You are on page 1of 7

NICHD/NHLBI/ORD Workshop Summary

Bronchopulmonary Dysplasia
ALAN H. JOBE and EDUARDO BANCALARI
Children’s Hospital Medical Center, Division of Pulmonary Biology, Cincinnati, Ohio; and Department of Pediatrics, University of Miami, Miami, Florida

BACKGROUND AND REASON FOR WORKSHOP ture gas-exchanging airways can already be identified at this point.
Burri proposed that a mantle of mesenchyme over the saccular
Bronchopulmonary Dysplasia (BPD) was first described by
lung may be producing undifferentiated cells that become inte-
Northway and colleagues in 1967 as a lung injury in preterm in-
grated in the differentiating lung. The alveolar stage of lung devel-
fants resulting from oxygen and mechanical ventilation (1). A
opment in the human is from about 36 wk gestation to 18 mo post-
National Heart, Lung and Blood Institute (NHLBI)-sponsored
natally, but the majority of alveolarization occurs within 5 to 6 mo
workshop further defined the disease and suggested research in-
of term birth. The current concept is that primary septation forms
itiatives in 1978 (2). The pathophysiology of BPD was extensively
saccules and that secondary septal crests indicate alveolarization.
reviewed by O’Brodovich and Mellins in 1985 (3). Subsequent
Burri questions whether these are separable processes rather than
research with animal models has shown that the very preterm
a continuum of septation, with some potential for alveolar devel-
lung can be acutely injured by either oxygen or mechanical ven-
opment even after most alveolarization has ceased. The arboriza-
tilation, resulting in interference with or inhibition of lung alve-
tion of the pulmonary microvasculature is intense as the lung
olar and vascular development (4, 5). A change in the pathol-
grows, even after completion of the major phase of alveolarization
ogy of the lungs of infants who have died of BPD has also been
(14). An in-depth understanding of the interdependence of alveo-
found as smaller and more immature infants have come to con-
lization and microvascular development is needed for a better un-
stitute the majority of the infants who develop BPD (6, 7). A re-
derstanding of the pathophysiology of BPD.
cently published book contains multiple reviews of all aspects of
Hussain characterized the “new” BPD on the basis of pathol-
BPD (8). This workshop was organized by the National Insti-
ogy found in infants dying from BPD (15). Before the surfactant
tute of Child Health and Human Development (NICHD) and
treatment era, airway injury, inflammation, and parenchymal fi-
the NHLBI, together with the Office of Rare Diseases (ORD),
brosis were the prominent findings in BPD. More recently, the
to review the definition of BPD and lung injury in very preterm
lungs of infants dying from BPD have shown less fibrosis and
infants, to identify gaps in knowledge about lung development
more uniform inflation. The large and small airways are remark-
and the best indicators of outcome for infants with BPD, and to
ably free of epithelial metaplasia, smooth-muscle hypertrophy,
determine priorities for future research.
and fibrosis. However, there are fewer and larger alveoli, indi-
BPD is now infrequent in infants of more than 1,200 g birth
cating an interference with septation despite an increase in elas-
weight or with gestations exceeding 30 wk (9). Gentler ventila-
tic tissue that is proportionate to the severity and duration of the
tion techniques, antenatal glucocorticoid therapy, and surfac-
respiratory disease before death (16). Some specimens also have
tant treatments have minimized severe lung injury in larger
decreased pulmonary microvascular development. The few bi-
and more mature infants. However, some patients who de-
opsy specimens available from surviving infants show a similar
velop BPD are more enigmatic. These consist of very low
decrease in alveolarization (5). More information is needed
birth weight infants who initially have minimal or no lung dis-
about the progression of the lung injury in survivors of BPD.
ease but who develop increasing oxygen and ventilatory needs
Mechanical ventilation and oxygen can interfere with alve-
over the first several weeks of life (9, 10). Some of these in-
olar and vascular development in preterm baboons and sheep
fants with minimal lung disease that progresses to BPD may
(5, 17). Coalson demonstrated that 7 d of mechanical ventila-
have been exposed to chronic chorioamnionitis (11). The inci-
tion of preterm baboons of 140 d gestation with 100% oxygen
dence of BPD defined as an oxygen need at 36 wk postmen-
severely reduced the numbers of alveoli (4). The same inter-
strual age is about 30% for infants with birth weights  1,000 g
ference with septation of the more preterm 125-d–gestation
(6). Some of these infants have severe lung disease requiring
baboon lung occurs after surfactant treatment and ventilation
ventilation and/or supplemental oxygen for months or years.
but without exposure to large amounts of supplemental oxygen
(5). The large decrease in surface area is associated with a de-
PATHOLOGY OF THE NEW BPD
creased and dysmorphic pulmonary microvasculature. These
Burri reviewed normal lung development and identified a number anatomic changes are associated with persistent increases in
of questions and controversies surrounding it (12, 13). The concept white blood cells and cytokine levels in airway samples. Al-
that only conducting airways are formed during the pseudoglan- though ventilation of preterm baboons from birth with high-
dular period of lung development may not be correct, because fu- frequency oscillatory ventilation resulted in somewhat better gas
exchange, better lung mechanics, and lower proinflammatory
cytokine levels than did conventional ventilation, both ventila-
(Received in original form November 24, 2000 and in revised form February 9, 2001) tion techniques interfered with septation (18). The relationships
National Institute of Child Health and Human Development/National Heart, between the lung injury and inflammatory responses and lung
Lung and Blood Institute/Office of Rare Diseases Workshop on Bronchopulmo-
nary Dysplasia. Held June 1 and 2, 2000, Bethesda, Maryland.
vascular and alveolar hypoplasia need to be better characterized.
Infants with severe BPD have pulmonary hypertension and
Correspondence and requests for reprints should be addressed to Dr. Alan H.
Jobe, Children’s Hospital Medical Center, Division of Pulmonary Biology, 3333 abnormal vascular development. In model systems of pulmo-
Burnet Avenue, Cincinnati, OH 45229. E-mail: jobea@chmcc.org nary hypertension caused by chronic hypoxia in calves, Sten-
Am J Respir Crit Care Med Vol 163. pp 1723–1729, 2001 mark has identified vascular adventitial fibroblasts that prolif-
Internet address: www.atsjournals.org erate and migrate into the media of resistance vessels in the
1724 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 163 2001

presence of hypoxia (19). These hypoxia-sensitive cells express produced by activated white blood cells in the lungs may con-
matrix metalloproteinase-2, and inhibitors of this metallopro- tribute to the progression of lung injury, as suggested by initial
teinase block migration of the cells into the vessels. These fi- evaluations of 1-antitrypsin to decrease the risk of BPD (31).
broblasts of adventitial origin may be sentinel cells that can The recruitment of neutrophils to the lungs soon after birth in-
transdifferentiate and contribute to pulmonary hypertension. dicates that the events surrounding birth have consequences
The molecular signals causing hypoxic activation of adventi- that can increase the risk of BPD. More information is needed
tial fibroblasts are being identified (20). The relationship be- about what modulates neutrophil sequestration in the preterm
tween the decrease in septation and vascular development in lung, and about the downstream events that result in lung injury.
BPD is not understood, nor is the potential for recovery from The theme that inflammation is central to the development
either of these abnormalities. of BPD was further developed by Speer. Multiple proinflam-
Genetics may contribute to BPD at multiple levels. Genetic matory and chemotactic factors are present in the air spaces
polymorphisms in the population may result in increased risks of ventilated preterm infants, and these factors are found in
for developing BPD, as was recently shown for respiratory dis- higher concentrations in the air spaces of infants who subse-
tress syndrome in the Finnish population (21). Through the quently develop BPD (32). Factors such as macrophage in-
use of gene ablation in animal models, factors such as fibro- flammatory protein-1 and interleukin (IL)-8 persist in the air
blast growth factor-10, Bmp-4, and NKx2.1 were shown to be spaces, and counterregulatory cytokines such as IL-10 may be
essential for early lung development (22). This signaling cir- decreased, resulting in unregulated and persistent inflamma-
cuitry of morphoregulators of early lung development is likely tion. Infants exposed to antenatal infection/inflammation or
to be equally important and more complex in location and in fetal colonization with Ureaplasma urealyticum have proin-
developmental timing for alveolar and vascular development. flammatory indicators in their air spaces at delivery (33). In-
Newly developed techniques to regulate genes at precise times flammatory cells are prominent in the interstitium as well as in
during development will provide critical information about the the air spaces, and lung epithelial cells also may synthesize in-
effects of specific genes on lung developmental stages relevant flammatory mediators. Free radical production, enhanced by
to BPD. Minoo emphasized that there will undoubtedly be free iron in the air spaces, can result in production of TGF-
complex interactions between morphoregulators of lung devel- production and fibrosis. The relative importance of the different
opment and the inflammatory mediators present in the injured factors discussed here to the pathophysiology of BPD remains to
lung (23). Factors such as transforming growth factor (TGF)- be defined, and multiple pathways to injury are plausible.
exhibit sexual dimorphism and may predict the development of Sunday has evaluated bombesin-like peptides (BLP) pro-
BPD of sufficient severity as to need home oxygen therapy (24). duced by neuroendocrine cells as mediators in BPD (34). In-
Advances in expression technology and proteinomics should be fants and baboons with BPD have increased numbers of neu-
applied to lung injury in the preterm infant to begin to identify roendocrine cells, mast cells, and eosinophils in their lungs,
those genes that contribute to the injury sequence. and treatment of preterm baboons with an anti-BLP blocking
antibody decreases the numbers of these “immunologic” cells
and results in less lung injury. BLP and other factors may elicit
MECHANISMS OF LUNG INJURY
or promote proinflammatory responses that progress to BPD.
Multiple factors contribute to BPD, and probably act additively Urinary BLP levels correlate with the severity of BPD in pre-
or synergistically to promote injury. The traditional view has term baboons, and infants destined to develop BPD have in-
been that BPD is caused primarily by oxidant- and ventilation- creased urinary levels of BLP. BLP may be a useful early indi-
mediated injury. Oxygen alone can arrest septation of lungs that cator for the identification of infants at risk for BPD.
are in the saccular stage of development (4, 25). Infants with Because decreased numbers of alveoli are so striking in the
BPD who were exposed to higher levels of supplemental oxygen lungs of very preterm infants who die of BPD, understanding
to achieve higher levels of oxygen saturation were found to have the developmental regulation of septation and alveolarization is
more persistent lung disease (26). Mechanical ventilation of pre- a high priority in understanding the pathology of BPD. In ex-
term animals without simultaneous exposure to high levels of perimental models, hyperoxia, hypoxia, or poor nutrition can
supplemental oxygen also results in the pathologic lesion of decrease septation, as can glucocorticoid treatment (35). In
BPD (5, 17). The initiation of mechanical ventilation in surfac- transgenic mice, overexpression of the cytokines tumor necro-
tant-treated preterm animals causes a proinflammatory re- sis factor-, TGF-, IL-6, or IL-11 also can interfere with alve-
sponse, suggesting that any mechanical ventilation of the pre- olarization, suggesting that the proinflammatory environment
term lung may be injurious (27). The avoidance of intubation of the air space of the preterm infant may contribute to the al-
and mechanical ventilation with the use of continuous positive tered septation (7). Massaro noted that all-trans retinoic acid
airway pressure (CPAP) in the delivery room was associated can increase septation in newborn rodents, and promote sep-
with a lower incidence of BPD, although this has not been vali- tation in adult rats with elastase-induced emphysema (36).
dated by randomized trials (28). Therefore, the development of These findings have been extended to the observations that
ventilation and oxygen-exposure strategies that minimize lung mice lacking the retinoic acid receptor (RAR) have early-
injury is a priority for improving outcomes. onset septation, and that treatment of rats with an RAR ago-
Continuing the theme of the possible importance of early nist inhibits septation (37). There are several classes of RAR,
postnatal events as contributors to lung injury in the preterm and their signaling relative to septation will need to be under-
fetus, D. Carlton noted that the preterm lung contains very stood. The inhibition of septation induced by glucocorticoids
few mature macrophages or granulocytes, and that granulo- in neonatal rats also can be reversed with retinoic acid (38).
cytes appear in the lung soon after the initiation of ventilation These studies demonstrate that alveolarization can be regulated
in animal models (29). The appearance of granulocytes in al- once the signaling pathways involved in it are understood.
veolar washes correlates with pulmonary edema and with the
appearance of early indicators of injury, and occurs in parallel
INTERVENTIONS FOR BPD
with a decrease in circulating granulocytes. Preterm infants
who have a decrease in circulating granulocytes at about 1 h of Nutrition plays an important supportive role in the process of
age have an increased risk of developing BPD (30). Proteases normal lung development and maturation. Sosenko noted that
NHLBI Workshop Summary 1725

general undernutrition, and specifically insufficient protein in- berg questioned the rationale for the high doses of dexameth-
take, may increase the vulnerability of the preterm infant to asone frequently used in BPD. Cortisol is a key factor in the
oxidant-induced lung injury. Decreased glutathione levels may response of the lung to injury, and cortisol synthesis is sup-
impair the response to oxidant-induced lung injury, and pro- pressed until relatively late in fetal life. Very preterm infants
tein undernutrition may interfere with lung growth and DNA may lack the capacity to produce enough cortisol to respond
synthesis. A protective effect of polyunsaturated fatty acids to extrauterine stress, and infants who develop BPD have low
against lung injury was reported in experimental animals (39). cortisol levels and a decreased response to adrenocorticotropic
However, several randomized, controlled clinical trials of poly- hormone (50). In a pilot study, low-dose hydrocortisone, when
unsaturated fatty acid administration soon after birth failed to begun shortly after birth and given for 12 d, increased survival
show protection against BPD in preterm infants (40). The lack without BPD at 36 wk postmenstrual age (51). Further studies
of effect may have been related to the presence of toxic lipid are needed to evaluate low dose glucocorticoid therapy.
peroxidation products in the lipid preparations. The commer- Pulmonary vascular resistance (Rpv) is often increased in in-
cial lipid preparations now available have reduced hydroper- fants with BPD, and decreased vascular development may be an
oxide contents, and new trials may be justified. Vitamin A is a important component of the pathophysiology of the disease (52).
nutrient that is important to cell growth and differentiation Abman reported that the administration of antiangiogenesis
and to airway epithelial cell integrity. In a recent randomized, drugs such as fumagillin and thalidomide impaired pulmonary
controlled multicenter clinical trial, vitamin A supplementation vascular and alveolar development in rats, demonstrating that
caused a small but significant reduction in BPD (41). Addi- angiogenesis can be a regulator of alveolar septation (53). Nitric
tional nutrients, such as inositol, sulfur-containing amino acids, oxide (NO) is an important regulator of pulmonary vascular
and selenium may provide the premature infant with addi- tone, and NO synthase is expressed in the vascular endothelium
tional protection against the development of BPD (42). Evalu- and bronchiolar epithelium. NOS increases in the early canalicu-
ations of nutritional interventions are warranted. lar stage of development, and inhaled NO increases pulmonary
Many premature infants are exposed to increased oxygen blood flow and decreases Rpv in fetal lambs. NO also decreases
concentrations, and endogenous antioxidant enzyme activity Rpv in infants with severe RDS and can improve oxygenation
is relatively deficient at birth. Hyperoxic lung injury can be (54). Although possible side effects of NO in the preterm infant
ameliorated both in cell culture and in animals by the adminis- have not been adequately evaluated, NO may decrease the risk
tration of recombinant human superoxide dismutase (rhSOD) of BPD by decreasing intrapulmonary and extrapulmonary
(43). Davis reviewed a placebo-controlled multicenter trial of shunting and by decreasing inflammation in infants with severe
the safety and efficacy of rhSOD in preventing BPD (44). Pla- hyaline membrane disease. Clinical trials are needed to evaluate
cebo or rhSOD was instilled into the trachea after the first NO therapy to prevent and treat BPD.
dose of exogenous surfactant, and the treatment was contin- Mechanical ventilation alone can cause severe lung injury.
ued for up to 28 d or for as long as infants were ventilated. Al- Carlo noted that insufficient positive end-expiratory pressure
though there was no difference in the primary outcome of (PEEP) and large tidal volumes are the main causes of acute
death and/or BPD, the administration of rhSOD was associ- lung damage with mechanical ventilation (55). In adults with
ated with less severe intraventricular hemorrhage and periven- acute respiratory distress syndrome, ventilation at low tidal
tricular leukomalacia. At 6 mo and at 12 mo of corrected age, volume (VT) was associated with improved outcomes and de-
infants treated with rhSOD had a reduced need for respira- creased mortality (56). This ventilation strategy has not been
tory medications as compared with infants receiving placebo evaluated in infants. No detrimental effects were observed in
(45). Antioxidant administration for the prevention and treat- an initial randomized, controlled trial of permissive hypercap-
ment of BPD will need to be further evaluated. nia in preterm infants, although no benefit was demonstrated
Ballard (46) noted that lung development results from the (57). Because arterial carbon droxide tension may not be a
balance between stimulatory and inhibitory influences, and that good substitute for VT, a prospective study, assessing varying
two of the key regulators are glucocorticoids and TGF-. Glu- levels of VT and PEEP, will be required.
cocorticoids accelerate the maturation of parenchymal struc-
ture, increase surfactant production and lung compliance, re-
EPIDEMIOLOGY AND EVALUATIONS OF BPD
duce vascular permeability, and increase lung water clearance.
The net results are improved lung function, better responses to Preterm births continue to be the major challenge in obstetrics
surfactant, and improved survival (46). Glucocorticoids modu- and neonatology, accounting for most of the perinatal mortality
late both the transcriptional and posttranscriptional regulation and long-term neurologic morbidity among newborns (58). An-
of surfactant components, and the effects are reversible after drews noted that approximately half of all preterm births result
treatment (47). Maturation of the surfactant system is also in- from the spontaneous onset of preterm labor, and that about a
duced by analogs of cyclic adenosine monophosphate. In con- third result from preterm premature rupture of the amniotic
trast, TGF- is an inhibitor of lung development (48). TGF- membranes. The remaining 20% of preterm births are medi-
isoforms and receptors are expressed by fibroblasts and epithe- cally indicated for specific maternal or fetal conditions. The rate
lial cells in lung during early gestation. In cultured fetal lung, of clinically asymptomatic colonization of the chorioamnion
TGF- inhibits branching and blocks differentiation of type II and the amniotic fluid increases as the gestational age at deliv-
cells. TGF- increases in tracheal aspirates on the first day of life ery decreases. In one study, positive cultures of chorioamnion
in premature infants who subsequently develop BPD, suggesting were reported in 73% of women with spontaneous preterm
that TGF- is involved in initiation of the injury in BPD. births occuring prior to 30 wk gestation, and in 83% who had
Watterberg discussed the role of postnatal glucocorticoid newborns with birth weights under 1 kg. The colonizing bacte-
therapy for BPD. Many infants at risk for developing BPD are ria initiate an inflammatory cascade and the release of numer-
treated with high doses of glucocorticoids, a therapy associ- ous cytokines, chemokines, prostaglandins, and other bioactive
ated with adverse effects, such as gastrointestinal perforation, substances that can induce cervical ripening, preterm labor, and
cardiac hypertrophy, short- and long-term growth failure, and membrane rupture (59). This inflammatory response may also
the possibility of neurodevelopmental compromise (49). Gluco- cause adverse neonatal outcomes, such as neurologic damage
corticoids impair alveolar septation in animal models. Watten- and cerebral palsy, necrotizing enterocolitis, and BPD.
1726 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 163 2001

Palta presented the results at 8 yr of a six-center follow-up of best name of the disease referred to as BPD in the older liter-
infants with birth weights below 1.5 kg in an effort to identify ature, and more recently as chronic lung disease, was dis-
predictive factors for early and late respiratory and functional cussed. The consensus was to retain the name BPD because it
outcomes (60, 61). Radiographic evidence of BPD at 36 wk was is clearly distinct from the multiple chronic lung diseases of
predictive for rehospitalization at ages 0 to 1 yr, of the need for later life. A new definition, which categorizes the severity of
asthma medications, and of wheezing at age 8 yr, but the predic- BPD, is proposed (Table 1). The definition for infants with
tive value of the radiographic changes diminished as the children gestational ages  32 wk was validated preliminarily with the
became older. On the other hand, patent ductus arteriosus in- NICHD Neonatal Network data base and Palta’s data (61),
creased in predictive importance as the children aged. Oxygen but extensive validation will be needed to determine whether
use at 36 wk was not predictive of outcomes after 2 yr of age. A this definition is superior to previous definitions of BPD. Ra-
family history of asthma was predictive of most respiratory out- diographic findings of BPD are inconsistently interpreted and
comes among children without BPD. Many infants with BPD not routinely available at precise ages, and did not contribute
seem to recover without long-term respiratory problems. to the resolution of the new definition.
Tepper reviewed techniques to evaluate pulmonary out- Research and training initiatives that are critical to better
comes. The main determinant of chronic morbidity in patients understanding of the pathophysiology of BPD, and for explor-
with BPD is the development of obstructive airway disease. This ing therapies for it, are outlined in Table 2. The rapid progress
is demonstrated by a decreased forced expiratory flow (FEF), in understanding the developmental biology of the lung will
increased airway reactivity, and increased RV with a normal provide the essential information about the major signaling
TLC (62–64). In addition, carbon monoxide diffusion capacity pathways for lung-structural development. The stages of par-
may be decreased. All abnormalities may normalize during the ticular interest for BPD will be saccular-to-alveolar develop-
first 3 yr of life, except for that in FEF, which may remain de- ment, with the associated extracellular matrix and vascular de-
creased to adulthood (65). Patients with BPD have increased velopment. Once signaling pathways for alveolar and vascular
airway reactivity to bronchoconstricting agents, as well as persis- development are identified, studies of how oxidants, mechani-
tent respiratory symptoms. Curves of FEF versus volume, gener- cal stress, and inflammation may alter that signaling will be
ated with the squeeze or the forced suction method, are helpful critical to understanding BPD. Expression arrays developed
in detecting airway abnormalities even in patients who are clini- with preterm models of BPD and from human tissue should
cally asymptomatic. High-resolution computed tomographic complement mouse models in which manipulation of specific
scans may provide information on airway size, wall thickness, genes is possible. Such studies will ultimately define what hap-
and hyperinflation caused by gas trapping, and on heterogeneity pens when lung injury is superimposed on a prealveolized and
within the airways and lung parenchyma. Longitudinal studies of minimally vascularized developing lung.
large cohorts of infants with BPD, from the neonatal period
through infancy, childhood, and to adulthood, are needed. The
development of new techniques to evaluate pulmonary structure TABLE 1. DEFINITION OF BRONCHOPULMONARY DYSPLASIA:
and function in different age groups will be crucial to identifying DIAGNOSTIC CRITERIA
the underlying mechanisms for persistent lung-functional abnor- Gestational Age  32 wk  32 wk
malities in survivors of BPD.
With the change in clinical presentation of BPD, the origi- Time point of 36 wk PMA or discharge to  28 d but  56 d
assessment home, whichever comes postnatal age or discharge
nal description now applies only to a minority of patients (1). first to home, whichever
A variety of definitions of BPD have been used in the litera- comes first
ture. The most widely used was that of the 1979 BPD work- Treatment with oxygen  21% for at least 28 d plus
shop, which defined BPD as 28 d of oxygen therapy with ra- Mild BPD Breathing room air at 36 wk Breathing room air by 56 d
diographic changes (2). The oxygen requirement at 36 wk PMA or discharge, whichever postnatal age or discharge,
postmenstrual age was suggested as a better predictor of long- comes first whichever comes first
term respiratory outcomes (66). These definitions have the Moderate BPD Need* for  30% oxygen at Need* for  30% oxygen at
limitation that oxygen administration may vary according to 36 wk PMA or discharge, 56 d postnatal age or dis-
clinical practice among different centers. In an attempt to find whichever comes first charge, whichever comes
a better definition, Ehrenkranz analyzed the NICHD Neona- first
tal Network data base for all infants with birth weights under Severe BPD Need* for  30% oxygen Need* for  30% oxygen
1 kg and gestational ages under 32 wk who were born between and/or positive pressure, and/or positive pressure
January 1995 and December 1997. Oxygen administration for (PPV or NCPAP) at 36 wk (PPV or NCPAP) at 56 d
the first 28 d resulted in the highest sensitivity, specificity, and PMA or discharge, whichever postnatal age or discharge,
comes first whichever comes first
positive and negative predictive values for oxygen administra-
tion at 36 wk. With respect to predicting oxygen use at dis- Definition of abbreviations: BPD  bronchopulmonary dysplasia; NCPAP  nasal continu-
charge, oxygen administration at 36 wk postmenstrual age had ous positive airway pressure; PMA  postmenstrual age; PPV  positive-pressure ventilation.
* A physiologic test confirming that the oxygen requirement at the assessment time
the highest values for sensitivity, specificity, and the percent of point remains to be defined. This assessment may include a pulse oximetry saturation
infants correctly classified. Rehospitalization for respiratory range.
causes, and the use of pulmonary medications after discharge, BPD usually develops in neonates being treated with oxygen and positive pressure
ventilation for respiratory failure, most commonly respiratory distress syndrome. Persis-
were predicted similarly by the 28-d and the 36 wk oxygen re- tence of clinical features of respiratory disease (tachypnea, retractions, rales) are con-
quirements. Given the importance of a consistent definition of sidered common to the broad description of BPD and have not been included in the
BPD, a subcommittee was asked to develop a new definition. diagnostic criteria describing the severity of BPD. Infants treated with oxygen  21%
and/or positive pressure for nonrespiratory disease (e.g., central apnea or diaphrag-
matic paralysis) do not have BPD unless they also develop parenchymal lung disease
GENERAL DISCUSSION, NEW DEFINITION OF BPD, and exhibit clinical features of respiratory distress. A day of treatment with oxygen  21%
means that the infant received oxygen  21% for more than 12 h on that day. Treat-
AND RESEARCH PRIORITIES ment with oxygen  21% and/or positive pressure at 36 wk PMA, or at 56 d postnatal
age or discharge, should not reflect an “acute” event, but should rather reflect the in-
All workshop participants contributed to a discussion of gaps fant’s usual daily therapy for several days preceding and following 36 wk PMA, 56 d
in knowledge and research priorities relating to BPD. The postnatal age, or discharge.
NHLBI Workshop Summary 1727

TABLE 2. PRIORITIES FOR BETTER CHARACTERIZING THE to be identified and subsequently evaluated with intervention
PATHOPHYSIOLOGY OF AND DEVELOPING EFFECTIVE PREVENTIVE trials. Factors that are thought to contribute to BPD (delivery-
AND TREATMENT STRATEGIES FOR BRONCHOPULMONARY DYSPLASIA
room care, oxygen, ventilation, macro- and micronutrient de-
Research Priorities ficiencies, antenatal and postnatal infection) will need to be
Understand the developmental processes of septation, evaluated with clinical trials.
alveolarization, and vascularization The pathologic description of the new BPD is based on tis-
Regulatory genes
Signaling pathways
sue from infants who have died. Much of this tissue is not opti-
Learn how inflammation and injury are expressed by the mally collected and prepared. Better pathologic information
developing lung depends on the careful collection of tissue from infants who
Differential gene expression in uninjured and injured lungs die of BPD and infants with BPD who die from other causes.
Identify modulators of inflammation and injury The collection of tissue prepared according to protocol by a
Use animal models of BPD to test new treatments centralized tissue bank could provide tissue with which inves-
Clinical research priorities
Characterization of BPD
tigators could test for possible pathophysiologic factors as
Establish a resource for tissue from infants dying with BPD they are identified in experimental models. The clinical re-
Develop new clinical tests for lung function in infants and children search and development of tests of pulmonary function for in-
Study genetic contributors to BPD in human populations fants, and the wide application of such tests, require trained
Prevention of BPD personnel. There is a severe lack of physicians with expertise
Develop standards of care as a basis for clinical trials in the evaluation of lung function of infants and children, and
Evaluate delivery room procedures and ventilation techniques
Evaluate nutritional, antioxidant, and antiinflammatory interventions
training programs in this area are needed.
Training priority
Train physicians and physiologists with expertise in evaluating lung function
in infants and children References
Definition of abbreviations: BPD  bronchopulmonary dysplasia. 1. Northway WH Jr, Rosan RC, Porter DY. Pulmonary disease following
respirator therapy of hyaline-membrane disease: bronchopulmonary
dysplasia. N Engl J Med 1967;276:357–368.
2. Report of Workshop on Bronchopulmonary Dysplasia; NIH Publication
No. 80-1660. Wasthington, DC: National Institutes of Health; 1979.
New insights into how the lung develops may yield useful
3. O’Brodovich HM, Mellins RB. Bronchopulmonary dysplasia: unre-
strategies for maturing the preterm lung that are more selective solved neonatal acute lung injury. Am Rev Respir Dis 1985;132:694–709.
and have fewer adverse effects than glucocorticoids. Also, 4. Coalson JJ, Winter V, deLemos RA. Decreased alveolarization in ba-
knowledge of the specific signaling pathways that interfere with boon survivors with bronchopulmonary dysplasia. Am J Respir Crit
normal alveolar and vascular development should yield treat- Care Med 1995;152:640–646.
ment options to better promote lung development despite pre- 5. Coalson JJ, Winter VT, Siler-Khodr T, Yoder BA. Neonatal chronic
lung disease in extremely immature baboons. Am J Respir Crit Care
term birth and the inevitable oxygen and ventilation exposures
Med 1999;160:1333–1346.
necessary for clinical care. Optimizing this information will re- 6. Stevenson DK, Wright LL, Lemons JA, Oh W, Korones SB, Papile LA,
quire research targeted toward the specific abnormalities of Bauer CR, Stoll BJ, Tyson JE, Shankaran S, et al. Very low birth
BPD, including unique characteristics of inflammation and tis- weight outcomes of the National Institute of Child Health and Human
sue responses in the preterm infant. These studies will require Development Neonatal Research Network, January 1993 through De-
animal models that are appropriate to the questions being cember 1994. Am J Obstet Gynecol 1998;179:1632–1639.
7. Jobe AH. The new BPD: an arrest of lung development. Pediatr Res
asked. Ultimately, the biological question is whether normal
1999;46:641–643.
lung development can be altered by injury or by treatment 8. Bland RD, Coalson JJ, editors. Chronic lung disease in early infancy.
strategies without adversely affecting subsequent development. New York: Marcel Dekker; 2000.
A risk is that interventions targeted at one component of the 9. Bancalari E, Gonzalez A. Clinical course and lung function abnormali-
system (e.g., the type II cell) may have a dichotomous effect ties during development of neonatal chronic lung disease. In: Bland
and adversely alter another component such as vascular devel- RD, Coalson JJ editors. Chronic lung disease in early infancy. New
York: Marcel Dekker; 2000. p. 41-64.
opment. Animal models of BPD will be critical for testing treat-
10. Charafeddine L, D’Angio CT, Phelps DL. Atypical chronic lung disease
ment strategies that affect lung-developmental sequences. patterns in neonates. Pediatrics 1999;103:759–765.
Clinical research is needed to better characterize the long- 11. Watterberg KL, Demers LM, Scott SM, Murphy S. Chorioamnionitis
term outcomes of infants with BPD as newly defined. Airway and early lung inflammation in infants in whom bronchopulmonary
disease and reactivity can be measured from early infancy. dysplasia develops. Pediatrics 1996;97:210–215.
However, there are no tests with which to evaluate the abnor- 12. Burri PH. Structural aspects of prenatal and postnatal development and
growth of the lung. In: McDonald JA, editor. Lung growth and devel-
malities resulting from the arrested alveolar and vascular de-
opment. New York: Marcel Dekker; 1997. p. 1–35.
velopment in BPD, or for monitoring how these abnormalities 13. Tschanz SA, Burri PH. Postnatal lung development and its impairment
change with time. Tests of gas diffusion with exercise may pro- by glucocorticoids. Pediatr Pulmonol Suppl 1997;16:247–249.
vide information about the function of the alveolar–capillary 14. Tschanz SA, Damke BM, Burri PH. Influence of postnatally adminis-
barrier. High-resolution imaging techniques may be useful. tered glucocorticoids on rat lung growth. Biol Neonate 1995;68:229–245.
Poor neurodevelopmental outcomes are associated with BPD, 15. Hussain NA, Siddiqui NH, Stocker JR. Pathology of arrested acinar de-
velopment in postsurfactant bronchopulmonary dysplasia. Hum Pathol
and their possible pathophysiologic link to the disease via
1998;29:710–717.
proinflammatory mediators needs to be explored (58). Surro- 16. Thibeault DW, Mabry SM, Ekekezie II, Truog WE. Lung elastic tissue
gate indicators for the anatomic and functional consequences maturation and perturbations during the evolution of chronic lung dis-
of early lung injury resulting in BPD need to be developed to ease. Pediatrics 2000;106:1452–1459.
facilitate treatment-directed clinical research. 17. Albertine KH, Jones GP, Starcher BC, Bohnsack JF, Davis PL, Cho S,
Because BPD results from multiple insults to the preterm Carlton DP, Bland RD. Chronic lung injury in preterm lambs. Am J
Respir Crit Care Med 1999;159:945–958.
lung that probably cause additive or synergistic injurious re-
18. Yoder BA, Siler-Khodr T, Winter VT, Coalson JJ. High-frequency oscilla-
sponses, multiple aspects of care need careful assessment. tory ventilation: effects on lung function, mechanics, and airway cyto-
Given the wide variation in incidence of BPD among neonatal kines in the immature baboon model for neonatal chronic lung disease.
care units (28), different elements of care in current use need Am J Respir Crit Care Med 2000;162:1867–1876.
1728 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 163 2001

19. Frid M, Aldashev A, Nemenoff R, Higashito R, Westcott J, Stenmark K. mentation in premature infants with respiratory distress syndrome. N
Subendothelial cells from normal bovine arteries exhibit autonomous Engl J Med 1992;326:1233–1239.
growth constitutively activated intracellular signaling. Arterioscler 43. Davis JM, Rosenfeld WN, Sanders RJ, Gonenne A. Prophylactic effects
Thromb Vasc Biol 1999;19:2884–2893. of recombinant human superoxide dismutase in neonatal lung injury.
20. Das M, Dempsey EC, Bouchey D, Reyland ME, Stenmark KR. Chronic J Appl Physiol 1993;74:2234–2241.
hypoxia induces exaggerated growth responses in pulmonary artery 44. Davis J, Rosenfeld W, Parad R. Improved pulmonary outcome at one
adventitial fibroblasts: potential contribution of specific protein ki- year corrected age in premature neonates treated with recombinant
nase c isozymes. Am J Respir Cell Mol Biol 2000;22:15–25. human superoxide dismutase. Pediatr Res 2000;47:395A.
21. Ramet M, Haataja R, Marttila R, Floros J, Hallman M. Association be- 45. Davis JM, Richter SE, Biswas S, Rosenfeld WN, Parton L, Gewolb IH,
tween the surfactant protein A (SP-A) gene locus and respiratory-dis- Parad R, Carlo W, Couser RJ, Baumgart S, et al. Long-term follow-up
tress syndrome in the Finnish population. Am J Hum Genet 2000;66: of premature infants treated with prophylactic, intratracheal recombi-
1569–1579. nant human CuZn superoxide dismutase [In Process Citation]. J Peri-
22. Perl AK, Whitsett JA. Molecular mechanisms controlling lung morpho- natol 2000;20:213–216.
genesis. Clin Genet 1999;56:14–27. 46. Ballard PL. Scientific rationale for the use of antenatal glucocorticoids
23. Minoo P, Su G, Drum H, Bringas P, Kimura S. Defects in tracheoesoph- to promote fetal development. Pediatr Rev (Online) 2000;1:E83–E90.
ageal and lung morphogenesis in Nkx2.1(-/-) mouse embryos. Dev 47. Ballard PL, Ning Y, Polk D, Ikegami M, Jobe A. Glucocorticoid regula-
Biol 1999;209:60–71. tion of surfactant components in immature lambs. Am J Physiol 1997;
24. Lecart C, Cayabyab R, Buckley S, Morrison J, Kwong KY, Warburton 273:L1048–L1057.
D, Ramanathan R, Jones CA, Minoo P. Bioactive transforming 48. Beers MF, Solarin KO, Guttentag SH, Rosenbloom J, Kormilli A,
growth factor-beta in the lungs of extremely low birthweight neonates Gonzales LW, Ballard PL. TGF-beta1 inhibits surfactant component
predicts the need for home oxygen supplementation. Biol Neonate expression and epithelial cell maturation in cultured human fetal lung.
2000;77:217–223. Am J Physiol 1998;275:L950–L960.
25. Warner BB, Stuart LA, Papes RA, Wispe JR. Functional and pathologi- 49. Finer NN, Craft A, Vaucher YE, Clark RH, Sola A. Postnatal steroids:
cal effects of prolonged hyperoxia in neonatal mice. Am J Physiol short term gain, long term pain? J Pediatr 2000;137:9–13.
1998;275:L110–L117. 50. Watterberg KL, Scott SM. Evidence of early adrenal insufficiency in ba-
26. Supplemental Therapeutic Oxygen for Prethreshold Retinopathy Of bies who develop bronchopulmonary dysplasia. Pediatrics 1995;95:
Prematurity (STOP-ROP), a randomized, controlled trial: I. primary 120–125.
outcomes. Pediatrics 2000;105:295–310. 51. Watterberg KL, Gerdes JS, Gifford KL, Lin HM. Prophylaxis against
27. Naik A, Kallapur S, Bachurski CJ, Michna J, Jobe AH, Ikegami M. Ef- early adrenal insufficiency to prevent chronic lung disease in prema-
fects of different styles of ventilation on cytokine expression in pre- ture infants. Pediatrics 1999;104:1258–1263.
term lamb lung. Pediatr Res 2000;47:370A. 52. Abman S. Pulmonary hypertension in chronic lung disease in infancy:
28. Van Marter LJ, Allred EN, Pagano M, Sanocka U, Parad R, Moore M, pathogenesis, pathophysiology and treatment. In: Bland R, Coalson J,
Susser M, Paneth N, Leviton A. Do clinical markers of barotrauma editors. Chronic lung disease of early infancy, New York: Marcel Dek-
and oxygen toxicity explain interhospital variation in rates of chronic ker; 1999. p. 619–668.
lung disease? Pediatrics 2000;105:1194–1201. 53. Jakkula M, Le Cras TD, Gebb S, Hirth KP, Tuder RM, Voelkel NF, Ab-
29. Carlton DP, Albertine KH, Cho SC, Lont M, Bland RD. Role of neutro- man SH. Inhibition of angiogenesis decreases alveolarization in the
phils in lung vascular injury and edema after premature birth in lambs. developing rat lung. Am J Physiol 2000;279:L600–L607.
J Appl Physiol 1997;83:1307–1317. 54. Kinsella J, Walsh W, Bose C, Gerstmann D, Labella J, Sardesai S,
30. Ferreira PJ, Bunch TJ, Albertine KH, Carlton DP. Circulating neutro- Walsh-Sukys M, McCaffrey M, Cornfield D, Bhutani V, et al. Ran-
phil concentration and respiratory distress in premature infants. J Pe- domized controlled trial of inhaled nitric oxide in premature neonates
diatr 2000;136:466–472. with severe hypoxemic respiratory failure. Lancet 1999;354:1061–1065.
31. Stiskal JA, Dunn MS, Shennan AT, O’Brien KKE, Kelly EN, Koppel 55. Tremblay LN, Slutsky AS, Dreyfuss D, Saumon G. Ventilator-induced
RI, Cox DW, Ito S, Chappel SL, Rabinovitch M. Alpha1-proteinase lung injury: mechanisms and clinical correlates. In: Marini JJ, Slutsky
inhibitor therapy for the prevention of chronic lung disease of prema- AS, editors. Physiological basis of ventilatory support. New York:
turity: a randomized, controlled trial. Pediatrics 1998;101:89–94. Marcel Dekker; 1998. p. 395–451.
32. Groneck P, Speer CP. Inflammatory mediators and bronchopulmonary 56. ARDSNetwork. Ventilation with lower tidal volumes as compared with
dysplasia. Arch Dis Child 1995;73:F1–F3. traditional tidal volumes for acute lung injury and the acute respira-
33. Hannaford K, Todd DA, Jeffery H, John E, Blyth K, Gilbert GL. Role tory distress syndrome. N Engl J Med 2000;342:1301–1308.
of ureaplasma urealyticum in lung disease of prematurity. Arch Dis 57. Mariani G, Cifuentes J, Carlo WA. Randomized trial of permissive hy-
Child (Fetal Neonatal Ed) 1999;81:F162–F167. percapnia in preterm infants. Pediatrics 1999;104:1082–1088.
34. Sunday ME, Yoder BA, Cuttitta F, Haley KJ, Emanuel RL. Bombesin- 58. Goldenberg RL, Hauth JC, Andrews WW. Intrauterine infection and
like peptide mediates lung injury in a baboon model of bronchopul- preterm delivery. N Engl J Med 2000;342:1500–1507.
monary dysplasia. J Clin Invest 1998;102:584–594. 59. Andrews WW, Hauth JC, Goldenberg RL, Gomez R, Romero R, Cas-
35. Massaro GD, Massaro D. Formation of pulmonary alveoli and gas-ex- sell GH. Amniotic fluid interleukin-6: correlation with upper genital
change surface area: quantitation and regulation. Annu Rev Physiol tract microbial colonization and gestational age in women delivered
1996;58:73–92. after spontaneous labor versus indicated delivery. Am J Obstet Gy-
36. Massaro GD, Massaro D. Retinoic acid treatment abrogates elastase- necol 1995;173:606–612.
induced pulmonary emphysema in rats. Nat Med 1997;3:675–677. 60. Palta M, Sadek M, Barnet JH, Evans M, Weinstein MR, McGuiness G,
37. Massaro GD, Massaro D, Chan WY, Clerch LB, Ghyselinck N, Cham- Peters ME, Gabbert D, Fryback D, Farrell P. Evaluation of criteria
bon P, Chandraratna RA. Retinoic acid receptor-beta: an endogenous for chronic lung disease in surviving very low birth weight infants. J
inhibitor of the perinatal formation of pulmonary alveoli [In Process Pediatr 1998;132:57–63.
Citation]. Physiol Genomics 2000;4:51–57. 61. Palta M, Sadek-Badawi M, Evans M, Weinstein MR, McGuinnes G. Func-
38. Massaro GD, Massaro D. Retinoic acid treatment partially rescues failed tional assessment of a multicenter very low-birth-weight cohort at age 5
septation in rats and in mice. Am J Physiol (Lung Cell Mol Physiol) years: Newborn Lung Project. Arch Pediatr Adolesc Med 2000;154:23–30.
2000;278:L955–L960. 62. Tepper RS, Morgan WJ, Cota K, Taussig LM. Expiratory flow limitation
39. Sosenko IR, Innis SM, Frank L. Intralipid increases lung polyunsatu- in infants with bronchopulmonary dysplasia. J Pediatr 1986;109:1040–
rated fatty acids and protects newborn rats from oxygen toxicity. Pedi- 1046.
atr Res 1991;30:413–417. 63. Gerhardt T, Hehre D, Feller R, Reifenberg L, Bancalari E. Serial deter-
40. Sosenko IR, Rodriguez-Pierce M, Bancalari E. Effect of early initiation mination of pulmonary function in infants with chronic lung disease. J
of intravenous lipid administration on the incidence and severity of Pediatr 1987;110:448–456.
chronic lung disease in premature infants. J Pediatr 1993;123:975–982. 64. Baraldi E, Filippone M, Trevisanuto D, Zanardo V, Zacchello F. Pulmo-
41. Tyson JE, Wright LL, Oh W, Kennedy KA, Mele L, Ehrenkranz RA, nary function until two years of life in infants with bronchopulmonary
Stoll BJ, Lemons JA, Stevenson DK, Bauer CR, et al. Vitamin A sup- dysplasia. Am J Respir Crit Care Med 1997;155:149–155.
plementation for extremely-low-birth-weight infants: National Insti- 65. Northway W, Moss R, Carlisle K, Parker B, Popp R, Pitlick P, Eichler I,
tute of Child Health and Human Development Neonatal Research Lamm R, Brown B Jr. Late pulmonary sequelae of bronchopulmo-
Network. N Engl J Med 1999;340:1962–1968. nary dysplasia. N Engl J Med 1990;323:1793–1799.
42. Hallman M, Bry K, Hoppu K, Lappi M, Pohjavuori M. Inositol supple- 66. Shennan AT, Dunn MS, Ohlsson A, Lennox K, Hoskins EM. Abnormal
NHLBI Workshop Summary 1729

pulmonary outcomes in premature infants: prediction from oxygen re- Children’s Hospital at Buffalo; Richard A. Ehrenkranz, M.D.,
quirement in the neonatal period. Pediatrics 1988;82:527–532. Yale University; Dorothy Gail, Ph.D., NHLBI; Fabio Ghezzi,
M.D., University of Insubria-Varese; Thomas Hazinski, M.D.,
APPENDIX Vanderbilt University; William A. Hodson, M.D., University of
Washington; Aliya Hussain, M.D., Loyola University Medical
PARTICIPANTS OF THE CONFERENCE
Center; Alan H. Jobe, M.D., Ph.D., Children’s Hospital Medi-
Soraya Abassi, M.D., Pennsylvania Hospital; Steven Abman, cal Center, Cincinnati; Sheldon B. Korones, M.D., University
M.D., University of Colorado; Duane Alexander, M.D., Insti- of Tennessee, Memphis; Gloria D. Massaro, M.D., George-
tute of Child Health and Human Development; Bill Andrews, town University; Parviz Minoo, Ph.D., University of Southern
M.D., Ph.D., University of Alabama at Birmingham; Phillip California; Mari Palta, Ph.D., University of Wisconsin-Madison;
Ballard, M.D., Ph.D., Children’s Hospital of Philadelphia; Lu-Ann Papile, M.D., University of New Mexico; April Pilon,
Roberta Ballard, M.D., Children’s Hospital of Philadelphia; Ph.D., Claragen, Inc.; Rangasamy Ramanathan, M.D., Uni-
Eduardo Bancalari, M.D., University of Miami; Beverly versity of Southern California; Jonelle Rowe, M.D., NICHD,
Banks, M.D., The Children’s Hospital of Philadelphia; Charlie NIH; Rashmin C. Savani, M.D., Children’s Hospital of Phila-
R. Bauer, M.D., University of Miami; Mary Anne Berberich, delphia; Gregory M. Sokol, M.D., Indiana University; Ilene
Ph.D., NHLBI; Richard Bland, M.D., University of Utah; Pe- Sosenko, M.D., University of Miami; Christian Speer, M.D.,
ter H. Burri, M.D., University of Bern; Waldemar Carlo, University of Wurzburg; Kurt Stenmark, M.D., University of
M.D., University of Alabama at Birmingham; David P. Carl- Colorado; Cleide Y. Suguihara, M.D., Ph.D., University of
ton, M.D., University of Utah; Reese H. Clark, M.D., Pedia- Miami,; Mary E. Sunday, M.D., Ph.D., Harvard University;
trix Medical Group, Inc.; Jacqueline Coalson, Ph.D., Univer- Robert Tepper, M.D., Indiana University; John S. Torday,
sity of Texas; Jonathan Davis, M.D., State University of NY at Ph.D., Harbor-UCLA Medical Center; Michele Walsh-Sukys,
Stony Brook; Francesco J. DeMayo, Ph.D., Baylor College of M.D., Rainbow Babies & Children’s Hospital; Kristi Watter-
Medicine; Mary Demory, Office of Rare Diseases, NIH; Shah- berg, M.D., Pennsylvania State University; Mary C. Williams,
naz Duara, M.D., University of Miami; Manuel Durand, M.D., Ph.D., Boston University; Linda L. Wright, M.D., NICHD,
University of Southern California; Edmund A. Egan, M.D., NIH; Stephen L. Young, M.D., Duke University

You might also like