Article

Drug Interactions in Pulmonary Arterial Hypertension and Their Implications

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

Medical intervention is necessary in the treatment of pulmonary arterial hypertension (PAH) in order to prevent chronic disease progression and clinical deterioration. Recent years have seen the development of new disease-specific drugs such as endothelin receptor antagonists (ERAs). However, there remains the potential for drug–drug interactions (DDIs), which can adversely affect drug metabolism and therefore treatment efficacy, an especially significant factor to consider in light of the increasing use of combination therapy in PAH and the aging patient population, who may also suffer age-related diseases requiring concomitant drug therapy. The ERA ambrisentan has demonstrated lower liver toxicity and fewer DDIs than other ERAs. The lack of any significant drug interactions with commonly used adjunct therapies such as warfarin or sildenafil could potentially improve treatment efficacy as well as patient safety.

Disclosure:Hossein-Ardeschir Ghofrani, MD, has received educational and research grants from Bayer Schering, Pfizer, Encysive (until June 2009), and Ergonex and has provided consulting services for Bayer Schering, Pfizer, Actelion, Encysive, Nycomed, Ergonex, and GlaxoSmithKline. In addition, he has performed lectures supported by Bayer Schering, Pfizer, Actelion, and Encysive. Ralph Schermuly, PhD, has received either educational or research grants or speaker honoraria or performed consulting services for Bayer Schering, Pfizer, Actelion, Encysive, Nycomed, Ergonex, GlaxoSmithKline, Novartis, and Solvay Pharmaceuticals. Werner Seegar, MD, has received educational and research grants from Gilead Colorado, Lung Rx, Myogen Inc. Westminster, and Schering Deutschland and has provided consultancy services to Altana Pharma, Bayer Schering, Lung Rx, and Schering AG. In addition, he has received speaker honoraria from Bayer Shering, Encysive, and Lung Rx. The remaining authors have no conflicts of interest to declare.

Received:

Accepted:

Correspondence Details:Hossein-Ardeschir Ghofrani, MD, Medical Clinic II/V, University Hospital Giessen and Marburg GmbH, Klinikstrasse 36, 35385 Giessen, Germany. E: Ardeschir.Ghofrani@innere.med.uni-giessen.de

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Pulmonary arterial hypertension (PAH) has an estimated prevalence of 15–25 cases/million population.1 This chronic, progressive disease is defined by a mean pulmonary arterial pressure >25mmHg in conjunction with normal pulmonary capillary wedge pressure <15mmHg.2 The disease is characterized by increased vascular resistance of the pulmonary microvasculature, ultimately resulting in right ventricular overload, right heart failure, and death.3

Severity of PAH is graded by the New York Heart Association functional classifications (NYHA FC, see Table 1), a modification of the original classification by the World Health Organization (WHO);4 symptomatic severity has long since been recognized to be related to prognosis.5 Not only is PAH a rapidly evolving disease, but it is also asymptomatic in the early stages. As such, approximately 75% of patients present with NYHA FC III and marked functional impairment at diagnosis.1 PAH is classified as idiopathic (IPAH), familial (FPAH), or occurring in association with other conditions or risk factors.6

These subgroups share similar clinical and pathological features, but the precise etiology of PAH remains unknown. This article will consider the treatments available in PAH, the increasing use of combination therapies, and the implications of resulting drug–drug interactions (DDIs) in PAH, with a special focus on endothelin receptor antagonists (ERAs).

Current Management of Pulmonary Arterial Hypertension

Treatment of PAH can be non-specific or disease-specific (see Table 2). Anticoagulants are commonly used as conventional therapy in PAH, despite the fact that there has been no evidence from prospective, randomized, placebo-controlled studies in support of this approach. Studies that support the use of anticoagulants have largely been retrospective or non-randomized, with small study numbers of patients with IPAH.7,8 A target international normalized ratio (INR) for oral anticoagulation of between 1.5 and 2.5 is recommended.9

Diuretics and low-sodium diets are able to relieve hypervolemia and the associated symptoms, but it is not known whether this approach reduces the burden of right ventricular overload and improves prognosis. Digoxin has also been used as PAH therapy due to its ability to increase cardiac output, but doubts regarding its long-term efficacy have restricted its use to cases of PAH associated with atrial fibrillation.10 The vasodilatory activity of calcium-channel blockers can be used in a minority of PAH patients to counteract vasoconstriction, which was originally assumed to be the underlying cause of PAH. Favorable response rates to an acute vasodilator are achieved in fewer than 10% of patients, only half of whom are able to maintain long-term responses.11

Studies of the underlying molecular mechanism in PAH have allowed the development of disease-specific therapies in the three established molecular pathways of PAH pathophysiology: the prostacyclin pathway, the nitric oxide (NO) pathway, and the endothelin (ET) pathway.

Prostacyclin Pathway and Prostanoids

Prostacyclin is a metabolite of arachidonic acid produced by the vascular endothelium and acts as a potent pulmonary and systemic vasodilator through the effects of the secondary messenger cyclic adenosine monophosphate (cAMP).12,13 Prostacyclin also has antiproliferative properties and inhibitory effects on platelet aggregation. Deficiencies of prostacyclin due to reduced expression of prostacyclin synthase in PAH led to the development of prostanoid replacement therapy, of which there are three prostacyclin analogs available on the market (see Table 2). Although effective in improving exercise capacity, cardiopulmonary hemodynamics, and symptoms,14–18 the short half-lives of these drugs mean that drug delivery is via continuous intravenous (IV) or subcutaneous (SC) administration, which can cause infection from venous catheters and site pain, respectively, or frequent inhalation therapy.19

Nitric Oxide Pathway and Phosphodiesterase Inhibitors

NO has potent pulmonary vasodilatory effects and inhibits platelet activation and vascular smooth-muscle cell proliferation; vasodilation is achieved by activation of the soluble guanylate cyclase by NO and production of the second messenger cyclic guanosine monophosphate (cGMP).20 However, increased expression of phosphodiesterase-5 (PDE5) in the lungs in PAH leads to enhanced cGMP degradation. Therefore, selective inhibition of PDE5 by sildenafil blocks cGMP degradation in the pulmonary vasculature, leading to increased vasodilatory activity of endogenous NO.21 Sildenafil has also been shown to improve the six-minute walk distance (6MWD), hemodynamic variables, and NYHA FC.21

Endothelin Pathway and Endothelin Receptor Antagonists

Levels of ET-1, a potent vasoconstrictor and smooth-muscle mitogen, are elevated in the plasma and lung tissue of patients with PAH.22 The effects of ET are mediated by two ET receptor isoforms: ET type A (ETA) and ET type B (ETB). Activation of ETA receptors mediates vasoconstriction and cellular proliferation of pulmonary vascular smooth-muscle cells; in normal pulmonary vasculature, ETB receptors mediate vasodilation primarily through clearance of ET-1 circulating in the vascular beds of the lungs and kidneys, and increased production of prostacyclin and NO.23 Bosentan is a non-selective (dual) ERA; ambrisentan and sitaxentan are specific antagonists of the ETA receptor. Currently, there is no clear evidence suggesting that receptor selectivity confers any advantage in terms of the clinical efficacy of these drugs.

Large-scale clinical studies have shown that ERAs have proven efficacy in mediating vasoconstriction in PAH. In the Bosentan Randomized Trial of Endothelin Antagonised Therapy (BREATHE-1), patients receiving bosentan exhibited improvements in FC and exercise capacity as measured by 6MWD and prolonged time to clinical worsening compared with patients receiving placebo.24 The Sitaxentan to Relieve Impaired Exercise (STRIDE-1) trial showed that patients receiving sitaxentan benefited in terms of improvements of 6MWD, NYHA FC, and pulmonary hemodynamics.25 The Ambrisentan in Pulmonary Arterial Hypertension, Randomized, Double-Blind, Placebo- Controlled, Multicenter, Efficacy Study 1 and 2 (ARIES-1 and ARIES-2) found that patients who were randomized to the ambrisentan groups had significant improvements in increased 6MWD, time to clinical worsening, FC, Borg dyspnea score, and quality of life score.26 The ERAs are associated with hepatoxicity requiring monthly liver function testing; bosentan and sitaxentan have both been found to induce a dose-dependent increase in hepatic transaminase levels to more than three times the upper limit of normal (ULN) in 11 and 5% of patients, respectively.24,27

Clinical trials have found a lower incidence of acute hepatotoxicity with ambrisentan, with fewer than 3% of patients experiencing elevations in hepatic aminotransferases more than three times ULN.28–30 This lower incidence of liver function abnormalities has been attributed to the fact that, unlike the other ERAs, ambrisentan does not inhibit the bile salt export pump (BSEP) in the liver.31–33

Combination Therapy and Drug–Drug Interactions

With disease-specific therapeutic strategies targeting the multiple pathophysiological pathways and mechanisms of PAH, it is possible that the addition of one agent to another could confer additive or synergistic benefits. Patients can receive disease-specific agents alongside general background therapies, but continued disease progression may eventually lead patients to require a combination of disease-specific therapies in order to effectively manage the disease. The European Society of Cardiology (ESC) advocates such an approach for patients with advanced disease who are not responsive to or exhibit deterioration with first-line treatment.34 The Registry to EValuate Early And Long-term PAH Disease Management (REVEAL) showed that combination therapy is frequently used to treat PAH in the US, with 36% of patients undergoing dual combination and 9% receiving triple combination.35

Combination therapy can provide benefits for patients with PAH, addressing more than one of the disease mechanisms and perhaps even allowing for dose reductions and lowered risk for side effects due to enhanced efficacy.36 However, combination therapy also presents the risk for DDIs, which can compromise disease efficacy or increase the incidence or severity of adverse effects, thereby negatively affecting the health of the patient. Furthermore, the aging population means an increase in the proportion of elderly patients with PAH,1,37 many of whom will also require concomitant drug therapy for the prevention or treatment of other age-related diseases, such as diabetes and hypertension.38 PAH is also frequently associated with other comorbidities that require concomitant medical treatments, such as scleroderma, HIV, and congenital heart disease.39–41

Studies have shown favorable outcomes in patients receiving combination therapy; sildenafil plus IV epoprostenol conferred improvements in 6MWD and hemodynamic parameters over placebo, with a significant reduction in the number of patients exhibiting clinical worsening, and improved survival.42 Significant improvements were achieved in 6MWD, NYHA FC, and post-inhalation hemodynamic parameters in patients with IPAH or associated PAH with NYHA FC III who received the combination of bosentan and iloprost over those who received bosentan and placebo,43 although another study found no significant change in 6MWD upon addition of iloprost to bosentan.44

Other studies have documented encouraging results with additional benefits upon sequential addition of bosentan or sildenafil to prostanoids.45–48 The combination of the orally available bosentan and sildenafil has also provided an interesting outcome; patients with mild PAH (WHO FC II) who received this combination were shown to experience decreased pulmonary vascular resistance, but no improvements in 6MWD.49 Other studies have reported improved 6MWD and NYHA FC in patients with IPAH.50,51 However, there are pharmacological interactions affecting the metabolism and efficacy of these drugs. Currently, there are no long-term data available concerning combination therapy.

DDIs are a result of one medicine altering the pharmacokinetics or pharmacodynamics of another. The most notable DDI is that involving the cytochrome (CYP) P450 oxidases, where modulation of the various CYP isozymes of this enzyme system by one drug can invariably effect the metabolism of another. A number of P450 isozymes with important roles in drug metabolism have been identified; approximately 90% of drug metabolisms by the CYP system can be accounted for by CYP1A2, CYP2C19, CYP2C9, CYP2D6, CYP2E1, and CYP3A4.52–54 Of the highly expressed CYP3A family, CYP3A4 is the most abundant isoform expressed in the liver and gut,55 while CYP2D6 and the CYP2C family are also responsible for metabolizing a majority of other clinically relevant drugs.56 Drug pharmacokinetics can be influenced by the rate of absorption, the distribution into bodily areas, biotransformation, and clearance rates; additional variables of age, genetics, and disease mean that DDIs can present in myriad ways in clinical practice. These DDIs can be subtle and go unnoticed unless there is reason to suspect a DDI. Therapeutic ranges of drugs are generally designated such that the lower spectrum is approximately equal to the concentration at which half of the greatest possible therapeutic effect is achieved, while the higher end of the spectrum will limit the number of toxicities to 5–10% of patients.57

DDIs can then be problematic if the affected drug has a narrow therapeutic window as—in the case of warfarin58—minute changes in plasma concentration could lead to sub-therapeutic effects or toxicity. Furthermore, DDIs are associated with substantial financial effects on healthcare resources; investigations into potential DDIs can prolong the duration and rate of hospitalization, increase the need for therapeutic monitoring, and require extensive laboratory and clinical testing.59,60

Drug–Drug Interactions with Endothelin Receptor Antagonists

A number of cytochrome P450 pathways are involved in the metabolism of the ERAs and PDE-5 inhibitor sildenafil (see Table 3). However, ambrisentan, a non-sulphonamide, propanoic-acid-based ERA, is principally metabolized through hepatic glucuronidation, with a minor route through the cytochrome P450 system. These characteristics confer a low risk for DDIs with ambrisentan.31 Drug-induced inhibition of cytochrome isozymes can potentially increase the plasma concentration of the drug, whereas induction would decrease the plasma concentration. The DDIs associated with ERAs are of special interest because many of these interactions are with medications that are taken alongside intercurrent illnesses or conditions (see Table 4).36 The use of some of these drugs is associated with potential DDIS: bosentan is an inducer of hepatic CYP isozymes, and sitaxentan inhibits hepatic CYP isozymes.36 Some of these potential clinically significant DDIs are identified in Table 5.

Bosentan and sitaxentan are known to compromise the level and therefore the efficacy of CYP3A4 substrates, including cyclosporin, oral estrogens, and simvastatin. Strong CYP3A4 inhibitors such as ketoconazole and cyclosporine increase plasma levels of bosentan; the combination of cyclosporine with bosentan or sitaxentan is contraindicated and cautioned with ambrisentan.31,61,62 Although metabolized by CYP3A4, studies suggest a lack of inductive effect for ambrisentan on the CYP3A4 isozyme.31 Furthermore, ambrisentan does not appear to interact with the commonly used PAH therapies of sildenafil, a CYP3A4 substrate, and warfarin, a CYP2C9 substrate.28,63,64 Co-administration of bosentan and sildenafil induces a two-fold increase in sildenafil clearance,65 while increasing bosentan levels by about 50%.36,65,66

The resultant decreased dose of sildenafil could lead to a lack of effect (especially in the indicated dose of 20mg twice a day), while increased bosentan plasma concentration can cause hepatic toxicity. Small but not clinically significant elevations of sildenafil have been observed when co-administered with sitaxentan and ambrisentan separately.67–69 Induction of CYP2C9 by bosentan or inhibition by sitaxentan can modify warfarin metabolism and alter the bioavailability of the drug,25,27,36,70,71 which has considerable implications for patients with PAH who use warfarin as background therapy. Indeed, an up to 80% reduction of warfarin was necessary in clinical trials with sitaxentan to prevent over-anticoagulation and account for potential bleeding.27 Close INR monitoring is recommended when introducing these ERAs. The combination of ambrisentan and warfarin in healthy volunteers and PAH patients did not cause any clinically relevant changes in coagulation, INR, or pharmacokinetics of either drug.64

Pregnancy in women with PAH is associated with an increased risk for maternal mortality of 30–50%.72 Therefore, women of child-bearing age are encouraged to use both hormonal and mechanical contraception.34 However, co-administration of PAH drugs can compromise the reliability of hormonal contraceptives. Failure of contraception is possible with bosentan co-administration owing to partial metabolism of estrogens and progestogens by CYP3A4 and CYP2C9. Conversely, sitaxentan increases estrogen levels and can increase contraceptive-associated side effects, such as the risk for thromboembolism, particularly in women who smoke. The combination of ambrisentan and the combined oral contraceptive pill (COCP) did not cause any clinically relevant changes in either component of the COCP.31 All three ERAs are contraindicated in pregnancy owing to their teratogenic profiles.31,61,62 To control hypercholesterolemia and prevent cardiovascular disease, simvastatin is commonly prescribed. Comorbidities present in the aging PAH patient population mean that many of these patients may also require simvastatin therapy. However, co-administration of bosentan and simvastatin significantly reduces the plasma concentration of simvastatin; there was no effect on the plasma concentration of bosentan.71 Patients receiving bosentan in addition to simvastatin therefore need careful monitoring of cholesterol levels and subsequent dose adjustments.

Antiviral drugs used in the treatment of HIV also present potential problems if co-administered with PAH therapies. The HIV protease inhibitors ritonavir and saquinovir are known inhibitors of CYP3A4, which could interfere with the metabolism of CYP450 isozymes and, thus, bosentan, sitaxentan, and ambrisentan. However, DDI studies with antiviral drugs have not been performed.73

The Outlook for Pulmonary Arterial Hypertension Management

The aging PAH patient population presents a number of complications. Not only are patients older at diagnosis, but they are also more likely to suffer from both disease- and age-related comorbidities, necessitating the use of safe and effective PAH therapies in addition to medications that are required for intercurrent illnesses. Indeed, the use of combination therapy is on the rise, and there is a need for PAH therapies that produce minimal DDIs.

It is important to address the matter of DDIs, and their potential to perturb the achievement of therapeutic goals. There are various benefits of reducing DDIs: increased treatment efficacy, enhanced ability to implement combination therapies, and reductions in complications and investigations to allow healthcare resources to be better spent. Although monotherapy in PAH has undergone great medical advances in recent years, patients with PAH still experience poor quality of life and survival, prompting physicians to attempt combination therapies. The limited data available concerning combinations of bosentan or sildenafil plus prostanoids are encouraging, but there is clearly a need for further studies in order to evaluate the role of other combinations of classes of therapies targeting different pathways of PAH: it is necessary to conduct such studies to ascertain which combinations are beneficial and which have the potential for adverse DDIs.

Current data have shown that each of the ERAs has different safety profiles and DDIs. Although each of the ERAs requires monthly liver test monitoring, ambrisentan has demonstrated the least liver toxicity. Head-to-head studies comparing ambrisentan directly with other standard therapies would help to define the role of this drug in treating PAH. As yet, there are no such studies that directly compare the efficacy and side-effect profile and incidence between ERAs; any conferred advantage of one agent over the other in PAH is only speculative.

However, ambrisentan offers a clinically important advantage compared with other ERAs in that there is a lack of any significant drug interactions with warfarin and sildenafil, thus potentially improving patient safety for the population of PAH patients requiring multiple therapies.

References

  1. Humbert M, Sitbon O, Chaouat A, et al., Pulmonary arterial hypertension in France: results from a national registry, Am J Respir Crit Care Med, 2006;173:1023–30.
    Crossref | PubMed
  2. Gaine SP, Rubin LJ, Primary pulmonary hypertension, Lancet, 1998;352:719–25.
    Crossref | PubMed
  3. Levine DJ, Diagnosis and management of pulmonary arterial hypertension: Implications for respiratory care, Respir Care, 2006;51:368–81.
    PubMed
  4. Executive Summary from the World Symposium on Primary Pulmonary Hypertension, 1998.
  5. D’Alonzo GE, Barst RJ, Ayres SM, et al., Survival in patients with primary pulmonary hypertension. Results from a national prospective registry, Ann Intern Med, 1991;115:343–9.
    Crossref | PubMed
  6. Simonneau G, Galie N, Rubin LJ, et al., Clinical classification of pulmonary hypertension, J Am Coll Cardiol, 2004;43:5S–12S.
    Crossref | PubMed
  7. Fuster V, Steele PM, Edwards WD, et al., Primary pulmonary hypertension: natural history and the importance of thrombosis, Circulation, 1984;70:580–87.
    Crossref | PubMed
  8. Rich S, Kaufmann E, Levy PS, The effect of high doses of calcium-channel blockers on survival in primary pulmonary hypertension, N Engl J Med, 1992;327:76–81.
    Crossref | PubMed
  9. Badesch DB, Abman SH, Ahearn GS, et al., Medical therapy for pulmonary arterial hypertension: ACCP evidence-based clinical practice guidelines, Chest, 2004;126: 35S–62S.
    Crossref | PubMed
  10. Rich S, Seidlitz M, Dodin E, et al., The short-term effects of digoxin in patients with right ventricular dysfunction from pulmonary hypertension, Chest, 1998;114:787–92.
    Crossref | PubMed
  11. Sitbon O, Humbert M, Jais X, et al., Long-term response to calcium channel blockers in idiopathic pulmonary arterial hypertension, Circulation, 2005;111:3105–11.
    Crossref | PubMed
  12. Humbert M, Morrell NW, Archer SL, et al., Cellular and molecular pathobiology of pulmonary arterial hypertension, J Am Coll Cardiol, 2004;43:13S–24S.
    Crossref | PubMed
  13. Badesch DB, McLaughlin VV, Delcroix M, et al., Prostanoid therapy for pulmonary arterial hypertension, J Am Coll Cardiol, 2004;43:56S–61S.
    Crossref | PubMed
  14. Barst RJ, Rubin LJ, McGoon MD, et al., Survival in primary pulmonary hypertension with long-term continuous intravenous prostacyclin, Ann Intern Med, 1994;121:409–15.
    Crossref | PubMed
  15. Barst RJ, Rubin LJ, Long WA, et al., A comparison of continuous intravenous epoprostenol (prostacyclin) with conventional therapy for primary pulmonary hypertension. The Primary Pulmonary Hypertension Study Group, N Engl J Med, 1996;334:296–302.
    Crossref | PubMed
  16. Simonneau G, Barst RJ, Galie N, et al., Continuous subcutaneous infusion of treprostinil, a prostacyclin analogue, in patients with pulmonary arterial hypertension: a double-blind, randomized, placebo-controlled trial, Am J Respir Crit Care Med, 2002;165:800–804.
    Crossref | PubMed
  17. Tapson VF, Gomberg-Maitland M, McLaughlin VV, et al., Safety and efficacy of IV treprostinil for pulmonary arterial hypertension: a prospective, multicenter, open-label, 12-week trial, Chest, 2006;129:683–8.
    Crossref | PubMed
  18. Olschewski H, Simonneau G, Galie N, et al., Inhaled iloprost for severe pulmonary hypertension, N Engl J Med, 2002;347: 322–9.
    Crossref | PubMed
  19. Rubin LJ, Badesch DB, Evaluation and management of the patient with pulmonary arterial hypertension, Ann Intern Med, 2005;143:282–92.
    Crossref | PubMed
  20. Ghofrani HA, Pepke-Zaba J, Barbera JA, et al., Nitric oxide pathway and phosphodiesterase inhibitors in pulmonary arterial hypertension, J Am Coll Cardiol, 2004;43:68S–72S.
    Crossref | PubMed
  21. Galie N, Ghofrani HA, Torbicki A, et al., Sildenafil citrate therapy for pulmonary arterial hypertension, N Engl J Med, 2005;353:2148–57.
    Crossref | PubMed
  22. Rubens C, Ewert R, Halank M, et al., Big endothelin-1 and endothelin-1 plasma levels are correlated with the severity of primary pulmonary hypertension, Chest, 2001;120:1562–9.
    Crossref | PubMed
  23. Channick RN, Sitbon O, Barst RJ, et al., Endothelin receptor antagonists in pulmonary arterial hypertension, J Am Coll Cardiol, 2004;43:62S–7S.
    Crossref | PubMed
  24. Rubin LJ, Badesch DB, Barst RJ, et al., Bosentan therapy for pulmonary arterial hypertension, N Engl J Med, 2002;346: 896–903.
    Crossref | PubMed
  25. Barst RJ, Langleben D, Frost A, et al., Sitaxentan therapy for pulmonary arterial hypertension, Am J Respir Crit Care Med, 2004;169:441–7.
    Crossref | PubMed
  26. Galie N, Olschewski H, Oudiz RJ, et al., Ambrisentan for the treatment of pulmonary arterial hypertension: results of the ambrisentan in pulmonary arterial hypertension, randomized, double-blind, placebo-controlled, multicenter, efficacy (ARIES) study 1 and 2, Circulation, 2008;117:3010–19.
    Crossref | PubMed
  27. Barst RJ, Langleben D, Badesch D, et al., Treatment of pulmonary arterial hypertension with the selective endothelin-A receptor antagonist sitaxentan, J Am Coll Cardiol, 2006;47:2049–56.
    Crossref | PubMed
  28. Galie N, Badesch D, Oudiz R, et al., Ambrisentan therapy for pulmonary arterial hypertension, J Am Coll Cardiol, 2005; 46:529–35.
    Crossref | PubMed
  29. Olschewski H, Galie N, Ghofrani HA, et al., Ambrisentan improves exercise capacity and time to clinical worsening in patients with pulmonary arterial hypertension: Results of the ARIES-2 study, Proc Am Thorac Soc, 2006;3:A728.
  30. Oudiz R, Torres F, Frost A, et al., ARIES-1: A placebo-controlled efficacy and safety study of ambrisentan in patients with pulmonary arterial hypertension, Chest, 2006;130:121S.
    Crossref
  31. Volibris, Summary of Product Characteristics, GlaxoSmithKline, 2009.
  32. Fattinger K, Funk C, Pantze M, et al., The endothelin antagonist bosentan inhibits the canalicular bile salt export pump: a potential mechanism for hepatic adverse reactions, Clin Pharmacol Ther, 2001;69:223–31.
    Crossref | PubMed
  33. Mano Y, Usui T, Kamimura H, Effects of bosentan, an endothelin receptor antagonist, on bile salt export pump and multidrug resistance-associated protein 2, Biopharm Drug Dispos, 2007;28:13–18.
    Crossref | PubMed
  34. Galie N, Torbicki A, Barst R, et al., Guidelines on diagnosis and treatment of pulmonary arterial hypertension. The Task Force on Diagnosis and Treatment of Pulmonary Arterial Hypertension of the European Society of Cardiology, Eur Heart J, 2004;25:2243–78.
    Crossref | PubMed
  35. McGoon MD, Barst RJ, Doyle RL, et al., REVEAL Registry: Treatment History and Treatment at Baseline, Chest, 2007;2007:631S.
  36. Consensus statement on the management of pulmonary hypertension in clinical practice in the UK and Ireland, Heart, 2008;94(Suppl. 1):i1–41.
    Crossref | PubMed
  37. Thenappan T, Shah SJ, Rich S, et al., A USA-based registry for pulmonary arterial hypertension: 1982–2006, Eur Respir J, 2007;30:1103–10.
    Crossref | PubMed
  38. Elliot GC, Farber H, Frost A, et al., REVEAL Registry: Medical History and Time to Diagnosis of Enrolled Patients, Chest, 2007;2007:631S.
  39. Hoeper MM, Pulmonary hypertension in collagen vascular disease, Eur Respir J, 2002;19:571–6.
    Crossref | PubMed
  40. McLaughlin VV, McGoon MD, Pulmonary arterial hypertension, Circulation, 2006;114:1417–31.
    Crossref | PubMed
  41. Speich R, Jenni R, Opravil M, et al., Primary pulmonary hypertension in HIV infection, Chest, 1991;100:1268–71.
    Crossref | PubMed
  42. Simonneau G, Rubin LJ, Galie N, et al., Addition of sildenafil to long-term intravenous epoprostenol therapy in patients with pulmonary arterial hypertension: a randomized trial, Ann Intern Med, 2008;149:521–30.
    Crossref | PubMed
  43. McLaughlin VV, Oudiz RJ, Frost A, et al., Randomized study of adding inhaled iloprost to existing bosentan in pulmonary arterial hypertension, Am J Respir Crit Care Med, 2006;174: 1257–63.
    Crossref | PubMed
  44. Hoeper MM, Leuchte H, Halank M, et al., Combining inhaled iloprost with bosentan in patients with idiopathic pulmonary arterial hypertension, Eur Respir J, 2006;28: 691–4.
    Crossref | PubMed
  45. Ghofrani HA, Rose F, Schermuly RT, et al., Oral sildenafil as long-term adjunct therapy to inhaled iloprost in severe pulmonary arterial hypertension, J Am Coll Cardiol, 2003; 42:158–64.
    Crossref | PubMed
  46. Gomberg-Maitland M, McLaughlin V, Gulati M, et al., Efficacy and safety of sildenafil added to treprostinil in pulmonary hypertension, Am J Cardiol, 2005;96:1334–6.
    Crossref | PubMed
  47. Kataoka M, Satoh T, Manabe T, et al., Oral sildenafil improves primary pulmonary hypertension refractory to epoprostenol, Circ J, 2005;69:461–5.
    Crossref | PubMed
  48. Hoeper MM, Taha N, Bekjarova A, et al., Bosentan treatment in patients with primary pulmonary hypertension receiving nonparenteral prostanoids, Am J Cardiol, 2003;22:330–34.
    Crossref | PubMed
  49. Galie N, Rubin L, Hoeper M, et al., Treatment of patients with mildly symptomatic pulmonary arterial hypertension with bosentan (EARLY study): a double-blind, randomised controlled trial, Lancet, 2008;371:2093–2100.
    Crossref | PubMed
  50. Hoeper MM, Faulenbach C, Golpon H, et al., Combination therapy with bosentan and sildenafil in idiopathic pulmonary arterial hypertension, Eur Respir J, 2004;24:1007–10.
    Crossref | PubMed
  51. Mathai SC, Girgis RE, Fisher MR, et al., Addition of sildenafil to bosentan monotherapy in pulmonary arterial hypertension, Eur Respir J, 2007;29:469–75.
    Crossref | PubMed
  52. Pharmacotherapy: A Pathophysiologic Approach, 4th Ed., 1999.
  53. Cupp MJ, Tracy TS, Cytochrome P450: new nomenclature and clinical implications, Am Fam Physician, 1998;57:107–16.
    PubMed
  54. Guengerich FP, Cytochrome P450s and other enzymes in drug metabolism and toxicity, AAPS J, 2006;8:E101–11.
    Crossref | PubMed
  55. Eichelbaum M, Burk O, CYP3A genetics in drug metabolism, Nat Med, 2001;7:285–7.
    Crossref | PubMed
  56. Flockhart DA, Tanus-Santos JE, Implications of cytochrome P450 interactions when prescribing medication for hypertension, Arch Intern Med, 2002;162:405–12.
    Crossref | PubMed
  57. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 11th Ed., 2005.
  58. Kuruvilla M, Gurk-Turner C, A review of warfarin dosing and monitoring, Proc (Bayl Univ Med Cent), 2001;14:305–6.
    PubMed
  59. Sandson N, Economic Grand Rounds: Drug–Drug Interactions: The Silent Epidemic, Psychiatr Serv, 2005;56:22–4.
    Crossref | PubMed
  60. Shad MU, Marsh C, Preskorn SH, The economic consequences of a drug–drug interaction, J Clin Psychopharmacol, 2001;21:119–20.
    Crossref | PubMed
  61. Tracleer, Summary of Product Characteristics, Actelion Pharmaceuticals UK Ltd, 2008.
  62. Thelin, Summary of Product Characteristics, Encysive UK Ltd, 2009.
  63. Dufton C, Gerber M, Yin O, et al., No clinically relevant pharmacokinetic interaction between ambrisentan and sildenafil, Chest, 2006;(Suppl. 130):256S.
  64. Gerber M, Dufton C, Pentikis H, et al., Ambrisentan has no clinically relevant effects on the pharmacokinetics or pharmacodynamics of warfarin, Chest, 2006;(Suppl. 130): 256S.
  65. Paul GA, Gibbs JS, Boobis AR, et al., Bosentan decreases the plasma concentration of sildenafil when coprescribed in pulmonary hypertension, Br J Clin Pharmacol, 2005;60: 107–12.
    Crossref | PubMed
  66. Burgess G, Hoogkamer H, Collings L, et al., Mutual pharmacokinetic interactions between steady-state bosentan and sildenafil, Eur J Clin Pharmacol, 2008;64:43–50.
    Crossref | PubMed
  67. Benza R, Frost A, Combination of sildenafil and sitaxentan for treatment of pulmonary arterial hypertension, Chest, 2008;134:161004S.
  68. Coyne TC, Garces PC, Kramer W, No clinical interaction between sitaxentan and sildenafil, Am J Respir Crit Care Med, 2005;171:A201.
  69. Spence R, Mandagere A, Dufton C, et al., Pharmacokinetics and safety of ambrisentan in combination with sildenafil in healthy volunteers, J Clin Pharmacol, 2008;48:1451–9.
    Crossref | PubMed
  70. Barst RJ, Rich S, Widlitz A, et al., Clinical efficacy of sitaxentan, an endothelin-A receptor antagonist, in patients with pulmonary arterial hypertension: open-label pilot study, Chest, 2002;121:1860–68.
    Crossref | PubMed
  71. Dingemanse J, van Giersbergen PL, Clinical pharmacology of bosentan, a dual endothelin receptor antagonist, Clin Pharmacokinet, 2004;43:1089–1115.
    Crossref | PubMed
  72. Expert consensus document on management of cardiovascular diseases during pregnancy, Eur Heart J, 2003;24:761–81.
    Crossref | PubMed
  73. Sitbon O, Gressin V, Speich R, et al., Bosentan for the treatment of human immunodeficiency virus-associated pulmonary arterial hypertension, Am J Respir Crit Care Med, 2004;170:1212–17.
    Crossref | PubMed
  74. Flolan, Summary of Product Characteristics, GlaxoSmithKline UK, 2006.
  75. Remodulin, Summary of Product Characteristics, United Therapeutics Corp, 2008.
  76. Revatio, Summary of Product Characteristics, Pfizer Ltd, 2008.
  77. Ventavis, Summary of Product Characteristics, Bayer Schering Pharma, 2008.