Pulmonary Hypertension in Heart Failure

Journal of Cardiac Failure Vol. 16 No. 6 2010 Review Article Pulmonary Hypertension in Heart Failure MAYA GUGLIN, MD, PhD,1 AND HAMMAD KHAN, MD1 Tam...
0 downloads 1 Views 408KB Size
Journal of Cardiac Failure Vol. 16 No. 6 2010

Review Article

Pulmonary Hypertension in Heart Failure MAYA GUGLIN, MD, PhD,1 AND HAMMAD KHAN, MD1 Tampa, Florida

ABSTRACT Background: Pulmonary hypertension occurs in 60% to 80% of patients with heart failure and is associated with high morbidity and mortality. Methods and Results: Pulmonary artery pressure correlates with increased left ventricular end-diastolic pressure. Therefore, pulmonary hypertension is a common feature of heart failure with preserved as well as reduced systolic function. Pulmonary hypertension is partially reversible with normalization of cardiac filling pressures. Pulmonary vasculature remodeling and vasoconstriction create a second component, which does not reverse immediately, but has been shown to improve with vasoactive drugs and especially with left ventricular assist devices. Conclusion: Many drugs used for idiopathic pulmonary arterial hypertension are being considered as treatment options for heart failureerelated pulmonary hypertension. This is of particular significance in the heart transplant population. Randomized clinical trials with interventions targeting heart failure patients with elevated pulmonary artery pressure would be justified. (J Cardiac Fail 2010;16:461e474) Key Words: Heart failure, pulmonary hypertension, pulmonary reversibility testing, pulmonary vascular resistance, left ventricular assist device, heart transplant.

Heart failure (HF) is the most common cause of pulmonary hypertension (PH). Elevated left heart filling pressures, whether from systolic dysfunction, diastolic dysfunction, or valvular heart disease, can result in elevated pulmonary artery pressures.1 The proportion of HF patients who also have PH varies depending on the subset of patients, but remains invariably high. In 1992, Abramson et al2 reported that 26% of their cardiomyopathy cohort had a systolic pulmonary artery (PA) pressure of 40 mm Hg or higher. In 2001, Ghio et al3 found PH in more than 60% of their patients with moderate or severe HF and left ventricular (LV) dysfunction. In 2009, Lam et al4 presented the Mayo Clinic cohort of HF patients with preserved systolic function, more than 80% of which had PH. However, although the literature is abundant on the much less common primary pulmonary arterial hypertension, it is

relatively scarce on PH in HF, or type 2 PH by the World Health Organization classification. This review will focus on PH in HF. Definitions PH is usually defined as a chronic elevation of a mean PA pressure above 25 mm Hg, measured invasively by a PA catheter.5 By echocardiography, PA systolic pressure is commonly estimated from the velocity of tricuspid regurgitation. PH is considered mild if the PA systolic pressure is 35 to 45 mm Hg, moderate if it is 46 to 60 mm Hg, and severe if it is greater than 60 mm Hg.6 The driving pressure across the pulmonary circulation, or the difference of mean PA pressure and pulmonary capillary wedge pressure, is referred to as the transpulmonary gradient (TPG). Pulmonary vascular resistance (PVR) is derived by dividing the difference of mean PA pressure and left atrial pressure (pulmonary capillary wedge pressure) by the cardiac output. PVR index is used to correct PVR for body size and to avoid underestimation of PH in obesity. The key feature differentiating PA hypertension and PH resulting from HF is elevated pulmonary capillary wedge pressure, which is present in HF and absent in pulmonary arterial hypertension. The latest diagnostic algorithm is presented by Hoeper et al (Fig. 1).7

From the 1Department of Cardiology, University of South Florida, Tampa. Manuscript received July 7, 2009; revised manuscript received January 14, 2010; revised manuscript accepted January 19, 2010. Reprint requests: Maya Guglin, MD, PhD, Director, Heart Failure, University of South Florida, 2A Columbia Drive Suite 5074, Tampa, FL 33618. Tel: 813-259-0992; E-mail: [email protected] See page 470 for disclosure information. 1071-9164/$ - see front matter Ó 2010 Elsevier Inc. All rights reserved. doi:10.1016/j.cardfail.2010.01.003

461

462 Journal of Cardiac Failure Vol. 16 No. 6 June 2010

Fig. 1. Diagnostic algorithm for PH.7 EF, ejection fraction; DHF, diastolic heart failure; PVR, pulmonary vascular resistance; PCWP, pulmonary capillary wedge pressure; PH, pulmonary hypertension; PAH, pulmonary arterial hypertension; RHC, right heart catheterization; WU, Wood units.

Pathophysiology In HF, there are 2 major components of PH: hydrostatic and vasoreactive (Fig. 2). Hydrostatic, or the passive component, reflects the backward transmission of elevated LV end-diastolic pressure. Therefore, PA systolic pressure correlates tightly with pulmonary capillary wedge pressure and

Fig. 2. Pathophysiology of pulmonary hypertension in heart failure.

is roughly twice the wedge pressure.8 The reactive component represents vasoconstriction and remodeling of pulmonary vasculature. Normally, the pulmonary vasculature is characterized by low pressure, low resistance, and high distensibility. It can accommodate a significant increase in blood flow with a minimal elevation of PA pressure. When this compensatory capacity is exceeded, PA pressure increases at first on exertion and then at rest. The right ventricle is very sensitive to changes in afterload. With chronic elevation in afterload, the right ventricle begins to fail. Excessive sympathetic adrenergic stimulation as well as activation of the endothelin and renin-angiotensin-aldosterone system contributes to the pathophysiology of right ventricular failure.9e11 Right ventricular dilatation results in a leftward shift of the septum causing a change in LV geometry, decrease in LV distensibility, and thus contributing to the low cardiac output state.12 The measured severity of PH depends, in part, on the performance of the right ventricle. Right ventricular failure as a result of cardiomyopathy, infarct, or ischemia may occur at pulmonary pressures lower than 45 to 50 mm Hg; severe PH may never develop, despite an increase in PVR. An acute increase in pulmonary vascular tone can also result in right ventricular failure as is seen in heart transplant recipients with PH and thus there is the necessity to decrease PA pressure both before and after heart transplant. Structural changes occur at the capillary level including swelling of the pulmonary capillary endothelial cells, thickening of the basal lamina, and proliferation of reticular and elastic fibrils. These structural changes contribute toward

Pulmonary Hypertension in Heart Failure

increasing PVR, decreasing permeability of the vascular bed, and ultimately lower the possibility of development of pulmonary edema13 and increase the likelihood of right ventricular failure. These changes are mostly reversible if there is a successful reduction in cardiac filling pressures, although the reversal may take several months.14

Factors Determining PH Although related to LV failure, PH in the context of HF does not seem to correlate with the severity of LV systolic dysfunction. On the contrary, it depends on the elevation of filling pressure echocardiographically determined by severity of diastolic dysfunction.15e18 The E wave deceleration rate, E/A ratio,15,19 and ratio of E/E0 have been shown to reflect end-diastolic pressure more accurately than other echocardiographic parameters.20 According to the results of various studies, the restrictive filling pattern,21 the E wave deceleration rate, and the degree of mitral regurgitation were the strongest independent predictors of PH.19 A reduction in PA systolic pressure during therapy is determined, to the great extent, by wedge pressure reduction.8 Moreover, the reversibility of PH was apparent only in those patients who exhibited improvement of diastolic dysfunction from the restrictive or pseudonormal to impaired relaxation pattern likely representing improvement in filling pressures. Among patients with normal systolic function, Neuman et al22 found a significant increase in PA pressure for each step up in diastolic dysfunction grade. The mean PA pressure was 31.1 6 6 mm Hg for normal diastolic function, 35.6 6 10.2 mm Hg for grade 1 diastolic dysfunction (impaired relaxation), 38.9 6 10.6 mm Hg for grade 2 (pseudo normal), and 55.1 6 11.4 mm Hg for grade 3 (restrictive pattern) (P ! .001). Interestingly, pulmonary venous hypertension has been shown to be highly associated with the metabolic syndrome in patients with normal LV systolic function.23 Age strongly predicts PH as well.16 PH is highly prevalent among patients with end-stage renal disease on hemodialysis. In 1 study, diastolic dysfunction was present in three fourths of patients with PH and end-stage renal disease. A higher PA pressure correlated with a higher flow through arteriovenous fistula and the brain natriuretic peptide level.24 In exercise, wedge pressure rises in parallel with PA mean pressure; both correlate well with maximal oxygen consumption, unlike resting cardiac index and resting systemic arterial mean pressure, which did not demonstrate correlation with PA pressure.25 However, according to other data, ejection fraction, or degree of systolic dysfunction, is a determinant of PH on exercise. LV ejection fraction (P 5 .03) and mitral effective regurgitant orifice (P 5 .008) were independently associated with PA systolic pressure. Furthermore, dyspnea on exertion was more pronounced in patients with greater exercise-related increase in PA systolic pressure.26



Guglin and Khan

463

Compared with patients with idiopathic pulmonary arterial hypertension, patients with pulmonary venous hypertension have been found to have a higher frequency of metabolic syndrome.23 Clinical Significance In patients with HF, PH is a predictor of poor outcome. It is associated with increased short- and long-term mortality2,27 in patients with both reduced and preserved LV ejection fraction.28 Moreover, it was recently discovered that PH is associated with poor prognosis and high mortality, not only in HF, but also in general population.29 Patients with a higher velocity of tricuspid regurgitation (greater than 2.5 m/s) had a 40% higher mortality rate and a 50% higher rate of HF admissions than those with normal pulmonary pressures.2 Shalaby et al30 demonstrated that elevated baseline systolic PA pressure in patients who undergo cardiac resynchronization therapy independently predicted all-cause mortality or transplantation and HF admission. A post-resynchronization decrease in pulmonary pressure was an independent positive prognostic marker. In another study, in which more than 1000 patients with cardiomyopathy were followed for an average of 4.4 years, mean PA pressure appeared to be 1 of the most important hemodynamic predictors of death.27 In a cohort of 400 patients with known or presumed HF and PH, a Cox proportional-hazards model apportioned a 9% increase in mortality per 5 mm Hg increase in right ventricular systolic pressure.28 PH also impacts the exercise capacity in HF patients. In treadmill exercise testing with hemodynamic monitoring, increased pulmonary vascular resistance was associated with significantly lower peak exercise oxygen consumption (VO2) and lower peak exercise cardiac output, suggesting that PH impairs exercise performance in HF.31 Role of PH in Heart Transplantation PH is a risk factor for early postoperative mortality in heart transplantation, mostly from fatal right ventricular failure. According to data from the International Society for Heart and Lung Transplantation registry, approximately 20% of early deaths after cardiac transplantation can be attributed to right ventricular failure.32 Several studies have identified increased PVR O2.5 Woods units (WU), which is present in about 30% of heart transplant candidates, as an important risk factor for early death after transplant. Other groups reported a 2.5-fold increase in 3-month mortality in patients with a PVR O2.5 and an increase in 3-, 6-, and 12-month mortality with a TPG O15 mm Hg.33e35 Retrospectively studied transplant patients had a 1-year mortality of 5.6% if their PVR and TPG were below these levels and 24.4% if they exceeded these numbers before the transplant.35 These and other observations are reflected in the listing criteria for cardiac transplantation published by the International Society for Heart and Lung Transplantation.

464 Journal of Cardiac Failure Vol. 16 No. 6 June 2010 According to them, PA hypertension and elevated PVR should be considered as a relative contraindication to cardiac transplantation when the PVR is O5 WU or the PVR index O6 or the TPG exceeds 16 to 20 mm Hg. If PA systolic pressure exceeds 60 mm Hg in conjunction with any 1 of the preceding 3 variables, the risk of right HF and early death is increased. If the PVR can be reduced to !2.5 with a vasodilator but the systolic blood pressure falls !85 mm Hg, the patient remains at high risk of right HF and mortality after cardiac transplantation.36 The determination of TPG in combination with PVR is a more reliable predictor of early posttransplant survival than PVR alone.37 Butler et al reported that PA systolic pressure and TPGdbut not PVR pretransplantdwere associated with poor posttransplant survival.38 Interestingly, some investigators did not see the increased risk when performing transplants with PH. Tenderich et al39 could not confirm increased mortality in patients with elevated PVR. In their 83 patients with preoperative PVR $5 WU and TPG O15 mm Hg, 30-day mortality and cumulative survival after 1 and 5 years was not different from patients with more favorable from the hemodynamic profile.40 Addonizio et al41 transplanted 33 patients with PVR index $6, and 28 of them survived. After heart transplantation, 80% of survivors normalize their PVR at 1 year,42e44 indicating that even ‘‘fixed’’ PH reverses eventually with adequate blood flow and normal filling pressures. Similar favorable changes occur in LV assist device (LVAD) recipients. Reversibility Tests PH is considered reversible if PVR can decrease to #2.5 WU with pharmacological testing, which is commonly done as a part of the transplant evaluation process in an attempt to predict and improve outcomes. Patients with reactive PH can be considered suitable for orthotropic cardiac transplantation. Different factors influence the reversibility of PH, which is less reversible in patients with ischemic versus nonischemic cardiomyopathy and in ex-smokers versus nonsmokers.45 Reversibility of PH translates into improved 30-day survival rate after heart transplantation, but does not entirely eliminate the risk. Pharmacological tests to predict the reversibility of PH do not always reflect the posttransplant response. Cardiopulmonary bypass and intraoperative blood products may lead to further increase in PVR, which might explain why even patients with ‘‘reversible’’ PH still have a markedly higher mortality compared with patients with normal PVR.34 According to Costard-Jackie et al,46 candidates for transplant with a PVR O2.5 WU had a 3-month posttransplant mortality rate of 17.9% versus 6.9% in patients with PVR #2.5 WU. However, if their PVR could be reduced to #2.5 WU with nitroprusside without a significant drop in systemic blood pressure, their 3-month mortality after the transplant was only 3.8%, as opposed to 40.6% in those who did not respond to nitroprusside and 27.5% in those who responded but

with a drop in systolic blood pressure to below 85 mm Hg. After these data were published in 1992, it became a common practice to conduct acute pharmacological testing in patients with HF and PH before heart transplant. Historically, sodium nitroprusside was used as a challenge in the acute setting. Intravenous nitroprusside therapy is begun with a dose of approximately 1 mcg kg min and titrated until (1) PVR decreases to below 2.0 Wood units, (2) PA systolic pressure decreases to below 50 mm Hg, or (3) mean blood pressure decreases to below 65 mm Hg.34 Currently, multiple other agents are used for PH reversibility testing. Table 1 has medications and doses used for this purpose. It is difficult to say which agents are better than others because direct comparison of all of them in a randomized manner was never done. To a great extent, the choice of an agent depends on the experience of a particular center. An intravenous bolus of 0.1 mg of nitroglycerin can be followed by increasing doses according to the criteria used for nitroprusside.47 An intravenous bolus of milrinone followed by continuous infusion can be used acutely and continued for a prolonged time.48,49 Among patients participating in the study by Pamboukian et al,49 none of those who responded to milrinone in pretransplant testing died after the transplant. Milrinone was also tested as a nebulizer.50 Inhaled nitric oxide (NO) is a potent, rapidly acting, and selective pulmonary vasodilator that causes pulmonary vasodilatation with minimal systemic effects, thereby reducing TPG and PVR.51,52 Compared with nitroprusside, NO does not drop systemic pressure, although it can sometimes cause an elevation in pulmonary capillary wedge pressure.53 Intravenous dipyridamole augments and prolongs the pulmonary vasodilator effects of inhaled NO in congestive HF patients with severe PH; when administered in combination with NO inhalation, it can identify PH reversibility in potential cardiac transplant recipients in whom no pulmonary vasodilator response to inhalation of NO alone is observed.54 Prostaglandins are also used for PH reversibility testing with good results. Prostacyclin is infused at a starting dose of 1 to 2 ng kg min and increased until a decrease in systolic arterial blood pressure, increase in heart rate, or side effects such as nausea and headache. Haraldsson and colleagues55 described a good selective dose-dependent decrease in PVR and TPG with inhaled prostacyclin with no effects on systemic circulation. It effectively improves pulmonary hemodynamics, with an almost 50% reduction in PVR (P 5 .0003) and a doubling of PA compliance (P ! .0001), reflecting improvement in pulmonary vascular tone. Furthermore, it produces a positive inotropic effect.56 Intravenous prostaglandin E1 have also safely and successfully lowered PVR and TPG in 99% of patients without any significant side effects.57 In the Muenster experience, all patients with PH experienced a successful reduction in PVR by using prostaglandin E1 or prostacyclin; their 30-day and 10-year survival rates after orthotopic cardiac transplantation were similar to patients without PH.58

Pulmonary Hypertension in Heart Failure



Guglin and Khan

465

Table 1. Pharmacologic Agents Used for Testing of Reversibility of PH Agent

Dose

Route

Author, Year

Milrinone Enoximone Dobutamine Sodium nitroprusside

50 mcg/kg 0.5e2.5 mg/kg 7.5e15 mcg/kg/min 1e1.5 mcg/kg/min to maximally tolerated

IV IV IV IV

Prostaglandin E1 Prostaglandin E1 Prostacyclin Prostacyclin Sildenafil

O50 ng/kg/min !50 ng/kg/min 5e13 ng/kg/min 50e1000 mcg 25e100 mg

IV IV IV Inhaled PO

Sildenafil Sildenafil Nitroglycerin Nitric oxide

0.43 mg/kg 0.05 mg/kg 15e25 mcg/kg/min 5e80 parts per million

IV IV IV Inhaled

Dipyridamole

0.2-mg/kg bolus, then infusion of 0.0375 mg/kg/min Bolus 2 mcg/kg then 0.01 mcg/kg/min

IV

Givertz et al 1996,48 Pamboukian et al 1999,49 Botha 200968 Murali et al 199164 Murali et al 199164 Murali et al 1991,64 Semigran et al 1994,53 Kieler-Jensen 1994,51 Pagano et al 1996,146 Weston et al 200160 von Scheidt et al 2006,57 Radovancevic 200563 Murali et al 1992,147 Murali et al 1991,64 Wasler 1993148 Kieler-Jensen 1994,51 Pagano et al 1996,146 Trautnitz 1999149 Weston et al 2001,60 Haraldsson et al 1998,62 Sablotzki et al 200261 Angel Gomez-Sanchez et al 2004,67 Alaeddini et al 2004,66 Lepore et al 2005,83 De Freitas 2009150 Botha 200968 Botha 200968 Murali et al 199164 Kieler-Jensen N 1994,51 Lepore et al 2005,54,83 Mahajan et al 2007,151 Haraldsson et al 1998,62 Pagano et al 1996,146 Sablotzki et al 2002,61 Radovancevic et al 2005, 63 Semigran et al 1994,53 Fojon 2006,152 Loh 1994153 Lepore et al 200554

IV

O’Dell et al 2005136

Nesiritide

PO, orally; IV, intravenously.

Some evidence suggests that prostaglandins may be more effective than other agents in the acute reversal of PH. Prostacyclin, compared with sodium nitroprusside, induced a greater increase in cardiac output and decrease in systemic and pulmonary vascular resistances as well as lesser decrease in cardiac filling pressures.59 Inhaled prostacyclin effectively reversed PH in patients resistant to sodium nitroprusside,60 and caused a significantly greater reduction in PA pressure and an increase in cardiac output than inhaled NO.61,62 Other data have indicated that patients unresponsive to NO may be responsive to prostaglandin E1 and vice versa.63 Murali et al64 compared nitroglycerin and nitroprusside to prostaglandin E1, dobutamine, and enoximone. All of the drugs significantly increased cardiac output and decreased PVR. In addition, all of the drugs except dobutamine significantly lowered PA pressure and wedge pressure. However, prostaglandin E1 was the only drug that significantly lowered TPG. The magnitude of decline in PVR and TPG was greatest with prostaglandin E1. In a retrospective comparison of vasopressors (dobutamine, dopamine, or combination of both) with vasodilators (nitroglycerin, sodium nitroprusside) and prostacyclin, no significant difference in the magnitude of decrease of PA pressure, TPG, or PVR, although cardiac output increased more with prostacyclin. Similar proportion of patients (45% to 50%) demonstrated reversibility of PH with each intervention.65 Recently, oral sildenafil has been used to assess for reversibility of PH, reducing PA pressure in a dose-dependent fashion without significant change in mean pulmonary capillary wedge pressure, cardiac output, systemic vascular resistances, or mean blood pressure.66,67 It provided an even stronger effect when used in combination with inhaled NO.54 In a randomized comparison of intravenous milrinone and intravenous sildenafil, milrinone produced greater decrease in mean PA pressure and wedge pressure, as well

as increase in heart rate. The reductions in systemic and pulmonary vascular resistance were similar.68 When an acute vasodilator challenge is unsuccessful, hospitalization with continuous inotropes or vasodilators and hemodynamic monitoring may be required. PVR may normalize in days or weeks; it almost always eventually normalizes with LVADs.42 Treatment PH resulting from LV failure can be improved using medications that improve LV function. LV unloading results in decreased PA pressure, which in turn leads to favorable effects on systemic circulation. b-blockers that normally do not directly change pulmonary pressure may reverse PH, which was considered fixed after post-nitroprusside, because of the improved LV function.69 Cardiac resynchronization therapy increases cardiac output and decreases pulmonary capillary wedge pressure, thereby partially reversing hemodynamic abnormalities that lead to secondary PH in many HF patientsdsometimes to the point that they become eligible for cardiac transplantation.70 Many treatment options available for pulmonary arterial hypertension have been tested in PH secondary to HF, some of them successfully. Clinical studies in HF population assessing agents used in treatment of pulmonary arterial hypertension are summarized in Table 2. Studies in which patients were exposed to a single dose of a drug are not included in Table 2. Phosphodiesterase Inhibitors (Sildenafil)

Sildenafil is a specific phosphodiesterase type 5 inhibitor that increases NO availability and NO-mediated vasodilation in HF.71 In HF, phosphodiesterase type 5 activity is

Treatment, Duration 114

Bosentan 500 mg twice daily Bosentan154 62.5 mg twice daily for 4 weeks, then 125 mg twice daily Darusentan109 varying daily doses: 10 mg, 25 mg, 50 mg, 100 mg, 300 mg or placebo Tezosentan155 50 or 100 mg/hour IV Bosentan110 8e125 mg Twice a day Epoprostenol93 4.0 ng.kg.min IV Prostaglandin E197 10 ng/kg/min IV for a total of 24 hours over 3 consecutive days every 3 months

Study Acronym and Characteristics

Patients Population, Duration

PH is an Inclusion Criterion

Outcome

REACH-1 Double-blind, randomized, placebo controlled ENABLE Placebo controlled, randomized

n 5 370 NYHA III/IV 26 weeks n 5 1600 1.5 years

No

Terminated early because of liver function test abnormalities

No

No improvement with bosentan

EARTH Placebo controlled, randomized, parallel

n 5 642 NYHA II-IV 6 months

No

No change in left ventricular end systolic volume

RITZ-5 Placebo controlled, randomized, blinded Multicenter, double-blind, randomized placebo controlled

n 5 84 Acute pulmonary edema 24 hours n 5 94 NYHA III-IV, EF !35% 20 weeks Treatment þ30 days’ Follow-up n 5 471 NYHA III-IV, EF !25% 2 years n 5 22 NYHA III-IV, EF !35%.

No

No change in oxygen saturation Outcome worse with higher dose

Yes Systolic PAP O40 mm Hg

No difference in systolic PAP change, cardiac index shift, or any of the other 22 echocardiographic measurements. More patients in the bosentan arm experienced adverse and serious adverse events

No

Increased mortality rates and no improvement in quality of life with Flolan

Yes Tricuspid regurgitant velocity O3 m/s by echo confirmed by right heart catheterization

36 months’ survival: 72.7% in the PGE1 group and 56% in the control group (NS). The mean EF increased from 25.78% to 32.1% in the PGE1 group and from 23.38% to 26.15 in the control group (P ! .001); the NYHA mean class improved from 3.18 to 2.24 in the PGE1 group and from 3.46 to 3.38 in the control group (P ! 05). The PA pressure decreased from 57.7 to 40.8 mm Hg (P ! 001) Increase in 6-minute walk distance 72 6 40 vs. -39 6 32 m, P 5 .033

The Flolan International Randomized Survival Trial (FIRST) Prospective controlled not randomized 36 months

Prostacyclin98 continuous IV infusion

Prospective controlled nonrandomized

Sildenafil77 25 mg, 3 times/ day

Not controlled, not randomized

Sildenafil85 50 mg twice per day

Prospective, randomized

Sildenafil86 25 to 75 mg orally 3 times daily

Prospective randomized placebo controlled 12 weeks

n 5 33 NYHA III-IV EF !30% 12 weeks n 5 40 HF 6 months n 5 46 !65 years old NYHA II-III EF !45% 6 months

No

n 5 34 NYHA II-IV, EF !40%

Yes Mean PA pressure O25 mm Hg by catheter

No

Improved heart rate recovery (baseline: 17.5 6 3.5 beats per minute versus post: 20.6 6 3.2 beats/min)

No

Reduction of systolic PAP (from 33.7 to 23.9 mm Hg), decrease in ventilation to CO2 production slope (from 35.5 to 29.8), and breathlessness score (from 23.6 to 17.2 Increase in peak VO2 (from 14.8 to 18.7 ml  min(-1)  kg(-1)), and ratio of VO2 to work rate changes (from 7.7 to 9.3 and 10.1). All changes were significant at P ! .01 The change in peak VO2 from baseline was greater in the sildenafil group (1.8 6 -0.7 mL  kg(-1)  min(-1)) than in the placebo group (-0.27 mL  kg(-1)  min(-1); P 5 .02). Sildenafil reduced pulmonary vascular resistance and increased cardiac output with exercise (P ! .05 vs. placebo for both). Sildenafil improved in 6minute walk distance (29 m vs. placebo; P 5 .047) and Minnesota Living With Heart Failure score (-14 versus placebo; P 5 .01). Subjects in the sildenafil group experienced fewer hospitalizations for HF

PH, pulmonary hypertension; HF, heart failure; PAP, pulmonary arterial pressure; IV, intravenously; NYHA, New York Heart Association; EF, ejection fraction.

466 Journal of Cardiac Failure Vol. 16 No. 6 June 2010

Table 2. Clinical Trials with PH Medications in HF Patients

Pulmonary Hypertension in Heart Failure

increased in the systemic and pulmonary vasculature and in the kidney. It is partially responsible for natriuretic peptide desensitization in HF, and many effects of phosphodiesterase type 5 inhibition produce effects similar to brain natriuretic peptide infusion.72 Basic studies suggest that phosphodiesterase inhibition has potentially favorable direct myocardial effects that may block adrenergic, hypertrophic, and proapoptotic signaling.73 Sildenafil protects cardiomyocytes against necrosis and apoptosis in the ischemia-reperfusion model,74 and prevents LV dysfunction in the anthracycline-induced cardiomyopathy model.75 In humans, it has been found to enhance endothelial function76 and improve arterial stiffness.77 Sildenafil increases myocardial contractility and reduces LV afterload,78 blunts adrenergic stimulation,79 and improves lung diffusion capacity as well as pulmonary hemodynamics at rest and during exertion. Measurements on right cardiac catheterization have shown that sildenafil acutely reduced resting and exercise pulmonary arterial pressure, systemic vascular resistance, and PVR while increasing the resting and exercise cardiac index (P ! .05 for all) without altering mean arterial pressure, heart rate, or pulmonary capillary wedge pressure, thereby acting as a selective pulmonary vasodilator.80 These effects are sustained if sildenafil is used regularly.81 It also reduces fluid transition to the alveolar interstitium.80,82 In addition, sildenafil augmented and prolonged the hemodynamic effects of inhaled NO in patients with HF and PH.83 In exercise, sildenafil improves exercise ventilation efficiency and aerobic performance,84 as well as heart rate recovery after exercise.77 It increases peak VO2 and decreases ventilatory response to carbon dioxide output during a cardiopulmonary stress test.80,85 Sildenafil’s ability to augment peak VO2 is directly correlated with baseline resting PVR and indirectly correlated with baseline resting right ventricular ejection fraction.86 Some early concerns about potential arterial oxygen desaturation with sildenafil have yet to be confirmed. Sildenafil is usually well tolerated without desaturation or significant changes in heart rate or blood pressure.87 The only common side effect is flushing.85 Favorable physiologic effects of sildenafil translate into better outcomes in HF patients. A randomized, doubleblind, placebo-controlled trial demonstrated that sildenafil in HF patients with severely reduced systolic function decreased PA systolic pressure at 60 minutes and at 4 weeks compared with a placebo.88 Symptomatically, stable HF patients on optimal therapy experienced less breathlessness after adding sildenafil as their systolic PA pressure decreased from 33.7 to 25.2 mm Hg and 23.9 mm Hg (P ! .01) in 3 and 6 months, respectively.85 Sildenafil treatment has also been associated with improvement in right ventricular systolic function, 6-minute walk distance, and Minnesota Living with Heart Failure score.86 Sildenafil may be effectively used not only for acute hemodynamic testing of PH reversibility, but also for



Guglin and Khan

467

treatment of secondary, irreversible PH in potential heart transplant recipients because it allows patients who may otherwise have been excluded because of PH to be transplanted. In 1 study, 6 patients with PH nonresponsive to nitroprusside were put on oral sildenafil. After 1 month, 3 patients had normalized PVR and TPG and 2 patients became responsive to nitroprusside.89 Moreover, sildenafil appears to not only make patients eligible for heart transplant, but also provide a smooth postoperative course in transplanted patients. Six patients received 50 mg of sildenafil before the transplant, followed by 50 or 25 mg 3 times per day after heart transplantation; good short-term outcomes occurred in 4 of them. Sildenafil treatment was discontinued 10 to 14 days after surgery, using a stepwise dose reduction.90 After LVAD implantation, sildenafil provides additional reduction of PA pressure and facilitates weaning from inhaled NO and inotropes, which can otherwise cause a rebound PH.91 All these small studies provide a good background for future randomized controlled clinical trials in HF population with PH. Prostaglandins

Prostaglandins are potent vasodilators that have been used in the treatment of primary (arterial) PH. They have multiple effects that seem to benefit HF patients. Available prostaglandins include prostaglandin I2 (prostacyclin, epoprostenol, Flolan) and prostaglandin E1. Although the former is a potent systemic vasodilator with consequent deleterious effects on ischemic myocardium, the latter is degraded in the pulmonary vessels and has a systemic effect only at high doses.92,93 Intravenous prostaglandin E1 reduces atrial natriuretic peptide and norepinephrine 94 while increasing renal perfusion and diuresis.95 Despite their vasoactive properties, prostaglandins did not perform well in the HF setting. The acute effects of intravenous prostacyclin in HF patients include a decrease in pulmonary capillary wedge pressure and PVR and an increase in cardiac index; however, at the same time, they can cause a drop in arterial pressure and systemic vascular resistance, with a subsequent rise in plasma epinephrine, norepinephrine, renin, and aldosterone concentrations.96 Some small nonrandomized trials provided encouraging results with improved hemodynamics and symptoms, showing a trend toward better outcomes through intermittent infusion of prostaglandin E1.97 Epoprostenol (prostacyclin), added to a maximal conventional treatmentdincluding dobutamine in 30% of patientsdresulted in an increase in 6-minute walk distance by 30 meters in 12 weeks when compared with standard treatment only. Long-term outcomes were not studied.98 One case report described a longer successful use of prostaglandins. Intravenous prostacyclin and dobutamine combined with inhaled iloprost given for 21 days stabilized a patient with PH for heart transplantation, which was uncomplicated.99 A direct prospective and randomized

468 Journal of Cardiac Failure Vol. 16 No. 6 June 2010 Table 3. Reversal of Pulmonary Hypertension with Left Ventricular Assist Devices

142

Zimpfer et al 2007 Liden et al 2009144 Etz et al 2007143 Martin et al 2004137 Salzberg et al 2005156 Gallagher et al 1991157 Nguyen et al 2001158 Smedira et al 1996159

N

Time, Days

35 11 10 6 6 16 3 63

42 239 182 130 61

Mean PA Pressure before, mm Hg

Mean PA Pressure after, mm Hg

42 64 47

24 21 29

56 41

26 30

PVR before, WU

PVR after, WU

5.1 4.3 4.8 5.7 5.0 3.8 6.3 5.0

2.0 2.0 2.2 2.0 2.1 1.5 3.6 3.7

PA, pulmonary artery; PVR, pulmonary vascular resistance; WU, Wood units.

comparison of prostaglandin E1 versus prostacyclin versus low-dose dobutamine in patients awaiting heart transplantation resulted in treatment failure in 13% of patients on prostaglandin E1, 50% on prostacyclin, and 43% on dobutamine (P ! .05).100 Intermittent infusion of prostaglandin E1 has been shown to decrease PA pressure in patients with advanced HF.97 Some data indicate that prostaglandins can benefit shortterm management of PH during the early postoperative period after cardiac transplantation. When 9 patients with PH after cardiac surgery or heart transplantation and an elevated PVR treated with inotropic support were given inhaled prostacyclin, a dose-dependent decrease occurred in PVR and TPG (which decreased by 29% and 26%, respectively), right ventricular performance improved, and no changes were evident in systemic vascular resistance.55 In patients with PH undergoing mitral valve surgery, inhaled iloprost was superior to intravenous nitroglycerin for preventing acute right ventricular failure during weaning from cardiopulmonary bypass.102 Aerosolized iloprost in a patient after heart transplant with early postoperative right ventricular failure resulted in a decrease of PVR (-23.5%), whereas cardiac index (þ24.0%) and mixed venous saturation (þ9.0%) increased, without affecting systemic vascular resistance.101 However, in clinical trials, patients with severe HF and decreased LV ejection fraction randomly assigned to epoprostenol demonstrated a strong trend toward decreased survival, despite improvements in cardiac index and pulmonary capillary wedge pressure. The Flolan International Randomized Survival Trial was prematurely stopped because of increased mortality in the epoprostenol arm.93 This increased mortality could have resulted from a positive inotropic effect.56 It is important to realize though that PH was not among inclusion criteria in the Flolan International Randomized Survival Trial. Therefore, treatment with Flolan in this study did not target subset of patients with HF and PH, where the benefit from Flolan could be expected. Endothelin-receptor Blockers

Endothelin, one of the most potent natural vasoconstrictors, plays an important role in the regulation of vascular tone. Vasodilatation is in turn mediated by the release of

NO and prostacyclin. Studies in both experimental models and patients suggest that NO-dependent pulmonary vasodilatation (which depends on endothelial functions) is impaired in HF.102,103 Production of endothelin-1 is markedly increased in HF, correlating with both prognosis and symptoms.104 Endothelin-receptor blockage provides hemodynamic benefits in experimental and clinical HF. Endothelin receptor blockage after experimental myocardial infarction also reduced PH and right ventricular hypertrophy.105 Elevated plasma endothelin levels in patients with HF correlate with PA pressures and PVR.106 Acute effects of intravenous bosentan in HF have included a reduction in mean arterial and pulmonary pressures, right atrial and wedge pressure, and PVR as well as an increased cardiac and stroke volume index without change in heart rate. Short-term oral therapy has demonstrated that oral bosentan provides similar but enhanced hemodynamic effects: PVR normalized and dyspnea improved.107 In a multicenter, double-blind, placebocontrolled trial, Givertz et al109 demonstrated similar hemodynamic responses to sitaxsentan. However, vascular resistance also significantly decreased, which can create problems in HF.108 And indeed, the outcomes of the use of endothelin antagonists in HF trials thus far have been discouraging. In the randomized, double-blind, placebo-controlled EndothelinA Receptor Antagonist Trial in Heart Failure (ie, EARTH), endothelin blockage with darusentan did not improve cardiac remodeling or clinical symptoms or outcomes in patients with chronic HF.109 In another multicenter, double-blind, randomized, and placebo-controlled trial comparing bosentan to a placebo in HF patients with PH, no difference emerged in PA pressure and cardiac indexdor any of the other 22 echocardiographic measurements obtained. The frequency of adverse events requiring drug discontinuation was higher in the bosentan group.110 In the Endothelin Antagonist Bosentan for Lowering Cardiac Events in Heart Failure (ie, ENABLE) study, treatment with bosentan increased the risk of worsening HF, necessitating hospitalization.111 In the Heart Failure ET(A) Receptor Blockade Trial (ie, HEAT), short-term administration of darusentan increased the cardiac index, but did not change pulmonary capillary wedge pressure,

Pulmonary Hypertension in Heart Failure

PA pressure, PVR, or right atrial pressure. Systemic vascular resistance decreased significantly, and a trend toward more adverse events (including death and early exacerbation of HF) was also evident.112 Tezosentan, an intravenous short-acting endothelin receptor antagonist tested in the Value of Endothelin Receptor Inhibition With Tezosentan in Acute Heart Failure Studies (ie, VERITAS), neither improved dyspnea nor reduced the incidences of death or worsening HF.113 Packer et al114 found that bosentan-treated HF patients had an increased risk of HF during the first month of treatment, but a decreased risk of HF during the fourth, fifth, and sixth months of therapy when compared with a placebo. The major noncardiac adverse effects of bosentan included an increase in hepatic transaminases (in 15.6% of patients) and a decrease in hemoglobin. There may be still a place for short-term treatment with endothelin antagonists in the setting of pretransplant or early posttransplant PH. Indeed, in 5 of 7 patients considered ineligible for heart transplantation because of high PVR (post-nitroprusside infusion), bosentan therapy significantly reduced PVR.115 However, in the only long-term randomized control trial where PH was in inclusion criterion, patients on bosentan experienced more serious adverse events than controls.110 Inhaled NO

NO is synthesized endogenously from L-arginine and oxygen through a family of 3 NO synthasesdall of which are expressed in the lungdand provides vasorelaxation and bronchodilation as well as the inhibition of mitochondrial respiration, inhibition of platelet and leukocyte activation, and modulation of vascular smooth muscle cell proliferation.116 Normal basal pulmonary resistance is maintained in part by continuous local production of NO.117 Drugs that generate NO, such as nitroglycerin and sodium nitroprusside, can dilate the pulmonary vasculature, but they also cause systemic hypotension. In 1991, Frostell et al118 described the decrease in PA pressure with inhaled NO in experimental PH and found that doses up to 80 parts per million did not alter systemic blood pressure. In 1999, it was approved by the Food and Drug Administration for the treatment of hypoxemic infants with PH.119 Since then, it has been widely used in adult cardiology as well. Inhaled NO diffuses rapidly across the alveolar-capillary membrane into the smooth muscle of pulmonary vessels and provides effects similar to endogenous NOdnamely, vasodilatory, bronchodilatory, anti-inflammatory, and antiproliferatory.120 Inhaled NO has facilitated the weaning off of right ventricular assist devices in animal experiments.121 In addition, in cardiopulmonary stress test, NO has been shown to improve oxygen consumption at the anaerobic threshold.122 Inhaled NO in concentrations from 5 to 80 parts per million has been used for the treatment of PH in HF patients,



Guglin and Khan

469

especially post-ventricular assist device placement and posttransplant. When inhaled NO was started before the termination of cardiopulmonary bypass, patients had better 30-day survival rate and decreased incidence of right ventricular dysfunction compared with the historical cohort.123 Their hemodynamics were also better,124e126 and the weaning from bypass was easier.127 After LVAD implantation, inhaled NO reduced PA pressure and increased LVAD flow.128 When several drugs (inhaled NO, intravenous prostacyclin, prostaglandin E1, and sodium nitroprusside) were compared in posttransplant PH management, cardiac output, right ventricular end-diastolic volume, and central filling pressures were highest, whereas systemic and pulmonary vascular resistance were lowest with prostacyclin. Only inhaled NO induced a selective decrease in pulmonary vascular resistance, with no change in systemic vascular resistance. Cardiac output increased with NO, whereas mean pulmonary arterial pressure, transpulmonary pressure gradient, and central venous pressure decreased, had the most pronounced effect at an inhaled concentration of 20 parts per million.92 Because of its short half-life, NO has to be administered continuously; even brief interruptions may cause a dangerous rebound of PH, leading to a decreased cardiac output and systemic hypotension.129 The risk can be minimized by gradually weaning the concentration of inhaled NO.119 The downregulation of endogenous NO synthesis or elevated endothelin-1 levels by inhaled NO may be responsible for this phenomenon.120 Inhaled NO also carries a small risk of methemoglobinemia, which is rarely an issue in adult population. Testing for methemoglobin regularly, Ardehali et al123 and Beck et al127 did not observe any abnormalities even after 4 days of continuous NO. Some authors still advocate routine methemoglobin checks especially in patients inhaling concentrations of 80 parts per million,119 although we did not find any mentioning of actual occurrence of methemoglobinemia in any reviewed papers. Another potential side effect of inhaled NO is an increase in LV filling pressure in patients with HF from an increased pulmonary venous return to a poorly compliant left ventricle, resulting in an acute pulmonary edema.53,130 This finding has not been confirmed by other authors. Thus, Sablotzki et al61 found no increase of pulmonary capillary wedge pressure during NO inhalation. To the contrary, they observed a falling trend in wedge pressure with NO inhalation. In their study, the main disadvantage of NO inhalation was an increase in PA pressure and PVR in 4 patients (28.6%). Some success has occurred with inhaled NO as a supplement to other treatments. Of particular interest is the combination of inhaled NO and sildenafil. After the Food and Drug Administration approved sildenafil for the treatment of erectile dysfunction, reports of severe and possibly fatal hypotension in patients taking sildenafil and nitrates led to changes in drug labeling that warned against the administration of nitrates in patients using sildenafil. An expert panel of the American College of Cardiology/American Heart Association concluded that concomitant use of sildenafil and nitrates should be avoided unless the benefits are

470 Journal of Cardiac Failure Vol. 16 No. 6 June 2010 determined to far outweigh the risks.131 However, Parker et al132 found that the actual risk is low. In HF patients with PH, when sildenafil did not result in the desired magnitude of pulmonary vasodilation, adding systemic nitrates (oral isosorbide dinitrate) led to a reduction in mean PA pressure of 11 mm Hg, whereas mean systemic arterial pressure decreased by only 3 mm Hg. The ratio of pulmonary vascular resistance to systemic vascular resistance was reduced by 45%, suggesting some pulmonary selectivity of the combination. Treatment with sildenafil and nitrates was continued for 2 to 8 months, with no episodes of marked systemic hypotension, syncope, or lightheadedness.133 Inotropes and Vasodilators

Milrinone, a type 3 phosphodiesterase inhibitor with vasodilating and positive inotropic properties, has been shown to lower PVR. Milrinone produces sustained inotropic effect with increase in contractility and cardiac output and venous and pulmonary vasodilation with decrease in right and left heart filling pressures and systemic and PVR without significant change in TPG.48 In right ventricular failure due to residual PH after the implantation of LVAD, milrinone caused a significant reduction in PVR and an increase in LVAD flow.134 Inhaled milrinone also can be useful for decrease of PA pressure in the postoperative setting, alone or in combination with prostacyclin.135 Nesiritide decreases pulmonary capillary wedge pressure and therefore decreases pulmonary pressure. A 31% decrease in wedge pressure was accompanied by 15.6% reduction in mean PA pressure.136 LVAD

When pharmacological interventions fail to decrease PVR sufficiently to allow heart transplantation, it can be achieved with LVADs, which reverse many pathological processes in HF, including PH.137 Unloading the left ventricle alleviates left atrial hypertension and decreases pulmonary capillary wedge pressure, thereby inducing a decline in the PA pressure. In addition, improved cardiac output reduces hypoxia at the tissue level, resulting in less pulmonary vasoconstriction. Initial anecdotal reports of normalization of ‘‘fixed’’ PH after LVAD support138e140 have been replaced by case series, as summarized in the Table 3. Even in patients with fixed PH, long-term LVAD therapy usually results in normalization of pulmonary pressures within several months, and patients become eligible for transplant, regardless of the type of LVAD used. Pulsatile and nonpulsatile flow devices appear equally effective in PH reduction.141e143 Subsequent heart transplantations result in good outcomes.137 However, according to some data, LVAD before heart transplant in the presence of PHdalthough decreasing PVR from 4.3 6 1.6 to 2.0 6 0.6WU, P ! .05ddid not provide better long-term outcomes. The 30-day survival rate in patients who received LVADs pretransplant was 82% versus 91% in

patients with similar degree of PH who did not receive LVADs; the 4-year survival was 64% and 82%, respectively.144 Sometimes heterotopic heart transplant is used with the donor heart functioning as an LVAD. This also leads to normalization of PA pressure.145 Conclusions PH is highly prevalent in HF. It is associated with high morbidity and mortality. PA pressure correlates with increased LV end-diastolic pressure and diastolic dysfunction, but not with LV ejection fraction. Many drugs currently used for pulmonary arterial hypertension are being tested as treatment options for PH in HF. Although the short-term benefit of such therapies is well established in the peritransplant setting, the effects on longterm outcomes are unclear. Randomized clinical trials with endothelin antagonists and prostaglandins did not target HF patients with PH and are therefore inconclusive. Sildenafil appears to be a promising agent based on small studies with surrogate outcomes. Randomized clinical trials designed to elucidate its potential role would be justified. Pulmonary hypertension in HF may be a potentially important treatment target. Advances in its management could potentially improve the clinical outcomes in heart failure population. Disclosures None. References 1. McLaughlin VV, Archer SL, Badesch DB, et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association developed in collaboration with the American College of Chest Physicians; American Thoracic Society, Inc.; and the Pulmonary Hypertension Association. J Am Coll Cardiol 2009;53:1573e619. 2. Abramson SV, Burke JF, Kelly JJ, Jr, et al. Pulmonary hypertension predicts mortality and morbidity in patients with dilated cardiomyopathy. Ann Intern Med 1992;116:888e95. 3. Ghio S, Gavazzi A, Campana C, et al. Independent and additive prognostic value of right ventricular systolic function and pulmonary artery pressure in patients with chronic heart failure. J Am Coll Cardiol 2001;37:183e8. 4. Lam CS, Roger VL, Rodeheffer RJ, Borlaug BA, Enders FT, Redfield MM. Pulmonary hypertension in heart failure with preserved ejection fraction: a community-based study. J Am Coll Cardiol 2009;53:1119e26. 5. Barst RJ, McGoon M, Torbicki A, et al. Diagnosis and differential assessment of pulmonary arterial hypertension. J Am Coll Cardiol 2004;43:40Se7S. 6. Roldan C. The ultimate echo guide. Philadelphia, PA: Lippincott Williams and Wilkins; 2005. 7. Hoeper MM, Barbera JA, Channick RN, et al. Diagnosis, assessment, and treatment of non-pulmonary arterial hypertension pulmonary hypertension. J Am Coll Cardiol 2009;54:S85e96. 8. Drazner MH, Hamilton MA, Fonarow G, Creaser J, Flavell C, Stevenson LW. Relationship between right and left-sided filling

Pulmonary Hypertension in Heart Failure

9.

10.

11.

12.

13.

14. 15.

16.

17.

18.

19.

20.

21.

22.

23.

24. 25.

26.

27.

28.

pressures in 1000 patients with advanced heart failure. J Heart Lung Transplant 1999;18:1126e32. Bolger AP, Sharma R, Li W, et al. Neurohormonal activation and the chronic heart failure syndrome in adults with congenital heart disease. Circulation 2002;106:92e9. Fan TH, Liang CS, Kawashima S, Banerjee SP. Alterations in cardiac beta-adrenoceptor responsiveness and adenylate cyclase system by congestive heart failure in dogs. Eur J Pharmacol 1987;140:123e32. Kiely DG, Cargill RI, Lipworth BJ. Angiotensin II receptor blockade and effects on pulmonary hemodynamics and hypoxic pulmonary vasoconstriction in humans. Chest 1996;110:698e703. Haddad F, Doyle R, Murphy DJ, Hunt SA. Right ventricular function in cardiovascular disease, part II: pathophysiology, clinical importance, and management of right ventricular failure. Circulation 2008;117:1717e31. Huang W, Kingsbury MP, Turner MA, Donnelly JL, Flores NA, Sheridan DJ. Capillary filtration is reduced in lungs adapted to chronic heart failure: morphological and haemodynamic correlates. Cardiovasc Res 2001;49:207e17. Rich S, Rabinovitch M. Diagnosis and treatment of secondary (noncategory 1) pulmonary hypertension. Circulation 2008;118:2190e9. Enriquez-Sarano M, Rossi A, Seward JB, Bailey KR, Tajik AJ. Determinants of pulmonary hypertension in left ventricular dysfunction. J Am Coll Cardiol 1997;29:153e9. Tribouilloy CM, Enriquez-Sarano M, Rossi A, Tajik AJ, Seward JB. Determinants of the pulmonary artery pressure rise in left ventricular dysfunction. Cardiologia 1997;42:1051e8. Capomolla S, Febo O, Guazzotti G, et al. Invasive and non-invasive determinants of pulmonary hypertension in patients with chronic heart failure. J Heart Lung Transplant 2000;19:426e38. Faggiano P, Antonini-Canterin F, Ribichini F, et al. Pulmonary artery hypertension in adult patients with symptomatic valvular aortic stenosis. Am J Cardiol 2000;85:204e8. Dini FL, Nuti R, Barsotti L, Baldini U, Dell’Anna R, Micheli G. Doppler-derived mitral and pulmonary venous flow variables are predictors of pulmonary hypertension in dilated cardiomyopathy. Echocardiography 2002;19:457e65. Stein JH, Neumann A, Preston LM, et al. Echocardiography for hemodynamic assessment of patients with advanced heart failure and potential heart transplant recipients. J Am Coll Cardiol 1997;30: 1765e72. Straburzynska-Migaj E, Szyszka A, Trojnarska O, Cieslinski A. Restrictive filling pattern predicts pulmonary hypertension and is associated with increased BNP levels and impaired exercise capacity in patients with heart failure. Kardiol Pol 2007;65:1049e55; discussion 1056e7. Neuman Y, Kotliroff A, Bental T, Siegel RJ, David D, Lishner M. Pulmonary artery pressure and diastolic dysfunction in normal left ventricular systolic function. Int J Cardiol 2008;127:174e8. Robbins IM, Newman JH, Johnson RF, et al. Association of the metabolic syndrome with pulmonary venous hypertension. Chest 2009; 136:3e4. Abdelwhab S, Elshinnawy S. Pulmonary hypertension in chronic renal failure patients. Am J Nephrol 2008;28:990e7. Franciosa JA, Baker BJ, Seth L. Pulmonary versus systemic hemodynamics in determining exercise capacity of patients with chronic left ventricular failure. Am Heart J 1985;110:807e13. Tumminello G, Lancellotti P, Lempereur M, D’Orio V, Pierard LA. Determinants of pulmonary artery hypertension at rest and during exercise in patients with heart failure. Eur Heart J 2007;28:569e74. Cappola TP, Felker GM, Kao WH, Hare JM, Baughman KL, Kasper EK. Pulmonary hypertension and risk of death in cardiomyopathy: patients with myocarditis are at higher risk. Circulation 2002; 105:1663e8. Kjaergaard J, Akkan D, Iversen KK, et al. Prognostic importance of pulmonary hypertension in patients with heart failure. Am J Cardiol 2007;99:1146e50.



Guglin and Khan

471

29. Lam CS, Borlaug BA, Kane GC, Enders FT, Rodeheffer RJ, Redfield MM. Age-associated increases in pulmonary artery systolic pressure in the general population. Circulation 2009;119:2663e70. 30. Shalaby A, Voigt A, El-Saed A, Saba S. Usefulness of pulmonary artery pressure by echocardiography to predict outcome in patients receiving cardiac resynchronization therapy heart failure. Am J Cardiol 2008;101:238e41. 31. Butler J, Chomsky DB, Wilson JR. Pulmonary hypertension and exercise intolerance in patients with heart failure. J Am Coll Cardiol 1999;34:1802e6. 32. Stobierska-Dzierzek B, Awad H, Michler RE. The evolving management of acute right-sided heart failure in cardiac transplant recipients. J Am Coll Cardiol 2001;38:923e31. 33. Erickson KW, Costanzo-Nordin MR, O’Sullivan EJ, et al. Influence of preoperative transpulmonary gradient on late mortality after orthotopic heart transplantation. J Heart Transplant 1990;9:526e37. 34. Chen JM, Levin HR, Michler RE, Prusmack CJ, Rose EA, Aaronson KD. Reevaluating the significance of pulmonary hypertension before cardiac transplantation: determination of optimal thresholds and quantification of the effect of reversibility on perioperative mortality. J Thorac Cardiovasc Surg 1997;114:627e34. 35. Delgado JF, Gomez-Sanchez MA, Saenz de la Calzada C, et al. Impact of mild pulmonary hypertension on mortality and pulmonary artery pressure profile after heart transplantation. J Heart Lung Transplant 2001;20:942e8. 36. Mehra MR, Kobashigawa J, Starling R, et al. Listing criteria for heart transplantation: International Society for Heart and Lung Transplantation guidelines for the care of cardiac transplant candidatesd2006. J Heart Lung Transplant 2006;25:1024e42. 37. Gorlitzer M, Ankersmit J, Fiegl N, et al. Is the transpulmonary pressure gradient a predictor for mortality after orthotopic cardiac transplantation? Transpl Int 2005;18:390e5. 38. Butler J, Stankewicz MA, Wu J, et al. Pre-transplant reversible pulmonary hypertension predicts higher risk for mortality after cardiac transplantation. J Heart Lung Transplant 2005;24:170e7. 39. Tenderich G, Koerner MM, Stuettgen B, et al. Does preexisting elevated pulmonary vascular resistance (transpulmonary gradient O 15 mm Hg or O 5 wood) predict early and long-term results after orthotopic heart transplantation? Transplant Proc 1998;30:1130e1. 40. Tenderich G, Koerner MM, Stuettgen B, et al. Pre-existing elevated pulmonary vascular resistance: long-term hemodynamic follow-up and outcome of recipients after orthotopic heart transplantation. J Cardiovasc Surg (Torino) 2000;41:215e9. 41. Addonizio LJ, Gersony WM, Robbins RC, et al. Elevated pulmonary vascular resistance and cardiac transplantation. Circulation 1987;76: V52e5. 42. Klotz S, Deng MC, Hanafy D, et al. Reversible pulmonary hypertension in heart transplant candidatesdpretransplant evaluation and outcome after orthotopic heart transplantation. Eur J Heart Fail 2003;5: 645e53. 43. Lindelow B, Andersson B, Waagstein F, Bergh CH. High and low pulmonary vascular resistance in heart transplant candidates. A 5year follow-up after heart transplantation shows continuous reduction in resistance and no difference in complication rate. Eur Heart J 1999;20:148e56. 44. Bhatia SJ, Kirshenbaum JM, Shemin RJ, et al. Time course of resolution of pulmonary hypertension and right ventricular remodeling after orthotopic cardiac transplantation. Circulation 1987;76: 819e26. 45. Chau EM, Bailey KR, Mahoney DW, et al. Predictors of reversibility of pulmonary hypertension in cardiac transplant recipients in the first postoperative year. Circulation 1996;94:II267e72. 46. Costard-Jackle A, Fowler MB. Influence of preoperative pulmonary artery pressure on mortality after heart transplantation: testing of potential reversibility of pulmonary hypertension with nitroprusside is useful in defining a high risk group. J Am Coll Cardiol 1992;19: 48e54.

472 Journal of Cardiac Failure Vol. 16 No. 6 June 2010 47. Gavazzi A, Ghio S, Scelsi L, et al. Response of the right ventricle to acute pulmonary vasodilation predicts the outcome in patients with advanced heart failure and pulmonary hypertension. Am Heart J 2003;145:310e6. 48. Givertz MM, Hare JM, Loh E, Gauthier DF, Colucci WS. Effect of bolus milrinone on hemodynamic variables and pulmonary vascular resistance in patients with severe left ventricular dysfunction: a rapid test for reversibility of pulmonary hypertension. J Am Coll Cardiol 1996;28:1775e80. 49. Pamboukian SV, Carere RG, Webb JG, et al. The use of milrinone in pretransplant assessment of patients with congestive heart failure and pulmonary hypertension. J Heart Lung Transplant 1999;18:367e71. 50. Sablotzki A, Starzmann W, Scheubel R, Grond S, Czeslick EG. Selective pulmonary vasodilation with inhaled aerosolized milrinone in heart transplant candidates. Can J Anaesth 2005;52:1076e82. 51. Kieler-Jensen N, Ricksten SE, Stenqvist O, et al. Inhaled nitric oxide in the evaluation of heart transplant candidates with elevated pulmonary vascular resistance. J Heart Lung Transplant 1994;13:366e75. 52. Balzer DT, Kort HW, Day RW, et al. Inhaled Nitric Oxide as a Preoperative Test (INOP Test I): the INOP Test Study Group. Circulation 2002;106:I76e81. 53. Semigran MJ, Cockrill BA, Kacmarek R, et al. Hemodynamic effects of inhaled nitric oxide in heart failure. J Am Coll Cardiol 1994;24: 982e8. 54. Lepore JJ, Dec GW, Zapol WM, Bloch KD, Semigran MJ. Combined administration of intravenous dipyridamole and inhaled nitric oxide to assess reversibility of pulmonary arterial hypertension in potential cardiac transplant recipients. J Heart Lung Transplant 2005;24: 1950e6. 55. Haraldsson A, Kieler-Jensen N, Ricksten SE. Inhaled prostacyclin for treatment of pulmonary hypertension after cardiac surgery or heart transplantation: a pharmacodynamic study. J Cardiothorac Vasc Anesth 1996;10:864e8. 56. Montalescot G, Drobinski G, Meurin P, et al. Effects of prostacyclin on the pulmonary vascular tone and cardiac contractility of patients with pulmonary hypertension secondary to end-stage heart failure. Am J Cardiol 1998;82:749e55. 57. von Scheidt W, Costard-Jaeckle A, Stempfle HU, et al. Prostaglandin E1 testing in heart failure-associated pulmonary hypertension enables transplantation: the PROPHET study. J Heart Lung Transplant 2006;25:1070e6. 58. Klotz S, Wenzelburger F, Stypmann J, et al. Reversible pulmonary hypertension in heart transplant candidates: to transplant or not to transplant. Ann Thorac Surg 2006;82:1770e3. 59. Kieler-Jensen N, Houltz E, Ricksten SE. A comparison of prostacyclin and sodium nitroprusside for the treatment of heart failure after cardiac surgery. J Cardiothorac Vasc Anesth 1995;9:641e6. 60. Weston MW, Isaac BF, Crain C. The use of inhaled prostacyclin in nitroprusside-resistant pulmonary artery hypertension. J Heart Lung Transplant 2001;20:1340e4. 61. Sablotzki A, Hentschel T, Gruenig E, et al. Hemodynamic effects of inhaled aerosolized iloprost and inhaled nitric oxide in heart transplant candidates with elevated pulmonary vascular resistance. Eur J Cardiothorac Surg 2002;22:746e52. 62. Haraldsson A, Kieler-Jensen N, Nathorst-Westfelt U, Bergh CH, Ricksten SE. Comparison of inhaled nitric oxide and inhaled aerosolized prostacyclin in the evaluation of heart transplant candidates with elevated pulmonary vascular resistance. Chest 1998;114:780e6. 63. Radovancevic B, Vrtovec B, Thomas CD, et al. Nitric oxide versus prostaglandin E1 for reduction of pulmonary hypertension in heart transplant candidates. J Heart Lung Transplant 2005;24:690e5. 64. Murali S, Uretsky BF, Reddy PS, Tokarczyk TR, Betschart AR. Reversibility of pulmonary hypertension in congestive heart failure patients evaluated for cardiac transplantation: comparative effects of various pharmacologic agents. Am Heart J 1991;122:1375e81. 65. Torres Macho J, Delgado Jimenez JF, Sanz Salvo J, et al. Effect of different pharmacologic agents to reverse severe pulmonary

66.

67.

68.

69.

70.

71.

72.

73. 74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

hypertension among end-stage heart failure patients. Transplant Proc 2009;41:2477e9. Alaeddini J, Uber PA, Park MH, Scott RL, Ventura HO, Mehra MR. Efficacy and safety of sildenafil in the evaluation of pulmonary hypertension in severe heart failure. Am J Cardiol 2004;94:1475e7. Angel Gomez-Sanchez M, Saenz De La Calzada C, Escribano Subias P, et al. Pilot assessment of the response of several pulmonary hemodynamic variables to sublingual sildenafil in candidates for heart transplantation. Eur J Heart Fail 2004;6:615e7. Botha P, Parry G, Dark JH, Macgowan GA. Acute hemodynamic effects of intravenous sildenafil citrate in congestive heart failure: comparison of phosphodiesterase type-3 and -5 inhibition. J Heart Lung Transplant 2009;28:676e82. Crilley JG, Dark JH, Hall JA. Reversal of severe pulmonary hypertension with beta blockade in a patient with end stage left ventricular failure. Heart 1998;80:620e2. Healey JS, Davies RA, Tang AS. Improvement of apparently fixed pulmonary hypertension with cardiac resynchronization therapy. J Heart Lung Transplant 2004;23:650e2. Katz SD, Balidemaj K, Homma S, Wu H, Wang J, Maybaum S. Acute type 5 phosphodiesterase inhibition with sildenafil enhances flow-mediated vasodilation in patients with chronic heart failure. J Am Coll Cardiol 2000;36:845e51. Forfia PR, Lee M, Tunin RS, Mahmud M, Champion HC, Kass DA. Acute phosphodiesterase 5 inhibition mimics hemodynamic effects of B-type natriuretic peptide and potentiates B-type natriuretic peptide effects in failing but not normal canine heart. J Am Coll Cardiol 2007;49:1079e88. Guazzi M. Sildenafil and phosphodiesterase-5 inhibitors for heart failure. Curr Heart Fail Rep 2008;5:110e4. Das A, Xi L, Kukreja RC. Phosphodiesterase-5 inhibitor sildenafil preconditions adult cardiac myocytes against necrosis and apoptosis. Essential role of nitric oxide signaling. J Biol Chem 2005;280: 12944e55. Fisher PW, Salloum F, Das A, Hyder H, Kukreja RC. Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation 2005;111:1601e10. Patel MD, Katz SD. Phosphodiesterase 5 inhibition in chronic heart failure and pulmonary hypertension. Am J Cardiol 2005;96: 47Me51M. Guazzi M, Arena R, Pinkstaff S, Guazzi MD. Six months of Sildenafil therapy improves heart rate recovery in patients with heart failure. Int J Cardiol 2008;136:141e3. Hirata K, Adji A, Vlachopoulos C, O’Rourke MF. Effect of sildenafil on cardiac performance in patients with heart failure. Am J Cardiol 2005;96:1436e40. Borlaug BA, Melenovsky V, Marhin T, Fitzgerald P, Kass DA. Sildenafil inhibits beta-adrenergic-stimulated cardiac contractility in humans. Circulation 2005;112:2642e9. Lewis GD, Lachmann J, Camuso J, et al. Sildenafil improves exercise hemodynamics and oxygen uptake in patients with systolic heart failure. Circulation 2007;115:59e66. Jabbour A, Keogh A, Hayward C, Macdonald P. Chronic sildenafil lowers transpulmonary gradient and improves cardiac output allowing successful heart transplantation. Eur J Heart Fail 2007;9:674e7. Guazzi M, Tumminello G, Di Marco F, Guazzi MD. Influences of sildenafil on lung function and hemodynamics in patients with chronic heart failure. Clin Pharmacol Ther 2004;76:371e8. Lepore JJ, Maroo A, Bigatello LM, et al. Hemodynamic effects of sildenafil in patients with congestive heart failure and pulmonary hypertension: combined administration with inhaled nitric oxide. Chest 2005;127:1647e53. Guazzi M, Casali M, Berti F, Rossoni G, Colonna VD, Guazzi MD. Endothelium-mediated modulation of ergoreflex and improvement in exercise ventilation by acute sildenafil in heart failure patients. Clin Pharmacol Ther 2008;83:336e41.

Pulmonary Hypertension in Heart Failure 85. Guazzi M, Samaja M, Arena R, Vicenzi M, Guazzi MD. Long-term use of sildenafil in the therapeutic management of heart failure. J Am Coll Cardiol 2007;50:2136e44. 86. Lewis GD, Shah R, Shahzad K, et al. Sildenafil improves exercise capacity and quality of life in patients with systolic heart failure and secondary pulmonary hypertension. Circulation 2007;116:1555e62. 87. Herrmann HC, Chang G, Klugherz BD, Mahoney PD. Hemodynamic effects of sildenafil in men with severe coronary artery disease. N Engl J Med 2000;342:1622e6. 88. Behling A, Rohde LE, Colombo FC, Goldraich LA, Stein R, Clausell N. Effects of 5’-phosphodiesterase four-week long inhibition with sildenafil in patients with chronic heart failure: a doubleblind, placebo-controlled clinical trial. J Card Fail 2008;14:189e97. 89. Zakliczynski M, Maruszewski M, Pyka L, et al. Effectiveness and safety of treatment with sildenafil for secondary pulmonary hypertension in heart transplant candidates. Transplant Proc 2007;39: 2856e8. 90. Maruszewski M, Zakliczynski M, Przybylski R, Kucewicz-Czech E, Zembala M. Use of sildenafil in heart transplant recipients with pulmonary hypertension may prevent right heart failure. Transplant Proc 2007;39:2850e2. 91. Klodell CT, Jr, Morey TE, Lobato EB, et al. Effect of sildenafil on pulmonary artery pressure, systemic pressure, and nitric oxide utilization in patients with left ventricular assist devices. Ann Thorac Surg 2007;83:68e71; discussion 71. 92. Kieler-Jensen N, Lundin S, Ricksten SE. Vasodilator therapy after heart transplantation: effects of inhaled nitric oxide and intravenous prostacyclin, prostaglandin E1, and sodium nitroprusside. J Heart Lung Transplant 1995;14:436e43. 93. Califf RM, Adams KF, McKenna WJ, et al. A randomized controlled trial of epoprostenol therapy for severe congestive heart failure: the Flolan International Randomized Survival Trial (FIRST). Am Heart J 1997;134:44e54. 94. Hulsmann M, Stanek B, Frey B, et al. Hemodynamic and neurohumoral effects of long-term prostaglandin E1 infusions in outpatients with severe congestive heart failure. J Heart Lung Transplant 1997; 16:556e62. 95. Packer M. Interaction of prostaglandins and angiotensin II in the modulation of renal function in congestive heart failure. Circulation 1988;77:I64e73. 96. Yui Y, Nakajima H, Kawai C, Murakami T. Prostacyclin therapy in patients with congestive heart failure. Am J Cardiol 1982;50:320e4. 97. Serra W, Musiari L, Ardissino D, Gherli T, Montanari A. Benefit of prostaglandin infusion in severe heart failure Preliminary clinical experience of repetitive administration. Int J Cardiol 2009. 98. Sueta CA, Gheorghiade M, Adams KF, Jr, et al. Safety and efficacy of epoprostenol in patients with severe congestive heart failure. Epoprostenol Multicenter Research Group. Am J Cardiol 1995;75: 34Ae43A. 99. Wittwer T, Pethig K, Struber M, et al. Aerosolized iloprost for severe pulmonary hypertension as a bridge to heart transplantation. Ann Thorac Surg 2001;71:1004e6. 100. Stanek B, Sturm B, Frey B, et al. Bridging to heart transplantation: prostaglandin E1 versus prostacyclin versus dobutamine. J Heart Lung Transplant 1999;18:358e66. 101. Langer F, Wendler O, Wilhelm W, Tscholl D, Schafers HJ. Treatment of a case of acute right heart failure by inhalation of iloprost, a longacting prostacyclin analogue. Eur J Anaesthesiol 2001;18:770e3. 102. Porter TR, Taylor DO, Cycan A, et al. Endothelium-dependent pulmonary artery responses in chronic heart failure: influence of pulmonary hypertension. J Am Coll Cardiol 1993;22:1418e24. 103. Cooper CJ, Jevnikar FW, Walsh T, Dickinson J, Mouhaffel A, Selwyn AP. The influence of basal nitric oxide activity on pulmonary vascular resistance in patients with congestive heart failure. Am J Cardiol 1998;82:609e14. 104. Stewart DJ, Cernacek P, Costello KB, Rouleau JL. Elevated endothelin-1 in heart failure and loss of normal response to postural change. Circulation 1992;85:510e7.



Guglin and Khan

473

105. Lucas M, Jasmin JF, Dupuis J. Effect of ET(A) receptor antagonist on pulmonary hypertension and vascular reactivity in rats with congestive heart failure. Pulm Pharmacol Ther 2001;14:307e14. 106. Cody RJ, Haas GJ, Binkley PF, Capers Q, Kelley R. Plasma endothelin correlates with the extent of pulmonary hypertension in patients with chronic congestive heart failure. Circulation 1992;85:504e9. 107. Sutsch G, Kiowski W, Yan XW, et al. Short-term oral endothelinreceptor antagonist therapy in conventionally treated patients with symptomatic severe chronic heart failure. Circulation 1998;98: 2262e8. 108. Kiowski W, Sutsch G, Oechslin E, Bertel O. Hemodynamic effects of bosentan in patients with chronic heart failure. Heart Fail Rev 2001; 6:325e34. 109. Anand I, McMurray J, Cohn JN, et al. 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 2004; 364:347e54. 110. Kaluski E, Cotter G, Leitman M, et al. Clinical and hemodynamic effects of bosentan dose optimization in symptomatic heart failure patients with severe systolic dysfunction, associated with secondary pulmonary hypertensionea multi-center randomized study. Cardiology 2008;109:273e80. 111. Kalra PR, Moon JC, Coats AJ. 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 2002;85:195e7. 112. Luscher TF, Enseleit F, Pacher R, et al. Hemodynamic and neurohumoral effects of selective endothelin A (ET(A)) receptor blockade in chronic heart failure: the Heart Failure ET(A) Receptor Blockade Trial (HEAT). Circulation 2002;106:2666e72. 113. McMurray JJ, Teerlink JR, Cotter G, et al. Effects of tezosentan on symptoms and clinical outcomes in patients with acute heart failure: the VERITAS randomized controlled trials. JAMA 2007;298: 2009e19. 114. Packer M, McMurray J, Massie BM, et al. Clinical effects of endothelin receptor antagonism with bosentan in patients with severe chronic heart failure: results of a pilot study. J Card Fail 2005;11: 12e20. 115. Perez-Villa F, Cuppoletti A, Rossel V, Vallejos I, Roig E. Initial experience with bosentan therapy in patients considered ineligible for heart transplantation because of severe pulmonary hypertension. Clin Transplant 2006;20:239e44. 116. Fagan KA, Tyler RC, Sato K, et al. Relative contributions of endothelial, inducible, and neuronal NOS to tone in the murine pulmonary circulation. Am J Physiol 1999;277:L472e8. 117. Cooper CJ, Landzberg MJ, Anderson TJ, et al. Role of nitric oxide in the local regulation of pulmonary vascular resistance in humans. Circulation 1996;93:266e71. 118. Frostell C, Fratacci MD, Wain JC, Jones R, Zapol WM. Inhaled nitric oxide. A selective pulmonary vasodilator reversing hypoxic pulmonary vasoconstriction. Circulation 1991;83:2038e47. 119. Bloch KD, Ichinose F, Roberts JD, Jr, Zapol WM. Inhaled NO as a therapeutic agent. Cardiovasc Res 2007;75:339e48. 120. Ichinose F, Roberts JD, Jr, Zapol WM. Inhaled nitric oxide: a selective pulmonary vasodilator: current uses and therapeutic potential. Circulation 2004;109:3106e11. 121. Endo GJ, Kojima K, Nakamura K, Matsuzaki Y, Onitsuka T. Nitric oxide inhalation prompts weaning from the right ventricular assist device: evaluation under continuous-flow biventricular assistance. J Thorac Cardiovasc Surg 2002;124:739e49. 122. Koelling TM, Kirmse M, Di Salvo TG, Dec GW, Zapol WM, Semigran MJ. Inhaled nitric oxide improves exercise capacity in patients with severe heart failure and right ventricular dysfunction. Am J Cardiol 1998;81:1494e7. 123. Ardehali A, Hughes K, Sadeghi A, et al. Inhaled nitric oxide for pulmonary hypertension after heart transplantation. Transplantation 2001;72:638e41.

474 Journal of Cardiac Failure Vol. 16 No. 6 June 2010 124. Girard C, Durand PG, Vedrinne C, et al. Case 4-1993. Inhaled nitric oxide for right ventricular failure after heart transplantation. J Cardiothorac Vasc Anesth 1993;7:481e5. 125. Rich GF, Murphy GD, Jr, Roos CM, Johns RA. Inhaled nitric oxide. Selective pulmonary vasodilation in cardiac surgical patients. Anesthesiology 1993;78:1028e35. 126. Fullerton DA, Jones SD, Jaggers J, Piedalue F, Grover FL, McIntyre RC, Jr. Effective control of pulmonary vascular resistance with inhaled nitric oxide after cardiac operation. J Thorac Cardiovasc Surg 1996;111:753e62; discussion 762e3. 127. Beck JR, Mongero LB, Kroslowitz RM, et al. Inhaled nitric oxide improves hemodynamics in patients with acute pulmonary hypertension after high-risk cardiac surgery. Perfusion 1999;14:37e42. 128. Argenziano M, Choudhri AF, Moazami N, et al. Randomized, double-blind trial of inhaled nitric oxide in LVAD recipients with pulmonary hypertension. Ann Thorac Surg 1998;65:340e5. 129. Miller OI, Tang SF, Keech A, Celermajer DS. Rebound pulmonary hypertension on withdrawal from inhaled nitric oxide. Lancet 1995;346:51e2. 130. Bocchi EA, Bacal F, Auler Junior JO, Carmone MJ, Bellotti G, Pileggi F. Inhaled nitric oxide leading to pulmonary edema in stable severe heart failure. Am J Cardiol 1994;74:70e2. 131. Cheitlin MD, Hutter AM, Jr, Brindis RG, et al. ACC/AHA expert consensus document. Use of sildenafil (Viagra) in patients with cardiovascular disease. American College of Cardiology/American Heart Association. J Am Coll Cardiol 1999;33:273e82. 132. Parker JD, Bart BA, Webb DJ, et al. Safety of intravenous nitroglycerin after administration of sildenafil citrate to men with coronary artery disease: a double-blind, placebo-controlled, randomized, crossover trial. Crit Care Med 2007;35:1863e8. 133. Stehlik J, Movsesian MA. Combined use of PDE5 inhibitors and nitrates in the treatment of pulmonary arterial hypertension in patients with heart failure. J Card Fail 2009;15:31e4. 134. Kihara S, Kawai A, Fukuda T, et al. Effects of milrinone for right ventricular failure after left ventricular assist device implantation. Heart Vessels 2002;16:69e71. 135. Haraldsson s A, Kieler-Jensen N, Ricksten SE. The additive pulmonary vasodilatory effects of inhaled prostacyclin and inhaled milrinone in postcardiac surgical patients with pulmonary hypertension. Anesth Analg 2001;93:1439e45. 136. O’Dell KM, Kalus JS, Kucukarslan S, Czerska B. Nesiritide for secondary pulmonary hypertension in patients with end-stage heart failure. Am J Health Syst Pharm 2005;62:606e9. 137. Martin J, Siegenthaler MP, Friesewinkel O, et al. Implantable left ventricular assist device for treatment of pulmonary hypertension in candidates for orthotopic heart transplantation-a preliminary study. Eur J Cardiothorac Surg 2004;25:971e7. 138. Adamson RM, Dembitsky WP, Jaski BE, et al. Left ventricular assist device support of medically unresponsive pulmonary hypertension and aortic insufficiency. ASAIO J 1997;43:365e9. 139. Petrofski JA, Hoopes CW, Bashore TM, Russell SD, Milano CA. Mechanical ventricular support lowers pulmonary vascular resistance in a patient with congenital heart disease. Ann Thorac Surg 2003;75: 1005e7. 140. Al-Khaldi A, Ergina P, DeVarennes B, Lachappelle K, Cecere R. Left ventricular unloading in a patient with end-stage cardiomyopathy and medically unresponsive pulmonary hypertension. Artif Organs 2004; 28:158e60. 141. Andrea G, Giuseppe B, Tiziano C, Maria F, Ettore V. Is fixed severe pulmonary hypertension still a contraindication to heart transplant in the modern era of mechanical circulatory support? A review. J Cardiovasc Med (Hagerstown) 2008;9:1059e62.

142. Zimpfer D, Zrunek P, Roethy W, et al. Left ventricular assist devices decrease fixed pulmonary hypertension in cardiac transplant candidates. J Thorac Cardiovasc Surg 2007;133:689e95. 143. Etz CD, Welp HA, Tjan TD, et al. Medically refractory pulmonary hypertension: treatment with nonpulsatile left ventricular assist devices. Ann Thorac Surg 2007;83:1697e705. 144. Liden H, Haraldsson A, Ricksten SE, Kjellman U, Wiklund L. Does pretransplant left ventricular assist device therapy improve results after heart transplantation in patients with elevated pulmonary vascular resistance? Eur J Cardiothorac Surg 2009. 145. Bleasdale RA, Banner NR, Anyanwu AC, Mitchell AG, Khaghani A, Yacoub MH. Determinants of outcome after heterotopic heart transplantation. J Heart Lung Transplant 2002;21:867e73. 146. Pagano D, Townend JN, Horton R, Smith C, Clutton-Brock T, Bonser RS. A comparison of inhaled nitric oxide with intravenous vasodilators in the assessment of pulmonary haemodynamics prior to cardiac transplantation. Eur J Cardiothorac Surg 1996;10:1120e6. 147. Murali S, Uretsky BF, Armitage JM, et al. Utility of prostaglandin E1 in the pretransplantation evaluation of heart failure patients with significant pulmonary hypertension. J Heart Lung Transplant 1992;11:716e23. 148. Wasler A, Iberer F, Tscheliessnigg KH, Auer T, Petutschnigg B. Prostaglandin E1 in the pretransplantation period in patients with pulmonary hypertension. J Heart Lung Transplant 1993;12:884. 149. Trautnitz M, Pehlivanli S, Behr J, et al. [Pharmacological testing of the reversibility of increased pulmonary vascular resistance before heart transplantation with prostaglandin I2 (prostacyclin)]. Z Kardiol 1999;88:133e40. 150. de Freitas AF, Jr, Bacal F, Oliveira Jde L, Jr, et al. Impact of sublingual sildenafil on pulmonary hypertension in patients with heart failure. Arq Bras Cardiol 2009;92:116e26. 151. Mahajan A, Shabanie A, Varshney SM, Marijic J, Sopher MJ. Inhaled nitric oxide in the preoperative evaluation of pulmonary hypertension in heart transplant candidates. J Cardiothorac Vasc Anesth 2007;21: 51e6. 152. Fojon S, Fernandez-Gonzalez C, Sanchez-Andrade J, et al. Inhaled nitric oxide through a noninvasive ventilation device to assess reversibility of pulmonary hypertension in selecting recipients for heart transplant. Transplant Proc 2005;37:4028e30. 153. Loh E, Stamler JS, Hare JM, Loscalzo J, Colucci WS. Cardiovascular effects of inhaled nitric oxide in patients with left ventricular dysfunction. Circulation 1994;90:2780e5. 154. ENABLE. Effects of the Endothelin Receptor Antagonist Bosentan on the Morbidity and Mortality in Patients with Chronic Heart Failure Cardiosource 2002;Clinical trials. 155. Kaluski E, Kobrin I, Zimlichman R, et al. RITZ-5: randomized intravenous TeZosentan (an endothelin-A/B antagonist) for the treatment of pulmonary edema: a prospective, multicenter, double-blind, placebo-controlled study. J Am Coll Cardiol 2003;41:204e10. 156. Salzberg SP, Lachat ML, von Harbou K, Zund G, Turina MI. Normalization of high pulmonary vascular resistance with LVAD support in heart transplantation candidates. Eur J Cardiothorac Surg 2005;27: 222e5. 157. Gallagher RC, Kormos RL, Gasior T, Murali S, Griffith BP, Hardesty RL. Univentricular support results in reduction of pulmonary resistance and improved right ventricular function. ASAIO Trans 1991;37:M287e8. 158. Nguyen DQ, Ormaza S, Miller LW, et al. Left ventricular assist device support for medically unresponsive pulmonary hypertension from left ventricular failure. J Heart Lung Transplant 2001;20:190. 159. Smedira NG, Massad MG, Navia J, et al. Pulmonary hypertension is not a risk factor for RVAD use and death after left ventricular assist system support. ASAIO J 1996;42:M733e5.