You are on page 1of 8

11th Bengt Robertson Memorial Lecture

Neonatology 2019;115:384–391 Published online: April 11, 2019


DOI: 10.1159/000497422

Bronchopulmonary Dysplasia: 50 Years


after the Original Description
Eduardo Bancalari Deepak Jain
Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA

Keywords modulators, and growth factors, but they are still in preclini-
BPD · Premature · Prevention · Management cal stage. The future challenges include finding ways to de-
fine BPD based on the severity of lung pathology, which can
better predict long-term outcomes, development of early
Abstract predictors of lung disease, and finding innovative and evi-
Bronchopulmonary dysplasia (BPD) is one of the few diseas- dence-based preventive and management strategies.
es in neonatal medicine that has continued to evolve since © 2019 S. Karger AG, Basel
its first description about 50 years ago. Over these years, ad-
vancements in neonatal medicine such as antenatal steroids
and exogenous surfactant therapy have significantly re- Introduction
duced neonatal mortality and lowered the limits of viability
for preterm infants. Although the incidence of BPD contin- More than 50 years after its initial description by
ues to be high, especially in extremely low birth weight in- Northway et al. [1], bronchopulmonary dysplasia (BPD)
fants, the clinical picture has evolved into a milder disease continues to be one of the most significant chronic mor-
with low mortality or significant morbidities. This new BPD is bidities affecting very preterm infants. Many advances in
the result of complex interactions between altered alveolar the fields of obstetrics and neonatal medicine over these
and vascular development, injury by ante- and postnatal years have resulted in better survival of very preterm in-
pathogenic factors, and reparative processes in the lung. fants who are at higher risk of developing complications
There has been significant progress in our understanding of associated with extreme prematurity. This has resulted in
risk factors for BPD, but challenges persist in its definition, significant change in the epidemiology, clinical course,
and in finding effective preventive strategies. There are and pathologic picture of BPD posing new challenges for
promising developments with newer preventive interven- clinicians and researchers vested in improving the out-
tions such as mesenchymal stem cells, exosomes, immuno- come of these preterm infants.
130.236.82.7 - 4/12/2019 11:22:16 AM

© 2019 S. Karger AG, Basel Eduardo Bancalari


Linköpings Universitetsbibliotek

Division of Neonatology, Department of Pediatrics


University of Miami Miller School of Medicine
E-Mail karger@karger.com
1611 NW 12th Avenue, Miami, FL 33136 (USA)
www.karger.com/neo
Downloaded by:

E-Mail ebancalari @ med.miami.edu


Progression from “Original” to “New BPD” subsequent days to weeks, many of these infants have
gradual worsening of their respiratory function necessi-
When first described in 1967, the term BPD was used tating escalation in respiratory support [6]. Though the
to define a group of relatively mature preterm infants exact cause of this deterioration is not always clear, often
with severe respiratory failure who survived their initial it coincides with a hemodynamically significant patent
respiratory distress syndrome after receiving aggressive ductus arteriosus (PDA) or infection. Most of these in-
respiratory support with high oxygen and positive pres- fants undergo a slow and steady improvement in their
sure ventilation. Their clinical course was characterized respiratory status and are eventually weaned from respi-
by severe chronic respiratory failure with a radiographic ratory support prior to discharge. A minority of them
picture showing areas of hyperinflation alternating with have a more severe respiratory course with progressive
adjacent increased densities. Many of these infants died worsening of respiratory failure commonly associated
later due to respiratory failure with postmortem lung ex- with pulmonary hypertension and signs of right heart
amination showing widespread emphysema, interstitial failure. The radiographic picture of these infants some-
fibrosis, and airway epithelial changes. times resembles that of the “Original BPD.”
During the ensuing decades, improvements in the
fields of obstetrics and neonatal medicine such as ante-
natal corticosteroids, surfactant therapy, and advance- Evolution of BPD Definition
ments in neonatal care resulted in increased survival of
infants at lower gestational ages. This led to a decline in BPD is one of the few diseases in neonatology where
BPD in the more mature infants and to changes in the multiple definitions have been proposed, reflecting
clinical presentation into a milder form of the respira- changes in the at risk population, clinical presentation,
tory disease in the extremely preterm infants, called and the continued quest to better define the severity of
“New BPD” [2]. These infants are born at the late cana- lung disease and predict long-term outcome. The early
licular or early saccular stages of lung development, and definitions of BPD consisted of oxygen requirement at 28
exposure to various antenatal and postnatal insults can days of age along with chest radiographic changes re-
result in disruption of normal alveolar and vascular de- flecting chronic lung pathology [7, 8]. Over the next de-
velopment. As opposed to the original BPD, the patho- cade as more infants at lower gestational age survived
logic picture in these infants is characterized by alveolar with less severe lung disease and inconsistent radio-
simplification with fewer and dysmorphic capillaries, but graphic picture, Shennan et al. [9] proposed a simpler
less evidence of emphysema, fibrosis, and airway chang- definition of oxygen need at 36 weeks’ post menstrual age
es [3]. as a better predictor of later respiratory outcomes.
In 2001, a workshop sponsored by the National Heart,
Lung, and Blood Institute proposed a definition that for
Clinical Presentation of BPD the first time categorized BPD into mild, moderate, or
severe based on the oxygen or respiratory support re-
The overall incidence of BPD in extremely preterm quirement at 36 weeks’ postmenstrual age. It also in-
infants has not changed significantly over the last decade cluded the need for supplemental oxygen for at least 28
despite improvements in clinical care. This is likely due days to reflect the chronicity of the lung derangement
to the increasing survival of sicker infants at lower gesta- [10]. This characterization of BPD into different severi-
tional age [4]. Infants born at >28 weeks’ gestation, how- ties improves prediction of respiratory and neurological
ever, have lower incidence of BPD and when occurs, it is outcomes [11]. Since then, there have been some modi-
usually milder. The incidence of BPD in extremely low fications with attempts to standardize the need for oxy-
birth weight infants is about 40%, but this varies among gen at 36 weeks by an oxygen reduction test as a more
different centers likely due to variations in clinical prac- physiological definition of BPD [12]. One of the criti-
tices, population characteristics, and definitions used [5]. cisms of the National Heart, Lung, and Blood Institute
The early clinical course of BPD has evolved with a definition of BPD has been the need for determining the
majority of infants now presenting with mild or no initial number of days on supplemental oxygen. This has re-
respiratory disease. These infants are usually managed sulted in different variants of BPD definitions being
with noninvasive respiratory support, and if intubated, used for benchmarking as well as in clinical trials mak-
require low ventilator pressures and oxygen. Over the ing any comparison of outcomes very difficult [13–15].
130.236.82.7 - 4/12/2019 11:22:16 AM

BPD: 50 Years after the Original Neonatology 2019;115:384–391 385


Linköpings Universitetsbibliotek

Description DOI: 10.1159/000497422


Downloaded by:
Bronchopulmonary dysplasia pathogenesis

Extreme prematurity
Antenatal factors
– IUGR, placental dysfunction Postnatal factors
– Chorioamnionitis – Ventilator-induced lung injury
– Bacterial colonization Lung injury/developmental arrest – Supplemental oxygen
– Maternal hypertension/ Airway-vascular damage – Infection/Sepsis
preeclampsia Decreased alveoli and capillaries  – Patent ductus arteriosus
– Maternal smoking/drugs – Nutritional deficiencies 
– Genetic predisposition
Respiratory failure
Pulmonary hypertension

Early respiratory death

Repair/Regeneration

Disease resolution

Chronic respiratory disease: BPD

Fig. 1. Algorithm for pathogenesis of BPD. Modified from Abman et al. [55].

Some of the other shortcomings of current definitions veolar and vascular development, injury by ante- and
of BPD include potentially inaccurate classification of postnatal pathogenic factors, and reparative processes in
infants requiring respiratory support for nonpulmonary the lung (Fig. 1).
causes such as central apnea, or misclassification of in-
fants on newer modes of respiratory support such as Risk Factors
high flow nasal cannula. To overcome some of these Immaturity is the most important risk factor for BPD
shortcomings, a recent Eunice Kennedy National Insti- with its incidence being inversely related to gestational
tute of Child health and Human Development work- age. The role of genetic predisposition for BPD has been
shop proposed some refinements to the 2001 definition suggested by twin pair studies, but the search for specific
of BPD. This proposal attempts to categorize infants on candidate genes has not been very successful so far [17].
nasal cannula oxygen therapy, infants dying due to se- There is increasing epidemiological and experimental ev-
vere lung disease prior to 36 weeks, as well as excluding idence that antenatal factors such as maternal smoking,
infants with transitory need for respiratory support at 36 hypertension, white race, and male gender are important
weeks or nonpulmonary causes of respiratory failure contributors to not only the risk for BPD but also poor
[16]. childhood respiratory health [18]. Intrauterine growth
restriction has been shown to increase the risk for BPD.
The effect of chorioamnionitis on the risk for BPD is not
Changes in the Pathogenesis of BPD conclusive, suggesting that variability in definition of
chorioamnionitis, causative organism and inflammatory
With the changes in epidemiology, clinical picture and response of the fetus may modulate this risk [19].
antenatal and postnatal management of preterm neo- There is clear evidence from animal studies suggesting
nates over the last half century, the pathogenesis of BPD mechanical trauma from high pressure or tidal volume
has also evolved significantly. As opposed to the “original causes lung injury, but the clinical evidence is weak [20,
BPD” resulting mainly from lung injury from high airway 21]. The role of oxygen toxicity in the development of
pressures and oxygen toxicity, the “new BPD” seen today BPD has been shown in both experimental models as well
is the result of complex interactions between altered al- as in clinical trials [22, 23].
130.236.82.7 - 4/12/2019 11:22:16 AM

386 Neonatology 2019;115:384–391 Bancalari/Jain


Linköpings Universitetsbibliotek

DOI: 10.1159/000497422
Downloaded by:
Table 1. Strategies for prevention of BPD
Vitamin A [48] RR 0.88 (95% CI 0.77–0.99)**
Noninvasive respiratory support [38] OR 0.83 (95% CI 0.71–0.96)**, #
Lower oxygen target [23] RR 0.81 (95% CI 0.74–0.9)**, τ
High-frequency oscillatory ventilation [40] RR 0.86 (95% CI 0.78–0.96)**
Postnatal dexamethasone
Early [54] RR 0.79 (95% CI 0.71–0.88)**
Late [42] RR 0.76 (95% CI 0.66–0.88)**
Postnatal hydrocortisone [44] OR 1.48 (95% CI 1.02–2.16)*, ##
Postnatal surfactant + budesonide [45] RR 0.58 (95% CI 0.44–0.77)*, #
Strategies with inconclusive evidence
Antenatal steroids
Exogenous surfactant
Caffeine for apnea or extubation€
Volume-targeted ventilation€
Inhaled nitric oxide
Early PDA closure
Stem cell treatment

* Randomized controlled trial.


** Meta-analysis.
# BPD or death.
##
Survival without BPD.
τ BPD: oxygen at 36 weeks.
€ Lacking data from trials with BPD as primary outcome.

BPD, bronchopulmonary dysplasia.

While postnatal systemic or pulmonary infections in- and epidemiological studies have suggested the associa-
crease the risk for BPD, the role of antenatal colonization tion between a significant PDA and lung damage, but
with Ureaplasma urealyticum is still controversial. Data prospective clinical trials evaluating different PDA clo-
from animal studies show that colonization of fetal lung sure strategies have failed to show a clear effect on inci-
with Ureaplasma increases inflammation and alters al- dence of BPD [27–29].
veolar development. Several epidemiological studies have
suggested an association between Ureaplasma coloniza-
tion of the respiratory tract and an increased risk for BPD Strategies for Prevention of BPD: “Hit – or – Miss”
[24]. Despite this, the treatment of Ureaplasma coloniza-
tion with macrolides has not been consistently successful The continual evolution of BPD as a disease process has
in reducing BPD. presented significant challenges to development of effec-
There is increasing interest in the possible role of lung tive strategies for prevention with increasing realization
microbiome in the risk for BPD with several studies that absolute prevention of BPD may not be achievable
showing that multiple antenatal and postnatal factors, and effort may need to be focused on steps to minimize
such as antibiotic use, nutrition, and sepsis can alter the lung injury and promote alveolar and vascular develop-
normal development of lung microbiome [25, 26]. The ment. Some of these strategies are presented in Table 1.
potential role of lung dysbiosis in the development of Since prematurity is a prerequisite for development of
BPD needs to be further elucidated. BPD, any strategy that may decrease preterm birth is cru-
The role of the PDA in the pathogenesis of BPD has cial in reducing the incidence of BPD. Unfortunately,
been suggested for a long time and is still highly contro- this has not been very successful and in fact, the inci-
versial. Increased pulmonary blood flow due to left-to- dence of preterm birth has been stable or increasing re-
right ductal shunting can decrease lung compliance and cently in many countries [30]. Antenatal corticosteroids
increase airway resistance, which in turn may prolong have been used for accelerating lung maturity since the
need for mechanical ventilation and oxygen supplemen- 1970s, and many randomized controlled trials have
tation and increase the risk for lung injury. Several animal shown reduction in mortality, and other neonatal mor-
130.236.82.7 - 4/12/2019 11:22:16 AM

BPD: 50 Years after the Original Neonatology 2019;115:384–391 387


Linköpings Universitetsbibliotek

Description DOI: 10.1159/000497422


Downloaded by:
bidities except BPD, which is not significantly affected Since oxygen is both lifesaving and potentially injuri-
[31]. This may be explained by increased survival of ex- ous to the lung and other organs, its use needs to be ti-
tremely preterm infants born after antenatal steroids ex- trated very carefully. The ability to continuously and non-
posure [32]. Though antenatal steroids are now standard invasively monitor arterial oxygen saturation (SpO2) has
of care for preterm infants, several questions including been an important advancement in neonatal medicine.
their effect on long-term outcomes in extremely preterm The question of optimal oxygen saturation target range
infants, ideal formulation, and dosing schedule remain for preterm infants has been partially answered recently
to be answered. with 3 large randomized controlled trials comparing the
There is growing interest in optimizing respiratory effect of 2 different targets, low (85–89%) and high (91–
support strategies soon after birth with increasing avail- 95%), on neonatal mortality and morbidities. Meta-anal-
ability of respiratory function and arterial oxygen satura- ysis of these trials showed a small decrease in the inci-
tion monitoring. Currently, there is no clear evidence that dence of BPD in low-target groups but higher risk of NEC
the use of respiratory function monitoring, or targeting and mortality compared to the high-target range [41].
different tidal volumes or oxygen supplementation at Postnatal steroid administration for prevention of
birth affects BPD [33]. Another strategy that showed BPD has been extensively studied and facilitates weaning
promise in reducing lung injury in animal models and from mechanical ventilation and reduces BPD but with
early clinical studies is sustained inflation at birth to im- the potential risk of worse neurologic outcomes in infants
prove functional residual capacity. However, recent trials exposed to high-dose dexamethasone. There is increasing
have not shown beneficial effect on BPD and possibly evidence that short courses with lower doses of dexa-
higher mortality with sustained inflation [34]. methasone after the first postnatal week in infants at high
Exogenous surfactant is one of the landmark therapies risk of BPD may be safe and improve outcomes [42, 43].
in the field of neonatology, which has been shown to de- To avoid potential neurotoxic side effects of systemic
crease severity of RDS and mortality but without any re- dexamethasone, other strategies that have been evaluated
duction in the incidence of BPD in surviving infants [35]. include early systemic hydrocortisone and intra-tracheal
As with antenatal steroids, this is most likely due to in- instillation of budesonide with surfactant [44, 45]. The
creased survival of extremely preterm infants with the use initial results of these interventions have been encourag-
of surfactant [36]. Recently, there has been increasing in- ing, but further studies in different patient populations
terest in novel ways of delivering surfactant avoiding the and effect on long-term outcomes are needed.
need for endotracheal intubation and associated risks of Caffeine is a methylxanthine commonly used for ap-
lung injury. These include less invasive, and nebulized nea of prematurity, and it decreases the incidence of BPD
surfactant administration with some evidence of effec- compared to controls not exposed to the drug [46]. As a
tiveness in decreasing the risk for BPD [37]. result, there has been increasing use of caffeine outside
Respiratory support has evolved significantly since the the established indication for apnea of prematurity or pri-
original description of BPD with an emphasis on provid- or to extubation. This is clearly an area that needs further
ing the least amount of support required for adequate gas investigation [47]. Other therapies investigated for pre-
exchange and oxygenation. One way for achieving this is vention of BPD include vitamin A prophylaxis resulting
avoidance of invasive mechanical ventilation by using al- in a small but significant reduction in BPD compared to
ternatives such as nasal continuous positive airway pres- controls [48] and prophylactic use of nitric oxide that has
sure with some evidence of a small reduction in BPD or not shown a consistent effect on BPD [49].
death [38]. Other noninvasive respiratory support strate- Some innovative strategies for prevention of BPD are
gies such as nasal intermittent positive pressure ventila- mesenchymal stem cells (MSCs) or their secretory prod-
tion or high flow nasal cannula do not consistently reduce ucts. Many studies have shown MSCs to be effective in
the incidence of BPD. Use of invasive mechanical breaths prevention and treatment of lung injury in animal models
synchronized with spontaneous breaths and the accurate of BPD [50]. Early clinical studies have shown promising
monitoring and delivery of appropriate tidal volumes results, but extensive safety and efficacy studies need to
have reduced the duration of exposure to mechanical be performed prior to clinical use. There is increasing ev-
ventilation. There is some evidence that the use of high- idence that the main effects of MSCs may be paracrine
frequency oscillatory ventilation and volume-targeted with secretion of anti-inflammatory and trophic factors
modes of ventilation may also lead to a small reduction in that are contained in nano-sized vesicles like exosomes.
BPD, but the evidence is not strong [39, 40]. Early studies using these MSCs derived exosomes in ani-
130.236.82.7 - 4/12/2019 11:22:16 AM

388 Neonatology 2019;115:384–391 Bancalari/Jain


Linköpings Universitetsbibliotek

DOI: 10.1159/000497422
Downloaded by:
mal models of BPD have shown promising results in re- ical practice or had limited effect on the incidence of BPD
ducing lung injury and preservation of lung structure and [53]. Whether it is due to oversimplification of BPD in
function [51]. Other molecular therapies with promising animal models or limitation of clinical definition of BPD
results in early clinical studies include Clara Cell protein needs to be further evaluated. In addition, there is in-
with anti-inflammatory and immunomodulatory prop- creasing concern that prevention of BPD should be based
erties, and insulin-like growth factor-1, a growth factor on longer-term outcomes rather than outcomes prior to
involved in alveolar and vascular development [52]. discharge from hospital.
Other challenges for optimal management of BPD in-
clude development of evidence-based treatment strate-
Future Challenges and Opportunities in Prevention gies such as respiratory care for chronic respiratory fail-
and Management of BPD ure, evaluation and management of associated pulmo-
nary hypertension, or postdischarge multidisciplinary
One of the major challenges for clinicians and re- care of these infants.
searchers in the field of BPD is the need for a definition,
which can accurately reflect the degree of lung pathology
and differentiate a preterm infant with “normal” alveolar Conclusion
and vascular development. A single, all-encompassing
definition of BPD is difficult to achieve, as the criteria for Fifty years since its first description, the clinical pic-
ideal definition for different stakeholders may be contra- ture, diagnosis, and pathogenesis of BPD have evolved
dictory. While an epidemiologist may prefer to have a significantly making the current disease very different
simple definition that accurately reflects the disease pro- from that described originally. Due to significant ad-
cess, for researchers developing therapeutic strategies, the vancements in obstetrics and neonatal–perinatal medi-
effects on long-term outcomes may be paramount. Some cine, most infants with BPD today have a milder form of
of the newer strategies including biomarkers for evalua- respiratory failure with low mortality and less significant
tion of lung injury, innovative imaging studies, such as morbidity. However, it is clear that there remain signifi-
functional magnetic resonance imaging and pulmonary cant gaps in our understanding of its pathogenesis, opti-
function studies, may provide tools to move toward a mal definition, effective preventive and management
pathophysiology-based definition of BPD. strategies, which will require a concerted effort from re-
Development of an effective preventive strategy for searchers and clinicians alike.
BPD has been a major challenge so far. Multiple interven-
tions acting on different pathways leading to BPD, such
as blocking inflammatory response, preventing oxidative Disclosure Statement
injury, or providing growth factors, have been evaluated
The authors declare that no conflicts of interest related to this
in various animal models of BPD with great success. Most
paper exist.
of these interventions have either not transitioned to clin-

References
  1 Northway WH Jr, Rosan RC, Porter DY. Pul-   4 Stoll BJ, Hansen NI, Bell EF, Walsh MC, Carlo   6 Laughon M, Allred EN, Bose C, O’Shea TM,
monary disease following respirator therapy WA, Shankaran S, et al.; Eunice Kennedy Shriv- Van Marter LJ, Ehrenkranz RA, et al.; ELGAN
of hyaline-membrane disease. Bronchopul- er National Institute of Child Health and Hu- Study Investigators. Patterns of respiratory
monary dysplasia. N Engl J Med. 1967 Feb; man Development Neonatal Research Net- disease during the first 2 postnatal weeks in
276(7):357–68. work. Trends in care practices, morbidity, and extremely premature infants. Pediatrics. 2009
  2 Jobe AJ. The new BPD: an arrest of lung de- mortality of extremely preterm neonates, 1993- Apr;123(4):1124–31.
velopment. Pediatr Res. 1999 Dec;46(6):641– 2012. JAMA. 2015 Sep;314(10):1039–51.   7 Bancalari E, Abdenour GE, Feller R, Gannon
3.   5 Lapcharoensap W, Gage SC, Kan P, Profit J, J. Bronchopulmonary dysplasia: clinical pre-
 3 Bancalari E, Jain D. Pathophysiology of Shaw GM, Gould JB, et al. Hospital variation sentation. J Pediatr. 1979 Nov;95(5 Pt 2):819–
Bronchopulmonary Dysplasia. In: Polin RA, and risk factors for bronchopulmonary dys- 23.
Abman SH, Rowitch DH, Benitz WE, Fox plasia in a population-based cohort. JAMA  8 Tooley WH. Epidemiology of bronchopul-
WW, editors. Fetal and Neonatal Physiolo- Pediatr. 2015 Feb;169(2):e143676. monary dysplasia. J Pediatr. 1979 Nov;95(5 Pt
gy. 5th ed. Vol 2. Elsevier; 2017. p. 1625– 2):851–8.
1631.
130.236.82.7 - 4/12/2019 11:22:16 AM

BPD: 50 Years after the Original Neonatology 2019;115:384–391 389


Linköpings Universitetsbibliotek

Description DOI: 10.1159/000497422


Downloaded by:
  9 Shennan AT, Dunn MS, Ohlsson A, Lennox 22 Buczynski BW, Maduekwe ET, O’Reilly MA. 33 Oei JL, Vento M, Rabi Y, Wright I, Finer N,
K, Hoskins EM. Abnormal pulmonary out- The role of hyperoxia in the pathogenesis of Rich W, et al. Higher or lower oxygen for de-
comes in premature infants: prediction from experimental BPD. Semin Perinatol. 2013 livery room resuscitation of preterm infants
oxygen requirement in the neonatal period. Apr;37(2):69–78. below 28 completed weeks gestation: a meta-
Pediatrics. 1988 Oct;82(4):527–32. 23 Askie LM, Darlow BA, Finer N, Schmidt B, analysis. Arch Dis Child Fetal Neonatal Ed.
10 Jobe AH, Bancalari E. Bronchopulmonary Stenson B, Tarnow-Mordi W, et al.; Neonatal 2017 Jan;102(1):F24–30.
dysplasia. Am J Respir Crit Care Med. 2001 Oxygenation Prospective Meta-analysis (NeO- 34 Fischer HS, Schmolzer GM, Cheung PY,
Jun;163(7):1723–9. ProM) Collaboration. Neonatal Oxygenation Buhrer C. Sustained inflations and avoiding
11 Ehrenkranz RA, Walsh MC, Vohr BR, Jobe Prospective Meta-analysis (NeOProM) Collab- mechanical ventilation to prevent death or
AH, Wright LL, Fanaroff AA, et al.; National oration: association between oxygen saturation bronchopulmonary dysplasia: a meta-analy-
Institutes of Child Health and Human Devel- targeting and death or disability in extremely sis. Eur Resp Rev. 2018;27(150).
opment Neonatal Research Network. Valida- preterm infants in the Neonatal Oxygenation 35 Seger N, Soll R. Animal derived surfactant ex-
tion of the National Institutes of Health con- Prospective Meta-analysis Collaboration. tract for treatment of respiratory distress syn-
sensus definition of bronchopulmonary dys- JAMA. 2018 Jun;319(21):2190–201. drome. Cochrane Database Syst Rev. 2009
plasia. Pediatrics. 2005 Dec;116(6):1353–60. 24 Viscardi RM, Kallapur SG. Role of ureaplas- Apr;(2):CD007836.
12 Walsh MC, Yao Q, Gettner P, Hale E, Collins ma respiratory tract colonization in broncho- 36 Polin RA, Carlo WA; Committee on Fetus
M, Hensman A, et al.; National Institute of pulmonary dysplasia pathogenesis: current and Newborn; American Academy of Pediat-
Child Health and Human Development Neo- concepts and update. Clin Perinatol. 2015 rics. Surfactant replacement therapy for pre-
natal Research Network. Impact of a physio- Dec;42(4):719–38. term and term neonates with respiratory dis-
logic definition on bronchopulmonary dyspla- 25 Lal CV, Travers C, Aghai ZH, Eipers P, Jilling tress. Pediatrics. 2014 Jan;133(1):156–63.
sia rates. Pediatrics. 2004 Nov;114(5):1305–11. T, Halloran B, et al. The airway microbiome 37 Aldana-Aguirre JC, Pinto M, Featherstone
13 Beam KS, Aliaga S, Ahlfeld SK, Cohen- at birth. Sci Rep. 2016 Aug;6(1):31023. RM, Kumar M. Less invasive surfactant ad-
Wolkowiez M, Smith PB, Laughon MM. A 26 Pammi M, Lal CV, Wagner BD, Mourani PM, ministration versus intubation for surfactant
systematic review of randomized controlled Lohmann P, Luna RA, et al. Airway microbi- delivery in preterm infants with respiratory
trials for the prevention of bronchopulmona- ome and development of bronchopulmonary distress syndrome: a systematic review and
ry dysplasia in infants. J Perinatol. 2014 Sep; dysplasia in preterm infants: a systematic re- meta-analysis. Arch Dis Child Fetal Neonatal
34(9):705–10. view. J Pediatr. 2019 Jan;204:126–133.e2. Ed. 2017 Jan;102(1):F17–23.
14 Isayama T, Lee SK, Yang J, Lee D, Daspal S, 27 McCurnin D, Seidner S, Chang LY, Waleh N, 38 Fischer HS, Bührer C. Avoiding endotracheal
Dunn M, et al.; Canadian Neonatal Network Ikegami M, Petershack J, et al. Ibuprofen-in- ventilation to prevent bronchopulmonary
and Canadian Neonatal Follow-Up Network duced patent ductus arteriosus closure: phys- dysplasia: a meta-analysis. Pediatrics. 2013
Investigators. Revisiting the definition of iologic, histologic, and biochemical effects on Nov;132(5):e1351–60.
bronchopulmonary dysplasia: effect of chang- the premature lung. Pediatrics. 2008 May; 39 Klingenberg C, Wheeler KI, McCallion N,
ing panoply of respiratory support for pre- 121(5):945–56. Morley CJ, Davis PG. Volume-targeted versus
term neonates. JAMA Pediatr. 2017 Mar; 28 Liebowitz M, Clyman RI. Prophylactic indo- pressure-limited ventilation in neonates. Co-
171(3):271–9. methacin compared with delayed conserva- chrane Database Syst Rev. 2017 Oct;
15 Bancalari E, Jain D. Bronchopulmonary dys- tive management of the patent ductus arterio- 10:CD003666.
plasia: can we agree on a definition? Am J Per- sus in extremely preterm infants: effects on 40 Cools F, Offringa M, Askie LM. Elective high
inatol. 2018 May;35(6):537–40. neonatal outcomes. J Pediatr. 2017 Aug; 187: frequency oscillatory ventilation versus con-
16 Higgins RD, Jobe AH, Koso-Thomas M, Ban- 119–126.e1. ventional ventilation for acute pulmonary
calari E, Viscardi RM, Hartert TV, et al. Bron- 29 Clyman RI, Liebowitz M, Kaempf J, Erdeve O, dysfunction in preterm infants. Cochrane Da-
chopulmonary dysplasia: executive summary Bulbul A, Hakansson S, et al.; PDA-TOLER- tabase Syst Rev. 2015 Mar;(3):CD000104.
of a workshop. J Pediatr. 2018 Jun;197:300–8. ATE (PDA: To LEave it alone or Respond 41 Askie LM, Darlow BA, Davis PG, Finer N,
17 Shaw GM, O’Brodovich HM. Progress in un- And Treat Early) Trial Investigators: PDA- Stenson B, Vento M, et al. Effects of targeting
derstanding the genetics of bronchopulmo- TOLERATE Trial: An exploratory random- lower versus higher arterial oxygen satura-
nary dysplasia. Semin Perinatol. 2013 Apr; ized controlled trial of treatment of mod- tions on death or disability in preterm infants.
37(2):85–93. erate-to-large patent ductus arteriosus at 1 Cochrane Database Syst Rev. 2017 Apr;
18 Manuck TA, Levy PT, Gyamfi-Bannerman C, week of age. J Pediatr. 2019 Feb;205:41–48.e6. 4:CD011190.
Jobe AH, Blaisdell CJ. Prenatal and perinatal 30 Blencowe H, Cousens S, Chou D, Oestergaard 42 Doyle LW, Ehrenkranz RA, Halliday HL. Late
determinants of lung health and disease in M, Say L, Moller AB, et al.; Born Too Soon (>7 days) postnatal corticosteroids for chron-
early life: A National Heart, Lung, and Blood Preterm Birth Action Group. Born too soon: ic lung disease in preterm infants. Cochrane
Institute Workshop Report. JAMA Pediatr. the global epidemiology of 15 million preterm Database Syst Rev. 2014 May;(5):CD001145.
2016 May;170(5):e154577. births. Reprod Health. 2013;10 Suppl 1:S2. 43 Doyle LW, Halliday HL, Ehrenkranz RA, Da-
19 Jobe AH. Effects of chorioamnionitis on the fe- 31 Roberts D, Brown J, Medley N, Dalziel SR. vis PG, Sinclair JC. Impact of postnatal sys-
tal lung. Clin Perinatol. 2012 Sep;39(3):441–57. Antenatal corticosteroids for accelerating fe- temic corticosteroids on mortality and cere-
20 Björklund LJ, Ingimarsson J, Curstedt T, John tal lung maturation for women at risk of pre- bral palsy in preterm infants: effect modifica-
J, Robertson B, Werner O, et al. Manual ven- term birth. Cochrane Database Syst Rev. 2017 tion by risk for chronic lung disease.
tilation with a few large breaths at birth com- Mar;3:CD004454. Pediatrics. 2005 Mar;115(3):655–61.
promises the therapeutic effect of subsequent 32 Chawla S, Natarajan G, Shankaran S, Pappas 44 Baud O, Maury L, Lebail F, Ramful D, El
surfactant replacement in immature lambs. A, Stoll BJ, Carlo WA, et al.; National Institute Moussawi F, Nicaise C, et al.; PREMILOC
Pediatr Res. 1997 Sep;42(3):348–55. of Child Health and Human Development trial study group. Effect of early low-dose
21 Mokres LM, Parai K, Hilgendorff A, Ertsey R, Neonatal Research Network. Association of hydrocortisone on survival without bron-
Alvira CM, Rabinovitch M, et al. Prolonged neurodevelopmental outcomes and neonatal chopulmonary dysplasia in extremely pre-
mechanical ventilation with air induces apop- morbidities of extremely premature infants term infants (PREMILOC): a double-blind,
tosis and causes failure of alveolar septation with differential exposure to antenatal ste- placebocontrolled, multicentre, rando­ mi­
and angiogenesis in lungs of newborn mice. roids. JAMA Pediatr. 2016 Dec; 170(12): sed trial. Lancet. 2016 Apr; 387(10030):
Am J Physiol Lung Cell Mol Physiol. 2010 Jan; 1164–72. 1827–36.
298(1):L23–35.
130.236.82.7 - 4/12/2019 11:22:16 AM

390 Neonatology 2019;115:384–391 Bancalari/Jain


Linköpings Universitetsbibliotek

DOI: 10.1159/000497422
Downloaded by:
45 Yeh TF, Chen CM, Wu SY, Husan Z, Li TC, 49 Donohue PK, Gilmore MM, Cristofalo E, 52 Ley D, Hallberg B, Hansen-Pupp I, Dani C,
Hsieh WS, et al. Intratracheal administration Wilson RF, Weiner JZ, Lau BD, et al. Inhaled Ramenghi LA, Marlow N, et al. rhIGF-1/
of budesonide/surfactant to prevent broncho- nitric oxide in preterm infants: a systematic rhIGFBP-3 in preterm infants: a phase 2 ran-
pulmonary dysplasia. Am J Respir Crit Care review. Pediatrics. 2011 Feb;127(2):e414–22. domized controlled trial. J Pediatr. 2019 Mar;
Med. 2016 Jan;193(1):86–95. 50 Augustine S, Avey MT, Harrison B, Locke T, 206:56-65.e8.
46 Schmidt B, Roberts RS, Davis P, Doyle LW, Ghannad M, Moher D, et al. Mesenchymal 53 Jobe AH. Animal models, learning lessons to
Barrington KJ, Ohlsson A, et al.; Caffeine for stromal cell therapy in bronchopulmonary prevent and treat neonatal chronic lung dis-
Apnea of Prematurity Trial Group. Caffeine dysplasia: systematic review and meta-analy- ease. Front Med (Lausanne). 2015 Aug;2:49.
therapy for apnea of prematurity. N Engl J sis of preclinical studies. Stem Cells Transl 54 Doyle LW, Ehrenkranz RA, Halliday HL. Ear-
Med. 2006 May;354(20):2112–21. Med. 2017 Dec;6(12):2079–93. ly (<8 days) postnatal corticosteroids for pre-
47 Jobe AH. Caffeine: a lung drug for all very low 51 Lesage F, Thébaud B. Nanotherapies for mi- venting chronic lung disease in preterm in-
birth weight preterm infants? Am J Respir cropreemies: stem cells and the secretome in fants. Cochrane Database Syst Rev. 2014
Crit Care Med. 2017 Nov;196(10):1241–3. bronchopulmonary dysplasia. Semin Perina- May;(5):CD001146.
48 Araki S, Kato S, Namba F, Ota E. Vitamin A tol. 2018 Nov;42(7):453–8. 55 Abman SH, Bancalari E, Jobe A. The Evolu-
to prevent bronchopulmonary dysplasia in tion of Bronchopulmonary Dysplasia after 50
extremely low birth weight infants: a system- Years. Am J Respir Crit Care Med. 2017 Feb;
atic review and meta-analysis. PLoS One. 195(4):421–4.
2018 Nov;13(11):e0207730.

130.236.82.7 - 4/12/2019 11:22:16 AM

BPD: 50 Years after the Original Neonatology 2019;115:384–391 391


Linköpings Universitetsbibliotek

Description DOI: 10.1159/000497422


Downloaded by:

You might also like