throbber
Journal of the American College of Cardiology
`© 2004 by the American College of Cardiology Foundation
`Published by Elsevier Inc.
`
`Vol. 43, No. 12 Suppl S
`ISSN 0735-1097/04/$30.00
`doi:10.1016/j.jacc.2004.02.036
`
`Prostanoid Therapy for
`Pulmonary Arterial Hypertension
`David B. Badesch, MD,* Vallerie V. McLaughlin, MD,† Marion Delcroix, MD,‡
`Carmine Dario Vizza, MD,§ Horst Olschewski, MD,㛳 Olivier Sitbon, MD,¶ Robyn J. Barst, MD#
`Denver, Colorado; Ann Arbor, Michigan; Leuven, Belgium; Rome, Italy; Giessen, Germany; Clamart, France;
`and New York, New York
`
`Prostanoids have played a prominent role in the treatment of pulmonary arterial hypertension
`(PAH). Several compounds and methods of administration have been studied: chronic
`intravenously infused epoprostenol, chronic subcutaneously infused treprostinil,
`inhaled
`iloprost, and oral beraprost. Chronic intravenous epoprostenol therapy has had a substantial
`impact on the clinical management of patients with severe PAH. It improves exercise
`capacity, hemodynamics, and survival in patients with idiopathic pulmonary arterial hyper-
`tension (IPAH). It also improves exercise capacity and hemodynamics in patients with PAH
`occurring in association with scleroderma. The complexity of epoprostenol therapy (chronic
`indwelling catheters, reconstitution of the drug, operation of the infusion pump, and others)
`has led to attempts to develop other prostanoids with simpler modes of delivery. Treprostinil,
`a stable prostacyclin analogue with a half-life of 3 h, has been developed for subcutaneous
`delivery. It has beneficial effects on exercise and hemodynamics, which depend somewhat on
`the dose achieved. This, in turn, is determined by the patient’s ability to tolerate the drug’s
`side effects, including pain and erythema at the infusion site. Inhaled iloprost therapy may
`provide selectivity of the hemodynamic effects to the lung vasculature, thus avoiding systemic
`side effects. In a randomized and controlled trial, iloprost resulted in improvement in a
`combined end point incorporating the New York Heart Association functional class, 6-min
`walk test, and deterioration or death. Beraprost is the first orally active prostacyclin analogue.
`In the first of two randomized controlled trials, beraprost increased exercise capacity in
`patients with IPAH, with no significant changes in subjects with associated conditions.
`Hemodynamics did not change significantly, and no difference in survival was detected
`between the two treatment groups. The second study showed that beraprost-treated patients
`had less disease progression at six months and confirmed the results of the previous trial.
`However, this improvement was no longer present at 9 or 12 months. In conclusion, though
`treatment with prostanoids is complicated by their generally short half-lives and complicated
`drug delivery systems, they continue to play an important role in the treatment of
`PAH.
`(J Am Coll Cardiol 2004;43:56S–61S) © 2004 by the American College of
`Cardiology Foundation
`
`A metabolite of arachidonic acid, prostacyclin is endog-
`enously produced by vascular endothelium. It is a potent
`vasodilator in both the pulmonary and systemic circulations,
`and has antiplatelet aggregatory activity. A relative defi-
`ciency of prostacyclin may contribute to the pathogenesis of
`pulmonary arterial hypertension (PAH). Clinical studies
`have explored the possibility that chronic therapy with
`exogenous prostacyclin analogues might be of long-term
`benefit in patients with moderately severe to severe PAH.
`To date, the following compounds and methods of admin-
`istration have been studied: chronic intravenously infused
`epoprostenol, chronic subcutaneously infused treprostinil,
`inhaled iloprost, and oral beraprost. This report summarizes
`the rationale for therapy utilizing each of these prostanoids,
`and it provides currently available evidence supporting the
`use of each in the treatment of PAH.
`
`From the *University of Colorado Health Sciences Center, Denver, Colorado;
`†University of Michigan, Ann Arbor, Michigan; ‡University Hospital Gasthuisberg,
`Leuven, Belgium; §University of Roma “La Sapienza,” Rome, Italy; 㛳University
`Hospital, Justus-Liebig-University, Giessen, Germany; ¶Hoˆpital Antoine Be´cle`re,
`Clamart, France; and #Columbia University College of Physicians and Surgeons,
`New York, New York.
`Manuscript received November 26, 2003; accepted February 3, 2004.
`
`EPOPROSTENOL
`
`Rationale. Christman et al. (1) reported a deficiency of
`prostacyclin and an excess of thromboxane in patients with
`PAH. Tuder et al. (2) showed decreased expression of
`prostacyclin synthase in lungs from patients with severe
`PAH. Exogenously administered prostanoid analogues
`might help to overcome the adverse effects of decreased
`endogenously produced prostacyclin. Epoprostenol has a
`very short half-life in the bloodstream, requiring constant
`intravenous (IV) administration.
`Treatment. In a multicenter, randomized, controlled trial
`in 81 patients with severe idiopathic pulmonary arterial
`hypertension (IPAH, formerly known as primary pulmo-
`nary hypertension or PPH), continuously intravenously
`infused epoprostenol plus conventional therapy (oral vaso-
`dilators, anticoagulation, others) was compared to conven-
`tional therapy alone. The epoprostenol-treated group dem-
`onstrated improved survival and exercise tolerance,
`increased cardiac output, and decreased pulmonary vascular
`resistance (3). The beneficial effects of epoprostenol therapy
`are often sustained. Barst et al. (4) reported long-term
`
`WATSON LABORATORIES, INC. , IPR2017-01621, Ex. 1033, p. 1 of 6
`
`

`

`JACC Vol. 43, No. 12 Suppl S
`June 16, 2004:56S–61S
`
`Badesch et al.
`Prostanoid Therapy for PAH
`
`57S
`
`Abbreviations and Acronyms
`IPAH ⫽ idiopathic pulmonary arterial hypertension
`⫽ intravenous
`IV
`⫽ nitric oxide
`NO
`NYHA ⫽ New York Heart Association
`PAH ⫽ pulmonary arterial hypertension
`PPH ⫽ primary pulmonary hypertension
`
`benefit in a small group of patients from centers involved in
`the earliest clinical use of epoprostenol. Shapiro et al. (5)
`and McLaughlin et al. (6) have described sustained benefit
`in larger numbers of patients. McLaughlin et al. (7) more
`recently reported experience with long-term epoprostenol
`therapy in 162 consecutive patients with IPAH followed for
`a mean of 36.3 months (median, 31 months) (Fig. 1). Data
`followed included functional class, exercise tolerance, and
`hemodynamics. Observed survival with epoprostenol ther-
`apy at one, two, and three years was 87.8%, 76.3%, and
`62.8%, respectively, and was significantly greater than the
`expected survival of 58.9%, 46.3%, and 35.4%, respectively,
`based on historical data. Baseline predictors of survival
`included exercise tolerance, functional class, right atrial
`pressure, and vasodilator response to adenosine. Predictors
`of survival after the first year of therapy included functional
`class and improvement in exercise tolerance, cardiac index,
`and mean pulmonary artery pressure.
`Sitbon et al. (8) evaluated the factors associated with
`long-term survival in patients with PPH/IPAH treated with
`continuous epoprostenol infusion. A total of 178 patients in
`New York Heart Association (NYHA) functional class III
`
`Figure 1. McLauglin et al. (7) reported 162 consecutive patients diagnosed
`with primary pulmonary hypertension and treated with epoprostenol who
`were followed for a mean of 36.3 months (median, 31 months). Observed
`survival (diamonds) with epoprostenol therapy at one, two, and three years
`was 87.8%, 76.3%, and 62.8%, respectively, and was significantly greater
`than the expected survival (squares) of 58.9%, 46.3%, and 35.4%, respec-
`tively, based on historical data. Baseline predictors of survival included
`exercise tolerance, functional class, right atrial pressure, and vasodilator
`response to adenosine. Predictors of survival after the first year of therapy
`included functional class and improvement in exercise tolerance, cardiac
`index, and mean pulmonary artery pressure. Reprinted with permission
`from Circulation (Lippincott Williams & Wilkins).
`
`or IV were treated with epoprostenol. Survival rates at one,
`two, three, and five years were 85%, 70%, 63%, and 55%,
`respectively. Baseline variables associated with a poor out-
`come were a history of right-sided heart failure, NYHA
`functional class IV, 6-min walk test ⱕ250 m (median
`value), right atrial pressure ⱖ12 mm Hg, and, paradoxically,
`mean pulmonary artery pressure ⱕ65 mm Hg. Multivariate
`analysis, including both baseline variables and those mea-
`sured after three months on epoprostenol, demonstrated
`that a history of right-sided heart failure, persistence of
`NYHA functional class III or IV at three months, and the
`absence of a decline in total pulmonary resistance of ⱖ30%,
`relative to baseline, were associated with poor survival. The
`investigators concluded that survival of patients with IPAH
`treated with epoprostenol depends both on severity of
`disease at baseline and the response to three months of
`therapy.
`A multicenter, randomized, and controlled study of
`chronic IV epoprostenol in patients with PAH occurring in
`association with the scleroderma spectrum of disease
`showed improvement in exercise capacity and hemodynam-
`ics at 12 weeks as compared to the control group (9). Trends
`were seen toward greater improvement in severity of the
`Raynaud phenomenon and fewer new digital ulcers in the
`epoprostenol group. A survival difference between groups
`was not seen in this population over the period of study, but
`the study was not adequately powered to detect such a
`difference.
`Epoprostenol therapy requires continuous IV infusion.
`The drug has a very short half-life (⬍6 min), is unstable at
`acidic PpH, and cannot be taken orally. It is unstable at
`room temperature, and is generally kept cold prior to
`infusion. Patients are usually begun on a low dosage of
`epoprostenol (1 to 2 ng/kg/min), and gradually titrated
`upward in increments of 1 to 2 ng/kg/min, based upon side
`effects and tolerance. Many patients seem to reach a
`“plateau” dose and may not require continued up-titration
`from that point. Whereas this dose may be between 20 and
`40 ng/kg/min for many patients, the dose range is wide,
`with considerable interindividual variability.
`Common side effects of epoprostenol therapy include
`flushing, headache, jaw pain with the first bite of food,
`(which is usually tolerable), diarrhea, nausea, a blotchy
`erythematous rash, and musculoskeletal aches and pain
`(predominantly involving the legs and feet). These side
`effects tend to be dose dependent, and they often respond to
`a cautious reduction in dose. Abrupt or inadvertent inter-
`ruption of the epoprostenol infusion should be avoided, as
`this may lead to a rebound worsening of pulmonary hyper-
`tension with symptomatic deterioration and perhaps even
`death. Complications of chronic IV therapy with epopro-
`stenol include line-related infections (which range from exit
`site reactions, to tunnel infections and cellulitis, to bactere-
`mia or sepsis), catheter-associated venous thrombosis,
`thrombocytopenia, and ascites (although this may also be a
`manifestation of severe disease).
`
`WATSON LABORATORIES, INC. , IPR2017-01621, Ex. 1033, p. 2 of 6
`
`

`

`58S
`
`Badesch etal.
`Prostanoid Therapy for PAH
`
`JACC Vol. 43, No. 12 Suppl S
`June 16, 2004:56S–61S
`
`Chronic IV epoprostenol therapy has had a substantial
`impact on the treatment of patients with moderately severe
`to severe PAH. It has been best studied in patients with
`IPAH and PAH occurring in association with the sclero-
`derma spectrum of disease. Because of the complexity of
`epoprostenol therapy (chronic indwelling catheters, recon-
`stitution of the drug, operation of the infusion pump, and so
`forth), and the relative rarity of severe PAH, strong con-
`sideration should be given to referral to centers of excel-
`lence.
`
`TREPROSTINIL
`
`Rationale. Although epoprostenol is effective therapy for
`PAH, the nature of the delivery system has a number of
`potential complications, which range in severity from local
`exit site infections easily treated with oral antibiotics to
`life-threatening sepsis. Because of the short half-life of
`epoprostenol, interruptions in therapy related to catheter
`displacement or pump malfunction may be life-threatening.
`Rare adverse events associated with the delivery system
`include pneumothorax, deep venous thrombosis, and para-
`doxical embolus. The efficacy of epoprostenol, coupled with
`the limitations of the delivery system, has led to the
`development of prostacyclin analogues with alternative
`routes of delivery. Treprostinil, a stable prostacyclin ana-
`logue with a half-life of 3 h, has been developed for
`subcutaneous delivery.
`Treatment. In 14 patients with IPAH, treatment acutely
`with IV epoprostenol and IV treprostinil had similar hemo-
`dynamic effects (10). To test the alternative subcutaneous
`delivery method, the effects of IV treprostinil and subcuta-
`neous treprostinil were compared in 25 patients with IPAH.
`Acute hemodynamic effects were similar. An eight-week,
`placebo-controlled, 2:1 randomized trial of subcutaneous
`treprostinil was subsequently performed in 26 patients with
`IPAH. An improvement of 37 ⫾ 17 m in the 6-min walk
`distance occurred in patients receiving the active therapy
`(from 373 m to 411 m), compared to a 6 ⫾ 28-m reduction
`in those receiving placebo (379 m vs. 384 m), a nonstatis-
`tically significant trend. There was a favorable, but not
`statistically significant, trend in hemodynamics, with a 20%
`reduction in pulmonary vascular resistance index over the
`eight-week period in the group receiving active treprostinil.
`Adverse events including headache, diarrhea, flushing, jaw
`pain, and foot pain were common in the treprostinil group,
`similar to what had been previously reported with epopro-
`stenol. Pain (occasionally severe), erythema, and induration
`at the site of the subcutaneous infusion occurred frequently.
`A large international, placebo-controlled, randomized
`study was conducted assessing the efficacy of chronic sub-
`cutaneously delivered treprostinil in patients with IPAH or
`PAH occurring in association with collagen vascular disease
`or congenital systemic-to-pulmonary shunts (11). Four
`hundred-seventy patients enrolled between November 1998
`and October 1999 in 24 centers in North America and 16
`
`centers in Europe, Australia, and Israel were randomly
`assigned to receive either continuous subcutaneously infused
`treprostinil plus conventional therapy or continuous infu-
`sion of placebo plus conventional therapy. Owing to the
`infusion-site pain and reaction that occurred in the proof-
`of-concept trial, the dosing strategy called for lower doses at
`initiation, with a maximal allowable dose at the end of 12
`weeks of 22.5 ng/kg/min. The primary end point was
`exercise capacity as measured by the 6-min walk distance,
`which improved in the treprostinil group and was un-
`changed with placebo. The median difference between
`treatment groups was 16 m (p ⫽ 0.006), and the effect on
`exercise tolerance appeared to be dose related. The patients
`in the lowest two dosing quartiles experienced little im-
`provement in the 6-min walk distance, whereas patients in
`the highest quartile in terms of dose (⬎13.8 ng/kg/min)
`demonstrated an improvement of 36 m. Treprostinil ther-
`apy was also associated with a significant improvement in
`mean right atrial pressure, mean pulmonary artery pressure,
`cardiac index, pulmonary vascular resistance, and mixed
`venous oxygen saturation. Common side effects included
`headache, diarrhea, nausea, rash, jaw pain, and infusion-site
`pain. Of the patients, 85% complained of pain at the
`infusion site, and 83% had erythema or induration at the
`infusion site. Although statistically significant,
`improve-
`ment in the 6-min walk distance was relatively modest. The
`reasons for this may be multifactorial. Entry criteria for the
`treprostinil trial were much more broad than for either of
`the epoprostenol trials.
`
`INHALED ILOPROST
`
`Rationale. Inhaled therapy for pulmonary hypertension
`may provide selectivity of the hemodynamic effects to the
`lung vasculature, thus avoiding systemic side effects. Pul-
`monary selective vasodilation has been described for inhaled
`nitric oxide (NO), but this agent has several drawbacks.
`Most importantly, there are no data demonstrating im-
`proved survival during therapy with inhaled NO, and this
`agent possesses less vasodilative potency than do prostanoids
`in IPAH patients (12,13). In contrast, prostacyclin (epopro-
`stenol) has been shown to improve survival, exercise capac-
`ity, and hemodynamics in patients with severe IPAH
`(3,4,6), and the continuous IV infusion of this drug has been
`approved for therapy in certain groups of patients in the
`U.S. and several European countries.
`Treatment. Iloprost is a prostacyclin analogue and has the
`same biologic profile as the natural substance with respect to
`prostaglandin receptor binding and cellular effects (14). Its
`effects and its side effects are similar to those seen during
`epoprostenol infusion (15). In contrast, the chemical stabil-
`ity of iloprost is considerably better. Where epoprostenol
`has to be freshly dissolved, continuously cooled, and pro-
`tected from light to provide full activity, iloprost is stable at
`room temperature, at pH 7.4, and normal light. Epopro-
`stenol has a half-life of ⬍6 min, whereas iloprost has a
`
`WATSON LABORATORIES, INC. , IPR2017-01621, Ex. 1033, p. 3 of 6
`
`

`

`JACC Vol. 43, No. 12 Suppl S
`June 16, 2004:56S–61S
`
`Badesch et al.
`Prostanoid Therapy for PAH
`
`59S
`
`serum half-life of 20 to 25 min (16). Iloprost has been
`approved for the treatment of PAH in New Zealand, and
`was recently approved for the treatment of IPAH in
`Europe. Although epoprostenol is available in the United
`States and Europe, iloprost is not approved in the U.S.
`Owing to the fact that the intra-acinar pulmonary arteries
`are tightly surrounded by alveolar surfaces, it is possible to
`vasodilate these vessels by means of alveolar deposition of a
`prostanoid. For long-term therapy, repetitive inhalations of
`iloprost are administered six to nine times daily. Each
`inhalation requires 10 to 15 min. With alternative devices it
`is possible to reduce the inhalation time to about 4 min (17)
`and to avoid any noise by use of ultrasound energy for
`nebulization.
`In patients with severe PAH, inhalation of aerosolized
`iloprost resulted in a substantial decrease in pulmonary
`artery pressure and resistance, concomitant with an increase
`in cardiac output, in the absence of significant systemic
`artery pressure drop and ventilation–perfusion mismatch
`(18,19). In severe lung fibrosis, an increase of the pulmonary
`shunt blood flow may limit the use of IV epoprostenol (19),
`whereas inhaled iloprost can safely be administered. In
`uncontrolled studies, inhaled iloprost was effective in de-
`compensated right heart failure (20) and showed favorable
`long-term hemodynamic improvement (21).
`A large randomized, double-blind, placebo-controlled
`European multicenter study with inhaled iloprost (Aerosol-
`ized Iloprost Randomized, AIR [22]) involved a total of 203
`patients in NYHA functional class III or class IV with
`IPAH or PAH occurring in association with appetite-
`suppressant use, collagen vascular disease, or nonoperable
`thromboembolic disease. In both the iloprost and placebo
`groups, approximately 50% suffered from IPAH, and about
`60% were in NYHA functional class III and 40% in NYHA
`functional class IV. The primary end point of the study,
`defined as an improvement in NYHA functional class
`combined with at least 10% improvement in the 6-min
`walking test, and no deterioration or death (combined
`clinical end point), was reached by 3.4 ⫻ more patients in
`the iloprost versus the placebo group (16.8% vs. 4.9%; p ⫽
`0.007). This effect was achieved with a mean inhaled dose of
`0.37 ng/kg/min. In the 6-min walk test, the treatment effect
`was 36.4 m in favor of iloprost (p ⬍ 0.01). Hemodynamics
`significantly deteriorated in the placebo group, whereas in
`the iloprost group preinhalation values were unchanged
`compared to baseline, and postinhalation values were sig-
`nificantly improved. Importantly, the number of patients
`remaining on study medication, a measure corresponding to
`event-free survival, was significantly higher in the iloprost
`than in the placebo group. Over three months of therapy,
`there was no indication of tachyphylaxis. In the iloprost
`group, one patient (1.0%) died during the double-blind
`study period versus four patients (4.0%) in the placebo
`group. Overall, the therapy was well tolerated. Cough
`occurred more frequently in the iloprost compared to the
`placebo group (38.6% vs. 25.5%) as well as headache (29.7%
`
`vs. 19.6%) and flushing (26.7% vs. 8.8%). These adverse
`events were mild and mostly transient. Syncope occurring in
`the iloprost group was more often rated as serious than in
`the placebo group, but was not commonly associated with
`clinical deterioration.
`An open-label multicenter study of the two-year effects of
`inhaled iloprost in pulmonary hypertension with an initial
`three-month controlled randomized phase was performed
`in Germany. Inhaled iloprost treatment was administered
`for up to two years to 63 patients (40 with IPAH and 23
`with PAH occurring in association with underlying disor-
`ders). The median daily aerosolized dose was 100 ␮g (total
`inhaled dose ⬇24 ␮g) divided into six inhalations. At study
`entry, 66.6% of patients were in NYHA functional class III
`or class IV, and 33.4% in functional class II. During the
`two-year study period, five IPAH patients were switched to
`alternative therapy (mostly IV iloprost) but remained in the
`study, and 13 patients discontinued the study prematurely (7
`with IPAH; 6 with other forms of PAH). After two years,
`37 patients received inhaled iloprost treatment (25 with
`IPAH; 12 with other forms of PAH). During the course of
`the study, eight patients died: three IPAH and five with
`other forms of PAH. Two of these patients died before
`receiving inhaled iloprost treatment. The survival rate ac-
`cording to Kaplan-Meier analysis was 0.850 for all patients
`and 0.914 (95% confidence interval: 0.81; ⬇1) for IPAH
`patients for the two-year study period, including the ran-
`domized phase. For IPAH patients, the predicted survival
`rate according to D’Alonzo et al. (23) was 0.631, which
`corresponds to ⬇14.8 deaths. In contrast, only three IPAH
`patients died. This suggests that survival on inhaled iloprost
`treatment is substantially higher than expected.
`In addition to treatment of IPAH, the pulmonary selec-
`tivity of inhaled iloprost provides the opportunity to apply
`prostanoids to patients who are prone to decrease in
`systemic arterial pressure, as in portopulmonary hyperten-
`sion, and in emergency situations. The intrapulmonary
`selectivity also allows prostanoid application to patients who
`are prone to intrapulmonary right to left shunt, like hepa-
`topulmonary syndrome and lung fibrosis (19).
`Inhaled iloprost may provide an alternative to the use of
`IV epoprostenol. When the clinical effects of
`inhaled
`iloprost and IV epoprostenol are compared, inhaled iloprost
`has some advantages but also certain drawbacks. Most
`importantly, the inhalation provides potent pulmonary va-
`sodilation with little systemic side effects and no risk of
`catheter complications. Additionally, it allows therapy in
`patients with pre-existent ventilation-perfusion mismatch
`and in those who are prone to develop such a mismatch
`during systemic prostanoid application. The most important
`drawback is the fact that the hemodynamic effect of inhaled
`iloprost plateaus within 30 to 90 min, and that six to nine
`inhalations per day are needed to achieve satisfactory clinical
`results. Inhaled iloprost is currently approved in Europe for
`functional class III IPAH. Long-term survival data are
`needed.
`
`WATSON LABORATORIES, INC. , IPR2017-01621, Ex. 1033, p. 4 of 6
`
`

`

`60S
`
`Badesch etal.
`Prostanoid Therapy for PAH
`
`BERAPROST
`
`Rationale. Beraprost is the first orally active prostacyclin
`analogue (24). It is rapidly absorbed during fasting, peak
`concentration is reached after 30 min, and the elimination
`half-life is 35 to 40 min after oral administration (25). In the
`monocrotaline-induced pulmonary hypertension model,
`beraprost has been shown to have a protective effect on the
`development of pulmonary hypertension lesions (26). High
`doses of beraprost appear to have inotropic and chrono-
`tropic effects in the isolated guinea pig myocardium (27).
`Beraprost has also been studied in peripheral vascular
`diseases such as intermittent claudication (28), Raynaud
`phenomenon, and digital necrosis in systemic sclerosis (29),
`with variable results.
`Treatment. Beraprost has been used to treat PAH since
`1995 in Japan; several small open uncontrolled studies have
`reported beneficial hemodynamic effects with beraprost in
`patients with IPAH (24,30). Functional class also improved
`in the majority of the patients after a mean of two months,
`and pulmonary vascular resistance was reduced by 26% (24,
`30). Nagaya et al. (31), in a retrospective open uncontrolled
`study, reported improved survival
`in 24 IPAH patients
`treated with beraprost compared to a similar group of 34
`patients on conventional therapy. The three-year survival
`rate was 76% in the beraprost group and 44% in the
`conventional-therapy group.
`Two randomized, double-blind, placebo-controlled trials
`of beraprost in PAH have been performed. The first was a
`12-week double-blind, randomized, placebo-controlled trial
`performed in 130 NYHA functional class II and class III
`patients with PAH of various etiologies (IPAH, and PAH
`associated with collagen vascular diseases, congenital
`systemic-to-pulmonary shunts, portal hypertension, or hu-
`man immunodeficiency virus infection) (32). At a median
`dose of 80 ␮g administered orally four times daily, beraprost
`increased exercise capacity assessed by the 6-min walk test:
`treatment effect was 25 m in the overall population, and
`45 m in the IPAH patients, with no significant changes in
`the exercise capacity of subjects with the associated condi-
`tions. Hemodynamics had no statistically significant
`changes, and no difference in survival was detected between
`the two treatment groups. Side effects related to systemic
`vasodilation were frequent, mainly in the initial titration
`period.
`A second trial evaluated the effects of beraprost therapy
`for PAH in 116 NYHA functional class II and class III
`patients. This study was 12 months in duration, double-
`blind, randomized, and placebo-controlled (33). It showed
`that the beraprost-treated patients had less disease progres-
`sion at six months and confirmed the results of the previous
`trial (32): improved 6-min walk distance at three months
`(⫹22 m from baseline) and six months (⫹31 m), as
`compared to placebo. However, this improvement was no
`longer present at 9 or 12 months (33). No significant
`changes occurred in hemodynamics or survival at month 12
`
`JACC Vol. 43, No. 12 Suppl S
`June 16, 2004:56S–61S
`
`versus baseline. It is possible that the beneficial effects of
`beraprost may attenuate with time. Beraprost is approved
`for treatment of PAH in Japan, and is currently under
`evaluation by the European Agency for the Evaluation of
`Medicinal Products (EMEA).
`
`SUMMARY
`
`Chronic IV epoprostenol therapy has had a substantial
`impact on the clinical management of patients with severe
`PAH. The complexity of epoprostenol therapy has led to
`attempts to develop other prostanoids with simpler modes
`of delivery. Subcutaneous treprostinil, inhaled iloprost, and
`oral beraprost have all been studied, and they have various
`relative advantages and disadvantages. In conclusion, al-
`though treatment with prostanoids is complicated by their
`generally short half-lives and complicated drug-delivery
`systems, they continue to play an important role in the
`treatment of PAH. Together with the other major classes of
`therapeutic agents currently utilized or under investigation
`for the treatment of PAH (including anticoagulants, sup-
`plemental oxygen, calcium channel antagonists, endothelin
`receptor antagonists, phosphodiesterase inhibitors, and NO
`or NO donors), prostanoids remain an important therapeu-
`tic option.
`
`Reprint requests and correspondence: Dr. David B. Badesch,
`Divisions of Pulmonary Sciences and Critical Care Medicine, and
`Cardiology, University of Colorado Health Sciences Center, Box
`C-272, 4200 East Ninth Avenue, Denver, Colorado 80262.
`E-mail: David.Badesch@UCHSC.edu.
`
`REFERENCES
`
`1. Christman BW, McPherson CD, Newman JH, et al. An imbalance
`between the excretion of thromboxane and prostacyclin metabolites in
`pulmonary hypertension. N Engl J Med 1992;327:70–5.
`2. Tuder RM, Cool CD, Geraci MW, et al. Prostacyclin synthase
`expression is decreased in lungs from patients with severe pulmonary
`hypertension. Am J Respir Crit Care Med 1999;159:1925–32.
`3. 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 Hyperten-
`sion Study Group. N Engl J Med 1996;334:296–302.
`4. Barst RJ, Rubin LJ, McGoon MD, Coldwell EJ, Long WA, Levy PS.
`Survival in primary pulmonary hypertension with long-term continu-
`ous intravenous prostacyclin. Ann Intern Med 1994;121:409–15.
`5. Shapiro SM, Oudiz RJ, Cao T, et al. Primary pulmonary hypertension:
`improved long-term effects and survival with continuous intravenous
`epoprostenol infusion. J Am Coll Cardiol 1997;30:343–9.
`6. McLaughlin VV, Genthner DE, Panella MM, Rich S. Reduction in
`pulmonary vascular resistance with long-term epoprostenol (prostacy-
`clin) therapy in primary pulmonary hypertension. N Engl J Med
`1998;338:273–7.
`7. McLaughlin VV, Shillington A, Rich S. Survival in primary pulmo-
`nary hypertension: the impact of epoprostenol therapy. Circulation
`2002;106:1477–82.
`8. Sitbon O, Humbert M, Nunes H, et al. Long-term intravenous
`epoprostenol infusion in primary pulmonary hypertension: prognostic
`factors and survival. J Am Coll Cardiol 2002;40:780–8.
`9. Badesch DB, Tapson VF, McGoon MD, et al. Continuous intrave-
`nous epoprostenol for pulmonary hypertension due to the scleroderma
`spectrum of disease. A randomized, controlled trial. Ann Intern Med
`2000;132:425–34.
`
`WATSON LABORATORIES, INC. , IPR2017-01621, Ex. 1033, p. 5 of 6
`
`

`

`JACC Vol. 43, No. 12 Suppl S
`June 16, 2004:56S–61S
`
`Badesch et al.
`Prostanoid Therapy for PAH
`
`61S
`
`10. McLaughlin VV, Gaine SP, Barst RJ, et al. Efficacy and safety of
`treprostinil: an epoprostenol analog for primary pulmonary hyperten-
`sion. J Cardiovasc Pharmacol 2003;41:293–9.
`11. 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;165:800–4.
`12. Pepke-Zaba J, Higenbottam TW, Dinh-Xuan AT, Stone D, Wall-
`work J. Inhaled nitric oxide as a cause of selective pulmonary
`vasodilatation in pulmonary hypertension. Lancet 1991;338:1173–4.
`13. Hoeper MM, Olschewski H, Ghotrani HA, et al. A comparison of the
`acute hemodynamic effects of inhaled nitric oxide and aerosolized
`iloprost in primary pulmonary hypertension. German PPH Study
`Group. J Am Coll Cardiol 2000;35:176–82.
`14. Olschewski H, Olschewski A, Rose F, et al. Physiologic basis for the
`treatment of pulmonary hypertension. J Lab Clin Med 2001;138:287–97.
`15. Higenbottam T, Bult AY, McMahon A, Westerbeck R, Sharples L.
`Long-term intravenous prostaglandin (epoprostenol or iloprost) for
`treatment of severe pulmonary hypertension. Heart 1998;80:151–5.
`16. Krause W, Krais T. Pharmacokinetics and pharmacodynamics of the
`prostacyclin analogue iloprost in man. Eur J Clin Pharmacol 1986;30:
`61–8.
`17. Gessler T, Schmehl T, Hoeper MM, et al. Ultrasonic versus jet
`nebulization of iloprost in severe pulmonary hypertension. Eur Respir
`J 2001;17:14–9.
`18. Olschewski H, Walmrath D, Schermully R, Ghofrani A, Glimminger
`F, Seeger W. Aerosolized prostacyclin and iloprost in severe pulmo-
`nary hypertension. Ann Intern Med 1996;124:820–4.
`19. Olschewski H, Ghofrani HA, Walmrath D, et al. Inhaled prostacyclin
`and iloprost in severe pulmonary hypertension secondary to lung
`fibrosis. Am J Respir Crit Care Med 1999;160:600–7.
`20. Olschewski H, Ghofrani HA, Schmehl T, et al. Inhaled iloprost to
`treat severe pulmonary hypertension. An uncontrolled trial. German
`PPH Study Group. Ann Intern Med 2000;132:435–43.
`21. Hoeper MM, Schwarze M, Ehlerding S, et al. Long-term treatment
`of primary pulmonary hypertension with aerosolized iloprost, a pros-
`tacyclin analogue. N Engl J Med 2000;342:1866–70.
`22. Olschewski H, Simonneau G, Galie N, et al. Inhaled iloprost for
`severe pulmonary hypertension. N Engl J Med 2002;347:322–9.

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