+ All Categories
Home > Documents > 06-Seminars in Fetal development

06-Seminars in Fetal development

Date post: 04-Jun-2018
Category:
Upload: ganesh-subramanian-mohan
View: 215 times
Download: 0 times
Share this document with a friend

of 75

Transcript
  • 8/13/2019 06-Seminars in Fetal development

    1/75

    Seminars in Fetal &Neonatal MedicineAmsterdam Boston London New York Oxford Paris Philadelphia San Diego St. Louis

    Aims and ScopeSeminars in Fetal & Neonatal Medicine (formerly Seminars in Neonatology) is a

    bi-monthly journal which publishes topic-based issues, including current Hot Topics on the

    latest advances in fetal and neonatal medicine. The change in title relates to the growing

    interest amongst obstetricians, midwives and fetal medicine specialists.

    The Journal commissions review-based content covering current clinical opinion on the

    care and treatment of the neonate and draws on the necessary specialist knowledge,

    including that of the respiratory physician, the infectious disease physician, the surgeon,as well as the paediatrician and obstetrician.

    Each topic-based issue is edited by an authority in their field and contains 810 articles.

    Current and forthcoming events can be viewed on the Internet at:

    http://www.elsevier.com/locate/siny

    Seminars in Fetal & Neonatal Medicine provides:

    coverage of major developments in neonatal care;

    value to practising neonatologists, consultant and trainee paediatricians,

    obstetricians, midwives and fetal medicine specialists wishing to extend their

    knowledge in this field;

    up-to-date information in an attractive and relevant format.

    Editorial BoardEditor-in-Chief

    Professor M I LeveneUniversity of Leeds

    School of Medicine

    D Floor, Clarendon Wing

    The General Infirmary at Leeds

    Leeds LS2 9NS, UK

    Associate EditorsM Blennow, Huddinge, Sweden K Marsa l, Lund, SwedenL Cornette, Brugge, Belgium D Peebles, London, UKD J Field, Leicester, UK S Sinha, Middlesbrough, UKI Laing, Edinburgh, UK A M Weindling, Liverpool, UK

    Advisory Board

    F A Chervenak, USA P C NG, Hong KongS M Donn, USA J M Perlman, USAN Evans, Australia E Saliba, FranceV Fellman, Sweden M Vento, AlicanteN N Finer, USA L de Vries, The Netherlands

  • 8/13/2019 06-Seminars in Fetal development

    2/75

    Editorial

    Prologue: Advances in Bronchopulmonary Dysplasia

    It has been 42 years since our first published report of broncho-

    pulmonary dysplasia (BPD)1; it is still a problem for premature

    infants. The original goal of using mechanical ventilation to treat

    premature infants with respiratory distress syndrome and respira-

    tory failure was to decrease the significant mortality. During the

    ensuing decades, a decrease in mortality has indeed occurred.Once recognized, it was hoped that a reduction of supplemental

    oxygen concentrations and ventilatory pressure would eliminate

    or decrease the incidence of BPD. This has, for the most part,

    been achieved in the 33 week gestational age infants originally

    described. Advances in neonatal care and respiratory therapy since

    1967 have resulted in the successful ventilation of increasingly

    more immature infants. As a result the original radiographic picture

    and pathology have been modified.

    The understanding of the growth and development of the

    extremely premature lung and the genetic, intrauterine, biochem-

    ical, and infectious factors that influence the susceptibility and

    severity of BPD has also advanced. This has led to improvements

    in prevention strategies and both ventilatory and non-ventilatory

    treatment of BPD. Unfortunately, it has not eliminated BPD or led

    to a more precise diagnosis. The chest radiographic changes are

    much more subtle and are seldom used in the diagnosis. Infants

    are now delivered while their lungs are in the late canalicular to

    early saccular stage of development. It has yet to be determined

    whether there is an oxygen concentration or ventilator pressure

    below which these treatments are non-injurious. Prevention of

    premature delivery remains an elusive and persistent problem.

    The long-term pulmonary function of premature infants with

    or without BPD surviving beyond adolescence to older adulthood

    is also unknown. Whereas some of the original premature

    infants with BPD displayed persistent pulmonary dysfunction as

    adolescents, it is not known if these changes regressed, persisted,

    or exacerbated in older adulthood. These original infants were

    less premature than todays neonatal intensive care unit popula-

    tion. They also were treated with higher concentrations of supple-

    mental oxygen, and higher ventilator pressures than are currentlyused, and prior to the widespread use of antenatal corticosteroids

    and surfactant therapy. The persistence of BPD in more immature

    premature infants indicates the need for long-term clinical and

    pulmonary function testing. Physicians who deal with these

    patients as adults will need to be aware of their possible

    late pulmonary disability. Knowledge of the evolving pulmonary

    function of infants born very prematurely might best be obtained

    if pediatric and adult oriented pulmonologists combine their

    interest and talents.

    We should not forget, however, that the continuing morbidity

    from BPD is the result of the successful application of modern

    neonatal intensive care to very premature infants to improve their

    survival.

    Reference

    1. Northway Jr WH, Rosan RC, Porter DY. Pulmonary disease following respiratortherapy of hyaline-membrane disease. Bronchopulmonary dysplasia. N Engl JMed1967;276:35768.

    William H. Northway Jr.

    Stanford University, Lucile Packard Childrens Hospital, Palo Alto,

    California, USA

    E-mail address: [email protected]

    Contents lists available atScienceDirect

    Seminars in Fetal & Neonatal Medicine

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / s i n y

    1744-165X/$ see front matter 2009 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.siny.2009.08.008

    Seminars in Fetal & Neonatal Medicine 14 (2009) 331

    mailto:[email protected]://www.sciencedirect.com/science/journal/1744165Xhttp://www.elsevier.com/locate/sinyhttp://www.elsevier.com/locate/sinyhttp://www.sciencedirect.com/science/journal/1744165Xmailto:[email protected]
  • 8/13/2019 06-Seminars in Fetal development

    3/75

    Editorial

    Despite the myriad of advances in neonatal intensive care in the

    more than 40 years since Prof. Northway and colleagues first coined

    the term bronchopulmonary dysplasia to describe the aftermath

    of neonatal mechanical ventilation, the incidence of chronic lung

    disease has not appreciably changed. Approximately 3040% of

    infants weighing less than 1500 g at birth sustain chronic lung

    disease, evenwith the advent of antenatal corticosteroid treatment,surfactant replacement therapy, and sophisticated techniques for

    both non-invasive and invasive mechanical ventilation. What has

    changed, however, is the demographic composition of affected

    infants, many of whom received only mild or modest respiratory

    support, suggesting that chronic lung disease may now reflect an

    alteration in lung development and growth. This issue ofSeminars

    in Fetal & Neonatal Medicine examines recent advances in the

    understanding and management of bronchopulmonary dysplasia.

    We are most grateful to our distinguished group of contribu-

    tors, headed by Professor Northway, himself, who kindly wrote

    the Prologue for this issue. Our first article was written by

    Professor Philip, who puts another historical context to chronic

    lung disease. His interest in the subject dates back to his seminal

    paper published in 1975. Next, Professor Greenough presentsa novel perspective on the potential prenatal factors which might

    predispose an infant to the development of chronic lung disease.

    Professor Merritt and colleagues provide an interesting commen-

    tary on whether the new BPD is actually different from the

    old BPD and examine the challenges that are ahead. Dr. Van

    Marter subsequently re-examines the epidemiology of chronic

    lung disease and evaluates its multifactorial etiology. Dr. Laughon

    and colleagues critically evaluate the evidence for preventive

    strategies. Our own chapter on ventilation follows, where we

    summarize both lung protective strategies and ventilatory

    management of BPD. Professor Wiswell and Dr. Tin examine in

    detail many of the medical myths that surround BPD and presentthe evidence to date. Prof. Chiswick has written a thoughtful and

    provocative piece on the ethics of managing end-stage BPD.

    Finally, Professor Doyle and colleagues examine the long term

    outcomes of affected infants.

    We hope that the readers will realise that BPD is a multi-faceted

    disorder and not merely the effect of ventilator-induced lung injury.

    The genetic, cellular, developmental, and nutritional milieu of the

    fetus and newborn are all contributing factors and deserve to be

    thoroughly investigated. If this issue is a stimulus for such, it will

    be an unqualified success.

    Steven M. Donn*

    C S Mott Childrens Hospital, Div. of Neonatal-Perinatal Medicine,

    Dept. of Pediatrics, Ann Arbor, Michigan, USA Corresponding author.

    E-mail address: [email protected](S.M. Donn)

    Sunil K. Sinha

    Middlesbrough, UK

    Contents lists available atScienceDirect

    Seminars in Fetal & Neonatal Medicine

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / s i n y

    1744-165X/$ see front matter 2009 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.siny.2009.08.006

    Seminars in Fetal & Neonatal Medicine 14 (2009) 332

    mailto:[email protected]://www.sciencedirect.com/science/journal/1744165Xhttp://www.elsevier.com/locate/sinyhttp://www.elsevier.com/locate/sinyhttp://www.sciencedirect.com/science/journal/1744165Xmailto:[email protected]
  • 8/13/2019 06-Seminars in Fetal development

    4/75

    Chronic lung disease of prematurity: A short history

    Alistair G.S. Philip

    Stanford University School of Medicine, Division of Neonatal and Developmental Medicine, Suite 315, 750 Welch Road, Palo Alto, CA 94304, USA

    Keywords:

    Bronchopulmonary dysplasia

    Chronic lung disease

    Genetic predisposition

    Prematurity

    WilsonMikity syndrome

    s u m m a r y

    Chronic lung disease of prematurity (CLD) is commonly considered to be a consequence of assisted

    ventilation. However, prior to the description in 1967 of bronchopulmonary dysplasia (BPD), following

    ventilator therapy for respiratory distress syndrome, WilsonMikity syndrome (WMS) had beendescribed in very preterm infants on minimal oxygen supplementation. In the 1970s and 1980s, many

    infants treated with assisted ventilation required prolonged mechanical ventilation after developing

    radiographic features of coarse infiltrates, severe hyperinflation, and microcystic changes, associated

    with hypercarbemia and the need for increased inspired oxygen concentrations. Some infants died and

    showed evidence of pulmonary fibrosis, obstructive bronchiolitis, and dysplastic change. The role of

    supplemental oxygen, positive pressure ventilation, and the immaturity of the lung have long been

    considered important in the etiology of CLD/BPD. More recently, the role of inflammation (particularly

    antenatal exposure to cytokines) and individual susceptibility (genetic predisposition) have assumed

    greater etiologic importance. The historical setting into which corticosteroid treatment for BPD was

    introduced is also discussed. After the licensing of exogenous surfactant to treat RDS in the early 1990s

    and more widespread use of prenatal corticosteroids in the mid-1990s, severe BPD became an unusual

    event. Gradually, the diagnosis of CLD, still often referred to as BPD, was based on an oxygen requirement

    at 36 weeks postmenstrual age. However, it is not clear that this new BPD is substantially different from

    WMS. It is difficult to make prognostications about long-term lung function of these infants based on

    oxygen requirement at 36 weeks, since supplemental oxygen is frequently used unnecessarily.

    2009 Elsevier Ltd. All rights reserved.

    1. Introduction

    Prior to the introduction of assisted (mechanical) ventilation,

    comparatively few very low birth weight (VLBW) infants (

  • 8/13/2019 06-Seminars in Fetal development

    5/75

    same medical center in 1969,6 and many other cases were reported

    from around the world.7 WMS was observed in very preterm

    infants approximately two weeks after birth. For the most part,

    these were infants who initially had little or no oxygen require-

    ment, but later developed the need for increasing supplemental

    oxygen concentrations of up to 40% to prevent cyanosis. Radio-

    graphically, microcystic changes, with some degree of hyperinfla-

    tion and flattening of the diaphragms were seen. Some infants

    recovered spontaneously over weeks to months, but others died

    and demonstrated hyper-aeration and reduced alveolar septa at

    postmortem examination. A few had evidence of pulmonary

    fibrosis and obstructive bronchiolitis.

    3. Oxygen toxicity in adults, animal models and in vitro

    The terms pulmonary oxygen toxicity or pulmonary poisoning

    were used at about this time, both in animals and in humans.8

    Pathologic findings were described in adult humans treated with

    oxygen and artificial (mechanical) ventilation.9 The additive roles of

    oxygen and assisted ventilation were noted in lambs10 and

    pulmonary oxygen toxicity was reported in monkeys.11 Somewhat

    later, findings similar to those in WMS were noted when lunghistology was evaluated in extremely premature baboons exposed

    to carefully controlled assisted ventilation and oxygen adminis-

    tration.12 Loss of muco-ciliary function was also noted when

    cultured human neonatal respiratory epithelium was exposed to

    80% oxygen for 4896 h in vitro.13

    4. First description of bronchopulmonary dysplasia

    Soon after the introduction of mechanical ventilation to manage

    RDS in the mid-1960s, reports began to appear of radiographic and

    pathologic abnormalities that seemed to result from exposure to

    high concentrations of oxygen using mechanical ventilation. The

    first description of bronchopulmonary dysplasia (BPD) is generally

    attributed to Northway et al. in 1967.14

    They used the term bron-chopulmonary dysplasia to describe findings of pulmonary

    disease following respirator therapy of hyaline membrane disease.

    However, almost identical findingswere described in the same year

    by Hawker et al. from London, noting infants who died after

    respirator treatment for severe hyaline membrane disease.15

    Northway et al. believed that the critical factor appeared to be

    exposure to an inspired oxygen concentration>80% for longer than

    150 h.14 Hawker et al. linked the lung findings to >60% oxygen for

    more than 5 days (120 h).15 In a review of pulmonary oxygen

    toxicity in 1969, Nelson stated that there seemed to be general

    agreement that oxygen partial pressures of less than 0.6

    atmospheres are not toxic, over any time span.16 On the other

    hand, Pusey et al. (also in 1969) suggested that neither HMD nor

    concentrations of oxygen >80% were needed to produce

    pulmonary fibroplasia, the key feature of BPD, and that assisted

    ventilation was more important.17

    A comparison of the principal features of BPD (in the late 1960s)

    and WMS is provided inTable 1.

    5. Negative pressure respirators (ventilators)

    Although the introduction of mechanical ventilation in the

    1960s was predominantly concerned with positive pressure

    machines, some investigators, including Stahlman and Stern,

    believed that there might be advantages in using negative pressure

    devices. They reported that it was unusual to observe pulmonary

    oxygen toxicity when negative pressure respirators were used,

    even with prolonged exposure to high oxygen concentrations.

    18

    This apparent advantage of the negative pressure respirator didnot gain general acceptance. The negative pressure device available

    at the time looked a little like a modified incubator, based on the

    iron lung devices used to manage severe cases of poliomyelitis in

    older children and adults (Fig.1). Themost difficult part about using

    the machines was to createan adequate seal at the neck to generate

    adequate negative pressure around the baby, without disrupting

    venous return from the brain. It was also difficult to use in infants

  • 8/13/2019 06-Seminars in Fetal development

    6/75

    oxygen toxicity, we attempted to minimize exposure of preterm

    infants to high oxygen concentrations. Nevertheless, we encoun-

    tered many infants treated with positive pressure who developed

    radiographic features of BPD. It seemed that the problem could not

    simply be duration of exposure to high oxygen concentrations. Two

    other factors seemed to be important: the immaturity of the lung,

    and the role of the endotracheal tube. Since negative pressure

    ventilation was rarely associated with chronic lung disease, it

    seemed possible that the endotracheal tube allowed pressure to be

    applied more directly to the immature lung (later referred to as

    barotrauma). In a series of ten infants who developed BPD that I

    reported in 1975,19 only two required >80% oxygen for more than

    24 h, and only one required 60% oxygen for more than 100 h. I

    suggested that the etiology of BPD might be a complicated

    interaction between oxygen plus pressure plus time and that

    immature lungs when exposed to oxygen concentrations>40% for

    as little as three days, via positive pressure ventilation, might

    develop BPD. It also seemedpossible that WMSmight form one end

    of a spectrum, where the very immature lung might react adversely

    to even minor increases in oxygen exposure (since they should not

    have been exposed to any). I also suggested that there might be

    individual susceptibility.19 Much evidence now supports these

    hypotheses.

    7. National Institutes of Health (NIH) consensus conference

    In the late 1970s, there was increasing attention paid to the

    development of lung injury in preterm infants, most of whom had

    received assisted ventilation. The NIH convened a consensus

    conference in late 1978. The results were published in 1979.20

    Northway was the opening speaker. Some of those in attendance

    were inclined to use the term chronic lung disease of prematurity

    (CLD), but the majority favored the term bronchopulmonary

    dysplasia (BPD). This seemed somewhat unusual to me, since the

    term RDS (a clinical definition) had been favored over HMD (a

    pathological definition) at an earlier consensus conference. BPD,

    like HMD, suggests knowledge of histopathology, whereas CLDexpresses what is happening clinically.

    There was considerable discussion of possible etiologic factors,

    the clinical and radiographic findings and diagnostic criteria.20 Key

    features of BPD/CLD were prolonged oxygen dependency and

    associated radiographic features of hyperinflation, usually

    accompanied by increased pCO2. Not infrequently, in the 1970s and

    1980s, prolonged assisted ventilation was required and hospital

    stays of many months were common. Severe cases of BPD

    frequently resulted in death from cor pulmonale.

    8. Oxygen radical disease

    Although the etiology of BPD is likely multifactorial, the concept

    of pulmonary oxygen toxicity received additional support in the1980s. In a series of papers, Saugstad promoted the concept that

    oxygen free radicals could result in pulmonary damage.21,22 He had

    earlier documented the importance of hypoxanthine and xanthine

    oxidase in asphyxial insult.23 He extended these observations and

    proposed that several problems frequently encountered in very

    preterm infants might be the result of a common pathway. He

    proposed that BPD, retinopathy of prematurity (ROP), necrotizing

    enterocolitis (NEC), patent ductus arteriosus (PDA), periventricular

    leukomalacia (PVL), and possibly intraventricular hemorrhage

    (IVH) might all be part of an entity he called oxygen radical disease

    of the newborn.22 The initial proposal was that a burst of oxygen

    free radicals was generated in the re-oxygenation phase following

    an acute hypoxic insult, which could cause widespread injury. This

    idea has continued to be supported, rather than refuted, in recent

    years. Attempts arenow made to minimize the exposureof preterm

    infants to high oxygen saturations.

    9. Chorioamnionitis, cytokines and chemokines

    Before considering the role that exogenous surfactant has

    played in decreasing the severity of BPD, other etiologic factors in

    the production of BPD/CLD emerged in the late 1970s and 1980s.

    Excessive fluid administration was implicated, although this may

    have been because it resulted in exposure to higher oxygen

    concentrations.24 Cytomegalovirus (CMV) infection was also

    implicated in late-onset chronic lung disease25 in an era before

    CMV screening of blood for transfusion became routine.

    The possibility that chorioamnionitis might predispose to BPD

    seemed unlikely when it was first reported, but the role of

    inflammation gained increasing acceptance as a variety of

    cytokines and chemokines were reported to be circulating

    following chorioamnionitis. One of the first reports came from

    Japan in 1983.26 Elevated levels of immunoglobulin M (IgM) were

    noted in low birth weight infants with chronic respiratory insuffi-

    ciency. Subsequently, the same group reported higher IgM levels in

    infants evaluated within 72 h of birth who went on to develop

    WMS.27 Somewhat surprisingly, much lower levels were seen inthose who developed BPD or unexplained chronic lung disease.

    Over the next decade, lung inflammation was increasingly

    recognized, and the role of cytokines was reported in evolving

    BPD.28,29 By the turn of the millennium, inflammation was firmly

    established in the etiology of BPD30,31 and currently seems to be

    even more entrenched as an antenatal precursor to lung injury and

    the development of BPD.32,33

    10. Status of BPD in the 1980s

    During the 1970s, neonatologists became more adept at

    managing mechanical ventilation, and survival of VLBW infants

    improved. This improvement was partly the result of the learning

    curve as increasing numbers of people learned to manage VLBWinfants successfully. This was, in part, attributed to the develop-

    ment of ventilators specifically designed to be used in infants, and

    partly from other improvements (e.g. parenteral nutrition). The

    possibility that undernutrition was a contributing factor in the

    pathogenesis of BPD was proposed.34

    In 1980, the first report of successful treatment of RDS with

    exogenous surfactant came from Fujiwara et al. in Japan.35 Within

    a few years, there was an explosion of interest in exogenous

    surfactants, which were evaluated at the end of the decade in

    multiple randomized controlled studies around the world, with

    spectacular success. By 1991, both Exosurf (colfosceril, a synthetic

    surfactant) and Survanta (beractant, a bovine-extract surfactant)

    had been approved by the Federal Drug Administration. Although

    the introduction of exogenous surfactant did not eliminate thedevelopment of BPD, it substantially altered its severity, so that by

    1999 Jobe referred to chronic lung disease of preterm infants

    observed at that time as the new BPD.36

    During the 1980s, our ability to keep babies alive with

    successful mechanical ventilation came at considerable expense

    (Fig. 2). This was an era of chronic ventilator dependence, with

    some infants being ventilated for several months. Whereas many

    were eventually ableto be weaned, some were pulmonary cripples

    and others died of cor pulmonale after several months in the

    neonatal intensive care unit (NICU). We tried to remain optimistic

    about the future for these preterm infants by reminding ourselves

    (and parents) that lung growth continues for many months after

    birth (for as long as three years), and we tried to minimize the

    duration of exposure to ventilation as much as possible.

    A.G.S. Philip / Seminars in Fetal & Neonatal Medicine 14 (2009) 333338 335

  • 8/13/2019 06-Seminars in Fetal development

    7/75

    11. Prevention and therapeutic strategies

    Prior to surfactant therapy, therewere few prevention strategies.Antioxidants were promoted, but initial enthusiasm for vitamin E

    and superoxide dismutase were tempered by negative results. 37,38

    On the other hand, vitamin A was more convincing 39 and seems to

    have a place in standard care of most VLBW infants. More recently,

    a large, prospective, randomized study of caffeine to prevent apnea

    documented a significantreduction in CLD/BPD.40 In 1972,reduction

    in the incidence and severity of RDS was reported by Liggins and

    Howie using antenatal betamethasone.41 Although used by many,

    widespread acceptance of this strategy did not occur for more than

    20 years.42 However, maternal antenatal glucocorticoid adminis-

    tration was shown to reduce the risk of BPD in 1990.43

    Since cor pulmonale was frequently observed, diuretics and

    digoxin were often used, but the most impressive improvement was

    noted withpostnatal corticosteroids(see below).In order todecreasehospital stay, some infants were discharged home on nasal cannula

    oxygen (Fig. 3). Initially, this was facilitated by transcutaneous

    oxygen monitoring44 and later by pulse oximetry.45 Another

    suggestion was to use early continuous positive airway pressure, as

    the incidence of CLD was much lower in one center adopting this

    strategy compared with seven that did not.46 This approach has

    gained increased support in recent years, although it may be difficult

    to separate it from the effects of early surfactant administration.47

    12. Introduction of corticosteroids for BPD

    There has been increasing concern that using corticosteroids to

    prevent BPD may be creating other problems, particularly

    neurodevelopmental delay.48 It seems important to establish the

    circumstances into which this treatment was introduced. Some

    infants who developed CLD/BPD required assisted ventilation for

    months. Against this backdrop, it is a little easier to understand why

    steroid use had considerable appeal. Steroids certainly seemed to

    shorten the duration of mechanical ventilation and the link

    between mechanical ventilation and CLD/BPD appeared to be

    well-established.

    My own experience may have contributed to more widespread

    use of corticosteroids. In 1974, I presented a paper at the Western

    Society for Pediatric Research documenting pulmonary improve-

    ment and rapid weaning from assisted ventilation in ten infants

    with severe BPD.49 In the first two patients, steroids had been used

    for other reasons (cerebral edema in one and laryngeal edema at

    attempted extubation in the other), with serendipitous pulmonary

    improvement. Although I prepared a manuscript describing the

    results, it was not published because reviewers were concernedabout possible long-term consequences, based on animal data. This

    was not something to which I had given much thought. My use of

    steroids for CLD/BPD was subsequently limited to late treatment in

    severe cases with prolonged requirements of assisted ventilation.

    In the ensuing years, there was considerable discussion. We all

    wrestled with how to minimize the long-term effects of assisted

    ventilation. The first randomized controlled study of corticosteroids

    for BPD was published in 1983 by Mammel et al.50 and a second

    study appeared in 1985.51 Protocols for both of these studies had

    comparatively short courses of steroids.

    The most influential study appeared in 1989, with earlier

    administration and a prolonged weaning course.52 After this,

    corticosteroid use became more popular, until concerns about long-

    term sequelae were expressed.48 Exogenous surfactant was beingevaluated at about the same time that this report appeared. 52 Any

    decrease in severity of CLD/BPD may have been attributed, by some,

    more to steroids than to surfactant, because there was a tendency

    to use steroids earlier than before. Even when surfactant was being

    used more routinely, it was still difficult to wean VLBW infants from

    assisted ventilation and even short courses of steroids (seven days)

    seemed to be helpful in facilitating extubation and decreasing the

    duration of mechanical ventilation and the incidence of CLD.53

    13. Surfactant protein deficiency states

    One group of infants who developed CLD that was difficult to

    understand was term infants who appeared to have RDS.

    Fig. 2. An example of the chest radiograph findings of bronchopulmonary dysplasia

    commonly observed in the 1970s and 1980s, demonstrating coarse infiltrates, hyper-

    inflation and microcystic changes. (Courtesy of Steven M. Donn, MD.).

    Fig. 3. An infant on nasal cannula oxygen supplementation being assessed as an

    outpatient with a transcutaneous oxygen monitor placed on the back.

    A.G.S. Philip / Seminars in Fetal & Neonatal Medicine 14 (2009) 333338336

  • 8/13/2019 06-Seminars in Fetal development

    8/75

    Frequently, it was believed that mistakes had been made in deter-

    mining gestational age. In recent years, surfactant proteins have

    been categorized, and many of these infants probably had a specific

    surfactant protein (SP) deficiency, usually SP-B or SP-C.54 There is

    also evidence that there are associations between genetic variants

    of surfactant proteins and BPD.55

    14. Individual susceptibility

    While debate continued about the relative importance of

    oxygen or assisted ventilation in the etiology of CLD/BPD, 20 at or

    soon after birth, it was difficult to predict which infants would go

    on to develop CLD/BPD. Thus, it seemed possible that there might

    be some factor or factors that contributed to individual suscepti-

    bility. One of the first associations, reported in 1982, was between

    CLD and HLA (human lymphocyte antigen) phenotypes.56 In the

    ensuing 25 years, the idea that there may be a genetic predispo-

    sition to develop CLD/BPD has become generally accepted.57,58

    Further details can be found in Chapter 3 of this issue.

    15. The new BPD

    With the widespread use of surfactant, in conjunction withgreater use of antenatal betamethasone, following another NIH

    consensus development conference in 1994,42 the outcome for

    most preterm infants was altered substantially. Not only did

    survival improve for each gestational age, but the severity of

    residual lung damage also decreased.59 Today, we almost never

    encounter the severe hyperinflation of the lungs with flattened

    diaphragms and associated severe hypercarbemia seen in the 1970s

    and 1980s. The concern now is whether or not an infant with

    continuing oxygen requirement does or does not have CLD/BPD.

    In many NICUs, the nursing staff became very reliant on pulse

    oximetery monitors, introduced in the mid to late 1980s.60 They

    would frequently adjust inspired oxygen concentration to maintain

    SaO2at 98 to 100%, in order to decrease the amount of apnea and

    reduce the frequency of alarms. However, early supplementaloxygen may have contributed to the development of chronic lung

    disease because of oxygen free radicals. Recent years have seen

    changes in delivery room and NICU management to limit exposure

    to supplemental oxygen.61 There has been a move away from using

    100%oxygenas the standard method of delivery roomresuscitation,

    since many infants respond to room air and may even do better.

    Oxygen blenders have become more common in delivery rooms.

    The radiographic features of old BPD are nowseen infrequently

    and rarely do babies die of BPD, so that the pathologic features are

    almost never seen. Jobe coined the term the new BPD in 1999. 36 A

    second National Institute of Child Health and Human Development

    (NICHD) consensus conference followed.62 The new definition of

    BPD became a supplemental oxygen need at 36 weeks post-

    menstrual age. However, such an oxygen requirement may not bereal. The NICHD neonatal network centers demonstrated that many

    babies who required oxygen, according to the nursing staff, were

    able to maintain SaO2> 90% on room air.63 Of 1598 infants with

    birth weights 5011249 g, 560 (35%) had clinical BPD (oxygen use

    at 36 weeks), but only 398 (25%) had physiologic BPD (SaO 2< 90%

    in room air).63

    16. Final thoughts

    The late Joan Hodgman (who collaborated with Mikity in the

    1960s) questioned whether or not the new BPD was indeed

    another name for WMS.7 Even more recently, Lefkowitz and

    Rosenberg pointed out that it is now unfeasible to directly examine

    the relationship between the histopathologic disease and its

    long-term pulmonary outcome.64 It might be possible if one were

    willing to perform lung biopsies on these infants (and some were

    done in the past), but few would recommend this now. As they

    state, The term bronchopulmonary dysplasia (BPD) is an overused

    catchall for all aspects of chronic lung disease in the neonatal

    population.64 They noted that the main reason for labeling an

    infant as having BPD was to predict long-term pulmonary outcome,

    but also observed that Shennan et al.65 had suggested that many

    infants with a functional abnormality as neonates had normal

    long-term outcomes. They further added . it is important to

    recognize that from Shennan et al. forward, the clinical character-

    istic of some degree of supplemental oxygen use at 36 weeks

    post-menstrual age was considered simply a possible screening test

    for abnormal long-term pulmonary outcome, not a marker of

    a particular histopathology.64

    Because neonatology has changed substantially in the past 40

    years, CLD may be one entity (a clinical description) and BPD

    another (a histopathologic description). It still seems probable that

    WMS and BPD are parts of a continuum of CLD of prematurity.

    Conflict of interest statement

    None declared.Funding sources

    None.

    References

    1. Behrman RE, Babson GS, Lassel R. Fetal and neonatal mortality in white middleclass infants: mortality risks by gestational age and weight. Am J Dis Child1971;121:4869.

    2. Silverman WA, Fertig JW, Berger AP. The influence of the thermal environmentupon the survival of newly born premature infants. Pediatrics1958;22:87686.

    3. Usher R. Reduction of mortality from respiratory distress syndrome ofprematurity with early administration of intravenous glucose and sodiumbicarbonate. Pediatrics 1963;32:96675.

    4. Wilson MJ, Mikity VG. A new form of respiratory disease in premature infants.Am J Dis Child1960;99:48999.

    5. Sykes MK. Intermittent positive pressure respiration in tetanus neonatorum.Anesthesia1960, Oct;15:40110.

    6. Hodgman JE, Mikity VG, Tatter D, Cleland RS. Chronic respiratory distress in thepremature infant: WilsonMikity syndrome.Pediatrics 1969;44:17995.

    7. Hodgman JE. Relationship between WilsonMikity syndrome and the newbronchopulmonary dysplasia.Pediatrics 2003;112:14145.

    8. Giamonna ST, Kerner D, Bondurant S. Effect of oxygen breathing at atmosphericpressure on pulmonary surfactant. J Appl Physiol 1965;20:8558.

    9. Nash G, Blennerhassett JB, Pontoppidan H. Pulmonary lesions associated withoxygen therapy and artificial ventilation. N Eng J Med 1967;276:36874.

    10. de Lemos R, Wolfsdorf J, Nachman R, et al. Lung injury from oxygen in lambs:the role of artificial ventilation. Anesthesiology 1969;30:60918.

    11. Kaplan HP, Robinson FR, Kapanci Y, Weibel ER. Pathogenesis and reversibility ofthe pulmonary lesions of oxygen toxicity in monkeys. 1: clinical and lightmicroscopic studies.Lab Invest1969;20:94100.

    12. Coalson JJ, Winter VT, Siler-Khodr T, Yoder BA. Neonatal chronic lung disease inextremely immature baboons. Am J Respir Crit Care Med 1999;160:133346.

    13. Boat TF, Kleinerman JI, Fanaroff AA, Matthews LW. Toxic effects of oxygen oncultured human neonatal respiratory epithelium. Pediatr Res 1973;7:60715.

    14. Northway Jr WH, Rosan RC, Porter DY. Pulmonary disease following respiratortherapy of hyaline membrane disease: bronchopulmonary dysplasia. N Eng

    J Med1967;276:35768.15. Hawker JM, Reynolds EOR, Taghizadeh A. Pulmonary surface tension and

    pathological changes in infants dying after respirator treatment for severehyaline membrane disease.Lancet1967;ii:757.

    16. Nelson NM. Chronology, morphology and physiology of pulmonary oxygentoxicity. In: Lucey JF, editor. Problems of neonatal intensive care units. Report ofthe 59th Ross Conference on Pediatric Research. Columbus, Ohio: Ross Labora-tories; 1969. p. 44.

    17. Pusey VA, MacPherson RI, Chernick V. Pulmonary fibroplasia followingprolonged artificial ventilation of newborn infants. Can Med Assoc J 1969;100:4517.

    18. Swyer PR. Symposium on artificial ventilation. Summary of conferenceproceedings. Biol Neonate 1970;16:1915.

    19. Philip AGS. Oxygen plus pressure plus time: the etiology of bronchopulmonarydysplasia.Pediatrics1975;55:4550.

    20. Workshop on bronchopulmonary dysplasia. J Pediatr 1979;95(5, part 2):

    815920.

    A.G.S. Philip / Seminars in Fetal & Neonatal Medicine 14 (2009) 333338 337

  • 8/13/2019 06-Seminars in Fetal development

    9/75

    21. Saugstad OD. Oxygen radicals and pulmonary damage. Pediatr Pulmonol1985;1:16775.

    22. Saugstad OD. Oxygen toxicity in the neonatal period. Acta Pediatr Scand1990;79:88192.

    23. Saugstad OD, Gluck L. Plasma hypoxanthine levels in newborn infants:a specific indicator of hypoxia. J Perinat Med 1982;10:26672.

    24. Brown ER, Stark A, Sosenko I, Lawson EE, Avery ME. Bronchopulmonarydysplasia: possible relationship to pulmonary edema. J Pediatr1978;92:9824.

    25. Ballard RA, Drew WL, Hufnagle KG, Riedel PA. Acquired cytomegalovirusinfection in preterm infants. Am J Dis Child 1979;133:4825.

    26. Fujimura M, Takeuchi T, Ando M, et al. Elevated immunoglobulin M levels inlow birth-weight neonates with chronic respiratory insufficiency. Early HumDev1983;9:2733.

    27. Fujimura M, Takeuchi T, Kitajima H, Nakajima M. Chorioamnionitis and serumimmunoglobulin M in WilsonMikity syndrome. Arch Dis Child 1989;64:137983.

    28. Watterberg KL, Carmichael DF, Gerdes JS, Werner S, Backstrom C, Murphy S.Secretory leukocyte protease inhibitor and lung inflammation in developingBPD. J Pediatr1994;125:2649.

    29. Yoon BH, Romero R, Jun JK, et al. Amniotic fluid cytokines (interleukin-6, tumornecrosis factor-alpha, interleukin-1-beta and interleukin-8) and the risk fordevelopment of bronchopulmonary dysplasia. Am J Obstet Gynecol1997;177:82530.

    30. Lyon A. Chronic lung disease of prematurity: the role of intra-uterine infection.Eur J Pediatr2000;159:798802.

    31. Speer CP. Inflammation and bronchopulmonary dysplasia. Semin Neonatol2003;8:2938.

    32. Kramer BW. Antenatal inflammation and lung injury: prenatal origin ofneonatal disease. J Perinatol 2008;28(Suppl. 1):S217.

    33. Bose CL, Dammann CF, Laughon MM. Bronchopulmonary dysplasia andinflammatory biomarkers in the premature neonate. Arch Dis Child FetalNeonatal Ed2008;93:F45561.

    34. Frank L, Sosenko IR. Undernutrition as a major contributing factor in thepathogenesis of bronchopulmonary dysplasia. Am Rev Respir Dis 1988;138:7259.

    35. Fujiwara T, Maeta H, Chida S, Morita T, Watabe Y, Abe T. Artificial surfactanttherapy in hyaline membrane disease. Lancet1980;i:559.

    36. Jobe AH. The new BPD: an arrest of lung development. Pediatr Res1999;66:6413.

    37. Saldanha RI, Cepeda EE, Poland RL. The effect of vitamin E prophylaxis on theincidence and severity of bronchopulmonary dysplasia. J Pediatr 1982;101:8993.

    38. Thomas W, Speer CP. Non-ventilator strategies for prevention and treatment ofbronchopulmonary dysplasiawhat is the evidence? Neonatology2008;94:1509.

    39. Shenai JP, Kennedy KA, Chytil F, Stahlman MT. Clinical trial of vitamin Asupplementation in infants susceptible to bronchopulmonary dysplasia.

    J Pediatr1987;111:26977.40. Schmidt B, Roberts R, Miller D, Kirpalani H. Evidence-based neonatal drugtherapy for prevention of bronchopulmonary dysplasia in very low birth-weight infants. Neonatology2008;93:2847.

    41. Liggins GC, Howie RN. A controlled trial of antepartum glucocorticoid treat-ment for prevention of the respiratory distress syndrome in premature infants.Pediatrics1972;50:51525.

    42. McCarthy MUS. Recommendations for antenatal corticosteroids. Lancet1994;343:726.

    43. van Marter LJ, Leviton A, Kuban KCK, et al. Maternal glucocorticoid therapyand reduced risk of bronchopulmonary dysplasia. Peditatrics 1990;86:3316.

    44. Philip AGS, Peabody JL, Lucey JF. Transcutaneous pO2 monitoring in the homemanagement of bronchopulmonary dysplasia. Pediatrics 1978;61:6557.

    45. Hudak BB, Allen MC, Hudak ML, Loughlin GM. Home oxygen therapy forchronic lung disease in extremely low birth-weight infants. Am J Dis Child1989;143:35760.

    46. Avery ME, Tooley WH, Keller JE, et al. Is chronic lung disease in low birthweight infants preventable? A survey of eight centers. Pediatrics 1987;79:

    2630.47. Patel D, Greenough A. Does nasal CPAP reduce bronchopulmonary dysplasia

    (BPD)? Acta Paediatr2008;97:13147.48. Stark AR. Risks and benefits of post-natal corticosteroids. NeoReviews

    2005;6:e99103.49. Philip AGS. Treatment of bronchopulmonary dysplasia with corticosteroids.

    Clin Res 1974;22:242A.50. Mammel MC, Green TP, Johnson DE, Thompson TR. Controlled trial of dexa-

    methasone therapy in infants with bronchopulmonary dysplasia. Lancet1983;i:13568.

    51. Avery GB, Fletcher AB, Kaplan M, Brudno DS. Controlled trial of dexamethasonein respirator-dependent infants with bronchopulmonary dysplasia. Pediatrics1985;75:10611.

    52. Cummings JJ, DEugenio DB, Gross SJ. A controlled trial of dexamethasone inpreterm infants at high risk for bronchopulmonary dysplasia. N Engl J Med1989;320:150510.

    53. Durand M, Sardesai S, McEvoy C. Effects of early dexamethasone therapy onpulmonary mechanics and chronic lung disease in very low birth weightinfants: a randomized controlled trial. Pediatrics 1995;95:58490.

    54. Gower WA, Wert SE, Nogee LM. Inherited surfactant disorders. NeoReviews2008;9:e45867.

    55. Pavlovic J, Papagaroufalis C, Xanthou M, et al. Genetic variants of surfactantproteins A, B, C and D in bronchopulmonary dysplasia. Dis Markers2006;22:27791.

    56. Clark DA, Pincus LG, Oliphant M, Hubbell C, Oates RP, Davey FR. HLA-A2 andchronic lung disease in neonates. J Am Med Assoc1982;248:18689.

    57. Bhandari V, Gruen JR. The genomics of bronchopulmonary dysplasia. NeoRe-views2007;8:e33644.

    58. Abman SH, Mourani PM, Sontag M. Bronchopulmonary dysplasia: a geneticdisease.Pediatrics 2008;122:6589.

    59. Horbar JD, Badger GJ, Carpenter JH, et al. Trends in mortality and morbidity forvery low birth weight infants, 19911999. Pediatrics 2002;110:14351.

    60. Hay Jr WW, Thilo E, Curlander JB. Pulse oximetry in neonatal medicine. ClinPerinatol1991;18:44172.

    61. Saugstad OD. Room air resuscitation two decades of neonatal research.EarlyHum Dev2005;81:1116.

    62. Jobe AH, Bancalari E. NICHD/NIH Workshop summary: bronchopulmonary

    dysplasia.Am J Respir Crit Care Med2001;163:17239.63. Walsh MC, Yao Q, Gettner P, et al. NICHD Neonatal Research Network: impact ofa physiologic definition on bronchopulmonary dysplasia rates. Pediatrics2004;114:130511.

    64. Lefkowitz W, Rosenberg SH. Bronchopulmonary dysplasia: pathway fromdisease to long-term outcome. J Perinatol 2008;28:83740.

    65. Shennan AT, Dunn MS, Ohlsson A, Lennox K, Hoskins EM. Abnormal pulmonaryoutcomes in premature infants: prediction from oxygen requirement in theneonatal period. Pediatrics 1988;82:52732.

    A.G.S. Philip / Seminars in Fetal & Neonatal Medicine 14 (2009) 333338338

  • 8/13/2019 06-Seminars in Fetal development

    10/75

    Prenatal factors in the development of chronic lung disease

    Anne Greenough*

    Division of Asthma, Allergy and Lung Biology, Kings College London School of Medicine, London, UK

    Keywords:

    Bronchopulmonary dysplasia

    Cytokines

    Glucocorticoids

    Infection

    Prematurity

    s u m m a r y

    Chronic lung disease (CLD), defined as chronic oxygen dependency, is a common outcome of neonatal

    intensive care. It occurs most frequently in infants born very prematurely, but also in infants born at term

    who had severe lung disease and those with abnormal antenatal lung growth due particularly toreduction in fetal breathing movements, amniotic fluid volume or intrathoracic space. There are,

    however, other causes and the importance of antenatal infection/inflammation regarding impairment of

    antenatal lung growth is increasingly recognised. Affected infants can suffer chronic respiratory

    morbidity including an excess of respiratory symptoms and lung function abnormalities even in adult-

    hood. Antenatal interventions directed at improving lung growth are available, but require testing

    inappropriately designed trials with pulmonary function at follow-up as an outcome.

    2009 Elsevier Ltd. All rights reserved.

    1. Introduction

    Chronic lung disease (CLD), defined as chronic oxygen depen-

    dency, is a common adverse outcome of neonatal intensive care. It

    occurs most frequently in infants born very prematurely; morethan 40% of one series of infants born before 29 weeks of gestation

    were affected.1 Such infants are now usually described as having

    bronchopulmonary dysplasia (BPD). Infants born at term, however,

    can remain chronically oxygen dependent, particularly if they have

    had severe lung disease as evidenced by a requirement for extra-

    corporeal membrane oxygenation (ECMO).2 The other major group

    of infants who can suffer chronic oxygen dependency are those

    with abnormal antenatal lung growth (Box 1). The aims of this

    review are to emphasise the importance of CLD by briefly

    describing the associated chronic respiratory morbidity and then to

    discuss the evidence as to whether certain prenatal factors

    predispose to the development of CLD and if there are effective

    antenatal interventions.

    2. Chronic respiratory morbidity

    2.1. Respiratory symptoms

    Recurrent respiratory symptoms requiring treatment are

    common in prematurely born children, particularly those who had

    BPD. At preschool age, 28% of a BPD cohort coughed and 7%

    wheezed more than once a week.3 Prematurely born infants, even

    without BPD, are more at risk of being symptomatic at follow-up

    than children born at term. In 78-year-olds, 30% of BPD children

    and 24% of prematurely born children without BPD were wheezing,whereas only 7% of term controls were so affected.4 This adverse

    respiratory outcome, particularly in those who had BPD, can persist

    into adulthood. In a follow-up study in The Netherlands, the

    prevalence of doctor-diagnosed asthma was significantly higher in

    19-year-olds born prior to 32 weeks of gestational age than in age-

    matched controls5; the females not the males had BPD, and

    were more likely to wheeze without a cold (35% vs 13%) and be

    short of breath on exercise (43% vs 16%).

    2.2. Lung function abnormalities

    Prematurely born infants, particularly those with wheezing at

    follow-up, have evidence of airways obstruction (a raised airways

    resistance and gas trapping) in the first 2 years after birth.6 Even atschool age, particularly in those with ongoing recurrent respiratory

    symptoms, evidence of poor airway growth persists.7 A strong

    correlation was demonstrated between the maximum flow at

    functional residual capacity at 2 years of age and forced expiratory

    volume in one second at school age, suggesting persistent airflow

    limitation at least in some patients with BPD.7 Adolescents who had

    BPD have evidence of airways obstruction and hyper-reactivity,

    with an increased responsiveness to histamine8 and apparently

    asymptomatic BPD patients have been demonstrated to desaturate

    on exercise.9 It is important to emphasise, however, that those

    adolescents received intensive care many years before and that the

    * Newborn Unit, 4th Floor Golden Jubilee Wing, Kings College Hospital, Denmark

    Hill, London SE5 9RS, UK. Tel.: 44 20 3299 3037; fax: 44 20 3299 8284.

    E-mail address: [email protected]

    Contents lists available atScienceDirect

    Seminars in Fetal & Neonatal Medicine

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / s i n y

    1744-165X/$ see front matter 2009 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.siny.2009.08.001

    Seminars in Fetal & Neonatal Medicine 14 (2009) 339344

    mailto:[email protected]://www.sciencedirect.com/science/journal/1744165Xhttp://www.elsevier.com/locate/sinyhttp://www.elsevier.com/locate/sinyhttp://www.sciencedirect.com/science/journal/1744165Xmailto:[email protected]
  • 8/13/2019 06-Seminars in Fetal development

    11/75

    long-term pulmonary function of those currently receiving inten-

    sive care is not known. Such infants are described as suffering from

    new BPD and, although they have less inflammation and fibrosis,

    they have an arrest in acinar development resulting in fewer and

    larger alveoli and reduction in the number of arteries.10 Whether

    such patients have appropriate catch-up lung growth requires

    careful investigation, particularly in infants with BPD, as there is

    evidence that their small airway function can decline over the first

    year.11

    There is a spectrum of severity in survivors with abnormal lung

    growth; some require many months of supplementary oxygen and

    recurrent respiratory symptoms are common, whereas those

    mildly affected may have only a raised respiratory rate in infancy.12

    Those patients who had surgically repaired congenital diaphrag-

    matic hernia in infancy have lung function abnormalities in

    adolescence, including reduction in forced expiratory volume,

    airways obstruction and increased airways hyper-responsiveness.13

    In addition, perfusion to the ipsilateral lung is decreased,14

    reflecting a persistent reduction in the number of branches or

    generations of pulmonary arteries. Some studies have suggested

    that these abnormalities are not associated with persisting symp-

    toms, but others have reported reduced exercise tolerance, with

    only 50% having enough stamina to take part in sport.15

    3. Prenatal factors predisposing to CLD

    3.1. Immaturity

    In the past, prematurely born infants who developed CLD

    frequently had had severe respiratory failure requiring high pres-

    sure mechanical ventilation with high concentrations of supple-

    mentary oxygen. Nowadays, CLD can occur in very prematurely

    born infants who initially had minimal or even no signs of lung

    disease,16 so-called new BPD. It has been suggested that in such

    infants abnormal vascular development may lead to the abnor-

    malities of lung growth. It has been proposed that the new BPDmay be a maldevelopment sequence resulting from interference/

    interruption of normal development signalling for terminal matu-

    ration and alveolarisation of the lungs of very preterm infants.17

    3.1.1. Antenatal infection/inflammation

    Certain evidence suggests that CLD may be more common if

    there has been antenatal infection/inflammation. Chorioamnionitis

    reduces the incidence of respiratory distress syndrome (RDS), but

    has been shown to be associated with an increase in CLD.18 In

    another series,19 however, histological chorioamnionitis was only

    associated with an increased risk of BPD if the infant subsequently

    developed postnatal infection or required mechanical ventilation

    for more than 7 days. It was therefore suggested that antenatal

    infection and/or inflammation is protective, unless there is post-natal sepsis or prolonged ventilation, and a multi-hit model for BPD

    development was proposed.19 A strong joint effect of prematurity

    and chorioamnionitis was demonstrated on early childhood

    wheezing in the Boston Birth cohort (n 1096), who were followed

    from birth to a mean age of 2.2 years.20

    Results from animal models demonstrate mechanisms by which

    antenatal infection/inflammation may result in BPD. For example,

    in preterm fetal lambs, a single dose of intra-amniotic endotoxin

    given before preterm delivery at 125 days of gestation resulted in

    an increased expression of mRNA for transforming growth factor

    (TGF)-b1 and a reduction in the expression of connective tissue

    growth factor (CTGF).21 TGF-b1 is a regulator of lung development,

    angiogenesis and airway remodelling22; transfer of TGF-b1 genes

    by an adenovirus vector to newborn rat pups resulted in enlarged

    alveolar airspaces.23 Decreased CTGF expression inhibits vascular

    development.

    3.1.2. Antenatal glucocorticoids

    It has been suggested that antenatal glucocorticoids are the first

    hit taken by the fetal lung, which primes the lung for more venti-

    lation-induced injury.24 The impact of antenatal endotoxin and

    betamethasone on the structure and function of preterm sheep

    lungs has been compared.25 Both treatments led to thinning of the

    alveolar walls, but the average alveolar volume increased by about

    20% and the total alveolar number decreased by almost 30%. The

    impact of antenatal betamethasone on fetal inflammation has been

    investigated in a sheep model.26 Ewes were treated at 108110 days

    of gestation (term being 150 days) with intra-amniotic endotoxin,

    intramuscular betamethasone, both or saline (control). At five days,

    the lambs who had received the combined intervention had

    increased alveolar neutrophils and proinflammatory cytokine

    mRNA expression, hence it was hypothesised that glucocorticoids

    impair the ability of the preterm lung to downregulate endotoxin-

    induced inflammation.26

    3.2. Reduction in amniotic fluid volume

    3.2.1. Renal abnormalities

    Reduction in amniotic fluid is associated with bladder outlet

    obstruction, bilateral renal dysplasia/hypoplasia and multicystic

    kidneys.27 Affected infants may be further predisposed to pulmo-

    nary hypoplasia by reduced renal proline production or thoracic

    compression.

    3.2.1.1. Antenatal interventions. Antenatal interventions to try to

    prevent pulmonary hypoplasia in infants with renal anomalies

    include relieving the oligohydramnios by amnioinfusion or by

    bypassing the urinary tract obstruction. A review of 169 cases of

    vesico-amniotic shunting (placing a pigtail shunt between the fetal

    bladder and amniotic cavity under ultrasound guidance) demon-

    strated only 47% perinatal survival and 45% shunt-related compli-cations.28 Oligohydramnios present before shunt replacement (56%

    mortality) and failure to restore amniotic fluid volume (100%

    mortality) were signs of a poor prognosis.28 An alternative

    approach has been to undertake laser therapy during fetal cystos-

    copy to disrupt posterior urethral valves, but this may result in

    damage to the nearby bowel. The impact of neither intervention on

    pulmonary development has been determined.

    3.2.2. Oligohydramnios

    The timing of onset of oligohydramnios in pregnancies

    complicated by premature, prelabour rupture of the membranes

    (PPROM) is critical; pulmonary hypoplasia only occurs if membrane

    rupture is prior to 26 weeks of gestation.29 Abnormal lung devel-

    opment, however, is not an invariable consequence of early-onsetoligohydramnios; 23% of one cohort with membrane rupture prior

    to 20 weeks of gestation had no signs of pulmonary hypoplasia.30

    3.2.2.1. Antenatal interventions. Serial amnio-infusion has been

    studied in pregnancies complicated by oligohydramnios resulting

    from PPROM. Unfortunately, the infused fluid was retained only in

    30% of one series of patients and in the remaining 70% oligohy-

    dramnios recurred within at least 48 h of the procedure.31

    3.2.3. Invasive antenatal diagnostic procedures

    Several groups have reported an excess of lung function

    abnormalities in the neonatal period and early infancy following

    first32 or second33 trimester amniocentesis. In addition, in a rand-

    omised trial the occurrence of neonatal RDS and pneumonia was

    A. Greenough / Seminars in Fetal & Neonatal Medicine 14 (2009) 339344340

  • 8/13/2019 06-Seminars in Fetal development

    12/75

    doubled in infants whose mother had undergone amniocentesis at

    a mean gestation of 16 weeks.34 Infants whose mothers who had

    undergone first trimester amniocentesis had more neonatal unit

    admissions35 and were more likely to be symptomatic at follow-

    up36 compared not only with controls, but also with infants whose

    mothers had had first trimester chorion villous sampling. Those

    data36 suggest that removal of even a small amount of amniotic

    fluid at a critical stage during pregnancy can adversely affect lung

    growth.

    3.3. Reduction in fetal breathing movements

    Selective destruction of the upper cervical cord between the

    lower medulla and the level of the phrenic nucleus results in

    cessation of fetal breathing movements and arrested lung growth

    and development.37 Cessation or reduction of fetal breathing

    movements may be responsible for the abnormal lung growth seen

    in such conditions as WerdigHoffman Disease and myotonic

    dystrophy. It has been suggested that the persistent absence of fetal

    breathing movements in pregnancies complicated by oligohy-

    dramnios due to premature and prolonged rupture of the

    membranes is a poor prognostic sign.38 Infants with anterior

    abdominal wall defects (AWD) can have abnormal lung growth.Stillbirths with exomphalos have been demonstrated to have small

    chests and infants with giant exomphalos reduced chest wall

    widths and lung areas on chest radiography. At follow-up, infants

    with either gastroschisis or exomphalos had significantly lower

    lung volumes than controls and five of the 13 had lung volumes

    below the normal range.39 Fetuses with AWD have a reduction in

    viscera in the upper part of the abdominal cavity and hence an

    inadequate framework for chest wall development. The low intra-

    abdominal pressure experienced by such patients could result in

    impaired diaphragmatic development. Indeed, in the neonatal

    period, the trans-diaphragmatic pressure generated by magnetic

    stimulation of the phrenic nerves has been demonstrated to be

    significantly lower in infants with gastroschisis than controls.40

    Those data40 suggest that the abnormal lung growth seen in certainAWD infants may be a reduction in fetal breathing activity.

    3.4. Reduction in intrathoracic space

    This can occur because of a small chest, particularly asphyxi-

    ating thoracic dystrophy, malformations of the lung [e.g. cystic

    adenomatoid malformation (CCAM) or lung cysts], pleural effusions

    and congenital diaphragmatic hernia (CDH). Fetal pleural effusions

    which spontaneously resolve have a good prognosis, but they can

    progress to non-immune hydrops because of impaired venous

    return and congestive cardiac failure due to compression. Fatal

    pulmonary hypoplasia also occurs in fetuses with rhesus iso-

    immunisation and results from chronic compression of the lungs by

    fetal ascites and pleural effusions. There may also be a directimmune-mediated injury affecting lung growth.41 Follow-up

    infants who had rhesus isoimmunisation demonstrated that those

    who had lower lung volumes had the first fetal blood sampling and

    intrauterine transfusion at a significantly earlier gestation.42

    3.4.1. Antenatal interventions

    3.4.1.1. Thoraco-amniotic shunting. Thoraco-amniotic shunting was

    initially used to decompress a large cyst in a fetus with a CCAM;

    subsequently further affected fetuses have been so treated, with

    70% survival in fetuses with macrocystic CCAM in one series.43 This

    intervention, however, is inappropriate for fetuses with solid or

    multicystic CCAMs. Pleural effusions can also be treated by thoraco-

    amniotic shunting with a pigtail catheter being placed under local

    anaesthetic into the effusion, which is then drained into the

    amniotic cavity. This can achieve reversal of the associated hydrops

    and chronic drainage with fetal lung expansion facilitating resus-

    citation at birth. Placement of thoraco-amniotic shunts was asso-

    ciatedwith 57% survival in fetuses with hydrops in one series; all 31

    survivors of 54 treated fetuses had chylothorax.44 Follow-up lung

    function studies45 demonstrated that the majority of infants who

    had had thoraco-amniotic shunting had lung volumes within the

    normal range,but the procedure was usuallyperformed in the third

    trimesterand hence probably toolate to influence lung growth. The

    UK National Institute for Health and Clinical Excellence (NICE) 46

    supports insertion of pleura-amniotic shunts with appropriate

    patient selection and counselling. An alternative strategy to

    manage fetal pleural effusions is to cause pleurodesis by an intra-

    pleural injection of OK-432. OK-432 is derived from a low virulence

    Su strain of type 3 Group A Streptococcus pyrogens. There are case

    reports of successful treatment.47 Rusticos review of the literature

    suggests that in-utero intervention (repeated thoracocentesis,

    intrauterine shunting or pleurodesis) improves the chances of

    survival in fetuses with persistent effusions48; nevertheless, rand-

    omised trials are required to determine if long-term respiratory

    outcome is improved.

    3.4.1.2. In-utero surgery. Open resection via maternal hysterotomyhas been associated with 5060% survival rates for fetuses with

    CCAM and associated hydrops.49 In animal models of CDH,

    pulmonary hypoplasia was reversed following repair of surgically

    created diaphragmatic defects; direct repair was performed by

    maternal hysterotomy and subsequent fetal thoracotomy.50 In

    a prospective trial in infants, however, no benefit over standard

    postnatal therapy was demonstrated51; fetal surgery was particu-

    larly inappropriate if there was liver herniation, as liver reduction

    resulted in kinking of the umbilical vein compromising venous

    return.51 Clinical observations of distended, hyperplastic lungs in

    cases of congenital high airway obstruction syndrome (CHAOS)

    prompted investigation of temporary tracheal occlusion in animal

    models as a potential method of preventing lung hypoplasia.

    Prevention of lung fluid egress causes lung tissue stretch,promoting airway and pulmonary vessel growth.52 Initial attempts

    at tracheal occlusion were via hysterotomy, but with poor survival

    rates; the limited data available demonstrated that, although open

    fetal tracheal occlusion was associated with increased lung growth

    as evidenced by an increase in lung weight, there was no

    improvement in the lung parenchymal lung structure or reduction

    in muscularisation of the pulmonary arteries.53 Fetoscopic inter-

    vention was then used which involved fetal neck dissection and

    temporary occlusive titanium clips; unfortunately this was associ-

    ated with complications including vocal cord paresis.54 As

    a consequence, the technique has now been modified such that the

    endoscopic placement of a balloon is used to temporarily obstruct

    the trachea (FETO). The balloon is retrieved by fetal tracheoscopy at

    34 weeks or is punctured during ultrasound guidance. Ultrasoundexaminations after FETO have demonstrated an increase in the

    echogenicity of the lungs within 48 h.55 Survival to discharge was

    50% following FETO in a multicentre study compared to 8% in

    eligible contemporary controls,56 but the cases were not rando-

    mised. Lung function of infants entered into a randomised trial of

    tracheal occlusion by an external clip or a balloon demonstrated

    only modest improvements in neonatal pulmonary function, but

    only 20 infants were studied.57 Complications of FETO include

    PPROM. The efficacy of FETO needs to be studied in a randomised

    trial including long term pulmonary outcomes.

    3.4.2. Prediction of pulmonary hypoplasia

    Antenatal lung:head circumference ratio (LHR) has been used as

    a prognostic indicator for CDH outcome. In a retrospective

    A. Greenough / Seminars in Fetal & Neonatal Medicine 14 (2009) 339344 341

  • 8/13/2019 06-Seminars in Fetal development

    13/75

    multicentre review, LHR measurements and position of the liver

    were obtained in 134cases of left-sided CDH between 24 and 28

    weeks of gestation.

    58

    When the LHR was 52 mmHg (minimal ventilation) or

  • 8/13/2019 06-Seminars in Fetal development

    48/75

    infants

  • 8/13/2019 06-Seminars in Fetal development

    49/75

    treatment with systemic corticosteroids, have proven efficacy

    based on RCTs. Although not used specifically for the prevention

    of BPD, the treatment of apnea of prematurity with caffeine and

    the use of aggressive phototherapy in ELBW infants are also

    associated with reductions in BPD. Individually, these treatments

    reduce risk by 711%. Unfortunately, treatment with systemic

    corticosteroids (particularly dexamethasone), although effective

    in reducing the risk of BPD, is associated with an increased rate

    of neurodevelopmental impairment. The benefits of treatment

    with systemic corticosteroids may outweigh the risks in infants

    with high baseline risk of BPD. The use of CPAP in selected

    populations of infants, in lieu of mechanical ventilation, may also

    decrease the incidence of BPD, but further data are needed to

    define this population. The use of quality improvement meth-

    odologies has potential for reducing BPD, but will rely upon

    high quality evidence from RCTs that support bundles of

    best practices.

    Because of the economic impact and long-term consequences of

    BPD, new preventive therapies are desirable. Future trials that test

    these therapies should incorporate strategies to systematically

    quantify the risk of BPD prior to enrollment. To date, a method for

    predicting BPD with sufficiently high sensitivity and specificity has

    not been available. A new tool using clinical and demographicvariables appears promising.103 Because of the heterogeneity of the

    causal pathways that lead to the development of BPD, it is unlikely

    that a single preventive strategy will have a major impact on its

    reduction. Rather, several strategies, with the expectation that each

    will contribute to a modest reduction in BPD, will need to be tested

    in RCTs.

    Conflict of interest statement

    None declared.

    Funding sources

    None.

    References

    1. McGuire W, FowlieP, Soll R. What hasthe Cochranecollaborationever done fornewborn infants?Arch Dis Child Fetal Neonatal Ed 2009. [Epub ahead of print].

    2. Gambrill E. Evidence-based clinical practice, [corrected] evidence-basedmedicine and the Cochrane collaboration. J Behav Ther Exp Psychiatry1999;30:114.

    3. Soll RF, Morley CJ. Prophylactic versus selective use of surfactant forpreventing morbidity and mortality in preterm infants.Cochrane Database SystRev; 2000. CD000510.

    4. Soll RF. Synthetic surfactant for respiratory distress syndrome in preterminfants.Cochrane Database Syst Rev; 2000. CD001149.

    5. Soll RF. Prophylactic synthetic surfactant for preventing morbidity andmortality in preterm infants. Cochrane Database Syst Rev; 2000. CD001079.

    6. Soll RF. Prophylactic natural surfactant extract for preventing morbidity andmortality in preterm infants. Cochrane Database Syst Rev; 2000. CD000511.

    7. Stevens TP, Harrington EW, Blennow M, Soll RF. Early surfactant administra-tion with brief ventilation vs selective surfactant and continued mechanicalventilation for preterm infants with or at risk for respiratory distresssyndrome. Cochrane Database Syst Rev; 2007. CD003063.

    8. Verder H, Albertsen P, Ebbesen F, et al. Nasal continuous positive airwaypressure and early surfactant therapy for respiratory distress syndrome innewborns of less than 30 weeks gestation. Pediatrics 1999;103:E24.

    9. Verder H, Robertson B, Greisen G, et al. Surfactant therapy and nasal contin-uous positive airway pressure for newborns with respiratory distresssyndrome. DanishSwedish Multicenter Study Group. N Engl J Med1994;331:10515.

    10. Reininger A, Khalak R, Kendig JW, et al. Surfactant administration by transient

    intubation in infants 29 to 35 weeks gestation with respiratory distresssyndrome decreases the likelihood of later mechanical ventilation:a randomized controlled trial. J Perinatol 2005;25:7038.

    11. Stevens TP, Blennow M, Soll RF. Early surfactant administration with briefventilation vs selective surfactant and continued mechanical ventilation forpreterm infants with or at risk for RDS. Cochrane Database Syst Rev; 2002.CD003063.

    12. Rojas MA, Lozano JM, Rojas MX, et al. Very early surfactant without manda-tory ventilation in premature infants treated with early continuous positiveairway pressure: a randomized, controlled trial. Pediatrics 2009;123:13742.

    13. Merrill JD, Ballard RA, Cnaan A, et al. Dysfunction of pulmonary surfactant inchronically ventilated premature infants.Pediatr Res 2004;56:91826.

    14. Laughon M, Bose C, Moya F, et al. A pilot randomized, controlled trial of latertreatment with a peptide-containing, synthetic surfactant for the preventionof bronchopulmonary dysplasia. Pediatrics 2009;123:8996.

    15. Marshall DD, Kotelchuck M, Young TE, et al. Risk factors for chronic lungdisease in the surfactant era: a North Carolina population-based study of verylow birth weight infants. North Carolina Neonatologists Association.Pediatrics1999;104:134550.

    16. Rojas MA, Gonzalez A, Bancalari E, et al. Changing trends in the epidemiologyand pathogenesis of neonatal chronic lung disease. J Pediatr1995;126:60510.

    17. Stefano JL, Abbasi S, Pearlman SA, et al. Closure of the ductus arteriosus withindomethacin in ventilated neonates with respiratory distress syndrome.Effects of pulmonary compliance and ventilation. Am Rev Respir Dis1991;143:2369.

    18. Yeh TF, Luken JA, Thalji A, et al. Intravenous indomethacin therapy inpremature infants with persistent ductus arteriosus a double-blindcontrolled study. J Pediatr1981;98:13745.

    19. Van Marter LJ, Pagano M, Allred EN, et al. Rate of bronchopulmonary dysplasiaas a function of neonatal intensive care practices. J Pediatr1992;120:93846.

    20. Clyman RI. Recommendations for the postnatal use of indomethacin: ananalysis of four separate treatment strategies. J Pediatr1996;128:6017.

    21. Fowlie PW. Intravenous indomethacin for preventing mortality and morbidityin very low birth weight infants.Cochrane Database Syst Rev; 200 0. CD00 0174.

    22. Cooke L, Steer P, Woodgate P. Indomethacin for asymptomatic patent ductusarteriosus in preterm infants. Cochrane Database Syst Rev; 2003. CD003745.

    23. Schmidt B, Davis P, Moddemann D, et al. Long-term effects of indomethacinprophylaxis in extremely-low-birth-weight infants. N Engl J Med 2001;344:196672.

    24. Knight DB. The treatment of patent ductus arteriosus in preterm infants. Areview and overview of randomized trials. Semin Neonatol 2001;6:6373.

    25. Ohlsson A, Walia R, Shah S. Ibuprofen for the treatment of patent ductusarteriosus in preterm and/or low birth weight infants. Cochrane Database SystRev; 2008. CD003481.

    26. Cassady G, Crouse DT, Kirklin JW, et al. A randomized, controlled trial of veryearly prophylactic ligation of the ductus arteriosus in babies who weighed1000 g or less at birth. N Engl J Med 1989;320:15116.

    27. Clyman R, Cassady G, Kirklin JK, et al. The role of patent ductus arteriosusligation in bronchopulmonary dysplasia: reexamining a randomized controlledtrial.J Pediatr2009;154:8736.

    28. Schmidt B, Roberts RS, Davis P, et al. Long-term effects of caffeine therapy forapnea of prematurity. N Engl J Med 2007;357:1893902.

    29. Schmidt B, Roberts RS, Davis P, et al. Caffeine therapy for apnea of prematurity.N Engl J Med 2006;354:211221.

    30. Stocker R, Glazer AN, Ames BN. Antioxidant activity of albumin-bound bili-rubin.Proc Natl Acad Sci U S A 1987;84:591822.

    Practice points

    BPD is an important morbidity associated with prema-

    ture birth.

    The prevention strategies with the highest qualityevidence with most favorable benefit/risk ratio include

    vitamin A and caffeine.

    Corticosteroids reduce the incidence of BPD, butincrease the risk of abnormal neurologic examination.

    Research directions

    A simple, clinically relevant, predictive model that

    objectively assesses the risk of BPD needs to be

    developed.

    Well-powered trials of surfactant therapy with briefventilation, and later surfactant therapy with the primary

    endpoint of CLD, are needed.

    An RCT of systemic corticosteroids versus placeboamong patients at high risk of bronchopulmonary

    dysplasia, with appropriate neurodevelopmental follow-

    up, is needed.

    M.M. Laughon et al. / Seminars in Fetal & Neonatal Medicine 14 (2009) 374382380

  • 8/13/2019 06-Seminars in Fetal development

    50/75

    31. Fardy CH, Silverman M. Antioxidants in neonatal lung disease. Arch Dis ChildFetal Neonatal Ed 1995;73:F1127.

    32. Schrod L, von Stockhausen HB. Serum bilirubin and development of chroniclung disease in very low birth weight infants. Eur J Pediatr 1999;158:16970.

    33. Morris BH, Oh W, Tyson JE, et al. Aggressive vs conservative phototherapy forinfants with extremely low birth weight. N Engl J Med 2008;359:188596.

    34. Bancalari E. Corticosteroids and neonatal chronic lung disease. Eur J Pediatr1998;157(Suppl. 1):S317.

    35. Halliday HL, Ehrenkranz RA, Doyle LW. Early (7 days) postnatal corticoste-roids for chronic lung disease in preterm infants. Cochrane Database Syst Rev;2009. CD001145.

    37. Doyle LW, Halliday HL, Ehrenkranz RA, et al. Impact of postnatal systemiccorticosteroids on mortality and cerebral palsy in preterm infants: effectmodification by risk for chronic lung disease. Pediatrics 2005;115:65561.

    38. Walsh M, Laptook A, Kazzi SN, et al. A cluster-randomized trial of bench-marking and multimodal quality improvement to improve rates of survivalfree of bronchopulmonary dysplasia for infants with birth weights of less than1250 grams.Pediatrics 2007;119:87690.

    39. Shah V, Ohlsson A, Halliday HL, Dunn MS. Early administration of inhaledcorticosteroids for preventing chronic lung disease in ventilated very lowbirth weight preterm neonates.Cochrane Database Syst Rev; 2007. CD001969.

    40. Finer NN, Barrington KJ. Nitric oxide for respiratory failure in infants born ator near term. Cochrane Database Syst Rev; 2006. CD000399.

    41. Abman SH, Kinsella JP, Schaffer MS, Wilkening RB. Inhaled nitric oxide in themanagement of a premature newborn with severe respiratory distress andpulmonary hypertension. Pediatrics1993;92:6069.

    42. Kinsella JP, Ivy DD, Abman SH. Inhaled nitric oxide improves gas exchange andlowers pulmonary vascular resistance in severe experimental hyalinemembrane disease.Pediatr Res1994;36:4028.

    43. McCurnin DC, Pierce RA, Chang LY, et al. Inhaled NO improves early pulmo-nary function and modifies lung growth and elastin deposition in a baboonmodel of neonatal chronic lung disease. Am J Physiol Lung Cell Mol Physiol2005;288:L4509.

    44. Barrington KJ, Finer NN. Inhaled nitric oxide for respiratory failure in preterminfants.Cochrane Database Syst Rev; 2007. CD000509.

    45. Schreiber MD, Gin-Mestan K, Marks JD, et al. Inhaled nitric oxide inpremature infants with the respiratory distress syndrome. N Engl J Med2003;349:2099107.

    46. Kinsella JP, Cutter GR, Walsh WF, et al. Early inhaled nitric oxide therapy inpremature newborns with respiratory failure.N Engl J Med2006;355:35464.

    47. Subhedar NV, Ryan SW, Shaw NJ. Open randomised controlled trial of inhalednitric oxide and early dexamethasone in high risk preterm infants. Arch DisChild Fetal Neonatal Ed 1997;77:F18590.

    48. Ballard RA, Truog WE, Cnaan A, et al. Inhaled nitric oxide in preterm infantsundergoing mechanical ventilation.N Engl J Med 2006;355:34353.49. Davis JM, Rosenfeld WN, Sanders RJ, Gonenne A. Prophylactic effects of

    recombinant human superoxide dismutase in neonatal lung injury. J ApplPhysiol1993;74:223441.

    50. Rosenfeld W, Evans H, Concepcion L, et al. Prevention of bronchopulmonarydysplasia by administration of bovine superoxide dismutase in preterminfants with respiratory distress syndrome. J Pediatr1984;105:7815.

    51. Davis JM, Rosenfeld WN, Richter SE, et al. Safety and pharmacokinetics ofmultiple doses of recombinant human CuZn superoxide dismutase adminis-tered intratracheally to premature neonates with respiratory distresssyndrome. Pediatrics 1997;100:2430.

    52. Davis JM, Parad RB, Michele T, et al. Pulmonary outcome at 1 year correctedage in premature infants treated at birth with recombinant human CuZnsuperoxide dismutase. Pediatrics 2003;111:46976.

    53. Speer CP. New insights into the pathogenesis of pulmonary inflammation inpreterm infants. Biol Neonate 2001;79:2059.

    54. Ahola T, Levonen AL, Fellman V, Lapatto R. Thiol metabolism in preterminfants during the first week of life. Scand J Clin Lab Invest2004;64:64958.

    55. Ahola T, Lapatto R, Raivio KO, et al. N-Acetylcysteine does not prevent bron-chopulmonary dysplasia in immature infants: a randomized controlled trial.

    J Pediatr2003;143:7139.56. Veness-Meehan KA. Effects of retinol deficiency and hyperoxia on collagen

    gene expression in rat lung. Exp Lung Res 1997;23:56981.57. Veness-Meehan KA, Bottone Jr FG, Stiles AD. Effects of retinoic acid on airspace

    development and lung collagen in hyperoxia-exposed newborn rats. PediatrRes2000;48:43444.

    58. Kennedy KA. Epidemiology of acute and chronic lung injury.Semin Perinatol1993;17:24752.

    59. Darlow BA, Graham PJ. Vitamin A supplementation to prevent mortality andshort and long-term morbidity in very low birthweight infants. CochraneDatabase Syst Rev; 2007. CD000501.

    60. Tyson JE, Wright LL, Oh W, et al. Vitamin A supplementation for extremely-low-birth-weight infants. National Institute of Child Health and HumanDevelopment Neonatal Research Network. N Engl J Med 1999;340:19628.

    61. Ambalavanan N, Tyson JE, Kennedy KA, et al. Vitamin A supplementation forextremely low birth weight infants: outcome at 18 to 22 months. Pediatrics2005;115:e24954.

    62. Cotton RB, Hazinski TA, Morrow JD, et al. Cimetidine does not prevent lunginjury in newborn premature infants. Pediatr Res 2006;59:795800.

    63. Schelonka RL, Katz B, Waites KB, Benjamin Jr DK. Critical appraisal of the roleof Ureaplasma in the development of bronchopulmonary dysplasia withmetaanalytic techniques. Pediatr Infect Dis J2005;24:10339.

    64. Mabanta CG, Pryhuber GS, Weinberg GA, Phelps DL. Erythromycin for theprevention of chronic lung disease in intubated preterm infants at risk for, orcolonized or infected with Ureaplasma urealyticum. Cochrane Database SystRev; 2003. CD003744.

    65. Ballard HO, Anstead MI, Shook LA. Azithromycin in the extremely low birth

    weight infant for the prevention of bronchopulmonary dysplasia: a pilotstudy. Respir Res 2007;8:41.

    66. Jobe AH, Ikegami M. Mechanisms initiating lung injury in the preterm.EarlyHum Dev 1998;53:8194.

    67. Kraybill EN, Runyan DK, Bose CL, Khan JH. Risk factors for chronic lungdisease in infants with birth weights of 751 to 1000 grams. J Pediatr1989;115:11520.

    68. Garland JS, Buck RK, Allred EN, Leviton A. Hypocarbia before surfactanttherapy appears to increase bronchopulmonary dysplasia risk in infants withrespiratory distress syndrome. Arch Pediatr Adolesc Med1995;149:61722.

    69. Mariani G, Cifuentes J, Carlo WA. Randomized trial of permissive hypercapniain preterm infants. Pediatrics 1999;104:10828.

    70. Carlo WA, Stark AR, Wright LL, et al. Minimal ventilation to prevent bron-chopulmonary dysplasia in extremely-low-birth-weight infants. J Pediatr2002;141:3704.

    71. Thome U, Kossel H, Lipowsky G, et al. Randomized comparison of high-frequency ventilation with high-rate intermittent positive pressure ventilationin preterm infants with respiratory failure. J Pediatr1999;135:3946.

    72. Clark RH, Gerstmann DR, Null Jr DM, deLemos RA. Prospective randomizedcomparison of high-frequency oscillatory and conventional ventilation inrespiratory distress syndrome. Pediatrics 1992;89:512.

    73. Ogawa Y, Miyasaka K, Kawano T, et al. A multicenter randomized trial of highfrequency oscillatory ventilation as compared with conventional mechanicalventilation in preterm infants with respiratory failure. Early Hum Dev1993;32:110.

    74. Frantz 3rd ID, Close RH. Alveolar pressure swings during high frequencyventilation in rabbits. Pediatr Res 1985;19:1626.

    75. High-frequency oscillatory ventilation compared with conventionalmechanical ventilation in the treatment of respiratory failure in preterminfants. The HIFI Study Group. N Engl J Med 1989;320:8893.

    76. Henderson-Smart DJ, Cools F, Bhuta T, Offringa M. Elective high frequencyoscillatory ventilation versus conventional ventilation for acute pulmonarydysfunction in preterm infants. Cochrane Database Syst Rev; 2007. CD000104.

    77. Courtney SE, Durand DJ, Asselin JM, et al. High-frequency oscillatory ventila-tion versus conventional mechanical ventilation for very-low-birth-weightinfants.N Engl J Med 2002;347:64352.

    78. Johnson AH, Peacock JL, Greenough A, et al. High-frequency oscillatory

    ventilation for the prevention of chronic lung disease of prematurity. N Engl JMed2002;347:63342.79. Keszler M, Modanlou HD, Brudno DS, et al. Multicenter controlled clinical trial

    of high-frequency jet ventilation in preterm infants with uncomplicatedrespiratory distress syndrome. Pediatrics 1997;100:5939.

    80. Wiswell TE, Graziani LJ, Kornhauser MS, et al. High-frequency jet ventilationin the early management of respiratory distress syndrome is associated witha greater risk for adverse outcomes. Pediatrics 1996;98:103543.

    81. Craft AP, Bhandari V, Finer NN. The sy-fi study: a randomized prospective trialof synchronized intermittent mandatory ventilation versus a high-frequencyflow interrupter in infants less than 1000 g. J Perinatol 2003;23:149.

    82. Morley CJ, Davis PG, Doyle LW, et al. Nasal CPAP or intubation at birth for verypreterm infants. N Engl J Med 2008;358:7008.

    83. Davis PG, Henderson-Smart DJ. Nasal continuous positive airways pressureimmediately after extubation for preventing morbidity in preterm infants.Cochrane Database Syst Rev; 2003. CD000143.

    84. Van Marter LJ, Allred EN, Pagano M, et al. Do clinical markers of barotraumaand oxygen toxicity explain interhospital variation in rates of chronic lungdisease? The Neonatology Committee for the Developmental Network. Pedi-

    atrics2000;105:1194201.85. Finer NN, Carlo WA, Duara S, et al. Delivery room continuous positive airway

    pressure/positive end-expiratory pressure in extremely low birth weightinfants: a feasibility trial. Pediatrics 2004;114:6517.

    86. Surfactant Positive Airway Pressure and Pulse Oximetry Trial (SUPPORT).Available at: http://clinicaltrials.gov/ct2/show/NCT00233324. AccessedAugust 31, 2009.

    87. Frank L, Sosenko IR. Undernutrition as a major contributing factor in the path-ogenesis of bronchopulmonary dysplasia.Am Rev Respir Dis1988;138:7259.

    88. Wilson DC, McClure G, Halliday HL, et al. Nutrition and bronchopulmonarydysplasia.Arch Dis Child 1991;66:378.

    89. Wilson DC, Cairns P, Halliday HL, et al. Randomised controlled trial of anaggressive nutritional regimen in sick very low birthweight infants. Arch DisChild Fetal Neonatal Ed 1997;77:F411.

    90. Sosenko IR, Innis SM, Frank L. Polyunsaturated fatty acids and protection ofnewborn rats from oxygen toxicity. J Pediatr1988;112:6307.

    91. Sosenko IR, Rodriguez-Pierce M, Bancalari E. Effect of early initiation ofintravenous lipid administration on the incidence and severity of chronic lungdisease in premature infants. J Pediatr1993;123:97582.

    M.M. Laughon et al. / Seminars in Fetal & Neonatal Medicine 14 (2009) 374382 381

    http://clinicaltrials.gov/ct2/show/NCT00233324http://clinicaltrials.gov/ct2/show/NCT00233324
  • 8/13/2019 06-Seminars in Fetal development

    51/75

    92. Friis-Hansen B. Body water compartments in children: changes during growthand related changes in body composition. Pediatrics 1961;28:16981.

    93. Oh W, Poindexter BB, Perritt R, et al. Association between fluid intake andweight loss during the first ten days of life and risk of bronchopulmonarydysplasia in extremely low birth weight infants. J Pediatr2005;147:78690.

    94. Bell EF, Acarregui MJ. Restricted versus liberal water intake for preventingmorbidity and mortality in preterm infants.Cochrane Database Syst Rev; 2008.CD000503.

    95. Hartnoll G, Betremieux P, Modi N. Randomised controlled trial of postnatalsodium supplementation on oxygen dependency and body weight in 2530

    week gestational age infants. Arch Dis Child Fetal Neonatal Ed2000;82:F1923.96. Brion LP, Primhak RA, Ambrosio-Perez I. Diuretics acting on the distal renal

    tubule for preterm infants with (or developing) chronic lung disease.CochraneDatabase Syst Rev; 2002. CD001817.

    97. Brion LP, Primhak RA. Intravenous or enteral loop diuretics for preterm infantswith (or developing) chronic lung disease. Cochrane Database Syst Rev; 2002.CD001453.

    98. Segar JL,ChemtobS, BellEF.Changesin bodywatercompartmentswith diuretictherapy in infants with chronic


Recommended