Sie sind auf Seite 1von 9

Archives of Cardiovascular Disease (2013) 106, 169—177

Available online at

www.sciencedirect.com

REVIEW

Pathophysiology of persistent pulmonary


hypertension of the newborn: Impact of the
perinatal environment
Physiopathologie de l’hypertension artérielle pulmonaire persistante du
nouveau-né : rôle de l’environnement périnatal

Laurent Storme a,b,∗, Estelle Aubry a,c,


Thameur Rakza a,b, Ali Houeijeh b,
Véronique Debarge a,b, Pierre Tourneux d,
Philippe Deruelle a,b, Thomas Pennaforte b ,
French Congenital Diaphragmatic Hernia Study Group

a
EA4489, Environnement Périnatal et Croissance, Faculté de Médecine, Université Lille-2,
Lille, France
b
Pôle Femme—Mère-Nouveau-né, Hôpital Jeanne-de-Flandre, CHRU de Lille, Lille, France
c
Pôle Enfant, Clinique de Chirurgie et Orthopédie de l’Enfant, CHRU de Lille, Lille, France
d
Pôle Mère—Enfant, CHU d’Amiens, Amiens, France

Received 26 July 2012; received in revised form 8 December 2012; accepted 11 December 2012
Available online 29 March 2013

KEYWORDS Summary The main cause of pulmonary hypertension in newborn babies results from the
Pulmonary failure of the pulmonary circulation to dilate at birth, termed ‘persistent pulmonary hyper-
hypertension; tension of the newborn’ (PPHN). This syndrome is characterized by sustained elevation of
Newborn; pulmonary vascular resistance, causing extrapulmonary right-to-left shunting of blood across
Inhaled NO; the ductus arteriosus and foramen ovale and severe hypoxaemia. It can also lead to life-
Hypoxaemia threatening circulatory failure. There are many controversial and unresolved issues regarding
the pathophysiology of PPHN, and these are discussed. PPHN is generally associated with factors

Abbreviations: CDH, congenital diaphragmatic hernia; cGMP, cyclic guanosine monophosphate; ECMO, extracorporeal oxygenation;
eNOS, endothelial nitric oxide synthase; iNO, inhaled nitric oxide; NO, nitric oxide; NOS, nitric oxide synthase; PaCO2 , partial pressure of
carbon dioxide in arterial blood; PO2 , oxygen pressure; PPHN, persistent pulmonary hypertension of the newborn; PUFA, polyunsaturated
fatty acid; PVR, pulmonary vascular resistance; SpO2 , saturation of peripheral oxygen; SSRI, selective serotonin reuptake inhibitor; VEGF,
vascular endothelial growth factor.
∗ Corresponding author. Pôle Femme—Mère-Nouveau-né, Hôpital Jeanne-de-Flandre, CHRU de Lille, 1, avenue Eugène-Avinée, 59035 Lille

cedex, France. Fax: +33 3 20 44 62 36.


E-mail address: lstorme@chru-lille.fr (L. Storme).

1875-2136/$ — see front matter © 2013 Elsevier Masson SAS. All rights reserved.
http://dx.doi.org/10.1016/j.acvd.2012.12.005
170 L. Storme et al.

such as congenital diaphragmatic hernia, birth asphyxia, sepsis, meconium aspiration and respi-
ratory distress syndrome. However, the perinatal environment—exposure to nicotine and certain
medications, maternal obesity and diabetes, epigenetics, painful stimuli and birth by Caesarean
section—may also affect the maladaptation of the lung circulation at birth. In infants with
PPHN, it is important to optimize circulatory function. Suggested management strategies for
PPHN include: avoidance of environmental factors that worsen PPHN (e.g. noxious stimuli, lung
overdistension); adequate lung recruitment and alveolar ventilation; inhaled nitric oxide (or
sildenafil, if inhaled nitric oxide is not available); haemodynamic assessment; appropriate fluid
and cardiovascular resuscitation and inotropic and vasoactive agents.
© 2013 Elsevier Masson SAS. All rights reserved.

MOTS CLÉS Résumé La principale cause d’hypertension pulmonaire du nouveau-né résulte d’une vaso-
Hypertension dilatation pulmonaire insuffisante à la naissance, appelée « hypertension artérielle pulmonaire
pulmonaire ; persistante du nouveau-né » (HTAPP). Ce syndrome est caractérisé par une élévation des résis-
Nouveau-né ; tances vasculaires pulmonaires, responsable d’un shunt droit-gauche par le foramen ovale
NO inhalé ; et le canal artériel et d’une profonde hypoxémie. L’HTAPP est à risque vital lorsque qu’elle
Hypoxémie s’accompagne d’une défaillance circulatoire. La prise en charge nécessite un recrutement pul-
monaire adéquat, l’inhalation de monoxyde d’azote et un support cardiovasculaire adapté.
Néanmoins, la physiopathologie et la prise en charge sont toujours l’objet de recherches inno-
vantes. Ainsi, de plus en plus d’arguments existent pour penser que l’environnement périnatal
joue un rôle déterminant dans la genèse et l’aggravation de ce syndrome.
© 2013 Elsevier Masson SAS. Tous droits réservés.

Background
Pulmonary hypertension in newborns results from the fail-
ure of the pulmonary circulation to dilate at birth. Termed
‘persistent pulmonary hypertension of the newborn’ (PPHN),
it occurs in an estimated 1—2 infants per 1000 live births
[1]. This syndrome is characterized by sustained elevation
of pulmonary vascular resistance (PVR), causing extrapul-
monary right-to-left shunting of blood across the ductus
arteriosus and foramen ovale and severe hypoxaemia [2,3]
(Fig. 1). PPHN is frequently associated with low systemic
pressure and low cardiac output because of increased
right ventricular afterload and myocardial dysfunction [2,4].
PPHN-induced circulatory failure is a life-threatening condi-
tion. Cardiac failure further impairs oxygen delivery to the
tissues and contributes to significant mortality and morbidity
in newborn infants with PPHN [2—4].
Management requires adequate lung recruitment and
alveolar ventilation, inhaled nitric oxide (iNO), and appro-
priate fluid and cardiovascular resuscitation [5]. Early
initiation of inotropic and vasoactive agents is commonly
used to increase cardiac output, maintain adequate blood
pressure and enhance oxygen delivery to the tissue [4,6].
Nevertheless, there are many controversial and unresolved
issues regarding the pathophysiology and the most effective
management of PPHN. Growing evidence is emerging that
indicates that the perinatal environment plays a key role in
this syndrome.
Figure 1. Schematic representation of the circulation in PPHN.
Sustained elevation of PVR contributes to low pulmonary blood flow
Pathophysiology and high pulmonary pressure, causing extrapulmonary right-to-left
shunting of blood across the ductus arteriosus and foramen ovale
Foetal circulation and severe hypoxaemia. LA: left atrium; LV: left ventricle; PPHN:
persistent pulmonary hypertension of the newborn; PV: pulmonary
The foetal pulmonary circulation is characterized by high vein; PVR: pulmonary vascular resistance; RA: right atrium; RV: right
PVR and low blood flow. Despite high pulmonary artery ventricle.
Impact of the perinatal environment on newborn persistent pulmonary hypertension 171

pressure, the lungs are perfused with less than 10% of


the combined ventricular output during late gestation [7].
Owing to high PVR in the foetus, most of the right ventricu-
lar output crosses the ductus arteriosus into the descending
aorta, thereby increasing umbilical—placental flow and gas
exchange.
Mechanisms that maintain high PVR in utero are incom-
pletely understood, but include low foetal oxygen pressure
(PO2 ) [7], lack of a gas—liquid interface [8] and pro-
duction of vasoconstrictor mediators such as endothelin-1
[9]. Nitric oxide (NO) production also modulates foetal
pulmonary vascular tone in response to diverse stimuli,
including acute changes in haemodynamic forces [8]. In
foetal lambs, acute compression of the ductus arteriosus
abruptly increases blood flow and pressure, and causes acute
pulmonary vasodilation through the shear stress-induced
Figure 2. NO/cGMP pathway. NO, produced by endothelial NOS,
release of NO [10—13]. However, despite maintenance of
diffuses towards the smooth muscle cell in which it stimulates sol-
constant pressure, pulmonary blood flow progressively falls
uble guanylate cyclase to produce cGMP. The mechanism whereby
and PVR increases over time [14,15]. Therefore, the foetal increased cGMP regulates pulmonary vascular tone includes acti-
pulmonary circulation is characterized by an ability to vation of K+ channels, causing smooth muscle cell membrane
oppose vasodilation over time and to regulate its flow. hyperpolarization and inactivation of VOCC. Closure of VOCC causes
a decrease in cytosolic Ca++ concentration and vasorelaxation.
Pulmonary adaptation at birth Intracellular concentration of cGMP is downregulated by PDE5 activ-
ities. Ca++ : calcium; cGMP: cyclic guanosine monophosphate; eNOS:
At birth, pulmonary blood flow increases dramatically, by endothelial nitric oxide synthase; GMP: guanosine monophosphate;
8—10-fold, and PVR drops immediately. Several stimuli, K+ : potassium; NO: nitric oxide; NOS: nitric oxide synthase; PDE
including drainage and absorption of foetal lung liquid, 5: phosphodiesterase 5; SMC: smooth muscle cell; VOCC: voltage-
operated calcium channels.
rhythmic distension of the lung, increased PO2 and altered
production of several vasoactive products, including NO,
are known to contribute to pulmonary vasodilation at birth function. A rigorous clinical and echocardiographic assess-
[8]. The effects of several birth-related stimuli, including ment is required for early diagnosis of circulatory failure.
increased blood flow or shear stress, increased PO2 and ven- More relevant than changes in postductal SpO2 , reflecting
tilation are partly dependent upon activation of nitric oxide right-to-left ductus arteriosus shunting, the decrease in pre-
synthase (NOS) [8,10—13]. ductal SpO2 results from an increase in the right-to-left
The increase in NO production accounts for nearly 50% shunting through the foramen ovale. It occurs when the
of the abrupt fall in PVR at birth in foetal lambs [8]. NO pulmonary venous return decreases (which reduces the left
mediates vasodilatation by stimulating soluble guanylate atrial pressure) or in case of right cardiac failure (leading
cyclase-induced cyclic guanosine monophosphate (cGMP) to elevation of right atrial pressure). Both are markers for
production [16]. cGMP is downregulated by phosphodieste- potential circulatory failure. In most cases, low postductal
rase 5 activity. Phosphodiesterase 5, which is abundantly SpO2 alone (i.e. with normal preductal SpO2 ) does not cause
expressed in lung tissue, particularly during foetal life, tissue hypoxia. In contrast, a drop in preductal SpO2 is usu-
is a key regulator of perinatal pulmonary circulation [17] ally associated with symptoms of shock and an increased
(Fig. 2). lactate concentration.
Experimental studies of chronic pulmonary hypertension
Maladaptation of the pulmonary circulation at in newborn animals have demonstrated impaired endothelial
birth release of NO and increased production of vasoconstric-
tors (e.g. endothelin-1) [18]. In normal foetal lambs, acute
PPHN is a clinical syndrome that is associated with compression of the ductus arteriosus causes progressive
diverse neonatal cardiopulmonary diseases, including birth pulmonary vasodilation, which is blocked by NOS inhibi-
asphyxia, sepsis, meconium aspiration and respiratory dis- tion, suggesting that NO mediates shear stress-induced
tress syndrome, or it can be idiopathic [1—3]. vasodilation. After NOS inhibition, acute ductus arterio-
The usual clinical expression of PPHN consists of an sus compression causes a marked pulmonary vasoconstrictor
unstable refractory hypoxaemia with pre- and postductal response, suggesting that NOS blockade unmasks a potent
saturation of peripheral oxygen (SpO2 ) gradient. Pulmonary myogenic response in the normal foetal lung [14]. In con-
hypertension can be associated with a systemic hypoten- trast, exposure to chronic pulmonary hypertension for 2 days
sion and symptoms of shock, termed ‘obstructive’ shock, impairs flow-induced vasodilation and enhances the myo-
the clinical and biological signs of which are non-specific: genic response during acute ductus arteriosus compression
grey colour, tachycardia, refill capillary time more than in the absence of NOS inhibition [19,20]. Therefore, with
3 seconds, oliguria, systemic hypotension and lactic acido- prolonged pulmonary hypertension during the perinatal
sis. Doppler echocardiography is required to determine the period, pulmonary blood flow is autoregulated and indepen-
main component of the shock. Management priority is not to dent of pulmonary artery pressure. The most striking feature
normalize postductal SpO2 , but to optimize the circulatory of perinatal pulmonary hypertension is the loss of the normal
172 L. Storme et al.

vasodilator response to birth-related stimuli causing a para- smoke causes a potent and sustained pulmonary vasocon-
doxical vasoconstriction to acute haemodynamic stress. striction in the foetus, and blunts the vasodilator response
Several investigators have suggested that an increase to an increase in oxygen tension [30]. These effects are
in PDE5 activity may contribute to this phenomenon. Lung associated with a marked decrease in foetal oxygenation.
PDE5 activity increased by 150% in foetal lambs with As the postnatal circulatory adaptation is highly dependent
ductus arteriosus ligation for 8 days compared to control on the decrease in PVR and the vasodilator response to
lambs [21]. Sildenafil attenuates the progressive eleva- birth-related stimuli, such as oxygen, we speculate that pre-
tion of basal pulmonary vascular tone observed during natal exposure to tobacco smoke increases the risk of PPHN
chronic pulmonary hypertension, and preserves, at least through failure of the pulmonary circulation to dilate at
in part, the physiological pulmonary vascular response to birth.
vasodilators [22].
More recently, the role of vascular endothelial growth Role of stress or pain
factor (VEGF) in foetal lung development and the patho-
genesis of PPHN have been reported. VEGF is a potent Corticosteroids and catecholamine are the main stress hor-
endothelial cell mitogen and regulator of angiogenesis. mones. Their effects on lung parenchyma maturation, lung
In vivo inhibition of VEGF receptors in normal foetal fluid clearance and surfactant release at birth have been
sheep results in impaired vascular growth and pul- widely investigated. Evidence has emerged that the stress
monary hypertension [23]. Impaired alveolarization and hormones promote normal circulatory adaptation at birth.
vessel growth in chronic intrauterine pulmonary hyper- Several experimental and clinical studies have shown that
tension are associated with decreased VEGF protein norepinephrine improves circulation in the perinatal lung.
expression [24]. In foetal lambs, norepinephrine has been shown to increase
lung blood flow and reduce PVR [31]. Norepinephrine induces
an NO-dependent pulmonary vasodilation in the ovine foetus
through activation of ␣2 -adrenoceptors [32—35]. In a large
Risk factors for PPHN: impact of the retrospective review of nearly 30,000 consecutive deliver-
environment ies over 7 years, the incidence of PPHN in newborn infants
delivered by elective Caesarean was almost five times higher
PPHN is usually associated with pulmonary parenchymal dis- than among those delivered vaginally [36]. A likely hypoth-
ease, such as meconium aspiration, pneumonia or hyaline esis for PPHN after Caesarean is that there might be an
membrane disease; or with disease-related lung hypoplasia advantage to labour and vaginal delivery for the pulmonary
including congenital diaphragmatic hernia (CDH) or prema- vascular bed of the neonate. Mechanisms explaining the
ture rupture of the membrane-mediated oligohydramnios increase in PPHN after Caesarean remain unclear. A surge
[1—3]. However, growing evidence indicates that the foetal of catecholamines, especially norepinephrine, is observed
environment plays a critical role in the maladaptation of the at birth. However, neonatal norepinephrine concentrations
lung circulation at birth. Evidence of a role for epigenetics in are significantly lower after Caesarean section than after
the development of pulmonary arterial hypertension is also vaginal delivery. Lower levels of circulating norepinephrine
emerging. Epigenetic mechanisms are involved in the reg- after Caesarean section may explain, at least in part, the
ulation of gene expression, which are controlled, at least high incidence of PPHN in newborn infants delivered by Cae-
in part, by environment. In newborn rats with pulmonary sarean section [37].
hypertension, the increased expression of endothelial NOS is In contrast, evidence exists that painful stimuli impair
associated with changes in epigenetic regulation [25]. In the normal adaptation at birth and promote PPHN. Hypox-
same way, pulmonary hypertension has been associated with aemia is usually observed during stressful intensive care
abnormal epigenetic regulation of superoxide dismutase and procedures in newborn infants with respiratory failure,
hypoxia inducible factor [26]. the duration of which may be reduced by opioid anal-
gesia [38]. In foetal lambs, nociceptive stimuli increase
Antenatal smoke exposure pulmonary artery pressure and PVR [39,40]. This vaso-
constrictive response is abolished by analgesia and by
Cotinine concentrations in cord blood — a biological marker ␣1 -adrenoceptor blockade [41]. These results provide new
for nicotine exposure — have been found to be higher in insights into the mechanisms by which stressful events may
infants with PPHN than in healthy control newborn infants worsen hypoxaemia in newborn infants with PPHN. Noci-
[27]. Prenatal cigarette smoke has been found to increase ceptive stimulation, through ␣1 -adrenoceptor activation,
the risk of PPHN among premature infants less than 30 weeks may elevate pulmonary vascular tone leading to increased
gestational age, suggesting a toxic effect on the pulmonary right-to-left shunting through the ductus arteriosus and the
vascular development and maturation [28]. Studies of lungs foramen ovale and to severe hypoxaemia.
exposed in utero to cigarette smoke have shown marked
structural and functional changes, such as decreased alve- Drug exposure
olar attachments to the airway wall and increased airways
responsiveness. Endothelial dysfunction in intrapulmonary Drug administration during pregnancy may increase the risk
arteries has been found during exposure to tobacco smoke of maladaptation at birth.
contributing, at least in part, to vasoconstriction and smooth The likelihood of PPHN increases after antenatal expo-
muscle cell proliferation [29]. In an experimental model of sure to aspirin or other non-steroidal anti-inflammatory
foetal lambs, we found that prenatal exposure to cigarette drugs [42]. Mechanisms of antenatal non-steroidal
Impact of the perinatal environment on newborn persistent pulmonary hypertension 173

anti-inflammatory-induced PPHN may include antena- at birth, in particular in conditions with prolonged pul-
tal closure of the ductus arteriosus or decreased production monary hypertension such as CDH or pulmonary hypoplasia.
of vasodilator prostaglandins. Similar risks exist after birth:
ibuprofen administration has the potential to cause PPHN Oxygen and hyperoxia
in preterm infants with patent ductus arteriosus [43].
Selective serotonin reuptake inhibitors (SSRIs), used to While oxygen stimulates endothelial nitric oxide synthase
treat maternal depression, elevate the basal vascular tone (eNOS) and NO production and contributes to pulmonary
in the foetal lung [44]. Pulmonary vascular remodelling and adaptation at birth [8], high concentrations of oxygen,
right ventricular hypertrophy have been observed in rat pups such as are used to treat PPHN, may produce reactive
after maternal exposure to fluoxetine [45]. A case-control oxygen species. For instance, hydrogen peroxide (H2 O2 )
study to assess whether PPHN is associated with exposure to could decrease eNOS promoter activity, associated with
SSRIs has been conducted during late pregnancy [46]. A total an endothelin-1-mediated downregulation of eNOS expres-
of 377 women whose infants had PPHN and 836 matched con- sion [52]. Uncoupled eNOS has been shown to increase
trol women were enrolled in the study. Fourteen infants with superoxide radicals, which rapidly combine with NO to
PPHN had been exposed to antenatal SSRI, as compared with form peroxynitrite, a potent pulmonary vasoconstrictor
six control infants (adjusted odds ratio 6.1, 95% confidence and potential cellular toxin. Furthermore, hyperoxia may
interval 2.2—16.8) [46]. These data support an association blunt NO-mediated pulmonary vasodilation through increas-
between the maternal use of SSRIs in late pregnancy and ing phosphodiesterase 5 activity. In accordance with these
PPHN in the offspring. studies, recombinant human superoxide dismutase enhances
In case of severe respiratory failure, early initiation of vasodilation after birth in experimental models of PPHN
inotropic and vasoactive agents is warranted to increase [53].
cardiac output, maintain adequate blood pressure and
enhance oxygen delivery to the tissue [47]. Dopamine is
the sympathomimetic amine most frequently used for sep- Principles of management
tic shock in newborn infants. The ability of dopamine to
raise systemic blood pressure has been clearly documented. Basic principles
However, dopamine has also been suggested to increase
pulmonary artery pressure and pulmonary/systemic mean In PPHN with right-to-left shunt through the ductus arterio-
arterial pressure ratio in experimental models and in human sus, the contribution of hypoxaemia to tissue oxygenation
[31,48]. Caution should therefore be advocated in the use (low partial pressure of oxygen in arterial blood [PaO2 ]) is
of dopamine in newborn infants at risk of, or with, PPHN. likely to be modest, provided SpO2 in the preductal area
is greater than 80%, circulatory function is adequate and
haemoglobin concentration is normal. Similarly, hypoxaemia
Perinatal nutrition during foetal life (SpO2 60—75%) or during cyanotic congen-
ital heart diseases is not associated with tissue hypoxia,
It has been well established that maternal obesity and as long as cardiac function is normal. However, there is
diabetes are associated with increased risks of maladapta- some evidence to suggest that cardiac dysfunction may
tion at birth [49]. The exact mechanisms are unclear, but play a major role in PPHN. Severe pulmonary hyperten-
include perinatal asphyxia, parenchymal disease and poly- sion increases right ventricular afterload that may result
cythaemia. Studies have highlighted that infants with PPHN in right ventricular failure. The resulting elevation of right
are deficient in the amino acid, L-arginine, which is required ventricular telediastolic pressure causes right-to-left shunt-
for NO synthesis [50]. Evidence exists that differential vas- ing through the foramen ovale and worsens hypoxaemia.
cular effects can be expected according to lipid intake. Because of a high degree of interdependence between the
Indeed, dietary fish oils have beneficial effects on cardiac right and the left ventricles due to the presence of common
and vascular function, including effects on platelet/vessel structures (the interventricular septum and the inextensible
wall interactions, endothelial function and inhibition of pericardium), changes in the right ventricle size and geome-
smooth muscle cell proliferation. The n3 polyunsaturated try may alter the left ventricular function. This may explain
fatty acids (PUFAs) are metabolized by several enzymes, why PPHN is usually associated with low systemic pressure
including cyclooxygenase, to produce prostaglandins (PGE3 , and low cardiac output requiring the use of cardiac sup-
PGI3 ), which are known potential vasodilator mediators. In port [2—4]. Right ventricle failure occurs when the ductus
addition, n3 PUFA competes with arachidonic acid (n6 PUFA) arteriosus is closed or restricted. In severe PPHN with right-
for enzymatic conversion. This competition decreases for- to-left shunting through the ductus arteriosus, both the left
mation of vasoactive arachidonic acid metabolites such as and right ventricle contribute to systemic blood flow (Fig. 3).
thromboxane A2, a potent pulmonary vasoconstrictor. Thus, Therefore, management of the newborn infant with PPHN
some evidence suggests that n3 PUFA may be beneficial in requires both lowering of PVR and support of cardiovascular
conditions associated with pulmonary hypertension. Also, n3 function (Table 1).
PUFA induces a potent pulmonary vasodilatation in the peri-
natal lung [51]. This effect is abolished by cytochrome 450 Environmental factors
epoxygenase, but not by NOS inhibitors. Our data provide
evidence for potential beneficial effects of n3 PUFA on the First, exposure to environmental factors that worsen PPHN
pulmonary circulation through production of epoxides. n3 should be avoided. Noxious stimuli, including tactile stimuli,
PUFA supplementation may help to prevent maladaptation tracheal suction and heel pricks have to be limited as much
174 L. Storme et al.

Figure 3. Schematic representation of the circulation in PPHN. Because of elevated PVR, part of the right ventricular outflow is directed
towards the systemic circulation through the ductus arteriosus, contributing to systemic blood flow. Despite a right-to-left ductus arteriosus
shunting-mediating drop in postductal SpO2 , oxygen supply is usually adequate as long as the ductus arteriosus is widely patent. Both aortic
and pulmonary artery pressures are closely related when the ductus arteriosus is patent (low ductal resistance). Thus, aortic pressure plays
a critical role in pulmonary circulation. A drop in aortic pressure is associated with a decrease in pulmonary artery pressure, and since
pulmonary artery pressure is the driving force for lung blood flow, a decrease in aortic pressure-induced drop in pulmonary artery pressure
may worsen hypoxaemia and oxygen delivery. This electrical representation further highlights that a patent ductus arteriosus and normal
right ventricle function are required for sustaining systemic blood flow and oxygen supply. F(PAP): proportional to pulmonary artery pressure;
PA: pulmonary artery; PAP: pulmonary artery pressure; PPHN: persistent pulmonary hypertension of the newborn; PVR: pulmonary vascular
resistance; SVR: systemic vascular resistance.

as possible. A specific pain scale for newborns should be maintaining preductal oxygen saturation greater than 85%
used to titrate analgesia and optimize their environment and partial pressure of carbon dioxide in arterial blood
(reduce noise and light, ‘cocooning’). Overdistension of the (PaCO2 ) of 40—55 mmHg. Centres with better-than-expected
lungs contributes to a decrease in pulmonary blood flow and CDH survival report the combination of establishing clini-
should be prevented, in particular in conditions associated cal care guidelines that set limits on ventilatory pressures
with pulmonary hypoplasia (CDH and premature rupture of to avoid lung overdistension and accepting adequate blood
the membranes). Polycythaemia-mediating increased blood gas rather than optimal PaCO2 and PaO2 , as long as
viscosity and increased pulmonary artery pressure should be there is evidence of adequate cardiac output and organ
corrected. function [54].
Infants with significant right-to-left shunting require pul-
Mechanical ventilation monary vasodilator therapy. Currently, iNO is recommended
for infants with PPHN [55]. This improves outcomes in hypox-
Management requires adequate lung recruitment and aemic term and near-term infants by reducing the incidence
alveolar ventilation, appropriate fluid and cardiovascular of the combined endpoint of death or need for extracor-
resuscitation and use of pulmonary vasodilators [5]. In poreal oxygenation (ECMO) [55]. Oxygenation improves in
CDH, mask ventilation should be avoided during resus- approximately 50% of infants receiving iNO [55]. Improve-
citation in the delivery room to prevent gut distension ment in oxygenation has also been reported with the use of
located in the thoracic cavity. High airway pressures should iNO in newborns with CDH and PPHN [56]. However, early use
also be avoided, to prevent pulmonary barotraumas while of iNO does not appear to improve the combined endpoint of
Impact of the perinatal environment on newborn persistent pulmonary hypertension 175

Table 1 Steps in the management of PPHN. ECMO


Step Action ECMO may be required to ensure effective oxygenation and
1 Prevent exposure to environmental factors that decarboxylation, to limit the baro-volotrauma of the lungs
may worsen PPHN (e.g. stress, painful stimuli, and to improve right cardiac failure. ECMO is indicated when
noise, excessive light, lung overdistension) hypoxaemia persists despite optimal medical management.
2 Provide adequate lung expansion and ventilation ECMO may be indicated based on the following criteria [1]:
• preductal SpO2 less than 80% despite peak inspiratory
3 Provide inhaled nitric oxide
4 Assess for haemodynamics (clinical examination, pressure greater than 28 cmH2 O (or mean airway pres-
chest X-ray, cardiothoracic index, Doppler sure greater than 15 cmH2 O in high frequency oscillatory
echocardiography) ventilation);
• PPHN and circulatory failure resistant to adequate man-
5 Provide appropriate fluid expansion and
vasoactive support according to the main agement;
• gestational age greater than 34 weeks;
component of the circulatory failure
• birth weight greater than 2 kg.
(obstructive, hypovolaemic, distributive or
cardiogenic)
6 ECMO may be required in life-threatening Pulmonary vasodilator drugs
obstructive shock
There is limited experience in PPHN with other pharmaco-
ECMO: extracorporeal oxygenation; PPHN: persistent pulmonary logical pulmonary vasodilators.
hypertension of the newborn. Phosphodiesterase inhibitors reduce the degradation of
cGMP produced by endogenous or inhaled NO. Sildenafil has
been found to improve cardiac output and respiratory func-
tion by reducing pulmonary hypertension refractory to iNO in
death/ECMO in infants with CDH alone [57]. Usual concen- seven newborns with CDH [58]. In chronic pulmonary hyper-
trations of iNO for the treatment of PPHN are 5—20 ppm. tension associated with CDH, sildenafil has been found to
improve pulmonary vascular function and promoted lung
Supporting cardiovascular function growth [59]. Evidence exists that sildenafil is well tolerated
in the newborn infant with PPHN [60]. A meta-analysis of
Early initiation of inotropic and vasoactive agents is warr- three trials including 77 newborn infants with PPHN has indi-
anted to increase cardiac output, maintain adequate blood cated that sildenafil may improve oxygenation and reduce
pressure and enhance oxygen delivery to the tissues. mortality [61]. The studies were performed in resource-
Systemic hypotension is usually poorly tolerated, with limited settings where iNO was not available. The results of
worsening of hypoxia explained by: the meta-analysis suggest that sildenafil in the treatment
• a decrease in left ventricle filling causing a decrease in of PPHN has significant potential, especially in resource-
oxygen delivery to the tissue; limited settings.
• a decrease in coronary flow; The use of prostacyclin and analogues is an established
• a drop in pulmonary pressure resulting in a decrease in therapy for children and adults with primary pulmonary
pulmonary blood flow. hypertension. In a recent population-based study, the use of
Indeed, as the ductus arteriosus is usually patent in prostacyclin, along with various other measures, has been
newborns with PPHN, pulmonary artery pressure and aortic associated with a high survival rate [62]. Subcutaneous tre-
pressure are closely related (Fig. 3). A decrease in systemic prostinil has been found to improve functional symptoms in
pressure-mediated drop in pulmonary artery pressure causes young children with refractory pulmonary arterial hyperten-
a decrease in pulmonary blood flow, as Flow = f(Pressure). sion [63]. However, its use in PPHN has not been reported.
Therefore, careful assessment of haemodynamics (clini- Similarly, there is evidence to suggest that endothe-
cal examination, chest X-ray, cardiothoracic index and lin receptor blockade may improve pulmonary blood flow
echocardiography) is required in PPHN to provide optimal in PPHN. In an experimental model of foetal pulmonary
treatment. hypertension, intrauterine endothelin receptor blockade
Four types of shock, according to the main mechanism decreased pulmonary artery pressure, decreased right ven-
responsible for the cardiocirculatory failure, can be distin- tricular hypertrophy and distal muscularization of small
guished in the newborn: pulmonary arteries and increased the fall in PVR at deliv-
• hypovolaemic; ery [64]. Bosentan is a non-specific endothelin-1 receptor
• vasoplegic; inhibitor that improves pulmonary hypertension in adult
• cardiogenic; patients. Although recent reports suggest that bosentan may
• obstructive. improve PPHN [65], there is still insufficient evidence to
support the use of bosentan in the management of PPHN.
Fluid resuscitation should only be used if there is evi-
dence for hypovolaemia. Cardiogenic shock — a rare event
in PPHN as long as the ductus arteriosus is widely patent — Conclusions
may require inotropic drug infusion (dobutamine). The use
of vasoactive drug is usually warranted in vasoplegic or Growing evidence indicates that the perinatal environment
obstructive shock. plays a key role in the failure of cardiopulmonary transition
176 L. Storme et al.

to adequate pulmonary circulation at birth. Reduction of pulmonary circulation. Am J Physiol Lung Cell Mol Physiol
exposure to risk factors should be the first step in the pre- 2001;281:L1157—63.
vention of PPHN. New insights showing that PPHN may be [13] Rairigh RL, Storme L, Parker TA, et al. Role of neuronal nitric
associated with epigenetic modifications suggest that novel oxide synthase in regulation of vascular and ductus arteriosus
pharmacological interventions targeting modulation of epi- tone in the ovine fetus. Am J Physiol Lung Cell Mol Physiol
2000;278:L105—10.
genetic regulation may be beneficial.
[14] Storme L, Rairigh RL, Parker TA, Kinsella JP, Abman SH. In vivo
PPHN causes hypoxaemia through extrapulmonary right- evidence for a myogenic response in the fetal pulmonary cir-
to-left shunting. The management priority should be to culation. Pediatr Res 1999;45:425—31.
optimize circulatory function rather than normalize post- [15] Abman SH, Accurso FJ. Acute effects of partial compression of
ductal SpO2 , because PPHN-induced circulatory failure is a ductus arteriosus on fetal pulmonary circulation. Am J Physiol
life-threatening condition. 1989;257:H626—34.
Management of PPHN requires adequate lung recruit- [16] Deruelle P, Grover TR, Storme L, Abman SH. Effects of BAY
ment and alveolar ventilation, iNO and appropriate fluid 41-2272, a soluble guanylate cyclase activator, on pulmonary
and cardiovascular resuscitation. Early initiation of inotropic vascular reactivity in the ovine fetus. Am J Physiol Lung Cell
and vasoactive agents is commonly used to increase car- Mol Physiol 2005;288:L727—33.
[17] Jaillard S, Larrue B, Deruelle P, et al. Effects of phosphodieste-
diac output, maintain adequate blood pressure and enhance
rase 5 inhibitor on pulmonary vascular reactivity in the fetal
oxygen delivery to the tissue. However, a rigorous clinical lamb. Ann Thorac Surg 2006;81:935—42.
and echocardiographic assessment is required for optimal [18] Villamor E, Le Cras TD, Horan MP, Halbower AC, Tuder
management of PPHN-associated circulatory failure. RM, Abman SH. Chronic intrauterine pulmonary hypertension
impairs endothelial nitric oxide synthase in the ovine fetus.
Am J Physiol 1997;272:L1013—20.
Disclosure of interest [19] Storme L, Rairigh RL, Parker TA, Kinsella JP, Abman SH. Acute
intrauterine pulmonary hypertension impairs endothelium-
The authors declare that they have no conflicts of interest dependent vasodilation in the ovine fetus. Pediatr Res
1999;45:575—81.
concerning this article.
[20] Storme L, Parker TA, Kinsella JP, Rairigh RL, Abman SH. Chronic
hypertension impairs flow-induced vasodilation and augments
the myogenic response in fetal lung. Am J Physiol Lung Cell Mol
References Physiol 2002;282:L56—66.
[21] Hanson KA, Ziegler JW, Rybalkin SD, Miller JW, Abman
[1] Farrow KN, Fliman P, Steinhorn RH. The diseases treated with SH, Clarke WR. Chronic pulmonary hypertension increases
ECMO: focus on PPHN. Semin Perinatol 2005;29:8—14. fetal lung cGMP phosphodiesterase activity. Am J Physiol
[2] Kinsella JP, Abman SH. Recent developments in the pathophy- 1998;275:L931—41.
siology and treatment of persistent pulmonary hypertension of [22] Larrue B, Jaillard S, Lorthioir M, et al. Pulmonary vascular
the newborn. J Pediatr 1995;126:853—64. effects of sildenafil on the development of chronic pulmonary
[3] Abman SH. New developments in the pathogenesis and treat- hypertension in the ovine fetus. Am J Physiol Lung Cell Mol
ment of neonatal pulmonary hypertension. Pediatr Pulmonol Physiol 2005;288:L1193—200.
Suppl 1999;18:201—4. [23] Grover TR, Parker TA, Zenge JP, Markham NE, Kinsella JP,
[4] Tourneux P, Rakza T, Bouissou A, Krim G, Storme L. Pulmonary Abman SH. Intrauterine hypertension decreases lung VEGF
circulatory effects of norepinephrine in newborn infants with expression and VEGF inhibition causes pulmonary hyperten-
persistent pulmonary hypertension. J Pediatr 2008;153:345—9. sion in the ovine fetus. Am J Physiol Lung Cell Mol Physiol
[5] Weinberger B, Weiss K, Heck DE, Laskin DL, Laskin JD. Pharma- 2003;284:L508—17.
cologic therapy of persistent pulmonary hypertension of the [24] Gien J, Seedorf GJ, Balasubramaniam V, Markham N, Abman
newborn. Pharmacol Ther 2001;89:67—79. SH. Intrauterine pulmonary hypertension impairs angiogenesis
[6] Walsh-Sukys MC, Tyson JE, Wright LL, et al. Persistent pul- in vitro: role of vascular endothelial growth factor nitric oxide
monary hypertension of the newborn in the era before signaling. Am J Respir Crit Care Med 2007;176:1146—53.
nitric oxide: practice variation and outcomes. Pediatrics [25] Xu XF, Ma XL, Shen Z, Wu XL, Cheng F, Du LZ. Epigenetic regula-
2000;105:14—20. tion of the endothelial nitric oxide synthase gene in persistent
[7] Rudolph AM, Heymann MA. Pulmonary circulation in fetal pulmonary hypertension of the newborn rat. J Hypertens
lambs. Pediatr Res 1972;6:341 [abstract]. 2010;28:2227—35.
[8] Cornfield DN, Chatfield BA, McQueston JA, McMurtry IF, [26] Kim GH, Ryan JJ, Marsboom G, Archer SL. Epigenetic mecha-
Abman SH. Effects of birth-related stimuli on L-arginine- nisms of pulmonary hypertension. Pulm Circ 2011;1:347—56.
dependent pulmonary vasodilation in ovine fetus. Am J Physiol [27] Bearer C, Emerson RK, O’Riordan MA, Roitman E, Shack-
1992;262:H1474—81. leton C. Maternal tobacco smoke exposure and persistent
[9] Ivy DD, Kinsella JP, Abman SH. Physiologic characterization of pulmonary hypertension of the newborn. Environ Health Per-
endothelin A and B receptor activity in the ovine fetal pul- spect 1997;105:202—6.
monary circulation. J Clin Invest 1994;93:2141—8. [28] Muraskas JK, Juretschke LJ, Weiss MG, Bhola M, Besinger RE.
[10] Storme L, Rairigh RL, Parker TA, Cornfield DN, Kinsella JP, Neonatal-perinatal risk factors for the development of per-
Abman SH. K+-channel blockade inhibits shear stress-induced sistent pulmonary hypertension of the newborn in preterm
pulmonary vasodilation in the ovine fetus. Am J Physiol newborns. Am J Perinatol 2001;18:87—91.
1999;276:L220—8. [29] Wright JL, Churg A. Short-term exposure to cigarette smoke
[11] Rairigh RL, Storme L, Parker TA, et al. Inducible NO synthase induces endothelial dysfunction in small intrapulmonary arter-
inhibition attenuates shear stress-induced pulmonary vasodila- ies: analysis using guinea pig precision cut lung slices. J Appl
tion in the ovine fetus. Am J Physiol 1999;276:L513—21. Physiol 2008;104:1462—9.
[12] Zenge JP, Rairigh RL, Grover TR, et al. NO and prostaglandin [30] Houfflin-Debarge V, Sabbah-Briffaut E, Aubry E, Deruelle P,
interactions during hemodynamic stress in the fetal ovine Alexandre C, Storme L. Effects of environmental tobacco
Impact of the perinatal environment on newborn persistent pulmonary hypertension 177

smoke on the pulmonary circulation in the ovine fetus. Am J [49] Nohr EA, Villamor E, Vaeth M, Olsen J, Cnattingius S. Mortal-
Obstet Gynecol 2011;204:450 [e8-50 e14]. ity in infants of obese mothers: is risk modified by mode of
[31] Jaillard S, Houfflin-Debarge V, Riou Y, et al. Effects of cate- delivery? Acta Obstet Gynecol Scand 2012;91:363—71.
cholamines on the pulmonary circulation in the ovine fetus. [50] Vosatka RJ, Kashyap S, Trifiletti RR. Arginine deficiency accom-
Am J Physiol Regul Integr Comp Physiol 2001;281:R607—14. panies persistent pulmonary hypertension of the newborn. Biol
[32] Magnenant E, Jaillard S, Deruelle P, et al. Role of the alpha2- Neonate 1994;66:65—70.
adrenoceptors on the pulmonary circulation in the ovine fetus. [51] Houeijeh A, Aubry E, Coridon H, et al. Effects of n-3 polyunsat-
Pediatr Res 2003;54:44—51. urated fatty acids in the fetal pulmonary circulation. Crit Care
[33] Jaillard S, Elbaz F, Bresson-Just S, et al. Pulmonary vasodilator Med 2011;39:1431—8.
effects of norepinephrine during the development of chronic [52] Wedgwood S, Black SM. Endothelin-1 decreases endothelial
pulmonary hypertension in neonatal lambs. Br J Anaesth NOS expression and activity through ETA receptor-mediated
2004;93:818—24. generation of hydrogen peroxide. Am J Physiol Lung Cell Mol
[34] Houfflin-Debarge V, Deruelle P, Jaillard S, et al. Effects of ante- Physiol 2005;288:L480—7.
natal glucocorticoids on circulatory adaptation at birth in the [53] Kinsella JP, Parker TA, Davis JM, Abman SH. Superoxide dismu-
ovine fetus. Biol Neonate 2005;88:73—8. tase improves gas exchange and pulmonary hemodynamics in
[35] Deruelle P, Houfflin-Debarge V, Magnenant E, et al. Effects of premature lambs. Am J Respir Crit Care Med 2005;172:745—9.
antenatal glucocorticoids on pulmonary vascular reactivity in [54] Bagolan P, Casaccia G, Crescenzi F, Nahom A, Trucchi A, Gior-
the ovine fetus. Am J Obstet Gynecol 2003;189:208—15. landino C. Impact of a current treatment protocol on outcome
[36] Levine EM, Ghai V, Barton JJ, Strom CM. Mode of delivery of high-risk congenital diaphragmatic hernia. J Pediatr Surg
and risk of respiratory diseases in newborns. Obstet Gynecol 2004;39:313—8 [discussion 13—8].
2001;97:439—42. [55] Finer NN, Barrington KJ. Nitric oxide for respiratory failure
[37] Jaillard S, Houfflin-Debarge V, Storme L. Higher risk of persis- in infants born at or near term. Cochrane Database Syst Rev
tent pulmonary hypertension of the newborn after cesarean. J 2006:CD000399.
Perinat Med 2003;31:538—9. [56] Kinsella JP, Neish SR, Ivy DD, Shaffer E, Abman SH. Clini-
[38] Pokela ML. Pain relief can reduce hypoxemia in distressed cal responses to prolonged treatment of persistent pulmonary
neonates during routine treatment procedures. Pediatrics hypertension of the newborn with low doses of inhaled nitric
1994;93:379—83. oxide. J Pediatr 1993;123:103—8.
[39] Houfflin-Debarge V, Delelis A, Jaillard S, et al. Effects of noci- [57] The Neonatal Inhaled Nitric Oxide Study Group (NINOS). Inhaled
ceptive stimuli on the pulmonary circulation in the ovine fetus. nitric oxide and hypoxic respiratory failure in infants with con-
Am J Physiol Regul Integr Comp Physiol 2005;288:R547—53. genital diaphragmatic hernia. Pediatrics 1997;99:838—45.
[40] Houfflin-Debarge V, Bresson S, Jaillard S, et al. Development [58] Noori S, Friedlich P, Wong P, Garingo A, Seri I. Cardiovascular
of a new model to investigate the fetal nociceptive pathways. effects of sildenafil in neonates and infants with congenital
Fetal Diagn Ther 2005;20:415—9. diaphragmatic hernia and pulmonary hypertension. Neonatol-
[41] Houfflin-Debarge V, Sicot B, Jaillard S, et al. The mechanisms ogy 2007;91:92—100.
of pain-induced pulmonary vasoconstriction: an experimental [59] Keller RL, Moore P, Teitel D, Hawgood S, McQuitty J, Fine-
study in fetal lambs. Anesth Analg 2007;104:799—806. man JR. Abnormal vascular tone in infants and children
[42] Alano MA, Ngougmna E, Ostrea Jr EM, Konduri GG. Analysis with lung hypoplasia: Findings from cardiac catheterization
of nonsteroidal antiinflammatory drugs in meconium and its and the response to chronic therapy. Pediatr Crit Care Med
relation to persistent pulmonary hypertension of the newborn. 2006;7:589—94.
Pediatrics 2001;107:519—23. [60] Steinhorn RH, Kinsella JP, Pierce C, et al. Intravenous silden-
[43] Gournay V, Savagner C, Thiriez G, Kuster A, Roze JC. Pulmonary afil in the treatment of neonates with persistent pulmonary
hypertension after ibuprofen prophylaxis in very preterm hypertension. J Pediatr 2009;155:841—7 [e1].
infants. Lancet 2002;359:1486—8. [61] Shah PS, Ohlsson A. Sildenafil for pulmonary hypertension in
[44] Delaney C, Gien J, Grover TR, Roe G, Abman SH. Pulmonary neonates. Cochrane Database Syst Rev 2011:CD005494.
vascular effects of serotonin and selective serotonin reuptake [62] Betremieux P, Gaillot T, de la Pintiere A, et al. Congenital
inhibitors in the late-gestation ovine fetus. Am J Physiol Lung diaphragmatic hernia: prenatal diagnosis permits immediate
Cell Mol Physiol 2011;301:L937—44. intensive care with high survival rate in isolated cases. A
[45] Fornaro E, Li D, Pan J, Belik J. Prenatal exposure to fluoxetine population-based study. Prenat Diagn 2004;24:487—93.
induces fetal pulmonary hypertension in the rat. Am J Respir [63] Levy M, Celermajer DS, Bourges-Petit E, Del Cerro MJ, Bajolle
Crit Care Med 2007;176:1035—40. F, Bonnet D. Add-on therapy with subcutaneous treprostinil
[46] Chambers CD, Hernandez-Diaz S, Van Marter LJ, et al. Selective for refractory pediatric pulmonary hypertension. J Pediatr
serotonin-reuptake inhibitors and risk of persistent pulmonary 2011;158:584—8.
hypertension of the newborn. N Engl J Med 2006;354:579—87. [64] Ivy DD, Parker TA, Ziegler JW, et al. Prolonged endothelin A
[47] Seri I. Circulatory support of the sick preterm infant. Semin receptor blockade attenuates chronic pulmonary hypertension
Neonatol 2001;6:85—95. in the ovine fetus. J Clin Invest 1997;99:1179—86.
[48] Bouissou A, Rakza T, Klosowski S, Tourneux P, Vanderborght [65] Goissen C, Ghyselen L, Tourneux P, et al. Persistent pulmonary
M, Storme L. Hypotension in preterm infants with signifi- hypertension of the newborn with transposition of the great
cant patent ductus arteriosus: effects of dopamine. J Pediatr arteries: successful treatment with bosentan. Eur J Pediatr
2008;153:790—4. 2008;167:437—40.

Das könnte Ihnen auch gefallen