You are on page 1of 14

Pitfalls, Problems, and Progress in Bronchopulmonary Dysplasia

Anita Bhandari and Vineet Bhandari


Pediatrics 2009;123;1562-1573
DOI: 10.1542/peds.2008-1962

The online version of this article, along with updated information and services, is
located on the World Wide Web at:
http://www.pediatrics.org/cgi/content/full/123/6/1562

PEDIATRICS is the official journal of the American Academy of Pediatrics. A monthly


publication, it has been published continuously since 1948. PEDIATRICS is owned, published,
and trademarked by the American Academy of Pediatrics, 141 Northwest Point Boulevard, Elk
Grove Village, Illinois, 60007. Copyright © 2009 by the American Academy of Pediatrics. All
rights reserved. Print ISSN: 0031-4005. Online ISSN: 1098-4275.

Downloaded from www.pediatrics.org by on September 2, 2010


STATE-OF-THE-ART REVIEW ARTICLE

Pitfalls, Problems, and Progress in


Bronchopulmonary Dysplasia
Anita Bhandari, MDa, Vineet Bhandari, MD, DMb

aDivision of Pediatric Pulmonology, Connecticut Children’s Medical Center, Hartford, Connecticut; bDivision of Perinatal Medicine, Yale University School of Medicine,
New Haven, Connecticut

The authors have indicated they have no financial relationships relevant to this article to disclose.

ABSTRACT
Bronchopulmonary dysplasia is a chronic lung disease associated with premature
birth and characterized by early lung injury. In this review we discuss some pitfalls,
problems, and progress in this condition over the last decade, focusing mainly on www.pediatrics.org/cgi/doi/10.1542/
peds.2008-1962
the last 5 years, limited to studies in human neonates. Changes in the definition,
pathogenesis, genetic susceptibility, and recent biomarkers associated with broncho- doi:10.1542/peds.2008-1962
pulmonary dysplasia will be discussed. Progress in current management strategies, Key Words
chronic lung disease, prematurity,
along with novel approaches/therapies, will be critically appraised. Finally, recent pulmonary sequelae, noninvasive
data on long-term pulmonary and neurodevelopmental outcomes of infants with ventilation
bronchopulmonary dysplasia will be summarized. Pediatrics 2009;123:1562–1573 Abbreviations
BPD— bronchopulmonary dysplasia
GA— gestational age

T HE OLDER DEFINITION of bronchopulmonary dysplasia (BPD), the commonest


cause of chronic lung disease in infancy,1 was based solely on the duration of
supplemental oxygen requirement, which is intricately linked with the pathogenesis
BW— birth weight
PMA—postmenstrual age
TA—tracheal aspirate
IL—interleukin
and management of this disease.2 To surmount this problem, specific criteria were VEGF—vascular endothelial growth factor
RCT—randomized controlled trial
proposed for the diagnosis and severity of BPD (Table 1).3 CPAP— continuous positive airway
pressure
INCIDENCE FIO2—fraction of inspired oxygen
Earlier studies focused primarily on infants with a gestational age (GA) of ⬍32 weeks iNO—inhaled nitric oxide
SOD—superoxide dismutase
and birth weights (BWs) of ⬍1500 g.4 The bulk of cases today (“new” BPD) is seen CT— computed tomography
in infants ⬍30 weeks’ GA and ⬍1200 g BW. In a recent study, the incidence of BPD Accepted for publication Sep 24, 2008
(using the 36 weeks’ postmenstrual age [PMA] definition) was 42% (BW ⫽ 501–750 g), Address correspondence to Vineet Bhandari,
25% (BW ⫽ 751–1000 g), 11% (BW ⫽ 1001–1250 g), and 5% (BW ⫽ 1251–1500 g).5 MD, DM, Yale University School of Medicine,
Infants with a BW of ⬍1250 g account for 97% of all patients with BPD.6 Division of Perinatal Medicine, Department of
Pediatrics, LCI 401B, PO Box 208064, New
A physiologic definition based on an oxygen-reduction challenge at 36 weeks’ Haven, CT 06520-8064. E-mail: vineet.
PMA decreased the incidence of BPD by 10%.7 Adverse pulmonary and neurode- bhandari@yale.edu
velopmental outcomes are increased in infants with increasing BPD severity.8 PEDIATRICS (ISSN Numbers: Print, 0031-4005;
Online, 1098-4275). Copyright © 2009 by the
American Academy of Pediatrics
PATHOLOGY
With the shift in the population to infants with lower GA and BW, pathologic
findings of new BPD in the lung reveal more uniform inflation and less fibrosis with absence of small and large airway
epithelial metaplasia and smooth muscle hypertrophy, compared with “old” BPD.
One problem has been reaching a consensus about whether the pulmonary microvasculature is decreased9 or
increased.10,11 The controversy stems from findings in animal studies12 and the technical difficulties in obtaining and
conducting careful pathology on human samples.10,11 The pulmonary vasculature is undoubtedly dysregulated in
BPD.1,13 Vascular changes observed include marked angiogenesis proportionate to the growth of the air-exchanging
lung parenchyma,11 abnormal distribution of alveolar capillaries,14 prominent corner vessels with variable capillary
density in adjacent alveoli,15 and vessels that are more distant from the air surface.11,16

PATHOGENESIS
Although an immature lung on which a variety of “environmental” factors (infection, hyperoxia, barotrauma/
volutrauma) act to cause injury has been the cornerstone of the pathogenesis of BPD, an improved understanding
of this process has occurred over the last 5 years. Antenatal/postnatal factors contribute to the release of “proin-
flammatory and antiinflammatory” cytokines. An imbalance in these mediators leads to activation of the cellular
death pathways in the lung, which is followed by healing (resolution of injury to a normal lung architecture) or
repair.17,18 The latter is characterized by impaired alveolarization and dysregulated angiogenesis, which lead to fewer,
larger simplified alveoli and a dysmorphic pulmonary vasculature (Fig 1): the pathologic hallmarks of BPD.1

1562 BHANDARI, BHANDARI


Downloaded from www.pediatrics.org by on September 2, 2010
TABLE 1 Diagnostic Criteria for BPD
Age at Birth, Mild Moderate Severe
wk GA
⬍32 Supplemental O2 (for 28 d) and RA at Supplemental O2 (for 28 d) and ⬍0.3 FIO2 at Supplemental O2 (for 28 d) and ⱖ0.3 FIO2 or
36 wk corrected GA or at discharge 36 wk corrected GA or at discharge positive pressure support at 36 wk corrected GA
or at discharge
ⱖ32 RA by postnatal day 56 or at discharge ⬍0.3 FIO2 by postnatal day 56 or at discharge ⱖ0.3 FIO2 with or without positive pressure support
by postnatal day 56 or at discharge
RA indicates room air.

GENETICS ation in the smaller studies not yet replicated. This


Genetic and environmental factors that make an indi- makes sense if BPD is at least partly polygenic, because
vidual more or less likely to develop a disease constitute no single gene will have a large enough effect to be
liability or susceptibility. Heritability constitutes the ge- evident in small studies. Candidate-gene approaches
netic component of the disease. The large sample size of have been limited to only a few or single candidate genes
a multisite study that corrected for major covariates19 and, therefore, miss important molecular determinants
allowed the results to be generalizable to the preterm of disease that are not intuitively targeted for BPD. Fur-
population, because no additional risk for BPD could be thermore, publication of irreproducible results due to
attributed to twinning,20 independent of prematurity. small sample sizes will diminish the credibility and en-
Identification of a genetic component (contributing thusiasm for robust genetic studies in BPD.
⬎50% susceptibility) of BPD has transformed a disease
traditionally considered to be exclusively secondary to
“environmental” factors to one resulting from “gene- BIOMARKERS
environment” interactions.19,21 The establishment of a A multitude of biomarkers detected in tracheal aspirates
familial tendency and heritability in twin studies should (TAs), blood, and urine have been proposed for early
boost the efforts to identify reproducible allelic associa- identification for infants who subsequently develop
tion to the genetic susceptibility to BPD.21–24 BPD. TA specimens may be suitable substitutes for bron-
There are multiple pitfalls of the published studies. choalveolar lavage samples in preterm infants,25 but con-
Approximately half of the initial genetic-association troversy exists whether measurements of the markers in
studies, even those with strong effects (odds ratio: ⬎2.0) the TAs are valid without “normalization.” Although
or with low P values (⬍.001), have not been replicated. some have advocated use of total protein,26,27 secretory
The reasons include population admixture (unmatched component of immunoglobulin A,28–32 and serum urea,33,34
cases and controls), phenotypic heterogeneity (inclusion others have espoused that no normalization is required.35–38
of both genetic and nongenetic cases), and, commonly, TAs have the additional disadvantage that data from
underpowered studies. Follow-up studies in larger sam- nonintubated (healthier) infants cannot be obtained
ples will probably diminish the initial reports of associ- for comparison. TAs and blood analytes, usually

Postnatal factors: ventilator trauma, oxygen


Antenatal factors: chorioamnionitis
toxicity, pulmonary edema, sepsis

Cytokines Genetic susceptibility

Proinflammatory Antiinflammatory
(eg, IL-1) (eg, IL-10)

Immature lung

FIGURE 1
Pathogenesis of new BPD. Cell death

Normal lung Resolution Repair

Impaired alveolarization Dysregulated angiogenesis

Bronchopulmonary dysplasia

PEDIATRICS Volume 123, Number 6, June 2009 1563


Downloaded from www.pediatrics.org by on September 2, 2010
TABLE 2 Use of TA Analytes as Biomarkers for BPD TABLE 3 Use of Blood/Serum Analytes as Biomarkers for BPD
Analyte Total No. Association With Reference Analyte Total No. Association With Reference
of Infants BPD No. of Infants BPD No.
Angiopoietin 2 14; 60a Increased 27, 146 C-IV 39 Decreased 161
Cathepsin-K 13 Decreased 147 CD9 44 Decreased 162
CCSP 45 Present 148 ECP 44 Increased 162
ET-1 29 No association 35 8-isoprostanes 83 Increased 163
FGF-2 29 Increased 35 KL-6 42 Increased 164
HGF 22 Decreased 30 PICP 36 Decreased 111
IL-4 69 Absent/variable 149 PlGF 61 Increased 165
IL-6 75 Increased 150 Soluble E-selectin 30; 44a Increased 156, 166
IL-6/VEGF 17 Decreased 26 Soluble L-selectin 44 Decreased 166
IL-8 20; 31a Increased 151, 152 C-IV indicates type IV collagen; CD9, cellular surface antigen; ECP, eosinophilic cationic
IL-10 69 Absent/variable 149 protein; 8-isoprostanes, 8-epi-prostaglandin F2␣; KL-6, a circulating high molecular weight
IL-12 69 Absent/variable 149 mucinous glycoprotein that is the extracellular soluble region of MUC-1 mucin possessing
IL-1RA 35 Increased 153 an undefined sialylated carbohydrate chain that is recognized by the KL-6 antibody; PICP,
KGF 32; 91a Decreased 30, 154 C-terminal fragment of type I procollagen; PlGF, placental growth factor.
a Per study cited.
MCP-1 35; 56a Increased 31, 151
MCP-2 56 Increased 31
MCP-3 56 Increased 31
MIF 26 Decreased 155 has been replicated but showed variable results (Table
MMP-2 49 Decreased 32 2). Most of the comments made above are also true of
MMP-8 16 Increased 29 the blood and urinary markers. At the moment, there is
MMP-9/TIMP-1 49 Increased 32 no “magic” biomarker for the early identification of in-
Neutrophil counts 30 Increased 156
fants who subsequently develop BPD. Proteomics is a
NF-␬B 32 Increased 28
promising new approach in this field.
PAI-1 37 Increased 157
PTHrP 40 Decreased 158 Another pitfall is that the majority have only shown
TGF-␤1 69 Increased 149 an association with BPD. To prove a causal relationship,
TIMP-1 49 Increased 32 they need to be studied in developmentally appropriate
TIMP-2 16; 49a Decreased 29, 32 animal models and human lungs with BPD. Some
Trypsin 2 32 Increased 159 progress in this direction has been made with elastin,42,43
VEGF 29; 16a No association; phasic 35, 160 cathepsin-S,44,45 hepatocyte growth factor,46 keratinocyte
pattern growth factor,47 IL-1␤,48 IL-6,49 monocyte chemoattrac-
CCSP indicates Clara cells secretory protein; ET, endothelin; FGF, fibroblast growth factor; tant protein 1,50 transforming growth factor ␤1,51,52
HGF, hepatocyte growth factor; KGF, keratinocyte growth factor; MCP, monocyte che- VEGF,53–58 bombesin-like peptide,59 and angiopoietin 2,27
moattractant protein; MIF, macrophage migration inhibition factor; MMP, matrix metallo-
proteinase; NF-␬B, nuclear factor-␬B; PAI, plasminogen activator inhibitor; PTHrP, para-
but additional work is needed.
thyroid hormone-related protein; RA, receptor antagonist; TGF, transforming growth
factor; TIMP, tissue inhibitor of metalloproteinases. MANAGEMENT
a Per study cited.
The classification of robustness of the evidence and the
recommendation for use in the clinical setting guidelines
are shown in Table 4. The current status of the thera-
obtained in the first week of life, are summarized in peutic approaches in the 3 phases (early, evolving, and
Tables 2 and 3. established) of BPD are summarized in Table 5. In older
Urinary levels of F2-isoprostanes are not correlated (n ⫽ survivors of BPD, inhaled steroids and ␤-agonists are
24),39 but elevated urinary bombesin-like peptide levels are used for symptomatic relief irrespective of the severity of
associated with a 10-fold increased risk of developing BPD BPD during infancy. No randomized, controlled trials
(n ⫽ 50).40 (RCTs) exist to either support or negate this approach.
Infants with BPD (n ⫽ 15) have shown a significantly Prevention of BPD would be ideal. Among antenatal
lower blood vessel area and greater vascular network factors, prevention of premature birth is the single most
complexity in the lower gingival and vestibular oral effective preventive measure. Understanding the biology
mucosa than controls.41 of preterm labor60 and targeting therapies to inhibit it
The pitfall of these studies is that a majority com- safely would be important goals. The use of progesterone
prised small numbers of infants. Most attempts to in prevention of premature labor has shown promise,
identify biomarkers have been to access the pulmo- although the prolongation of gestation with this ap-
nary compartment. TAs collected at variable time points, proach has yet to improve infant outcomes.61 Tackling
measurements usually of single markers, with or with- the social and educational issues pertaining to preterm
out normalization and variable definitions of BPD are birth62 could potentially have a significant impact.
additional issues. Few have been replicated in different Antenatal steroids still remain the single most effec-
cohorts. Those with consistent results include angiopoi- tive intervention for lung maturation. The impact of
etin 2, interleukin 6 (IL-6), IL-8, monocyte chemoattrac- antenatal steroids on BPD has been controversial, with
tant protein 1, and tissue inhibitor of metalloproteinase studies reporting no or some benefit. This is not surpris-
2 (Table 2). Vascular endothelial growth factor (VEGF) ing given the complex nature of the disease (Fig 1), and

1564 BHANDARI, BHANDARI


Downloaded from www.pediatrics.org by on September 2, 2010
TABLE 4 Levels of Evidence and Recommendations for Clinical nasal continuous positive airway pressure (CPAP) com-
Use Based on the Guidelines Developed by the US pared with later selective surfactant replacement and
Preventive Services Task Force continued mechanical ventilation with extubation from
low ventilator support is associated with less need for
Levels of evidence
mechanical ventilation, lower incidence of BPD, and
I Evidence obtained from at least 1 properly
designed randomized, controlled trial fewer air-leak syndromes.76 Use of a cutoff value of
II-1 Evidence obtained from well-designed fraction of inspired oxygen (FIO2) of 0.45 to decide on
controlled trials without randomization transient intubation and surfactant replacement seems
II-2 Evidence obtained from well-designed to decrease the incidence of air leaks and BPD.76 The
cohort or case-control analytic studies, Cochrane review, however, did not include the results of
preferably from ⬎1 center or research the CPAP or intubation at birth (COIN) trial, which
group reported that in infants born at 25 to 28 weeks’ gesta-
II-3 Evidence obtained from multiple time series tion, early nasal CPAP did not significantly reduce the
with or without the intervention; dramatic
rate of death or BPD, compared with intubation, and the
results in uncontrolled trials might also be
CPAP group had a higher incidence of pneumothora-
regarded as this type of evidence
III Opinions of respected authorities based on ces.77 The Eunice Kennedy Shriver National Institute of
clinical experience, descriptive studies, or Child Health and Human Development–sponsored Neo-
reports of expert committees natal Research Network SUPPORT study, which is ex-
Levels of recommendations amining the same issue in addition to optimal oxygen-
for clinical use saturation ranges, is ongoing. Another approach is to use
A Good scientific evidence suggests that the nasal intermittent positive pressure ventilation, both in
benefits substantially outweigh the the synchronized78 and nonsynchronized79 modes that
potential risks have been found to decrease BPD in RCTs. The ongoing
B At least fair scientific evidence suggests that
nasal ventilation in preterms (NTP) trial is a large mul-
the benefits outweigh the potential risks
ticenter international RCT aimed at confirming the find-
C At least fair scientific evidence suggests that
there are benefits provided, but the ings of the initial studies. An attractive alternative to in-
balance between benefits and risks are tubation and delivery of intratracheal surfactant would
too close for making general be aerosol delivery. A new synthetic surfactant (lucinac-
recommendations tant) that contains the novel peptide sinapultide, a sur-
D At least fair scientific evidence suggests that factant-associated protein B mimic,80 seems to be similar
the risks outweigh potential benefits to animal-derived surfactant in 2 multicenter random-
I Scientific evidence is lacking, of poor quality, ized clinical trials when used intratracheally.81,82 A meta-
or conflicting, such that the risk/benefit analysis of these studies revealed no statistically different
balance cannot be assessed differences in the outcomes of death and BPD.83 To date,
however, no effective aerosolized surfactant preparation
is available for routine clinical use. Additional progress
other factors may mask their benefit.63 Weekly courses in this area is awaited.84,85 Inhaled nitric oxide (iNO)
of antenatal steroids are not recommended.64 Beta- may86,87 or may not88,89 be beneficial; hence, more stud-
methasone is preferred over dexamethasone.65,66 ies are needed to better identify potential benefits to
Considerable research has gone into understanding the target population likely to develop BPD.90 The use
the role of antenatal and postnatal inflammation in the of iNO does seem to be safe in this population,91
pathogenesis of BPD.67–69 Although use of maternal an- although it does not seem to affect surfactant compo-
tibiotics is recommended when intraamniotic infection is sition92 or pulmonary function.93 At present, routine
suspected/diagnosed,70 antenatal interventions to treat use of iNO to decrease BPD in the preterm population
chorioamnionitis have not decreased BPD; postnatal in- is not recommended,94 pending long-term outcome data.
terventions may be too late to be effective.71 The use of postnatal steroids, of which dexameth-
Novel approaches/therapies being used for man- asone has been the most well studied, has been quite
agement/prevention of BPD include ventilatory and controversial, with the pendulum swinging from one
pharmacologic strategies. Studies performed by using extreme to the other.71 Another steroid, hydrocorti-
patient-triggered ventilation have not shown any sone, did not improve survival without BPD in an
reduction in the incidence of BPD,72,73 although it has RCT, and the trial had to be terminated early because
been suggested that it may be more beneficial for infants hydrocortisone-treated infants who received indo-
with a BW of ⬍1000 g.74 In a meta-analysis that com- methacin had more gastrointestinal perforations.95
pared volume-targeted ventilation to pressure-limited The well-documented efficacy in extubation96 needs to
ventilation, there was no difference in the incidence of be balanced with the potential for causing neurode-
BPD between 2 groups.75 Research is ongoing to better velopmental harm,97 and more research is needed to
define the parameters that may be potentially benefi- find the “just-right” steroid preparation and its dosing
cial.73 Because none of the new devices have been and timing.
shown to be superior in preventing BPD, avoiding ven- In most circumstances, maintenance of target oxy-
tilation might be the best approach. Toward that end, gen-saturation ranges of ⬎90% to ⬍95% is prudent in
early surfactant-replacement therapy with extubation to preterm neonates to avoid oxygen toxicity in an attempt

PEDIATRICS Volume 123, Number 6, June 2009 1565


Downloaded from www.pediatrics.org by on September 2, 2010
TABLE 5 Management of BPD According to Phase (Early, Evolving, and Established)
Therapeutic Intervention Current Status Evidence Recommendation
Level Level
Early phase (up to 1 postnatal week)
Oxygen supplementation A wide variation in the acceptable oxygen-saturation levels exists across I A
centers but it is generally ⬍95%73
Ventilatory strategy Avoid intubation; if intubated, give ‘early’ surfactant167 I A
Use short inspiratory times (0.24–0.4 s),168 rapid rates (40–60 per min) and I A
low PIP (14–20 cm H2O), moderate PEEP (4–6 cm H2O), and tidal volumes III B
(3–6 mL/kg)72
Extubate early to SNIPPV/nasal CPAP143 III B
Blood gas targets: pH 7.25–7.35 I A
PaO2: 40–60 mm Hg; PaCO2: 45–55 mm Hg169 III B
High-frequency ventilation for ‘rescue’ if conventional ventilation fails73 I C
Methylxanthines Improves successful extubation rate170 I A
Decreases BPD144 I A
Vitamin A If considering use, dose is 5000 IU administered intramuscularly 3 times per I A
week for 4 wks; 1 additional infant survived without BPD for every 14–15
infants who received vitamin A171,172
Fluids Restrictive fluid intake may decrease BPD110,116,173 II-2 B
Nutrition Provide increased energy intake174 I B
Evolving phase (⬎1 postnatal week
to 36 weeks’ PMA)
Oxygen supplementation Same as for early phase I A
Ventilatory strategy Avoid endotracheal tube ventilation; maximize noninvasive ventilation I A
(SNIPPV/nasal CPAP) for respiratory support143
Blood gas targets: pH 7.25–7.35; PaO2: 50–70 mm Hg; PaCO2: 50–60 mm Hg72 III B
Methylxanthines Same as for early phase I A
Vitamin A Same as for early phase; if using, continue for 4 postnatal weeks I A
Steroids Dexamethasone is effective in weaning off mechanical ventilation when I A
used “moderately early” and “delayed”175,176
Increased incidence of neurologic sequelae with early use (⬍96 h)177 I D
Diuretics Furosemide: may use daily or every other day with transient improvement I B
in lung function178
Spironolactone and thiazides: chronic therapy improves lung function, I B
decreases oxygen requirements178
Nutrition Same as for early phase I B
Established phase (⬎36 weeks’ PMA)
Oxygen supplementation For prevention of pulmonary hypertension and cor pulmonale; a wide III C
variation in the acceptable oxygen saturation levels exists across centers,
but it is generally ⬃95%73,179
Ventilatory strategy Blood gas targets: pH 7.25–7.35; PaO2: 50–70 mm Hg; PaCO2: 50–65 mm Hg72 III B
Steroids Oral prednisolone may be helpful in weaning oxygen180 II-2 C
Diuretics Chronic therapy as for evolving phase I B
␤ -agonists Transient relief: increased compliance and reduced pulmonary resistance178; I C
no significant effect on incidence or severity of BPD181
Anticholinergics Used in combination with ␤ agonists in infants with bronchospasm; II-3 C
Increased compliance and decreased respiratory system resistance182
Nutrition Same as for early phase I B
Immunization Prophylaxis against RSV and influenza decreases incidence of I A
rehospitalization and morbidity
The references noted refer to the sources of information for the requisite level of evidence and recommendation for clinical use. PIP indicates peak inspiratory pressure; PEEP, positive end
expiratory pressure; SNIPPV, synchronized nasal intermittent positive pressure ventilation.

to prevent the development of BPD,98 although achiev- BPD was reported in 2 trials and was found to be signif-
ing intended targets may be difficult.99,100 Identification icantly reduced.101
of the optimal target oxygen-saturation ranges is an area
of intense research.
Other drugs studied include the following: Macrolide Antibiotics
Erythromycin has not been shown to decrease BPD in
Inositol infants when treated prophylactically or after known
In a meta-analysis of RCTs using inositol supplemen- Ureaplasma colonization.102 In a pilot study, azithromycin
tation in premature infants, the outcome of death or did not reduce the incidence of BPD.103

1566 BHANDARI, BHANDARI


Downloaded from www.pediatrics.org by on September 2, 2010
TABLE 6 Checklist of Specific Outstanding Questions in BPD
Question Comment
How is the pulmonary vasculature dysregulated, and what is the role of Understanding the causal role of specific molecular targets (including biomarkers
epithelium in the impaired alveolarization in BPD? listed in Table 2) in animal models of BPD is needed; proteomic approaches
may be useful to identify novel markers in humans
What are the candidate genes contributing to BPD? Unraveling the candidate genes and their validation will be important steps
before therapeutic targets can be addressed21; validation of experimental
findings in developmentally appropriate in vivo models, given the unique
responses of the newborn, is critical183
Should infants be managed primarily with noninvasive ventilatory The timing and use of intratracheal surfactant is a related issue
techniques to decrease BPD?
What is the most appropriate target oxygen-saturation ranges to This critical question still needs to be worked out184,185
prevent/manage infants with BPD?
Do steroids, diuretics, and iNO make any impact on The type, dose, and duration of steroids to be used needs to be worked
preventing/managing BPD? out71,96,97,185,186; although frequently used, diuretics have been the least studied
for efficacy in BPD185,187; and the efficacy, safety, and cost-effectiveness of iNO
in preterm neonates to prevent BPD is still unresolved90,94,185
What are the long-term outcomes of survivors with the new BPD? Pulmonary and neurodevelopmental outcome studies need to be performed to
assess the potential health impact of BPD up to adulthood

N-Acetyl Cysteine Morbidity


In a large RCT, use of a 6-day course of N-acetyl Infants with BPD have higher rates of rehospitalizations
cysteine did not prevent BPD or death in infants with (up to 50%) in the first year of life.118,119 Respiratory
BWs of ⬍1000 g.104 symptoms in patients with BPD persist beyond the first 2
years of life into the preschool years,120 adolescence, and
early adulthood.121,122 It is unclear whether BPD severity
Recombinant Clara Cell 10-Kilodalton Protein or prematurity per se influences the persistence and
Use of an antiinflammatory protein, recombinant hu- severity of symptoms.
man Clara cell 10-kilodalton protein (CC10), has shown
some initial promise.105 Radiologic Findings
Chest radiograph and computed tomography (CT) scan
abnormalities persist into adolescence and adulthood,
Recombinant Superoxide Dismutase
with CT being more sensitive.123,124 CT scans of infants
Although the antioxidant recombinant Cu-Zn super- with new BPD are not much different from those of old
oxide dismutase (SOD) has not shown any difference in BPD except for the absence of bronchial involvement.125
outcome,106 in one study infants ⬍27 weeks’ GA who A more severe clinical course correlates with greater
received SOD had decreased hospitalizations and emer- radiologic and pulmonary function abnormalities.125 A
gency department visits and less-frequent use of bron- CT scan scoring system may help with BPD prognosis.126
chodilator therapy at age 1 year compared with the
infants who did not receive SOD,107 suggesting that SOD
may have interrupted an inflammatory cascade involv- Pulmonary Function
ing reactive oxygen species and possibly conferring a Patients with BPD continue to have significant impair-
long-term benefit. ment and deterioration in lung function into late ado-
Given the complex nature of BPD (Fig 1), it is un- lescence.127,128 An inverse relationship has been reported
likely that a single “magic bullet” (with the possible between forced expiratory volume in 1 second at school
exception of prevention of preterm birth) will make a age and duration of supplemental oxygen.129 Conversely,
significant impact on BPD. Incremental improvements others have reported poor correlation between duration
possible by targeting multiple aspects of this condition of mechanical ventilation and duration of oxygen sup-
include early diagnosis and treatment of antenatal and plementation and forced expiratory flow in infants with
postnatal sepsis, optimal maintenance of oxygenation BPD.130 Another problem is that there have been few
and fluid/electrolyte status, and aggressive early paren- studies comparing various modes of ventilation with
teral/enteral nutrition, along with appropriate ventila- pulmonary function of BPD survivors. Infants who re-
tory strategies.108–116 The potential of using such a com- ceived either high-frequency oscillatory ventilation
bined approach has shown promise.117 (HFOV) or conventional mechanical ventilation (CMV)
demonstrated low expiratory flows at 6 and 12
months.131 At 12 months, however, infants who under-
PULMONARY OUTCOMES went HFOV had significantly better lung function.131
It is important to keep in mind the limitations of most of Another study compared lung function of preterm in-
these studies, which included preterm infants born be- fants with a history of respiratory distress syndrome or
fore extensive use of antenatal steroids and surfactant. BPD at 8 to 9 years of age treated with CMV to similar

PEDIATRICS Volume 123, Number 6, June 2009 1567


Downloaded from www.pediatrics.org by on September 2, 2010
groups who received HFOV.132 There was no significant ● The efficacy and safety of caffeine in the prevention of
difference in the degree of mild obstructive lung disease BPD144,145
as a function of ventilation mode, suggesting that other
factors influence long-term pulmonary outcome.132 Some specific outstanding questions are detailed in
Another problem is that not many studies have ex- Table 6.
amined whether abnormal lung function in infancy cor-
relates with extent of abnormality at a later stage. Filli-
CONCLUSIONS
pone et al133 measured lung function in the same group
Progress has been made, but problems persist, and addi-
of children at school age and reported a significant cor-
tional research is required to improve outcomes. Focus-
relation between the functional residual capacity (Vmax
ing on aspects of basic biology (lung development, iden-
[FRC]) measured at 24 months and both forced expira-
tification of biomarkers, genetics, animal models) and
tory volume in 1 second (FEV1) and forced midexpira-
pharmacotherapeutic approaches6 will have the maxi-
tory flow (FEF25%–75%). No serial data have been re-
mum potential to make such an impact. Significant im-
ported for adolescents and adults.
provements in the future will likely depend on our
Most studies have shown no reduction in exercise
ability to identify the genetic components of BPD and
capacity in children with BPD when compared with
target specific therapies.19
healthy term infants or preterm infants without lung
disease.134 From recent studies of large cohorts of pre-
mature infants, significant decrease in exercise capacity REFERENCES
was reported despite normal mean lung function or mild 1. Baraldi E, Filippone M. Chronic lung disease after premature
small airway obstruction and gas trapping,135 which sug- birth. N Engl J Med. 2007;357(19):1946 –1955
gests that impaired exercise tolerance was perhaps sec- 2. Bancalari E, Claure N. Definitions and diagnostic criteria for
ondary to poor fitness that may be improved with an bronchopulmonary dysplasia. Semin Perinatol. 2006;30(4):
exercise training program.136 164 –170
3. Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J
Lung abnormalities that may persist into adulthood
Respir Crit Care Med. 2001;163(7):1723–1729
include airway obstruction, airway hyperreactivity, and
4. Fanaroff AA, Hack M, Walsh MC. The NICHD neonatal re-
emphysema.123,128,135,137,138 search network: changes in practice and outcomes during the
first 15 years. Semin Perinatol. 2003;27(4):281–287
NEURODEVELOPMENTAL OUTCOMES 5. Fanaroff AA, Stoll BJ, Wright LL, et al. Trends in neonatal
Most studies of BPD have been cross-sectional in nature, morbidity and mortality for very low birthweight infants.
and the independent effect of BPD is difficult to assess Am J Obstet Gynecol. 2007;196(2):147.e1–147.e8
6. Walsh MC, Szefler S, Davis J, et al. Summary proceedings
given the high likelihood of the presence of additional
from the bronchopulmonary dysplasia group. Pediatrics. 2006;
medical complications in these infants. Although pre- 117(3 pt 2):S52–S56
term infants have an increased risk of neurodevelop- 7. Walsh MC, Yao Q, Gettner P, et al. Impact of a physiologic
mental impairment, BPD is an additional risk factor.139 definition on bronchopulmonary dysplasia rates. Pediatrics.
This is probably a result of multiple contributing factors 2004;114(5):1305–1311
including frequent episodes of hypoxia, poor growth, 8. Ehrenkranz RA, Walsh MC, Vohr BR, et al. Validation of the
and, potentially, postnatal steroids.139 Infants of ⬍1500 g National Institutes of Health consensus definition of broncho-
BW with BPD have greater fine and gross motor skill pulmonary dysplasia. Pediatrics. 2005;116(6):1353–1360
impairment as well as cognitive function and language 9. Thébaud B, Abman SH. Bronchopulmonary dysplasia: where
delay compared with those without BPD.140,141 Children have all the vessels gone? Roles of angiogenic growth factors
in chronic lung disease. Am J Respir Crit Care Med. 2007;
with severe BPD have worse outcomes and require more
175(10):978 –985
interventions at 8 years of age than do children with 10. De Paepe ME, Patel C, Tsai A, Gundavarapu S, Mao Q. En-
mild or moderate BPD.142 doglin (CD105) upregulation in pulmonary microvasculature
BPD does not seem to be associated with a specific of ventilated preterm infants. Am J Respir Crit Care Med. 2008;
neuropsychological but, rather, a global impairment.139 178(2):180 –187
The spectrum of neurodevelopmental impairment seems 11. De Paepe ME, Mao Q, Powell J, et al. Growth of pulmonary
to correlate well with BPD disease severity.8,119 microvasculature in ventilated preterm infants. Am J Respir
Crit Care Med. 2006;173(2):204 –211
12. Abman SH. The dysmorphic pulmonary circulation in bron-
SUMMARY chopulmonary dysplasia: a growing story. Am J Respir Crit Care
Over the last 5 years, progress has been made in the Med. 2008;178(2):114 –115
definition, pathogenesis, and management strategies for 13. Coalson JJ. Pathology of bronchopulmonary dysplasia. Semin
BPD: Perinatol. 2006;30(4):179 –184
14. Bhatt AJ, Pryhuber GS, Huyck H, Watkins RH, Metlay LA,
● A consensus-validated definition of BPD, with catego- Maniscalco WM. Disrupted pulmonary vasculature and de-
rization of severity (Table 1)8 creased vascular endothelial growth factor, Flt-1, and TIE-2 in
● Identification and quantification of the genetic contri- human infants dying with bronchopulmonary dysplasia. Am J
Respir Crit Care Med. 2001;164(10 pt 1):1971–1980
bution to BPD19
15. Coalson JJ. Pathology of new bronchopulmonary dysplasia.
● Resurgence of noninvasive ventilation in the preven- Semin Neonatol. 2003;8(1):73– 81
tion and management of BPD143 16. Thibeault DW, Mabry SM, Norberg M, Truog WE, Ekekezie,

1568 BHANDARI, BHANDARI


Downloaded from www.pediatrics.org by on September 2, 2010
II. Lung microvascular adaptation in infants with chronic lung aspirates of mechanically ventilated preterm neonates. Pediatr
disease. Biol Neonate. 2004;85(4):273–282 Res. 2003;53(2):240 –244
17. Bhandari A, Bhandari V. Pathogenesis, pathology and patho- 36. de Blic J, Midulla F, Barbato A, et al. Bronchoalveolar lavage
physiology of pulmonary sequelae of bronchopulmonary dys- in children. ERS Task Force on Bronchoalveolar Lavage in
plasia in premature infants. Front Biosci. 2003;8:e370 – e380 Children. European Respiratory Society. Eur Respir J. 2000;
18. Bhandari A, Bhandari V. Bronchopulmonary dysplasia: an 15(1):217–231
update. Indian J Pediatr. 2007;74(1):73–77 37. Cayabyab RG, Jones CA, Kwong KY, et al. Interleukin-1beta
19. Bhandari V, Bizzarro MJ, Shetty A, et al. Familial and genetic in the bronchoalveolar lavage fluid of premature neonates: a
susceptibility to major neonatal morbidities in preterm twins. marker for maternal chorioamnionitis and predictor of ad-
Pediatrics. 2006;117(6):1901–1906 verse neonatal outcome. J Matern Fetal Neonatal Med. 2003;
20. Parker RA, Lindstrom DP, Cotton RB. Evidence from twin 14(3):205–211
study implies possible genetic susceptibility to bronchopulmo- 38. Mahieu LM, De Dooy JJ, Ieven MM, Bridts CH, Stevens
nary dysplasia. Semin Perinatol. 1996;20(3):206 –209 WJ. Increased levels of tumor necrosis factor-alpha and de-
21. Bhandari V, Gruen JR. The genetics of bronchopulmonary creased levels of interleukin-12 p70 in tracheal aspirates,
dysplasia. Semin Perinatol. 2006;30(4):185–191 within 2 hrs after birth, are associated with mortality among
22. Hallman M, Marttila R, Pertile R, Ojaniemi M, Haataja R. ventilated preterm infants. Pediatr Crit Care Med. 2005;6(6):
Genes and environment in common neonatal lung disease. 682– 689
Neonatology. 2007;91(4):298 –302 39. Reuter SD, O’Donovan DJ, Hegemier SE, Smith EO, Heird
23. Parton LA, Strassberg SS, Qian D, Galvin-Parton PA, Cristea WC, Fernandes CJ. Urinary F2-isoprostanes are poor prog-
IA. The genetic basis for bronchopulmonary dysplasia. Front nostic indicators for the development of bronchopulmonary
Biosci. 2006;11:1854 –1860 dysplasia. J Perinatol. 2007;27(5):303–306
24. Bokodi G, Treszl A, Kovacs L, Tulassay T, Vasarhelyi B. 40. Cullen A, Van Marter LJ, Allred EN, Moore M, Parad RB,
Dysplasia: a review. Pediatr Pulmonol. 2007;42(10):952–961 Sunday ME. Urine bombesin-like peptide elevation precedes
25. D’Angio CT, Basavegowda K, Avissar NE, Finkelstein JN, clinical evidence of bronchopulmonary dysplasia. Am J Respir
Sinkin RA. Comparison of tracheal aspirate and bronchoal- Crit Care Med. 2002;165(8):1093–1097
veolar lavage specimens from premature infants. Biol Neonate. 41. de Felice C, Latini G, Parrini S, et al. Oral mucosal microvas-
2002;82(3):145–149 cular abnormalities: an early marker of bronchopulmonary
dysplasia. Pediatr Res. 2004;56(6):927–931
26. Nedrelow JH, Bhandari V. Interleukin (IL)-6 to vascular en-
42. Bland RD, Ertsey R, Mokres LM, et al. Mechanical ventilation
dothelial growth factor (VEGF) ratio predicts the develop-
uncouples synthesis and assembly of elastin and increases
ment of bronchopulmonary dysplasia (BPD)/death in prema-
apoptosis in lungs of newborn mice: prelude to defective
ture infants [abstract]. Pediatr Res. 2004;55(4):496A
alveolar septation during lung development? Am J Physiol
27. Bhandari V, Choo-Wing R, Lee CG, et al. Hyperoxia causes
Lung Cell Mol Physiol. 2008;294(1):L3–L14
angiopoietin 2-mediated acute lung injury and necrotic cell
43. Bland RD, Xu L, Ertsey R, et al. Dysregulation of pulmonary
death. Nat Med. 2006;12(11):1286 –1293
elastin synthesis and assembly in preterm lambs with chronic
28. Bourbia A, Cruz MA, Rozycki HJ. NF-kappaB in tracheal
lung disease. Am J Physiol Lung Cell Mol Physiol. 2007;292(6):
lavage fluid from intubated premature infants: association
L1370 –L1384
with inflammation, oxygen, and outcome. Arch Dis Child Fetal
44. Altiok O, Yasumatsu R, Bingol-Karakoc G, et al. Imbalance
Neonatal Ed. 2006;91(1):F36 –F39
between cysteine proteases and inhibitors in a baboon model
29. Cederqvist K, Sorsa T, Tervahartiala T, et al. Matrix metallo-
of bronchopulmonary dysplasia. Am J Respir Crit Care Med.
proteinases-2, -8, and -9 and TIMP-2 in tracheal aspirates
2006;173(3):318 –326
from preterm infants with respiratory distress. Pediatrics. 2001; 45. Hirakawa H, Pierce RA, Bingol-Karakoc G, et al. Cathepsin S
108(3):686 – 692 deficiency confers protection from neonatal hyperoxia-
30. Lassus P, Heikkila P, Andersson LC, von Boguslawski K, induced lung injury. Am J Respir Crit Care Med. 2007;176(8):
Andersson S. Lower concentration of pulmonary hepatocyte 778 –785
growth factor is associated with more severe lung disease in 46. Padela S, Cabacungan J, Shek S, et al. Hepatocyte growth
preterm infants. J Pediatr. 2003;143(2):199 –202 factor is required for alveologenesis in the neonatal rat. Am J
31. Baier RJ, Majid A, Parupia H, Loggins J, Kruger TE. CC Respir Crit Care Med. 2005;172(7):907–914
chemokine concentrations increase in respiratory distress 47. Frank L. Protective effect of keratinocyte growth factor
syndrome and correlate with development of bronchopulmo- against lung abnormalities associated with hyperoxia in pre-
nary dysplasia. Pediatr Pulmonol. 2004;37(2):137–148 maturely born rats. Biol Neonate. 2003;83(4):263–272
32. Ekekezie II, Thibeault DW, Simon SD, et al. Low levels of 48. Bry K, Whitsett JA, Lappalainen U. IL-1beta disrupts postna-
tissue inhibitors of metalloproteinases with a high matrix tal lung morphogenesis in the mouse. Am J Respir Cell Mol Biol.
metalloproteinase-9/tissue inhibitor of metalloproteinase-1 2007;36(1):32– 42
ratio are present in tracheal aspirate fluids of infants who 49. Choo-Wing R, Nedrelow JH, Homer RJ, Elias JA, Bhandari V.
develop chronic lung disease. Pediatrics. 2004;113(6): Developmental differences in the responses of IL-6 and IL-13
1709 –1714 transgenic mice exposed to hyperoxia. Am J Physiol Lung Cell
33. Munshi UK, Niu JO, Siddiq MM, Parton LA. Elevation of Mol Physiol. 2007;293(1):L142–L150
interleukin-8 and interleukin-6 precedes the influx of neu- 50. Vozzelli MA, Mason SN, Whorton MH, Auten RL Jr. Anti-
trophils in tracheal aspirates from preterm infants who de- macrophage chemokine treatment prevents neutrophil and
velop bronchopulmonary dysplasia. Pediatr Pulmonol. 1997; macrophage influx in hyperoxia-exposed newborn rat
24(5):331–336 lung. Am J Physiol Lung Cell Mol Physiol. 2004;286(3):
34. Kotecha S, Wangoo A, Silverman M, Shaw RJ. Increase in the L488 –L493
concentration of transforming growth factor beta-1 in bron- 51. Vicencio AG, Lee CG, Cho SJ, et al. Conditional overexpres-
choalveolar lavage fluid before development of chronic lung sion of bioactive transforming growth factor-beta1 in neonatal
disease of prematurity. J Pediatr. 1996;128(4):464 – 469 mouse lung: a new model for bronchopulmonary dysplasia?
35. Ambalavanan N, Novak ZE. Peptide growth factors in tracheal Am J Respir Cell Mol Biol. 2004;31(6):650 – 656

PEDIATRICS Volume 123, Number 6, June 2009 1569


Downloaded from www.pediatrics.org by on September 2, 2010
52. Nakanishi H, Sugiura T, Streisand JB, Lonning SM, Roberts continuing story. Semin Fetal Neonatal Med. 2006;11(5):
JD Jr. TGF-beta-neutralizing antibodies improve pulmonary 354 –362
alveologenesis and vasculogenesis in the injured newborn 69. Kallapur SG, Jobe AH. Contribution of inflammation to lung
lung. Am J Physiol Lung Cell Mol Physiol. 2007;293(1): injury and development. Arch Dis Child Fetal Neonatal Ed.
L151–L161 2006;91(2):F132–F135
53. Asikainen TM, Ahmad A, Schneider BK, White CW. Effect of 70. Fahey JO. Clinical management of intra-amniotic infection
preterm birth on hypoxia-inducible factors and vascular en- and chorioamnionitis: a review of the literature. J Midwifery
dothelial growth factor in primate lungs. Pediatr Pulmonol. Womens Health. 2008;53(3):227–235
2005;40(6):538 –546 71. Watterberg K. Anti-inflammatory therapy in the neonatal
54. Tambunting F, Beharry KD, Waltzman J, Modanlou HD. Im- intensive care unit: present and future. Semin Fetal Neonatal
paired lung vascular endothelial growth factor in extremely Med. 2006;11(5):378 –384
premature baboons developing bronchopulmonary dysplasia/ 72. Ambalavanan N, Carlo WA. Ventilatory strategies in the pre-
chronic lung disease. J Investig Med. 2005;53(5):253–262 vention and management of bronchopulmonary dysplasia.
55. Kunig AM, Balasubramaniam V, Markham NE, et al. Recom- Semin Perinatol. 2006;30(4):192–199
binant human VEGF treatment enhances alveolarization after 73. Greenough A. How has research in the past 5 years changed
hyperoxic lung injury in neonatal rats. Am J Physiol Lung Cell my clinical practice? Arch Dis Child Fetal Neonatal Ed. 2007;
Mol Physiol. 2005;289(4):L529 –L535 92(5):F404 –F407
56. Maniscalco WM, Watkins RH, Pryhuber GS, Bhatt A, Shea C, 74. Claure N, Bancalari E. New modes of mechanical ventilation
Huyck H. Angiogenic factors and alveolar vasculature: devel- in the preterm newborn: evidence of benefit. Arch Dis Child
opment and alterations by injury in very premature baboons. Fetal Neonatal Ed. 2007;92(6):F508 –F512
Am J Physiol Lung Cell Mol Physiol. 2002;282(4):L811–L823 75. McCallion N, Davis PG, Morley CJ. Volume-targeted versus
57. Lassus P, Turanlahti M, Heikkila P, et al. Pulmonary vascular pressure-limited ventilation in the neonate. Cochrane Database
endothelial growth factor and Flt-1 in fetuses, in acute and Syst Rev. 2005;(3):CD003666
chronic lung disease, and in persistent pulmonary hyperten- 76. Stevens TP, Harrington EW, Blennow M, Soll RF. Early sur-
sion of the newborn. Am J Respir Crit Care Med. 2001;164(10 pt factant administration with brief ventilation vs. selective sur-
1):1981–1987 factant and continued mechanical ventilation for preterm
58. Thébaud B, Ladha F, Michelakis ED, et al. Vascular endothe- infants with or at risk for respiratory distress syndrome. Co-
lial growth factor gene therapy increases survival, promotes
chrane Database Syst Rev. 2007;(4):CD003063
lung angiogenesis, and prevents alveolar damage in hyperoxia-
77. Morley CJ, Davis PG, Doyle LW, Brion LP, Hascoet JM, Carlin
induced lung injury: evidence that angiogenesis participates
JB. Nasal CPAP or intubation at birth for very preterm infants.
in alveolarization. Circulation. 2005;112(16):2477–2486
N Engl J Med. 2008;358(7):700 –708
59. Subramaniam M, Bausch C, Twomey A, et al. Bombesin-like
78. Bhandari V, Gavino RG, Nedrelow JH, et al. A randomized
peptides modulate alveolarization and angiogenesis in bron-
controlled trial of synchronized nasal intermittent positive
chopulmonary dysplasia. Am J Respir Crit Care Med. 2007;
pressure ventilation in RDS. J Perinatol. 2007;27(11):697–703
176(9):902–912
79. Kugelman A, Feferkorn I, Riskin A, Chistyakov I, Kaufman B,
60. Buhimschi CS, Weiner CP, Buhimschi IA. Clinical proteomics:
Bader D. Nasal intermittent mandatory ventilation versus
a novel diagnostic tool for the new biology of preterm labor,
nasal continuous positive airway pressure for respiratory dis-
part I: proteomics tools. Obstet Gynecol Surv. 2006;61(7):
tress syndrome: a randomized, controlled, prospective study.
481– 486
J Pediatr. 2007;150(5):521–526
61. Dodd JM, Crowther CA, Cincotta R, Flenady V, Robinson JS.
80. Donn SM. Lucinactant: a novel synthetic surfactant for the
Progesterone supplementation for preventing preterm birth: a
systematic review and meta-analysis. Acta Obstet Gynecol Scand. treatment of respiratory distress syndrome. Expert Opin Investig
2005;84(6):526 –533 Drugs. 2005;14(3):329 –334
62. Rosenberg L, Palmer JR, Wise LA, Horton NJ, Corwin MJ. 81. Sinha SK, Lacaze-Masmonteil T, Valls i Soler A, et al. A mul-
Perceptions of racial discrimination and the risk of preterm ticenter, randomized, controlled trial of lucinactant versus
birth. Epidemiology. 2002;13(6):646 – 652 poractant alfa among very premature infants at high risk for
63. Gagliardi L, Bellù R, Rusconi F, Merazzi D, Mosca F. Antena- respiratory distress syndrome. Pediatrics. 2005;115(4):
tal steroids and risk of bronchopulmonary dysplasia: a lack of 1030 –1038
effect or a case of over-adjustment? Paediatr Perinat Epidemiol. 82. Moya FR, Gadzinowski J, Bancalari E, et al. A multicenter,
2007;21(4):347–353 randomized, masked, comparison trial of lucinactant, colfos-
64. Guinn DA, Atkinson MW, Sullivan L, et al. Single vs weekly ceril palmitate, and beractant for the prevention of respiratory
courses of antenatal corticosteroids for women at risk of pre- distress syndrome among very preterm infants. Pediatrics.
term delivery: a randomized controlled trial. JAMA. 2001; 2005;115(4):1018 –1029
286(13):1581–1587 83. Pfister RH, Soll RF, Wiswell T. Protein containing synthetic
65. Feldman DM, Carbone J, Belden L, Borgida AF, Herson V. surfactant versus animal derived surfactant extract for the
Betamethasone vs dexamethasone for the prevention of mor- prevention and treatment of respiratory distress syndrome.
bidity in very-low-birthweight neonates. Am J Obstet Gynecol. Cochrane Database Syst Rev. 2007;(4):CD006069
2007;197(3):284.e1–284.e4 84. Donn SM, Sinha SK. Aerosolized lucinactant: a potential al-
66. Lee BH, Stoll BJ, McDonald SA, Higgins RD; National Institute ternative to intratracheal surfactant replacement therapy. Ex-
of Child Health and Human Development Neonatal Research pert Opin Pharmacother. 2008;9(3):475– 478
Network. Adverse neonatal outcomes associated with ante- 85. Bhandari V. Making babies breathe better: hopeful signals?
natal dexamethasone versus antenatal betamethasone. Pedi- Pediatr Res. 2008;64(2):123–124
atrics. 2006;117(5):1503–1510 86. Mestan KK, Marks JD, Hecox K, Huo D, Schreiber MD. Neu-
67. Ryan RM, Ahmed Q, Lakshminrusimha S. Inflammatory me- rodevelopmental outcomes of premature infants treated with
diators in the immunobiology of bronchopulmonary dyspla- inhaled nitric oxide. N Engl J Med. 2005;353(1):23–32
sia. Clin Rev Allergy Immunol. 2008;34(2):174 –190 87. Schreiber MD, Gin-Mestan K, Marks JD, Huo D, Lee G, Srisu-
68. Speer CP. Inflammation and bronchopulmonary dysplasia: a parp P. Inhaled nitric oxide in premature infants with the

1570 BHANDARI, BHANDARI


Downloaded from www.pediatrics.org by on September 2, 2010
respiratory distress syndrome. N Engl J Med. 2003;349(22): combinant human Clara cell protein in premature infants
2099 –2107 with respiratory distress syndrome. Pediatr Res. 2005;58(1):
88. Field D, Elbourne D, Truesdale A, et al. Neonatal ventilation 15–21
with inhaled nitric oxide versus ventilatory support without 106. Davis JM, Rosenfeld WN, Richter SE, et al. Safety and phar-
inhaled nitric oxide for preterm infants with severe respira- macokinetics of multiple doses of recombinant human CuZn
tory failure: the INNOVO Multicentre Randomised Controlled superoxide dismutase administered intratracheally to prema-
Trial (ISRCTN 17821339). Pediatrics. 2005;115(4):926 –936 ture neonates with respiratory distress syndrome. Pediatrics.
89. Hascoet JM, Fresson J, Claris O, et al. The safety and efficacy 1997;100(1):24 –30
of nitric oxide therapy in premature infants. J Pediatr. 2005; 107. Davis JM, Parad RB, Michele T, Allred E, Price A, Rosenfeld
146(3):318 –323 W. Pulmonary outcome at 1 year corrected age in premature
90. Barrington KJ, Finer NN. Inhaled nitric oxide for preterm infants treated at birth with recombinant human CuZn su-
infants: a systematic review [published correction appears in peroxide dismutase. Pediatrics. 2003;111(3):469 – 476
Pediatrics. 2008;121(2):451]. Pediatrics. 2007;120(5): 108. Akram Khan M, Kuzma-O’Reilly B, Brodsky NL, Bhandari V.
1088 –1099 Site-specific characteristics of infants developing bronchopul-
91. Ballard PL, Truog WE, Merrill JD, et al. Plasma biomarkers of monary dysplasia [published correction appears in J Perinatol.
oxidative stress: relationship to lung disease and inhaled nitric 2006;26(8):524]. J Perinatol. 2006;26(7):428 – 435
oxide therapy in premature infants. Pediatrics. 2008;121(3): 109. Kulkarni A, Ehrenkranz RA, Bhandari V. Effect of introduc-
555–561 tion of synchronized nasal intermittent positive-pressure ven-
92. Ballard PL, Merrill JD, Truog WE, et al. Surfactant function tilation in a neonatal intensive care unit on bronchopulmo-
and composition in premature infants treated with inhaled nary dysplasia and growth in preterm infants. Am J Perinatol.
nitric oxide. Pediatrics. 2007;120(2):346 –353 2006;23(4):233–240
93. Di Fiore JM, Hibbs AM, Zadell AE, et al. The effect of inhaled 110. Bhandari V, Brodsky N, Porat R. Improved outcome of ex-
nitric oxide on pulmonary function in preterm infants. J tremely low birth weight infants with Tegaderm application
Perinatol. 2007;27(12):766 –771 to skin. J Perinatol. 2005;25(4):276 –281
94. Dani C, Bertini G. Inhaled nitric oxide for the treatment of 111. Bhandari V, Fall P, Raisz L, Rowe J. Potential biochemical
preterm infants with respiratory distress syndrome. growth markers in premature infants. Am J Perinatol. 1999;
Neonatology. 2008;94(2):87–95 16(7):339 –349
95. Watterberg KL, Gerdes JS, Cole CH, et al. Prophylaxis of early
112. Buhimschi CS, Buhimschi IA, Abdel-Razeq S, et al. Proteomic
adrenal insufficiency to prevent bronchopulmonary
biomarkers of intra-amniotic inflammation: relationship with
dysplasia: a multicenter trial. Pediatrics. 2004;114(6):
funisitis and early onset sepsis in the premature neonate.
1649 –1657
Pediatr Res. 2007;61(3):318 –324
96. Doyle LW, Davis PG, Morley CJ, McPhee A, Carlin JB; DART
113. Walsh M, Laptook A, Kazzi SN, et al. A cluster-randomized
Study Investigators. Low-dose dexamethasone facilitates ex-
trial of benchmarking and multimodal quality improvement
tubation among chronically ventilator-dependent infants: a
to improve rates of survival free of bronchopulmonary dys-
multicenter, international, randomized, controlled trial. Pedi-
plasia for infants with birth weights of less than 1250 grams.
atrics. 2006;117(1):75– 83
Pediatrics. 2007;119(5):876 – 890
97. Doyle LW, Halliday HL, Ehrenkranz RA, Davis PG, Sinclair JC.
114. Payne NR, LaCorte M, Karna P, et al. Reduction of broncho-
Impact of postnatal systemic corticosteroids on mortality and
pulmonary dysplasia after participation in the Breathsavers
cerebral palsy in preterm infants: effect modification by risk
Group of the Vermont Oxford Network Neonatal Intensive
for chronic lung disease. Pediatrics. 2005;115(3):655– 661
Care Quality Improvement Collaborative. Pediatrics. 2006;
98. Askie LM, Henderson-Smart DJ, Irwig L, Simpson JM. Oxygen-
saturation targets and outcomes in extremely preterm infants. 118(suppl 2):S73–S77
N Engl J Med. 2003;349(10):959 –967 115. Buhimschi CS, Bhandari V, Hamar BD, et al. Proteomic pro-
99. Hagadorn JI, Furey AM, Nghiem TH, et al. Achieved versus filing of the amniotic fluid to detect inflammation, infection,
intended pulse oximeter saturation in infants born less than and neonatal sepsis. PLoS Med. 2007;4(1):e18
28 weeks’ gestation: the AVIOx Study. Pediatrics. 2006;118(4): 116. Bell EF, Acarregui MJ. Restricted versus liberal water intake
1574 –1582 for preventing morbidity and mortality in preterm infants.
100. Clucas L, Doyle LW, Dawson J, Donath S, Davis PG. Compli- Cochrane Database Syst Rev. 2008;(1):CD000503
ance with alarm limits for pulse oximetry in very preterm 117. Geary C, Caskey M, Fonseca R, Malloy M. Decreased inci-
infants. Pediatrics. 2007;119(6):1056 –1060 dence of bronchopulmonary dysplasia after early manage-
101. Howlett A, Ohlsson A. Inositol for respiratory distress syn- ment changes, including surfactant and nasal continuous pos-
drome in preterm infants. Cochrane Database Syst Rev. 2003; itive airway pressure treatment at delivery, lowered oxygen
(4):CD000366 saturation goals, and early amino acid administration: a his-
102. Mabanta CG, Pryhuber GS, Weinberg GA, Phelps DL. Eryth- torical cohort study. Pediatrics. 2008;121(1):89 –96
romycin for the prevention of chronic lung disease in intu- 118. Smith VC, Zupancic JA, McCormick MC, et al. Rehospitaliza-
bated preterm infants at risk for, or colonized or infected with tion in the first year of life among infants with bronchopul-
Ureaplasma urealyticum. Cochrane Database Syst Rev. 2003;(4): monary dysplasia. J Pediatr. 2004;144(6):799 – 803
CD003744 119. Jeng SF, Hsu CH, Tsao PN, et al. Bronchopulmonary dysplasia
103. Ballard HO, Anstead MI, Shook LA. Azithromycin in the predicts adverse developmental and clinical outcomes in
extremely low birth weight infant for the prevention of bron- very-low-birthweight infants. Dev Med Child Neurol. 2008;
chopulmonary dysplasia: a pilot study. Respir Res. 2007;8:41 50(1):51–57
104. Ahola T, Lapatto R, Raivio KO, et al. N-acetylcysteine does 120. Vrijlandt EJ, Boezen HM, Gerritsen J, Stremmelaar EF, Duiv-
not prevent bronchopulmonary dysplasia in immature erman EJ. Respiratory health in prematurely born preschool
infants: a randomized controlled trial. J Pediatr. 2003;143(6): children with and without bronchopulmonary dysplasia. J Pe-
713–719 diatr. 2007;150(3):256 –261
105. Levine CR, Gewolb IH, Allen K, et al. The safety, pharmaco- 121. Halvorsen T, Skadberg BT, Eide GE, Roksund OD, Carlsen
kinetics, and anti-inflammatory effects of intratracheal re- KH, Bakke P. Pulmonary outcome in adolescents of extreme

PEDIATRICS Volume 123, Number 6, June 2009 1571


Downloaded from www.pediatrics.org by on September 2, 2010
preterm birth: a regional cohort study. Acta Paediatr. 2004; Available at: www.pediatrics.org/cgi/content/full/112/5/
93(10):1294 –1300 e359. Accessed April 20, 2009
122. Vrijlandt EJ, Gerritsen J, Boezen HM, Duiverman EJ. Gender 141. Lewis BA, Singer LT, Fulton S, et al. Speech and language
differences in respiratory symptoms in 19-year-old adults outcomes of children with bronchopulmonary dysplasia.
born preterm. Respir Res. 2005;6:117 J Commun Disord. 2002;35(5):393– 406
123. Northway WH Jr, Moss RB, Carlisle KB, et al. Late pulmonary 142. Short EJ, Kirchner HL, Asaad GR, et al. Developmental se-
sequelae of bronchopulmonary dysplasia. N Engl J Med. 1990; quelae in preterm infants having a diagnosis of bronchopul-
323(26):1793–1799 monary dysplasia: analysis using a severity-based classifica-
124. Oppenheim C, Mamou-Mani T, Sayegh N, de Blic J, Schein- tion system. Arch Pediatr Adolesc Med. 2007;161(11):
mann P, Lallemand D. Bronchopulmonary dysplasia: value of 1082–1087
CT in identifying pulmonary sequelae. AJR Am J Roentgenol. 143. Bhandari V. Non-invasive ventilation of the sick neonate:
1994;163(1):169 –172 evidence-based recommendations. J Neonatol. 2006;20(3):
125. Mahut B, De Blic J, Emond S, et al. Chest computed tomog- 212–221
raphy findings in bronchopulmonary dysplasia and correla- 144. Schmidt B, Roberts RS, Davis P, et al. Caffeine therapy for
tion with lung function. Arch Dis Child Fetal Neonatal Ed. apnea of prematurity. N Engl J Med. 2006;354(20):2112–2121
2007;92(6):F459 –F464 145. Schmidt B, Roberts RS, Davis P, et al. Long-term effects of
126. Ochiai M, Hikino S, Yabuuchi H, et al. A new scoring system caffeine therapy for apnea of prematurity. N Engl J Med.
for computed tomography of the chest for assessing the clin- 2007;357(19):1893–1902
ical status of bronchopulmonary dysplasia. J Pediatr. 2008; 146. Aghai ZH, Faqiri S, Saslow JG, et al. Angiopoietin 2 concen-
152(1):90 –95, 95.e1–95.e3 trations in infants developing bronchopulmonary dysplasia:
127. Doyle LW. Respiratory function at age 8 –9 years in extremely attenuation by dexamethasone. J Perinatol. 2008;28(2):
low birthweight/very preterm children born in Victoria in 149 –155
1991–1992. Pediatr Pulmonol. 2006;41(6):570 –576 147. Knaapi J, Lukkarinen H, Kiviranta R, et al. Cathepsin K
128. Doyle LW, Faber B, Callanan C, Freezer N, Ford GW, Davis expression is diminished in infants with bronchopulmonary
NM. Bronchopulmonary dysplasia in very low birth weight dysplasia. Acta Paediatr. 2006;95(10):1298 –1300
subjects and lung function in late adolescence. Pediatrics. 148. Ramsay PL, DeMayo FJ, Hegemier SE, Wearden ME, Smith
2006;118(1):108 –113 CV, Welty SE. Clara cell secretory protein oxidation and
129. Kennedy JD, Edward LJ, Bates DJ, et al. Effects of birthweight expression in premature infants who develop bronchopulmo-
and oxygen supplementation on lung function in late child- nary dysplasia. Am J Respir Crit Care Med. 2001;164(1):
hood in children of very low birth weight. Pediatr Pulmonol. 155–161
2000;30(1):32– 40 149. Jónsson B, Li YH, Noack G, Brauner A, Tullus K. Downregu-
130. Robin B, Kim YJ, Huth J, et al. Pulmonary function in bron- latory cytokines in tracheobronchial aspirate fluid from in-
chopulmonary dysplasia. Pediatr Pulmonol. 2004;37(3): fants with chronic lung disease of prematurity. Acta Paediatr.
236 –242 2000;89(11):1375–1380
131. Hofhuis W, Huysman MW, van der Wiel EC, et al. Worsening 150. Choi CW, Kim BI, Kim HS, Park JD, Choi JH, Son DW.
of V’maxFRC in infants with chronic lung disease in the first Increase of interleukin-6 in tracheal aspirate at birth: a pre-
year of life: a more favorable outcome after high-frequency dictor of subsequent bronchopulmonary dysplasia in preterm
oscillation ventilation. Am J Respir Crit Care Med. 2002;166(12 infants. Acta Paediatr. 2006;95(1):38 – 43
pt 1):1539 –1543 151. Baier RJ, Loggins J, Kruger TE. Monocyte chemoattractant
132. Pianosi PT, Fisk M. High frequency ventilation trial: nine year protein-1 and interleukin-8 are increased in bronchopulmo-
follow up of lung function. Early Hum Dev. 2000;57(3): nary dysplasia: relation to isolation of Ureaplasma urealyticum.
225–234 J Investig Med. 2001;49(4):362–369
133. Filippone M, Sartor M, Zacchello F, Baraldi E. Flow limitation 152. Oei J, Lui K, Wang H, Henry R. Decreased interleukin-10 in
in infants with bronchopulmonary dysplasia and respiratory tracheal aspirates from preterm infants developing chronic
function at school age. Lancet. 2003;361(9359):753–754 lung disease. Acta Paediatr. 2002;91(11):1194 –1199
134. Bhandari A, Panitch HB. Pulmonary outcomes in broncho- 153. Kakkera DK, Siddiq MM, Parton LA. Interleukin-1 balance in
pulmonary dysplasia. Semin Perinatol. 2006;30(4):219 –226 the lungs of preterm infants who develop bronchopulmonary
135. Vrijlandt EJ, Gerritsen J, Boezen HM, Grevink RG, Duiver- dysplasia. Biol Neonate. 2005;87(2):82–90
man EJ. Lung function and exercise capacity in young adults 154. Danan C, Franco ML, Jarreau PH, et al. High concentrations
born prematurely. Am J Respir Crit Care Med. 2006;173(8): of keratinocyte growth factor in airways of premature infants
890 – 896 predicted absence of bronchopulmonary dysplasia. Am J Re-
136. Smith LJ, van Asperen PP, McKay KO, Selvadurai H, spir Crit Care Med. 2002;165(10):1384 –1387
Fitzgerald DA. Reduced exercise capacity in children born 155. Kevill KA, Bhandari V, Kettunen M, et al. A role for macro-
very preterm. Pediatrics. 2008;122(2). Available at: www. phage migration inhibitory factor in the neonatal respiratory
pediatrics.org/cgi/content/full/122/2/e287. Accessed April distress syndrome. J Immunol. 2008;180(1):601– 608
20, 2009 156. Kim BI, Lee HE, Choi CW, et al. Increase in cord blood soluble
137. Cutz E, Chiasson D. Chronic lung disease after premature E-selectin and tracheal aspirate neutrophils at birth and the
birth. N Engl J Med. 2008;358:743–745; author reply 745–746 development of new bronchopulmonary dysplasia. J Perinat
138. Wong PM, Lees AN, Louw J, et al. Emphysema in young adult Med. 2004;32(3):282–287
survivors of moderate-to-severe bronchopulmonary dyspla- 157. Cederqvist K, Sirén V, Petäjä J, Vaheri A, Haglund C, Anders-
sia. Eur Respir J. 2008;32(2):321–328 son S. High concentrations of plasminogen activator inhibi-
139. Anderson PJ, Doyle LW. Neurodevelopmental outcome of tor-1 in lungs of preterm infants with respiratory distress
bronchopulmonary dysplasia. Semin Perinatol. 2006;30(4): syndrome. Pediatrics. 2006;117(4):1226 –1234
227–232 158. Rehan VK, Torday JS. Lower parathyroid hormone-related
140. Short EJ, Klein NK, Lewis BA, et al. Cognitive and academic protein content of tracheal aspirates in very low birth weight
consequences of bronchopulmonary dysplasia and very low infants who develop bronchopulmonary dysplasia. Pediatr Res.
birth weight: 8-year-old outcomes. Pediatrics. 2003;112(5). 2006;60(2):216 –220

1572 BHANDARI, BHANDARI


Downloaded from www.pediatrics.org by on September 2, 2010
159. Cederqvist K, Haglund C, Heikkilä P, et al. Pulmonary tryp- preterm infants with (or developing) bronchopulmonary
sin-2 in the development of bronchopulmonary dysplasia in dysplasia/chronic lung disease. Cochrane Database Syst Rev.
preterm infants. Pediatrics. 2003;112(3 pt 1):570 –577 2006;(3):CD005093
160. Bhandari V, Choo-Wing R, Lee CG, et al. Developmental 175. Halliday HL, Ehrenkranz RA, Doyle LW. Delayed (⬎3 weeks)
regulation of NO-mediated VEGF-induced effects in the lung. postnatal corticosteroids for chronic lung disease in preterm
Am J Respir Cell Mol Biol. 2008;39(4):420 – 430 infants. Cochrane Database Syst Rev. 2003;(1):CD001145
161. Aghai ZH, Arevalo R, Lumicao L, et al. Basement membrane 176. Halliday HL, Ehrenkranz RA, Doyle LW. Moderately early
biomarkers in very low birth weight premature infants: asso- (7–14 days) postnatal corticosteroids for preventing chronic
ciation with length of NICU stay and bronchopulmonary lung disease in preterm infants. Cochrane Database Syst Rev.
dysplasia. Biol Neonate. 2002;81(1):16 –22 2003;(1):CD001144
162. Broström EB, Katz-Salamon M, Lundahl J, Halldén G, Win- 177. Halliday HL, Ehrenkranz RA, Doyle LW. Early postnatal (⬍96
bladh B. Eosinophil activation in preterm infants with lung hours) corticosteroids for preventing chronic lung disease in
disease. Acta Paediatr. 2007;96(1):23–28 preterm infants. Cochrane Database Syst Rev 2003;(1):
163. Ahola T, Fellman V, Kjellmer I, Raivio KO, Lapatto R. Plasma CD001146
8-isoprostane is increased in preterm infants who develop 178. Baveja R, Christou H. Pharmacological strategies in the pre-
bronchopulmonary dysplasia or periventricular leukomala- vention and management of bronchopulmonary dysplasia.
cia. Pediatr Res. 2004;56(1):88 –93 Semin Perinatol. 2006;30(4):209 –218
164. Ogihara T, Hirano K, Morinobu T, et al. Plasma KL-6 predicts 179. Khemani E, McElhinney DB, Rhein L, et al. Pulmonary artery
the development and outcome of bronchopulmonary dyspla- hypertension in formerly premature infants with broncho-
sia. Pediatr Res. 2006;60(5):613– 618 pulmonary dysplasia: clinical features and outcomes in the
165. Tsao PN, Wei SC, Su YN, et al. Placenta growth factor eleva- surfactant era. Pediatrics. 2007;120(6):1260 –1269
tion in the cord blood of premature neonates predicts poor 180. Bhandari A, Schramm CM, Kimble C, Pappagallo M, Hussain
pulmonary outcome. Pediatrics. 2004;113(5):1348 –1351 N. Effect of a short course of prednisolone in infants with
166. Ballabh P, Kumari J, Krauss AN, et al. Soluble E-selectin, oxygen-dependent bronchopulmonary dysplasia. Pediatrics.
soluble L-selectin and soluble ICAM-1 in bronchopulmonary 2008;121(2). Available at: www.pediatrics.org/cgi/content/
dysplasia, and changes with dexamethasone. Pediatrics. 2003; full/121/2/e344. Accessed April 20, 2009
111(3):461– 468 181. Ng GY, da S, Ohlsson A. Bronchodilators for the prevention
167. Yost CC, Soll RF. Early versus delayed selective surfactant and treatment of chronic lung disease in preterm infants.
treatment for neonatal respiratory distress syndrome. Co- Cochrane Database Syst Rev 2001;(3):CD003214
chrane Database Syst Rev. 2000;(2):CD001456 182. Brundage KL, Mohsini KG, Froese AB, Fisher JT. Bronchodi-
168. Kamlin CO, Davis PG. Long versus short inspiratory times in lator response to ipratropium bromide in infants with bron-
neonates receiving mechanical ventilation. Cochrane Database chopulmonary dysplasia. Am Rev Respir Dis. 1990;142(5):
Syst Rev. 2004;(4):CD004503 1137–1142
169. Miller JD, Carlo WA. Safety and effectiveness of permissive 183. Bhandari V, Elias JA. Cytokines in tolerance to hyperoxia-
hypercapnia in the preterm infant. Curr Opin Pediatr. 2007; induced injury in the developing and adult lung. Free Radic
19(2):142–144 Biol Med. 2006;41(1):4 –18
170. Henderson-Smart DJ, Davis PG. Prophylactic methylxan- 184. Cole CH, Wright KW, Tarnow-Mordi W, Phelps DL; Pulse
thines for extubation in preterm infants. Cochrane Database Oximetry Saturation Trial for Prevention of Retinopathy of
Syst Rev. 2003;(1):CD000139 Prematurity Planning Study Group. Resolving our uncer-
171. Tyson JE, Wright LL, Oh W, et al. Vitamin A supplementation tainty about oxygen therapy. Pediatrics. 2003;112(6 pt 1):
for extremely-low-birth-weight infants. National Institute of 1415–1419
Child Health and Human Development Neonatal Research 185. Tin W, Wiswell TE. Adjunctive therapies in chronic lung
Network. N Engl J Med. 1999;340(25):1962–1968 disease: examining the evidence. Semin Fetal Neonatal Med.
172. Darlow BA, Graham PJ. Vitamin A supplementation to pre- 2008;13(1):44 –52
vent mortality and short and long-term morbidity in very low 186. Doyle LW, Davis PG, Morley CJ, McPhee A, Carlin JB; DART
birthweight infants. Cochrane Database Syst Rev 2007;(4): Study Investigators. Outcome at 2 years of age of infants from
CD000501 the DART study: a multicenter, international, randomized,
173. Oh W, Poindexter BB, Perritt R, et al. Association between controlled trial of low-dose dexamethasone. Pediatrics. 2007;
fluid intake and weight loss during the first ten days of life 119(4):716 –721
and risk of bronchopulmonary dysplasia in extremely low 187. Thomas W, Speer CP. Nonventilatory strategies for preven-
birth weight infants. J Pediatr. 2005;147(6):786 –790 tion and treatment of bronchopulmonary dysplasia: what is
174. Lai NM, Rajadurai SV, Tan KH. Increased energy intake for the evidence? Neonatology. 2008;94(3):150 –159

PEDIATRICS Volume 123, Number 6, June 2009 1573


Downloaded from www.pediatrics.org by on September 2, 2010
Pitfalls, Problems, and Progress in Bronchopulmonary Dysplasia
Anita Bhandari and Vineet Bhandari
Pediatrics 2009;123;1562-1573
DOI: 10.1542/peds.2008-1962
Updated Information including high-resolution figures, can be found at:
& Services http://www.pediatrics.org/cgi/content/full/123/6/1562
References This article cites 169 articles, 78 of which you can access for
free at:
http://www.pediatrics.org/cgi/content/full/123/6/1562#BIBL
Citations This article has been cited by 1 HighWire-hosted articles:
http://www.pediatrics.org/cgi/content/full/123/6/1562#otherartic
les
Subspecialty Collections This article, along with others on similar topics, appears in the
following collection(s):
Neurology & Psychiatry
http://www.pediatrics.org/cgi/collection/neurology_and_psychia
try
Permissions & Licensing Information about reproducing this article in parts (figures,
tables) or in its entirety can be found online at:
http://www.pediatrics.org/misc/Permissions.shtml
Reprints Information about ordering reprints can be found online:
http://www.pediatrics.org/misc/reprints.shtml

Downloaded from www.pediatrics.org by on September 2, 2010

You might also like