Skip to main content

Main menu

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • COVID-19 submission information
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart
  • Log out

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • COVID-19 submission information
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions

Future perspectives in pulmonary arterial hypertension

Gérald Simonneau, Marius M. Hoeper, Vallerie McLaughlin, Lewis Rubin, Nazzareno Galiè
European Respiratory Review 2016 25: 381-389; DOI: 10.1183/16000617.0084-2016
Gérald Simonneau
1Université Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
2AP-HP, Service de Pneumologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
3INSERM U-999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: gerald.simonneau@bct.aphp.fr
Marius M. Hoeper
4Dept of Respiratory Medicine, Hannover Medical School and German Centre for Lung Research, Hannover, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Vallerie McLaughlin
5Dept of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lewis Rubin
6Dept of Medicine, University of California, San Diego, CA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nazzareno Galiè
7Dept of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

While there have been advances in the field of pulmonary arterial hypertension (PAH), disease management remains suboptimal for many patients. The development of novel treatments and strategies can provide opportunities to target other mechanisms that play a role in the complex pathobiology of PAH outside of the three main pathophysiological pathways. In this review, we highlight some of the potential PAH therapies or techniques that are being, or have been, investigated in phase II clinical trials. This review also discusses potential points for consideration in the development of novel therapies that target putative disease mediators or modifiers.

Abstract

Novel therapies and well-designed trials are important for improving the management of PAH patients http://ow.ly/YHPY304XdvH

Introduction

Pulmonary arterial hypertension (PAH) has evolved from a disease with limited treatment options to one where numerous therapies that target three key pathophysiological pathways in the disease (i.e. the prostacyclin, endothelin and nitric oxide pathways) are available [1]. All three pathways can now be targeted with approved drugs that significantly improve long-term outcomes [2–4]. As a result of recent developments in the field of PAH treatment, these treatment options are becoming more accessible. However, despite the advances, disease management remains unsatisfactory for many PAH patients [5] and a large number are still only treated with one PAH-specific therapy. To provide the best outcomes for patients with PAH, there is a clear need to ensure that patients are diagnosed as quickly and accurately as possible, and are optimally managed using the treatments that are currently available. One way to optimise the management of PAH patients is the use of combination therapy, which is discussed in greater detail in the article by Sitbon and Gaine [6]. Another approach to the optimisation of PAH management is the development of novel treatments and therapeutic strategies that will provide further opportunities to improve outcomes for PAH patients.

The diverse and complex mechanisms underlying the pathogenesis of PAH offer the potential for new therapies that target pathways beyond the three well-established pathways. However, only 7–8% of cardiopulmonary drugs reach the market [7], with >50% of failures occurring in early clinical development due to a lack of efficacy [8, 9]. Therefore, it is important that novel targets in PAH are examined carefully, using well-designed trials to enable the most meaningful data to be obtained. In this review, we will explore potential novel targets for the management of PAH, focusing on immune and inflammation pathways, mitochondrial dysfunction, bone morphogenetic protein receptor type 2 (BMPR2) signalling, iron-deficient states and pulmonary artery denervation (PADN). In particular, therapies that have progressed to, or are already being investigated in, phase II clinical trials will be highlighted. We will also discuss points to consider in the development of novel therapies for PAH, including lessons that can be learned from previous clinical trials.

Potential PAH therapies currently in development

Therapies targeting inflammation and immunity

A number of observations in PAH patients have implicated a dysfunctional immune/inflammatory response in the development of the disease [1]. First, perivascular inflammation has been frequently observed in patients with idiopathic PAH (IPAH) [10] and PAH associated with connective tissue disease (PAH-CTD) [11, 12]. Second, perivascular lesions are characterised by varying levels of infiltrating immune cells [13]. Third, circulating levels of cytokines and chemokines are increased in PAH patients [13, 14], which may lead to pulmonary vascular remodelling through alterations in pulmonary vascular cell proliferation, migration and differentiation [13]. Fourth, clinical studies have demonstrated that combining immunosuppressive and vasodilator therapies can improve pulmonary arterial pressure, prognosis and functional class in patients with PAH associated with some forms of CTD [15, 16]. As a result of these observations, the efficacy and safety of several immunomodulatory therapies are currently being assessed in clinical trials (summarised in table 1) [17–30].

View this table:
  • View inline
  • View popup
TABLE 1

Potential pulmonary arterial hypertension (PAH) therapies currently in development

Ubenimex is being developed as a potential treatment for PAH as it is an inhibitor of the inflammatory mediator leukotriene B4 found in the serum of some PAH patients [31]. A phase II trial and subsequent open-label extension study are currently planned to assess the efficacy and safety of ubenimex in PAH [17, 18]. Rituximab is a monoclonal antibody which can target the CD20+ B-cells found in plexiform lesions in PAH [32, 33]. Based on the results of case reports of improved outcomes in patients with advanced PAH associated with systemic sclerosis (PAH-SSc) [33], this drug is currently being investigated in an ongoing phase II trial in PAH-SSc patients [19]. Cytokines such as interleukin (IL)-6 have been linked to the pathogenesis of PAH through their ability to control cell migration, differentiation and proliferation [13]. As a result, tocilizumab (a monoclonal antibody to the IL-6 receptor) is currently being investigated in an open-label phase II trial in PAH patients [20].

Therapies targeting mitochondrial dysfunction

Impaired mitochondrial function may lead to vascular remodelling and thus contribute to the pathogenesis of PAH [34]. Therefore, drugs that may target mitochondrial dysfunction, such as bardoxolone methyl (an Nrf2 activator and inhibitor of the NF-κB pathway) and GS-4997 (an inhibitor of apoptosis signal regulating kinase 1), are being investigated as potential PAH therapies (summarised in table 1). Promising results were obtained for bardoxolone methyl in a phase II study in PAH patients on background therapy [21] and consequently a large phase III trial is now planned in a population of patients with PAH-CTD [22]. GS-4997 is also currently under investigation in an ongoing phase II trial in PAH patients [23].

Therapies targeting BMPR2 signalling

The critical role played by the BMPR2 pathway in the pathogenesis of PAH is illustrated by the fact that >80% of patients with heritable PAH and ∼20% of patients with sporadic IPAH have germline mutations resulting in loss of BMPR2 function [35, 36]. Furthermore, reduced expression of BMPR2 has also been reported in PAH patients without a mutation in this gene [37]. Thus, there is a clear rationale for activating BMPR2 signalling as a treatment option for PAH [38]. In order to identify an activator of BMPR2 signalling, a high-throughput screen of >3500 US Food and Drug Administration-approved drugs was performed [39]. Tacrolimus (a calcineurin inhibitor) was identified and subsequently demonstrated to reverse pulmonary hypertension in animal models. As a result, a phase II trial investigating the safety and efficacy of tacrolimus was initiated, but was ultimately terminated due to funding and recruitment issues (summarised in table 1) [24]. A follow-up phase IIb efficacy trial is planned [24].

Therapies targeting iron deficiency

There is evidence that iron homeostasis is important in PAH, with studies indicating that iron deficiency is common in PAH patients [40–43], and is associated with reduced exercise capacity [41, 42] and increased mortality [44]. Worsening iron deficiency has been shown to correlate with increased disease severity, as assessed by increased mean pulmonary artery pressure, reduced cardiac index and worsening functional class in IPAH patients [40]. Iron deficiency is also a finding in patients with heart failure, and likewise is associated with poor survival [45] and reduced exercise capacity [46]. This deficit in exercise capacity can be reversed by iron supplementation [46]. These findings provide a rationale for therapeutic interventions to address iron deficiency in PAH patients. To date, the clinical benefit of iron infusions in PAH patients has been demonstrated in two small exploratory studies [47, 48]. Although the results of these small studies are promising, it should be noted that low serum iron may offer a protective role in the development of pulmonary hypertension [49]. Thus, randomised controlled trials of iron supplementation in PAH patients are warranted to ensure that the impact of addressing iron deficiency in PAH can be fully elucidated.

Clinical trials investigating the efficacy of iron administration in PAH patients and healthy volunteers are summarised in table 1. A phase II trial is currently recruiting PAH patients to investigate the effect of an intravenous (i.v.) iron formulation, Ferinject (ferric carboxymaltose), on pulmonary vascular resistance (PVR) and exercise capacity [25]. Results from this trial are expected in December 2017. Ferinject was also investigated in a single-blind, interventional trial in iron-deficient or iron-replete healthy volunteers [26]. Ferinject prevented exaggerations in the hypertensive response to hypoxic exposure in iron-deficient, healthy volunteers [27]. An interventional, open-label study in IPAH patients investigated whether ferrous sulfate (oral dietary iron supplement) can improve clinical symptoms of PAH and relieve iron deficiency over 12 weeks [28]. No results have yet been reported for the latter two trials.

Pulmonary artery denervation

The neurohormonal axis is a potential therapeutic target in PAH as several studies have reported that plasma norepinephrine, muscle sympathetic nerve activity and the number of vessel sympathetic nerve endings are all elevated in IPAH patients [50]. Furthermore, it has been demonstrated in experimental models of pulmonary hypertension that disruption of sympathetic nerve fibres can improve pulmonary haemodynamics [50, 51]. The process of PADN involves ablating the nerves regulating sympathetic tone in the pulmonary artery, disrupting the nonvagal pulmonary baroreceptor reflex [50–52]. Results from the use of this technique in early clinical trials are preliminary, with a single patient case study [53] and a small proof-of-concept study demonstrating that PADN may result in improvements in 6-min walk distance (6MWD) and haemodynamic parameters in IPAH patients [52]. Although there were limitations associated with the design and analysis of the latter study [52, 54], the PADN procedure has been investigated in a phase II clinical trial, as described in table 1 [29]. However, it should be noted that this trial may be subject to similar design and analysis limitations as the earlier proof-of-concept study [55]. Another ongoing European multicentre study is investigating the efficacy of the PADN procedure in patients with PAH [30]. In all likelihood, the immediate changes in pulmonary artery pressure observed in pre-clinical and clinical studies of PADN cannot be explained by altered remodelling of the distal pulmonary arteries [50] and may result from improved pulmonary arterial compliance.

Clinical trial design in novel therapy development in PAH

Evolution of clinical trial design

Over the last two decades, the number of patients enrolled in PAH trials has risen significantly despite PAH being a rare disease. Only 81 patients were included in the pivotal randomised controlled trial in 1996 investigating epoprostenol, the first approved drug for the treatment of PAH. This trial utilised change in 6MWD from baseline to week 12 as its primary end-point [56]. Subsequent phase II/III PAH randomised controlled trials, which investigated the efficacy of a number of different PAH therapies, also used 6MWD as the primary end-point and included up to 350 patients [57–61]. Since these early trials, there has been an evolution to larger, event-driven studies [2–4, 62], with 1156 patients enrolled in the recently completed phase III GRIPHON trial [4]. From the first pivotal trial investigating epoprostenol in 1996, through to the GRIPHON trial in 2015, the evolving trial design in PAH has meant that the number of patients required for a phase III trial is now higher than ever. Therefore, important considerations must be taken into account when developing new drugs for PAH to reliably evaluate the preliminary efficacy, safety and the optimal dose range for the candidate drug prior to progressing to phase III trials.

Pre-clinical studies

The first step in drug development is the generation of pre-clinical data, which can provide a rationale for subsequent clinical development. However, a limitation of these pre-clinical studies is a possible lack of reproducibility, with one analysis suggesting that almost 80% of pre-clinical research data could not be reproduced [63]. This highlights the importance of using standardised techniques to ensure that data are as robust as possible. Animal models, while providing significant insights into the molecular and cellular pathways that underlie the development of PAH [64], are also subject to inherent limitations, including immutable species differences and the inability to completely replicate human heart failure causes and manifestations [65]. While animal models remain essential for pre-clinical drug development, the currently available acute models of pulmonary hypertension are not ideal as they do not fully represent the nature of the chronic disease in humans [64].

There is some indication that cells derived from PAH patients show increased inflammatory indices [66] and a hyperproliferative and apoptosis-resistant phenotype [67] compared with control cells. Ex vivo studies using these cells may potentially be useful in future studies as a “first-step” test of novel compounds [68]. Such ex vivo studies have revealed clinically important information regarding the manifestation of PAH in humans, which cannot be modelled in animals, including transcriptional, genetic and epigenetic pathogenic markers. It has also been suggested that drug efficacy in future pre-clinical studies should be demonstrated in blinded, randomised studies using relevant end-points such as changes in PVR and right ventricular function. These changes to pre-clinical study design would strengthen the evidence supporting the progression to clinical trials. However, even with the implementation of these changes, pre-clinical studies cannot demonstrate the whole-body effects of novel compounds in humans. In order to obtain this information, clinical trials remain essential.

Early clinical trial considerations

There is an obligation to terminate the development of ineffective drugs to reduce the number of patients exposed to them. Early-phase clinical trials are an essential bridge between pre-clinical research and large-scale human studies. Careful planning and execution of early clinical trials in PAH is important. Phase I trials are designed to assess the safety, tolerability, dose, pharmacokinetics and pharmacodynamics of the clinical candidate for administration in humans, before progressing to phase II clinical trials. Phase II or “proof-of-concept” trials, although smaller and less powerful than phase III trials, can generate critical information regarding dosing and safety in patients and can provide insight into whether a large phase III trial is likely to be successful [69, 70]. However, there are limitations associated with phase II trials in PAH, including small sample sizes, heterogeneity of study populations and the selection of appropriate end-points [69]. As a result, careful planning is warranted for phase II trials. In addition to providing safety data, phase II studies could include functional capacity and clinical worsening events, as well as N-terminal pro-brain natriuretic peptide and haemodynamic variables, such as PVR, cardiac index and right atrial pressure.

Despite strong pre-clinical data, candidates might not show efficacy in PAH clinical trials, as observed in studies of selective serotonin reuptake inhibitors (SSRIs). SSRIs were found to attenuate and even reverse the development of pulmonary hypertension induced by chronic hypoxia or monocrotaline in animal models [71]. However, one nested case–control study with SSRIs failed to show clinical efficacy [72], and results from a phase II trial suggested that SSRI use was associated with increased mortality and clinical worsening [73]. For therapies with a novel mechanism of action in a particular indication, it is critical that phase II studies are performed in the relevant patient population. This is demonstrated using the example of the tyrosine kinase inhibitor nilotinib. Despite being approved in chronic myelogenous leukaemia and preventing angioproliferation in a rodent model of pulmonary hypertension with systemic sclerosis [74], nilotinib has not progressed beyond phase II in PAH. A phase II trial of nilotinib in patients with PAH was terminated due to serious adverse events in this population [71].

Phase III clinical trial considerations

Phase III PAH trials require both high numbers of patients and significant resources. Therefore, it is important that data from pre-clinical studies and phase I and II trials are as robust and reliable as possible before initiating phase III studies, particularly when testing novel therapies that act upon putative disease targets. The value of obtaining consistent phase II data, ahead of progressing to phase III trials, is illustrated by the experience with the tyrosine kinase inhibitor imatinib. This drug was investigated in a phase III trial (enrolling 202 patients with PAH) [75] and subsequent open-label extension [76], although results from an earlier phase II study in 59 patients with PAH had been inconsistent with respect to efficacy end-points [77]. In the phase III study (IMPRES), imatinib treatment resulted in significant improvements in 6MWD (primary end-point) and also led to improvements in a number of haemodynamic parameters [75]. However, imatinib failed to delay the time to clinical worsening (composite secondary efficacy end-point), and was associated with a high incidence of drug discontinuation and a higher than expected incidence of subdural haematoma in the open-label extension [75, 76]. Consequently, the benefit/risk ratio was deemed unfavourable and the manufacturer withdrew its application for a licence for imatinib for PAH. The IMPRES study therefore highlights the increasing importance of using appropriate outcome-based end-points in PAH trials to ensure that clinically relevant improvements in patient outcomes can be determined, and the importance of collecting relatively long-term safety data. Prior to the phase III trials with macitentan and selexipag, PAH therapies had been granted regulatory approval based on the results of short-term studies measuring exercise capacity [56–61, 78]. However, with these more recent trials there has been a shift towards the use of a composite morbidity and mortality end-point [3, 4, 62], which is now recommended as the primary end-point in pivotal phase III PAH trials [79]. Recommended components of such a primary end-point include death, lung transplantation, PAH-related hospitalisation, initiation of parenteral prostacyclin analogue therapy, disease progression (measured by a decrease in functional class and exercise capacity) and worsening of PAH symptoms (including dyspnoea, chest pain, dizziness/syncope and fatigue) [69, 79]. To ensure the robustness of the data obtained from a composite end-point, all events should be adjudicated by a blinded committee to confirm that they meet the criteria set out in the protocol. Although it is desirable that the components of a composite end-point are equally weighted in terms of clinical relevance, an important consideration is the size of the patient population to be investigated. In a rare disease such as PAH, limiting the number of end-point components to include only those with similar clinical significance could reduce the number of events obtained. This in turn could lead to a decrease in the sensitivity or statistical power of the trial in question. The integrity of the composite end-point is dictated by each and every component, and as a result the more subjective component of disease progression must be both robust and well-defined. This is particularly important given that, due to the progressive nature of the disease, this component occurred more frequently than harder components such as death or initiation of i.v. prostacyclin therapy in several studies performed to date [3, 4]. A number of long-term studies have been completed recently using variations of a composite primary end-point, including SERAPHIN [3], GRIPHON [4], COMPASS-2 [62] and AMBITION [2]. Three of these studies met their primary end-point over an observation period of 15–27 months; however, for each one the treatment effect was evident at 12 months [2–4]. This observation should be taken into account for future studies in PAH. Looking ahead there may also be a move towards the use of a primary composite end-point that reflects disease improvement rather than worsening, as one important goal of PAH therapy is the clinical improvement of patients. Such an end-point may be of particular relevance for future studies that evaluate the comparative benefits of different treatment regimens.

Conclusion

Significant progress has been made in the treatment of PAH through the development of drugs that target three key pathophysiological pathways, although management remains suboptimal for many patients. It is important that research continues into novel treatments in PAH to further improve outcomes for patients. Such research should extend to investigating targets beyond the signalling pathways already subject to pharmacological intervention. Furthermore, evolving pre-clinical studies and clinical trial designs hold promise for the continued development of treatment strategies in PAH.

Supplementary material

Supplementary Material

Please note: supplementary material is not edited by the Editorial Office, and is uploaded as it has been supplied by the author.

N. Galie ERR-0084-2016_Galie

M. Hoeper ERR-0084-2016_Hoeper

V. McLaughlin ERR-0084-2016_McLaughlin

L. Rubin ERR-0084-2016_Rubin

G. Simonneau ERR-0084-2016_Simonneau

Acknowledgements

The authors would like to thank James Glasper (nspm Ltd, Meggen, Switzerland) for medical writing assistance, funded by Actelion Pharmaceuticals Ltd (Allschwil, Switzerland).

Footnotes

  • Editorial comment in Eur Respir Rev 2016; 25: 361–363.

  • Conflict of interest: Disclosures can be found alongside this article at err.ersjournals.com

  • Provenance: The European Respiratory Review received sponsorship from Actelion Pharmaceuticals Ltd, Allschwil, Switzerland, for the publication of these peer-reviewed articles.

  • Received August 18, 2016.
  • Accepted September 27, 2016.
  • Copyright ©ERS 2016.

ERR articles are open access and distributed under the terms of the Creative Commons Attribution Non-Commercial Licence 4.0.

References

  1. ↵
    1. Humbert M,
    2. Lau EM,
    3. Montani D, et al.
    Advances in therapeutic interventions for patients with pulmonary arterial hypertension. Circulation 2014; 130: 2189–2208.
    OpenUrlFREE Full Text
  2. ↵
    1. Galiè N,
    2. Barbera JA,
    3. Frost AE, et al.
    Initial use of ambrisentan plus tadalafil in pulmonary arterial hypertension. N Engl J Med 2015; 373: 834–844.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Pulido T,
    2. Adzerikho I,
    3. Channick R, et al.
    Macitentan and morbidity and mortality in pulmonary arterial hypertension. N Engl J Med 2013; 369: 809–818.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Sitbon O,
    2. Channick R,
    3. Chin KM, et al.
    Selexipag for the treatment of pulmonary arterial hypertension. N Engl J Med 2015; 373: 2522–2533.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Humbert M,
    2. Simonneau G,
    3. Rubin LJ
    . A decade of achievement in pulmonary hypertension. Eur Respir Rev 2011; 20: 215–217.
    OpenUrlFREE Full Text
  6. ↵
    1. Sitbon O,
    2. Gaine S
    . Beyond a single pathway – combination therapy in pulmonary arterial hypertension. Eur Respir Rev 2016; 25: 408–417.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Kaitin KI
    . Deconstructing the drug development process: the new face of innovation. Clin Pharmacol Ther 2010; 87: 356–361.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Arrowsmith J
    . Trial watch: phase III and submission failures: 2007–2010. Nat Rev Drug Discov 2011; 10: 87.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Lythgoe MP,
    2. Rhodes CJ,
    3. Ghataorhe P, et al.
    Why drugs fail in clinical trials in pulmonary arterial hypertension, and strategies to succeed in the future. Pharmacol Ther 2016; 164: 195–203.
    OpenUrl
  10. ↵
    1. Perros F,
    2. Dorfmuller P,
    3. Montani D, et al.
    Pulmonary lymphoid neogenesis in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 2012; 185: 311–321.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Dorfmuller P,
    2. Perros F,
    3. Balabanian K, et al.
    Inflammation in pulmonary arterial hypertension. Eur Respir J 2003; 22: 358–363.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Nicolls MR,
    2. Taraseviciene-Stewart L,
    3. Rai PR, et al.
    Autoimmunity and pulmonary hypertension: a perspective. Eur Respir J 2005; 26: 1110–1118.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Rabinovitch M,
    2. Guignabert C,
    3. Humbert M, et al.
    Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res 2014; 115: 165–175.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Cracowski JL,
    2. Chabot F,
    3. Labarere J, et al.
    Proinflammatory cytokine levels are linked to death in pulmonary arterial hypertension. Eur Respir J 2014; 43: 915–917.
    OpenUrlFREE Full Text
  15. ↵
    1. Miyamichi-Yamamoto S,
    2. Fukumoto Y,
    3. Sugimura K, et al.
    Intensive immunosuppressive therapy improves pulmonary hemodynamics and long-term prognosis in patients with pulmonary arterial hypertension associated with connective tissue disease. Circ J 2011; 75: 2668–2674.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Kommireddy S,
    2. Bhyravavajhala S,
    3. Kurimeti K, et al.
    Pulmonary arterial hypertension in systemic lupus erythematosus may benefit by addition of immunosuppression to vasodilator therapy: an observational study. Rheumatology (Oxford) 2015; 54: 1673–1679.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    NCT02664558. A Study of Ubenimex in Patients With Pulmonary Arterial Hypertension (WHO Group 1) (LIBERTY). 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  18. ↵
    NCT02736149. Open-Label Extension Study of Ubenimex in Patients With Pulmonary Arterial Hypertension (WHO Group 1) (LIBERTY2). 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  19. ↵
    NCT01086540. Rituximab for Treatment of Systemic Sclerosis-Associated Pulmonary Arterial Hypertension (SSc-PAH). 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  20. ↵
    NCT02676947. A Therapeutic Open Label Study of Tocilizumab in the Treatment of Pulmonary Arterial Hypertension (TRANSFORM-UK). 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  21. ↵
    1. Oudiz R,
    2. Meyer C,
    3. Chin M, et al.
    Initial data report from “LARIAT”: a phase 2 study of bardoxolone methyl in PAH patients on stable background therapy. Chest 2015; 148.
  22. ↵
    NCT02657356. Bardoxolone Methyl in Patients With Connective Tissue Disease-associated Pulmonary Arterial Hypertension – CATALYST. 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  23. ↵
    NCT02234141. GS-4997 in Adults With Pulmonary Arterial Hypertension (ARROW). 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  24. ↵
    NCT01647945. FK506 (Tacrolimus) in Pulmonary Arterial Hypertension (TransformPAH). 2016. https://clinicaltrials.gov 2016. Date last accessed: October 21, 2016.
  25. ↵
    NCT01447628. IV Iron Replacement for Iron Deficiency in Idiopathic Pulmonary Arterial Hypertension (IPAH) Patients. 2016. https://clinicaltrials.gov 2016. Date last accessed: October 21, 2016.
  26. ↵
    NCT01847352. Iron Status and Hypoxic Pulmonary Vascular Responses. 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  27. ↵
    1. Frise MC,
    2. Cheng HY,
    3. Nickol AH, et al.
    Clinical iron deficiency disturbs normal human responses to hypoxia. J Clin Invest 2016; 126: 2139–2150.
    OpenUrl
  28. ↵
    NCT01446848. Oral Iron Supplementation in Pulmonary Hypertension. 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  29. ↵
    1. Chen SL,
    2. Zhang H,
    3. Xie DJ, et al.
    Hemodynamic, functional, and clinical responses to pulmonary artery denervation in patients with pulmonary arterial hypertension of different causes: phase II results from the Pulmonary Artery Denervation-1 study. Circ Cardiovasc Interv 2015; 8: e002837.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    NCT02525926. Pulmonary Artery Denervation for Treatment of Pulmonary Arterial Hypertension (DENERV'AP). 2016. https://clinicaltrials.gov Date last accessed: October 21, 2016.
  31. ↵
    1. Tian W,
    2. Jiang X,
    3. Sung YK, et al.
    Leukotrienes in pulmonary arterial hypertension. Immunol Res 2014; 58: 387–393.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Huertas A,
    2. Perros F,
    3. Tu L, et al.
    Immune dysregulation and endothelial dysfunction in pulmonary arterial hypertension: a complex interplay. Circulation 2014; 129: 1332–1340.
    OpenUrlFREE Full Text
  33. ↵
    1. Chaisson NF,
    2. Hassoun PM
    . Systemic sclerosis-associated pulmonary arterial hypertension. Chest 2013; 144: 1346–1356.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Wilkins MR
    . Pulmonary hypertension: the science behind the disease spectrum. Eur Respir Rev 2012; 21: 19–26.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Deng Z,
    2. Morse JH,
    3. Slager SL, et al.
    Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am J Hum Genet 2000; 67: 737–744.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Thomson JR,
    2. Machado RD,
    3. Pauciulo MW, et al.
    Sporadic primary pulmonary hypertension is associated with germline mutations of the gene encoding BMPR-II, a receptor member of the TGF-beta family. J Med Genet 2000; 37: 741–745.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Atkinson C,
    2. Stewart S,
    3. Upton PD, et al.
    Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor. Circulation 2002; 105: 1672–1678.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Xiong J
    . BMPR2 spruces up the endothelium in pulmonary hypertension. Protein Cell 2015; 6: 703–708.
    OpenUrl
  39. ↵
    1. Spiekerkoetter E,
    2. Tian X,
    3. Cai J, et al.
    FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension. J Clin Invest 2013; 123: 3600–3613.
    OpenUrlCrossRefPubMed
  40. ↵
    1. van Empel VP,
    2. Lee J,
    3. Williams TJ, et al.
    Iron deficiency in patients with idiopathic pulmonary arterial hypertension. Heart Lung Circ 2014; 23: 287–292.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Rhodes CJ,
    2. Wharton J,
    3. Howard L, et al.
    Iron deficiency in pulmonary arterial hypertension: a potential therapeutic target. Eur Respir J 2011; 38: 1453–1460.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. Ruiter G,
    2. Lankhorst S,
    3. Boonstra A, et al.
    Iron deficiency is common in idiopathic pulmonary arterial hypertension. Eur Respir J 2011; 37: 1386–1391.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Soon E,
    2. Treacy CM,
    3. Toshner MR, et al.
    Unexplained iron deficiency in idiopathic and heritable pulmonary arterial hypertension. Thorax 2011; 66: 326–332.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Krasuski RA,
    2. Hart SA,
    3. Smith B, et al.
    Association of anemia and long-term survival in patients with pulmonary hypertension. Int J Cardiol 2011; 150: 291–295.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Jankowska EA,
    2. Rozentryt P,
    3. Witkowska A, et al.
    Iron deficiency: an ominous sign in patients with systolic chronic heart failure. Eur Heart J 2010; 31: 1872–1880.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Brownlie Tt,
    2. Utermohlen V,
    3. Hinton PS, et al.
    Tissue iron deficiency without anemia impairs adaptation in endurance capacity after aerobic training in previously untrained women. Am J Clin Nutr 2004; 79: 437–443.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Ruiter G,
    2. Manders E,
    3. Happe CM, et al.
    Intravenous iron therapy in patients with idiopathic pulmonary arterial hypertension and iron deficiency. Pulm Circ 2015; 5: 466–472.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Viethen T,
    2. Gerhardt F,
    3. Dumitrescu D, et al.
    Ferric carboxymaltose improves exercise capacity and quality of life in patients with pulmonary arterial hypertension and iron deficiency: a pilot study. Int J Cardiol 2014; 175: 233–239.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Robinson JC,
    2. Graham BB,
    3. Rouault TC, et al.
    The crossroads of iron with hypoxia and cellular metabolism. Implications in the pathobiology of pulmonary hypertension. Am J Respir Cell Mol Biol 2014; 51: 721–729.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Rothman AM,
    2. Arnold ND,
    3. Chang W, et al.
    Pulmonary artery denervation reduces pulmonary artery pressure and induces histological changes in an acute porcine model of pulmonary hypertension. Circ Cardiovasc Interv 2015; 8: e002569.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    1. Juratsch CE,
    2. Jengo JA,
    3. Castagna J, et al.
    Experimental pulmonary hypertension produced by surgical and chemical denervation of the pulmonary vasculature. Chest 1980; 77: 525–530.
    OpenUrlCrossRefPubMed
  52. ↵
    1. Chen SL,
    2. Zhang FF,
    3. Xu J, et al.
    Pulmonary artery denervation to treat pulmonary arterial hypertension: the single-center, prospective, first-in-man PADN-1 study (first-in-man pulmonary artery denervation for treatment of pulmonary artery hypertension). J Am Coll Cardiol 2013; 62: 1092–1100.
    OpenUrlFREE Full Text
  53. ↵
    1. Zhang H,
    2. Zhang J,
    3. Xie DJ, et al.
    Pulmonary artery denervation for treatment of a patient with pulmonary hypertension secondary to left heart disease. Pulm Circ 2016; 6: 240–243.
    OpenUrlCrossRef
  54. ↵
    1. Galiè N,
    2. Manes A
    . New treatment strategies for pulmonary arterial hypertension: hopes or hypes? J Am Coll Cardiol 2013; 62: 1101–1102.
    OpenUrlFREE Full Text
  55. ↵
    1. Hoeper MM,
    2. Galie N
    . Letter by Hoeper and Galie regarding article, “Hemodynamic, functional, and clinical responses to pulmonary artery denervation in patients with pulmonary arterial hypertension of different causes: phase II results from the pulmonary artery denervation-1 study”. Circ Cardiovasc Interv 2016; 9: e003422.
    OpenUrlFREE Full Text
  56. ↵
    1. Barst RJ,
    2. Rubin LJ,
    3. Long WA, et al.
    A comparison of continuous intravenous epoprostenol (prostacyclin) with conventional therapy for primary pulmonary hypertension. N Engl J Med 1996; 334: 296–301.
    OpenUrlCrossRefPubMed
  57. ↵
    1. McLaughlin VV,
    2. Benza RL,
    3. Rubin LJ, et al.
    Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: a randomized controlled clinical trial. J Am Coll Cardiol 2010; 55: 1915–1922.
    OpenUrlFREE Full Text
    1. McLaughlin VV,
    2. Oudiz RJ,
    3. 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: 1257–1263.
    OpenUrlCrossRefPubMed
    1. Simonneau G,
    2. Rubin LJ,
    3. 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: 521–530.
    OpenUrlCrossRefPubMed
    1. Tapson VF,
    2. Torres F,
    3. Kermeen F, et al.
    Oral treprostinil for the treatment of pulmonary arterial hypertension in patients on background endothelin receptor antagonist and/or phosphodiesterase type 5 inhibitor therapy (the FREEDOM-C study): a randomized controlled trial. Chest 2012; 142: 1383–1390.
    OpenUrlCrossRefPubMed
  58. ↵
    1. Tapson VF,
    2. Jing ZC,
    3. Xu KF, et al.
    Oral treprostinil for the treatment of pulmonary arterial hypertension in patients receiving background endothelin receptor antagonist and phosphodiesterase type 5 inhibitor therapy (the FREEDOM-C2 study): a randomized controlled trial. Chest 2013; 144: 952–958.
    OpenUrlCrossRefPubMed
  59. ↵
    1. McLaughlin V,
    2. Channick RN,
    3. Ghofrani HA, et al.
    Bosentan added to sildenafil therapy in patients with pulmonary arterial hypertension. Eur Respir J 2015; 46: 405–413.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    1. Mullard A
    . Reliability of ‘new drug target’ claims called into question. Nat Rev Drug Discov 2011; 10: 643–644.
    OpenUrlCrossRefPubMed
  61. ↵
    1. Colvin KL,
    2. Yeager ME
    . Animal models of pulmonary hypertension: matching disease mechanisms to etiology of the human disease. J Pulm Respir Med 2014; 4: 198.
    OpenUrl
  62. ↵
    1. Chandrasekera PC,
    2. Pippin JJ
    . The human subject: an integrative animal model for 21st century heart failure research. Am J Transl Res 2015; 7: 1636–1647.
    OpenUrlPubMed
  63. ↵
    1. Ross DJ,
    2. Hough G,
    3. Hama S, et al.
    Proinflammatory high-density lipoprotein results from oxidized lipid mediators in the pathogenesis of both idiopathic and associated types of pulmonary arterial hypertension. Pulm Circ 2015; 5: 640–648.
    OpenUrl
  64. ↵
    1. Chelladurai P,
    2. Seeger W,
    3. Pullamsetti SS
    . Epigenetic mechanisms in pulmonary arterial hypertension: the need for global perspectives. Eur Respir Rev 2016; 25: 135–140.
    OpenUrlAbstract/FREE Full Text
  65. ↵
    1. Esch EW,
    2. Bahinski A,
    3. Huh D
    . Organs-on-chips at the frontiers of drug discovery. Nat Rev Drug Discov 2015; 14: 248–260.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Gomberg-Maitland M,
    2. Bull TM,
    3. Saggar R
    . New trial designs and potential therapies for pulmonary artery hypertension. J Am Coll Cardiol 2013; 62: D82–D91.
    OpenUrlFREE Full Text
  67. ↵
    1. Sheiner LB
    . Learning versus confirming in clinical drug development. Clin Pharmacol Ther 1997; 61: 275–291.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Hu J,
    2. Xu Q,
    3. McTiernan C, et al.
    Novel targets of drug treatment for pulmonary hypertension. Am J Cardiovasc Drugs 2015; 15: 225–234.
    OpenUrl
  69. ↵
    1. Dhalla IA,
    2. Juurlink DN,
    3. Gomes T, et al.
    Selective serotonin reuptake inhibitors and pulmonary arterial hypertension: a case-control study. Chest 2012; 141: 348–353.
    OpenUrlCrossRefPubMed
  70. ↵
    1. Sadoughi A,
    2. Roberts KE,
    3. Preston IR, et al.
    Use of selective serotonin reuptake inhibitors and outcomes in pulmonary arterial hypertension. Chest 2013; 144: 531–541.
    OpenUrl
  71. ↵
    1. Maurer B,
    2. Reich N,
    3. Juengel A, et al.
    Fra-2 transgenic mice as a novel model of pulmonary hypertension associated with systemic sclerosis. Ann Rheum Dis 2012; 71: 1382–1387.
    OpenUrlAbstract/FREE Full Text
  72. ↵
    1. Hoeper MM,
    2. Barst RJ,
    3. Bourge RC, et al.
    Imatinib mesylate as add-on therapy for pulmonary arterial hypertension: results of the randomized IMPRES study. Circulation 2013; 127: 1128–1138.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Frost AE,
    2. Barst RJ,
    3. Hoeper MM, et al.
    Long-term safety and efficacy of imatinib in pulmonary arterial hypertension. J Heart Lung Transplant 2015; 34: 1366–1375.
    OpenUrlCrossRefPubMed
  74. ↵
    1. Ghofrani HA,
    2. Morrell NW,
    3. Hoeper MM, et al.
    Imatinib in pulmonary arterial hypertension patients with inadequate response to established therapy. Am J Respir Crit Care Med 2010; 182: 1171–1177.
    OpenUrlCrossRefPubMed
  75. ↵
    1. Hoeper MM,
    2. Leuchte H,
    3. Halank M, et al.
    Combining inhaled iloprost with bosentan in patients with idiopathic pulmonary arterial hypertension. Eur Respir J 2006; 28: 691–694.
    OpenUrlAbstract/FREE Full Text
  76. ↵
    1. McLaughlin VV,
    2. Badesch DB,
    3. Delcroix M, et al.
    End points and clinical trial design in pulmonary arterial hypertension. J Am Coll Cardiol 2009; 54: S97–S107.
    OpenUrlFREE Full Text
PreviousNext
Back to top
View this article with LENS
Vol 25 Issue 142 Table of Contents
European Respiratory Review: 25 (142)
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Future perspectives in pulmonary arterial hypertension
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Future perspectives in pulmonary arterial hypertension
Gérald Simonneau, Marius M. Hoeper, Vallerie McLaughlin, Lewis Rubin, Nazzareno Galiè
European Respiratory Review Dec 2016, 25 (142) 381-389; DOI: 10.1183/16000617.0084-2016

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Future perspectives in pulmonary arterial hypertension
Gérald Simonneau, Marius M. Hoeper, Vallerie McLaughlin, Lewis Rubin, Nazzareno Galiè
European Respiratory Review Dec 2016, 25 (142) 381-389; DOI: 10.1183/16000617.0084-2016
Reddit logo Technorati logo Twitter logo Connotea logo Facebook logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Abstract
    • Introduction
    • Potential PAH therapies currently in development
    • Clinical trial design in novel therapy development in PAH
    • Conclusion
    • Supplementary material
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • Pulmonary vascular disease
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Bariatric surgery: a potential cure for asthma?
  • Regimens to treat MDR-TB: past, present and future
Show more Mini-Review

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERR

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising
  • Sponsorship

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN: 0905-9180
Online ISSN: 1600-0617

Copyright © 2023 by the European Respiratory Society