throbber
Review
`
`For reprint orders, please contact reprints@future-drugs.com
`
`Medical therapeutics for
`pulmonary arterial hypertension:
`from basic science and clinical trial
`design to evidence-based medicine
`Roxana Sulica and Michael Poon†
`Pulmonary arterial hypertension is a severe disease with poor prognosis, caused by
`obliteration of the pulmonary vasculature as a result of pulmonary-vascular remodeling,
`active vasoconstriction and in situ thrombosis. Left untreated, pulmonary arterial
`hypertension results in right-ventricular failure and death. There has been dramatic
`progress in the treatment of pulmonary arterial hypertension during recent years. A
`remarkable number of randomized-controlled trials with agents known to target specific
`abnormalities present in pulmonary arterial hypertension have been completed. Most
`commonly, therapeutic efficacy was judged by the ability of the drug under study to
`improve exercise capacity and to decrease the rate of severe complications. Completed
`clinical trials have mainly evaluated patients with relatively advanced disease. Despite
`these advances, responses to therapy in pulmonary arterial hypertension are not uniformly
`favorable and frequently incomplete. In addition, the methods of delivery and the adverse
`effect profile of the currently available pulmonary arterial hypertension-specific drugs
`create further management difficulties. Based on newly identified pathobiologic
`abnormalities in the pulmonary vasculature, future studies are likely to focus on the
`discovery of new therapeutic targets. Clinical trial design will continue to evolve in an
`attempt to enable inclusion of patients with less advanced disease and evaluation of
`treatment combinations or comparisons of the currently approved drugs.
`
`Expert Rev. Cardiovasc. Ther. 3(2), 347–360 (2005)
`
`Definition & classification
`(PAH)
`Pulmonary
`arterial hypertension
`encompasses a heterogeneous group of dis-
`orders characterized by increased pulmonary
`artery pressure (PAP) and pulmonary vascular
`resistance (PVR). PAH classification includes
`idiopathic PAH (IPAH; formerly primary pul-
`monary hypertension), familial PAH, PAH
`associated with collagen vascular disease, con-
`genital systemic-to-pulmonary shunts, portal
`hypertension, HIV infection, drug and toxins,
`and other conditions (thyroid disorders, gly-
`cogen storage disease, Gaucher disease, here-
`ditary hemorrhagic telangiectasia, hemoglob-
`inopathies, myeloproliferative disorders and
`splenectomy), PAH associated with significant
`venous or capillary involvement and persistent
`pulmonary hypertension of the newborn [1].
`
`Without intervention, PAH has a progressive
`course and poor prognosis. Estimated median
`survival of untreated IPAH patients
`is
`2.8 years [2]. Survival is primarily determined
`by the level of the right-ventricular dysfunc-
`tion and the most common cause of death is
`right-ventricular failure. Appropriate therapy
`may alter the natural course of the disease,
`but does not offer a definitive cure. To a cer-
`tain degree, all forms of PAH share patho-
`logic characteristics, clinical presentation,
`diagnostic modalities
`and
`therapeutic
`options. In PAH, the small pulmonary arter-
`ies are occluded by a combination of active
`in situ
`vasoconstriction,
`thrombosis and,
`most importantly, vascular proliferation and
`remodeling. PAH treatment is directed at all
`of these processes (FIGURE 1).
`
`CONTENTS
`Definition & classification
`Conventional treatment
`Specific pulmonary arterial
`hypertension treatment
`Expert opinion
`Five-year view
`Key issues
`References
`Affiliations
`
`†Author for correspondence
`Mount Sinai School of Medicine,
`Director of Cardiology, Cabrini
`Medical Center, New York,
`NY 10003, USA
`Tel.: +1 212 995 6865
`Fax: +1 212 979 3474
`mpoon@cabrininy.org
`
`KEYWORDS:
`ambrisentan, bosentan,
`epoprostenol, exercise capacity,
`hemodynamics, iloprost,
`pulmonary arterial hypertension,
`sildenafil, sitaxsentan, treprostinil
`
`www.future-drugs.com
`
`10.1586/14779072.3.2.347
`
`© 2005 Future Drugs Ltd
`
`ISSN 1477-9072
`
`347
`
`Liquidia's Exhibit 1104
`Page 1
`
`

`

`Sulica & Poon
`
`In situ thrombosis
`
`Vascular endothelium
`(cid:127) Endothelial dysfunction
`(cid:127) Endothelial proliferation
`
`Prostacyclin replacement:
`epoprostenol, treprostinil,
`iloprost and beraprost
`
`Anticoagulation:
`warfarin
`
`Endothelin receptor
`antagonists: bosentan,
`sitaxsentan and
`ambrisentan
`
`Prostacyclin ↓
`
`Endothelin-1 ↑
`
`Nitric oxide ↓
`
`Nitric oxide replacement
`enhancers or
`donors: sildenafil
`
`Calcium channel blockers
`
`output. This response is present in less
`than 10% of PAH patients, mainly IPAH
`and PAH associated with anorexigen
`use [5]. Initiation of oral calcium channel-
`blocker therapy is restricted to patients
`who manifest acute vasoreactivity and
`requires frequent monitoring to document
`long-term clinical efficacy. Indiscriminate
`use of oral calcium channel blockers in
`patients with PAH is strongly discouraged
`to avoid precipitation of hypotension
`and heart failure with potentially fatal
`consequences.
`
`Oral anticoagulation
`Although no randomized, double-blind,
`placebo-controlled
`trials
`of
`anti-
`coagulation were performed in patients
`with PAH, two retrospective studies and
`one prospective report in patients with
`IPAH and PAH associated with anorexi-
`gen drug use have suggested improved
`outcome in patients receiving long-term
`warfarin therapy [3,6,7]. In the absence of
`contraindications, IPAH patients should
`receive long-term anticoagulant therapy,
`with a goal of keeping the international
`normalized ratio between 1.8 and 2.5.
`Some experts extend this recommen-
`dation to other forms of PAH, although
`this is controversial, particularly in cases
`with an increased risk of bleeding, such as
`PAH patients with scleroderma (erosive
`esophagitis), portopulmonary hyper-
`tension (gastrointestinal bleed) or congenital
`heart disease (hemoptysis).
`
`Vascular smooth muscle cells
`(cid:127) Vasoconstriction
`(cid:127) Proliferation
`(cid:127) Dysfunction of voltage-dependent K +
` channels and Ca++ channels
`
`Figure 1. Treatment of pulmonary arterial hypertension.
`
`Conventional treatment
`Oral calcium channel blockers
`Active vasoconstriction is detectable at right-heart catheterization
`during acute vasoreactivity testing. Patients who manifest acute
`vasoreactivity may have a good long-term response to oral cal-
`cium channel-blocker therapy [3]. Therefore, testing for acute
`vasoreactivity is recommended during initial evaluation in all
`patients with PAH [4]. During right heart catheterization,
`patients are administered short acting vasodilators such as
`inhaled nitric oxide (NO), intravenous epoprostenol or intra-
`venous adenosine. Acute vasoreactivity is present if, after vaso-
`dilator challenge, the mean PAP decreases by at least 10 mmHg
`and to less than 40 mmHg, with preserved or increased cardiac
`
`Specific pulmonary arterial
`hypertension treatment
`Basic pathogenesis of pulmonary
`arterial hypertension
`Vascular wall remodeling and proliferation
`are the hallmarks of pulmonary arterial obstruction in PAH [8].
`Central to the pathogenesis of vascular remodeling is the dys-
`function of the pulmonary vascular endothelium with an
`imbalance between endothelial mediators with opposing
`actions on the pulmonary vasculature [9–13]. There is an over-
`expression and/or activation of vasoconstricting, mitogenic and
`prothrombotic factors (endothelin [ET]-1, thromboxane and
`serotonin), while prostacyclin, NO and heparin-like sub-
`stances, which promote vasodilatation and have antiprolifera-
`tive and antithrombotic properties, are decreased. Specific PAH
`treatment entails either replacing deficient vasodilator factors or
`inhibiting mediators that induce vasoconstriction and vascular
`proliferation (FIGURE 1).
`
`348
`
`Expert Rev. Cardiovasc. Ther. 3(2), (2005)
`
`Liquidia's Exhibit 1104
`Page 2
`
`

`

`Pulmonary arterial hypertension
`
`Specific pulmonary arterial hypertension therapeutic agents
`Recent developments in the management of PAH focus on the
`introduction of new drugs with different intracellular mecha-
`nisms of action and the use of alternative routes of administra-
`tion. In the USA, the agents approved by the US Food and
`Drug Administration (FDA) for the specific treatment of PAH
`are continuous intravenous epoprostenol (prostacyclin and
`subcutaneous
`treprostinil
`I2), continuous
`prostaglandin
`(prostacyclin analog) and oral bosentan (ET receptor antago-
`nist). Agents approved by the European Agency for the Evalua-
`tion of Medicinal Products are intravenous epoprostenol, oral
`bosentan and inhaled iloprost (prostacyclin analog). Other oral
`drugs recently investigated or currently studied in clinical trials
`include beraprost, sildenafil, sitaxsentan and ambrisentan.
`
`Expert opinion
`Evidence-based management
`During the past 5 years, more than 2500 patients with PAH
`have been studied in a randomized, controlled fashion in more
`than ten completed major clinical trials. At least nine more
`randomized, controlled trials are planned or ongoing. TABLES 1–4
`summarize the findings of the major clinical trials
`in
`PAH [14–36]. Sildenafil, a phosphodiesterase-5 inhibitor with
`pulmonary vasodilatory action, has also been evaluated in a
`3-month double blind, placebo-controlled, multicenter, rand-
`omized trial in patients with PAH (63% IPAH, 30% PAH
`associated with connective tissue disease and 6% PAH associ-
`ated with surgically corrected congenital heart disease) [37]. This
`trial included 278 patients with PAH in World Health Organi-
`zation (WHO) functional Classes II to IV (39% Class II, 58%
`Class III and 3% Class IV) and evaluated three sildenafil doses
`(20, 40 and 80 mg three-times daily). The primary end point
`was the change from baseline in the 6-min walk test (6MWT)
`distance. There was a significant improvement in the exercise
`capacity as measured by the 6MWT, in hemodynamics and in
`WHO class for all three doses studied [37].
`
`Multiple new findings in PAH pathobiology, genetics,
`diagnosis and therapy were assessed during the Third World
`Symposium on Pulmonary Arterial Hypertension (Venice,
`2003) and proceedings of this meeting were published in a sup-
`plement
`to
`the Journal of
`the American College of
`Cardiology [38]. In addition, the American College of Chest
`Physicians has recently published evidence-based guidelines for
`PAH diagnosis and management [39]. Therapeutic choices in
`PAH depend on the etiopathogenic particularities of the disease
`and the severity of the functional impairment (FIGURE 2). A new
`treatment algorithm has been devised based on data from pub-
`lished randomized, controlled trials (FIGURE 3). This algorithm is
`mainly restricted to patients in WHO functional Classes III
`and IV and patients with IPAH and PAH due to the sclero-
`derma spectrum of disease, who represented the majority of
`studied patients. Extrapolation of current recommendations to
`other PAH patient populations requires caution.
`In the face of substantial achievements in the therapeutic
`armamentarium, PAH remains a disease with particularly
`poor prognosis, even among treated patients. Untreated
`IPAH patients from the National Institutes of Health regis-
`try had 1-, 3- and 5-year survival rates of 68, 48 and 34%,
`respectively [2]. Intravenous epoprostenol is the only PAH-
`specific therapy that has been shown to improve survival in a
`3-month, randomized, controlled trial in IPAH [15], while
`newer drugs have been shown to reduce clinical deterio-
`ration during the same time interval [29–31]. IPAH is more
`responsive to treatment compared with other forms of PAH
`[21,31,34] and intravenous epoprostenol is still considered the
`most efficacious therapy available. Retrospective studies of
`IPAH patients treated with
`intravenous epoprostenol,
`demonstrated improved long-term outcome with 1-, 3- and
`5-year survival rates of 85, 63 and 55%, respectively [19,20].
`Response to available therapy is not universally favorable
`and there is no cure for PAH. Therefore, there is a continual
`need for discovery of novel therapeutic strategies.
`
`Table 1. Major randomized controlled clinical trials with intravenous epoprostenol in patients with PAH.
`Control* Primary
`Treatment effect
`Trial
`n PAH WHO
`Route Duration
`Dose of
`end point
`class
`(months)
`active drug
`(%)
`achieved
`(ng/kg/min)
`
`II
`
`III
`
`IV
`
`Rubin
`(1990)
`
`Barst
`(1996)
`
`Badesch
`(2000)
`
`24 IPAH 9 65 26 Iv.
`
`81 IPAH
`
`75 25 Iv.
`
`111 SPAH 5 78 17 Iv.
`
`2
`
`3
`
`3
`
`N/A
`
`9.2
`
`11.2
`
`Conv.
`therapy
`
`Conv.
`therapy
`
`Conv.
`therapy
`
`H/dyn
`
`H/dyn
`
`6MWT
`
`6MWT
`(m)
`+45
`(mean)
`
`+47
`(mean)
`
`+94
`(median)
`
`H/dyn WHO
`class
`Better N/A
`
`Symptoms QOL
`
`Survival
`
`Better
`
`N/A
`
`No
`change
`
`Better Better Better
`
`Better Better
`
`Better Better Better
`
`N/A
`
`No
`Change
`
`Ref.
`
`[14]
`
`[15]
`
`[16]
`
`*None of these studies were placebo-controlled for ethical reasons; patients in the active group received intravenous epoprostenol in addition to conventional therapy, while the control
`group was represented by patients receiving conventional therapy alone.
`6MWT: 6-min walk test; Conv.: Conventional; H/dyn: Hemodynamics; IPAH: Idiopathic pulmonary arterial hypertension; Iv.: Intravenous; N/A: Not applicable; PAH: Pulmonary arterial
`hypertension; QOL: Quality of life; SPAH: Scleroderma pulmonary arterial hypertension; WHO: World Health Organization.
`
`www.future-drugs.com
`
`349
`
`Liquidia's Exhibit 1104
`Page 3
`
`

`

`Sulica & Poon
`
`Table 2. Other clinical trials with intravenous epoprostenol in patients with PAH.
`Trial
`Findings
`Comments
`Barst 1994
`Effect on survival
`Improved long-term survival in IPAH patients treated with epoprostenol compared
`Shapiro 1997
`with historic controls
`
`Retrospective
`reviews:
`McLaughlin (2002)
`Sitbon (2002)
`Kuhn (2003)
`
`McLaughlin (1998)
`
`Long-term outcome (3–5-year
`follow-up) and predictors of
`response to therapy
`
`Baseline hemodynamics and right-ventricular function predict response to therapy.
`Patients who improve at 3–12-month follow-up have better long-term prognosis.
`Patients with scleroderma PAH have worse long-term prognosis, even if treated
`with intravenous epoprostenol. Treated patients had better survival compared with
`historic controls or to expected survival derived from the NIH equation
`
`Insights into mechanism
`of action
`
`With epoprostenol, long-term decrease in PVR is more pronounced
`compared with the acute response, suggesting an effect on pulmonary
`vascular remodeling
`
`Rich 1999
`
`Dose adjustment
`
`Intravenous epoprostenol rate should be adjusted to avoid deleterious
`hyperdynamic cardiovascular effects
`
`Ref.
`[17,18]
`
`[19–21]
`
`[22]
`
`[23]
`
`Use of intravenous
`epoprostenol in other forms
`of PAH
`
`Hemodynamic and symptomatic improvement with intravenous epoprostenol in
`patients with other forms of PAH
`
`[24–28]
`
`Case series:
`McLaughlin 1999
`Robbins 2000
`Horn 2000
`Krowka 1999
`Rosenzweig 1999
`
`IPAH: Idiopathic pulmonary hypertension; NIH: National Institutes of Health; PAH: Pulmonary arterial hypertension; PVR: Pulmonary vascular resistance.
`
`Alternative approaches to therapy with currently
`available drugs
`Several therapeutic strategies aimed at improving the control of
`the disease or the risk–benefit ratio of the treatment were
`employed in small clinical trials and retrospective series.
`
`Combination therapy
`Since multiple and complex pathogenetic mechanisms are
`implicated in the development and progression of pulmonary
`hypertension, various combinations of drugs with already
`proven benefit may represent alternative therapeutic strategies
`in PAH. Prostacyclins, ET receptor antagonists and drugs act-
`ing through a NO-dependent pathway (such as sildenafil, a
`phosphodiesterase inhibitor) have different intracellular signal
`transduction pathways with potential synergistic effect. A
`small multicenter clinical trial on the combined use of epo-
`prostenol and bosentan, initiated simultaneously, suggested
`that bosentan may provide a small additional hemodynamic
`benefit to severe PAH patients who require epoprostenol treat-
`ment [40]. In retrospective studies of patients on chronic epo-
`prostenol or treprostinil, the addition of bosentan was safe and
`increased vasodilatory efficacy, which allowed for prostacyclin
`dose reduction or even discontinuation [41–43]. Small series
`have demonstrated acute and chronic benefit from combined
`use of various prostacyclins (intravenous epoprostenol, subcu-
`taneous
`treprostinil
`and
`inhaled
`iloprost)
`and oral
`sildenafil [44–47]. A multicenter trial investigating the effect of
`combined oral sildenafil and intravenous epoprostenol is cur-
`rently ongoing. The possibility of unexpected interactions
`between these drugs and the absence of efficacy and safety data
`in large clinical trials preclude recommendation of routine use
`of combination PAH therapy.
`
`Transition from intravenous epoprostenol to oral bosentan or
`subcutaneous treprostinil
`Continuous intravenous epoprostenol administration, although
`known to improve exercise capacity, symptoms, hemodynamics
`and right-ventricular function in severe PAH and to offer survival
`benefit in IPAH patients, requires a complicated delivery system.
`It may be associated with potentially life-threatening side effects,
`such as line-related sepsis and thrombosis or rebound pulmonary
`hypertension and acute right heart failure from inadvertent dis-
`continuation. Newer PAH drugs have also been shown to have
`beneficial effects on exercise capacity, symptoms, hemodynamics
`and clinical events in patients with severe PAH. Small published
`series from centers with extensive experience in PAH manage-
`ment have demonstrated the feasibility and safety of transition-
`ing selected patients from intravenous epoprostenol to other
`therapeutic alternatives, such as subcutaneous treprostinil and
`oral bosentan [43,48,49]. In addition, a current multicenter trial is
`evaluating the safety of transitioning PAH patients from intra-
`venous epoprostenol to subcutaneous treprostinil. At the
`present time, however, the authors caution against indiscrimi-
`nate discontinuation and substitution of intravenous epopros-
`tenol with other available drugs. There is no guarantee that, in
`the case of clinical deterioration, reinstitution of previous ther-
`apy will reverse disease progression, which may have fatal con-
`sequences. Larger clinical investigations are warranted to estab-
`lish enrollment criteria and transitioning methods to assure
`both the efficacy and safety of such an approach.
`
`Use of alternative methods of delivery
`The main difficulty with subcutaneous treprostinil therapy
`is the development of pain and reactions at the infusion
`site [29]. To avoid this side effect of treprostinil, intravenous
`
`350
`
`Expert Rev. Cardiovasc. Ther. 3(2), (2005)
`
`Liquidia's Exhibit 1104
`Page 4
`
`

`

`Pulmonary arterial hypertension
`
`PAH diagnosis
`Suggested/suspected/screening*
`
`Diagnosis confirmation and
`determination of severity
`(cid:127) Right heart catheterization ‡
`(cid:127) Vasoreactivity testing
`
`Determination of the
`diagnostic class§
`(cid:127) Echocardiography
`(cid:127) Electrocardiography
`(cid:127) Pulmonary function test
`(cid:127) HIV test
`(cid:127) Ventilation–perfusion scan
`(cid:127) Chest imaging tests
`(cid:127) Connective tissue-disease
` screening
`(cid:127) Pulmonary angiogram
`(cid:127) Cardiac MRI
`
`Evaluation of the
`functional impairment¶
`(cid:127) WHO class
`(cid:127) Echocardiography
`(cid:127) Right-heart
` catheterization
`(cid:127) 6 min walk test
`(cid:127) Cardiopulmonary
` exercise test
`(cid:127) Cardiac MRI
`
`Treatment
`
`Follow-up
`Transplant referral
`
`and inhalatory routes of administration
`were recently investigated. Treprostinil has
`a longer half-life compared with epo-
`prostenol, which decreases the risk of
`rebound pulmonary hypertension and
`right heart failure in the case of abrupt dis-
`continuation. In comparison to intrave-
`nous epoprostenol, intravenous trepros-
`tinil administration alleviates the need for
`a large infusion pump, drug reconstitution
`and the use of ice packs. Continuous
`intravenous treprostinil administration has
`been evaluated for safety and efficacy in a
`multicenter trial, both as transition from
`intravenous epoprostenol and as de novo
`therapy [50]. Intravenous treprostinil was
`associated with an improvement in exer-
`cise performance and hemodynamics,
`with added advantages of safety and con-
`venience [50]. It has recently received FDA
`approval for this route of administration.
`In a recent report from Germany,
`inhaled treprostinil demonstrated sub-
`stantial pulmonary vasodilatory efficacy
`in acute administration, as well as symp-
`tomatic and
`functional benefit
`in
`chronic use in a small number of PAH
`patients [51].
`
`Head-to-head comparisons between
`currently available drugs
`With the advent of multiple therapeutic
`options for PAH patients, questions of
`relative efficacy, safety and therapy of
`choice are likely to arise. Head-to-head
`comparison trials are the most meaning-
`ful and accurate method of detecting effi-
`cacy and safety differences among various
`PAH treatments with already proven benefit. Experimental
`data from tissue culture and animal studies are not necessarily
`predictive of human response, and data from individual clini-
`cal trials cannot be directly compared. An example is repre-
`sented by ET receptor antagonists. There are two types of ET
`receptors: ETA receptors, located in the vascular smooth mus-
`cle cells, and ETB receptors, found on both endothelial (ETB1
`subtype) and smooth muscle cells (ETB2 subtype) [52]. ET
`binding to receptors located on the vascular smooth muscle
`cell (ETA and ETB2) induces vasoconstriction and smooth
`muscle-cell proliferation [53,54]. The relative contribution of
`ETA and ETB2 receptors to the ET-mediated vasoconstriction
`depends on the species, experimental conditions and vascular
`bed studied. ETB1 receptors mediate vasodilatation and are
`responsible for ET clearance from the circulation [55]. Three
`ET receptor antagonists were recently evaluated in PAH trials:
`bosentan, a mixed ETA/ETB receptor antagonist already
`
`Figure 2. PAH management. Investigations used to characterize the disease process.
`*Strongly consider early referral to specialized centers.
`‡Right heart catheterization is required to confirm the diagnosis of PAH. Vasoreactivity testing by
`inexperienced operators may place patients at unduly increased risk.
`§Type and extent of work-up varies on individual basis, but all patients with suspected PAH diagnosis
`should undergo a battery of essential tests, which include echocardiography, chest radiography,
`ventilation-perfusion scan, HIV testing and screening for connective tissue disease, and pulmonary
`function test with oximetry.
`¶Prior to institution of therapy, it is essential to evaluate functional parameters with prognostic
`significance, such as exercise capacity and right-ventricular function.
`MRI: Magnetic resonance imaging; PAH: Pulmonary arterial hypertension; WHO: World
`Health Organization.
`
`approved for clinical use, and the selective ETA receptor
`antagonists, sitaxsentan and ambrisentan. Theoretically, selec-
`tive blockade of the ETA receptors may be more advantageous
`compared with nonselective ETA/ETB antagonism, by preser-
`ving ETB1-mediated vasodilatation and ET clearance. How-
`ever, in isolated pulmonary arteries and in animal models,
`combined ETA/ETB receptor blockade was superior to selec-
`tive ETA antagonism in reducing vasoconstriction and in
`improving right-ventricular hypertrophy and animal survival,
`respectively [56,57]. In randomized clinical trials in patients
`with PAH, all three ET receptor antagonists improved exer-
`cise capacity, hemodynamics, symptoms and WHO func-
`tional class [33–36]. It is impossible to accurately compare their
`relative efficacy on the basis of the available trials, because the
`PAH patient populations and trial designs were different.
`However, conducting large-scale head-to-head comparison
`studies would require more refined methodology and increased
`
`www.future-drugs.com
`
`351
`
`Liquidia's Exhibit 1104
`Page 5
`
`

`

`Sulica & Poon
`
`Table 3. Major clinical trials with newer prostanoids.
`Trial
`Drug/
`n
`PAH (%)
`WHO class (%)
`route
`active/placebo
`active/placebo
`
`Duration
`(months)
`
`Control
`
`Primary
`end point
`
`Dose
`
`Simmoneau
`(2002)
`
`SQ
`treprostinil
`
`Olschewski
`(2002)
`AIR
`
`Inhaled
`iloprost
`
`Galie
`(2002)
`ALPHABET
`
`Oral
`beraprost
`
`470 IPAH: 58/58
`CTDz: 17/21
`CHDz: 25/22
`
`203 IPAH: 50.5/50
`CTDz: 12.9/21.6
`Anorex.: 4/4.9
`CTEPH: 32.7/23.5
`
`130 IPAH: 53.9/43.1
`CTDz: 7.7/12.3
`CHDz: 13.8/23/1
`Portal HTN: 18.5/13.8
`HIV: 6.2/7.7
`
`Barst
`(2003)
`
`Oral
`beraprost
`
`116 IPAH: 78/70
`CTDz: 10/11
`CHDz: 12/20
`
`II
`
`III
`
`IV
`
`11/12
`
`82/81
`
`8/7
`
`3
`
`Placebo
`
`6MWT
`
`9.3 ng/kg/min
`(mean)
`
`59.4/57.8 40.6/42.2 3
`
`Placebo
`
`Combined
`clinical
`end point*
`
`30 µg/day
`(median)
`6–9 inhal./day
`
`47.7/50.8 52.3/49.2
`
`3
`
`Placebo
`
`6MWT
`
`80 µg
`four-times daily
`
`55/50
`
`45/50
`
`12
`
`Placebo
`
`Disease
`progression*
`
`120 µg
`four-times daily
`
`*Combined clinical end point was met if there was an improvement in the WHO class by at least one unit and in the 6MWT distance by at least 10% compared with baseline
`in the absence of clinical deterioration or death.
`‡Disease progression was represented by death, transplantation, epoprostenol rescue or more than 25% decrease in the peak VO2. Beraprost-treated patients exhibited less
`disease progression at 6 months, but no such effect was evident at shorter or longer duration of follow-up. The 6MWT distance improved at 3- and 6-month follow-up, but
`there were no further treatment effects noted at 9- and 12-month intervals.
`§Treatment effect on the 6MWT distance is the difference between active and placebo groups.
`¶Clinical worsening, variously defined in studies, includes clinical events consistent with PAH progression, such as death, (need for) transplantation, hospital admission for
`right heart failure, or need for additional PAH therapy.
`6MWT: 6-min walk test; AIR: Aerosolized Iloprost Randomized study; ALPHABET: ArteriaL Pulmonary Hypertension And BeraprosT; CHDz: Congenital heart disease; CTDz: Connective
`tissue disease; CTEPH: Chronic thromboembolic pulmonary hypertension; H/dyn: Hemodynamics; HTN: Hypertension; QOL: Quality of life; WHO: World Health Organization.
`
`cost. Such a study is expected to involve a longer duration,
`larger number of patients and more elaborate and sensitive end
`points to detect significant differences between groups. With
`the exception of an ongoing trial on the use of sitaxsentan in
`PAH (i.e., Sitaxsentan To Relieve ImpaireD Exercise [STRIDE]
`II), which has a bosentan comparison arm, the authors are not
`aware of any other direct comparison trials being conducted.
`
`Particularities associated with clinical trial design in pulmonary
`arterial hypertension
`Current design of therapeutic trials in PAH is challenged by
`ethical and logistic dilemmas, particularly with respect to the
`study design and the choice of appropriate end points. The
`scientific community and regulatory agencies require perform-
`ance in randomized, controlled trials of investigational PAH
`drugs as definitive proof of efficacy and safety. Ethical difficul-
`ties may arise from enrolling patients in double-blind, pla-
`cebo-controlled, randomized studies in the face of available
`therapeutic options known to improve outcome. Most of the
`recently completed randomized trials were blinded and pla-
`cebo-controlled for 3 to 4 months, followed by open-label or
`blinded, dose-ranging, active treatment extensions for long-
`term data collection. Specific mechanisms to protect patients
`in case of clinical worsening were incorporated, such as
`
`switching to open-label active treatment or using rescue therapy
`with already approved drugs. The relatively short duration of
`the placebo-controlled period, however, may not detect possi-
`ble attenuation of the clinical effect [32], and it may be
`inadequate for trials enrolling patients with less advanced dis-
`ease or evaluating combination therapies.
`The most commonly used primary end point in recent PAH
`clinical trials was the 6MWT distance as a measure of the exer-
`cise capacity. Peak oxygen consumption during formal cardio-
`pulmonary exercise test (CPET), WHO/New York Heart Asso-
`ciation (NYHA) functional class or measures of disease
`progression were also used as primary end points. Resting
`hemodynamics, symptoms (Borg score), general clinical status
`(e.g., dyspnea-fatigue rating), WHO/NYHA class, the time to
`clinical worsening (a composite of death, transplant, hospital
`admission for right heart failure or need for rescue therapy with
`intravenous epoprostenol) and quality of life measures were
`employed as secondary end points. The 6MWT is reliable, easy
`to administer and clinically meaningful, since it correlates with
`prognosis and CPET parameters in IPAH [58,59]. The 6MWT is
`validated for studies in PAH patients with moderate and severe
`disease. However, it may be insensitive in patients with less
`advanced disease and to date it is not validated in patients with
`certain forms of PAH, such as portopulmonary hypertension.
`
`352
`
`Expert Rev. Cardiovasc. Ther. 3(2), (2005)
`
`Liquidia's Exhibit 1104
`Page 6
`
`

`

`Table 3. Major clinical trials with newer prostanoids (cont.).
`Treatment effect
`
`Comments
`
`Ref.
`
`Pulmonary arterial hypertension
`
`6MWT
`(m)
`+16
`(median)
`
`+36.4
`(median)
`
`+25.1
`(mean)
`
`Peak VO2 H/dyn
`
`N/A
`
`Better
`
`Clinical
`worsening
`No effect
`
`Survival Symptoms WHO
`class
`N/A
`
`Better
`
`No
`effect
`
`QOL
`
`Better
`
`N/A
`
`Better
`
`Better
`
`No
`effect
`
`Better
`
`Better
`
`Better
`
`Treatment effect was dependent on
`the baseline 6MWT distance and the
`dose achieved
`
`Absolute change in the mean 6MWT
`distance was larger in the IPAH group
`
`N/A
`
`No effect No effect
`
`No
`effect
`
`Better
`
`No
`change
`
`N/A
`
`Subgroup analysis showed greater
`6MWT improvement in IPAH
`
`+31
`(median)*
`
`Trend to
`increase
`
`No effect Better
`
`No
`effect
`
`No effect
`
`No effect No
`effect
`
`The study was prematurely terminated
`to accelerate data review. Less disease
`progression in the treated group at
`6 months only
`
`[29]
`
`[30]
`
`[31]
`
`[32]
`
`*Combined clinical end point was met if there was an improvement in the WHO class by at least one unit and in the 6MWT distance by at least 10% compared with baseline
`in the absence of clinical deterioration or death.
`‡Disease progression was represented by death, transplantation, epoprostenol rescue or more than 25% decrease in the peak VO2. Beraprost-treated patients exhibited less
`disease progression at 6 months, but no such effect was evident at shorter or longer duration of follow-up. The 6MWT distance improved at 3- and 6-month follow-up, but
`there were no further treatment effects noted at 9- and 12-month intervals.
`§Treatment effect on the 6MWT distance is the difference between active and placebo groups.
`¶Clinical worsening, variously defined in studies, includes clinical events consistent with PAH progression, such as death, (need for) transplantation, hospital admission for
`right heart failure, or need for additional PAH therapy.
`6MWT: 6-min walk test; AIR: Aerosolized Iloprost Randomized study; ALPHABET: ArteriaL Pulmonary Hypertension And BeraprosT; CHDz: Congenital heart disease; CTDz: Connective
`tissue disease; CTEPH: Chronic thromboembolic pulmonary hypertension; H/dyn: Hemodynamics; HTN: Hypertension; QOL: Quality of life; WHO: World Health Organization.
`
`Compared with the 6MWT, CPET parameters may provide
`more objective and detailed evaluation of the functional
`capacity and the state of the pulmonary circulation in
`PAH [60–62]. In one trial with beraprost and another with
`sitaxsentan, the 6MWT distance and peak oxygen consump-
`tion were used in parallel as study end points [32,35].
`Although a treatment effect was detected by improvement in
`the 6MWT distance, there were no consistent significant
`changes in peak oxygen consumption. Such a difference in
`findings is most likely due to variable interpretation of the
`CPET results at different centers, unpredictable degree of
`technical skill and failure to weight-adjust the 6MWT dis-
`tance [63]. Use of follow-up right heart catheterization to
`document the treatment effect, although objective and accu-
`rate, may impact on the safety of study participants and
`increase the cost of investigation. Time to clinical worsening
`as a measure of severe complications has been a useful clini-
`cal end point in trials evaluating patients in WHO/NYHA
`Classes III and IV. It may be less sensitive in combination
`and direct comparison trials and even unethical in patients
`with less advanced disease. WHO/NYHA classification corre-
`lates well with disease severity and prognosis in IPAH, but it is
`a self-reported measure, subjected to interpersonal variability
`and observer bias. The impact of therapy on quality of life
`
`measures is an important end point from the perspective of
`both

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