shoag, G. C. Rowe, N. Sawada, S. Raghuram, and Z. Arany. 2009. The transcriptional coactivator PGC-1alpha mediates exercise-induced angiogenesis in skeletal muscle. Proc.Natl.Acad.Sci.U.S.A. 106:2140121406.

140. Tang, A. S., G. A. Wells, M. Talajic, M. O. Arnold, R. Sheldon, S. Connolly, S. H. Hohnloser, G. Nichol, D. H. Birnie, J. L. Sapp, et al. 2010. Cardiac-resynchronization therapy for mild-to-moderate heart failure. N.Engl.J.Med. 363:2385-2395.

141. Goldenberg, I., V. Kutyifa, H. U. Klein, D. S. Cannom, M. W. Brown, A. Dan, J. P. Daubert, N. A. Estes, III, E. Foster, H. Greenberg, et al. 2014. Survival with cardiacresynchronization therapy in mild heart failure. N.Engl.J.Med. 370:1694-1701.

142. Marcus, J. T., C. T. Gan, J. J. Zwanenburg, A. Boonstra, C. P. Allaart, M. J. Gotte, and A. Vonk-Noordegraaf. 2008. Interventricular mechanical asynchrony in pulmonary arterial hypertension: left-to-right delay in peak shortening is related to right ventricular overload and left ventricular underfilling. J Am Coll Cardiol. 51:750-757.

143. Handoko, M. L., R. R. Lamberts, E. M. Redout, F. S. de Man, C. Boer, W. S. Simonides, W. J. Paulus, N. Westerhof, C. P. Allaart, and A. Vonk-Noordegraaf. 2009. Right ventricular pacing improves right heart function in experimental pulmonary arterial hypertension: a study in the isolated heart. Am J Physiol Heart Circ Physiol. 297:H1752H1759.

144. Hardziyenka, M., S. Surie, J. R. de Groot, H. A. de Bruin-Bon, R. E. Knops, M. Remmelink, Z. Y. Yong, J. Baan, Jr., B. J. Bouma, P. Bresser, et al. 2011. Right ventricular pacing improves haemodynamics in right ventricular failure from pressure

46

Copyright © 2014 by the American Thoracic Society

Page 47 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

overload: an open observational proof-of-principle study in patients with chronic thromboembolic pulmonary hypertension. Europace. 13:1753-1759.

145. Sandoval, J., J. Gaspar, T. Pulido, E. Bautista, M. L. Martinez-Guerra, M. Zeballos, A. Palomar, and A. Gomez. 1998. Graded balloon dilation atrial septostomy in severe primary pulmonary hypertension. A therapeutic alternative for patients nonresponsive to vasodilator treatment. J.Am.Coll.Cardiol. 32:297-304.

146. Espinola-Zavaleta, N., J. Vargas-Barron, J. I. Tazar, J. M. Casanova, C. Keirns, A. R. Cardenas, J. Gaspar, and J. Sandoval. 1999. Echocardiographic Evaluation of Patients with Primary Pulmonary Hypertension Before and After Atrial Septostomy. Echocardiography. 16:625-634.

147. Ciarka, A., J. L. Vachiery, A. Houssiere, M. Gujic, E. Stoupel, S. Velez-Roa, R. Naeije, and P. van de Borne. 2007. Atrial septostomy decreases sympathetic overactivity in pulmonary arterial hypertension. Chest 131:1831-1837.

148. Sandoval, J. and A. Torbicki. 2014. Atrial Septostomy. In: Voelkel NF and Schranz D (Eds.).Right Ventricle in Health and disease.

149. Esch, J. J., P. B. Shah, B. A. Cockrill, H. W. Farber, M. J. Landzberg, M. R. Mehra, M. P. Mullen, A. R. Opotowsky, A. B. Waxman, J. E. Lock, et al. 2013. Transcatheter Potts shunt creation in patients with severe pulmonary arterial hypertension: initial clinical experience. J.Heart Lung Transplant. 32:381-387.

150. Berman, M., S. Tsui, A. Vuylsteke, A. Klein, and D. P. Jenkins. 2008. Life-threatening right ventricular failure in pulmonary hypertension: RVAD or ECMO? J.Heart Lung Transplant. 27:1188-1189.

151. Berman, M., S. Tsui, A. Vuylsteke, A. Snell, S. Colah, R. Latimer, R. Hall, J. E. Arrowsmith, J. Kneeshaw, A. A. Klein, et al. 2008. Successful extracorporeal membrane

47

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 48 of 55

oxygenation support after pulmonary thromboendarterectomy. Ann.Thorac.Surg. 86:1261-1267.

152. Furukawa, K., T. Motomura, and Y. Nose. 2005. Right ventricular failure after left ventricular assist device implantation: the need for an implantable right ventricular assist device. Artif.Organs 29:369-377.

153. Kaul, T. K. and D. R. Kahn. 2000. Postinfarct refractory right ventricle: right ventricular exclusion. A possible option to mechanical cardiac support, in patients unsuitable for heart transplant. J.Cardiovasc.Surg.(Torino) 41:349-355.

154. Klima, U., S. Ringes-Lichtenberg, G. Warnecke, A. Lichtenberg, M. Struber, and A. Haverich. 2005. Severe right heart failure after heart transplantation. A single-center experience. Transpl.Int. 18:326-332.

155. Moazami, N., M. K. Pasque, M. R. Moon, R. L. Herren, M. S. Bailey, J. S. Lawton, and R. J. Damiano, Jr. 2004. Mechanical support for isolated right ventricular failure in patients after cardiotomy. J.Heart Lung Transplant. 23:1371-1375.

156. Bhama, J. K., R. L. Kormos, Y. Toyoda, J. J. Teuteberg, K. R. McCurry, and M. P. Siegenthaler. 2009. Clinical experience using the Levitronix CentriMag system for temporary right ventricular mechanical circulatory support. J.Heart Lung Transplant. 28:971-976.

157. Takagaki, M., C. Wurzer, R. Wade, R. Lee, S. C. Malaisrie, P. M. McCarthy, and E. C. McGee, Jr. 2008. Successful conversion of TandemHeart left ventricular assist device to right ventricular assist device after implantation of a HeartMate XVE. Ann.Thorac.Surg. 86:1677-1679.

158. Giesler, G. M., J. S. Gomez, G. Letsou, M. Vooletich, and R. W. Smalling. 2006. Initial report of percutaneous right ventricular assist for right ventricular shock secondary to right ventricular infarction. Catheter.Cardiovasc.Interv. 68:263-266.

48

Copyright © 2014 by the American Thoracic Society

Page 49 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

159. Prutkin, J. M., J. A. Strote, and K. K. Stout. 2008. Percutaneous right ventricular assist device as support for cardiogenic shock due to right ventricular infarction. J.Invasive.Cardiol. 20:E215-E216.

160. Cheung, A. W., C. W. White, M. K. Davis, and D. H. Freed. 2014. Short-term mechanical circulatory support for recovery from acute right ventricular failure: Clinical outcomes. J.Heart Lung Transplant.(In press)

161. Goldstein, J. A. and M. J. Kern. 2012. Percutaneous mechanical support for the failing right heart. Cardiol.Clin. 30:303-310.

162. Yusen, R. D., J. D. Christie, L. B. Edwards, A. Y. Kucheryavaya, C. Benden, A. I. Dipchand, F. Dobbels, R. Kirk, L. H. Lund, A. O. Rahmel, et al. 2013. The Registry of the International Society for Heart and Lung Transplantation: thirtieth adult lung and heartlung transplant report--2013; focus theme: age. J.Heart Lung Transplant. 32:965-978.

163. Schaffer, J. M., S. K. Singh, D. L. Joyce, B. A. Reitz, R. C. Robbins, R. T. Zamanian, and H. R. Mallidi. 2013. Transplantation for idiopathic pulmonary arterial hypertension: improvement in the lung allocation score era. Circulation 127:2503-2513.

49

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 50 of 55

Table 1: Proposed direct cardiac effects of current PAH therapies and their impact on the right ventricle Proposed Direct Cardiac

Impact on the right

Effects

ventricle

Treatment ↑Angiogenesis Prostacyclin Analogues

Positive effect ↓Fibrosis ↑Maladaptive hypertrophy

Endothelin receptor blockers

Negative effect ↑Cardiomyocyte apoptosis ↓Apoptosis

PDE-5 inhibitors/sGC inducers

↑Myocardial relaxation

Positive effect

↑Contractility ↓Angiogenesis Tyrosine kinase inhibitors

↑Mitochondrial Dysfunction

Negative effect

↑Apoptosis ↓Apoptosis Positive effect ↓Inflammation Rho-kinase inhibitors

↓Contractility Excessive reduction of

Negative effect

hypertrophy ↓ROS production Statins

↑Nitric oxide production

Positive effect

↓Inflammation

50

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Page 51 of 55

Table 2: Potential RV-targeted therapies and the direct or indirect effects on the right ventricle Drug

Carvedilol, bisoprolol

Proposed effects on RV

Evidence for use in

function

PAH

↑Angiogenesis

Only experimental

↓Heart rate

evidence in animal

↓Fibrosis

models

Metabolic modulator ↓Maladaptive

Natriuretic peptides

No evidence

hypertrophy ↓RV Volume-overload (by diuresis) ↓Apoptosis ↓Fibrosis Metabolic modulator Systemic vasodilators Blockers of the Renin-Angiotensin-

↓Remodelling of

Aldosterone Axis

extracellular matrix

No evidence

↓Fibrosis HDAC inhibitors (Trichostatin-A

↑Fibrosis

Only experimental

and Valproic acid)

↓Angiogenesis

evidence in animal

↑Cardiomyocyte

models

apoptosis Antioxidants (BH4, protandim)

↓Fibrosis

Only experimental

51

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

↓Apoptosis

evidence in animal

↓ROS-induced damage

models

Exercise

Page 52 of 55

One clinical Trial: Improved exercise capacity and increase six-minute walk distance

52

Copyright © 2014 by the American Thoracic Society

Page 53 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

Legends to the Figures

Figure 1. A) Inhibition of fatty acid oxidation with etomoxir treatment had no impact in mean pulmonary artery pressure (MPAP) in comparison to vehicle-treated rats. B) Paired analysis demonstrated that two-weeks treatment with etomoxir was insufficient to prevent deterioration in RV function, as assessed by tricuspid annulus plane systolic excursion (TAPSE). C-E) Echocardiographic analysis demonstrates no difference in RV diastolic area between controls, vehicle and etomoxir treated rats.

Figure 2. Treatment goals for prevention of a deterioration of RV function. After the initial hemodynamic changes in the lung circulation (reduced blood flow, increased resistance and increased pulmonary artery pressure) the RV is capable of adaptation as long as there is sufficient hypertrophy, adequate capillary density, adequate substrate utilization and a controlled amount of reactive oxygen species. RV function decompensation may eventually occur leading to severe RV dysfunction and failure. Treatment goals should be oriented towards maintenance of contractility with reduced energy consumption, prevention of metabolic remodeling, prevention of fibrosis, increased capillarisation (induction of angiogenesis), control of reactive oxygen species, adequate cell growth and inhibition of cardiomyocyte apoptosis.

53

Copyright © 2014 by the American Thoracic Society

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

190x254mm (96 x 96 DPI)

Copyright © 2014 by the American Thoracic Society

Page 54 of 55

Page 55 of 55

ANNALSATS Articles in Press. Published on 31-July-2014 as 10.1513/AnnalsATS.201312-425FR

190x254mm (96 x 96 DPI)

Copyright © 2014 by the American Thoracic Society

Treatment for pulmonary arterial hypertension-associated right ventricular dysfunction.

Pulmonary arterial hypertension (PAH) includes a heterogeneous group of diseases characterized by pulmonary vasoconstriction and remodeling of the lun...
774KB Sizes 0 Downloads 12 Views