Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Basic & Clinical Pharmacology & Toxicology, 2018, 123, 392–406 Doi: 10.1111/bcpt.

13051

MiniReview

Current and Future Treatments for Persistent Pulmonary


Hypertension in the Newborn
Jonas Pedersen1, Elise R. Hedegaard1, Ulf Simonsen1, Marcus Kr€
uger2, Manfred Infanger2 and Daniela Grimm1,2
1
Department of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark and 2Clinic for Plastic, Aesthetic and Hand Surgery,
Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
(Received 20 March 2018; Accepted 27 May 2018)

Abstract: Persistent pulmonary hypertension in newborn (PPHN) is a serious and possibly fatal syndrome characterized by sus-
tained foetal elevation of pulmonary vascular resistance at birth. PPHN may manifest secondary to other conditions as meconium
aspiration syndrome, infection and congenital diaphragmatic hernia. This MiniReview provides the reader with an overview of
current and future treatment options for patients with PPHN without congenital diaphragmatic hernia. The study is based on sys-
tematic searches in the databases PubMed and Cochrane Library and registered studies on Clinicaltrials.gov investigating PPHN.
Inhaled nitric oxide (iNO) is well documented for treatment of PPHN, but 30% fail to respond to iNO. Other current treatment
options could be sildenafil, milrinone, prostaglandin analogues and bosentan. There are several ongoing trials with sildenafil, but
evidence is lacking for the other treatments and/or for the combination with iNO. Currently, there is no evidence for effect in
PPHN of other treatments, for example tadalafil, macitentan, ambrisentan, riociguat and selexipag used for pulmonary arterial
hypertension in adults. Experimental studies in animal models for PPHN suggest effect of a series of approaches including
recombinant human superoxide dismutase, L-citrulline, Rho-kinase inhibitors and peroxisome proliferator-activated receptor-c
agonists. We conclude that iNO is the most investigated and the only approved pulmonary vasodilator for infants with PPHN. In
the iNO non-responders, sildenafil currently seems to be the best alternative either alone or in combination with iNO. Systematic
and larger clinical studies are required for testing the other potential treatments of PPHN.

Persistent pulmonary hypertension of the newborn (PPHN) is by nitric oxide. Thus, PDE 5 limits the drop in pulmonary
a serious syndrome characterized by sustained foetal elevation vascular resistance. PDE 5 is highly expressed in lung tissue
of pulmonary vascular resistance at birth. The syndrome is during foetal life and a key regulator of perinatal pulmonary
seen in two of 1000 live-born infants and is associated with a circulation along with nitric oxide [3].
normal or low systemic vascular resistance [1]. Newborns with The pathophysiology of PPHN is based on changes in pul-
PPHN are at risk of severe complications as asphyxia, chronic monary vascular structure and function. An elevated pul-
lung disease, neurodevelopmental sequelae and death [2]. monary vascular resistance may result from pulmonary
The pulmonary transition at birth relies on an immediate vasoconstriction, structural remodelling of the pulmonary mus-
drop in pulmonary vascular resistance and an eight to 10 times culature, intravascular obstruction and lung hypoplasia [1]. It
increase in pulmonary blood flow. Stimuli known to con- may be secondary to other diseases as meconium aspiration
tribute to the pulmonary vasodilation and hence the drop in syndrome, infection and congenital diaphragmatic hernia. Pre-
pulmonary vascular resistance are increased PO2, rhythmic term and birth by Caesarean versus natural delivery increase
distension of the lungs, drainage and absorption of foetal lung the risk of PPHN [3]. Antenatal exposure to selective sero-
liquid, and increased production of different vasoactive media- tonin-reuptake inhibitors used to treat maternal depression has
tors. The effects of especially increased PO2, increased blood been associated with the development of PPHN [4]. Another
flow or shear stress and ventilation are partly dependent on known risk factor is antenatal exposure to smoke. The use of
the activation of endothelial nitric oxide synthase. The activa- non-steroidal anti-inflammatory drugs, especially ibuprofen,
tion of endothelial nitric oxide synthase resulting in the pro- naproxen and aspirin during pregnancy is associated with
duction of the vasoactive mediator nitric oxide is responsible PPHN [5]. This is problematic because of the easy access to
for nearly 50% of the fall in pulmonary vascular resistance. over-the-counter non-steroidal anti-inflammatory drugs. Fur-
Phosphodiesterase type 5 (PDE 5) is an enzyme that hydroly- thermore, stress or pain, perinatal malnutrition and hyperoxia
ses cyclic GMP and hence inhibits the vasodilatation induced may impair the pulmonary vascular adaptation at birth [3].
Infants with PPHN secondary to congenital diaphragmatic her-
Author for correspondence: Daniela Grimm, Department of Biomedi-
cine, Pharmacology, Aarhus University, Wilhelm Meyers Alle 4, nia may respond differently to treatment and should be anal-
DK-8000 Aarhus C, Denmark (e-mail dgg@biomed.au.dk). ysed separately [6].

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 393

Clinically, PPHN shows varying degrees of hypoxemic The last literature search was performed on 14 February 2018. On
respiratory failure. PPHN is characterized by labile hypox- PubMed, the search strategy was ‘Persistent Pulmonary Hypertension
aemia and pre–post ductal saturation gradient which mani- of the Newborn’. This resulted in 2130 items. With a ‘Clinical Trial’
filter, 111 items were found. With no filter and the addition of MeSH
fests itself as differential cyanosis [7]. Hypoxaemia is due to
Term ‘Animals, Newborn’, 153 items were collected. For finding in-
intrapulmonary shunting secondary to V/Q mismatch and/or depth literature describing each treatment, a search for ‘*Drug Name*
extrapulmonary right-to-left shunting across persistent foetal AND Persistent Pulmonary Hypertension of the Newborn’ was carried
channels because of the increased pulmonary vascular resis- out, for example ‘Treprostinil AND Persistent Pulmonary Hyperten-
tance/systemic vascular resistance ratio [1]. Echocardiography sion of the Newborn’. Searching for ‘Persistent Pulmonary Hyperten-
is the gold standard investigation for the diagnosis of PPHN sion of the Newborn’ in Cochrane Library gave nine items and using
the same search on Clinicaltrials.gov resulted in 31 items.
[8]. The echocardiographic findings are right ventricular
Study selection: Cf. the objective of this MiniReview, only studies
hypertrophy, deviation of the interventricular septum to the investigating treatments for PPHN without congenital diaphragmatic
left, tricuspid regurgitation and right-to-left or bidirectional hernia were included. To be included, the individual study should
shunting across patent foramen ovale, and patent ductus arte- clearly state the diagnosis of PPHN without congenital diaphragmatic
riosus [7]. hernia in the patient group investigated. Systemic reviews and meta-
Inhaled nitric oxide (iNO) is the only approved specific pul- analysis found on The Cochrane Library were excluded if no eligible
study had been found. Studies from Clinicaltrials.gov with the status
monary vasodilator for infants in the USA [9]. No literature
of ‘withdrawn’ or with an objective different from investigating a
describing other approved vasodilators has been found; how- treatment were excluded.
ever, literature describing clinical use of other treatments than
iNO has been found. It is a costly intervention, and approxi-
mately 30% of neonates with PPHN fail to respond to iNO. Therapy of PPHN
Some may even experience a rebound pulmonary hypertension General management.
due to suppression of endogenous nitric oxide production The general management of PPHN comprises the treatment of
[9,10]. Therefore, knowledge about the effects and the clinical hypoxaemia with oxygen supplementation and the underlying
use of other treatments than iNO may be important for the condition besides maintaining haemoglobin, glucose, intravas-
survival of the infant [7–9]. Some treatment options for PPHN cular volume, temperature and electrolytes at normal levels
have previously been described [9], but without a description [2,8]. High-frequency ventilation allows ventilation with small
of the quality of evidence or implications for future research tidal volumes. Combined with iNO in a randomized multi-cen-
for each treatment. This MiniReview provides the reader with tre trial, high-frequency ventilation was shown to be more suc-
an updated systematic overview of current available treat- cessful than treatment with high-frequency ventilation or iNO
ments. Furthermore, implications for future research for each alone [11]. This management may be considered in neonates
treatment will be discussed. Adverse events of each drug with PPHN before advancing to extra corporeal membrane
choice will be reviewed. This is important information in situa- oxygenation (ECMO). ECMO is an extremely complex and
tions where non-responders to iNO may be fatally challenged invasive technique providing life support in severe, but poten-
by adverse events of the treatment. tially reversible respiratory failure. The technique oxygenates
The objective of this MiniReview was to examine the cur- blood outside the body. Infants with PPHN are well suited to
rent available and emerging future treatment options for per- ECMO as the improved oxygenation and physiological stabil-
sistent pulmonary hypertension of the newborn without ity improves pulmonary blood flow without risking further
congenital diaphragmatic hernia. barotrauma [12]. When compared to conventional ventilation
support as presented in a Cochrane review by Mugford
Methods et al.[12], ECMO was shown to reduce mortality without an
Eligibility criteria: Following the Oxford Centre for Evidence-Based
increased risk of severe disability in infants with PPHN.
Medicine 2011 Levels of Evidence, all studies rated levels 1–5 were
considered eligible. As far as possible, eligible studies should be from
Inhaled nitric oxide.
2008 or newer, but if the available data were limited due to the low
prevalence of PPHN, older studies would be considered eligible, too. Nitric oxide (NO) is produced from the nitrogen of L-arginine
Information sources: PubMed – by the National Center for Biotech- and molecular oxygen by the endothelial nitric oxide synthase
nology Information, U.S. National Library of Medicine. The Cochrane enzyme (eNOS) in vascular endothelial cells. NO diffuses
Library – a collection of six databases that contain different types of through the endothelium to the vascular smooth muscle and
high-quality, independent evidence to inform healthcare decision-mak- activates soluble guanylyl cyclase (sGC) leading to the pro-
ing, and a seventh database that provides information about Cochrane duction of cyclic guanyl monophosphate (cGMP), which acti-
groups. Clinicaltrials.gov is a database of privately and publicly
funded clinical studies conducted around the world, run by U.S
vates the cGMP-dependent protein kinase or protein kinase G
National Library of Medicine. Pro.medicin.dk is a website and data- (PKG). PKG stimulates myosin phosphatase associated with
base containing information about drugs and therapeutic instructions. dephosphorylation of myosin light chain and activates potas-
It is issued by DLI A/S (Dansk Lægemiddel Information A/S). Pub- sium channels followed by hyperpolarization, and reduction of
Chem is an open chemistry database at the National Institutes of the influx of calcium. This leads to vasodilation. iNO acts
Health, collecting information on chemical structures, identifiers,
locally as pulmonary vasodilator (fig. 1). Thus, iNO is an
chemical and physical properties, biological activities, patents, health,
safety, toxicity data and many others. obvious choice in the treatment of PPHN.

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
394 JONAS PEDERSEN ET AL. MiniReview

Barrington et al.[6] compared iNO with control in neonates Currently, iNO is not believed to have systemic adverse
with PPHN by meta-analysing 17 randomized controlled trials events due to systemic inactivation by interactions with hae-
(RCTs). Participants were newborn infants (<1 month of age) moglobin [6] (table 1). That said, an interaction between iNO
with hypoxaemia suspected to be due to lung disease, pul- and haemoglobin may result in the creation of methaemoglo-
monary hypertension with right-to-left shunting or both. The bin. Methaemoglobin levels should be carefully monitored as
inclusion criterion was studies in term and near-term infants it is toxic and causes a left shift in the O2-Haemoglobin disso-
(>34 weeks of gestational age). Exclusions were infants with ciation curve resulting in worsening of hypoxia [13]. Signifi-
intracardiac shunting due to structural congenital heart disease. cant methaemoglobinaemia has been reported in a case of an
They reported no significant difference in death before hospi- overdose of iNO in a neonate [14]. Increased toxicity has not
tal discharge between the groups. A significant reduction in been shown after long-term treatment iNO. A reaction
the use of ECMO before hospital discharge was found, indi- between a superoxide and nitric oxide creates peroxynitrite,
cating that the number needed to treat with iNO for an addi- which may result in membrane lipid peroxidation. The forma-
tional beneficial outcome to prevent 1 neonate from requiring tion of the end product nitrite may induce bronchoconstriction
ECMO was 5. Oxygenation was significantly improved in and cough. Furthermore, NO inhibits platelet aggregation and
infants treated with iNO, and the oxygenation index 30– adhesion influencing the hemostasis.
60 min. after the start of treatment was significantly lower in
the iNO group. PaO2 30–60 min. after treatment was signifi-
Phosphodiesterase inhibitors.
cantly higher when treated with iNO. Furthermore, neonates
PDE 5 is responsible for breaking down cGMP in the smooth
who received iNO were not at increased risk of neurodevelop-
muscle cell. Inhibiting PDE 5 extends the duration of activity
mental sequelae and did not experience increased post-dis-
of cGMP resulting in relaxation of the smooth vascular muscle.
charge pulmonary complications. The meta-analysis reported
Thus, the inhibition of PDE 5 leads to vasodilation. Dipyri-
that clinical or echocardiographic evidence of PPHN was not
damole is a PDE 5 inhibitor but has shown significant systemic
present in all the included studies. Therefore, it was concluded
adverse effects in newborn lamb models with persistent pul-
that iNO should be considered for near-term infants with
monary hypertension [15]. Sildenafil (fig. 2A) is also a PDE 5
hypoxic respiratory failure with or without clear evidence of
inhibitor and has been studied in neonatal pig models of pul-
PPHN. The quality of evidence was rated moderate to high.
monary hypertension secondary to meconium aspiration, show-
To the author’s knowledge, iNO is the most investigated treat-
ing no adverse systemic effects and a vasodilating efficiency at
ment for PPHN.

Fig. 1. Pharmacodynamics of current and emerging treatments. PDE5, phosphodiesterase-5; PDE3, phosphodiesterase-3; GMP, guanylyl monophos-
phate; AMP, adenosine monophosphate; cGMP, cyclic guanylyl monophosphate; cAMP, cyclic adenosine monophosphate; sGC, soluble guanylyl
cyclase; AC, adenylyl cyclase; IP, PGI2-receptor; EP, PGE1-receptor; PGIS, prostacyclin synthase; COX, cyclooxygenase; eNOS, nitric oxide
synthase; ET-1, endothelin; rETB, relaxant endothelin receptor; cETB, contractile endothelin receptor and ETA, endothelin receptor; rhSOD,
recombinant human superoxide dismutase; L-arg., L-arginine; SNAT1, sodium-coupled neutral amino acid transporter.

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 395

least as good as iNO [16]. Sildenafil has become a common (table 1). Other known adverse events of PDE 5 inhibitors are
agent in the chronic management of pulmonary hypertension in visual, musculoskeletal, hearing and vestibular disorders [19].
adults [17] and is the most common enteral pulmonary The available literature regarding sildenafil as treatment of
vasodilator for children with pulmonary arterial hypertension PPHN is limited and low-rated regarding quality of evidence
[9]. Therefore, sildenafil may be a promising choice of treat- due to the imprecision of small studies and lack of methodologi-
ment for PPHN when iNO is not available (fig. 1). cal features. Despite the lack of literature, sildenafil is currently
Kelly et al.[10], a Cochrane review, investigated the efficacy used as acute adjuvant to iNO in NO-resistant PPHN and as an
and safety of sildenafil for treatment of PPHN. They included acute primary treatment of PPHN when iNO is not available or
term and near-term neonates (postnatal age <28 days after contraindicated [9]. Further RCTs investigating sildenafil should
reaching gestational age of 40 weeks) with either primary or be performed to increase the quality of evidence. Sildenafil
secondary PPHN. The included studies based the diagnosis on could either be investigated versus placebo or iNO. Double-
clinical findings with or without echocardiographic confirma- blinding of future studies would be preferred to raise the evi-
tion. Exclusion criteria were infants with structural genital heart dence, and the studies should be conducted in such manner that
disease with exception of patent foramen ovale and patent duc- follow-up will be possible for investigating long-term effects.
tus arteriosus. Sildenafil was compared to either placebo or no Tadalafil is also a PDE-5 inhibitor approved for use in
treatment, versus another pulmonary vasodilator, and sildenafil adults with pulmonary arterial hypertension and has larger
and another pulmonary vasodilator versus another pulmonary selectivity for PDE5 and longer half-life. One animal study,
vasodilator and placebo. This resulted in five included studies. investigating tadalafil in seven piglets with induced acute pul-
When compared to placebo, a significant reduction in mortality monary hypertension, showed a reduction in PVR and lung
in the sildenafil group was found based on three studies. No tri- shunt fraction as well as an increase in cardiac output and
als reported significant difference in mortality upon comparison arterial oxygenation, when oral tadalafil was administered
of the sildenafil group versus the active control group, or when [20]. No human study investigating tadalafil for use in PPHN
iNO was administered to both groups. Oxygenation index and has been found. One open-label study investigating tadalafil
PaO2 suggested steady improvement after the first sildenafil for use in 25 children with pulmonary arterial hypertension
dose, but without statistical significance. No adverse events has been found. Nineteen patients were administered tadalafil
were reported. In a small open-label study testing the pharma- and six patients had been on sildenafil for longer than
cokinetics of sildenafil in infants with PPHN, the following 6 months. A transition from sildenafil to tadalafil showed a
adverse events were reported: hypotension, development of significant clinical improvement. This study found that tadala-
patent ductus arteriosus with left-to-right shunting, pneumotho- fil may be more effective than sildenafil. The most frequent
rax, total anomalous pulmonary venous return, hypotension reported adverse event was nausea (table 1) [21]. Even though
associated with trisomy 21 and adrenal insufficiency [18] tadalafil has a low affinity for phosphodiesterase-6 in

Table 1.
Adverse event profiles.
Endothelin receptor
Modulation of NO/cGMP pathway Modulation of cAMP pathway antagonists Rho-kinase inhibitors
iNO: PDE 3 inhibitor:
• Cough • Hypotension
• Hepatotoxicity • Systemic hypotension
• Bronchoconstriction • Thrombocytopenia
• Nasopharyngitis
• Methaemoglobinaemia • Arrhythmias
• Headache
• Tolerance development Prostaglandin analogues:
• Flushing
• Change in haemostasis • Hypotension
• Peripheral oedema
PDE 5 inhibitor: • Drug tolerance
• Nausea • Nausea/vomiting
• Hypotension • Headache
• Hearing, visual, musculoskeletal, • Cough
and vestibular disorders • Jaw pain
• Adrenal insufficiency • Diarrhoea
• Pneumothorax • Catheter-related infections
• Patent ductus arteriosus • Fatigue
with right-to-left shunting • Skin symptoms
• Total anomalous pulmonary venous return • Thrombosis
• Hypotension associated • Flushing
with trisomy 21 • Infusion-site erythema and pain
sGC stimulator: • Pain in extremity
• Hypotension
• Headache
• Nasopharyngitis
• Peripheral oedema
• Musculoskeletal, gastrointestinal, vestibular
and haemorrhagic disorders

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
396 JONAS PEDERSEN ET AL. MiniReview

O CH3
A B H
N N O
O O N
N
S
N N
H N
N N
H3C O CH3

CH3
C D
O O O
S
OH NH O
O
H H N
N
N O

OH OH N OH

E
N
N
N F

N N

H2 N NH2
N O

Fig. 2. The chemical structures of sildenafil (A), milrinone (B), iloprost (C), bosentan (D) and riociguat (E).

photoreceptors, visual disturbances have been reported, too. events of hypotension, thrombocytopenia and arrhythmia have
Musculoskeletal, hearing and vestibular disorders have been been reported after the use of milrinone, but these were not
reported, but these are suggested to be less likely associated confirmed due to no reports of adverse events in the study
with tadalafil than sildenafil [19]. To investigate tadalafil in (table 1) [25]. Based on these data, milrinone should not be
PPHN, future studies must be conducted. interpreted as an alternative to iNO due to the lack of a con-
Phosphodiesterase-3 (PDE 3) is responsible for the break- trol group. Future RCTs should be conducted to investigate
down of cyclic adenosine monophosphate (cAMP) in vascular mortality, oxygenation index improvement or requirement for
smooth muscle cells and myocardium. By inhibiting PDE 3, ECMO. Double-blinding would be preferred as well as a long-
the intracellular cAMP level increases leading to vasodilation term follow-up for investigating sequelae.
and may increase cardiac contractility. Milrinone (fig. 2B) is a
PDE 3 inhibitor and vasodilator (fig. 1). In rat models of pul- Prostaglandin analogues.
monary hypertension, milrinone has shown to have an additive The arachidonic acid-prostacyclin pathway plays an important
pulmonary vasodilating effect when administered with iNO role in vascular pulmonary muscle tonus. By activating adenylyl
[22]. In lamb models with PPHN, milrinone exhibited an cyclase prostaglandins increase the intracellular cAMP concen-
enhancing effect of the vasodilation induced by prostacyclin. tration in vascular smooth muscle cells and lead to vasodilation
Thus, a combination therapy of milrinone and prostacyclin (fig. 1). Prostacyclin derivates (e.g. Iloprost; fig. 2C) are used
may be a new option of treatment for PPHN [23,24]. An to treat pulmonary arterial hypertension in adults.
open-label study [25] investigated milrinone in neonates with Janjindamai et al.[26] conducted a retrospective medical
PPHN and suboptimal response to iNO. Inclusion criteria were records review of 33 infants diagnosed with PPHN by echocar-
neonates with a gestational age larger than 34 weeks and <10 diography. They found that intravenous iloprost significantly
postnatal days of life, echocardiographic diagnosis of PPHN, reduced the oxygenation index. They showed that the need of
indwelling arterial line for obtaining blood samples, and inotropic support increased. When administered in adult patients
absence of a congenital heart defect excluding small atrial sep- with pulmonary arterial hypertension in an open-label study, the
tal defects and ventricular septal defects. A total of 11 neo- most common adverse events were headache and cough
nates were included. The study showed that the administration (table 1). One patient experienced hypotension [27]. The use of
of milrinone was associated with improvement in pulmonary aerosolised iloprost has shown a subsequent rapid tolerance
arterial hypertension and blood flow. Previously, adverse development to the vasodilatory effect [28].

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 397

In another retrospective study of 47 infants with PPHN pulmonary arterial hypertension, the most frequently reported
[29], iloprost seemed to be more effective than sildenafil in adverse events leading to treatment interruption were head-
terms of time to and duration of a clinical response. ache, diarrhoea, nausea, vomiting and pain in extremity
A retrospective cohort study [30] of 142 infants between (table 1) [50].
≤10 days and ≥34 weeks of gestation revealed the use of pros- Although prostaglandins are well-known pulmonary
taglandin E1 in infants with PPHN is significantly associated vasodilators, sufficient literature regarding treatment of PPHN
with a shortened length of hospitalization. No statistically sig- is non-existent.
nificant difference in mortality between PGE1-treated and non-
PGE1-treated infants was found. The final diagnosis of PPHN
was based on echocardiography. Endothelin receptor antagonists.
In a small phase I/II open-label clinical trial [31], 21 infants Endothelin-1 (ET-1) is a physiological potent vasoconstrictor
with PPHN born at >34 weeks of gestation at an age of produced by vascular endothelial cells. Endothelin-1 activates
<2 weeks were administered inhaled alprostadil, which is a smooth muscle ETA receptors and smooth muscle ETB recep-
PGE1 analogue. Echocardiography was not a required diagno- tors. The smooth muscle ETB receptor is well known as con-
sis for enrolment, and all infants were treated with surfactant tractile ETB receptor. The activation of both the ETA and
prior to enrolment. Alprostadil treatment revealed an improve- contractile ETB receptor leads to vasoconstriction. Further-
ment in PaO2 without toxic adverse events. No serious adverse more, endothelin-1 activates an endothelial ETB receptor well
events associated to alprostadil were reported. A RCT tried to known as relaxant ETB receptor. The activation of the
investigate this further but failed to include enough patients endothelial relaxant ETB receptors in large pulmonary arteries
[32]. In bovine coronary artery smooth muscle cells, prosta- is associated with release of NO and vasodilatation. Bosentan
glandin E2 has shown not to develop tolerance to antimito- (fig. 2D) is an ET-1 antagonist acting at both receptors
genic actions within 24 hr, as it was shown for iloprost (fig. 1). It has proven to be efficient in adults with pulmonary
[33,34]. When administered to patients with impaired renal hypertension [51]. In newborn lamb models, PPHN was asso-
function, alprostadil has shown adverse events as hypotension, ciated with increased ETA-mediated vasoconstriction and a
nausea, vomiting, fatigue and skin symptoms (table 1) [35]. loss of ETB-mediated vasodilation [52,53]. Furthermore, ele-
Epoprostenol is a prostacyclin analogue used for treatment vated plasma levels of ET-1 had been reported in newborn
of pulmonary arterial hypertension in adults. The found data infants with pulmonary hypertension [54,55]. Therefore, the
regarding the use of epoprostenol in neonates with PPHN are use of bosentan or other endothelin receptor antagonists to
limited to two old and small studies and case reports. The two treat PPHN may be an effective alternative to iNO.
studies were consistent in reporting beneficial outcome of the More et al.[56] identified in a systematic review of random-
use of epoprostenol in neonates with PPHN [36,37]. When ized, cluster-randomized or quasi-randomized controlled trials
administered in healthy adults, the most common adverse two studies that evaluated the efficacy and safety of endothelin
event of epoprostenol was headache [38]. When administered receptor antagonists in term and late pre-term infants with
intravenously in adults with pulmonary arterial hypertension, PPHN. Both studies reported that bosentan was well tolerated
adverse events of jaw pain, diarrhoea, catheter-related infec- and that there were no major adverse events. The problem
tions and thrombosis have been reported (table 1) [39]. Drug with the studies investigating PPHN is the low amounts of
tolerance was reported for continuous intravenous treatment patients. Thus, more rare and serious adverse events are not
with epoprostenol in pulmonary hypertension [40]. detected. In adults, treatment with bosentan for pulmonary
Treprostinil is a known prostacyclin analogue. The data on arterial hypertension has shown to cause an abnormal liver
the use of treprostinil are limited. Two case reports described function and elevations of alanine aminotransferase and aspar-
the administration of treprostinil in two neonates with early tate aminotransferase, indicating hepatotoxicity [57–59].
onset sepsis and PPHN who did not respond to iNO. Within Adverse events of nasopharyngitis have also been reported in
6–12 hr after the treatment with treprostinil, both patients patient groups treated with bosentan [59]. Other reported
showed clinical improvement. No systemic adverse events adverse events are headache, flushing and peripheral oedema
were observed [41]. When administered intravenously in (table 1) [60].
adults with pulmonary arterial hypertension, the most common In the first study, bosentan was used as the sole pulmonary
reported adverse events were infusion-site erythema and pain vasodilator. iNO and ECMO were not available in the setting.
[42]. When administered orally in adults, adverse events were Mohamed et al.[61] reported no significant difference in death
headache, diarrhoea, nausea, flushing and jaw pain (table 1) from any cause prior to hospital discharge or prior to 28 days
[43]. of life between bosentan-treated infants and a placebo control
Selexipag, a prostacyclin receptor agonist, is an orally avail- group. The infants treated with bosentan showed a significant
able prodrug which has shown significantly improvements in benefit when comparing at least 20% improvement in oxy-
the treatment of pulmonary arterial hypertension in adults genation index within 72 hr of treatment to the placebo con-
[44,45]. Thus, selexipag may have a future potential in the trol group. Furthermore, the bosentan group showed a
treatment of PPHN [46–49]. No animal studies investigating significant benefit in improved oxygenation index by at least
selexipag in PPHN have been found. In a large double-blinded 20% at the end of the treatment. The total duration of mechan-
and randomized global study testing selexipag used in ical ventilation was significantly lower in the bosentan group.

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
398 JONAS PEDERSEN ET AL. MiniReview

There was no difference in development of cerebral palsy or of 3 years. In adult patients with chronic thromboembolic pul-
hearing impairment between the groups. However, a trend to a monary hypertension, the most common adverse events of
higher incidence of other adverse neurodevelopmental seque- treatment with riociguat were hypotension, headache,
lae was found in the placebo group. They did not report the nasopharyngitis and peripheral oedema [74,75]. Other reported
length of hospitalization, other adverse events or other infor- adverse events of riociguat are musculoskeletal, gastrointesti-
mation on cognitive and neurodevelopmental disability at 18– nal, vestibular and haemorrhagic disorders (table 1) [19].
24 months. This study had no available iNO for treatment. No Other known compounds that simulate sGC are YC-1, BAY
hepatotoxicity was reported. 41-2272, BAY 41-8543, CFM-1571 and A-350619. Known
In the FUTURE-4 trial, a multi-centre study where iNO and compounds that activate sGC are BAY 58-2667 and HMR-
ECMO were available, bosentan was used as an adjunct ther- 1766 (related derivative: S-3448) [76]. In neonatal lamb mod-
apy to iNO. Steinhorn et al.[62] reported no significant differ- els with PPHN, sGC activators has been shown to cause a sig-
ence in the requirement for ECMO prior to hospital discharge nificantly greater fall in PVR than either 100% O2 or iNO
between the groups. There was no significant difference in [77,78]. Furthermore, a study in foetal lambs has indicated an
duration of iNO therapy or total duration of mechanical venti- impaired sGC activity as a contributing factor in the pathogen-
lation. The authors reported that bosentan did not affect sys- esis of PPHN [77].
temic blood pressure adversely, but no details were given. No These findings suggest that sGC stimulators and activators
hepatotoxicity was reported. may have a therapeutic potential as alternative or adjuvant
However, for both studies, the overall quality rating of evi- therapy for infants with PPHN who are non-responsive to iNO
dence was low due to small sample size of the included stud- and O2. By conducting a double-blinded RCT in the future,
ies, the numerical imbalance between the groups due to the efficacy of riociguat in the treatment of PPHN without
randomization and attrition, and unclear risk of bias on some CDH could be enlightened. A subsequent follow-up would be
of the important domains [56]. More RCTs regarding the effi- necessary to evaluate long-term effects.
cacy and safety of bosentan as alternative to iNO in the treat-
ment for PPHN should be conducted.
Rho-kinase inhibitors.
Macitentan is a new dual ETRA showing potential in the
RhoA is a small GTPase increasing Rho-kinase activity lead-
treatment of pulmonary arterial hypertension. Macitentan has
ing to phosphorylation of myosin light chain and vasoconstric-
been reported to have less adverse effects e.g. hepatotoxicity
tion in vascular smooth muscle (fig. 1). Rho-kinase activity
compared to bosentan (table 1); therefore, macitentan may
has shown to modulate contraction of vascular smooth muscle
have a future potential in the treatment of PPHN [63–65].
cells in the systemic and pulmonary circulation [79]. In foetal
That said, one case report describing serious liver injury after
lambs, Rho-kinase activity has been shown to maintain the
macitentan treatment for adult pulmonary arterial hypertension
high PVR [80].
has been found [66], indicating the importance of registering
The RhoA-Rho-kinase pathway is involved in vasoconstric-
adverse events of ETRAs in future studies.
tion, when eNOS is inhibited which may indicate Rho-kinase
Ambrisentan is an ETRA used in the treatment of pul-
inhibitors as a future treatment [80]. Furthermore, increased
monary arterial hypertension in functional WHO class II–III
Rho-kinase activity was found to lead to impaired angiogene-
[67,68]. No studies investigating ambrisentan in neonates with
sis in foetal lambs [79]. Therefore, the activity of the Rho-
PPHN have been found. In an ongoing study (NCT01406327)
kinase may be a potential future target for treatment of PPHN.
of 702 patients with pulmonary arterial hypertension treated
A newer animal study using foetal lambs with PPHN has sug-
with ambrisentan, interim results have shown that common
gested interactions between the Rho-kinase activity and perox-
adverse events are anaemia, peripheral oedema, headache, face
isome proliferator-activated receptor-c (PPAR-c). By
oedema, abnormal hepatic function and epistaxis [69].
inhibiting the Rho-kinase in foetal lambs with decreased
PPAR-c activity, the PPAR-c activity was restored to normal.
Soluble guanylyl cyclase activators/stimulators. These findings suggest that an inhibition of the Rho-kinase
As previously described, iNO acts on sGC, but resistance to combined with activation of PPAR-c results in a restored
NO and tolerance may limit the vasodilation. Thus, by activat- growth of pulmonary arterial smooth muscle cells [81]. Com-
ing sGC directly with an activator, a relaxation of vascular pounds inhibiting the Rho-kinase are Y-27632 and fasudil
smooth muscle cells may occur independently of NO (fig. 1). (HA-1077) [80]. Fasudil is registered for the treatment of pul-
The sGC stimulator riociguat (fig. 2E) (BAY 63-2521 [70]) is monary arterial hypertension in Japan. Infused fasudil has
used for pulmonary arterial hypertension as monotherapy or in shown to improve pulmonary hemodynamics without signifi-
combination with ETRA or prostanoids [71]. Information cant toxicity [82]. Inhaled fasudil has shown to be at least as
regarding the use of riociguat in children with pulmonary arte- effective as iNO [83]. Administered in a double-blinded, pla-
rial hypertension is missing [71,72]. No studies investigating cebo-controlled clinical trial, fasudil hydrochloride (AT-
sGC activators/stimulators as treatment of PPHN in neonates 877ER) was shown to improve hemodynamics in adult
were found. Spreemann et al.[73] reported positive results in a patients with pulmonary arterial hypertension [84]. No studies
case using oral riociguat in combination with bosentan in a investigating fasudil in PPHN have been found. Fasudil is also
child with supra-systemic pulmonary hypertension at the age used for preventing cerebral vasospasms in patients

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 399

undergoing surgery for subarachnoid haemorrhage and when recoupling of eNOS which improves NO production [94]. In
investigated versus placebo in these patients, no significant piglet models, L-citrulline is transported into pulmonary arte-
difference in adverse events was found [85]. Some studies rial endothelial cells (PAEC) by a sodium-coupled neutral
report no clinical significant systemic hypotension caused by amino acid transporter (SNAT1). The activity of SNAT1 mod-
fasudil [82,86,87] whereas a newer animal study has con- ulates the L-citrulline-induced NO production, which increases
cluded fasudil is not a useful targeting drug due to a signifi- in hypoxic PAEC due to an increased expression of SNAT1
cant systemic hypotension (table 1) [88]. No larger clinical within 24, 48 and 72 hr of hypoxic conditions in piglets
trial investigating fasudil has been found. Future studies of [95,96]. Furthermore, it has been demonstrated that an
fasudil could determine the profile of adverse events. Fasudil impaired L-citrulline-L-arginine-NO pathway is involved in
has a potential in the treatment of pulmonary arterial hyperten- the pathogenesis of pulmonary hypertension [97]. Another
sion, and future research should address the effect of fasudil study in piglet models has shown that rescue treatment with
versus placebo or iNO in PPHN in a large double-blinded L-citrulline improves pulmonary hypertension induced by
RCT. hypoxia [94]. These findings suggest that by increasing NO
production, supplemental L-citrulline and modulators of
SNAT1 may be emerging alternate therapies for infants with
Clinical trials.
PPHN. In 25 adult patients with idiopathic pulmonary hyper-
Clinical trials concerning the pharmaceutical treatment on PPHN
tension or Eisenmenger syndrome, oral L-citrulline has shown
can be found on Clinicaltrials.gov. Details regarding studies reg-
a reduction in PVR [98]. No clinical trials regarding L-citrul-
istered on Clinicaltrials.gov are presented in table 2 for sildenafil,
line use in infants had been performed until today.
bosentan, prostaglandins, milrinone and for iNO. Most of these
studies aim to assess the safety and treatment effect of the drugs
in infants with PPHN to find optimal starting doses as well as PPAR-c agonists.
therapy timescales. Further, some trials in adults have investi- PPAR-c regulates smooth muscle cell proliferation and inhi-
gated an additive effect of combining prostacyclins with silde- bits vascular remodelling (fig. 1). In a mouse model, it has
nafil or bosentan. First results seem to be promising with been shown that a selective deletion of the PPAR-c gene
improvement in hemodynamic parameters [43]. Emerging targets results in spontaneously developed pulmonary arterial hyper-
and therapies for PPHN are currently under investigation. tension [99]. Furthermore, activation of PPAR-c resulted in a
Table 3 summarizes implications of future research. decreased proliferation of pulmonary arterial smooth muscle
cells in vitro [99]. In foetal lamb models with PPHN, ET-1
has shown to decrease PPAR-c signalling which contributes to
Future Treatments
dysfunction of pulmonary arterial endothelial cells [100]. In
This section will describe possible future treatments based on the same study, PPAR-c agonists increased eNOS activity in
findings in animal studies. These compounds could be normal PAEC and in PAEC from lambs with PPHN. These
researched in future phase 0–1 studies. Some drugs are used findings may indicate that a combination of PPAR-c agonists
in other treatments, for example rosiglitazone in diabetes mel- and bosentan could be a future treatment option for PPHN.
litus type II meaning that some adverse events and pharma- Currently, no studies involving human beings had been con-
cokinetics already are known. ducted. As described in the section above, PPAR-c agonists
may be a future treatment combined with Rho-kinase inhibi-
tors. Compounds that act as PPAR-c agonists are rosiglitazone
Antioxidants.
and 15D-prostaglandin-J2 [100].
Mechanical ventilation as treatment option for PPHN may
result in formation of reactive oxygen species which can inac-
tivate NO and cause further vasoconstriction (fig. 1). In foetal Discussion
lamb models with PPHN, intratracheal recombinant human
According to the literature, sildenafil is the most investigated
superoxide dismutase (rhSOD) has shown to improve oxy-
drug together with iNO in the treatment of PPHN. Some clini-
genation and reduce oxidation and vasoconstriction [89]. In
cal trials investigating the treatment of PPHN can be found on
another study, inhaled rhSOD was shown to enhance the
Clinicaltrials.gov [101] (table 2). Six studies investigated the
effect of iNO in foetal lambs with PPHN [90]. In foetal lambs,
use of sildenafil either as monotherapy compared to placebo,
rhSOD has also been shown to restore eNOS coupling [91].
or as adjuvant therapy to iNO. But sildenafil has the disadvan-
These findings indicate that rhSOD could be a future therapy
tage of requiring endogenous NO or natriuretic peptides to
adjuvant to iNO for PPHN.
work in the pulmonary circulation.
The development of increased pulmonary vascular resis-
L-citrulline. tance may rely on malfunctioning intracellular pathways due
L-citrulline is a L-arginine-NO precursor. Through a two-step to genetic variations, for example a malfunction of eNOS.
enzymatic process, L-citrulline is converted to L-arginine Therefore, other treatments instead of sildenafil should be
which is directly channelled to eNOS for efficient NO produc- investigated. For instance, sGC activators may be promising
tion [92,93] (fig. 1). Furthermore, L-citrulline stimulates the drugs, because they are potentially better than sildenafil when

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
400 JONAS PEDERSEN ET AL. MiniReview

Table 2.
An overview of clinical trials registered at Clinicaltrials.gov investigating sildenafil, bosentan, prostaglandins, milrinone or iNO in PPHN.
Drug Title Design Objective Status
Sildenafil A multi-centre, randomized, placebo- Interventional, estimated This study will evaluate whether Recruiting
controlled, double-blind, two-armed, enrolment: 64, randomized, IV sildenafil can reduce the
parallel group study to evaluate efficacy masking: double, phase III time on iNO treatment and
and safety of iv sildenafil in the treatment reduce the failure rate of
of neonates with PPHN or hypoxic available treatments for PPHN
respiratory failure and at risk for PPHN,
with a long-term follow-up investigation
of developmental progress 12 and
24 months after completion of study
treatment.
ID: NCT01720524
Oral Sildenafil in Persistent Pulmonary Interventional, actual The purpose of this study was to Recruitment
Hypertension Secondary to Meconium enrolment: 96, randomized, study the role of oral sildenafil completed
Aspiration Syndrome in Newborns: A parallel assignment, masking: in PPHN secondary to
Randomized Placebo Controlled Trial. single, phase IV meconium aspiration syndrome
ID: NCT01757782 [102] (MAS) in newborns and to
study risk factors of MAS
developing into PPHN
Pharmacokinetics of Sildenafil in Premature Interventional actual The purpose of this study was to Recruitment
Infants enrolment: 34 non- learn more about the safety and completed
ID: NCT01670136 randomized single group dosing of sildenafil in infants
assignment open label phase 1
A Single Arm Single Centre Study To Interventional, actual The purpose of this study was to Terminated
Investigate Safety And Efficacy Of enrolment: 4, single group learn more about the safety and
Sildenafil In Near Term and Term assignment, open label, phase dosing of sildenafil in infants
Newborns With Persistent Pulmonary II
Hypertension Of The Newborn
ID: NCT01069861
Phase II Trial of Sildenafil in Newborns Interventional, actual The purpose of this study was to Terminated
With Persistent Pulmonary Hypertension enrolment: 4, randomized, determine whether intravenous
ID: NCT01409031 parallel assignment, masking: sildenafil reduces pulmonary
triple, phase II artery pressure and improves
oxygenation in near-term and
term infants with PPHN
A Follow-Up Investigation For Patients Observational, actual This study would monitor Completed
Completing Study A1481276 To enrolment: 1, prospective, developmental progress of
Investigate Developmental Progress 12 phase II PPHN patients for 2 years
and 24 Months Following Completion Of following study treatment,
Sildenafil Treatment using Bayley III and
ID: NCT01801982 Hammersmith tools
Bosentan Multi-centre, Double-Blind, Placebo- Interventional, actual The study was a phase 3 study Terminated
Controlled, Randomized, Prospective enrolment: 23, randomized, to investigate whether adding
Study of Bosentan as Adjunctive Therapy parallel assignment, m bosentan to iNO in newborns
to Inhaled Nitric Oxide in the asking: quadruple, phase III with PPHN was a supporting
Management of Persistent Pulmonary and safe therapy and to
Hypertension of the Newborn (PPHN) evaluate pharmacokinetics of
(FUTURE 4) bosentan and its metabolites
ID: NCT01389856 [62]
Prostaglandins Intravenous Remodulin (Treprostinil) as Interventional, estimated This study aims to assess the Recruiting
Add-on Therapy for the Treatment of enrolment: 70, randomized, safety and treatment effect of
Persistent Pulmonary Hypertension of the parallel assignment, masking: acute dosing with IV remodulin
Newborn: A Randomized, Placebo- quadruple, phase II in neonates with PPHN
Controlled, Safety and Efficacy Study.
ID: NCT02261883
Pilot Study: The Effect of Inhaled Iloprost Interventional, actual The purpose of this study was to Terminated
on Oxygenation in Term and Near Term enrolment: 1, single group test safety and efficacy of
infants With Pulmonary Hypertension. assignment, open label, phase inhaled iloprost in reducing
Testing Two Doses. IV pulmonary artery pressure in
ID: NCT00409526 near-term infants with PPHN

(continued)

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 401

Table 2. (continued)

Drug Title Design Objective Status


Milrinone Milrinone in Congenital Diaphragmatic Interventional, estimated This pilot trial determines Recruiting
Hernia enrolment: 66, randomized, whether milrinone infusion in
ID: NCT02951130 [103] parallel assignment, masking: neonates with CDH would lead
quadruple, phase II to an increase in PaO2 with a
corresponding decrease in OI
by itself or in conjunction with
other pulmonary vasodilators
24 hr post-infusion
Milrinone Pharmacokinetics and Interventional, actual The purpose of this pilot study Terminated
Pharmacodynamics in Newborns with enrolment: 12, randomized, was to determine a safe dose of
Persistent Pulmonary Hypertension of the parallel assignment, masking: milrinone to use in a larger
Newborn – a Pilot Study to Enable a single study of babies with PPHN
Randomized Trial of Intervention
ID: NCT01088997
iNO Effect of Early iNO on Oxidative Stress, Interventional, estimated The purpose of this study is to Recruiting
Vascular Tone and Inflammation in Term enrolment: 34, randomized, evaluate if giving the inhaled
and Late-Preterm Infants With Hypoxic parallel assignment, masking: nitric oxide earlier in the
Respiratory Failure triple, phase IV course of disease improves the
ID: NCT01891500 effectiveness of the drug,
reduces the amount of cellular
injury from oxygen exposure,
and decreases the total amount
of time a patient requires
supplemental oxygen
Early Inhaled Nitric Oxide Therapy in Term Interventional, actual This study tested whether Terminated
and Near Term Infants With Respiratory enrolment: 302, randomized, initiating iNO therapy earlier
Failure parallel assignment, masking: would reduce death and reduce
ID: NCT00005773 [104] quadruple, phase III the use of ECMO compared to
standard recommendation
threshold
The Randomized Inhaled Nitric Oxide Interventional, actual The primary objective is to Terminated
Study (NINOS) in Full-Term and Nearly enrolment: 235, randomized, determine whether inhaled
Full-Term Infants With Hypoxic parallel assignment, masking: nitric oxide would reduce
Respiratory Failure quadruple, phase III mortality or the initiation of
ID: NCT00005776 [105] extracorporeal membrane
oxygenation in infants with
hypoxic respiratory failure
NO Need to Ventilate: A Trial of Non- Interventional, estimated The primary objective of the Completed
invasive iNO in Persistent Pulmonary enrolment: 400, randomized, trials is to determine the
Hypertension of the Newborn single group assignment, feasibility and clinical safety
ID: NCT00139217 open label and efficacy of non-invasive
inhaled iNO in infants with
PPHN without significant
pulmonary parenchymal disease
who would normally receive
iNO only after placement of a
tracheal tube and the institution
of mechanical ventilation
Inhaled Nitric Oxide in Neonates With Interventional, actual This study was to determine of Completed
Elevated A-aDO2 Gradients Not Requiring enrolment: 8, randomized, iNO given into an oxygen
Mechanical Ventilation masking: triple, phase IV hood is effective in improving
ID: NCT00732537 [106] oxygenation in term and near-
term infants who have poor
oxygenation but who are not
yet mechanically ventilated

eNOS is malfunctional. No studies on Clinicaltrials.gov cur- investigate genetic variations which may contribute to devel-
rently have investigated sGC activators. One study on Clini- opment of targeted treatment for PPHN. The other drugs that
caltrials.gov investigated a specific PTGS1 genetic variation are currently studied on Clinicaltrials.gov are bosentan, milri-
and the risk of developing PPHN. Future research should none, iNO and prostaglandins. No other treatments under

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
402 JONAS PEDERSEN ET AL. MiniReview

Table 3.
Treatments in need of research and implications for future research regarding PPHN.
Treatments Implications for future research
Sildenafil
Milrinone • Mortality compared to placebo or iNO
Prostaglandins • Requirement for ECMO compared to placebo or iNO
Bosentan • OI Improvements within, for example 24, 48 and 72 hr of administration compared to placebo or iNO
sGC activators/stimulators • Adverse effects compared to placebo or iNO
rhSOD • Long-term sequelae
• Methodological information to increase quality of evidence
L-citrulline
• Information about whether infants have received surfactant or supportive care affecting primary outcome.
Rho-kinase inhibitors
PPAR-c agonists
• PPHN with CDH should be investigated separately

investigation have been found. This MiniReview and the find- technique which has shown to reduce mortality when used for
ings on Clinicaltrials.gov are consistent in pointing out that treating PPHN. iNO is the most investigated choice of treat-
implications for future research are to investigate every aspect ment and the only approved specific pulmonary vasodilator
of each described current and future treatment for PPHN for newborns. iNO should be considered for infants with
except iNO (fig. 1 and table 3). More double-blinded RCTs hypoxaemic respiratory failure with or without PPHN; 30% of
should be conducted, focusing on mortality, requirement for the patients are non-responsive to iNO. Literature reports the
ECMO, frequently measured oxygenation index improvements use of sildenafil, prostaglandins, milrinone and bosentan as
compared to iNO within the first days of administration, alternative treatment to iNO. These drug choices have different
adverse events and long-term sequelae especially neurodevel- profiles of adverse events (table 3). A common adverse event
opmental. The overall problem with describing adverse effects for each drug is hypotension, which may be unwanted in a
in the treatment of PPHN is the low prevalence, which may hypoxic infant with PPHN due to the risk of developing shock
result in an underestimation of adverse effects due to a small and hypoperfusion. On Clinicaltrials.gov, sildenafil is the most
sample size. Nevertheless, some adverse events to the treat- investigated non-iNO treatment. Animal studies have proven
ments have been reported. The adverse event profiles are sum- that other treatments of PPHN may emerge in the future.
marized in table 1. These adverse event profiles indicate that These promising choices consist of sGC activators and stimu-
some drugs may be superior to others in terms of adverse lators, rhSOD, L-citrulline, Rho-kinase inhibitors and PPAR-c
events when treating an infant with PPHN, who is a non- agonists. sGC activators and stimulators and Rho-kinase inhi-
responder to iNO. Hypotension is the common adverse event bitors are known in the treatment of pulmonary hypertension
of each drug; thus, blood pressure should be monitored to pre- in adults. This MiniReview and the findings on Clinicaltrials.-
vent development of shock and hypoperfusion in the hypoxic gov are consistent in pointing out that implications for future
infant. Future studies with sufficient sample sizes should research are to investigate every aspect of each one of the pre-
ensure to investigate adverse event profiles as secondary aim viously described current and future treatment options for
of the intervention drugs, as this is important knowledge for PPHN, except from iNO (fig. 1). The treatments and sug-
choosing a treatment alternative to iNO. To investigate the gested implications for future research are summarized in
adverse events of drugs used in the treatment of PHHN, we table 3. We suggest making a multi-centre global database for
suggest making a systematic database for registering the registering adverse events of treatments for PPHN to avoid
adverse events of treatments in PPHN. This would be an underestimations due to the low prevalence.
opportunity to investigate PPHN properly even though the
prevalence is low. Methodological information should be
Outlook
detailed to raise the quality of evidence if the RCTs are
included in future systematic reviews and meta-analyses. There are many small studies, and we lack a systematic and
Researchers should mention whether infants have received sur- prioritized effort to perform larger clinical studies in this area.
factant or not and be aware that PPHN with CDH should be One problem future researchers may encounter will be the
investigated separately (table 3). To evaluate long-term effects, long period of recruitment for a sufficient power due to the
for example neurodevelopment, follow-up is required. low incidence of PPHN. The use of sildenafil, prostaglandins,
milrinone and bosentan for PPHN as treatment alternate to
iNO is commonly reported. Current literature lacks sufficiently
Conclusions
conducted double-blinded RCTs investigating these choices
Nowadays, the general management of PPHN relies on treat- with a sufficient sample size and long-term follow-up. If
ing hypoxaemia and the underlying condition besides main- future RCTs testing these drugs are conducted, new beneficial
taining haemoglobin, glucose, intravascular volume, or adverse effects may be reported in the treatment of PPHN.
temperature and electrolytes. Infants with PPHN have shown These drugs could be tested either versus placebo or iNO.
to respond positively to the treatment of high-frequency venti- This will improve the possible choices of treatment when neo-
lation combined with iNO. ECMO is a complex and invasive nates with PPHN are non-responders to iNO. sGC activators

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 403

and stimulators, rhSOD, L-citrulline, Rho-kinase inhibitors pulmonary hypertension. Am J Respir Crit Care Med
and PPAR-c agonists all seem promising future choices based 2002;165:1098–102.
17 Wardle AJ, Tulloh RM. Paediatric pulmonary hypertension and
on known use in pulmonary arterial hypertension and animal
sildenafil: current practice and controversies. Arch Dis Child
models with PPHN. These treatments are summarized in Educ Pract Ed 2013;98:141–7.
table 3 with implications of future research. 18 Steinhorn RH, Kinsella JP, Pierce C, Butrous G, Dilleen M, Oakes
sGC stimulators as riociguat are already used as either M et al. Intravenous sildenafil in the treatment of neonates with
monotherapy or in combination with ETRA in the treatment persistent pulmonary hypertension. J Pediatr 2009;155:841–7 e1.
of arterial pulmonary hypertension in adults [76]. No paedi- 19 Khouri C, Lepelley M, Roustit M, Montastruc F, Humbert M,
Cracowski JL. Comparative safety of drugs targeting the nitric
atric studies using riociguat in the treatment of PPHN were
oxide pathway in pulmonary hypertension: a mixed approach
performed. combining a meta-analysis of clinical trials and a disproportional-
References ity analysis from the world health organization pharmacovigilance
database. Chest 2017, Doi: 10.1016/j.chest.2017.12.008.
1 Abman SH, Benitz WE, Rowitch D, Polin RA. Fetal and neonatal 20 Tessler RB, Zadinello M, Fiori H, Colvero M, Belik J, Fiori RM.
physiology. In: Laskminrusimha S, Steinhorn RH, editors. Fetal Tadalafil improves oxygenation in a model of newborn pul-
and Neonatal Physiology. Elsevier Health Science; Elsevier monary hypertension. Pediatr Crit Care Med 2008;9:330–2.
Health Sciences; Elsevier, New York, USA, 2016;1576–88. 21 Shiva A, Shiran M, Rafati M, Zamani H, Babazadeh K, Saeedi
2 Gleason CA, Devaskar SU, Avery ME. Avery’s diseases of the M et al. Oral tadalafil in children with pulmonary arterial hyper-
newborn. In: Steinhorn RH, Abman SH, editors. Avery’s Diseases tension. Drug Res (Stuttg) 2016;66:7–10.
of the Newborn. Saunders, Elsevier Health Sciences, Elsevier, 22 Deb B, Bradford K, Pearl RG. Additive effects of inhaled nitric
New York, USA, 2012;732–40. oxide and intravenous milrinone in experimental pulmonary
3 Storme L, Aubry E, Rakza T, Houeijeh A, Debarge V, Tourneux hypertension. Crit Care Med 2000;28:795–9.
P et al. Pathophysiology of persistent pulmonary hypertension of 23 Lakshminrusimha S. Milrinone enhances relaxation to prostacy-
the newborn: impact of the perinatal environment. Arch Cardio- clin and iloprost in pulmonary arteries isolated from lambs with
vasc Dis 2013;106:169–77. persistent pulmonary hypertension of the newborn. Pediatr Crit
4 Chambers CD, Hernandez-Diaz S, Van Marter LJ, Werler MM, Care Med 2009;10:106–12.
Louik C, Jones KL et al. Selective serotonin-reuptake inhibitors 24 Kumar VH, Swartz DD, Rashid N, Lakshminrusimha S, Ma C,
and risk of persistent pulmonary hypertension of the newborn. N Ryan RM et al. Prostacyclin and milrinone by aerosolization
Engl J Med 2006;354:579–87. improve pulmonary hemodynamics in newborn lambs with exper-
5 Alano MA, Ngougmna E, Ostrea EM Jr, Konduri GG. Analysis imental pulmonary hypertension. J Appl Physiol 2010;109:677.
of nonsteroidal antiinflammatory drugs in meconium and its rela- 25 McNamara PJ, Shivananda SP, Sahni M, Freeman D, Taddio A.
tion to persistent pulmonary hypertension of the newborn. Pedi- Pharmacology of milrinone in neonates with persistent pulmonary
atrics 2001;107:519–23. hypertension of the newborn and suboptimal response to inhaled
6 Barrington KJ, Finer N, Pennaforte T, Altit G. Nitric oxide for nitric oxide. Pediatr Crit Care Med 2013;14:74–84.
respiratory failure in infants born at or near term. Cochrane Data- 26 Janjindamai W, Thatrimontrichai A, Maneenil G, Chanvitan P,
base Syst Rev 2017;1:CD000399. Dissaneevate S. Effectiveness and safety of intravenous iloprost
7 Mathew B, Lakshminrusimha S. Persistent pulmonary hyperten- for severe persistent pulmonary hypertension of the newborn.
sion in the newborn. Children (Basel) 2017;4:63. Indian Pediatr 2013;50:934–8.
8 Bendapudi P, Rao GG, Greenough A. Diagnosis and management 27 Saji T, Myoishi M, Sugimura K, Tahara N, Takeda Y, Fukuda K
of persistent pulmonary hypertension of the newborn. Paediatr et al. Efficacy and safety of inhaled iloprost in japanese patients
Respir Rev 2015;16:157–61. with pulmonary arterial hypertension- insights from the IBUKI
9 Lakshminrusimha S, Mathew B, Leach CL. Pharmacologic strate- and AIR studies. Circ J 2016;80:835–42.
gies in neonatal pulmonary hypertension other than nitric oxide. 28 Schermuly RT, Schulz A, Ghofrani HA, Breitenbach CS, Weiss-
Semin Perinatol 2016;40:160–73. mann N, Hildebrand M et al. Comparison of pharmacokinetics
10 Kelly LE, Ohlsson A, Shah PS. Sildenafil for pulmonary hyperten- and vasodilatory effect of nebulized and infused iloprost in exper-
sion in neonates. Cochrane Database Syst Rev 2017;8:Cd005494. imental pulmonary hypertension: rapid tolerance development. J
11 Kinsella JP, Truog WE, Walsh WF, Goldberg RN, Bancalari E, Aerosol Med 2006;19:353–63.
Mayock DE et al. Randomized, multicenter trial of inhaled nitric 29 Kahveci H, Yilmaz O, Avsar UZ, Ciftel M, Kilic O, Laloglu F et al.
oxide and high-frequency oscillatory ventilation in severe, persis- Oral sildenafil and inhaled iloprost in the treatment of pulmonary
tent pulmonary hypertension of the newborn. J Pediatr hypertension of the newborn. Pediatr Pulmonol 2014;49:1205–13.
1997;131:55–62. 30 Gupta N. Prostaglandin E1 use during neonatal transfer: potential
12 Mugford M, Elbourne D, Field D. Extracorporeal membrane oxy- beneficial role in persistent pulmonary hypertension of the new-
genation for severe respiratory failure in newborn infants. born. Arch Dis Child Fetal Neonatal Ed 2013;98:F186–8.
Cochrane Database Syst Rev 2008;16:CD001340. 31 Sood BG, Delaney-Black V, Aranda JV, Shankaran S. Aeroso-
13 Gupta K, Jha M, Jadon RS, Sood R. Case of methaemoglobi- lized PGE1: a selective pulmonary vasodilator in neonatal hypox-
naemia caused by tree oils and kerosene. BMJ Case Rep 2017, emic respiratory failure results of a Phase I/II open label clinical
Doi: 10.1136/bcr-2017-220802. trial. Pediatr Res 2004;56:579–85.
14 Heal CA, Spencer SA. Methaemoglobinaemia with high-dose 32 Sood BG, Keszler M, Garg M, Klein JM, Ohls R, Ambalavanan
nitric oxide administration. Acta Paediatr 1995;84:1318–9. N et al. Inhaled PGE1 in neonates with hypoxemic respiratory
15 Dukarm RC, Morin FC 3rd, Russell JA, Steinhorn RH. Pul- failure: two pilot feasibility randomized clinical trials. Trials
monary and systemic effects of the phosphodiesterase inhibitor 2014;15:486.
dipyridamole in newborn lambs with persistent pulmonary hyper- 33 Zucker TP, Bonisch D, Hasse A, Grosser T, Weber AA, Schror
tension. Pediatr Res 1998;44:831–7. K. Tolerance development to antimitogenic actions of prostacyclin
16 Shekerdemian LS, Ravn HB, Penny DJ. Intravenous sildenafil but not of prostaglandin E1 in coronary artery smooth muscle
lowers pulmonary vascular resistance in a model of neonatal cells. Eur J Pharmacol 1998;345:213–20.

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
404 JONAS PEDERSEN ET AL. MiniReview

34 Grosser T, Bonisch D, Zucker TP, Schror K. Iloprost-induced pulmonary arterial hypertension: insights from the prostacyclin
inhibition of proliferation of coronary artery smooth muscle cells (PGI2) receptor agonist in pulmonary arterial hypertension (GRI-
is abolished by homologous desensitization. Agents Actions PHON) study. J Heart Lung Transplant 2018;37:401–8.
Suppl 1995;45:85–91. 51 Channick RN, Simonneau G, Sitbon O, Robbins IM, Frost A,
35 Koch JA, Plum J, Grabensee B, Modder U. Prostaglandin E1: a Tapson VF et al. Effects of the dual endothelin-receptor antago-
new agent for the prevention of renal dysfunction in high risk nist bosentan in patients with pulmonary hypertension: a ran-
patients caused by radiocontrast media? PGE1 Study Group. domised placebo-controlled study. Lancet 2001;358:1119–23.
Nephrol Dial Transplant 2000;15:43–9. 52 Ivy DD, Kinsella JP, Abman SH. Physiologic characterization of
36 Eronen M, Pohjavuori M, Andersson S, Pesonen E, Raivio KO. endothelin A and B receptor activity in the ovine fetal pulmonary
Prostacyclin treatment for persistent pulmonary hypertension of circulation. J Clin Invest 1994;93:2141–8.
the newborn. Pediatr Cardiol 1997;18:3–7. 53 Ivy DD, Parker TA, Ziegler JW, Galan HL, Kinsella JP, Tuder
37 De Jaegere AP, van den Anker JN. Endotracheal instillation of RM et al. Prolonged endothelin A receptor blockade attenuates
prostacyclin in preterm infants with persistent pulmonary hyper- chronic pulmonary hypertension in the ovine fetus. J Clin Invest
tension. Eur Respir J 1998;12:932–4. 1997;99:1179–86.
38 Nicolas LB. Comparative pharmacokinetic, pharmacodynamic, 54 Keller RL, Tacy TA, Hendricks-Munoz K, Xu J, Moon-Grady
safety, and tolerability profiles of 3 different formulations of epo- AJ, Neuhaus J et al. Congenital diaphragmatic hernia: endothelin-
prostenol sodium for injection in healthy men. Clin Ther 1, pulmonary hypertension, and disease severity. Am J Respir
2013;35:440–9. Crit Care Med 2010;182:555–61.
39 Barst RJ. A comparison of continuous intravenous epoprostenol 55 Kumar P, Kazzi NJ, Shankaran S. Plasma immunoreactive endothe-
(prostacyclin) with conventional therapy for primary pulmonary lin-1 concentrations in infants with persistent pulmonary hyperten-
hypertension. N Engl J Med 1996;334:296–301. sion of the newborn. Am J Perinatol 1996;13:335–41.
40 Shapiro SM, Oudiz RJ, Cao T, Romano MA, Beckmann XJ, 56 More K, Athalye-Jape GK, Rao SC, Patole SK. Endothelin receptor
Georgiou D et al. Primary pulmonary hypertension: improved antagonists for persistent pulmonary hypertension in term and late
long-term effects and survival with continuous intravenous epo- preterm infants. Cochrane Database Syst Rev 2016:Cd010531.
prostenol infusion. J Am Coll Cardiol 1997;30:343–9. 57 Simonneau G, Galie N, Jansa P, Meyer GM, Al-Hiti H, Kusic-
41 Park BY, Chung SH. Treprostinil for persistent pulmonary hyper- Pajic A et al. Long-term results from the EARLY study of bosen-
tension of the newborn, with early onset sepsis in preterm infant: tan in WHO functional class II pulmonary arterial hypertension
2 Case reports. Medicine (Baltimore) 2017;96:e7303. patients. Int J Cardiol 2014;172:332–9.
42 Skoro-Sajer N. Subcutaneous treprostinil in congenital heart dis- 58 Jing ZC, Strange G, Zhu XY, Zhou DX, Shen JY, Gu H et al.
ease-related pulmonary arterial hypertension. Heart 2018, Doi: 10. Efficacy, safety and tolerability of bosentan in Chinese patients
1136/heartjnl-2017-312143. with pulmonary arterial hypertension. J Heart Lung Transplant
43 Tapson VF, Jing ZC, Xu KF, Pan L, Feldman J, Kiely DG et al. 2010;29:150–6.
Oral treprostinil for the treatment of pulmonary arterial hyperten- 59 Galie N, Rubin L, Hoeper M, Jansa P, Al-Hiti H, Meyer G et al.
sion in patients receiving background endothelin receptor antago- Treatment of patients with mildly symptomatic pulmonary arterial
nist and phosphodiesterase type 5 inhibitor therapy (the hypertension with bosentan (EARLY study): a double-blind, ran-
FREEDOM-C2 study): a randomized controlled trial. Chest domised controlled trial. Lancet 2008;371:2093–100.
2013;144:952–8. 60 Jain M, Varga J. Bosentan for the treatment of systemic sclerosis-
44 Sørensen LM, Wehland M, Kr€uger M, Simonsen U, Nassef MZ, associated pulmonary arterial hypertension, pulmonary fibrosis
Infanger M et al. A special focus on selexipag - treatment of pul- and digital ulcers. Expert Opin Pharmacother 2006;7:1487–501.
monary arterial hypertension. Curr Pharm Des 2017;23:5191–99. 61 Mohamed WA, Ismail M. A randomized, double-blind, placebo-
45 Coghlan JG, Channick R, Chin K, Di Scala L, Galie N, Ghofrani controlled, prospective study of bosentan for the treatment of per-
HA et al. Targeting the prostacyclin pathway with selexipag in sistent pulmonary hypertension of the newborn. J Perinatol
patients with pulmonary arterial hypertension receiving double 2012;32:608–13.
combination therapy: insights from the Randomized Controlled 62 Steinhorn RH, Fineman J, Kusic-Pajic A, Cornelisse P, Gehin M,
GRIPHON Study. Am J Cardiovasc Drugs 2018;18:37–47. Nowbakht P et al. Bosentan as adjunctive therapy for persistent
46 Kuwano K, Hashino A, Asaki T, Hamamoto T, Yamada T, pulmonary hypertension of the newborn: results of the random-
Okubo K et al. 2-[4-[(5,6-diphenylpyrazin-2-yl)(isopropyl)amino] ized multicenter placebo-controlled exploratory trial. J Pediatr
butoxy]-N-(methylsulfonyl)acetam ide (NS-304), an orally avail- 2016;177:90–6.e3.
able and long-acting prostacyclin receptor agonist prodrug. J 63 Iglarz M, Binkert C, Morrison K, Fischli W, Gatfield J, Treiber
Pharmacol Exp Ther 2007;322:1181–8. A et al. Pharmacology of macitentan, an orally active tissue-tar-
47 Morrison K, Ernst R, Hess P, Studer R, Clozel M. Selexipag: a geting dual endothelin receptor antagonist. J Pharmacol Exp Ther
selective prostacyclin receptor agonist that does not affect rat gas- 2008;327:736–45.
tric function. J Pharmacol Exp Ther 2010;335:249–55. 64 Weiss J, Theile D, Ruppell MA, Speck T, Spalwisz A, Haefeli WE.
48 Fuchikami C, Murakami K, Tajima K, Homan J, Kosugi K, Kura- Interaction profile of macitentan, a new non-selective endothelin-1
moto K et al. A comparison of vasodilation mode among selexi- receptor antagonist, in vitro. Eur J Pharmacol 2013;701:168–75.
pag (NS-304; [2-{4-[(5,6-diphenylpyrazin-2-yl)(isopropyl)amino] 65 Pulido T, Adzerikho I, Channick RN, Delcroix M, Galie N, Gho-
butoxy}-N-(methylsulfonyl)aceta mide]), its active metabolite frani HA et al. Macitentan and morbidity and mortality in pul-
MRE-269 and various prostacyclin receptor agonists in rat, por- monary arterial hypertension. N Engl J Med 2013;369:809–18.
cine and human pulmonary arteries. Eur J Pharmacol 66 Tran TT. Serious liver injury associated with macitentan: a case
2017;795:75–83. report. Pharmacotherapy 2018;38:e22–4.
49 Simonneau G, Torbicki A, Hoeper MM, Delcroix M, Karlocai K, 67 Ambrisentan PubChem. https://pubchem.ncbi.nlm.nih.gov/com
Galie N et al. Selexipag: an oral, selective prostacyclin receptor pound/Ambrisentan#section=Top. 2018 (updated 2018; last
agonist for the treatment of pulmonary arterial hypertension. Eur accessed on 21 February 2018).
Respir J 2012;40:874–80. 68 Ambrisentan ProMedicin. https://pro.medicin.dk/Medicin/Praepara
50 Preston IR, Channick RN, Chin K, Di Scala L, Farber HW, Gaine ter/4466. 2018 (updated 2018; last accessed on 21 February
S et al. Temporary treatment interruptions with oral selexipag in 2018).

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
MiniReview PERSISTENT NEWBORN PULMONARY HYPERTENSION 405

69 Takahashi T, Hayata S, Kobayashi A, Onaka Y, Ebihara T, Hara subarachnoid hemorrhage. Surg Neurol 2007;68:126–31; discus-
T. Surveillance on the safety and efficacy of ambrisentan (volibris sion 31-2.
tablet 2.5 mg) in patients with pulmonary arterial hypertension in 86 Fukumoto Y, Matoba T, Ito A, Tanaka H, Kishi T, Hayashidani S
real clinical practice: post-marketing surveillance (interim analysis et al. Acute vasodilator effects of a Rho-kinase inhibitor, fasudil, in
report). Clin Drug Investig 2017;38:219–29. patients with severe pulmonary hypertension. Heart 2005;91:391–2.
70 Riociguat - PubChem. https://pubchem.ncbi.nlm.nih.gov/com 87 Ishikura K. Beneficial acute effects of rho-kinase inhibitor in
pound/Riociguat#section=Top. 2018 (updated 2018; last accessed patients with pulmonary arterial hypertension. Circ J
on 18 January 2018). 2006;70:174–8.
71 Riociguat - ProMedicin. https://pro.medicin.dk/Medicin/Praeparate 88 Moretta DT. Rho-kinase inhibitors targeted to the pulmonary vas-
r/7450. 2018 (updated 2018; last accessed on 24 January 2018). culature for treatment of pulmonary hypertension. Am J Respir
72 Wardle AJ, Seager MJ, Wardle R, Tulloh RM, Gibbs JS. Guany- Crit Care Med 2017;195:A4225.
late cyclase stimulators for pulmonary hypertension. Cochrane 89 Lakshminrusimha S, Russell JA, Wedgwood S, Gugino SF, Kaz-
Database Syst Rev 2016:Cd011205. zaz JA, Davis JM et al. Superoxide dismutase improves oxygena-
73 Spreemann T, Bertram H, Happel CM, Kozlik-Feldmann R, tion and reduces oxidation in neonatal pulmonary hypertension.
Hansmann G. First-in-child use of the oral soluble guanylate Am J Respir Crit Care Med 2006;174:1370–7.
cyclase stimulator riociguat in pulmonary arterial hypertension. 90 Steinhorn RH, Albert G, Swartz DD, Russell JA, Levine CR,
Pulm Circ 2018;8:2045893217743123. Davis JM. Recombinant human superoxide dismutase enhances
74 Halank M. Riociguat for pulmonary arterial hypertension and the effect of inhaled nitric oxide in persistent pulmonary hyper-
chronic thromboembolic pulmonary hypertension: results from tension. Am J Respir Crit Care Med 2001;164:834–9.
a phase II long-term extension study. Respir Med 2017;128: 91 Farrow KN, Lakshminrusimha S, Reda WJ, Wedgwood S, Czech
50–6. L, Gugino SF et al. Superoxide dismutase restores eNOS expres-
75 Ghofrani HA, Hoeper MM, Halank M, Meyer FJ, Staehler G, sion and function in resistance pulmonary arteries from neonatal
Behr J et al. Riociguat for chronic thromboembolic pulmonary lambs with persistent pulmonary hypertension. Am J Physiol
hypertension and pulmonary arterial hypertension: a phase II Lung Cell Mol Physiol 2008;295:L979–87.
study. Eur Respir J 2010;36:792–9. 92 Solomonson LP, Flam BR, Pendleton LC, Goodwin BL, Eichler
76 Evgenov OV, Pacher P, Schmidt PM, Hasko G, Schmidt HH, DC. The caveolar nitric oxide synthase/arginine regeneration sys-
Stasch JP. NO-independent stimulators and activators of soluble tem for NO production in endothelial cells. J Exp Biol
guanylate cyclase: discovery and therapeutic potential. Nat Rev 2003;206:2083–7.
Drug Discov 2006;5:755–68. 93 Erez A, Nagamani SC, Shchelochkov OA, Premkumar MH, Cam-
77 Chester M, Seedorf G, Tourneux P, Gien J, Tseng N, Grover T peau PM, Chen Y et al. Requirement of argininosuccinate lyase
et al. Cinaciguat, a soluble guanylate cyclase activator, augments for systemic nitric oxide production. Nat Med 2011;17:1619–26.
cGMP after oxidative stress and causes pulmonary vasodilation in 94 Fike CD, Dikalova A, Kaplowitz MR, Cunningham G, Summar
neonatal pulmonary hypertension. Am J Physiol Lung Cell Mol M, Aschner JL. Rescue treatment with L-citrulline inhibits hypox-
Physiol 2011;301:L755–64. ia-induced pulmonary hypertension in newborn pigs. Am J Respir
78 Chester M, Tourneux P, Seedorf G, Grover TR, Gien J, Abman Cell Mol Biol 2015;53:255–64.
SH. Cinaciguat, a soluble guanylate cyclase activator, causes 95 Dikalova A, Fagiana A, Aschner JL, Aschner M, Summar M,
potent and sustained pulmonary vasodilation in the ovine fetus. Fike CD. Sodium-coupled neutral amino acid transporter 1
Am J Physiol Lung Cell Mol Physiol 2009;297:L318–25. (SNAT1) modulates L-citrulline transport and nitric oxide (NO)
79 Gien J, Seedorf GJ, Balasubramaniam V, Tseng N, Markham N, signaling in piglet pulmonary arterial endothelial cells. PLoS
Abman SH. Chronic intrauterine pulmonary hypertension ONE 2014;9:e85730.
increases endothelial cell Rho kinase activity and impairs angio- 96 Fike CD, Sidoryk-Wegrzynowicz M, Aschner M, Summar M,
genesis in vitro. Am J Physiol Lung Cell Mol Physiol 2008;295: Prince LS, Cunningham G et al. Prolonged hypoxia augments L-
L680–7. citrulline transport by system A in the newborn piglet pulmonary
80 Parker TA, Roe G, Grover TR, Abman SH. Rho kinase activation circulation. Cardiovasc Res 2012;95:375–84.
maintains high pulmonary vascular resistance in the ovine fetal 97 Fike CD, Summar M, Aschner JL. L-citrulline provides a novel
lung. Am J Physiol Lung Cell Mol Physiol 2006;291:L976–82. strategy for treating chronic pulmonary hypertension in newborn
81 Gien J, Tseng N, Seedorf G, Roe G, Abman SH. Peroxisome pro- infants. Acta Paediatr 2014;103:1019–26.
liferator activated receptor-gamma-Rho-kinase interactions con- 98 Sharif Kashani B, Tahmaseb Pour P, Malekmohammad M,
tribute to vascular remodeling after chronic intrauterine Behzadnia N, Sheybani-Afshar F, Fakhri M et al. Oral l-citrulline
pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol malate in patients with idiopathic pulmonary arterial hypertension
2014;306:L299–308. and Eisenmenger Syndrome: a clinical trial. J Cardiol 2014;64:
82 Jiang X, Wang YF, Zhao QH, Jiang R, Wu Y, Peng FH et al. 231–5.
Acute hemodynamic response of infused fasudil in patients with 99 Hansmann G, de Jesus Perez VA, Alastalo TP, Alvira CM,
pulmonary arterial hypertension: a randomized, controlled, cross- Guignabert C, Bekker JM et al. An antiproliferative BMP-2/
over study. Int J Cardiol 2014;177:61–5. PPARgamma/apoE axis in human and murine SMCs and its role
83 Fujita H, Fukumoto Y, Saji K, Sugimura K, Demachi J, Nawata J in pulmonary hypertension. J Clin Invest 2008;118:1846–57.
et al. Acute vasodilator effects of inhaled fasudil, a specific Rho- 100 Wolf D, Tseng N, Seedorf G, Roe G, Abman SH, Gien J. Endothe-
kinase inhibitor, in patients with pulmonary arterial hypertension. lin-1 decreases endothelial PPARgamma signaling and impairs
Heart Vessels 2010;25:144–9. angiogenesis after chronic intrauterine pulmonary hypertension.
84 Fukumoto Y, Yamada N, Matsubara H, Mizoguchi M, Uchino K, Am J Physiol Lung Cell Mol Physiol 2014;306:L361–71.
Yao A et al. Double-blind, placebo-controlled clinical trial with a 101 Clinicaltrials.gov. https://clinicaltrials.gov/ct2/results?cond=Persis
rho-kinase inhibitor in pulmonary arterial hypertension. Circ J tent+Pulmonary+Hypertension+of+the+Newborn&term=&cntry1=
2013;77:2619–25. &state1=&recrs=. (last accessed on 23 May 2018).
85 Suzuki Y, Shibuya M, Satoh S, Sugimoto Y, Takakura K. A post- 102 Vargas-Origel A, Gomez-Rodriguez G, Aldana-Valenzuela C,
marketing surveillance study of fasudil treatment after aneurysmal Vela-Huerta MM, Alarcon-Santos SB, Amador-Licona N. The

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
406 JONAS PEDERSEN ET AL. MiniReview

use of sildenafil in persistent pulmonary hypertension of the new- nitric oxide therapy in term and near-term newborn infants with
born. Am J Perinatol 2010;27:225–30. hypoxic respiratory failure. Pediatrics 2004;113:559–64.
103 Lakshminrusimha S, Keszler M, Kirpalani H, Van Meurs K, Chess 105 Sokol GM, Ehrenkranz RA. Inhaled nitric oxide therapy in
P, Ambalavanan N et al. Milrinone in congenital diaphragmatic neonatal hypoxic respiratory failure: insights beyond primary out-
hernia - a randomized pilot trial: study protocol, review of literature comes. Semin Perinatol 2003;27:311–9.
and survey of current practices. Matern Health Neonatol Perinatol 106 Ambalavanan N, St John E, Carlo WA, Bulger A, Philips JB
2017;3:27. 3rd. Feasibility of nitric oxide administration by oxygen
104 Konduri GG, Solimano A, Sokol GM, Singer J, Ehrenkranz RA, hood in neonatal pulmonary hypertension. J Perinatol 2002;22:
Singhal N et al. A randomized trial of early versus standard inhaled 50–6.

© 2018 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

You might also like