Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Bronchopulmonary Dysplasia: Clinical Perspective

Deepak Jain and Eduardo Bancalari*

Since Northway’s original description of BPD almost 45 years ago, the clinical With better understanding of the complex and multifactorial pathogenesis of
presentation of BPD has evolved into a disease process, which mostly BPD, it is quite clear that any single therapy is very unlikely to eliminate this
involves extremely premature infants. This new form of BPD is the result of problem unless it reduces prematurity. Further development in prevention
multiple antenatal and postnatal factors that can cause injury to the and treatment of BPD will likely need a multi-pronged strategy with novel
developing lung leading to altered alveolar and vascular development. Over therapeutic agents acting at various stages of the disease process.
the years, there has been considerable increase in knowledge of factors that
contribute to the development of BPD. This has led to different strategies for Birth Defects Research (Part A) 100:134–144, 2014.
prevention as well as management of BPD. Some of these strategies have C 2014 Wiley Periodicals, Inc.
V
been successful and have withstood the test of clinical trials, such as vitamin
A supplementation, post-natal steroids, caffeine, and volume targeted Key words: BPD; clinical presentation; prevention; management; premature
ventilation. The evidence for other interventions has been weak or negative.

Introduction This final stage consisted of severe respiratory failure with


There have been significant advancements in the field of a radiographic picture characterized by increased densities
neonatal and perinatal medicine since Northway and col- secondary to fibrosis and atelectasis and adjacent large
leagues described “bronchopulmonary dysplasia” in their emphysematous areas (Northway et al., 1967) (Fig. 1).
original study almost 45 years ago. Although these
advancements have markedly increased survival rates of NEW BPD

extremely premature infants, BPD continues to be one of Over the years with increasing use of antenatal steroids,
the most common complications in these infants with inci- surfactant replacement therapy, and refinement in assisted
dence ranging from 25 to 42% in very low birth weight ventilation methods, the severe form of BPD has become
infants (Walsh et al., 2004; Stoll et al., 2010). less frequent. As more immature preterm infants are sur-
Since its description, the clinical presentation of BPD viving and care of preterm infants is improving, BPD now
has changed significantly with the original severe BPD in is seen most commonly in extremely low birth weight
relatively bigger infants being replaced by a milder form infants with structurally immature lungs and underdevel-
in extremely preterm infants with a very different clinical oped pulmonary vasculature. These infants typically have
and radiographic course. There has also been significant mild initial respiratory disease, which improves quickly
progress in the understanding of factors contributing to with noninvasive respiratory support or surfactant treat-
lung injury and repair, which can lead to development of ment. Within a few days, they are with minimal ventilator
preventive and therapeutic strategies in the near future. settings requiring low FiO2 or are extubated to noninva-
sive respiratory support. A large proportion of these
Clinical Presentation infants subsequently have progressive respiratory deterio-
When Northway and colleagues first introduced the term ration requiring increasing respiratory support, which may
BPD, they described the clinical, radiographic, and patho- be due to development of a symptomatic patent ductus
logic course of a group of infants with severe respiratory arteriosus (PDA) or pulmonary infections. These infants
distress syndrome (RDS). All of these infants had severe may go on to require mechanical ventilation and oxygen
respiratory failure and were ventilated with high pressures for several weeks or months but a large majority of them
and high oxygen concentrations. They had a high mortality are essentially symptom free at the time of discharge
rate (59%), and a significant number developed cor (Bancalari et al., 2003). This chronic lung disease process
pulmonale. has been termed as “New BPD” to differentiate from the
The respiratory course of these infants was described process described by Northway, “Original BPD.” The radio-
in different stages evolving from initial RDS to a final stage graphic picture in infants with “New BPD” usually pro-
of chronic lung disease at approximately 1 month of age. gress from hazy ground glass-opacity, which clears after
surfactant treatment to a hazy opacification by 1 to 2
weeks of age and eventually to a relatively uniform pat-
University of Miami, Miami, Florida tern of coarse interstitial opacities (Fig. 2) (Agrons et al.,
2005).
*Correspondence to: Eduardo Bancalari, University of Miami, PO Box 016960
(R-131), Miami, FL 33101. E-mail: ebancalari@med.miami.edu
SEVERE BPD

Published online 27 February 2014 in Wiley Online Library (wileyonlinelibrary. There remains a small group of preterm infants who have
com). Doi: 10.1002/bdra.23229 severe initial RDS, lung hypoplasia, or pneumonia who do

C 2014 Wiley Periodicals, Inc.


V
BIRTH DEFECTS RESEARCH (PART A) 100:134–144 (2014) 135

(Abman et al., 2008). These infants have increased pulmo-


nary vascular resistance and abnormal vascular reactivity,
leading to excessive vasoconstrictor response to acute
hypoxia (Abman et al., 1985; Mourani et al., 2004).
Clinically these changes in lung architecture lead to a
varying degree of hypoxemia, which increases during the
periods of physical activity, feeding, or pulmonary infec-
tions and edema. They also commonly have hypercapnia
secondary to alveolar hypoventilation and ventilation-
perfusion mismatch. This leads to compensatory metabolic
alkalosis, which is often exaggerated by the use of loop
diuretics.

Prevention of BPD
Bronchopulmonary dysplasia is the end result of multiple
factors that lead to injury to the developing lung and pul-
monary vasculature. Therefore, effective prevention of BPD
requires a multifactorial approach, which avoids factors
that are injurious to the lung and vasculature and pro-
motes normal growth and development of the lung. Over
the years, multiple strategies have been tried for decreas-
FIGURE 1. Original BPD with areas of hyperinflation and emphysema with ing the incidence of BPD with variable success (Table 1).
adjacent dense areas of atelectasis. Some of them, which have been more successful, are dis-
cussed here.

not respond adequately to surfactant administration. These ANTENATAL STEROIDS


infants continue to require mechanical ventilation with Corticosteroids are given prenatally to promote maturation
high airway pressure and inspired oxygen concentration. of the surfactant system and reduce the incidence and
Many of these infants develop air leak syndrome, second- severity of RDS (NIH Consensus Development Panel,
ary lung infections, and pulmonary edema secondary to 1995). Treatment with antenatal corticosteroids has been
inflammation or a PDA further aggravating the lung dam- associated with overall reduction in RDS (relative risk
age. These infants sometimes develop a radiographic pic- [RR], 0.66; 95% confidence interval [CI], 0.59–0.73), and
ture characteristic of “Original BPD” and are more likely to moderate to severe RDS (RR, 0.55; 95% CI, 0.43–0.71).
develop pulmonary hypertension and signs of right heart However, treatment with antenatal steroids has failed to
failure. Except for a minority, most of these infants can show statistically significant reduction in BPD (RR, 0.86;
eventually be weaned off from respiratory support but
require supplemental oxygen for prolonged periods of
time and many are discharged home on oxygen.

PULMONARY FUNCTION IN BPD


Infants with established BPD have varying degrees of
altered lung architecture due to disrupted alveolar and
vascular development. The infants with severe BPD com-
monly have increased airway resistance due to mucosal
hyperplasia, tracheomalacia, and bronchomalacia, resulting
in increased time constant and dead space ventilation.
There is heterogeneous damage to distal airways and dis-
tal airspaces, resulting in variable time constant for differ-
ent parts of the lungs. This can lead to areas of over
inflation and atelectasis and ventilation-perfusion mis-
match. These infants also have decreased functional resid-
ual capacity and lung compliance.
One of the major factors leading to pulmonary morbid-
ities associated with BPD is decreased pulmonary vascular FIGURE 2. New BPD with generalized homogenous opacities with an interstitial
growth and excessive pulmonary vascular remodeling pattern.
136 BRONCHOPULMONARY DYSPLASIA

approaches, early surfactant administration should be con-


TABLE 1. Strategies for Prevention of BPD sidered in all extremely low birth weight infants with RDS,
who require endotracheal intubation for management of
Proven strategies from randomized clinical trials: respiratory failure.
1. Caffeine

2. Vitamin A NONINVASIVE VENTILATION

3. Volume targeted ventilation


One of the major contributing factors for development of
BPD is injury to the premature lung due to respiratory
4. Postnatal Steroids
support required to sustain life. Therefore, use of noninva-
Strategies with equivocal evidence from clinical studies: sive ventilation, if it can provide adequate gas exchange,
1. Antenatal steroids may decrease the incidence of BPD. This was first shown
2. Exogenous surfactant by Avery et al. who described a lower incidence of BPD in
centers with highest use of continuous positive airway
3. Early PDA closure
pressure (CPAP) and avoidance of mechanical ventilation
4. High frequency ventilation
(Avery et al., 1987). Since then, numerous clinical trials
5. Non invasive respiratory support have examined use of early CPAP instead of mechanical
6. Inhaled nitric oxide ventilation on incidence of BPD with no significant reduc-
7. Reduced oxygen exposure
tion in incidence of BPD with use of early CPAP (Verder
et al., 1994; Morley et al., 2008). Recently, a meta-analysis
of studies evaluating strategies to avoid endotracheal
95% CI, 0.61–1.22) (Roberts and Dalziel, 2006). This could mechanical ventilation did show a small but statistically
be due to increased number of preterm infants at risk of significant reduction in incidence of BPD or death (odds
BPD being salvaged with use of antenatal steroids. ratio, 0.83; 95% CI, 0.71–0.96) by avoiding endotracheal
There has been conflicting evidence regarding the mechanical ventilation (Fischer and Buhrer, 2013).
advantage of multiple courses of antenatal steroids against Most clinical trials performed to evaluate use of early
single course. There are some animal data suggesting that CPAP have shown that 50 to 80% of these infants develop
repeated glucocorticoid administration may inhibit lung respiratory failure that requires endotracheal intubation
growth and development (Massaro and Massaro, 1992, (Morley et al., 2008; Finer et al., 2010a). High failure rate
Tschanz et al., 1995). A recent Cochrane review comparing of NCPAP has led to studies evaluating use of nasal inter-
multiple courses of antenatal steroids with single course mittent positive pressure ventilation (NIPPV) as the initial
showed reduced RDS but no improvement in BPD with mode of respiratory support and its effect on BPD. These
multiple courses (Crowther et al., 2011). studies have provided conflicting results with one study of
43 infants showing reduction in BPD (5% vs. 33%,
SURFACTANT TREATMENT p < 0.05, for infants with birth weight <1500 gm) (Kugel-
Surfactant treatment has improved survival of extremely man et al., 2007) and others showing no change in inci-
preterm infants reducing the severity of RDS, need for dence of BPD (Sai Sunil Kishore et al., 2009; Meneses
aggressive ventilation and prolonged oxygen therapy. How- et al., 2011). A recently published large RCT of 1009
ever, multiple randomized controlled trials (RCTs) have infants with birth weight less than 1000 gm comparing
failed to show a statistically significant effect of surfactant NIPPV with NCPAP failed to show reduction in incidence
on BPD, which may partly be due to increased survival of of death or BPD with nasal ventilation (38.4% vs. 36.7%,
the more immature infants resulting from surfactant adjusted odds ratio, 1.09; CI, 0.83–1.43; p 5 0.56) (Kirpa-
treatment. lani et al., 2013).
With the increasing use of nasal continuous positive
airway pressure (NCPAP) and noninvasive ventilation, MECHANICAL VENTILATION STRATEGIES FOR PREVENTION OF BPD
there have arisen new questions regarding need and tim- A significant number of ELBW infants require mechanical
ing of surfactant use. A recent meta-analysis comparing ventilation which, although required for gas exchange and
prophylactic vs. selective surfactant use concluded that a sustaining life, predisposes them to lung injury and
prophylactic approach was associated with increased risk increased risk of BPD.
of BPD (RR, 1.13; 95% CI, 1.00–1.28) (Rojas-Reyes et al., One of the strategies that has been proposed to limit
2012). ventilator-associated injury is permissive hypercapnia. The
One of the areas that have generated interest in recent evidence for effect of this strategy on BPD has been incon-
years is less invasive methods for administration of surfac- sistent with no clear reduction in BPD (Mariani et al.,
tant with multiple approaches being currently evaluated 1999; Carlo et al., 2002). A small trial has shown a possi-
(Finer et al., 2010b; Dargaville et al., 2011). Until there is ble increase in mortality and neurologic impairment in the
robust evidence on effectiveness and safety of these minimal ventilation group (Thome et al., 2006). Another
BIRTH DEFECTS RESEARCH (PART A) 100:134–144 (2014) 137

strategy that has been studied extensively by large RCTs is risk of RDS but increased risk of BPD (Watterberg et al.,
high frequency ventilation to decrease volutrauma. A 1996). Since then, antenatal and neonatal care for the
recent Cochrane review failed to show that high frequency infants exposed to chorioamnionitis has changed dramati-
oscillatory ventilation consistently decreases the incidence cally, which might explain why some recent studies have
of BPD (Soll, 2013). failed to show association of chorioamnionitis with
Volume targeted ventilation is one of the strategies to increased risk of BPD (Richardson et al., 2006; Andrews
limit volutrauma and achieve automatic weaning of PIP, et al., 2006).
which can potentially limit ventilator-associated lung Colonization of the respiratory tract with Ureaplasma
injury. A recent Cochrane review analyzed 12 RCTs com- urealyticum has been associated with increased incidence
paring pressure-limited ventilation with volume targeted of BPD (Schelonka et al., 2005). However, the effect of
ventilation and reported reduction in combined outcome treatment of this colonization with macrolide antibiotics
of death or BPD with the latter (Wheeler et al., 2010). on the incidence of BPD has been inconsistent. A Cochrane
Other strategies for mechanical ventilation that are review of two small RCTs evaluating the use of erythromy-
being evaluated are proportional assist ventilation cin did not find a decrease in incidence of BPD with this
(Schulze et al., 1999), addition of pressure support to therapy (Mabanta et al., 2003). Since then, some studies
synchronized intermittent mandatory ventilation (Reyes have evaluated the role of newer macrolides, such as azi-
et al., 2006), and neurally adjusted ventilator assist venti- thromycin and clarithromycin, on prevention of BPD.
lation (Beck et al., 2009). These still require well-designed These studies have had variable results with one study by
large RCTs to evaluate their effect on incidence of BPD. Ballard et al. showing decreased incidence of BPD with
azithromycin in Ureaplasma positive infants. However, this
OXYGEN THERAPY AND BPD PREVENTION study reported a very high incidence of BPD in both
Oxygen is the most commonly used therapy in the neona- groups, and clearance of Ureaplasma was unaffected by
tal ICU with large variation in target oxygen saturations. treatment with azithromycin (Ballard et al., 2011; Ozdemir
There is extensive evidence in experimental models docu- et al., 2011). There is a need for large RCTs evaluating the
menting oxygen induced lung injury, but until recently safety and efficacy of this treatment before it can be
actual evidence showing reduction in oxygen exposure recommended.
leads to decreased incidence of BPD had been lacking. In Other postnatal infections have also been associated
the past few years, three large multicentric RCTs have with increased risk of BPD. Coagulase negative staphylo-
been published comparing the effects of two different oxy- coccal sepsis has been associated with significantly higher
gen saturation targets on neonatal mortality and morbid- risk of BPD (64% vs. 24%; RR, 2.6; 95% CI, 1.5–4.6;
ities (Carlo et al., 2010; Stenson et al., 2013; Schmidt p 5 0.001) (Liljedahl et al., 2004). Sawyer et al. (1987)
et al., 2013). Two of these trials, SUPPORT and BOOST-II, evaluated the role of postnatal CMV infection in develop-
have shown significant reduction in incidence of BPD ment of BPD. They showed roentgenographic evidence of
defined as oxygen requirement at 36 weeks, and severe BPD in 24 of 32 postnatally CMV-infected premature
retinopathy of prematurity with use of oxygen saturation infants compared with 12 of 32 of the age-matched nonin-
targets between 85 and 89% against 91 and 95%. More fected control group.
importantly, the SUPPORT trial showed a significant
increase in mortality in the low saturation group (19.9% INHALED NITRIC OXIDE
vs. 16.2%; odds ratio, 1.27; 95% CI, 1.01–1.6), which was Inhaled nitric oxide (iNO) has been used extensively in the
confirmed by a recent meta-analysis of the three trials treatment of pulmonary hypertension due to its selective
(Saugstad and Dagfinn, 2014). Considering these results, it pulmonary vasodilatation and absence of short-term side
seems prudent to avoid targeting extremes by trying to effects. There is considerable evidence in animal models of
keep oxygen saturations between 90 and 95%. BPD that endogenous nitric oxide plays a vital role in alve-
In all of the above-mentioned studies, it was difficult olar and vascular development. Impaired endogenous NO
to maintain oxygen saturation targets within a tightly pre- production contributes in the pathogenesis of BPD (Afshar
scribed range. Therefore, it is likely that tools such as et al., 2003; Balasubramaniam et al., 2003) and adminis-
automated regulation of inspired oxygen using “closed tration of low dose NO attenuates the effects of factors
loop” systems will be used in the future to improve oxy- causing BPD in animal models (Horst et al., 2007; Lin
gen targeting (Claure et al., 2009). et al., 2005).
Clinically, there have been multiple RCTs evaluating the
PREVENTION OF INFECTION effect of iNO therapy on BPD, but results have been incon-
The association between antenatal infection and BPD has sistent. A recent systematic review of 14 RCTs, 7 follow-up
been extensively studied in the past two decades with studies, and 1 observational study by Donohue et al.
variable results. Watterberg et al. were first to report that (2011) failed to show a statistically significant difference
histologic chorioamnionitis is associated with decreased in incidence of BPD at 36 weeks (RR, 0.93; 95% CI,
138 BRONCHOPULMONARY DYSPLASIA

0.86–1.003), or mortality rate (RR, 0.97; 95% CI, 0.82– BPD, before caffeine use becomes a standard practice in
1.15) but did show a small difference in favor of iNO in all preterm infants without more robust evidence.
composite outcome of death or BPD (RR, 0.93; 95% CI,
0.87–0.99). MANAGEMENT OF PDA
There are currently at least four large multicentric PDA is present in up to 70% of infants born at less than
RCTs in progress evaluating the effect of iNO therapy on 28 weeks gestational age. There is significant evidence
BPD. Results of these trials are eagerly awaited, but until from animal models that left to right shunting through the
then, iNO to prevent BPD should only be used under trial PDA has both short- and long-term adverse effects on pul-
protocols. monary vasculature and alveolar architecture, which could
lead to development of BPD (McCurnin et al., 2008). How-
VITAMIN A ever, the evidence linking persistent PDA with BPD in
Vitamin A and its metabolites are important mediators in humans is not very robust, although there is some indirect
lung development and maturation (Mollard et al., 2000). evidence suggesting that hemodynamically significant PDA
They also play an important role in repair of respiratory over a prolonged period of time is associated with
epithelium after lung injury (McGowan, 2002). Preterm increased incidence of BPD (Rojas et al., 1995; Farstad
infants have low levels of vitamin A at birth. Darlow and et al., 2011).
Graham, in a recent systematic review, analyzed data from There is a wide variation in treatment practices for
eight vitamin A supplementation RCTs and showed statisti- PDA closure among different neonatal centers as well as
cally significant reduction in combined outcome of death within centers (Kulkarni et al., 2013). This is due to multi-
or oxygen use at 36 weeks (RR, 0.91; 95% CI, 0.82–1.00; ple reasons, including spontaneous closure of majority of
number needed to treat, 17), but no effect on death, reti- PDAs, no clear markers of hemodynamic significance of
nopathy of prematurity, or sepsis in infants who received PDA, inconsistent success rate of medical treatment in clo-
supplemental vitamin A (Darlow and Graham, 2007). sure of PDA, side effects of drugs used for PDA closure,
Despite vitamin A supplementation being one of the and a paucity of good quality RCTs documenting improved
only few strategies that have been proven to have some outcomes with early PDA closure.
effect on decreasing incidence of BPD, it is not universally There is some evidence that delayed closure of PDA
used (Ambalavanan et al., 2004). Some of the limiting fac- when compared with early closure does not result in
tors are limited availability of vitamin A, need for frequent adverse respiratory outcomes and leads to less need for
intramuscular injections, or perceived small benefit. There PDA treatment (Sosenko et al., 2012). There are increasing
is a need for further research to better understand vitamin data that PDA ligation may be associated with increased
A action at various stages of lung injury and explore alter- morbidities, with several studies showing acute hemody-
native routes of administration. namic side effects and long-term respiratory and neuro-
logic morbidities in infants who had PDA ligation (Kabra
CAFFEINE et al., 2007; Jhaveri et al., 2010; McNamara et al., 2010).
Caffeine is a competitive adenosine receptor antagonist There is a pressing need for a safer and more effective
resulting in stimulation of medullary respiratory center, treatment of PDA as well as large RCTs documenting if in
increased sensitivity to carbon dioxide, and increased dia- fact early closure of PDA improves morbidity and mortal-
phragmatic contractility. Its use in treatment of apnea of ity. Until then, treatment of PDA is very likely to continue
prematurity is well documented. to be individualized based on each patient and the physi-
Schmidt et al. (2006) showed a significant reduction in cian taking care of that infant.
BPD at 36 weeks (36.3% vs. 46.9%, p < 0.01), a secondary
outcome, in a large multicentric RCT in infants treated Management of Established BPD
with caffeine for treatment of apnea of prematurity or to As with the prevention of BPD, management of BPD is
support extubation within the first 10 days of life. Some of also multifactorial. Depending on severity of the BPD, it
the possible mechanisms to explain this beneficial effect could be a very challenging and frustrating task for the
may be the decreased duration of mechanical ventilation physicians taking care of these babies. There are a limited
in caffeine group, or the anti-inflammatory and diuretic number of strategies with variable success rates available
effects of this drug. to avoid further injury and promote development of
Bancalari raised a word of caution against generaliza- alveoli and pulmonary vasculature. Some of the most com-
tion of the results of this trial as these infants were com- mon strategies are discussed below.
paratively more mature and healthier and the effect on
BPD was one of the secondary outcomes for the trial (Ban- NUTRITION AND FLUID MANAGEMENT
calari, 2006). There is an urgent need for more RCTs to Infants at risk of developing BPD often are unable to get
evaluate whether caffeine supplementation since birth adequate nutrition in early postnatal life due to multiple
decreases the duration of ventilation and incidence of reasons, including being acutely ill, glucose intolerance,
BIRTH DEFECTS RESEARCH (PART A) 100:134–144 (2014) 139

attempts at fluid restriction for PDA, delay in initiation of of the methods currently used are CPAP, NIPPV, neurally
feeds, or feeding intolerance. Infants with established BPD adjusted ventilator assist, and high flow nasal cannula oxy-
also have increased demand due to increased work of gen with no clear advantage of one versus another. Fre-
breathing, inflammation, and need for catch up growth quently, choice of the method is decided by the individual
(Denne, 2001). physician’s familiarity with the method or the individual
There is some evidence from animal studies that patient’s response. The role of these different respiratory
under-nutrition, specifically protein and lipid, is associated support methods needs to be further explored in large
with increased risk of lung injury (Deneke et al., 1983; RCTs.
Sosenko et al., 1988) and impaired lung growth (Bhatia
and Parish, 2009). A recent study showed that infants who POSTNATAL CORTICOSTEROIDS
developed BPD had significantly less weight gain, and Postnatal corticosteroids for prevention or management of
received less calories and fat during the first postnatal BPD has been one of the most controversial topics in neo-
month (Akram Khan et al., 2006). natology. Corticosteroids may act by decreasing inflamma-
There is some evidence that high fluid intake during tion, reducing vascular permeability and lung edema,
the first days of life may lead to persistence of PDA (Ste- improving compliance, and decreasing lung fibrosis (Ban-
phens et al., 2008) and increase risk for development of calari, 1998; Rhen and Cidlowski, 2005).
BPD (Oh et al., 2005). This could be secondary to The role of dexamethasone in prevention of BPD was
increased fluid content in pulmonary interstitium leading first described almost 30 years ago, which showed sub-
to decreased compliance. This may increase the need for stantial improvement in lung function with faster weaning
respiratory support leading to lung injury and BPD. A from mechanical ventilation and earlier extubation (Avery
recent Cochrane review comparing varying water intake in et al., 1985). Subsequently multiple trials followed with
preterm infants found that restricted water intake signifi- similar results in improved short-term respiratory out-
cantly reduces the risk of PDA and NEC as well as trend comes, but as data for long-term neurological outcomes
toward decreased risk of BPD (Bell and Acarregui, 2008). were reported, they showed increased risk of cerebral
Therefore, it is prudent to carefully restrict fluid intake palsy in infants who received steroids.
during the first few days of life while ensuring adequate Studies evaluating use of dexamethasone for BPD have
nutrition and avoiding significant dehydration. used either an early or late strategy. A recent meta-
Even though there is no clear evidence from large analysis by Halliday et al. showed that early use of dexa-
RCTs (Lai et al., 2006), aggressive approach toward nutri- methasone within the first 7 days of birth leads to less
tion by early enteral nutrition, ensuring adequate caloric incidence of BPD, but has increased short-term adverse
and protein intake, while avoiding fluid overload to pre- effects such as gastrointestinal hemorrhage, bowel perfora-
vent pulmonary edema has sound physiological rationale. tion, as well as increased risk of cerebral palsy at follow
up (Halliday et al., 2010). In a similar analysis, late dexa-
RESPIRATORY SUPPORT methasone use after the first 7 days was associated with
Infants with severe BPD commonly have heterogeneous decreased risk of BPD, with no definite increase in long-
lung disease with areas of atelectasis and overdistension term adverse neurological outcomes (Halliday et al.,
leading to different time constants for different regions of 2009).
lungs. Due to prolonged mechanical ventilation, these One of the problems with combining different studies
infants commonly have airway injury leading to mucosal for meta-analysis is the inclusion of patient populations
edema, tracheomalacia, or bronchomalacia. with different baseline risk for BPD. Because BPD itself is
During mechanical ventilation, these infants may bene- associated with poor neurological outcomes, a selective
fit from low mechanical rates with longer inspiratory time group of patients with high risk of BPD may benefit from
and adequate expiratory time to ensure emptying of lung low dose dexamethasone. Doyle et al., who performed a
units with long time constants. This decreases the risk of risk-weighted meta-analysis, addressed this issue and
inadvertent positive end-expiratory pressure and overdis- showed that, in studies enrolling infants with high risk of
tension, which may worsen ventilation-perfusion mismatch BPD, use of dexamethasone was associated with decreased
and pulmonary hypertension. These infants also require risk of cerebral palsy or death (Doyle et al., 2005). Thus,
higher tidal volumes to account for increased anatomical in infants with high risk of BPD, such as those remaining
and physiological dead space and achieve adequate alveo- on mechanical ventilation after 2 to 3 weeks of life, the
lar ventilation and CO2 elimination. Although physiologi- beneficial effect of low dose dexamethasone may outweigh
cally sound, there have been no RCTs comparing a low its neurological adverse effects (Watterberg, 2012). In
rate, high tidal volume strategy to a high rate, low tidal keeping with these evidences, recent position statements
volume strategy in infants with established BPD. from both American Academy of Pediatrics and Canadian
Infants with BPD continue to require respiratory sup- Pediatric Society states that clinicians might consider a
port for prolonged periods of time after extubation. Some short course of low dose dexamethasone in individual
140 BRONCHOPULMONARY DYSPLASIA

infants at very high risk of BPD after 1 to 2 weeks of age important long-term outcomes. As a result, a recent meta-
(Jefferies, 2012; Watterberg et al., 2010). analysis concluded that routine or sustained use of sys-
Hydrocortisone is another corticosteroid, which has temic loop diuretics could not be recommended in infants
been studied as a replacement therapy in preterm infants with (or developing) BPD (Stewart and Brion, 2011). Furo-
with decreased inherent adrenal activity. These trials have semide has also been tried in aerosolized form to avoid
used hydrocortisone within the first week of life with systemic side effects, with no significant improvement in
mixed results and some increased risk of gastrointestinal long-term pulmonary outcomes (Brion et al., 2010).
perforation, especially when used in combination with Thiazide diuretics are commonly used along with
indomethacin (Watterberg et al., 2004; Doyle et al., 2010). potassium sparing diuretics in infants with BPD as they
There are two RCTs currently ongoing to evaluate the have decreased risk of electrolyte abnormalities when
effect of early hydrocortisone on BPD (PREMILOC trial, compared with loop diuretics. A double-blinded placebo
Onland et al., 2011). Until the results of these trials are RCT in BPD patients showed decreased oxygen require-
available, its use should be limited to protocol conditions. ment and improved lung function but no improvement in
Inhaled steroids have been used in the management of survival rate, duration of oxygen requirement, or length of
BPD with the possible advantages of direct topical affect hospital stay (Kao et al., 1994). There is a need for large
and avoiding systemic side effects. So far, the RCTs have RCTs before their routine use can be recommended.
failed to show any significant effect on BPD or an advant-
BRONCHODILATORS
age over systemic corticosteroids (Onland et al., 2012).
Some of the possible reasons could be low effective drug Infants with BPD have increased airway resistance due to
delivery due to particle size, or the type of inhaled steroid smooth muscle hypertrophy and hyper reactivity, which
used. Currently, there are two large RCTs under way to makes use of bronchodilators a promising prospect. How-
establish their efficacy and safety (Bassler et al., 2010; ever, there are no RCTs performed to evaluate the effect
QVAR trial). on important long-term pulmonary outcomes (Ng et al.,
2012). Because bronchodilator use has side effects such as
PULMONARY VASODILATORS
tachycardia and hypokalemia and there is no evidence that
Pulmonary hypertension is present in a large proportion long-term outcome is improved, their use in BPD cannot
of infants with severe BPD and is associated with signifi- be recommended at present.
cantly increased morbidity and mortality (Mourani and EMERGING THERAPIES
Abman, 2013). Many agents, including sildenafil and iNO, As our knowledge of mechanisms of alveolar and vascular
are commonly used clinically with no clear evidence of development, injury, and repair is increasing, new possibil-
their effect on pulmonary hypertension secondary to BPD. ities for prevention and treatment of BPD at the molecular
Sildenafil and iNO are currently used for pulmonary level are opening.
vasodilatation, and both drugs have also been shown to Stem cells are one of the most promising therapeutic
improve alveolar growth and angiogenesis in animal mod- approaches in various diseases, including BPD. In animal
els (Lin et al., 2005; Horst et al., 2007; Nyp et al., 2012). models of BPD, use of mesenchymal stem cells has been
Currently, there is limited evidence except case reports for shown to be effective in prevention as well as reversal of
their role in pulmonary hypertension associated with BPD neonatal lung injury (Zhang et al., 2012, Sutsko et al.,
(discussed in more detail in the article on Pulmonary Vas- 2013). There is still a significant amount of work that
cular Disease in BPD in the current issue of this journal). needs to be done to understand the mechanisms of action,
safety, and efficacy of this therapy before it can be used in
DIURETICS
routine clinical practice.
Infants with BPD are predisposed to development of pul- Other promising therapies include modulation of selec-
monary edema secondary to multiple reasons, such as vol- tive inflammatory pathways, targeted gene therapy, and
ume overload secondary to left to right shunt through use of molecules blocking signaling pathways leading to
PDA, capillary leak due to inflammation or ventilator asso- lung injury at the cellular level (Hummler et al., 2013).
ciated injury, or excessive fluid administration. Diuretics
have been used in BPD with the expectation of improving CONCLUSIONS
pulmonary edema, hence improving lung compliance Since Northway’s description almost 45 years ago, the
resulting in improved ventilation and oxygenation. clinical picture of BPD has evolved with New BPD being
There is some evidence that chronic administration of secondary to interplay of multiple factors causing lung
furosemide improves lung compliance and oxygenation in injury and altered lung and pulmonary vascular develop-
infants with BPD or a single dose of furosemide hastens ment. Even though there has been considerable increase
weaning from mechanical ventilation (McCann et al., in knowledge of these factors, BPD continues to be one of
1985). However, there is little evidence of reduction in the major challenges faced by neonatologists. Because BPD
duration of ventilator support, days on oxygen, or other is a multifactorial disease, it is unlikely that any single
BIRTH DEFECTS RESEARCH (PART A) 100:134–144 (2014) 141

therapy will be able to eliminate this problem. Further Bancalari E. 1998 Corticosteroids and neonatal chronic lung dis-
development in prevention and treatment of BPD will ease. Eur J Pediatr 157:S31–S37.
likely need a multi-pronged strategy with novel therapeu-
Bancalari E, Claure N, Sosenko IRS. 2003. Bronchopulmonary
tic agents acting at various stages of the disease process.
dysplasia: changes in pathogenesis, epidemiology and definition.
Semin Neonatol 8:63–71.
Disclosure
E. Bancalari has a patent on an algorithm for automated Bancalari E. 2006. Caffeine for apnea of prematurity. N Engl J
adjustment of inspired oxygen and a licensing agreement Med 354:2179–2181.
with Carefusion.
Bassler D, Halliday DL, Plavka R, et al. 2010. The Neonatal Euro-
pean Study of Inhaled Steroids (NEUROSIS): an eu-funded inter-
References national randomised controlled trial in preterm infants.
Neonatology 97:52–55.
Abman SH, Baker C, Balasubramaniam V. 2008. Growth and
development of the lung circulation: mechanisms and clinical
Beck J, Reilly M, Grasselli G, et al. 2009. Patient – Ventilator
implication. In: Bancalari E, Polin RA editors. The newborn lung.
Interaction during neurally adjusted ventilatory assist in low
St. Louis, MO: Saunders Elsevier. pp. 50–72.
birth weight infants. Pediatr Res 65:663–668.
Abman SH, Wolfe RR, Accuso FJ, et al. 1985. Pulmonary vascular Bell EF, Acarregui MJ. 2008. Restricted versus liberal water
response to oxygen in infants with severe bronchopulmonary intake for preventing morbidity and mortality in preterm infants.
dysplasia. Pediatrics 75:80–84.Afshar S, Gibson LL, Yuhanna IS, Cochrane Database Syst Rev 1:CD000503.
et al. 2003. Pulmonary NO synthase expression is attenuated in
a fetal baboon model of chronic lung disease. Am J Physiol Lung Bhatia J, Parish A. 2009 Nutrition and the lung. Neonatology 95:
Cell Mol Physiol 284:L749–L758. 362–367.

Agrons GA, Courtney SE, Stocker JT, Markowitz RI. 2005. From Brion LP, Primhak RA, Young W. 2010 Aerosolized diuretics for
the archives of the AFIP: lung disease in premature neonates: preterm infants with (or developing) chronic lung disease.
radiologic-pathologic correlation. Radiographics 25:1047–1073. Cochrane Database Syst Rev 3:CD001694.

Akram Khan M, Kuzma-O’Reilly B, Brodky NL, Bhandari V. 2006. Carlo WA, Finer NN, Walsh MC, et al for Support Study group.
Site-specific characteristics of infants developing bronchopulmo- 2010. Target ranges of oxygen saturation in extremely preterm
nary dysplasia. J Perinatol 26:428–435. infants. N Eng J Med 362:1959–1869.

Ambalavanan N, Kennedy K, Tyson J, Carlo WA. 2004. Survey of Carlo WA, Stark AR, Wright LL, et al. 2002. Minimal ventilation
vitamin A supplementation for extremely low birth weight to prevent bronchopulmonary dysplasia in extremely low birth
infants: is clinical practice consistent with evidence base? weight infants. J Pediatr 141:370–374.
J Pediatr 145:304–307.
Claure N, D’Ugard C, Bancalari E. 2009. Automated adjustment of
Andrews WW, Goldenberg RL, Faye-Peterson O, et al. 2006. The inspired oxygen in preterm infants with frequent fluctuations in
Alabama preterm birth study: polymorphonuclear and mononu- oxygenation: a pilot clinical trial. J Pediatr 155:640–645.
clear cell placental infiltrations, other markers of inflammation,
and outcomes in 23- to 32-week preterm newborn infants. Am J ClinicalTrials.gov inhaled extra-fine hydrofluoalkane-beclometha-
Obstet Gynecol 195:803–808. sone (QVAR) in premature infants with bronchopulmonary dys-
plasia (BPD). ClinicalTrials.gov Identifier NCT01373008.
Avery GB, Fletcher AB, Kaplan M, Brudno DS. 1985. Controlled
ClinicalTrials.gov PREMILOC trial to prevent bronchopulmonary
trial of dexamethasone in respirator-dependent infants with
dysplasia in very preterm neonates. Clinical.Trials.gov Identifier
bronchopulmonary dysplasia. Pediatrics 75:106–111.
NCT00623740.

Avery ME, Tooley WH, Keller JB, et al. 1987. Is chronic lung dis- Crowther CA, McKinlay CJD, Middleton P, Harding JE. 2011.
ease in low birth weight infants preventable? A survey of 8 cen- Repeat doses of prenatal corticosteroids for women at risk of
ters. Pediatrics 79:26–30. preterm birth for improving neonatal health outcomes. Cochrane
Database Syst Rev 6:CD003935.
Balasubramaniam V, Tang JR, Maxey A, et al. 2003. Mild hypoxia
impairs alveolarization in the endothelial nitric oxide synthase- Dargaville PA, Aiyappan A, Cornelius A, et al. 2011. Prelimi-
deficient mouse. Am J Physiol Lung Cell Mol Physiol 284:L964– nary evaluation of a new technique of minimally invasive
L971. surfactant therapy. Arch Dis Child Fetal Neonatal Ed 96:
F243–F248.
Ballard HO, Shook LA, Bernard P, et al. 2011. Use of Azithromy-
cin for the prevention of bronchopulmonary dysplasia in preterm Darlow BA, Graham PJ. 2007. Vitamin A supplementation to pre-
infants: a randomized, double-blind, placebo controlled trial. vent mortality and short term morbidity in very low birth weight
Pediatr Pulmonol 46:111–118. infants. Cochrane Database Syst Rev 10:CD000501.
142 BRONCHOPULMONARY DYSPLASIA

Deneke SM, Gershoff SN, Fanberg BL. 1983. Potentiation of oxy- Kabra NS, Schmidt B, Roberts RS, et al. Trial of Indomethacin Pro-
gen toxicity in rats by dietary protein or amino acid deficiency. phylaxis in Preterms Investigators. 2007. Neurosensory impair-
J Appl Physiol 54:147–151. ment after surgical closure of patent ductus arteriosus in
extremely low birth weight infants: results from the Trial of Indo-
Denne SC. 2001 Energy expenditure in infants with pulmonary methacin Prophylaxis in Preterms. J Pediatr 150:229–234 e1.
insufficiency: is there evidence for increased energy needs?
J Nutr 131:935S–937S. Kao LC, Durand DJ, McCrea RC, et al. 1994. Randomized trial of
long-term diuretic therapy for infants with oxygen-dependent
Donohue PK, Gilmore MM, Cristofalo E, et al. 2011. Inhaled nitric bronchopulmonary dysplasia. J Pediatr 124(Pt 1):772–781.
oxide in preterm infants: a systematic review. Pediatrics 127:
414–422. Kirpalani H, Millar D, Lemyre B, et al. NIPPV Study Group. 2013.
A trial comparing noninvasive ventilation strategies in preterm
Doyle LW, Ehrenkranz RA, Halliday HL. 2010. Postnatal hydrocor- infants. N Engl J Med 369:611–620.
tisone for preventing or treating bronchopulmonary dysplasia in
preterm infants: a systematic review. Neonatology 98:111–117. Kugelman A, Feferkorn I, Riskin A, et al. 2007. Nasal intermittent
mandatory ventilation versus nasal continuous positive airway
Doyle LW, Halliday HL, Ehrenkranz RA, et al. 2005. Impact of pressure for respiratory distress syndrome: a randomized, con-
postnatal systemic corticosteroids on mortality and cerebral trolled, prospective study. J Pediatr 150:521–526.
palsy in preterm infants: effect modification by risk for chronic
lung disease. Pediatrics 115:655–661. Kulkarni A, Richards J, Duffy D. 2013. Survey of management of
patent ductus arteriosus in neonatal units across England. Arch
Farstad T, Bratlid D, Medbo S, Markestad T; The Norwegian Dis Child Fetal Neonatal Ed 0:F1–F2.
Extreme Prematurity Study Group. 2011. Bronchopulmonary dys-
plasia—prevalence, severity and predictive factors in a national Lai NM, Rajadurai SV, Tan KH. 2006. Increased energy intake for
cohort of extremely premature infants. Acta Paediatr 100:53–58. preterm infants with (or developing) bronchopulmonary dyspla-
sia/chronic lung disease. Cochrane Database Syst Rev 19:
Finer NN, Carlo WA, Walsh MC, et al; SUPPORT Study group. CD005093.
2010a. Early CPAP versus surfactant in extremely preterm
infants. N Engl J Med 362:1970–1979. Liljedahl M, Bodin L, Schollin J. 2004. Coagulase negative staphy-
lococcal sepsis as a predictor of bronchopulmonary dysplasia.
Finer NN, Merritt TA, Bernstein G, et al. 2010b. An open label, Acta Pediatrica 93:211–215.
pilot study of Aerosurf combined with nCPAP to prevent RDS in
preterm neonates. J Aerosol Med Pulm Drug Deliv 23:303–309. Lin YJ, Markham NE, Balasubramaniam V, et al. 2005. Inhaled
nitric oxide enhances distal lung growth after exposure to hyper-
Fischer HS, Buhrer C. 2013. Avoiding endotracheal ventilation to oxia in neonatal rats. Pediatr Res 58:22–29.
prevent bronchopulmonary dysplasia: a meta-analysis. Pediatrics
132:e1351–e1360. Mabanta CG, Pryhuber GS, Weinberg GA, Phelps DL. 2003. Eryth-
romycin for the prevention of chronic lung disease in intubated
Halliday HL, Ehrenkranz RA, Doyle LW. 2009. Late (>7 days) preterm infants at risk for, or colonized or infected with Urea-
postnatal corticosteroids for chronic lung disease in preterm plasma urealyticum. Cochrane Database Syst Rev 4:CD003744.
infants. Cochrane Database Syst Rev 1:CD001145.
Mariani G, Cifuentes J, Carlo WA. 1999. Randomized trial of permis-
Halliday HL, Ehrenkranz RA, Doyle LW. 2010. Early (<8 days) sive hypercapnia in preterm infants. Pediatrics 104:1082–1088.
postnatal corticosteroids for preventing chronic lung disease in
preterm infants. Cochrane Database Syst Rev 1:CD001146. Massaro GD, Massaro D. 1992. Formation of alveoli in rats: post-
natal effect of prenatal dexamethasone. Am J Physiol 263(Pt 1):
Horst SA, Walther FJ, Poorthuis BJ, et al. 2007. Inhaled nitric 37–41.
oxide attenuates pulmonary inflammation and fibrin deposition
and prolongs survival in neonatal hyperoxic lung injury. Am J McCann EM, Lewis K, Deming DD, et al. 1985 Controlled trial of
Physiol Lung Cell Mol Physiol:L35–L44. furosemide therapy in infants with chronic lung disease. J
Pediatr 106:957–962.
Hummler SC, Rong M, Chen S, et al. 2013. Targeting glycogen
synthase kinase-3b to prevent hyperoxia-induced lung injury in McCurnin D, Seidner S, Chang LY, et al. 2008. Ibuprofen – induced
neonatal rats. Am J Respir Cell Mol Biol 48:578–588. patent ductus arteriosus closure: physiologic, histologic, and bio-
chemical effects on the premature lung. Pediatrics 121:945–956.
Jefferies AJ. 2012. Postnatal corticosteroids to treat or prevent
chronic lung disease in preterm infants. Position Statement – McGowan SE. 2002. Contributions of retinoids to the generation
CPS. Pediatr Child health 17:573. and repair of pulmonary alveolus. Chest 121(Suppl):206S–208S.

Jhaveri N, Moon-Grady A, Clyman RI. 2010. Early surgical ligation McNamara PJ, Stewart L, Shivananda SP, et al. 2010. Patent duc-
versus a conservative approach for management of patent ductus tus arteriosus ligation is associated with impaired left ventricular
arteriosus that fails to close after indomethacin treatment. systolic performance in premature infants weighing less than
J Pediatr 157:381–7.e1. 1000 g. J Thorac Cardiovasc Surg 140:150–157.
BIRTH DEFECTS RESEARCH (PART A) 100:134–144 (2014) 143

Meneses J, Bhandari V, Alves JG, Herrmann D. 2011. Noninvasive Richardson BS, Wakim E, daSilva O, Walton J. 2006. Preterm his-
ventilation for respiratory distress syndrome: a randomized con- tologic chorioamnionitis: impact on cord gas and pH values and
trolled trial. Pediatrics 127:300–307. neonatal outcome. Am J Obstet Gynecol 195:1357–1365.

Mollard R, Ghyselinck NB, Wendling O, et al. 2000. Stage-depend- Roberts D, Dalziel S. 2006. Antenatal corticosteroids for acceler-
ent responses of the developing lung to retinoic acid signaling. ating fetal lung maturation for women at risk of preterm birth.
Int J Dev Biol 44:457–462. Cochrane Database Syst Rev 3:CD004454.

Morley CJ, Davis PG, Doyle LW, et al. COIN Trial. 2008. Nasal Rojas MA, Gonzalez A, Bancalari E, et al. 1995. Changing trends
CPAP or intubation at birth for very preterm infants. N Engl J in the epidemiology and pathogenesis of neonatal chronic lung
Med 358:700–708. disease. J Pediatr 126:605–610.

Mourani PM, Abman SH. 2013. Pulmonary vascular disease in Rojas-Reyes MX, Morley CJ, Soll R. 2012. Prophylactic versus
bronchopulmonary dysplasia: pulmonary hypertension and selective use of surfactant in preventing morbidity and mortality
beyond. Curr Opin Pediatr 25:329–337. in preterm infants. Cochrane Database Syst Rev 3:CD000510.

Mourani PM, Ivy DD, Gao D, Abman SH. 2004. Pulmonary vascu- Sai Sunil Kishore M, Dutta S, Kumar P. 2009. Early nasal inter-
lar effects of inhaled nitric oxide and oxygen tension in broncho- mittent positive pressure ventilation versus continuous positive
pulmonary dysplasia. Am J Respir Care Med 170:1006–1013. airway pressure for respiratory distress syndrome. Acta Paediatr
98:1412–1415.
Ng G, Da Silva O, Ohlsson A. 2012. Bronchodilators for the pre-
vention and treatment of chronic lung disease in preterm infants. Saugstad OD, Dagfinn A. 2014. Optimal oxygenation of extremely
Cochrane Database Syst Rev 6:CD003214. low birth weight infants: a meta-analysis and systematic review
of the oxygen saturation target studies. Neonatology 105:55–63.
NIH Consensus Development Panel 1995: effect of corticoste-
roids for fetal maturation on perinatal outcomes. JAMA 273:413– Sawyer MH, Edwards DK, Spector SA. 1987 Cytomegalovirus
417. infection and bronchopulmonary dysplasia in premature infants.
Am J Dis Child 141:303–305.
Northway WH, Rosan RC, Porter DY. 1967. Pulmonary Disease
following respirator therapy of hyaline membrane disease. Bron- Schelonka RL, Katz B, Waites KB, Benjamin DK. 2005. Critical
chopulmonary Dysplasia. N Engl J Med 276:357–368. appraisal of the role of Ureaplasma in the development of bron-
chopulmonary dysplasia with metaanalytic techniques. Pediatr
Nyp M, Sandritter T, Poppinga N, et al. 2012. Sildenafil citrate, Infect Dis J 24:1033–1039.
bronchopulmonary dysplasia and disordered pulmonary gas
exchange: any benefits? J Perinatol 32:64–69. Schmidt B, Roberts RS, Davis P, et al. Caffeine for Apnea of Pre-
maturity Trial Group. 2006. Caffeine therapy for apnea of prema-
Oh W, Poindexter BB, Perritt R, et al. Neonatal Research Network. turity. N Engl J Med 354:2112–2121.
2005. Association between fluid intake and weight loss during the
first ten days of life and risk of bronchopulmonary dysplasia in Schmidt B, Whyte RK, Asztalos EV, et al. Canadian Oxygen Trial
extremely low birth weight infants. J Pediatr 147:786–790. (COT) group. 2013. Effects of targeting higher vs lower arterial
oxygen saturations on death or disability in extremely preterm
Onland W, Offringa M, Cools F, et al. 2011. Systemic hydrocorti- infants: a randomized clinical trial. JAMA 309:2011–2020.
sone to prevent bronchopulmonary dysplasia in preterm infants
(the SToP-BPD study); a multicenter randomized placebo con- Schulze A, Gerhardt T, Musante G, et al. 1999. Proportional assist
trolled trial. BMC Pediatr 11:102. ventilation in low birth weight infants with acute respiratory dis-
ease: a comparison to assist/control and conventional mechani-
Onland W, Offringa M, van Kaam A. 2012. Late ( 7days) inhala- cal ventilation. J Pediatr 135:339–344.
tion corticosteroids to reduce bronchopulmonary dysplasia in
preterm infants. Cochrane Database Syst Rev 4:CD002311. Soll RF. 2013. Elective high – frequency oscillatory ventilation
versus conventional ventilation for acute pulmonary dysfunction
Ozdemir R, Erdeve O, Dizdar EA, et al. 2011. Clarithromycin in in preterm infants. Neonatology 103:7–9.
preventing bronchopulmonary dysplasia in Ureaplasma urealyti-
cum positive preterm infants. Pediatrics 128:1496–1501. Sosenko IRS, Innis SM, Frank L. 1988. Polyunsaturated fatty acids
and protection of newborn rats from oxygen toxicity. J Pediatr
Reyes ZC, Claure N, Tauscher MK, et al. 2006. Randomized con- 112:630–637.
trolled trial comparing synchronized intermittent mandatory
ventilation and synchronized intermittent mandatory ventilation Sosenko IR, Fajardo MF, Claure N, Bancalari E. 2012 Timing of
plus pressure support in preterm infants. Pediatrics 118: patent ductus arteriosus treatment and respiratory outcome in
1409–1417. premature infants: a double-blind randomized controlled trial.
J Pediatr 160:929–935.
Rhen T, Cidlowski L. 2005. Anti-inflammatory action of glucocor-
ticoids: new mechanisms for old drugs. N Engl J Med 353: Stenson BJ, Tarnow-Mordi WO, Brockelhurst P, et al The BOOST
1711–1723. II United Kingdom, Australia, and New Zealand Collaborative
144 BRONCHOPULMONARY DYSPLASIA

Groups. 2013. Oxygen saturations and outcomes in preterm Walsh MC, Yao Q, Gettner P, National Institute of Child Health
infants. N Eng J Med May 368:2094–2104. and Human Development Neonatal Research Network. 2004.
Impact of a physiologic definition on bronchopulmonary dyspla-
Stephens BE, Gargus RA, Walden RV, et al. 2008. Fluid regimens in sia rates. Pediatrics 114:1305–1311.
the first week of life may increase risk of patent ductus arteriosus
in extremely low birth weight infants. J Perinatol 28:123–128. Watterberg K. 2012. Evidence based therapy – postnatal steroids.
Clin Perinatol 39:47–59.
Stewart A, Brion LP. 2011. Intravenous or enteral loop diuretics
for preterm infants with (or developing) chronic lung disease. Watterberg KL; American Academy of Pediatrics. Committee on
Cochrane Database Syst Rev 9:CD001453. Fetus and Newborn. 2010. Policy Statement – postnatal cortico-
steroids to prevent or treat bronchopulmonary dysplasia. Pedia-
Stoll BJ, Hansen NI, Bell EF. et al. 2010. Neonatal outcomes of trics 126:800–808.
extremely preterm infants from the NICHD Neonatal Research
Network. Pediatrics 126:443–456. Watterberg KL, Demers LM, Scott SM, Murphy S. 1996. Cho-
rioamnionitis and early lung inflammation in infants in whom
Sustko RP, Young KC, Ribeiro A, et al. 2013. Long term reparative bronchopulmonary dysplasia develops. Pediatrics 97:210–
effects of mesenchymal stem cell therapy following neonatal 2115.
hyperoxia-induced lung injury. Pediatr Res 73:46–53.
Watterberg KL, Gerdes JS, Cole CH, et al. 2004. Prophylaxis of
Thome UH, Carroll W, Wu TJ, et al. 2006. Outcome of extremely early adrenal insufficiency to prevent bronchopulmonary dyspla-
low preterm infants randomized at birth to different PaCO2 tar- sia: a multicenter trial. Pediatrics 114:1649–1657.
gets during the first seven days of life. Biol Neonate 90:218–225.
Wheeler K, Klingenberg C, McCallion N, et al. 2010. Volume-tar-
Tschanz SA, Damke BM, Burri PH. 1995. Influence of postnatally geted versus pressure-limited ventilation in the neonate.
administered glucocorticoids on rat lung growth. Biol Neonate Cochrane Database Syst Rev 11:CD003666.
68:229–245.
Zhang X, Wang H, Shi Y, et al. 2012. The role of bone-marrow
Verder H, Robertson B, Greisen G, et al. 1994. Surfactant therapy derived mesenchymal stem cells in the prevention of
and nasal continuous positive airway pressure for newborns with hyperoxia-induced lung injury in newborn mice. Cell Biol Int
respiratory distress syndrome. N Engl J Med 331:1051–1055. 36:589–594.

You might also like