throbber
Pharmacology & Therapeutics 114 (2007) 318 – 326
`
`www.elsevier.com/locate/pharmthera
`
`Associate editor: R.M. Wadsworth
`Therapy of pulmonary hypertension in neonates and infants
`Thomas Hoehn ⁎
`
`Neonatology and Pediatric Intensive Care Medicine, Department of General Pediatrics, Heinrich-Heine-University, Moorenstr. 5 D-40225 Duesseldorf, Germany
`
`Abstract
`
`Pulmonary hypertension (PH) in newborns and infants can present in its idiopathic form or complicate a long list of other diseases. Most of
`these conditions are either pulmonary or cardiovascular in origin. In the present review our currrent knowledge regarding pathophysiology,
`structural changes, diagnosis, and available treatment options for PH in the age group below 1 year of age is summarized. New treatment options
`available in adults including endothelin receptor antagonists (ETRA) and phosphodiesterase (PDE) inhibitors are presented and the need for
`randomized controlled trials in newborns and infants is emphasized. Future candidates for pharmacotherapy of PH in infants include among others
`vasoactive intestinal polypeptide (VIP), PDE-3 and PDE-4 inhibitors, hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, and
`adrenomedullin (ADM).
`© 2007 Elsevier Inc. All rights reserved.
`
`Keywords: Persistent pulmonary hypertension of the newborn; Pulmonary arterial hypertension; Nitric oxide; Prostacyclin; Sildenafil; Bosentan
`
`Abbreviations: 6MWD, 6-min walking distance; ADM, adrenomedullin; EPC, endothelial progenitor cells; ETRA, endothelin receptor antagonist; HMG-CoA,
`hydroxymethylglutaryl coenzyme A; MCT, monocrotaline; NO, nitric oxide; PAH, pulmonary arterial hypertension; PDE, phosphodiesterase; PH, pulmonary
`hypertension; PPHN, persistent pulmonary hypertension of the newborn; PVR, pulmonary vascular resistance; VEGF, vascular endothelial growth factor; VIP,
`vasoactive intestinal polypeptide.
`
`Contents
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Introduction .
`1.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Physiology of perinatal and postnatal changes .
`2.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Definition and incidence of pulmonary hypertension.
`4.
`Structural changes in persistent pulmonary hypertension of the newborn/pulmonary hypertension
`5.
`Functional changes in persistent pulmonary hypertension of the newborn/pulmonary hypertension
`6. Animal models .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`7. Neonatal disease .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`7.1.
`Primary persistent pulmonary hypertension of the newborn .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`7.2.
`Secondary persistent pulmonary hypertension of the newborn .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`7.3.
`Clinical presentation of persistent pulmonary hypertension of the newborn .
`.
`.
`.
`.
`.
`.
`.
`Pulmonary hypertension in infancy .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`8.
`9. Diagnosis of pulmonary hypertension .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`9.1. Newborns .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`9.2.
`Infants .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`319
`319
`320
`320
`320
`321
`321
`321
`322
`322
`322
`322
`322
`322
`
`⁎ Tel.: +49 211 81 18091; fax: +49 211 81 19786.
`E-mail address: thomas.hoehn@uni-duesseldorf.de.
`
`0163-7258/$ - see front matter © 2007 Elsevier Inc. All rights reserved.
`doi:10.1016/j.pharmthera.2007.03.006
`
`318
`
`

`
`T. Hoehn / Pharmacology & Therapeutics 114 (2007) 318–326
`
`319
`
`10.
`
`Therapy of pulmonary hypertension .
`10.1. Newborns .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`10.2.
`Infants.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Future options .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`11.
`Summary .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`12.
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`. 322
`. 322
`. 322
`. 323
`. 324
`. 324
`
`1. Introduction
`
`Pulmonary hypertension (PH) covers a broad clinical
`spectrum ranging from transient neonatal condition to perma-
`nent disabling disease in infancy or childhood. A variety of
`perinatal conditions can trigger persistent PH of the newborn
`(PPHN; Dakshinamurti, 2005). In the majority of cases, PPHN
`can successfully be reversed by specific treatment of the
`underlying condition in addition to treatment of PH. Primary
`PPHN is a rare event, in which cases no triggering condition of
`PPHN can be identified. Treatment of PPHN consists of
`preferably selective pulmonary vasodilation, which was first
`made available by the introduction of inhaled nitric oxide (NO)
`into neonatal clinical practice in the early 1990s (Kinsella et al.,
`1992; Roberts et al., 1992). While starting with considerable
`enthusiasm for the new drug, neonatologists began to realize,
`that a certain proportion of newborns with clinical PPHN did
`not respond favorably to NO (Hoehn & Krause, 2001; Travadi
`& Patole, 2003). Clinical management of this subgroup of
`patients combined with economic considerations and transient
`decreased availability of NO combined to boost the develop-
`ment of alternative drugs, like phosphodiesterase-5 inhibitors
`(PDE) or endothelin-1 receptor antagonists (ETRA) to success-
`fully treat PH. Advantages of the latter medications include a
`prolonged half-life and independence from inhalational equip-
`ment. Unfortunately, due to the low number of neonates and
`infants treated with these substances, there are currently neither
`
`randomized controlled trials nor much experience with long-
`term application of these drugs available.
`
`2. Physiology of perinatal and postnatal changes
`
`A prerequisite for efficient postnatal gas exchange in any
`newborn infant is the clearance of substantial amounts of fetal
`lung fluid (Jain & Dudell, 2006). Rapid clearance of fetal lung
`fluid is a key part of perinatal adaptation and is mediated in
`large part by transepithelial sodium reabsorption through amilo-
`ride-sensitive sodium channels in the alveolar epithelial cells
`(Jain & Eaton, 2006). Failure to achieve this adaptation results
`in respiratory morbidity presenting as transient tachypnea of the
`newborn (term infant) or respiratory distress syndrome (preterm
`infant), depending on gestational age of
`the newborn.
`Particularly when uterine contractions are absent immediately
`prior to delivery, as in selective Cesarean section, there is no
`activation of the amiloride-sensitive sodium channels in the
`alveolar epithelial cells and therefore an increased risk of
`respiratory morbidity.
`Regulation of pulmonary arterial resistance thus determining
`pulmonary blood flow is achieved by the interaction of 3 main
`players: NO, endothelin, and prostaglandins (Ziegler et al.,
`1995). Among these, endothelin increases pulmonary vascular
`resistance (PVR), whereas NO and prostaglandins lead to
`vasodilation and reduced vascular resistance (for details, see
`Fig. 1). Other genes and their products involved in the
`
`Fig. 1. Schematic interactions of endothelin, NO, and prostaglandins between pulmonary endothelial and smooth muscle cells.
`
`319
`
`

`
`320
`
`T. Hoehn / Pharmacology & Therapeutics 114 (2007) 318–326
`
`pathogenesis of PH include prostacyclin synthase, serotonin
`transporters, serine elastases, matrix metalloproteinases
`(MMP), voltage-gated potassium (Kv) channels, angiotensin-
`converting enzyme (ACE), vascular endothelial growth factor
`(VEGF), carbamoyl phosphate synthase, and plasminogen
`activator inhibitor type 1 (PAI-1; Runo & Loyd, 2003). The
`ultimate effect on pulmonary vascular remodeling is a result of
`the interplay of these genetic factors, modifying genes, and
`environmental factors.
`PVR is high in utero when pulmonary blood flow is limited
`to ∼ 8% of total cardiac output and fetal oxygen requirements
`are met by placental blood flow (Lakshminrusimha & Stein-
`horn, 1999).
`Immediately after birth PVR decreases and
`continues to do so for the following 3 months (Haworth,
`1995). The decrease in PVR leads to increases in pulmonary
`blood flow. Higher pressures in the left atrium and in the aorta
`lead to functional closure of the foramen ovale and reverse the
`intrauterine right-to-left shunting across the duct into a left-to-
`right shunting until the duct eventually closes. Any disruption in
`arterial oxygenation can reverse this process and lead to
`increased PVR and subsequent right-to-left shunt with clinical
`cyanosis.
`
`3. Definition and incidence of pulmonary hypertension
`
`The definition of PH is derived from adult patients and
`includes all individuals with mean pulmonary arterial pressures
`N25 mm Hg at rest or N30 mm Hg with exercise no matter what
`age (British Cardiac Society Guidelines and Medical Practice
`Committee, 2001). Since most of these measurements are
`performed by echocardiography, tricuspid regurgitation with a
`Doppler velocity of more than 2.5 m/sec has been used for the
`screening for PH. Most pediatric cardiologists would agree on a
`definition of PH where systolic pulmonary artery pressure
`exceeds 50% of systolic systemic pressure. These measure-
`ments are usually taken from either tricuspid regurgitation
`or from any known connection between systemic and pulmo-
`nary circulation (i.e., patent ductus arteriosus, ventricular septal
`defect; Tulloh, 2006).
`The WHO classification of PH, which has been modified
`lastly in 2003 (Proceedings of the 3rd World Symposium on
`Pulmonary Arterial Hypertension, 2004) is shown in Table 1.
`Conflicting data have been published concerning the in-
`cidence of PPHN. Whereas Farrow et al. (2005) quantified the
`incidence at 0.2% of live-born term infants, others gave a
`higher range of 0.43–6.8 per 1000 (Walsh-Sukys et al., 2000).
`The associated mortality rate of PPHN at the beginning of the
`21st century was given at 10–20%, whereas earlier investiga-
`tions reported mortality rates of up to 50% (Fox et al., 1977).
`According to a single center experience over almost 2 de-
`cades, a high proportion of infants who died from clinically
`suspected idiopathic PPHN were later found to have alveolar
`capillary dysplasia at autopsy (Tibballs & Chow, 2002). A
`decreasing incidence of PPHN could be expected from the
`falling incidence of meconium aspiration syndrome (Yoder
`et al., 2002), a disease which is frequently associated with
`PPHN.
`
`4. Structural changes in persistent pulmonary
`hypertension of the newborn/pulmonary hypertension
`
`Most prominent histologic changes in PPHN include hyper-
`trophy of the perivascular muscular layer in small and large
`pulmonary arteries (see Fig. 2A,B). Ultimately all 3 layers of the
`vascular wall are affected by thickening and extracellular matrix
`deposition, which is summarized by the term ‘pulmonary vascular
`remodeling’ (Jeffery & Wanstall, 2001). The latter condition
`consists of precocious development of muscle in intraacinar
`arteries, proliferation of adventitial connective tissue, and medial
`hypertrophy of preacinar arteries (Geggel et al., 1986).
`
`5. Functional changes in persistent pulmonary
`hypertension of the newborn/pulmonary hypertension
`
`Functional changes in PPHN/PH are mainly related to
`endothelial dysfunction and result in a dysbalance between
`
`Table 1
`Revised clinical classification of PH (adapted from Farber and Loscalzo, 2004;
`Simonneau et al., 2004)
`
`Group I
`Pulmonary arterial hypertension (PAH)
`Idiopathic (IPAH)
`Familial (FPAH)
`Associated with (APAH)
`•Collagen vascular disease
`•Congenital systemic-to-pulmonary shunts
`•Portal hypertension
`•HIV infection
`•Drugs and toxins
`•Other (thyroid disorders, glycogen storage disease, Gaucher disease,
`hereditary hemorrhagic telangiectasia, hemoglobinopathies,
`myeloproliferative disorders, splenectomy)
`Associated with significant venous or capillary involvement
`•Pulmonary veno-occlusive disease (PVOD)
`•Pulmonary capillary hemangiomatosis (PCH)
`PPHN
`
`Group II
`PH with left heart disease
`Left-sided atrial or ventricular heart disease
`Left-sided valvular heart disease
`
`Group III
`PH associated with lung diseases and/or hypoxemia
`Chronic obstructive pulmonary disease
`Interstitial lung disease
`Sleep-disordered breathing
`Alveolar hypoventilation disorders
`Chronic exposure to high altitude
`Developmental abnormalities
`
`Group IV
`PH due to chronic thrombotic and/or embolic disease
`Thromboembolic obstruction of proximal pulmonary arteries
`Thromboembolic obstruction of distal pulmonary arteries
`Nonthrombotic pulmonary embolism (tumor, parasites, foreign material)
`
`Group V
`Miscellaneous
`Sarcoidosis, histiocytosis X, lymphangiomatosis, compression of pulmonary
`vessels (adenopathy, tumor, fibrosing mediastinitis)
`
`320
`
`

`
`T. Hoehn / Pharmacology & Therapeutics 114 (2007) 318–326
`
`321
`
`by ductal ligation in the fetal lamb VEGF expression was
`markedly reduced (Grover et al., 2003). The postnatal model
`of hypoxia-induced PH found increased levels of VEGF
`but abolished vasodilation to VEGF potentially secondary to
`decreased expression of the VEGF receptor 2 (Nadeau et al.,
`2005).
`
`6. Animal models
`
`Several models of PH exist, the most frequently used among
`these are the injection of monocrotaline (MCT) and exposure
`to hypoxia (Campian et al., 2006). These animal models have
`not only been used to characterize the pathophysiology of PH
`and its sequelae such as right ventricular hypertrophy and
`failure but also to test novel therapeutic strategies. Others have
`used a specific rat strain, the Fawn-hooded rat, which tends to
`spontaneously develop PH under certain conditions (Le Cras
`et al., 1999; Tyler et al., 1999). The analysis of Campian et al.
`(2006) suggests that all approaches which have been
`successful in patients (most notably prostacyclin and ETRA)
`are also effective in various animal models. This is not true the
`other way around: results of animal experiments can often not
`been translated into efficacy in clinical studies, which presents
`a valid argument to perform the human studies in adults first,
`and thereafter in infants and children. Additional factors iden-
`tified to affect pulmonary vascular resistance include carbon
`dioxide and pH. In a neonatal lamb model it was shown that
`elevated pH rather than decreased PaCO2 during hyperventi-
`lation appears to be the major factor in moderating the response
`of the pulmonary vessels to acute hypoxia (Lyrene et al., 1985).
`Similar data have been obtained from infants after cardiopul-
`monary bypass for cardiac surgery. In this investigation in-
`creasing the arterial pH by the administration of sodium
`bicarbonate both lowered the pulmonary arterial pressure
`and increased the cardiac index, resulting in a decrease in
`pulmonary vascular resistance. These changes were observed
`without alteration in PaCO2 (Chang et al., 1995). These ob-
`servations should provide sufficient evidence to refrain from
`using hyperventilation in the clinical management of PH,
`whereas running the risk of decreasing cerebral blow flow
`below a critical threshold. Metabolic alkalosis apparently rep-
`resents a highly efficient modulator of PH.
`
`7. Neonatal disease
`
`7.1. Primary persistent pulmonary hypertension of the newborn
`
`The absence of any known trigger of PPHN in a term
`newborn presenting with cyanosis due to PH immediately
`postnatally suggests the presence of primary PPHN. In this
`condition PH appears to be caused by an abnormal pulmonary
`vascular bed rather than functional vasoconstriction due to other
`causes (Murphy et al., 1981). Intrauterine conditions like
`chronic hypoxia have been shown to result in muscularization
`of the pulmonary vasculature in animals and humans and to
`cause sustained hypertension after hypoxia is reversed (Rabi-
`novitch et al., 1981; Stenmark et al., 1987).
`
`Fig. 2. (A) Hematoxylin–eosin staining of small pulmonary artery in control
`infant (magnification 400×, own data). (B) Hematoxylin–eosin staining of
`small pulmonary artery in an infant with PPHN (magnification 400×, own
`data).
`
`vasodilation and vasoconstriction, in which vasoconstriction
`prevails. One factor contributing to vasoconstriction is high
`levels of endothelin-I, which have been shown in patients with
`PPHN (Vitali & Arnold, 2005). Another explanation for pul-
`monary arterial vasoconstriction in PPHN is the finding of
`diminished NO synthesis (Endo et al., 2001). Although endo-
`thelial NO synthase (eNOS) is upregulated in rapid PPHN
`(RPPHN), a particularly severe and fulminant form of PPHN
`(Hoehn et al., 2003), the net effect for the vessel diameter may
`still be vasoconstriction. Once hypoxia is present in PPHN it
`induces synthesis and release of VEGF (Liu et al., 1998).
`Increased levels of VEGF and smooth muscle proliferation
`can both be suppressed by NO. Others have shown beneficial
`effects of recombinant VEGF treatment in a lamb model of
`experimental PPHN (Grover et al., 2005). In this investigation
`VEGF improved endothelium-dependent vasodilation and
`reduced the severity of pulmonary vascular remodeling. Al-
`though VEGF expression is increased in newborns with
`PPHN (Lassus et al., 2001), the effects of VEGF may be
`altered by impaired signaling. An alteration of VEGF sig-
`naling has been suggested by animal studies of PH induced by
`either hypoxia or VEGF inhibition (Grover et al., 2003;
`Nadeau et al., 2005). In an intrauterine model of PH induced
`
`321
`
`

`
`322
`
`T. Hoehn / Pharmacology & Therapeutics 114 (2007) 318–326
`
`7.2. Secondary persistent pulmonary hypertension of the newborn
`
`10. Therapy of pulmonary hypertension
`
`A variety of perinatal and postnatal conditions can cause
`secondary PPHN: asphyxia, hypoxia, acidosis, hypoglycemia
`(Dakshinamurti, 2005), cold stress, infection, sepsis, meconium
`aspiration, lung hypoplasia, congenital diaphragmatic hernia
`(Perreault, 2006), respiratory distress syndrome, congenital
`pulmonary lymphangiectasia (Hoehn et al., 2006b), and
`congenital alveolar capillary dysplasia (Tibballs & Chow,
`2002). Symptoms occur either immediately postnatally, such
`as in perinatal asphyxia or can be delayed by hours, like in
`evolving neonatal sepsis.
`
`7.3. Clinical presentation of persistent
`pulmonary hypertension of the newborn
`
`Neonates with PPHN present with severe cyanosis due to
`extrapulmonary shunting across the duct and the foramen ovale.
`Depending on the underlying condition,
`the associated
`tachydyspnea is more or less pronounced.
`
`8. Pulmonary hypertension in infancy
`
`Symptoms of infantile PH include breathlessness, fainting or
`chest pain during exercise, and exercise-induced syncope
`(Tulloh, 2006). A minority of infants presents with cyanosis,
`hemoptysis, or
`right heart
`failure with ankle edema or
`hepatomegaly (Rosenzweig & Barst, 2005).
`
`9. Diagnosis of pulmonary hypertension
`
`9.1. Newborns
`
`PPHN can be most easily diagnosed by echocardiography
`even by the less experienced echocardiographer. Should this
`expertise be unavailable, simultaneous (or rapid sequential)
`transcutaneous measurements of pre- and postductal oxygen
`saturation can be used. A 5% or greater saturation decrease
`from pre- to postductal values is highly suggestive of the
`presence of extrapulmonary right-to-left shunting (Macdonald
`& Yu, 1992). High oxygen requirements without radiographic
`evidence of parenchymal pulmonary disease in any newborn
`infant should lead the clinician to suspect the presence of
`PPHN.
`
`9.2. Infants
`
`Echocardiography is the most frequently used investigation
`to screen for PH or to confirm or refute clinically suspected PH.
`Not only can cardiac causes of PH be identified (if present), the
`measurement of tricuspid regurgitation velocity enables the
`estimation of right ventricular pressures (Tulloh, 2006). Car-
`diac catheterization remains the gold standard for measurement
`of pulmonary artery pressures. This investigation specifical-
`ly allows the quantification of the effects of PVR-lowering
`medications like NO (Atz et al., 1999) or prostacyclin (Mikhail
`et al., 1997).
`
`10.1. Newborns
`
`Therapy of PPHN in preterm and term infants is primarily
`directed at correction of the underlying condition (e.g., sepsis,
`hypoxia). Additionally oxygen saturation can be kept higher
`than in healthy term infants in order to decrease PVR by making
`use of the vasodilatory effect of oxygen. Due to the toxic effects
`of hyperoxia (retinopathy of prematurity; McColm et al., 2004)
`this is not an option in preterm infants. Prostacyclin (PGI2) is a
`potent vasodilator of both pulmonary and systemic circulation
`acting via induction of cyclic adenosine monophosphate
`(Howard & Morrell, 2005). The nonselective effect of
`prostacyclins resulted in the development of the PGI2 analogue
`iloprost, which can be applied by nebulization and therefore acts
`predominantly on the pulmonary circulation. The discovery of
`the identity of endothelium-derived relaxing factor (EDRF) as
`NO (Ignarro et al., 1987; Furchgott & Vanhoutte, 1989) led to
`the rapid clinical application of NO (Kinsella et al., 1992;
`Roberts et al., 1992). This molecule causes selective pulmonary
`vasodilation not only in adults but also in near-term and term
`infants; data on a large number of newborns have been summa-
`rized in a recent Cochrane analysis (Finer & Barrington, 2006).
`Altogether 14 randomized controlled trials have been included
`in this analysis comparing inhaled NO to either standard care
`without NO or allowing NO for rescue treatment. Inhaled NO
`significantly reduced the combined endpoint need for extracor-
`poreal membrane oxygenation (ECMO) or death. Whereas
`mortality was hardly influenced, the main effect of inhaled NO
`was the avoidance of ECMO. No beneficial effect has been
`shown for subgroups of PPHN as for example infants with
`congenital diaphragmatic hernia (Finer & Barrington, 2006).
`Other medications studied in a small number of infants only
`include the PDE-5 inhibitor sildenafil (Baquero et al., 2006) and
`the nonselective ETRA bosentan (Galie et al., 2004). For both
`substances there is currently insufficient data in newborns in
`order to draw any conclusions for clinical practice.
`
`10.2. Infants
`
`Therapy of PH in children depends on the degree of PH in an
`individual patient and the extent of deterioration in specific
`situations (e.g., respiratory infection). One of the oldest drugs
`used for the treatment of PH is nifedipine, to which only a
`minority of infants respond favorably (Tulloh, 2006). Oxygen has
`been used traditionally for treatment not only under clinical
`conditions but also for home treatment. The vasodilatory response
`to high concentrations of oxygen belongs to the established tests
`during cardiac catheterization testing various vasodilatory condi-
`tions, including the inhalation of NO (Scheurer et al., 2006). The
`introduction of prostacyclin (PGI2) into clinical practice brought a
`sustained benefit for hemodynamics and reduced mortality in
`comparison with historical data (McLaughlin et al., 2002). The
`major disadvantage for prolonged treatment consisted of PGI2's
`short half-life of only 6 min and the necessity of an indwelling
`central line for continuous supply of PGI2. Therefore other PGI2
`
`322
`
`

`
`T. Hoehn / Pharmacology & Therapeutics 114 (2007) 318–326
`
`323
`
`analogues like treprostinil have been developed, which has a half-
`life of 3 hr and can be delivered subcutaneously (McLaughlin
`et al., 2003). Although treprostinil has been licensed for use in the
`United States since 2002, there is only limited experience in
`children. Iloprost is another PGI2 analogue which has a half-life of
`∼20 min and can be delivered both intravenously and by
`nebulization. Inhalation of iloprost can reverse pulmonary arterial
`hypertension (PAH) and vascular structural remodeling in MCT-
`treated rats (Schermuly et al., 2005) but unfortunately there seems
`to occur a rapid tolerance to this substance (Schermuly et al.,
`2006). Due to the half-life of iloprost it needs to be inhaled 6–9
`times/day, which makes it not very convenient to integrate into
`normal social life in infants. The only orally available PGI2
`analogue is beraprost, which has a half-life of ∼40 min and is
`currently licensed in Japan. Data of 2 randomized controlled trials
`in adults suggest neither significant hemodynamic effects in
`patients with PAH (Galie et al., 2002) nor was initial improvement
`in 6-min walking distance (6MWD) sustained at 9 and 12 months
`of therapy (Barst et al., 2003). There are currently no data of RCT
`of beraprost in infants; until now merely case series have been
`published (Limsuwan et al., 2005). In this case series from
`Thailand the authors found no significant effect on pulmonary
`arterial pressures. This suggests that beraprost
`is of limited
`suitability in infants since, it does not appear to reduce PH and
`reverse progression of morphologic pulmonary changes.
`Another class of medication is the group of ETRA. Most data
`exist on the nonselective ET-A/ET-B ETRA bosentan. In animal
`models of PH this substance can prevent and even reverse
`hypoxia-induced PH (Chen et al., 1995) and can also partially
`prevent MCT-induced PH (Hill et al., 1997). Human data in
`adults appear encouraging, although hepatic toxicity remains
`a serious concern (Cohen et al., 2004; Oldfield & Lyseng-
`Williamson, 2006). Retrospective data of bosentan treatment
`in children suggest safety and efficacy (Rosenzweig et al.,
`2005). Other ETRAs under investigation include sitaxsentan
`and ambrisentan; pediatric experience with both substances is
`limited (Abman, 2006). In view of the reversal of PH in animal
`models and the suggested safety and efficacy in the Rosenzweig
`data, bosentan appears a promising substance provided hepatic
`toxicity does not present a major obstacle for treatment of
`infants.
`Yet another substance class makes use of the NO-mediated
`mechanism of pulmonary vasodilation: phophodiesterases
`inactivate cyclic GMP (cGMP). cGMP in turn is generated by
`the activation of soluble guanylate cyclases and activates
`protein kinases to achieve vasodilation by relaxation of the
`smooth muscle cell. Since particularly PDE-5 is expressed in
`the pulmonary circulation, PDE-5 inhibitors, like sildenafil,
`have been shown to inhibit the development of PH in animal
`models of PAH (Sebkhi et al., 2003; Schermuly et al., 2004;
`Ladha et al., 2005; Ryhammer et al., 2006). Pediatric data
`suggest a beneficial effect of oral sildenafil on hemodynamics
`and exercise capacity (6MWD) in children with PAH (Humpl
`et al., 2005). In this investigation in 14 children 6MWD
`increased from 278 ± 114 m to 443 ± 107 m at 6 months and to
`432 ± 156 m at 12 months. Apparently a plateau was reached
`between 6 and 12 months of treatment. Mean pulmonary artery
`
`pressure decreased from a median of 60 mm Hg (range, 50–
`105) to 50 mm Hg (range, 38–84). Intravenous sildenafil has
`been shown to be as efficacious as NO in reducing pulmonary
`vascular resistance in children with congenital heart disease
`(Schulze-Neick et al., 2003). Unfortunately the number of
`infants included in these case series is too low to draw any
`reliable conclusions from these data.
`
`11. Future options
`
`Since the armamentarium of available medications for
`the treatment of PH has increased dramatically, the near future
`will bring various combinations of drugs being already on the
`market. The vast majority of ongoing trials includes patients
`above the age of 18 years (phase III study ‘Combination
`treatment with bosentan and sildenafil
`to patients with
`Eisenmengers syndrome’ from the Rigshospitalet, Denmark
`[ClinicalTrials.gov Identifier: NCT00303004]; phase IV study
`‘To assess the efficacy and safety of sildenafil when added to
`bosentan in the treatment of pulmonary arterial hypertension’
`from Pfizer Investigational Site, San Antonio, TX, United
`States [ClinicalTrials.gov Identifier: NCT00323297]; phase
`IV study ‘Combination therapy of bosentan and aerosolized
`iloprost in idiopathic pulmonary arterial hypertension (IPAH)’
`from Hannover Medical School, Germany [ClinicalTrials.gov
`Identifier: NCT00120380]). Others have also included pe-
`diatric patients above the age of 12 years (phase II study
`‘Trial of iloprost inhaled solution as add-on therapy with
`bosentan in subjects with pulmonary arterial hypertension
`(PAH)’ from CoTherix, United States [ClinicalTrials.gov
`Identifier: NCT00086463]; phase IV study ‘Effects of the
`combination of bosentan and sildenafil versus sildenafil
`monotherapy on pulmonary arterial hypertension (PAH)’
`from Actelion, United States [ClinicalTrials.gov Identifier:
`NCT00303459]). As far as the author is aware there are cur-
`rently no trials enrolling newborns or infants in studies of
`combined therapies of pulmonary antihypertensive drugs.
`Like other medications in newborns and infants, a substantial
`number of drugs is not licensed for this particular age group
`(Conroy et al., 1999). Reasons for this fact are numerous and
`include weight adapted low dosages, subsequently a negligible
`business volume for the pharmaceutical industry, and rela-
`tively high costs to conduct licensing trials in newborns and
`infants. Costs would be expected to be especially high due to
`specific outcome measures in this age group. Beyond cardio-
`vascular and echocardiographic parameters, follow-up would
`nowadays require long-term observations into school age in
`order to assess neurodevelopmental outcome of these infants
`(Hoehn et al., 2006a).
`Future trials in newborns and infants will need to address
`the efficacy of monotherapy (e.g., sildenafil, bosentan) versus
`combined therapies, including prostacyclins and NO. The adult
`experience and convincing data from animal experiments on the
`efficacy of these substances preclude the possibility of placebo-
`controlled trials in newborns and infants. Nevertheless it seems
`important to have the adult experience prior to introduction of
`medications in infants, both for numerical reasons (there are
`
`323
`
`

`
`324
`
`T. Hoehn / Pharmacology & Therapeutics 114 (2007) 318–326
`
`many more adult patients) and for reasons of safety of a given
`compound.
`The pathophysiology in this age group would not favor a
`specific combination of drugs. Potentially a combination of
`NO-dependent vasodilation (e.g., sildenafil) with NO-indepen-
`dent mechanisms of action (prostacyclin) might prove superior.
`At
`least
`in newborns and infants deposition of the active
`substance during inhalation is regarded as highly erratic
`(Everard, 1996). In order to answer the question of optimized
`drug therapy in a scientifically acceptable manner, randomized
`controlled trials are needed in this particular age group.
`Other
`treatments for PH include vasoactive intestinal
`polypeptide (VIP), which has been shown to induce both
`intracellular cAMP and cGMP and which requires intact
`endothelial function including synthesis of endothelial NO
`(Shahbazian et al., 2006). In an investigation in adults with
`PAH-inhaled VIP led to significant improvements in pulmonary
`hemodynamics and 6MWD (Petkov et al., 2003).
`Besides PDE-5 inhibitors significant pulmonary vasodilation
`can be achieved by PDE-3 and PDE-4 inhibitors when either
`combined with each other or in conjunction with a prostacyclin
`analogue (Phillips et al., 2005). In a case series of 4 PPHN
`patients, treatment with the PDE-3 inhibitor milrinone was
`reported to efficiently reduce PH following nonresponse to
`inhaled NO. However, 3 quarters of patients developed
`intraventricular hemorrhage, 2 of which were classified as
`severe (Bassler et al., 2006). This potential side effect of
`milrinone needs to be taken very seriously, since intraventric-
`ular hemorrhage is extremely uncommon in term newborns (as
`opposed to preterm infants).
`Hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase
`inhibitors, called as statins, have been shown to improve
`endothelial
`function and exert antiproliferative effects on
`vascular smooth muscle cells of systemic vessels (Rakotoniaina
`et al., 2006). In a rat model of MCT-induced PH, pravastatin
`was superior to atorvastatin regarding the degree of PH and the
`restoration of endothelium-dependent relaxation.
`Adrenomedullin (ADM) is a potent vasodilator and acts by
`cAMP and NO-dependent mechanisms. In patients with PAH,
`ADM decreases pulmonary arterial pressures and pulmonary
`vascular
`resistance following intratracheal or
`intravenous
`delivery (Murakami et al., 2006). Intravenously administered
`ADM gene-modified endothelial progenitor cells (EPC) were
`incorporated into lung tissues and attenuated MCT-induced PH
`in rats (Zhao et al., 2005). In this rat model of MCT-induced PH,
`endothelial-like progenitor cells were transduced with human
`eNOS, which

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