throbber
REVIEW
`PULMONARY ARTERIAL HYPERTENSION
`
`Epoprostenol and pulmonary arterial
`hypertension: 20 years of clinical
`experience
`
`Olivier Sitbon1,2,3 and Anton Vonk Noordegraaf4
`
`Affiliations: 1Universite Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
`2AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, Hôpital Bicêtre,
`Le Kremlin-Bicêtre, France. 3INSERM UMR_S 999, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France.
`4Dept of Pulmonology, Vrje Universiteit Medical Centre, Amsterdam, the Netherlands.
`
`Correspondence: Olivier Sitbon, Service de Pneumologie, Hôpital de Bicêtre, 78 rue Général Leclerc, 94270
`Le Kremlin-Bicêtre, France. E-mail: olivier.sitbon@aphp.fr
`
`@ERSpublications
`The evolution of the place of epoprostenol in the management of pulmonary arterial hypertension
`http://ow.ly/OkY3303N2CX
`
`Cite this article as: Sitbon O, Vonk Noordegraaf A. Epoprostenol and pulmonary arterial hypertension:
`20 years of clinical experience. Eur Respir Rev 2017; 26: 160055 [https://doi.org/10.1183/16000617.0055-
`2016].
`
`ABSTRACT Epoprostenol was the first therapy to be approved for the treatment of pulmonary arterial
`hypertension (PAH). In the 20 years since the introduction of this prostacyclin analogue, the outlook for
`patients with PAH has improved, with survival rates now double those from the era before the
`development of disease-specific treatments. Today, there are a large amount of data on the clinical role of
`prostacyclin treatments and a body of evidence attesting the efficacy of epoprostenol in improving exercise
`capacity, key haemodynamic parameters and PAH symptoms, as well as in reducing mortality. The place
`of epoprostenol in the therapeutic management of PAH continues to evolve, with the development of new
`formulations and use in combination with other drug classes. In this review, we provide a historical
`perspective on the first 20 years of epoprostenol, a therapy that led to evidence-based study of PAH-
`specific treatments and the subsequent expansion of treatment options for PAH.
`
`Introduction
`Pulmonary arterial hypertension (PAH) is a rare, progressive disease associated with significant morbidity
`[1–4]. The disease is characterised by elevated pulmonary artery pressure (PAP) and pulmonary vascular
`resistance (PVR). Left untreated, PAH leads to right-sided heart failure and premature death [1–4]. In the
`1980s, median survival was 2.8 years from diagnosis; the 5-year survival rate was 34% [5]. Although PAH
`remains
`incurable,
`insights
`into the underlying mechanisms have led to the development of
`disease-specific treatments that have approximately doubled survival rates [6–8]. Today there are 10
`approved PAH-specific therapies [9]. The first of these was the prostacyclin analogue epoprostenol, which
`was approved in 1995 in the USA before being licensed, a year later, in Europe. This treatment is still
`regarded as the gold standard to which other therapies should be compared [10–12].
`
`In this review, we take a historical perspective on epoprostenol and its place in PAH management over the
`past 20 years. We also consider the role of epoprostenol at a time when new formulations that are stable at
`room temperature are becoming more widely available. This review is based on our knowledge of the field,
`
`Received: June 02 2016 | Accepted after revision: Aug 28 2016
`
`Conflict of interest: Disclosures can be found alongside this article at err.ersjournals.com
`
`Provenance: Submitted article, peer reviewed.
`
`Copyright ©ERS 2017. ERR articles are open access and distributed under the terms of the Creative Commons
`Attribution Non-Commercial Licence 4.0.
`
`https://doi.org/10.1183/16000617.0055-2016
`
`Eur Respir Rev 2017; 26: 160055
`
`IPR2021-00406
`United Therapeutics EX2051
`
`

`

`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`supplemented by a methodical literature search designed to provide a comprehensive historical overview of
`milestones since the approval of epoprostenol. The literature review comprised searches of a database of
`PAH pdf
`files, PubMed, Scopus and the abstract database Searchlight for relevant publications on
`in PAH using “epoprostenol” and “pulmonary arterial hypertension” as keywords. The
`epoprostenol
`period for the PubMed, Scopus and Searchlight searches was 1994 to December 2014 and all English
`language citations were captured. Titles and abstracts of original articles were manually searched and
`potentially relevant articles selected; the authors reviewed the resulting publication list and agreed papers
`of
`interest. Review articles that capture the historical narrative of
`the development and study of
`epoprostenol have been included.
`
`Background
`Impact of epoprostenol on PAH treatment
`Before the approval of epoprostenol, PAH was treated using a combination of non-specific treatments
`including warfarin, calcium-channel blockers, digoxin, diuretics and supplemental oxygen. These therapies
`targeted specific aspects of the disease, but demonstrated little short-term or long-term benefit on major
`haemodynamic parameters or clinical outcomes (reviewed in [3, 13]), with the exception of long-term
`calcium channel blockers that improved outcomes in a majority of patients classed using strong criteria
`such as acute pulmonary vasodilator responders [14]. The introduction of epoprostenol transformed the
`care of patients with PAH [15]: epoprostenol improved exercise capacity, key haemodynamic parameters
`and PAH symptoms [16, 17] and, importantly, was the first pharmacological therapy to reduce mortality
`[18]. Twenty years later, epoprostenol remains the only treatment to have reduced mortality in patients
`with idiopathic PAH (IPAH) in a randomised study [17].
`
`Early studies of epoprostenol improved our understanding of pulmonary hypertension from associated
`causes, and led to evidence-based studies of PAH-specific treatments and the subsequent expansion of
`treatment options for PAH [19, 20]. Lessons learned from studying epoprostenol have informed the
`development of other inhaled, oral and subcutaneously administered prostanoid therapies. More recently,
`epoprostenol and some other prostacylins have been shown to be effective and well tolerated when used in
`combination with other PAH drug classes [21–25]. Therapies previously reserved for patients with severe
`disease are now being considered for use in those with earlier stage disease, in an attempt to further
`prolong life and improve patient outcomes [11, 12, 26, 27].
`
`Current recommendations and evolving terminology
`The 2015 European Society of Cardiology and European Respiratory Society guidelines for the diagnosis and
`treatment of pulmonary hypertension outline the continued place of epoprostenol within treatment options for
`patients with PAH (World Health Organization (WHO) group 1 pulmonary hypertension) [11, 12]. Based on
`level A evidence of efficacy (data derived from multiple randomised clinical
`trials or meta-analyses),
`intravenous epoprostenol is recommended as a class I monotherapy in patients with PAH (WHO group 1)
`with WHO functional class (FC) III or IV. It should also be considered (class IIa) for use in upfront
`combination therapy in patients with WHO FC III or IV alongside bosentan, and alongside bosentan and
`sildenafil, based on level C efficacy evidence (consensus of opinion of experts and/or small studies, retrospective
`studies and registries) [11, 12]. Today, epoprostenol is approved in many countries including the USA where it
`is indicated to improve exercise capacity in patients with WHO group 1 pulmonary hypertension (specifically,
`patients with IPAH, heritable PAH and PAH associated with coexisting conditions such as connective tissue
`disease (PAH–CTD)) based on studies including predominantly patients with New York Heart Association
`(NYHA) FC III–IV symptoms [28]. The term IPAH had not been developed at the time of initial approval of
`epoprostenol; thus, early studies describe patients with primary pulmonary hypertension (PPH) which was the
`preferred term at this time. Current terminology will be used within this review wherever appropriate. WHO
`FC and NYHA FC are used interchangeably when characterising patients with PAH.
`
`Early research and discovery
`The development of epoprostenol stemmed from the discovery of endogenous prostacyclins in the
`vasculature by MONCADA et al. [29] in the 1970s. Soon after this, epoprostenol was synthesised and shown
`to have anti-platelet activity and vasodilatory effects in humans [30–32]. One of the first patients given
`epoprostenol was a young, cyanotic, hospitalised and bed-bound woman with IPAH. Intravenous
`epoprostenol improved haemodynamic parameters and clinical symptoms and the patient was discharged
`to continue long-term treatment [33]. Another early proof-of-concept study involving seven patients with
`IPAH showed that epoprostenol increased cardiac output and reduced PAP and PVR [34]. These and
`other early explorations preceded the innovative clinical studies that
`led to the first approval of
`epoprostenol for the treatment of patients with IPAH in 1995 [16, 17].
`
`https://doi.org/10.1183/16000617.0055-2016
`
`2
`
`

`

`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`Epoprostenol in profile
`Pharmacology
`Epoprostenol is a synthetic analogue of the naturally occurring eicosanoid prostacyclin (prostaglandin I2
`or PGI2), which is the main metabolite of arachidonic acid [30, 35]. Endogenous prostacyclin is produced
`predominantly by endothelial cells and acts both on local vasculature and on blood cells that adhere to the
`endothelium [26]. In PAH, the normal release of endogenous prostacyclin is depressed and release of the
`vasoconstrictor thromboxane A2 is increased [36]. In addition, pulmonary endothelin-1 homeostasis is
`abnormal, and this may contribute to the progressive rise in PVR that typifies PAH [37].
`
`Prostacyclins (and related prostanoids) have direct and potent vasodilatory effects resulting from their
`action on vascular smooth muscle cells; they inhibit platelet aggregation and thrombus formation, and
`have antiproliferative and anti-inflammatory actions (figure 1) [26, 38]. These effects are mediated via
`G-protein-coupled prostanoid IP receptors
`in blood vessels,
`leukocytes and thrombocytes
`[26].
`Epoprostenol may also have indirect vasodilatory effects owing to inhibition of production of the potent
`vasoconstrictor endothelin-1 [39]. In patients with PAH, therapeutic use of prostanoids is associated with
`immediate vasodilatory action in the pulmonary and systemic circulation and resultant,
`longer-term
`haemodynamic changes that contribute to additional decreases in PVR [26]. It has been suggested that
`indirect positive inotropic effects of therapy may also ameliorate systemic hypotension [26]; however, such
`effects have not been established in any model of chronic pulmonary hypertension, and the effect of
`epoprostenol in chronic pressure overload on the right ventricle remains unknown.
`
`The pharmacokinetic properties of the original formulation of epoprostenol are dominated by the lability
`of the molecule in aqueous fluids at physiological temperature and pH. Epoprostenol has a short
`elimination half-life of approximately 3–6 min in human blood, which necessitates administration via
`continuous intravenous infusion [15]. Treatment has to be initiated by an experienced physician, and
`long-term use requires a permanent central venous catheter and portable infusion pump [11, 12].
`
`Clinical studies with epoprostenol in the treatment of PAH
`Today there is substantial evidence from randomised controlled trials (RCTs) supporting the use of
`prostacyclin treatments in PAH, while data from observational studies and registries provide real-world
`evidence and experiences of patient management (for reviews see [6, 40]). Table 1 provides an overview of
`key studies that have contributed to our understanding of the clinical profile of epoprostenol.
`
`RCTs with epoprostenol in PAH
`Epoprostenol was initially approved for use in patients with “PPH and moderate-to-severe functional
`status”, based on data from two RCTs [16, 17]. The first studied 24 patients with IPAH (NYHA FC II−IV),
`randomised to receive either intravenous epoprostenol or the conventional treatment of the time for
`8 weeks [16]. Epoprostenol was associated with a significant and sustained decrease in total pulmonary
`resistance (–7.9 units; p=0.022) but there was no change for patients on conventional treatment (–0.2
`units), and six out of 10 patients in the epoprostenol group compared with only one out of nine patients in
`the conventional treatment group had reductions in mean PAP (mPAP) of greater than 10 mmHg. This
`study also reported that continued epoprostenol treatment for up to 18 months was associated with
`
`Prostanoids
`
`Vessels
`
`Leukocytes
`
`SMC
`
`Fibroblasts
`
`EC
`
`Platelets
`
`Mono
`
`MA
`
`PMN
`
`T-cells
`
`Vaso-
`dilation
`
`Anti-
`proliferation
`
`Matrix
`secretion
`
`Anti-
`coagulation
`
`NF-κB
`TNF-α
`IL-1
`IL-10
`
`MAPK
`iNOS
`
`Burst
`Elastase/secretion
`Leukotrienes
`
`TNF-α
`IFN-α
`IL-2
`
`FIGURE 1 The effects of prostanoids on vasculature and blood cells; a variety of vascular cells, platelets and leukocytes have been identified as
`targets for the antiproliferative, anti-inflammatory and anti-aggregatory actions of prostaglandins. SMC: smooth muscle cells; EC: endothelial
`cells; Mono: mononuclear cells; NF: nuclear factor; TNF: transforming nuclear factor; IL: interleukin; MA: macrophages; MAPK: mitogen-
`activated protein kinase; iNOS: inducible nitric oxide synthase; PMN: polymorphonuclear neutrophils; Burst: generation of reactive oxygen
`species. Reproduced from [38] with permission from the publisher.
`
`https://doi.org/10.1183/16000617.0055-2016
`
`3
`
`

`

`TABLE 1 Overview of key studies that have contributed to our understanding of the clinical profile of epoprostenol
`
`Study, first author year [ref.]
`
`Aetiology of PAH
`
`Study design
`
`Treatments/intervention
`
`Patient characteristics
`
`Efficacy assessments/ primary
`endpoint and key outcomes
`
`PVR changes
`
`Safety data
`
`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`Headache (n=6); nausea (n=4);
`vomiting (n=2); cutaneous
`flushing (n=5); diplopia (n=1,
`resolved on discontinuation);
`systemic hypertension
`during dose-ranging (n=2;
`resolved on discontinuation);
`temporary significant
`reduction in systemic blood
`pressure during continuous
`infusion (n=1)
`Sterile pleural effusion ascites
`(treated with diuretics);
`cannula-associated
`Staphylococcal bacteraemia
`(resolved by cannula
`change)
`
`Loose stools (100%), jaw pain
`(57%) and photosensitivity
`(36%) were common with
`epoprostenol. One patient
`discontinued owing to
`pulmonary oedema; most
`complications were linked
`with the drug-delivery
`system
`
`Continued
`
`Early studies in patients
`with PAH
`RUBIN 1982 [34]
`
`IPAH
`
`Exploratory study
`
`Seven patients
`
`Dose-ranging protocol
`(starting dose
`2 ng·kg−1·min−1 to
`maximum
`12 ng·kg−1·min−1) and
`continuous i.v.
`epoprostenol in three
`patients for up to 48 h
`
`mPAP decreased in six out of seven
`patients and total pulmonary
`resistance decreased by >20%
`in all patients; cardiac output
`and stroke volume increased
`by >40%
`
`Total pulmonary
`resistance 17.1±8.7
`units at baseline versus
`9.7±5.9 units following
`epoprostenol infusion
`(mean±SD)
`
`HIGENBOTTAM 1983 [33]
`
`IPAH
`
`Key RCTs in patients
`with PAH
`RUBIN 1990 [16]
`
`IPAH
`
`Case: first report of
`long-term i.v.
`epoprostenol therapy
`
`Continuous i.v.
`epoprostenol 4–
`20 ng·kg−1·min−1
`
`Woman with uncontrolled
`post-partum PH
`
`Decreased PVR, improved
`oxygenation and exercise
`tolerance allowed patient to live
`independently at home
`
`PVR fell from baseline 25–
`30 units to 15 units;
`values maintained over
`10 months
`
`8-week RCT with an
`18-month non-RCT
`extension
`
`Total pulmonary
`resistance significantly
`decreased on
`epoprostenol
`
`Continuous i.v.
`epoprostenol (starting
`dose 1–2 ng·kg−1·min−1)
`versus conventional
`treatment (optimum
`doses of oral
`vasodilators,
`anticoagulants,
`supplemental oxygen,
`cardiac glycosides and
`diuretics)
`
`24 patients (NYHA FC II–IV) Epoprostenol significantly
`decreased total pulmonary
`resistance after 8 weeks
`(decrease of 7.9 units from
`baseline of 21.6 units) (p=0.022)
`versus conventional therapy
`(decrease of 0.2 units from
`baseline of 20.6)
`(non-significant). Six out of 10
`patients receiving epoprostenol
`showed >10 mmHg reductions in
`mPAP versus one out of nine
`patients on conventional
`treatment (p=0.057).
`Haemodynamic improvements
`were maintained over 18 months
`in nine patients
`
`https://doi.org/10.1183/16000617.0055-2016
`
`4
`
`

`

`TABLE 1 Continued
`
`Study, first author year [ref.]
`
`Aetiology of PAH
`
`Study design
`
`Treatments/intervention
`
`Patient characteristics
`
`Efficacy assessments/ primary
`endpoint and key outcomes
`
`PVR changes
`
`Safety data
`
`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`Exercise capacity by 6MWD
`(primary endpoint): patients on
`epoprostenol (n=41) showed
`improvements in median change
`in distance walked from baseline
`to week 12 (median increase,
`31 m; median distance of 315 m
`at baseline, 362 m at 12 weeks)
`versus conventional therapy
`(median decrease, 29 m; median
`distance of 270 m at baseline,
`204 m at 12 weeks) (p<0.002,
`nonparametric analysis). Mean
`distance walked increased by
`32 m in the epoprostenol group
`(316 m at baseline; 348 m at
`week 12) and decreased by 15 m
`in the conventional therapy
`group (272 m at baseline; 257 m
`at week 12) (p<0.003, parametric
`analysis). mPAP changes: –8%
`for epoprostenol versus +3% for
`conventional therapy (difference
`in mean change, −6.7 mmHg;
`95% CI, –10.7 to −2.6 mmHg;
`p<0.002). Indices of HRQoL
`improved in the epoprostenol
`group (p<0.01). Eight deaths in
`conventional therapy group
`versus no mortality in
`epoprostenol group (p=0.003)
`Exercise capacity by 6MWD
`(primary endpoint): patients on
`epoprostenol (n=56) showed
`improvements from baseline
`(median, 270 m) to week 12
`(316 m) versus conventional
`therapy (240 m at baseline;
`192 m at week 12) (difference in
`median distance walked, 108 m,
`95% CI, 55.2–180.0 m, p<0.001).
`Change from baseline in mPAP:
`epoprostenol, −5.03±1.09 mmHg
`versus conventional therapy,
`+0.94±1.10 mmHg (difference
`between groups, −5.97 mmHg;
`95% CI, −8.98 to −2.96). 21
`patients on epoprostenol versus
`zero patients on conventional
`therapy showed improvements in
`NYHA FC
`
`mPVR changes of −21%
`for epoprostenol versus
`+9% for conventional
`therapy (difference in
`mean change:
`−4.9 mmHg·L−1·min−1;
`95% CI: −7.6 to −2.3;
`p<0.001)
`
`Jaw pain, diarrhoea, flushing,
`headaches, nausea and
`vomiting were frequent.
`Four episodes of
`catheter-related sepsis; one
`thrombotic event. Delivery
`system-related issues
`included device malfunction
`(n=26) and irritation/
`infection (n=7), bleeding
`(n=4) and pain (n=4) at
`catheter site
`
`PVR change from baseline:
`epoprostenol −4.58
`±0.76 mmHg·L−1·min−1
`versus conventional
`therapy 0.92±0.56
`mmHg·L−1·min−1
`(mean±SE) (difference,
`−5.50 mmHg·L−1·min−1;
`95% CI, −7.33 to −3.67)
`
`Jaw pain (75% versus 0%),
`anorexia (66% versus 47%),
`nausea (41% versus 16%),
`diarrhoea (50% versus 5%)
`and depression (13% versus
`4%) more common in
`epoprostenol versus
`conventional therapy group,
`respectively. Drug-delivery
`system associated with eight
`catheter-related AEs,
`including sepsis, cellulitis,
`haemorrhage and
`pneumothorax (4% each)
`
`Continued
`
`BARST 1996 [17]
`
`IPAH
`
`12-week, prospective,
`multicentre, open-label
`RCT
`
`81 patients with
`severe disease
`(NYHA FC III–IV)
`
`Continuous i.v.
`epoprostenol (starting
`dose 2 ng·kg−1·min−1
`to maximum
`tolerated dose of
`9.2±0.5 ng·kg−1·min−1)
`plus conventional
`treatment
`(anticoagulants, oral
`vasodilators, diuretic
`agents, cardiac
`glycosides and
`supplemental oxygen)
`versus conventional
`treatment alone
`
`BADESCH 2000 [18]
`
`PAH secondary to
`scleroderma
`
`12-week, prospective,
`multicentre, open-label
`RCT
`
`111 patients with
`moderate-to-severe
`disease
`
`Continuous i.v.
`epoprostenol (starting
`dose ⩽2 ng·kg−1·min−1
`to mean dose
`11.2 ng·kg−1·min−1 at
`week 12) plus
`conventional treatment
`versus conventional
`treatment alone
`
`https://doi.org/10.1183/16000617.0055-2016
`
`5
`
`

`

`TABLE 1 Continued
`
`Study, first author year [ref.]
`
`Aetiology of PAH
`
`Study design
`
`Treatments/intervention
`
`Patient characteristics
`
`Efficacy assessments/ primary
`endpoint and key outcomes
`
`PVR changes
`
`Safety data
`
`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`Not reported in detail. One
`patient discontinued
`
`Local infections at catheter site
`(119 episodes; 0.24 per
`person-year), sepsis (70
`episodes; 0.14 per
`person-year), tunnel
`infections (10 episodes; 0.02
`per person-year), catheter
`replacement required (72
`instances; 0.15 per
`person-year)
`
`Jaw pain, headache, diarrhoea,
`flushing, leg pain and
`nausea/vomiting were
`common. Catheter-related
`sepsis (76 episodes in 53
`patients; 0.19 per
`patient-year); four deaths
`due to severe
`catheter-related infections
`(three were nosocomial
`infections acquired in
`intensive care); seven
`deaths due to severe
`pulmonary oedema
`N/A
`
`PVR at baseline (mean±SD):
`16.7±6.4 units versus
`10.2±5.4 units at first
`follow-up (p<0.0001)
`
`Total pulmonary
`resistance (mean±SD):
`37.3±10.5 units·m−2 at
`baseline; 25.4
`±6.6 units·m−2 at
`3 months; 25.0
`±6.9 units·m−2 at 1 year,
`(both p<0.0001 versus
`baseline)
`
`PVR decreased by
`700 dyn·s−1·cm–5
`(p<0.0001) and
`299 dyn·s−1·cm–5
`(p=0.009) in
`treatment-naïve and
`treatment-experienced
`patients, respectively
`
`Significant reduction in maximum
`systolic pressure gradient
`between right ventricle and right
`atrium (from mean±SD 84.1±24.1
`to 62.7±18.2 mmHg; p<0.01). 1-,
`2- and 3-year survival rates:
`80%, 76% and 49%, respectively
`Significant changes in mean right
`atrial pressure, mPAP, cardiac
`output, cardiac index and PVR
`between baseline and first
`follow-up (all p<0.0001). 1-, 2-
`and 3-year survival rates: 87.8%,
`76.3% and 62.8%, respectively
`versus expected survival of
`58.9%, 46.3% and 35.4%,
`respectively (p<0.001 at
`all time points)
`Baseline 6MWD (mean±SD): 251
`±144 m; 3 month 6MWD: 376
`±114 m (p<0.001), with 90% of
`patients achieving an
`improvement in 6MWD. Among
`patients on epoprostenol for
`1 year, mPAP, cardiac index,
`oxygen saturation and total
`pulmonary resistance
`significantly changed from
`baseline at 3 months and 1 year.
`1-, 2-, 3- and 5-year survival
`rates: 85%, 70%, 63% and 55%,
`respectively
`Improvements in haemodynamic
`parameters and clinical
`outcomes reported according to
`whether patients were
`treatment-naïve or
`treatment-experienced (previous
`PAH therapy). 4 months of
`epoprostenol increased 6MWD
`by 146 m (p<0.0001) and by 41 m
`(p=0.03) in treatment-naïve and
`treatment-experienced patients,
`respectively. 1-year and 3-year
`survival rates from treatment
`initiation were 84% and 69%,
`respectively. Greatest survival
`benefit was in treatment-naïve
`patients given upfront
`combination epoprostenol plus
`oral therapy (1-year survival,
`92%; 3-year survival, 88%)
`
`Continued
`
`Selected non-RCT studies in
`patients with PAH
`SHAPIRO 1997 [41]
`
`IPAH
`
`Observational,
`single-centre study
`
`Continuous i.v.
`epoprostenol increased
`to 1–2 ng·kg−1·min−1
`every 2 months
`
`69 patients (NYHA FC III–
`IV); 18 followed for
`>330 days
`
`MCLAUGHLIN 2002 [42]
`
`IPAH
`
`Observational,
`single-centre, registry
`database study
`
`Continuous i.v.
`epoprostenol
`
`162 patients (NYHA FC III–
`IV), followed for mean
`36.3 months
`
`SITBON 2002 [8]
`
`IPAH
`
`Observational,
`single-centre study
`
`Continuous i.v.
`epoprostenol (mean
`dose 14 ng·kg−1·min−1)
`
`178 patients (NYHA FC III–
`IV), followed for mean
`±SD 26±21 months
`
`BERGOT 2014 [43]
`
`IPAH, heritable or
`anorexigen-associated
`PAH
`
`Observational, French PH
`registry study (2006–
`2010)
`
`Continuous i.v.
`epoprostenol (dose not
`stated)
`
`209 patients (NYHA FC II–
`IV)
`
`https://doi.org/10.1183/16000617.0055-2016
`
`6
`
`

`

`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`TABLE 1 Continued
`
`Study, first author year [ref.]
`
`Aetiology of PAH
`
`Study design
`
`Treatments/intervention
`
`Patient characteristics
`
`Efficacy assessments/ primary
`endpoint and key outcomes
`
`PVR changes
`
`Safety data
`
`Selected clinical trials of
`reformulations
`TAMURA 2013 [81]
`
`IPAH or heritable PAH
`
`12-week, open-label,
`prospective,
`single-arm, two-centre
`exploratory study
`
`Switch to continuous i.v.
`epoprostenol AS
`(started on same dose;
`mean starting dose
`40.13 ng·kg−1·min−1)
`
`Eight patients (NYHA FC I–
`III) receiving stable
`epoprostenol dose
`
`SITBON 2014 [74]
`
`IPAH, heritable PAH,
`PAH-CTD or PAH
`associated with drugs/
`toxins
`
`3-month, open-label,
`prospective,
`multicentre, single-arm
`study
`
`41 patients (NYHA FC I–III)
`receiving stable
`epoprostenol dose
`
`Switch to continuous i.v.
`epoprostenol AS
`(starting dose±10% of
`previous epoprostenol
`dose; six patients
`required dose
`adjustment during
`study)
`
`PROVENCHER 2015 [80]
`
`IPAH, heritable PAH or
`PAH associated with
`concomitant conditions
`
`4-week, open-label,
`prospective,
`multicentre, single-arm
`study
`
`Switch to continuous i.v.
`pH-adjusted
`epoprostenol
`reformulation (started
`on same dose)
`
`16 patients (WHO FC II–III)
`receiving stable
`epoprostenol dose
`
`PVR was 448.3
`±158.1 dyn·s−1·cm−5 at
`baseline and 453.6
`±175.3 dyn·s−1·cm−5 at
`week 12 (mean±SD)
`
`No significant changes in
`haemodynamic factors or
`NT-proBNP concentrations from
`baseline to week 12; WHO FC
`also remained unchanged
`Patient-reported improvement in
`convenience on TSQM-9
`(p=0.0313). No unexpected safety
`or tolerability concerns after
`switching formulations
`
`PVR decreased by 8.0
`±116.8 dyn·s−1·cm–5 at
`month 3 (mean±SD)
`
`PVR was 8.4
`±5.5 mmHg·L−1·min−1
`at baseline and 6.9
`±3.6 mmHg·L−1·min−1
`2-h post transition
`(mean±SD)
`
`No clinically relevant changes in
`haemodynamics, exercise
`tolerance or NT-proBNP from
`baseline to month 3; NYHA FC
`improved in one patient and
`worsened in four patients.
`Patient-reported improvement in
`convenience on TSQM-9 (mean
`score change, +12.7±20.0 (95%
`CI, 6.1–19.3). Adverse events
`consistent with those described
`previously
`No significant changes in
`Short-Form 36 scores, 6MWD,
`Borg dyspnoea index,
`NT-proBNP or dose of
`epoprostenol after 4 weeks
`versus baseline; WHO FC
`improved in one patient. Small
`improvements in mean scores
`on most questions in 15-item
`study-specific HRQoL
`questionnaire; 14 out of 16
`patients preferred the
`reformulated product to the
`previous formulation. No
`significant changes in
`haemodynamic parameters in
`patient subgroup assessed 2 h
`post transition
`
`18 adverse events experienced
`in seven patients;
`gastrointestinal disorders
`(six events in five patients);
`infections/infestations (three
`events in three patients);
`skin/subcutaneous
`disorders (three events in
`two patients), ear-related
`(one event); musculoskeletal
`(one event); nervous system
`(one event); respiratory (one
`event); device-related (two
`events in two patients)
`Adverse events occurring in
`>5% of patients included:
`headache (n=12),
`nasopharyngitis (n=7), jaw
`pain (n=6), flushing/hot
`flush (n=6), dyspnoea (n=5),
`device connection issue
`(n=3), epistaxis (n=3),
`extremity pain (n=3) and
`palpitations (n=3)
`
`Three and nine patients
`experienced adverse events
`during run-in and treatment
`phases, respectively. Severe
`events reported were not
`considered related to study
`drug and included
`device-related infections
`(n=2) in run-in period and
`back pain (n=1) in treatment
`period; proportions with
`adverse events considered
`related to study drug were
`similar in run-in and
`treatment periods (one
`versus three subjects,
`respectively)
`
`This table does not represent a comprehensive list of all clinical studies on epoprostenol. PAH: pulmonary arterial hypertension; RCT; randomised clinical trial; IPAH: idiopathic PAH;
`mPAP: mean pulmonary arterial pressure; PH: pulmonary hypertension; PVR: pulmonary vascular resistance; NYHA FC: New York Heart Association Functional Class; 6MWD: 6-min
`walking distance; HRQoL: health-related quality of life; mPVR: mean PVR; AE: adverse event; N/A: not applicable; NT-proBNP: N-terminal prohormone of brain natriuretic peptide; PAH-
`CTD: PAH associated with connective tissue disease; TSQM-9: treatment satisfaction questionnaire for medication; WHO: World Health Organization.
`
`https://doi.org/10.1183/16000617.0055-2016
`
`7
`
`

`

`PULMONARY ARTERIAL HYPERTENSION | O. SITBON AND A. VONK NOORDEGRAAF
`
`persistent haemodynamic effects (table 1) [16]. The second, pivotal RCT was a study in 81 patients with
`IPAH (NYHA FC III or IV) that compared treatment with intravenous epoprostenol for 12 weeks in
`addition to conventional therapy with conventional therapy alone (table 1) [17]. This study demonstrated
`that epoprostenol treatment improved exercise capacity as shown by a median increase from baseline in
`6-min walking distance (6MWD) of 31 m for patients receiving epoprostenol compared with a decrease of
`29 m for those receiving conventional
`therapy alone (p<0.002). Key cardiopulmonary variables also
`improved significantly in the epoprostenol-treated group, with a change in mPAP of –8% compared with
`+3% in the conventional therapy group (p<0.002), and a significant mean change in PVR of –21% in those
`receiving epoprostenol versus +9% with conventional therapy alone (p<0.001). This study also reported that
`treatment with epoprostenol conferred a survival advantage over conventional therapy alone, with eight
`patients on conventional therapy dying during the study compared with none in the epoprostenol-treated
`group (p=0.003). This remains the only RCT in which a treatment approved for PAH reduced mortality.
`
`In 2000, another pivotal RCT in patients with moderate-to-severe PAH (patients with PAH due to scleroderma
`(PAH–CTD)), was published (table 1) [18]. This study involved 111 patients randomised to receive continuous
`epoprostenol plus conventional therapy or conventional therapy alone for 12 weeks. Epoprostenol treatment was
`associated with significant improvements in exercise capacity at 12 weeks as demonstrated by a median change
`from baseline in 6MWD of +63.5 m in the epoprostenol group compared with –36.0 m in the conventional
`treatment only group (p<0.001). Patients receiving epoprostenol experienced improvements in haemodynamic
`parameters compared with those on conventional therapy alone, achieving a change from baseline in mPAP of
`–5.03 mmHg compared with +0.94 mmHg in the conventional therapy only group, and a mean change from
`
`
`
`−1·min−1 compared with +0.92 mmHg·L−1·min−1 for the
`baseline in PVR at 12 weeks of –4.58 mmHg·L
`conventional treatment only group. Long-term outcome data showed improved survival in patients receiving
`epoprostenol during a 3-year extension period compared with historical controls [44].
`
`Non-RCTs and observational studies
`There is a strong body of evidence from non-RCTs showing that long-term treatment with continuous
`intravenous
`epoprostenol
`is
`associated with sustained improvements
`in exercise
`capacity and
`haemodynamic parameters. Some studies also reported improved survival in epoprostenol-treated patients
`compared with historical controls and have supported the role of epoprostenol as a bridge to lung
`transplantation or heart and lung transplantation [8, 41–43, 45–49]. One of the earliest non-RCTs to
`describe long-term outcomes of epoprostenol therapy was an open-label, multicentre, uncontrolled study
`in 18 patients with IPAH (NYHA FC II−IV) [45]. This study reported that improvements in the primary
`endpoint, change in 6MWD from baseline at 6 months, were sustained at 18 months, and demonstrated
`that haemodynamic improvements, such as an increase in cardiac index and reduction in total pulmonary
`resistance, were maintained over 12 months of treatment. Furthermore, in the 17 patients with NYHA FC
`III−IV followed for 37−69 months, survival was significantly improved compared with historical controls
`who had not received PAH-specific therapy (p=0.045) [45]. The literature also includes reports from
`small-scale studies of the short- and long-term benefits of continuous intravenous epoprostenol in patients
`with PAH–CTD associated with scleroderma and systemic lupus erythematosus [50–53].
`
`The survival benefits of long-term treatment with epoprostenol were also highlighted by an early US
`followed 69 patients with IPAH (NYHA FC III–IV)
`observational
`study that
`[41]. Continuous
`epoprostenol therapy decreased PAP and was associated with 1-, 2- and 3-year survival rates of 80%, 76%
`and 49%, respectively, compared with historical cohort survival rates of 56% at 20 months and 47% at
`30 months [41]. Subsequently, data from two other single-centre non-RCTs also reported improved
`survival in IPAH cohorts treated with long-term epoprostenol. One study that followed 162 consecutive
`patients with IPAH reported that continuous epoprostenol treatment for at least 1 year resulted in
`significantly greater survival rat

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket