You are on page 1of 12

Paediatric Respiratory Reviews 44 (2022) 19–30

Contents lists available at ScienceDirect

Paediatric Respiratory Reviews

Mini-symposium: What’s New in the NICU in 2022 (Part 2)

Fetal growth restriction and neonatal-pediatric lung diseases: Vascular


mechanistic links and therapeutic directions
Arvind Sehgal a,b,⇑, Theodore Dassios c,d, Marcel F. Nold a,b,e, Claudia A. Nold-Petry b,e, Anne Greenough c,f
a
Monash Newborn, Monash Children’s Hospital, Melbourne, Australia
b
Department of Pediatrics, Monash University, Melbourne, Australia
c
Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
d
Neonatal Intensive Care Centre, King’s College Hospital NHS Foundation Trust, London, United Kingdom
e
Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
f
NIHR Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, United Kingdom

Educational aims

The reader will come to appreciate that:

 An overview of the burden of disease for FGR and BPD.


 Vascular involvement in the placenta in FGR and BPD cohorts.
 Data from human FGR infants, and AGA infants with severe BPD, using high resolution vascular ultrasound.
 Therapeutic relevance of the overlapping vascular pathophysiology.
 The role of perinatal vascular insults in predisposition to pediatric age-group lung diseases.

a r t i c l e i n f o a b s t r a c t

Keywords: Bronchopulmonary dysplasia (BPD) is the most common respiratory sequela of prematurity, and infants
Fetal growth restriction born with fetal growth restriction (FGR) are disproportionately represented in BPD statistics, as factors
Bronchopulmonary dysplasia which affect somatic growth may also affect pulmonary growth. Effects of in-utero hypoxia underlying
Arterial stiffness FGR on lung parenchymal architecture predisposing to BPD are well documented, but the pulmonary vas-
Mechanisms
cular constructs are not well appreciated. Disruption of angiogenesis during critical periods of lung
Placenta
growth impairs alveolarization, contributing to BPD pathogenesis. Pulmonary artery thickness/stiffness
has been noted in FGR in the initial postnatal weeks, and also in well-grown infants with established
BPD. The lack of waveform cushioning by the major arteries exposes the pulmonary resistance vessels
to higher pulsatile stress, thereby accelerating microvascular disease. Reactive oxygen species, increased
sympathetic activity and endothelial dysfunction are common mediators in FGR and BPD; each putative
targets for prevention and/or therapeutics using interleukin (IL)-1 receptor antagonist (IL-1Ra), mela-
tonin or inhibition of renin–angiotensin–aldosterone system. While BPD is the archetypal respiratory dis-
ease of infancy, effects of FGR on pulmonary function are long-term, extending well into childhood. This
narrative links FGR in very/extremely preterm infants with BPD through the vascular affliction as a mech-
anistic and potentially, therapeutic pathway. Our objectives were to depict the burden of disease for FGR
and BPD amongst preterm infants, portray vascular involvement in the placenta in FGR and BPD cohorts,
provide high resolution vascular ultrasound information in both cohorts with a view to address therapeu-
tic relevance, and lastly, link this information with paediatric age-group lung diseases.
Ó 2022 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

INTRODUCTION
⇑ Corresponding author at: Neonatal Cardiovascular Research, Monash Children’s
Hospital, Adjunct Professor of Paediatrics, Monash University, 246, Clayton Road, Bronchopulmonary dysplasia (BPD) remains the most common
Clayton, Melbourne, VIC 3168, Australia. respiratory sequela of prematurity. For infants born <32 weeks
E-mail address: Arvind.Sehgal@monash.edu (A. Sehgal).

https://doi.org/10.1016/j.prrv.2022.09.002
1526-0542/Ó 2022 The Author(s). Published by Elsevier Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

gestational age (GA), BPD is defined as continued need for respira- pulmonary vascular development; these include FGR-fetal hypox-
tory support at 36 weeks post-menstrual age [1]. The NICHD clas- ia, pre-eclampsia, chorioamnionitis and exposure to intrauterine
sification divides BPD into moderate and severe categories based toxins produced by maternal smoking. Premature infants are still
on oxygen requirement of </30 % and positive pressure ventila- at a critical stage of lung maturation, making them particularly
tion [2]. A more recently published classification uses additional susceptible to pulmonary vascular injury mediated via hypoxic
parameters such as low-flow/high-flow nasal cannula oxygen, insults. Impairments in placental vascular perfusion further
nasal continuous positive airway pressure/nasal intermittent posi- increase the risk for development of BPD and pulmonary vascular
tive pressure ventilation and invasive positive pressure ventilation disease after birth [23–24]. This is corroborated by FGR sheep data
[3]. A significant proportion of infants with severe BPD are also which noted stunted pulmonary vascular growth, and impaired in-
affected by BPD associated pulmonary hypertension (PH) [4–6]. vitro pulmonary artery endothelial cell migration and nitric oxide
The additional complication contributes to a prolonged hospital production [25]. This in turn may be compounded by postnatal
stay, recurrent hospitalisations in the first two years as well as experiences such as episodes of hyperoxia, hypoxia, positive pres-
higher mortality. sure ventilation and postnatal infection.
Fetal growth restriction (FGR) is the inability of the developing The resulting respiratory morbidities are not limited to BPD, but
fetus to grow as per potential. In clinical terms, investigators have also include chronic oxygen dependency, lung function abnormal-
defined FGR as birthweight (BW) <10th centile for GA and sex [7]. ities at follow-up and rehospitalisation due to respiratory ailments
Abnormal Doppler patterns in fetal systemic arteries identify a during childhood. While FGR is the archetypal diagnosis that incor-
subset with especially poor short and long term outlooks. About porates cardiovascular and respiratory adaptation to chronic in-
two thirds of the late-onset small fetuses present severe forms of utero hypoxia, amongst premature infants, BPD is the archetypal
smallness, i.e. estimated fetal weight <3rd centile, or as abnormal setting for chronic post-natal hypoxia.
cerebro-placental ratio [8]. This group has features of vascular
mal-perfusion on placental pathology and poorer perinatal out-
SECTION A: BURDEN OF DISEASE
comes [9]. While most FGR infants are born preterm, the adverse
respiratory outcomes such as BPD attributable to the FGR state,
Data from Australian and New Zealand Neonatal Network noted
are independent of the degree of prematurity, indicating possible
that amongst infants <32 weeks in 2020, 668 (21 %) were born with
alignment of pathophysiology between FGR and BPD [10–11]. In
BW <10th centile for GA and sex; approximately 45 % amongst
a large multi-centric study on >1200 infants born <28 weeks GA,
them weighed <3rd centile [1]. In the same year, the incidence of
within all the antenatal, placental and neonatal strata variables,
BPD was high amongst infants <27 weeks GA (159/249, 64 %). A
preterm infants with FGR were at increased risk for BPD compared
3-year regional centre data in preterm infants <32 weeks GA noted
with appropriate for gestational age (AGA) infants [10]. This review
the incidence of FGR to be 27 % [26]. Table 1 summarises studies
focuses on very/extremely preterm FGR infants who develop BPD.
noting the incidence of FGR and the greater odds of developing BPD
in preterm FGR infants [26–33]. Combined, prematurity and FGR
increase the risk of BPD by 10–20 fold [34]. The Optum Neonatal
RATIONALE FOR REVIEW
database analysis of the results of 6708 infants from 717 US hospi-
tals demonstrated that the risk adjusted probability of dying or
FGR affects lung structure and function, impacting on both alve-
developing BPD or other morbidities amongst FGR infants was sim-
olar parenchymal and vascular components. The prevalent narra-
ilar to that of non-growth restricted infants who were born
tive linking in-utero events such as utero-placental insufficiency
approximately 2–3 weeks more immature [35]. BW <3rd centile
(UPI) with ‘subsequent’ respiratory sequela such as BPD is through
amongst extremely low BW infants increases the risk of developing
the involvement of pulmonary parenchyma and somatic growth.
BPD by five times [36]. A recent five-year study of all extremely
While the pathophysiology of BPD is multifactorial, the vascular
preterm infants admitted to neonatal intensive care in England
hypothesis behind it is increasingly being explored [12–13], The
noted that infants that developed BPD had a lower median BW z-
Barker Hypothesis, or the Developmental Origins of Health and
score (-0.44) compared to the ones without BPD (-0.23) (Fig. 1)
Disease Hypothesis, in its original proposition, explained fetal/de-
[37]. Research performed in singletons can be affected by a certain
velopmental priming of the cardiovascular system [14–15]. How-
level of bias caused by individual differences in genetic constitu-
ever, the underlying principles are equally relevant to the
tion, obstetric and maternal factors. However, recent data compar-
developing pulmonary vasculature [16]. In sheep, undernutrition
ing the larger and smaller twin in monochorionic twins provided
during the saccular and alveolar stage of lung development not
similar outcomes (Table 1) [33]. Finally, a meta-analysis of 211
only leads to prenatal altered surfactant protein expression and
studies demonstrated that when placental vascular dysfunction
reduction of the alveolar surface area in relation to lung volume,
was accompanied by FGR, it was associated with an increased risk
but also leads to decreased pulmonary vascular growth [17]. There
of developing BPD and PH [38].
are emerging experimental and clinical data linking in-utero events
with BPD via ‘pulmonary vascular’ structural and functional
changes (altered pulmonary vasoreactivity). SECTION B. OVERLAPPING VASCULAR AFFLICTION IN FGR AND
Angiogenesis is important for optimum alveolarization, espe- BPD COHORTS: PLACENTAL STUDIES
cially during critical periods of lung growth. Pulmonary alveolar
development is closely intertwined with vascular development to Vascular pathology in FGR placentae
the extent that specific disruption in alveolar development also
interferes with vascular development. Critical periods can occur Placental abnormalities (reduced feto-placental blood in UPI)
during fetal life where respective organs develop and differentiate are a major aetiology behind FGR [39–41]. Vascular adaptations
during sometimes-narrow windows of maturation. Impairments in are key in maintaining a healthy pregnancy with advancing gesta-
angiogenic signalling during critical fetal periods can reduce pul- tion. These include a drop in placental vascular resistance and the
monary vascular density, making them particularly susceptible to establishment of a high-flow circuit to optimise oxygen and nutri-
pulmonary vascular injury [13,18–21]. In essence, early-life vascu- ent delivery to the fetus. Angiogenesis and vasodilatation facilitate
lar disorders may impact alveolar development [12,22]. A wide this evolution and in severe cases of FGR, these adaptive mecha-
variety of prenatal conditions have been associated with impaired nisms fail, resulting in a persistently high placental vascular resis-
20
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

Table 1
Summary of studies noting higher incidence of BPD in FGR cohorts.

Study Subject Population Results Comment


Sehgal K, et al. 2021. (3 year Premature infants <32 weeks gestation. Greater risk of BPD when born FGR. Longer duration of respiratory support:
[2016–2018] single centre Of 973 infants, the incidence of FGR BPD: 53 (25.7 %) in FGR vs 28 (13.6 %) 37 ± 10 in FGR vs 23 ± 5 days in AGA
study) was 206 (27 %). 85 % FGR were born in AGA, (p = 0.002), respectively. (p = 0.016), home oxygen: 24 (11.6 %) in FGR
between 28 and 31+6 weeks.-FGR vs 8 (3.8 %) in AGA; (p = 0.005) after adjusting
(n = 206)-AGA (n = 206) for postnatal variables such as gestation, sex,
antenatal steroids, patent ductus arteriosus,
surfactant and infection.
Groene SG et al. 2021. (2010– 94 included pregnancies, median Incidence of BPD higher in the smaller Odds ratio to develop BPD in smaller twin 2.5
2019,single centre, gestation 32.4 (IQR 30.4–34.3) weeks. twin vs larger twin (16.7 % vs 6.7 %, (95 % CI 1.3–4.9).
retrospective) p = 0.008).
Boghossian NS et al. 2018. Data Data for preterm infants 23–29 weeks. BPD risk was increased with FGR, At 23 weeks (BPD relative risk: 1.84; 95 % CI:
from 2006 to 2014 on 156,587 FGR incidence/gestation starting at GA 23 weeks through 1.30–2.60); at 27 weeks (BPD relative risk:
infants from 852 American 9.95 % overall 27 weeks GA. 3.56; 95 % CI: 3.04–4.18).
centres: Vermont Oxford 10.1 % at 22 weeks;
Network. 9.4 % at 23 weeks;
10.2 % at 24 weeks;
10.2 % at 25 weeks;
9.7 % at 26 weeks;
10.% at 27 weeks;
10.% at 28 weeks;
9.9 % at 29 weeks.
Klinger G et al. 2013. Population- Preterm infants born between 24 and BPD was independently associated Greater odds for BPD when FGR (2.65, 95 % CI,
based observational study 32 weeks, surviving to 36 weeks. with FGR state. 2.24–3.12).
from Israel Neonatal Network
for data from 2000 to 2010.
Qiu X et al. 2012. Data from the Incidence of FGR amongst <33 weeks Greater risk for mortality and BPD. Adjusted odds for BPD (1.78; 95 % CI, 1.48–
Canadian Neonatal Network GA infants: 1249/11,909 (10.5 %). 2.13).
Ferdynus C, et al. 2009. (Regional Premature infants 28–31 weeks Greater risk of BPD when born FGR. FGR associated with a significantly increased
study-Burgundy, France [2000 gestation (n = 65). risk of BPD (adjusted odds: 2.6, CI 1.3–5.2).
to 2006]) 30.4 % infants were FGR-AGA-
(n = 52) -FGR
(n = 13)
Bose C, et al. 2009. (14-center, Premature infants <27 weeks gestation Increased risk for BPD when FGR Among antenatal factors, birthweight
observational study [2002– (n = 411) 25 % of infants without FGR provided the most information about BPD
2004]) -No FGR (n = 335)-Moderate FGR* 60 % of infants with moderate FGR risk. FGR was the only maternal or antenatal
(n = 55) -Severe FGR^ 90 % of infants with severe FGR characteristic which highly predicted BPD,
(n = 21) even after adjusting for other risk factors.
Aucott SW et al. 2004. Comparison between preterm infants Greater BPD in FGR (29 %) vs AGA Number of days on ventilator, FGR 12 ± 31 vs
Retrospective, single centre <32 weeks-FGR infants (15 %). AGA 6 ± 15.
(1991to1998) (n = 39) -AGA Number of days in oxygen, FGR 34 ± 53 vs
(n = 41) AGA 16 ± 27
Garite TJ et al. 2004. (Regional Singleton, inborn infants, 23–34 weeks Each measure of FGR was associated Respiratory support at 28 days of life:
data, 124 neonatal units [1997 gestation. with " mortality and greater BPD (need -No FGR: 4,068 (17.8 %) -FGR definition 1: 287
to 2001]) Compared FGR per 3 definitions: - for respiratory support at 28 days of (19.8 %) -FGR definition 2: 708
antenatally diagnosed FGR age) (24.1 %) -Either definition 1 or 2: 831
(1451 [4.8 %] of total); (22.4 %)
-birthweight <10th centile (2936
[9.8 %] of total); -either of the above
(3,708 [12.3 %] of total).
Lal MK et al. 2003. (Regional n = 401 (9.9 % of infants  32 weeks) FGR infants had higher mortality Odds of FGR infants developing BPD at
observational study-Trent, <28 days and 36 weeks postmenstrual 28 days’ and 36 weeks’ PMA (OR: 1.34, 95 %
United Kingdom [1995–1999]) age (PMA). Greater risk of developing CI: 1.03–1.74; and OR: 1.87, 95 % CI: 1.39 –
BPD compared to reference group. 2.51), respectively.
Reiss et al. 2003. (Regional data, Preterm infants <32 weeks of gestation. Mortality and BPD were significantly Mortality (23 % in FGR vs 11 % in AGA), BPD
Hesse, Germany [1990 to <32 weeks gestation. higher in FGR. (28 % in FGR vs 14 % in AGA). BPD odds
1996]) Analyse differences FGR and AGA. -FGR ratio = 3.80, 95 % CI 2.11, 6.84).
(n = 183) -AGA
(n = 1,182)
Bardin C et al. 1997. Comparison between preterm infants Higher risk for BPD (defined as a need Prolonged need for oxygen supplementation
Retrospective, single centre (24–266/7 weeks) -FGR for supplementary oxygen at 36 weeks and ventilator support (94 days for FGR vs
study (1983–1991) (n = 37) -AGA (65 % for FGR vs 32 % for AGA). 68 days for AGA, and 58 days for FGR vs
(n = 147) 40 days for AGA, respectively).

BPD-bronchopulmonary dysplasia, FGR-fetal growth restriction, *moderate FGR-birthweight <10th centile, ^severe FGR-birthweight.
<3rd centile, CI-confidence interval.

tance [39,42]. In comparison to human AGA placentae, the FGR pla- are pathognomonic features of hypoxia and reperfusion [44–45].
centae have significantly higher resistance, endothelial dysfunction Vascular aetiology (villous hypoplasia, increased syncytial knots)
and reduced flow-mediated vasodilatation [43]. underlies reduced supply of nutrients and oxygen in human FGR
Maternal Vascular Under-perfusion (MVU) is a group of histo- pregnancies [46]. This is corroborated by data from sheep models
logic lesions seen in births complicated by FGR, and affect flow of chronic fetal hypoxia and UPI where vascular remodelling with
dynamics such as high placental vascular resistance (Fig. 2). On small lumens and thick walls have been noted [25]. Chronic fetal
histopathology, vascular remodelling/ obstructed spiral arteries hypoxia owing to UPI, as indicated by the extent/severity of

21
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

AGA, mean GA 30 weeks in either group) using predefined


histopathological characteristics (Amsterdam Placental Workshop
Group Consensus Statement) [51–52], vascular mal-adaptations
in the FGR cohort included MVU, fetal mal-perfusion and acceler-
ated villous maturation. Of interest to the clinicians, the preterm
FGR cohort in this study had significantly longer duration of respi-
ratory support requirements and BPD, compared to gestation and
sex matched AGA infants [52]. In essence, vasculature was a key
‘organ’ to be affected in pregnancies affected by FGR. Fig. 3 depicts
antenatal and postnatal vascular constructs in FGR and BPD and
their influence on lung disease.

Vascular origins of BPD-clues from placental histopathology and cord


blood biomarkers

Placental histopathology and biomarkers have also been inves-


tigated in BPD for early clues of subsequent neonatal respiratory
events, with significant histopathology overlapping findings with
FGR. A study on 56 preterm AGA infants evaluated specific placen-
tal histopathological changes to assess their relationship with BPD
associated PH. Placental as well as echocardiographic assessments
(latter at 36 weeks post-menstrual age) were done by investigators
masked to the grouping (PH or no PH) [23]. The presence of placen-
tal features consistent with MVU increased the relative risk of sub-
Fig. 1. Boxplot of the birthweight z-score in infants that developed BPD compared
to infants that did not. The horizontal lines represent the 5th, 25th, 50th, 75th and sequent BPD-associated PH by 2.75 (95 % CI 1.56 to 4.85, P = 0.004);
95th centile of the values, outliers are depicted as single points. 11,806 included the significance persisting after adjustment for GA. Mestan et al.
infants. [24] noted similar findings in a 5-year retrospective cohort study
of premature infants with mean GA 26 weeks. Amongst the com-
MVU, may explain the associations with adverse infant outcomes
mon complications of prematurity, BPD was the only outcome that
[47–49]. MVU is associated with >4 times higher risk of delivering
was increased with MVU, persisting after adjustment for perinatal
an FGR infant [50]. In human data, where placental histopathology
variables (adjusted odds ratio 2.6; 95 % confidence interval [CI] 1.4,
was evaluated in a cohort of 40 preterm infants (20 FGR and 20
4.8). The antenatal vascular constructs of BPD were corroborated

Fig. 2. Placental histopathology in fetal growth restriction Fig. 2a. Maternal vascular perfusion: Placental infarct, H and E, original magnification Fig. 2b. Fetal vascular
malperfusion: villous stromal-vascular karyorrhexis (old term hemorrhagic endovasculitis), H and E, original magnification 100 Fig. 2c. Chronic intervillositis, H and E,
original magnification 400 Fig. 2d. Microscopic chorangioma, H and E, original magnification 50 (from, Sehgal A, Dahlstrom J, Chan Y, Allison BJ, Miller S, Polglase GR.
Placental histopathology in preterm fetal growth restriction. J Pediatr Ch Health 2019).

22
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

Fig. 3. Antenatal and postnatal vascular constructs in FGR and BPD and their influence on lung disease.

by the findings of significantly lower cord blood angiogenic factors monary dynamics in response to minimally invasive surfactant ther-
in placental MVU [53]. These included placental growth factor and apy (administered via a catheter followed by continuous positive
vascular endothelial growth factor-A. Decreased levels in the pla- airway pressure) was compared between FGR and AGA infants [55].
centa were predictive of subsequent BPD-PH. Based on the above In this study on preterm infants (mean GA 28 weeks in either group),
results, placental MVU may identify premature infants with BPD the baseline assessments at median age of 3 h noted higher PVR and
who were exposed to chronic intrauterine hypoxia–ischemia dur- lower right ventricular contractility in FGR infants, compared to
ing fetal life. AGA infants. The evolution of flow dynamics after surfactant noted a
In summary, placental histopathology and cord blood biomark- significantly lesser reduction in PVR in the FGR cohort, indicating that
ers indicate vascular origins of respiratory sequelae, and MVU is an FGR infants adapt differently in early postnatal life. Potentially, the
important common finding between FGR and BPD. altered pulmonary vascular mechanics reflect maladaptive changes
in response to UPI. A recent prospective study evaluated right pul-
monary artery in 40 preterm infants (mean GA 30 weeks in either
SECTION C. ARTERIAL STIFFENING IN FGR AND BPD COHORTS:
group) using colour flow tissue Doppler imaging, comparing infants
POSTNATAL DATA
with FGR with GA, gender and age matched AGA infants [13]. Vascular
assessments performed in the second week of life noted significantly
Vascular cross-sectional area is an important determinant of pul-
greater pulmonary artery wall thickness and reduced pulsatility in
monary vascular resistance (PVR). Putatively, a reduction of gas
infants with FGR, compared to AGA infants (Figs. 4 and 5). FGR infants
exchange area because of vascular remodelling may necessitate an
also had a persistently higher PVR. This high pulsatile stress is trans-
increase in pulmonary artery pressure to maintain pulmonary blood
mitted to pulmonary arterioles. As postulated earlier, this may lead
flow. Arterial stiffening due to remodelling contributes significantly
to microvascular leakage and decreased lung compliance. As evidence
to increased afterload and is an important index of disease progres-
of ventriculo-arterial coupling in these preterm infants, right ventric-
sion. The role of proximal conduit arteries such as main or branch
ular systolic function was also impaired in FGR infants [13]. This is not
pulmonary arteries is to dampen the pressure oscillations originat-
surprising as the cardiac inflow tract, pulmonary vascular smooth
ing from right ventricular ejection force. On vascular ultrasound,
muscle, and proximal vascular endothelium all arise from a common
increased thickness, and reduced pulsatility and distensibility may
in-utero multipotent cardiopulmonary mesoderm progenitor. Essen-
indicate impaired ability to dampen these waveforms. At the organ
tially, insults to the developing pulmonary vasculature could also
level, this exposes the pulmonary resistance vessels to higher pul-
affect the right ventricle directly or via back pressure changes [56].
satile stress, thereby accelerating microvascular disease [54]. Over
Antenatal hypoxia in particular, results in a cascade of maladaptive
a period, this may cause decreased pulmonary compliance and pul-
consequences, including altered pulmonary vasoreactivity and pul-
monary oedema. The resultant increased requirements for ventila-
monary artery muscularization [57]. Plausibly, involvement of the
tor support and baro/volutrauma contribute to a higher incidence
pulmonary vasculature is contributory towards a higher incidence
of BPD amongst preterm FGR infants.
of BPD in preterm FGR infants.

Arterial stiffening in preterm FGR cohorts


Arterial stiffening in non-FGR severe BPD cohorts: pulmonary
Recent studies using high-definition vascular ultrasound in human circulation and right heart
newborns have evaluated dynamic pulmonary vascular properties in
infants with FGR, as influences on perinatal transition. In a prospective Using Synchrotron micro-CT imaging in a murine model of BPD
echocardiographic study, the evolution of cardiac function and pul- and BPD-PH, we recently demonstrated that in addition to
23
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

Fig. 4. Pulmonary artery imaging using 2-dimensional and colour Doppler. A, 2-D image from modified short axis view showing the right pulmonary artery. B, colour Doppler
showing the right pulmonary artery. C, m-mode tissue Doppler of right pulmonary artery (from, Sehgal A, Gwini SM, Menahem S, Allison BJ, Miller S, Polglase GR. Preterm
growth restriction and bronchopulmonary dysplasia: the vascular hypothesis and related physiology. J Physiol. 2019).

Fig. 5. Right pulmonary artery. Scatterplot illustrating thickness of the right pulmonary artery. (from, Sehgal A, Gwini SM, Menahem S, Allison BJ, Miller S, Polglase GR.
Preterm growth restriction and bronchopulmonary dysplasia: the vascular hypothesis and related physiology. J Physiol. 2019).

dramatic rarefaction of pulmonary blood vessels of a small diame- ture architecture and vasoreactivity in infants with BPD set the
ter (4–7 lm) the percentage of larger vessels (40–60 lm) was up to tone for the development of higher PVR, and eventually, PH and
9-fold increased in mice with BPD-PH [58]) (Fig. 6. Whilst this right ventricular dysfunction. In a prospective echocardiographic
observation is yet to be explored in the FGR setting, this disease- study on preterm infants with severe BPD compared with those
related abnormality may contribute to the dynamic components with no BPD (comparable GA at birth; 26 weeks), infants with
of changes in the pulmonary circulation, which reflect as raised BPD had higher PVR and significantly lower right ventricular sys-
PVR. Reduced pulmonary vasculature density, abnormal vascula- tolic and diastolic function on tissue Doppler imaging [59].

24
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

Fig. 6. Representative reconstructed synchrotron micro-CT image of pulmonary vascular injury in a neonatal murine model bronchopulmonary dysplasia-pulmonary
hypertension. Filament filling is colour coded for diameter size highlighting the rarefaction of small pulmonary vessels and increase in the larger vessels (40–60 lm) in 28 day
old pups housed in either air (A) or hyperoxia (B) (from, Bui CB, Kolodziej M, Lamanna E, Elgass K, Sehgal A, Rudloff I, Schwenke DO, Tsuchimochi H, Kroon MAGM, Cho SX,
Maksimenko A, Cholewa M, Berger PJ, Young MJ, Bourke JE, Pearson JT, Nold MF, Nold-Petry CA. Interleukin-1 Receptor Antagonist Protects Newborn Mice Against Pulmonary
Hypertension. Front Immunol. 2019).

Systemic circulation and left heart higher systolic BP was noted at assessments done in the second
week of life [60]. Data on infants with BPD is limited but has noted
Systemic arteries (carotid/aorta) are similarly affected in FGR similarly results [65]. Fifty-seven infants with severe BPD were
cohorts in the initial weeks of postnatal life [60–63]. In a prospec- compared with 114 GA matched infants with no BPD. The inci-
tive echocardiographic study on preterm FGR infants, compared to dence of hypertension (systolic BP  95th centile) was higher in
AGA infants (GA 30 weeks in either group), aorta intima–media the BPD cohort vs no BPD cohort (40.3 % vs 2.6 %, p < 0.001). Higher
thickness, wall stiffness and peripheral resistance were noted to BP was associated with significantly longer duration of respiratory
be significantly greater in FGR infants [60]. Similar to the right support.
heart involvement, left ventricular systolic function was impaired In essence, vascular properties in the two cohorts (FGR and
with significant correlations between vascular mechanics and car- BPD) are quite similar, with demonstration of thickening and stiff-
diac function. There is evidence of similar involvement of the sys- ness. The overlapping vascular pathophysiology indicates a semi-
temic vasculature in preterm infants with severe BPD. A nal role for arterial remodelling and functional impairments in
prospective vascular ultrasound study evaluated aortic dynamic disease pathogenesis, making the mechanisms, attractive targets
properties in preterm AGA infants with severe BPD and equally for pharmacological interventions.
preterm infants with no BPD (GA 26 weeks in either group) [64].
Both, input impedance and stiffness index were significantly
higher in infants with BPD. The significance persisted after adjust- SECTION D. MECHANISTIC LINKS AND THE THERAPEUTIC
ing for GA and BW separately, and GA-BW combined. From the RELEVANCE OF OVERLAPPING VASCULAR PATHOPHYSIOLOGY.
clinical perspective, both populations have noted higher blood
pressure (BP) recordings [60,65]. In a prospective study on FGR Table 2 depicts overlapping mechanistic links between FGR and
and AGA infants (mean GA 30 weeks in both cohorts), significantly BPD; many of them could be potential therapeutic targets. Investi-

Table 2
Vascular mechanistic links between FGR & BPD.

 Maternal vascular under-perfusion on placental histopathology


 Chronic hypoxemia induced SMc proliferation and fibroblasts in the adventitia ? muscularization of precapillary ‘resistance vessels’
 Low VEGFA and VEGFR2 in PAECs (indicating ; alveolar and vascular growth)
 Thickened distal air space walls, ; pulmonary vascular growth ? ; pulmonary compliance and diffusing capacity
 ; elastin expression and elastin fibre deposition in the thickened ECM and blood gas barrier (replaced with collagen which is 100  stiffer)
 ; angiogenic signalling through direct inhibition of VEGF activity at birth ? ; lung eNOS protein content
 Pulmonary artery endothelial injury/dysfunction? ; production of nitric oxide
 Alterations in arterial extracellular matrix (; arterial elastin, " expression of metalloproteases, SMc proliferation) ? Thicker, stiffer arteries with narrow lumen ?
‘Adaptive dysmorphic pattern of vascular organization’
 Paucity of capillaries within the walls of the thinned abnormally enlarged alveoli
 Deficient endothelial progenitor cells in cord blood of infants who subsequently develop moderate or severe BPD; ; in cord blood of mothers with pre-eclampsia-
FGR
 Oxidative damage by reactive oxygen species

FGR-Fetal growth restriction, BPD-Bronchopulmonary dysplasia, SMc-Smooth muscle cell, ECM-Extracellular matrix, eNOS-Endothelial nitric oxide synthase, VEGFA- Vas-
cular endothelial growth factor A, VEGFR2- Vascular endothelial growth factor receptor, PAEC-Pulmonary artery endothelial cell.

25
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

gator interest in vasculature as a focus of therapeutics in the Sildenafil has been used for management of BPD associated PH,
human FGR cohorts has centred primarily on vasodilators and the incidence of which ranges from 17 to 40 % [70–72]). In addition
vasomodulators. Fig. 7 summarizes various therapeutic to its physiological effects detailed above, sildenafil also preserves
interventions. lung angiogenesis, and decreases PVR and media wall thickness
[73]. Its property of reducing pulmonary inflammatory response
and fibrin deposition makes it a physiologically very suited to
Pulmonary circulation/right heart focussed interventions BPD disease physiology management [74]. Echocardiographic and
clinical efficacy of sildenafil was evaluated in 22 infants (mean
Vasodilator GA 25 weeks) with BPD associated PH. Significant improvement
Phosphodiesterase-5 is produced in the lungs and breaks down in echocardiographic parameters of pH were noted, alongside
cyclic guanosine monophosphate. Sildenafil decreases the activity reduction in respiratory support requirements [72]. Mourani
of Phosphodiesterase-5, thereby increasing the levels of cyclic gua- et al. described 25 subjects (median GA at birth: 28 weeks) with
nosine monophosphate, which decreases pulmonary pressures by BPD who were administered with sildenafil at the age of <2 years
causing vasodilatation and relaxation. Nitric oxide, released from [75]. Only approximately half (13 [52 %]) the patients had paired
fetal trophoblasts, is a potent vasodilator by way of increasing cyc- echocardiographic data which showed significant reduction in pul-
lic adenosine monophosphate. However, decreased release is seen monary pressures. A recent systematic review and meta-analysis
in pregnancies complicated by pre-eclampsia or FGR, which evaluated the effectiveness of chronic use of sildenafil in preterm
explains the rationale to use sildenafil as a therapeutic option infants with BPD-associated PH. Five studies were included, yield-
[66]. However, a systematic review combining in-vitro studies, ing a total of 101 patients, with a pooled mean GA of 26 weeks. Pul-
experimental animal studies and human studies on women with monary arterial pressure improved by >20 % in majority of cases
fertility disorders was inconclusive [67]. A recent randomized con- within 1–6 months [76]. While physiologically appropriate in both
trolled trial investigating the effect of sildenafil (75 mg/day) com- settings (FGR and BPD), greater refinement of this promising ther-
pared with placebo in pregnancies with severe FGR showed no apeutic intervention is warranted to optimise gestation and dose at
differences on the main outcome of prolongation of pregnancy. A administration, as well as outcomes to be studied.
non-significant 2.3 % increase in BW was noted [68]. However, clin-
ical efficacy of the drug’s primary vasodilatory effect in these situ-
ations has not been systematically evaluated. Trapani et al. further Other therapies
tested this hypothesis in a trial of 100 women with early-onset Antioxidant therapy with Vitamin C and E improves endothelial
pre-eclampsia who were randomised to 50 mg sildenafil or pla- function and flow-mediated vasodilatation, and attenuates hypoxic
cebo, thrice daily. This study demonstrated lower mean BP and PH, suggesting a reversible component in dysregulation [77]. Mela-
improved utero-placental resistance, although no corresponding tonin is a neurohormone with antioxidant and vasodilator proper-
improvement in neonatal outcomes [69]. ties which are active at the pulmonary level. Recent data noted that

Fig. 7. Summary of various therapeutic interventions.

26
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

daily administration of melatonin mitigated PH and vascular thickening and reduced pulsatility of pulmonary and systemic
remodelling in chronically hypoxic rats. Melatonin treatment also arteries has been seen in FGR infants [13].
significantly attenuated right ventricular systolic pressures, thick- Although the physiological role of ACE inhibition in human FGR
ness of the arteriolar wall, and oxidative and inflammatory mark- cohorts has not yet been explored, recent work in sheep has
ers in the hypoxic animals [78]. Gonzaléz-Candia recently noted demonstrated proof of concept. This explains the physiological
that postnatal treatment with melatonin during the neonatal per- rationale that ACE inhibition may mitigate hyperfiltration. A sheep
iod in lambs with chronic hypoxia decreased pulmonary oxidative model of a single functioning kidney was administered enalapril
stress by inducing antioxidant enzymes and improved pulmonary for a period of 5 weeks [92], and compared to a control group.
vascular reactivity [79]. In essence, melatonin seems a promising ACE inhibition prevented albuminuria, resulted in approximately
drug which may stop, or possibly reverse these vascular effects, 22 % higher renal blood flow and approximately 35 % lower filtra-
and needs further exploration in human studies. Furthermore, tion fraction compared with vehicle-treated sheep. A greater
inflammation, and specifically the potent inflammatory cytokine recruitment of renal function reserve indicated a reduction in
IL-1, is now recognised as a key pathogenic culprit in BPD and glomerular hyper-filtration–mediated kidney dysfunction.
BPD-PH. Interleukin-1 receptor antagonist (IL-1Ra) is a natural pro- Increased urinary nitrite in ACE treated animals indicated elevated
tein that blocks IL-1, and has been safely used as a drug (Anakinra) nitric oxide bioavailability, which likely contributed to improve-
to treat many other diseases. Preclinical studies by us and others ments in kidney function and prevention of albuminuria. In
have shown that blocking inflammation, specifically the potent humans, nephrogenesis is complete before birth, and perinatal pro-
pro-inflammatory IL-1 with Anakinra, early and effectively, holds gramming influences reduction of nephron number and increased
great promise for preventing BPD, BPD-PH and also improving neu- renal apoptosis [93]. A poorer renal endowment, accompanied by
rodevelopmental outcomes [80–83]. Anakinra is currently being systemic arterial thickness in FGR could explain ongoing microal-
investigated for safety and feasibility in a Phase I/IIa dose escala- buminuria by way of glomerular hyper-filtration [94]. In essence,
tion trial. Importantly, there is a growing body of preclinical evi- ACE aimed at vascular stiffness has therapeutic benefits in FGR
dence highlighting the role of IL-1 driven inflammation in pathology as well. The above preliminary animal and human data
placental insufficiency and FGR. Interleukin-1b is elevated in the indicates the seminal role of arterial stiffness (and directed thera-
serum from women with FGR during the third trimester of preg- peutic potential) in disease physiology.
nancy [84]. Placental levels of IL-1Ra were found to negatively cor- In summary, a range of therapeutic options are currently under
relate with BW and GA in high risk pregnancies [85]. investigation. Efficacy of these interventions has the potential to
significantly change the neonatal disease paradigm.

Systemic circulation/left heart focussed interventions


SECTION E: PERINATAL VASCULAR INSULTS AND
PREDISPOSITION TO PEDIATRIC LUNG DISEASES
Vasomodulator
Angiotensin converting enzyme (ACE) inhibitors have pharma-
Earlier in the review we elaborated the range of pulmonary vas-
cological properties beyond being effective anti-hypertensive
cular architectural and functional impairments in preterm FGR
agents. By way of improving endothelial function, bradykinin
infants in response to perinatal insults. However, a significant com-
mediated vasodilatation, endothelial nitric oxide release, and
ponent of pulmonary vascular development occurs after birth,
change in extracellular matrix, they are physiologically very suited
making postnatal insults during infancy and childhood, important
to the proposed FGR-BPD pathophysiology of heightened renin-
contributors to abnormal lung development.
angiotensin-aldosterone system activity. The inhibition of renin-
angiotensin-aldosterone system by angiotensin receptor blockers
and ACE inhibitors could modify the inflammatory response in Long-term lung development: effects of prenatal factors and
the arterial wall [86–87]. accompanying hypoxia
Recent data has postulated the effect of systemic hypertension
and left heart ventricular dysfunction in BPD pathophysiology In a retrospective cohort study of 247 mother-neonatal pairs,
[88–89]. Termed as ‘post-capillary pathophysiology, enough left pre-eclampsia was associated with BPD, in part mediated by FGR
ventricular afterload may be generated, leading to back-pressure [95]. Abnormal vascular endothelial growth factor signalling might
cascade, raised end-diastolic left atrial pressure, increased pul- play a critical role as it has been described in the pathogenesis of
monary venous pressure leading and pulmonary edema. Address- pre-eclampsia [96] and in the ‘vascular hypothesis’ of BPD [12–
ing this mechanism, we administered captopril (ACE inhibition) 13,97]. Third trimester fetal blood flow patterns might inform
to extremely preterm infants with ‘severe’ BPD (GA and weight childhood respiratory health. In a population based prospective
at birth, 23–29 weeks and 505–814 g, respectively) [90]. All infants study of 903 children in whom Doppler measurements had been
required mechanical ventilation and had no improvement with made at a median GA of 30.3 weeks, persistently higher PVR and
postnatal steroids, sildenafil and diuretics. Captopril was adminis- a higher risk of late/persistent wheezing was noted at six years
tered for systemic hypertension and left heart dysfunction. After a of age [98]. Fetal inflammation is also implicated in FGR, while
five-week interval (required to reach steady-state), a significant hypoxia itself may give rise to inflammation. In one study, 36
reduction in oxygen requirements was noted. This prospective women with FGR fetuses had higher peripheral blood lymphocyte
study of AGA infants also demonstrated a reduction in aorta levels of pro-inflammatory cytokines Interleukin-6, Tumour necro-
intima-media thickness, and improved cardiac function and arte- sis Factor-a, and Interleukin-12 compared to 22 normal AGA preg-
rial pulsatility [90]. Interestingly, histopathology review of autop- nancies [99]. Arguably, management of fetal inflammation has the
sies with diagnosis of BPD over a 10-year period noted that potential to improve metabolic dysfunction and potentially, lung
capillary ACE endothelial staining is largely absent in BPD lungs developmental impairment.
[91]. This suggests that the improvements in respiratory support Maternal smoking during pregnancy is also associated with
requirements and cardiovascular parameters (especially PVR) in FGR; impact on growth trajectories persisting up to six months
our study were primarily mediated via the systemic (rather than after birth [100]. In a study evaluating 90 FGR and 180 AGA sub-
direct pulmonary) effects of ACE inhibition. This data warrants fur- jects, maternal smoking exposure was independently associated
ther exploration of ACE inhibition in selected cohorts as similar with asymmetric FGR, possibly indicating deleterious effects on
27
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

placental health [101]. Data from epidemiological and experimen- dren had lower mean FEV1, and FGR state was negatively
tal studies indicates the disruptive effects of intra-uterine toxin associated with FEV1 (B = 0.66; P = 0.004) [112].
exposure (including tobacco smoke) on pulmonary development In essence, disrupted vascular development in FGR alters pul-
[102–103]. Gonzales-Luis and colleagues performed a systematic monary function and respiratory outlook trajectory, well into the
review of studies reporting on tobacco smoking during pregnancy childhood. Secondary insults such as smoking and viral infections
being a potential risk factor for BPD [104]. A significant association can further impact pulmonary vascular endowment, and accentu-
between tobacco smoking during pregnancy and BPD at 36 weeks ate early ageing.
was noted (17 studies, RR 1.126, 95 % CI 1.008–1.259, p = 0.036).
Infants with severe BPD are known to have a high incidence of
CONCLUSIONS
PH (39 %) [4]. It is possible that maternal smoking during preg-
nancy might affect pulmonary development and predispose to a
From the cardiovascular perspective, multiple mechanistic links
higher incidence of persistent PH. Nicotinic acetylcholine receptors
between FGR and systemic hypertension have been explored.
on cells of the pulmonary vessel walls possibly mediate morpho-
Increasingly, a longitudinal and close association of lung health
logical alterations in fetal pulmonary vasculature. Experimental
with cardiovascular health in individuals born FGR is being consid-
studies in timed pregnant rhesus monkeys treated with nicotine
ered biologically plausible. Epidemiological and pathological link-
noted significantly greater thickness of the total wall and tunica
ages underscore the biological plausibility of vascular links
adventitia in airway associated vessels [105]. Nicotine exposure
between FGR state and BPD. The adverse respiratory outlook attri-
also increased collagen deposition (100 stiffer than elastin) and
butable to FGR and its independence from degree of prematurity
contrastingly, lower levels of elastin. Putatively, smoking during
(and linear trajectory with FGR severity), points towards possible
pregnancy may lead to nicotine being transported across the pla-
overlapping pathophysiology between FGR and BPD. Studies
centa and directly interacting with nicotinic receptors in pul-
depicting longitudinal and cross-sectional data provide distinctive
monary vessels and cause vascular structural alterations. Such
evidence for the notion that cardiac/ vascular function and respira-
experimental data strongly points towards a causal linkage [105].
tory function in these high-risk cohorts may be interlinked, and
In summary, infants born FGR already have poor lung reserve
affected well into adulthood. Understanding the pathways, and
due to lung parenchymal and vascular alterations. Interactions
the consequent therapeutic pointers, can significantly improve
with antenatal smoking further exacerbated by postnatal expo-
the clinical outlook of this cohort.
sures, may adversely affect long-term respiratory trajectory.

DIRECTIONS FOR FUTURE RESEARCH


Long term effects-population data
From the clinical perspective, further longitudinal data from
The above postulations are corroborated by population based neonatal networks are better placed in delineating the true impact
studies. In a twin study examining >500 children, the forced expi- of FGR. From an investigation point of view, arterial thickness is
ratory volume in the first second (FEV1) results post- being used in adults as an end-point in clinical trials assessing
bronchodilator therapy were significantly negatively associated the impact of antihypertensive medications on cardiovascular risk
with fetal growth [106]. Approximately 40 % of this cohort was [113–114]. Whether similar end-points could be used for experi-
preterm born; associations for FEV1 and forced vital capacity with mental trials using ACE inhibition in human FGR, remains to be
GA were non-significant. However, there was significant associa- tested. From the therapeutic perspective, choosing the appropriate
tion between restricted fetal growth and post-bronchodilator intervention and the timeline (in-utero: to prevent disease or after-
FEV1 and forced vital capacity. In a population based prospective birth: to reduce disease severity) needs prospective analysis.
cohort study of approximately 5000 children in The Netherlands,
school age children who had higher airway resistance had lower Declaration of Competing Interest
fetal growth and weight gain, and were more likely to have physi-
cian diagnosed asthma [107]). Approximately 5 % of this cohort The authors declare that they have no known competing finan-
was born preterm. Information from studies assessing older cial interests or personal relationships that could have appeared
cohorts is consistent with the above findings. Ronkainen and co- to influence the work reported in this paper.
workers studied 88 preterm-born children at a mean age of
11 years and reported that in preterm-born children exposed to References
FGR, both FEV1 and diffusing capacity for carbon monoxide were
lower compared to preterm-born children with appropriate in- [1] Chow SSW, Creighton P, Chambers GM, Lui K. Report of the Australian and
utero growth after adjustment for a diagnosis of BPD [108]. In a New Zealand Neonatal Network 2019. Sydney: ANZNN; 2021.
[2] Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J Respir Crit Care Med
cross-sectional study of 74 adolescents who were premature 2001;163:1723–9.
(<32 weeks GA) and had FGR, lower FEV1 (88.7 % versus 95.9 %) [3] Jensen EA, Dysart K, Gantz MG, McDonald S, Bamat NA, Keszler M, et al. The
and lower forced vital capacity (88.2 % versus 95.5 %) in compar- diagnosis of bronchopulmonary dysplasia in very preterm infants an
evidence-based approach. Am J Respir Crit Care Med 2019;200(6):751–9.
ison to those well grown at birth was noted [109]. Other investiga- [4] Revanna GK, Kunjunju A, Sehgal A. Bronchopulmonary dysplasia associated
tors however, have suggested that amongst children born very pulmonary hypertension: making the best use of bedside echocardiography.
preterm, and studied between 8 and 15 years of age, the impact Prog Pediatr Cardiol 2017;46:39–43.
[5] An HS, Bae EJ, Kim GB, Kwon BS, Beak JS, Kim EK, et al. Pulmonary
of FGR may be due to the higher risk of BPD in that group [110]. Hypertension in Preterm Infants with Bronchopulmonary Dysplasia. Korean
Harris et al. studied 159 subjects of 16–19 years of age born Circ J 2010;40:131–6.
<29 weeks of gestation, of whom 37 had FGR. Even at this later [6] Bhat R, Salas AA, Foster C, Carlo WA, Ambalavanan N. Prospective analysis of
pulmonary hypertension in extremely low birth weight infants. Pediatrics
age, the FGR cohort had poorer spirometric indices and exercise
2012;129:e682–9.
capacity compared with those with adequate fetal growth [111]. [7] Fenton TR, Kim JH. A systematic review and meta-analysis to revise the
In more recent data, the influence of FGR on lung function was Fenton growth chart for preterm infants. BMC Pediatr 2013;13:59. https://
tested in children born very preterm. Twenty-eight preterm infants doi.org/10.1186/1471-2431-13-59.
[8] Figueras F, Gratacós E. Update on the diagnosis and classification of fetal
born <32 weeks GA were cross-sectionally evaluated and com- growth restriction and proposal of a stage-based management protocol. Fetal
pared with 67 AGA infants at 8–15 years of age. Preterm-FGR chil- Diagn Ther 2014;36:86–98.

28
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

[9] Parra-Saavedra M, Crovetto F, Triunfo S, Savchev S, Peguero A, Nadal A, et al. Child Fetal Neonatal Ed 2021;106:386–91. https://doi.org/10.1136/
Figueras Placental findings in late-onset SGA births without Doppler signs of archdischild-2020-320816.
placental insufficiency. Placenta 2013;34:1136–41. [38] Pierro M, Villamor-Martinez E, van Westering-Kroon E, Alvarez-Fuente M,
[10] Bose C, Van Marter LJ, Laughon M, O’Shea TM, Allred EN, Karna P, et al. Fetal Abman SH, et al. Association of the dysfunctional placentation endotype of
growth restriction and chronic lung disease among infants born before the prematurity with bronchopulmonary dysplasia: a systematic review, meta-
28th week of gestation. Pediatrics 2009;124:e450–8. analysis and meta-regression. Thorax 2021 [Epub ahead of print]. 10.1136/
[11] Gortner L, Misselwitz B, Milligan D, Zeitlin J, Kollee L, Boerch K, et al. Rates of thoraxjnl-2020-216485.
bronchopulmonary dysplasia in very preterm neonates in Europe. [39] Chaddha V, Viero S, Huppertz B, Kingdom J. Developmental biology of the
Neonatology 2011;99:112–7. https://doi.org/10.1159/000313024. placenta and the origins of placental insufficiency. Semin Fetal Neonatal Med
[12] Abman SH. Bronchopulmonary dysplasia: ‘‘A vascular hypothesis”. Am J 2004;9:357–69.
Respir Crit Care Med 2001;164:1755–6. [40] Sibley CP, Turner MA, Cetin I, Ayuk P, Boyd CAR, D’Souza SW, et al. Placental
[13] Sehgal A, Gwini SM, Menahem S, Allison BJ, Miller S, Polglase GR. Preterm phenotypes of intrauterine growth. Pediatr Res 2005;58:827–32.
growth restriction and bronchopulmonary dysplasia: the vascular hypothesis [41] Kiserud T, Kessler J, Ebbing C, Rasmussen S. Ductus venosus shunting in
and related physiology. J Physiol 2019;597:1209–20. growth-restricted fetuses and the effect of umbilical circulatory compromise.
[14] Barker DJ. Fetal origins of coronary heart disease. BMJ 1995;311:171–4. Ultrasound Obstet Gynecol 2006;28:143–9.
[15] Barker DJ. The origins of the developmental origins theory. J Intern Med [42] Sala C, Campise M, Ambroso G, Motta T, Zanchetti A, Morganti A. Atrial
2007;261:412–7. natriuretic peptide and hemodynamic changes during normal human
[16] Maron BA, Abman SH. Translational advances in the field of pulmonary pregnancy. Hypertension 1995;25:631–6.
hypertension: focusing on developmental origins and disease inception for [43] Kingdom J, Huppertz B, Seaward G, Kaufmann P. Development of the
the prevention of pulmonary hypertension. Am J Respir Crit Care Med placental villous tree and its consequences for fetal growth. Eur J Obstet
2017;195:292–301. Gynecol Reprod Biol 2000;92:35–43.
[17] Arigliani M, Spinelli AM, Liguoro I, Cogo P. Nutrition and lung growth. [44] Brosens I, Pijnenborg R, Vercruysse L, The RR. ‘‘great obstetrical syndromes”
Nutrients 2018;10:919. https://doi.org/10.3390/nu10070919. are associated with disorders of deep placentation. Am J Obstet Gynecol
[18] Orgeig S, Crittenden TA, Marchant C, McMillen IC, Morrison JL. Intrauterine 2011;204:193e201.
growth restriction delays surfactant protein maturation in the sheep fetus. [45] Kovo M, Schreiber L, Bar J. Placental vascular pathology as a
Am J Physiol Lung Cell Mol Physiol 2010;298:L575–83. mechanism of disease in pregnancy complications. Thromb Res
[19] Maritz GS, Morley CJ, Harding R. Fetal growth restriction has long-term 2013;131:S18–21.
effects on postnatal lung structure in sheep. Pediatr Res 2004;55:287–95. [46] Veerbeek JH, Nikkels PG, Torrance HL, et al. Placental pathology in early
[20] Maritz GS, Cock ML, Louey S, Suzuki K, Harding R. Early developmental origins intrauterine growth restriction associated with maternal hypertension.
of impaired lung structure and function. Early Hum Dev 2005;81:763–71. Placenta 2014;35:696–701.
[21] O’Brien EA, Barnes V, Zhao L, McKnight RA, Yu X, Callaway CW, et al. [47] Hansen AR, Barnes CM, Folkman J, McElrath TF. Maternal preeclampsia
Uteroplacental insufficiency decreases p53 serine-15 phosphorylation in predicts the development of bronchopulmonary dysplasia. J Pediatr
term IUGR rat lungs. Am J Physiol Regul Integr Comp Physiol 2007;293(1): 2010;156:532–6.
R314–22. [48] Tang JR, Karumanchi SA, Seedorf G, Markham N, Abman SH. Excess soluble
[22] Jakkula M, Le Cras TD, Gebb S, Hirth KP, Tuder RM, Voelkel NF, et al. Inhibition vascular endothelial growth factor receptor-1 in amniotic fluid impairs lung
of angiogenesis decreases alveolarization in the developing rat lung. Am J growth in rats: linking preeclampsia with bronchopulmonary dysplasia. Am J
Physiol Lung Cell Mol Physiol 2000;279:L600–7. Physiol Lung Cell Mol Physiol 2012;302:L36–46.
[23] Kunjunju A, Gopagondanahalli K, Chan Y, Sehgal A. Bronchopulmonary [49] Vinnars MT, Nasiell J, Holmstrom G, Norman M, Westgren M,
dysplasia associated pulmonary hypertension: clues from placental Papadogiannakis N. Association between placental pathology and neonatal
pathology. J Perinatol 2017;37:1310–4. outcome in preeclampsia: a large cohort study. Hypertens Pregnancy
[24] Mestan KK, Check J, Minturn L, Yallapragada S, Farrow KN, Liu X, et al. 2014;33:145–58.
Placental pathologic changes of maternal vascular underperfusion in [50] Wright E, Audette MC, Ye XY, et al. Maternal vascular malperfusion and
bronchopulmonary dysplasia and pulmonary hypertension. Placenta adverse perinatal outcomes in low-risk nulliparous women. Obstet Gynecol
2014;35:570–4. 2017;130:1112–20.
[25] Rozance PJ, Seedorf GJ, Brown A, et al. Intrauterine growth restriction [51] Khong TY, Mooney EE, Ariel I, et al. Sampling and definitions of placental
decreases pulmonary alveolar and vessel growth and causes pulmonary lesions Amsterdam placental workshop group consensus statement. Arch
artery endothelial cell dysfunction in vitro in fetal sheep. Am J Physiol Lung Pathol Lab Med 2016;140:698–713.
Cell Mol Physiol 2011;301:L860–71. [52] Sehgal A, Dahlstrom J, Chan Y, Allison BJ, Miller S, Polglase GR. Placental
[26] Sehgal K, Sehgal K, Tan K. Increased incidence of chronic lung disease and histopathology in preterm fetal growth restriction. J Pediatr Ch Health
respiratory sequelae in growth restricted versus appropriately grown 2019;55:582–7.
preterms. Ind Journal Child Health 2021;8:280–3. [53] Mestan KK, Gotteiner N, Porta N, Grobman W, Su EJ, Ernst LM. Cord blood
[27] Ferdynus C, Quantin C, Abrahamowicz M, Platt R, Burguet A, Sagot P, et al. Can biomarkers of placental maternal vascular underperfusion predict
birth weight standards based on healthy populations improve the bronchopulmonary dysplasia-associated pulmonary hypertension. J Pediatr
identification of small-for-gestational-age newborns at risk of adverse 2017;185:33–41.
neonatal outcomes? Pediatrics 2009;123(2):723–30. [54] Wang Z, Chesler NC. Pulmonary vascular wall stiffness: An important
[28] Qiu X, Lodha A, Shah PS, Sankaran K, Seshia MMK, Yee W, et al. Neonatal contributor to the increased right ventricular afterload with pulmonary
outcomes of small for gestational age preterm infants in Canada. Am J hypertension. Pulm Circ 2011;1:212–23.
Perinatol 2012;29:87–94. [55] Sehgal A, Bhatia R, Roberts CT. Cardiovascular response and sequelae after
[29] Boghossian NS, Geraci M, Edwards EM, Horbar JD. Morbidity and mortality in minimally-invasive surfactant therapy in growth-restricted preterm infants. J
small for gestational age infants at 22 to 29 weeks’ gestation. Pediatrics Perinatol 2020;40:1178–84.
2018;141:e20172533. [56] Peng T, Tian Y, Boogerd CJ, Lu MM, Kadzik RS, Stewart KM, et al. Coordination
[30] Lal MK, Manktelow BN, Draper ES, Field DJ. Chronic lung disease of of heart and lung co-development by a multipotent cardiopulmonary
prematurity and intrauterine growth retardation: a population-based study. progenitor. Nature 2013;500:589–92.
Pediatrics 2003;111:483–7. [57] Papamatheakis DG, Blood AB, Kim JH, Wilson SM. Antenatal hypoxia and
[31] Reiss I, Landmann E, HeckmannM MB, Gortner L. Increased risk of pulmonary vascular function and remodeling. Curr Vasc Pharmacol
bronchopulmonary dysplasia and increased mortality in very preterm 2013;11:616–40.
infants being small for gestational age. Arch Gynecol Obstet 2003;269:40–4. [58] Lao JC, Bui CB, Pang MA, Cho SX, Rudloff I, Elgass K, et al. Type 2 immune
[32] Klinger G, Sokolover N, Boyko V, Sirota L, Lerner-Geva L, Reichman B. Israel polarization is associated with cardiopulmonary disease in preterm infants.
Neonatal Network. Perinatal risk factors for bronchopulmonary dysplasia in a Sci Transl Med. 2022;14(639):eaaz8454. 10.1126/scitranslmed.aaz8454.
national cohort of very-low-birthweight infants. Am J Obstet Gynecol Epub 2022 Apr 6.
2013;208(115):e1–9. https://doi.org/10.1016/j.ajog.2012.11.026. [59] Sehgal A, Malikiwi A, Paul E, Tan K, Menahem S. Right ventricular function in
[33] Groene SG, Spekman JA, Te Pas AB, Heijmans BT, Haak MC, van Klink JM, et al. infants with Bronchopulmonary Dysplasia: Association with respiratory
Respiratory distress syndrome and bronchopulmonary dysplasia after fetal sequelae. Neonatology 2016;109(4):289–96.
growth restriction: lessons from a natural experiment in identical twins. E- [60] Sehgal A, Allison B, Gwini S, Miller S, Polglase G. Cardiac morphology and
Clin Med 2021;32:. https://doi.org/10.1016/j.eclinm.2021.100725100725. function in preterm growth restricted infants: relevance for clinical sequelae.
[34] Rosenberg A. The IUGR newborn. Semin Perinatol 2008;32:219–24. J Pediatr 2017;188:128–34.
[35] Jensen EA, Foglia EE, Dysart KC, Simmons RA, Aghai ZH, Cook A, et al. Adverse [61] Sehgal A, Crispi F, Skilton M, De Boode W. Clinician performed ultrasound in fetal
effects of small for gestational age differ by gestational week among very growth restriction: fetal, neonatal, pediatric aspects. J Perinatol 2017;37:1251–8.
preterm infants. Arch Dis Child Fetal Neonatal Ed 2019;104:F192–8. https:// [62] Sehgal A, Murthy P, Dahlstrom JE. Vascular changes in fetal growth restriction:
doi.org/10.1136/archdischild-2017-314171. clinical relevance and future therapeutics. J Perinatol 2019;39:366–74.
[36] Soudée S, Vuillemin L, Alberti C, Mohamed D, Becquet O, Farnoux C, et al. [63] Sehgal A, Allison B, Gwini S, Menahem S, Miller S, Polglase G. Vascular ageing
Fetal growth restriction is worse than extreme prematurity for the and cardiac maladaptation in growth restricted preterm infants. J Perinatol
developing lung. Neonatology 2014;106:304–10. https://doi.org/10.1159/ 2018;38(1):92–7.
000360842. [64] Sehgal A, Malikiwi A, Paul E, Tan K, Menahem S. Systemic arterial stiffness in
[37] Dassios T, Williams EE, Hickey A, Bunce C, Greenough A. Bronchopulmonary infants with bronchopulmonary dysplasia: Potential cause of systemic
dysplasia and postnatal growth following extremely preterm birth. Arch Dis hypertension. J Perinatol 2016;36:564–9.

29
A. Sehgal, T. Dassios, M.F. Nold et al. Paediatric Respiratory Reviews 44 (2022) 19–30

[65] Sehgal A, Elsayed K, Nugent M, Varma S. Sequelae associated with systemic [91] Castro EC, Parks WT, Galambos C. The ontogeny of human pulmonary
hypertension in infants with severe bronchopulmonary dysplasia. J Perinatol angiotensin-converting enzyme and its aberrant expression may contribute
2022;42:775–80. to the pathobiology of bronchopulmonary dysplasia. Pediatr Pulmonol
[66] Motta-Mejia C, Kandzija N, Zhang W, Mhlomi V, Cerdeira AS, Burdujan A, 2014;49:985–90.
et al. Placental vesicles carry active endothelial nitric oxide synthase and [92] McArdle Z, Singh RR, Bielefeldt-Ohmann H, Moritz KM, Schreuder MF, Denton
their activity is reduced in preeclampsia. Hypertension 2017;70:372–81. KM. Brief early life angiotensin-converting enzyme inhibition offers
[67] Villanueva-Garcia D, Mota-Rojas D, Hernandez-Gonzalez R, Sanchez-Aparicio renoprotection in sheep with a solitary functioning kidney at 8 months of
P, Alonso-Spilsbury M, Trujillo-Ortega M, et al. A systematic review of age. J Am Soc Nephrol 2022; ASN.2021111534. 10.1681/ASN.2021111534.
experimental and clinical studies of sildenafil citrate for intrauterine growth Online ahead of print.
restriction and pre-term labour. J Obstet Gynaecol 2007;27:255e9. [93] Puddu M, Fanos V, Podda F, Zaffanello M. The kidney from prenatal to adult
[68] Sharp A, Cornforth C, Jackson R, Harrold J, Turner MA, Kenny LC, et al. life: perinatal programming and reduction of number of nephrons during
Maternal sildenafil for severe fetal growth restriction (STRIDER): a development. Am J Nephrol 2009;30(2):162–70.
multicentre, randomised, placebo-controlled, double-blind trial. Lancet [94] Zanardo V, Fanelli T, Weiner G, Fanos V, Zaninotto M & Visentin S. Intrauterine
Child Adolesc. Health 2018;2:93–102. growth restriction is associated with persistent aortic wall thickening and
[69] Trapani Jr A, Goncalves LF, Trapani TF, Vieira S, Pires M, Pires MM. Perinatal glomerular proteinuria during infancy. Kidney Int 2011; 80: 119-123.77.
and hemodynamic evaluation of sildenafil citrate for preeclampsia [95] Wilmink FA, Reijnierse J, Reiss IKM, Steegers EAP, de Jonge RCJ. Research
treatment: a randomized controlled trial. Obstet Gynecol 2016;128:253–9. Consortium Neonatology South-West of the Netherlands. Preeclampsia and
[70] Bhat R, Salas AA, Foster C, Carlo WA, Ambalavanan N. Prospective analysis of risk of developing bronchopulmonary dysplasia in very preterm neonates.
pulmonary hypertension in extremely low birth weight infants. Pediatrics Pregnancy Hypertens 2019;15:57–63. https://doi.org/10.1016/j.
2012; 129:e682–e689. preghy.2018.11.001.
[71] Samiee-Zafarghandy S, Smith PB, van den Anker JN. Safety of sildenafil in [96] Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, et al. Circulating
infants. Pediatr Crit Care Med 2014;15:362–8. angiogenic factors and the risk of preeclampsia. New Engl J Med
[72] Tan K, Krishnamurthy MB, O’Heney JL, Paul E, Sehgal A. Sildenafil therapy in 2004;350:672–83.
bronchopulmonary dysplasia associated pulmonary hypertension: A [97] Bhatt AJ, Pryhuber GS, Huyck H, Watkins RH, Metlay LA, Maniscalco WM.
retrospective study of efficacy and safety. Eur J Pediatr 2015;174:1109–15. Disrupted pulmonary vasculature and decreased vascular endothelial growth
[73] Ladha F, Bonnet S, Eaton F, Hashimoto K, Korbutt G, The’baud B. Sildenafil factor, Flt-1, and TIE-2 in human infants dying with bronchopulmonary
improves alveolar growth and pulmonary hypertension in hyperoxia-induced dysplasia. Am J Respir Crit Care Med 2001;164:1971–80.
lung injury. Am J Respir Crit Care Med 2005;172:750–6. [98] Kooijman MN, van Meel ER, Steegers EAP, Reiss IKM, de Jongste JC, Jaddoe VWV,
[74] de Visser YP, Walther FJ, Laghmani EH, Boersma H, van der Laarse A, et al. Fetal umbilical, cerebral and pulmonary blood flow patterns in relation to
Wagenaar GTM. Sildenafil attenuates pulmonary inflammation and fibrin lung function and asthma in childhood. The Generation R Study. Pediatr Allergy
deposition, mortality and right ventricular hypertrophy in neonatal Immunol 2019;30:443–50. https://doi.org/10.1111/pai.13044.
hyperoxic lung injury. Respir Res 2009;10:30. [99] Al-Azemi M, Raghupathy R, Azizieh F. Pro-inflammatory and anti-
[75] Mourani PM, Sontag MK, Ivy DD, Abman SH. Effects of long-term sildenafil inflammatory cytokine profiles in fetal growth restriction. Clin Exp Obstet
treatment for pulmonary hypertension in infants with chronic lung disease. J Gynecol 2017;44:98–103.
Pediatr 2009;154:379–84. [100] de Brito ML, Nunes M, Bernardi JR, Bosa VL, Goldani MZ, da Silva CH. Somatic
[76] van der Graaf M, Rojer LA, Helbing WA, Reiss IKM, Etnel JRG, et al. Sildenafil growth in the first six months of life of infants exposed to maternal smoking in
for bronchopulmonary dysplasia and pulmonary hypertension: a meta- pregnancy. BMC Pediatr 2017;17:67. https://doi.org/10.1186/s12887-017-0819-7.
analysis. Pulm Circ 2019; 9(3): 2045894019837875. [101] Delpisheh A, Brabin L, Drummond S, Brabin BJ. Prenatal smoking exposure
[77] Rimoldi SF, Sartori C, Rexhaj E, Bailey DM, de Marchi SF, McEneny J, et al. and asymmetric fetal growth restriction. Ann Hum Biol 2008;35(6):573–83.
Antioxidants improve vascular function in children conceived by assisted [102] Wagijo MA, Sheikh A, Duijts L, Been JV. Reducing tobacco smoking and smoke
reproductive technologies: A randomized double-blind placebo-controlled exposure to prevent preterm birth and its complications. Paediatr Respir Rev
trial. Eur J Prev Cardiol 2015;22:1399–407. 2017;22:3–10.
[78] Hung MW, Yeung HM, Lau CF, Poon AMS, Tipoe GL, et al. Melatonin [103] Gibbs K, Collaco JM, McGrath-Morrow SA. Impact of tobacco smoke and
attenuates pulmonary hypertension in chronically hypoxic rats. Int J Mol Sci nicotine exposure on lung development. Chest 2016;149:552–61.
2017; 18: 1125; 10.3390/ijms18061125. [104] González-Luis GE, van Westering-Kroon E, Villamor-Martinez E, Huizing MJ,
[79] Gonzaléz-Candia A, Candia AA, Figueroa EG, Feixes E, Gonzalez-Candia C, Kilani MA, Kramer BW, et al. Tobacco smoking during pregnancy is associated
Aguilar SA, et al. Melatonin long-lasting beneficial effects on pulmonary with increased risk of moderate/severe bronchopulmonary dysplasia: a
vascular reactivity and redox balance in chronic hypoxic ovine neonates. J systematic review and meta-analysis. Front Pediatr 2020;8:160. https://doi.
Pineal Res 2020;68:e12613. org/10.3389/fped.2020.00160.
[80] Bui CB, Kolodziej M, Lamanna E, Elgass K, Sehgal A, Rudloff I, et al. [105] Sekhon HS, Proskocil BJ, Clark JA, Spindel ER. Prenatal nicotine exposure
Interleukin-1 receptor antagonist protects newborn mice against increases connective tissue expression in foetal monkey pulmonary vessels.
pulmonary hypertension. Front Immunol 2019;11(10):1480. https://doi.org/ Eur Respir J 2004;23:906–15.
10.3389/fimmu.2019.01480. [106] Örtqvist AK, Ullemar V, Lundholm C, Kuja-Halkola R, Magnusson PKE,
[81] Rudloff I, Cho SX, Bui CB, McLean C, Veldman A, Berger PJ, et al. Refining anti- Lichtenstein P, et al. Fetal growth and childhood lung function in the
inflammatory therapy strategies for bronchopulmonary dysplasia. J Cell Mol Swedish twin study on prediction and prevention of asthma. Ann Am Thorac
Med 2017;21(6):1128–38. Soc 2017;14:1147–53. https://doi.org/10.1513/AnnalsATS.201611-908OC.
[82] Nold MF, Mangan NE, Rudloff I, Cho SX, Shariatian N, Samarasinghe TD, et al. [107] Sonnenschein-van der Voort AM, Gaillard R, de Jongste JC, Hofman A, Jaddoe
Interleukin-1 receptor antagonist prevents murine bronchopulmonary VW, Duijts L. Foetal and infant growth patterns, airway resistance and
dysplasia induced by perinatal inflammation and hyperoxia. Proc Natl Acad school-age asthma. Respirology 2016;21:674–82.
Sci 2013;110(35):14384–9. [108] Ronkainen E, Dunder T, Kaukola T, Marttila R, Hallman M. Intrauterine
[83] Kelly SB, Stojanovska V, Zahra VA, Moxham A, Miller SL, Moss TJM, et al. growth restriction predicts lower lung function at school age in children born
Interleukin-1 blockade attenuates white matter inflammation and very preterm. Arch Dis Child Fetal Neonatal Ed 2016;101:F412–7. https://doi.
oligodendrocyte loss after progressive systemic lipopolysaccharide org/10.1136/archdischild-2015-308922.
exposure in near-term fetal sheep. J Neuroinflammation 2021;18(1):189. [109] Arroyas M, Calvo C, Rueda S, Esquivias M, Gonzalez-Menchen C, Gonzalez-
https://doi.org/10.1186/s12974-021-02238-4. Carrasco E, et al. Asthma prevalence, lung and cardiovascular function in
[84] Berbets A, Koval H, Barbe A, Albota O, Yuzko O. Melatonin decreases and adolescents born preterm. Sci Rep 2020;10:19616. https://doi.org/10.1038/
cytokines increase in women with placental insufficiency. J Matern Fetal s41598-020-76614-0.
Neonatal Med 2021;34(3):373–8. [110] Arigliani M, Stocco C, Valentini E, De Pieri C, Castriotta L, Ferrari ME, et al.
[85] Girard S, Heazell AEP, Derricott H, Allan SM, Sibley CP, Abrahams VM, et al. Lung function between 8 and 15 years of age in very preterm infants with
Circulating cytokines and alarmins associated with placental inflammation in fetal growth restriction. Pediatr Res 2012;90:657–63. https://doi.org/
high-risk pregnancies. Am J Reprod Immunol 2014;72(4):422–34. 10.1038/s41390-020-01299-0.
[86] Mervaala EM, Muller DN, Park JK, Schmidt F, Löhn M, Breu V, et al. Monocyte [111] Harris C, Lunt A, Bisquera A, Peacock J, Greenough A. Intrauterine growth
infiltration and adhesion molecules in a rat model of high human renin retardation and lung function of very prematurely born young people. Pediatr
hypertension. Hypertension 1999;33:389–95. Pulmonol 2021;56:2284–91. https://doi.org/10.1002/ppul.25359.
[87] Dransfield MT, Cockcroft JR, Townsend RR, Coxson HO, Sharma SS, Rubin DB, [112] Arigliani M, Stocco C, Valentini E, De Pieri C, Castriotta L, Ferrari ME, et al.
et al. Effect of fluticasone propionate/salmeterol on arterial stiffness in Lung function between 8 and 15 years of age in very preterm infants with
patients with COPD. Respir Med 2011;105:1322–30. fetal growth restriction. Pediatr Res 2021;90:657–63.
[88] Sehgal A, Malikiwi A, Paul E, Tan K, Menahem S. A new look at [113] Zanchetti A, Bond MG, Hennig M, Neiss A, Mancia G, Dal Palu C, et al. Calcium
Bronchopulmonary dysplasia: Post-capillary pathophysiology and cardiac antagonist lacidipine slows down progression of asymptomatic carotid
dysfunction. Pulm Circ 2016;6(4). https://doi.org/10.1086/688641. atherosclerosis: principal results of the European Lacidipine Study on
[89] Sehgal A, Steenhorst JJ, Mclennan DI, Merkus D, Ivy D, McNamara PJ. The left Atherosclerosis (ELSA), a randomized, double-blind, long-term trial.
heart, systemic circulation and bronchopulmonary dysplasia: relevance to Circulation 2002;106:2422–7.
pathophysiology and therapeutics. J Pediatr 2020;225:13–22. [114] Lorenz MW, von Kegler S, Steinmetz H, Markus HS, Sitzer M. Carotid intima-
[90] Sehgal A, Krishnamurthy MB, Clark M, Menahem S. ACE inhibition for severe media thickening indicates a higher vascular risk across a wide age range:
bronchopulmonary dysplasia-An approach based on physiology. Physiol Rep prospective data from the Carotid Atherosclerosis Progression Study (CAPS).
2018;6(17):e13821. https://doi.org/10.14814/phy2.13821. Stroke 2006;37:87–92.

30

You might also like