Nothing Special   »   [go: up one dir, main page]

Academia.eduAcademia.edu

Prevention and Treatment of Bronchopulmonary Dysplasia: Contemporary Status and Future Outlook

2008, Lung

Lung (2008) 186:75–89 DOI 10.1007/s00408-007-9069-z STATE OF THE ART REVIEW Prevention and Treatment of Bronchopulmonary Dysplasia: Contemporary Status and Future Outlook Laura Cerny Æ John S. Torday Æ Virender K. Rehan Received: 17 December 2007 / Accepted: 27 December 2007 / Published online: 30 January 2008 Ó Springer Science+Business Media, LLC 2008 Abstract Despite tremendous advances in neonatology, bronchopulmonary dysplasia (BPD) remains a major cause of morbidity and mortality among premature infants. Any intervention that would reduce the risk of BPD or improve its outcome is likely to have substantial clinical and financial benefits. However, there is a clear lack of an effective agent for the treatment and/or prevention of BPD. This is due to an incomplete understanding of the molecular mechanisms involved in its pathogenesis. Taking a basic biological approach, our laboratory has discovered that disruption of normal alveolar homeostatic signaling is centrally involved in this process. Using a number of in vitro and in vivo models, our laboratory has demonstrated that stabilization of the normal alveolar homeostatic signaling pathway(s) can prevent and/or rescue the molecular injuries caused by the insults that lead to BPD. Here, we review the existing approaches to prevent and treat BPD L. Cerny Harbor-UCLA-CHOC Neonatal Perinatal Fellowship Program, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center, Torrance, California 90502, USA J. S. Torday Departments of Pediatrics and Obstetrics and Gynecology, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center, Torrance, California 90502, USA V. K. Rehan Department of Pediatrics and Neonatal Intensive Care Unit, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center, Torrance, California 90502, USA V. K. Rehan (&) Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA e-mail: vrehan@labiomed.org and then, based on our insights into the pathogenesis of BPD, we propose novel and innovative therapeutic options that impact the disease on a cell/molecular level, unlike most of the current treatments available for BPD. Keywords Bronchopulmonary dysplasia  Chronic lung disease  Hyperoxia  Inhaled nitric oxide  Parathyroid hormone-related protein  Peroxisome proliferator-activated receptor c  Prematurity  Surfactant  Volutrauma Abbreviations ADRP Adipocyte differentiation-related protein ATII Alveolar type II BPD Bronchopulmonary dysplasia CC10 Clara cell 10-kDa protein CLD Chronic lung disease FDA Food and Drug Administration GA Gestational age iNO Inhaled nitric oxide IVH Intraventricular hemorrhage NCPAP Nasal continuous positive airway pressure NEC Necrotizing enterocolitis PaCO2 Arterial partial pressure of carbon dioxide PDA Patent ductus arteriosus PTHrP Parathyroid hormone-related protein PVH Periventricular hemorrhage PVL Periventricular leukomalacia PPARc Peroxisome proliferator-activated receptor c ROP Retinopathy of prematurity RDS Respiratory distress syndrome RGZ Rosiglitazone SNIPPV Synchronized nasal intermittent positive pressure ventilation 123 76 SIMV TA VLBWI Wingless/ int Lung (2008) 186:75–89 Synchronized intermittent mechanical ventilation Tracheal aspirate Very-low-birth-weight Infant Wnt Introduction Bronchopulmonary dysplasia (BPD) is a chronic pulmonary disorder that is the consequence of abnormally repaired lung damage due to premature birth and lung immaturity. The clinical definition of BPD established by the National Institute of Child Health and Human Development Workshop on BPD in 2001 stratifies the definition based upon gestational age (GA) at birth. Infants born at less than 32 weeks postmenstrual age (PMA) are classified as having mild BPD if they require oxygen for the first 28 days, but are in room air at 36 weeks PMA or time of discharge; moderate BPD if they require oxygen for the first 28 days and need less than 30% oxygen at 36 weeks PMA or time of discharge; and severe BPD if they require oxygen for the first 28 days and are still requiring 30% or more oxygen and/or continuous positive airway pressure or mechanical ventilation at 36 weeks PMA or discharge. For infants born 32 weeks or more PMA, the categories of mild, moderate, and severe are defined the same; however, the time to evaluate the oxygen status is 56 days postnatal age or discharge, rather than 36 weeks. Of note, a physiologic test such as pulse oximetry should be performed to truly assess the need of oxygen because there is much variation in oxygen treatment practices. Furthermore, for a child to be classified as having BPD, he or she should display persistence of common clinical features of respiratory distress syndrome [1]. Despite tremendous advances in neonatal care in general, and neonatal respiratory care in particular, BPD still remains a major cause of morbidity and mortality among premature infants [2]. Although these medical advances may have decreased the severity of BPD, its incidence has not decreased, and may in fact have actually increased [1, 3]. The incidence of BPD increases with decreasing birth weight and currently affects approximately 30% of infants with birth weights less than 1000 g [2]. Affected infants have both short- and long-term clinical issues, including, but not limited to, a complicated initial neonatal intensive care course, poor growth, neurodevelopmental delays, and repeated hospitalizations during the first few years of life [4–6]. Therefore, any intervention that would reduce the risk of BPD or improve its outcome is likely to have substantial clinical and financial benefits. Despite this, 123 there is a clear lack of an effective agent for the treatment and/or prevention of BPD. ‘‘Old’’ versus ‘‘New’’ BPD In the 1960s, when Northway described BPD, he described it as a chronic lung disease (CLD) of relatively larger preterm infants who required ventilation with high pressures and oxygen for prolonged periods of time [7]. It was characterized by four well-defined clinical, radiologic, and histopathologic findings. The hallmarks of the now socalled ‘‘old BPD’’ were severe large airway injury, interstitial and alveolar edema, and extensive small airway disease with alternating areas of overinflation and fibrosis. With the introduction of antenatal steroids, routine surfactant replacement for respiratory distress syndrome (RDS), and modern respiratory care, in contrast, the ‘‘new’’ BPD occurs primarily in extremely premature infants, occurs after only modest ventilatory and oxygen needs, and is not accompanied by the classic clinical and radiologic stages of the ‘‘old’’ BPD. The most dramatic histopathologic finding is ‘‘arrested alveolarization’’ with minimal large or small airway disease, and relatively less inflammation and fibrosis [2, 8] (Table 1). Why Prevention and Treatment Have Not Worked Thus Far The primary prevention of BPD through elimination of the quintessential risk factor, prematurity, has not yet been achieved and is unlikely to be achievable in the near future because of the lack of understanding of the pathophysiology of preterm labor. Up until now, the available preventive/treatment strategies for BPD have also suffered the same limitations, i.e., lack of understanding of the Table 1 Comparsion of ‘‘old’’ versus ‘‘new’’ bronchopulmonary dysplasia Old BPD New BPD Larger preterm infants Extremely premature infants High ventilation and oxygen needs Modest ventilation and oxygen needs Severe large airway injury Interstitial and alveolar edema Minimal large airway disease Arrested alveolarization Extensive small airway disease with alternating areas of overinflation and fibrosis Minimal small airway disease with less inflammation and fibrosis Pulmonary artery muscularization Fewer and abnormal pulmonary arteries Lung (2008) 186:75–89 77 molecular processes involved in both normal and abnormal lung development. The preventive interventions such as antenatal and postnatal steroids, ‘‘kinder and gentler’’ ventilation, early closure of patent ductus arteriosus (PDA), treatment of prenatal and postnatal infections, fluid restriction, various nutrient supplements (vitamin A, inositol, polyunsaturated fatty acids), and, most recently, nitric oxide administration are either not effective or are associated with unacceptable side effects [9–12]. Most of the these interventions are of an empiric nature, failing to target the underlying molecular processes that lead to BPD, which may precisely be the reason that none of these interventions have been effective in preventing BPD. Therefore, the available preventive strategies for BPD are not effective, not safe, and not based on a thorough understanding of the pathogenesis of BPD. With recent advances in our understanding of the cell/molecular processes involved in both normal and abnormal lung development, in particular the mechanism of alveolar development and its failure in BPD, there is now new hope for not only preventing and treating BPD, but also possibly reversing established disease [13]. The intent of this article is to review some of the existing approaches to prevent and manage BPD, which target the symptoms (Table 2) and then describe a state-ofthe-art approach to the prevention and treatment of BPD that impacts the disease on a cell/molecular level, unlike most of the treatments we currently have. Table 2 Current treatments for bronchopulmonary dysplasia Therapy Strategy/mechanism of action Benefit/comment Ventilation Avoid ventilation when possible with the use of NCPAP or SNIPPV Minimizes volu/barotrauma caused by ventilation Minimize ventilator exposure time. Accept permissive hypercapnia Oxygen Accept oxygen saturations between 88 and 92% Prevents oxygen toxicity Surfactant Provide exogenous pulmonary surfactant to the deficient neonate to decrease alveolar surface tension and improve lung compliance Possible reduction in BPD or death at 28 days Inhaled nitric oxide Decreases V/Q mismatch, reduces inflammation, and restores normal growth patterns in premature lung No overall reduction in BPD but a trend toward reduction in BPD in select populations Caffeine Decreases apnea by acting as a central respiratory stimulant, by decreasing the threshold to CO2, and by decreasing diaphragmatic fatigue Decreases duration of PPV and possibly frequency of BPD. However, neurodevelopmental studies to ensure safety are awaited Induces lung maturation Reduces the incidence and severity of RDS and neonatal death but not the incidence of BPD Systemic Anti-inflammatory Significant reduction in the incidence of BPD but have negative neurodelevopmental consequences Inhaled Steroids Antenatal Postnatal Local anti-inflammatory with minimal systemic effects Ineffective in reducing incidence of BPD Antibiotics Treatment of pathogens causing infection/inflammation Treatment of Ureaplasma has not shown a decrease in BPD Diuretics Reduce pulmonary edema Improved lung function in short-term but no decrease in the incidence of BPD Bronchodilators Dilate airways by relaxation of bronchial smooth muscle to improve compliance/tidal volume and reduce airway resistance Provide adequate calories for somatic and pulmonary growth and injury/repair Insufficient data to evaluate safety and efficacy for prevention or treatment of BPD Allows providing adequate calories for improved growth in the face of fluid restriction Vitamin A Important for growth, immunity, and integrity of epithelial cells Decreases incidence of BPD but safety not fully established Inositol Essential nutrient for cell signaling, cell membrane maintenance, and maturation of surfactant Protect against inflammation and oxygen toxicity, both known contributors to BPD A trend toward reduction in the risk of death or BPD but needs more studies Have not proven successful in reducing the incidence of BPD Nutrition/fluid restriction Antioxidants and anti-inflammatory agents 123 78 Existing Approaches to Prevent and/or Treat BPD Kinder and Gentler Ventilatory Management Volutrauma/barotrauma from mechanical ventilation is one of the key risk factors for the development of BPD. A potential approach to prevent BPD, therefore, is to avoid mechanical ventilation, for example, by using noninvasive ventilatory support modalities such as nasal continuous positive airway pressure (NCPAP) and synchronized nasal intermittent positive pressure ventilation (SNIPPV). Support for early NCPAP and avoidance of mechanical ventilation was first provided by Avery et al. [14] in 1985 in a study in which it was shown that the incidence of BPD at Columbia-Presbyterian Medical Center (Columbia) in New York was significantly less than the other study centers. The strategy at Columbia encourages early use of NCPAP. A follow-up study by Van Marter et al. [15] in 2000 compared the experience at Columbia-New York to Harvard-Boston and again found a lower incidence of BPD at Columbia, which was attributed to lower rates of initiation of mechanical ventilation. Though the incidence of BPD using the Columbia approach is lower, the question as to its severity needs to be addressed, as those patients who are not intubated do not see the potential benefits of surfactant. The success of early NCPAP improves with gestational age and experience of the user, implying that it may be a feasible option in a subset of patients in experienced centers [16]. Although minimizing the use of mechanical ventilation by using early NCPAP has shown trends toward decreased incidence of BPD, further randomized controlled trials are needed to confirm these results [17, 18]. As described above, early/prophylactic NCPAP is a promising tool for reduction in ventilator use and incidence of BPD. NCPAP also has been evaluated in the postextubation setting to prevent reintubation. A meta-analysis of randomized trials has shown that extubation to NCPAP versus headbox oxygen significantly reduced the incidence of apnea and oxygen requirements [19]. In addition to continuous positive pressure ventilation, other modes of nasal ventilation are being used. One mode that has been studied is SNIPPV, which provides breaths in addition to the continuous pressure. A meta-analysis of SNIPPV versus NCPAP after extubation showed a significant benefit for the preterm infants extubated to SNIPPV in terms of prevention of failed extubation and a trend toward decreased CLD in the SNIPPV group [20]. Another recent study suggested that infants on SNIPPV versus NCPAP after extubation had a significantly lower need for supplemental oxygen and a lower incidence of BPD. This study, however, was not randomized [21], and further studies are needed to confirm the benefits of SNIPPV. 123 Lung (2008) 186:75–89 For those infants who are supported on mechanical ventilation, the ideal ventilation strategy is still not clear. The general strategy is to minimize pulmonary damage by minimizing time on the ventilator, reducing peak inspiratory pressure, restricting tidal volume to 3–6 cc/kg, and using short inspiratory times of 0.24–0.4 s [22, 23]. The concept of permissive hypercapnia has allowed us to rethink our target PaCO2 levels and subsequently rationalize lower ventilator settings. Studies have shown that aiming for PaCO2 goals of 45–55 mmHg versus 35– 45 mmHg resulted in shorter duration of ventilation. A multicenter trial conducted by the National Institute of Child Health and Human Development Neonatal Research Network showed that permissive hypercapnia resulted in a trend toward a decrease in the incidence of BPD. There were no trends toward worse short-term neurodevelopmental outcomes in the higher PaCO2 cohort and a metaanalysis revealed IVH actually decreased in the permissive hypercapnia group versus control. Findings suggesting decrease in the incidence of IVH are important, as some of the initial concerns of accepting higher PaCO2 levels included potential disruption of cerebral autoregulation of blood flow. Permissive hypercapnia may have other benefits in addition to decreased ventilator requirements, including more efficient PaCO2 elimination and a shift of the oxygen-dissociation curve to the right, allowing improved oxygen delivery to the tissues [24]. It should be noted, however, that a recent study found that infants randomized to target PaCO2 range of 55–65 mmHg versus 35–45 mmHg for the first 7 days of life were found to have trends toward higher incidence of BPD or death and significantly increased combined outcome of mental impairment at 18–22 months or death. The study was stopped before completion [25]. Although the concepts of permissive hypercapnia and minimizing the damage inflicted by mechanical ventilation remain promising, we must use caution in the application of permissive hypercapnia until more data are available. Use of high-frequency oscillatory ventilation (HFOV) has been evaluated as both a mode of ventilation for prophylaxis against BPD and as a rescue mode for infants in severe respiratory failure. A meta-analysis of 11 trials comparing early use of HFOV to conventional ventilation showed a modest reduction in BPD but no statistically significant effect on mortality, short-term neurologic outcomes, ICH, or PVL [26]. However, many other recent studies have provided conflicting data [27–29]. These conflicting data are likely to be due to differences in the study populations among various studies. HFOV has also been used as a rescue mode for infants in severe respiratory failure, but the evidence that this approach can reduce BPD or improve long-term outcomes in preterm infants is not yet available [18, 26]. Lung (2008) 186:75–89 Other ventilatory modes employed to reduce BPD include patient-triggered ventilation and volume-targeted ventilation. Patient-triggered ventilation modes such as assist control and synchronized intermittent mechanical ventilation (SIMV) were introduced with the hope that such ventilation modes would decrease the incidence of BPD. However, a meta-analysis of randomized trials evaluating patient-triggered modes does not show a significant decrease in the incidence of BPD. Use of patienttriggered modes, however, was shown to shorten the duration of ventilation if started in the recovery phase but not early in the acute phase of illness [26]. Another strategy being evaluated is volume-targeted versus pressure-control ventilation to determine if one is superior. A meta-analysis of four randomized trials comparing volume-targeted with pressure-limited ventilation showed that the use of volumetargeted ventilation significantly decreases the duration of mechanical ventilation as well as the rate of pneumothorax, but did not show a significant decrease in death. There was, however, a reduction in the incidence of BPD among the surviving infants. Further studies are needed to confirm these results and explore other modes that ventilators are now capable of delivering [26, 30]. Oxygen Supplementation Oxygen toxicity plays a major role in the pathogenesis of lung injury and BPD as well as other systemic diseases such as retinopathy of prematurity (ROP), necrotizing enterocolitis (NEC), and intraventricular/periventricular (IVH/PVH) hemorrhage, necessitating the need to establish a balance between appropriate oxygen delivery and toxicity. Although studies have shown that hyperoxia can have major effects on lung tissue, including, but not limited to, increased proliferation of alveolar type II (ATII) cells and fibroblasts, alterations in the surfactant system, increases in inflammatory cells and cytokines, increased collagen deposition, and decreased alveolarization and microvascular density, there is also concern that hypoxia can lead to decreased growth and poor neurodevelopmental outcomes. The lack of sound data has contributed, therefore, to the wide variation in practice between care providers in terms of the accepted oxygen saturation range. Recent data support, however, that aiming for saturation targets in the range of 85–93%, rather than [92%, decreases the incidence of ROP, oxygen use at 36 weeks, and use of postnatal steroids without increasing rates of NEC, IVH/ PVH, periventricular leukomalacia (PVL), or having a detrimental effect on developmental outcomes [31]. Furthermore, a recent study showed that targeting a high oxygen saturation goal of 95–98% in infants born at less than 30 weeks of GA and who were oxygen-dependent at 79 32 weeks had no significant benefit on growth or development [32]. Although determining appropriate saturation ranges is important, another issue that is now apparent is our inability to maintain infants within set target ranges, regardless of what saturations are set. Some studies have revealed that infants are within set ranges as little as 50% of the time. An important goal for the neonatal community to achieve, therefore, is to avoid frequent oxygen fluctuations to both hypoxic and hyperoxic ranges [33]. Medications Pulmonary surfactant The use of pulmonary surfactant has been instrumental in changing the face of the ‘‘old’’ BPD. The currently available surfactants are either animal-derived (natural) or synthetic. The original synthetic surfactants contained phospholipids and lacked the surfactant proteins present in natural surfactants. The current evidence suggests that the use of natural surfactant versus synthetic surfactant leads to greater improvement in initial ventilator requirements, lower incidence of pneumothorax, and a trend toward increased survival [34]. However, in some studies the use of natural surfactant had a slightly higher incidence of IVH, but a similar incidence of grades 3 and 4 IVH. Other concerns about the use of animal-derived products include transmission of infectious agents, immune reactions, and the presence of other impurities. These concerns, however, have not been realized [34, 35]. As a result of concerns about animal-derived products, there have been attempts to make an improved synthetic surfactant. Lucinactant is a new synthetic surfactant (also known as Surfaxin) that contains sinapultide, a peptide that mimicks human SP-B. Compared with porcine-derived poractant alfa (Curosurf), Surfaxin showed similar efficacy and safety for treatment of RDS [36]. Additional studies showed that Surfaxin was more effective than colfosceril palmitate (synthetic surfactant) for prevention of RDS, it reduced the incidence of BPD compared with colfosceril, and it decreased the RDS-related mortality rates compared with beractant (bovine-derived). The side-effect profile for Surfaxin was as good as or better than colfosceril palmitate and beractant for the secondary outcomes of air leaks, IVH, PVL, ROP, pulmonary hemorrhage, PDA, NEC, and apnea [37]. Furthermore, a 1-year follow-up comparing infants who received lucinactant versus other surfactants in two trials revealed that the infants who received lucinactant had similar neurologic outcomes compared with infants who received colfosceril palmitate, beractant, and poractant [38]. These initial trials of Surfaxin have yielded promising results and strongly suggest that this new surfactant functions better 123 80 clinically than previous synthetic surfactants. Unfortunately, questions about quality control during its production have hampered trials to further prove its superior efficacy and safety. Furthermore, a meta-analysis of two studies comparing protein-containing synthetic surfactants versus animal-derived surfactants for prevention and treatment of RDS showed that there were no statistically significant differences between the groups for mortality at 36 weeks, CLD, or the combined outcome of death or CLD [39]. In terms of the timing of surfactant administration and its efficacy, a meta-analysis of four randomized controlled studies evaluating early (within 2 h of life) versus late surfactant administration favored the use of early administration by showing reductions in the risk of pneumothorax, pulmonary interstitial emphysema, neonatal mortality, and CLD or death at 36 weeks. The use of early surfactant treatment also showed a trend toward reduction in the risk for BPD or death at 28 days [40]. It should be noted, however, that when evaluating the overall use of surfactant, there has not been a significant reduction in the incidence of BPD [38]. Inhaled nitric oxide (iNO) The properties of iNO, including its ability to decrease ventilation/perfusion mismatch, reduction of inflammation, and restoration of more normal patterns of growth in the premature lung, make it a candidate for the treatment of BPD. However, the available data on iNO for prevention or treatment of BPD is inconsistent [10, 41–44]. For example, a multicenter randomized trial by Kinsella et al. [41] in 2006 showed that in infants less than 34 weeks GA with respiratory failure requiring mechanical ventilation, there was no significant decrease in BPD with the use of lowdose iNO. In the subpopulation of infants weighing 1000– 1250 g, however, they did find a reduction in BPD. Ballard et al. [10] in 2006 found that infants less than 1250 g who received iNO had shorter hospital stays and shorter durations of oxygen use, but no decrease in BPD incidence. Taken together, the available studies are suggesting that iNO may have its place in the treatment of BPD, especially for particular subsets of infants such as those who are more than 1000 g and are older than 7 days. Again, however, further studies are needed to support this conclusion and to determine first safety and then optimal dose, timing, and duration of iNO use in preterm infants [42, 43]. Caffeine Caffeine has been shown to decrease the frequency of apnea of prematurity and mechanical ventilation during the 123 Lung (2008) 186:75–89 first week of life. A large randomized international study by Schmidt et al. [45] showed that in infants who received caffeine, positive pressure support was discontinued 1 week earlier than in the group of infants assigned to placebo. Furthermore, caffeine significantly reduced the frequency of BPD, defined as a need for supplemental oxygen at the age of 36 weeks PMA. However, the primary outcome of this study, i.e., evaluation of the neurologic outcome of this cohort at 18–21 months, is still pending. Results of long-term follow-up will be important in confirming the safety of caffeine. Potential risks of caffeine use include poor weight gain secondary to increased oxygen consumption and the fact that methyxanthines inhibit adenosine receptors, since adenosine has a neuroprotective role in the brain against hypoxia and ischemia [45]. Corticosteroids Antenatal Knowing that inflammation plays an important role in the pathogenesis of BPD, agents that control inflammation are a logical choice for treatment and/or prevention of BPD. Corticosteroids have strong anti-inflammatory properties and therefore have been extensively studied as agents to prevent and treat BPD. Many studies have shown that antenatal corticosteroids have benefits for the infant, including reduction in fetal or neonatal death, RDS, and IVH. In a large review of six studies including 818 infants, however, there was no statistically significant difference in BPD between those infants exposed to antenatal steroids and control [46, 47]. Systemic Steroids Initial data on the use of ‘‘early’’ systemic steroids were promising for the prevention of BPD. A meta-analysis of 21 randomized controlled trials comparing systemic steroids given at less than 96 h of life versus control showed significant decreases in the incidence of BPD at both 28 and 36 weeks and in death or BPD, ROP, and PDA. The incidences of mortality, IVH, PVL, NEC, and pulmonary hemorrhage were not significantly different between the two groups. The concerns, however, came with the evaluation of late neurodevelopmental outcomes. Infants receiving ‘‘early’’ systemic steroids were found to have a higher incidence of cerebral palsy, developmental delay, and abnormal neurologic examinations. The rate of combined death or major neurosensory disability in these studies, however, was not significantly increased in the steroid-exposed group [48]. Until we have further long- Lung (2008) 186:75–89 term follow-up on patients treated with systemic steroids, the neonatal community needs to avoid the use of ‘‘early’’ systemic steroids as much as possible. Another potential use of early systemic corticosteroids is for the treatment of adrenal insufficiency in premature infants. However, a study by Watterberg et al. [49] revealed that the use of low-dose hydrocortisone therapy did not improve survival without BPD. In this study, there were also significant concerns about the increase in gastrointestinal perforation in the treatment group. Furthermore, the combination of indomethacin and hydrocortisone appeared to have an additive risk of perforation. Attempts at finding the optimal timing for administration of systemic steroids that may provide pulmonary benefit without neurologic sequelae has led to the examination of ‘‘moderately early’’ (7–14 days) and ‘‘late’’ systemic (beyond 7 days) steroid use [50, 51]. Although infants receiving ‘‘moderately early’’ or ‘‘late’’ systemic steroids compared to controls show early extubation and decreased BPD at 28 days and 36 weeks, decreased combined outcome of death or BPD, and decreased number of children discharged home on oxygen, these regimens are associated with significant side effects, including hypertension, hyperglycemia, hypertrophic cardiomyopathy, and infection. Furthermore, the limited available long-term neurologic follow-up, again, puts the use of these regimens into question [49, 50]. Inhaled Steroids Due to the concerns of side effects of systemic steroids, inhaled steroids have been studied as a possible way to deliver local benefit with less systemic side effects. If given during the first 2 weeks of life to ventilated infants with birth weights less than 1500 g, inhaled corticosteroids do not reduce the incidence of BPD [52]. There is, however, some evidence to suggest that inhaled steroids reduce the use of systemic postnatal steroid use, but this has not been a consistent finding [52]. Studies comparing inhaled versus systemic corticosteroid use for both treatment and prevention of BPD in very-low-birth-weight infants have also been performed [53, 54]. Based on the available data, neither inhaled nor systemic steroids can currently be recommended for the treatment or prevention of BPD. Antibiotics for Infection/Chorioamnionitis Premature infants are commonly delivered in the setting of maternal chorioamnionitis. Infants born to mothers with chorioamnionitis tend to initially have accelerated lung maturation with less RDS, but are then at higher risk for 81 developing BPD [55]. Although we know that inflammation is a key element in the development of BPD, how postnatal inflammation affects development of the lung that has been exposed to chronic infection in utero is not known. For now, our goal should be to decrease proinflammatory exposures of the preterm lung both in utero and in the newborn period [56]. A logical approach to decreasing inflammation is to treat infection, which is a common source of inflammation in the neonatal population. One of the common pathogens that colonize the respiratory tract of preterm infants is Ureaplasma urealyticum. Colonization with Ureaplasma is thought to be a risk factor for the development of BPD [57]. One study evaluating lung specimens revealed that Ureaplasma-infected specimens were associated with more myofibroblast proliferation and interstitial fibrosis than lung specimens affected by other pathogens [58]. A few studies have been performed to see if in infants less than 30 weeks GA requiring ventilatory support, treatment of Ureaplasma would decrease the incidence of BPD. One study looked at prophylactic treatment with erythromycin in culture-unknown patients, while another study looked at erythromycin treatment in culture-positive infants. Neither study showed a decrease in BPD or death following treatment with erythromycin [59]. A promising pilot study by Ballard et al. [60] in 2007 showed that in infants with birth weight of less than 1000 g who had prophylactic treatment with azithromycin versus placebo had less postnatal steroid use and significantly shorter mechanical ventilation. Again, further larger prospective randomized controlled studies are needed to evaluate the effect of treating Ureaplasma infection on the incidence of BPD. Diuretics Infants with BPD have a tendency to retain fluid and develop pulmonary edema. To balance the fluid status of these infants with adequate calorie delivery, diuretics are often incorporated in the management plan. Review of the literature shows that aerosolized diuretics may temporarily improve pulmonary mechanics, but not enough data are available on long-term outcomes to show benefit [61]. Evaluation of the use of systemic diuretics that act on the distal tubule show that in infants older than 3 weeks with BPD there is improved lung compliance, but there is no evidence to support decreased need for ventilatory support or improved long-term outcomes [62]. A review of the use of loop diuretics shows that in infants less than 3 weeks of age furosemide has inconsistent effects, and in infants older than 3 weeks of age furosemide improves oxygenation and lung compliance. However, no data are available to show a decrease in BPD with the use of furosemide [63]. 123 82 Bronchodilators Bronchodilators are used to dilate small airways by relaxing bronchial smooth muscle. Bronchodilators are potentially useful in the prevention or treatment of BPD, as many studies of pulmonary mechanics have shown shortterm improvements in compliance and tidal volume and decreased airway resistance when bronchodilators are used in infants with CLD. However, in the only randomized controlled trial on use of bronchodilators for prevention or treatment of CLD, with mortality or CLD as the primary outcome, prophylactic use of salbutamol did not show significant differences in mortality, CLD, or duration of ventilation or oxygen requirement when compared to controls. Currently, there are insufficient data to evaluate the efficacy and safety of bronchodilators for prevention or treatment of BPD, and therefore their use to prevent BPD is not recommended [64]. Nutrition/Fluid Management Maximizing caloric intake helps injury repair. The optimal approach to feeding premature infants is not yet known and we have no available studies evaluating the benefits of increased ([135 kcal/kg/day) energy intake versus standard energy intake [65]. However, it is known that decreased protein and calorie intake can significantly decrease alveolar number [66]. Fluid management is another important aspect of BPD care. Fluid restriction, without compromise of calories, can improve lung function and decrease oxygen requirements and may decrease the risk of BPD [67]. Diuretics are often used to assist in achieving adequate kilocalories for growth without volume overload. Vitamin A Vitamin A plays an important role in vision, immunity, and growth, and in the integrity of epithelial cells such as in the respiratory tract. Preterm infants have low serum vitamin A levels and studies suggest that infants with low levels of vitamin A are at increased risk for BPD. A multicenter, blinded, randomized trial in infants with a mean birth weight of 770 g showed that 5000 IU of vitamin A administered intramuscularly three times per week for four weeks decreased death or CLD significantly in the treatment group (relative risk [RR] = 0.89). The study showed that one additional infant survived without CLD for every 14–15 infants treated and that the control and treatment groups had similar numbers of infants with potential signs of vitamin A toxicity [68]. Analysis of the vitamin A 123 Lung (2008) 186:75–89 cohort from this study at 18–24 months revealed that the vitamin A group had similar mortality and neurodevelopmental outcomes as the control group [9]. Furthermore, a meta-analysis of seven trials showed a reduction in death or oxygen requirement at 1 month of age (RR = 0.93) and a decrease in oxygen requirement at 36 weeks PMA (RR = 0.87) in infants with a birth weight of less than 1000 g receiving vitamin A supplementation [9]. Potential risks of vitamin A include toxicity leading to increased intracranial pressure, mucous membrane lesions, and emesis. Furthermore, vitamin A is administered intramuscularly, which induces pain, local inflammation, and risk of bleeding [68, 69]. Inositol Inositol is an essential nutrient that plays important biological roles, including cell signaling, cytoskeleton assembly, cell membrane maintenance, fat breakdown, and maturation of pulmonary surfactant. Inositol is present in human milk. It has been shown that infants with BPD have lower serum levels of inositol. Prior to the availability of surfactant for routine use, supplemental inositol was evaluated as a potential treatment for RDS and BPD [70]. Review of the literature suggests that there is a reduction in the risk of death or BPD in two trials (RR = 0.56) with inositol supplementation, but a multicenter, randomized, controlled trial of surfactant-treated infants is needed to confirm the benefits [71]. Risks of inositol supplementation were not identified, presumably because it is a naturally occurring compound. Antioxidants/Proteinase Inhibitors/Anti-inflammatory Proteins Given the deficiency of antioxidant defenses in the premature infant and oxidant exposure as an important predisposing factor for BPD, it is logical to expect that administration of antioxidants, both antenatally and postnatally, might reduce BPD. These potential therapies include using antioxidant vitamins, superoxide dismutase, alpha-1 proteinase inhibitor, and Clara cell protein. There is very limited information on antenatal antioxidant supplementation and BPD. Even after years of work by Davis et al. [72], this mode of treatment has not yet proven to be effective. However, maternal supplementation with antioxidant vitamins C and E has been shown to reduce the occurrence of pre-eclampsia and, therefore, should reduce the rate of preterm delivery and thus should indirectly reduce the incidence of BPD [73, 74]. However, at present, the impact of antenatal antioxidant supplementation in Lung (2008) 186:75–89 reducing BPD is not completely known. Use of postnatally administered antioxidants such as superoxide dismutase has also been disappointing. Superoxide dismutase is an enzyme produced in the human body that participates in protection against oxygen free radicals. Studies of exogenous superoxide dismutase suggest that it is safe; however, there is no clear benefit for preventing BPD [75]. Alpha-1 proteinase plays a protective role against tissue damage. Attempts to treat with alpha-1 proteinase showed a trend toward decreased oxygen dependency at 28 days postnatal age, but it did not reduce the risk of BPD [76, 77]. Clara cell 10-kDa protein (CC10), a naturally occurring antiinflammatory protein in the respiratory tract, was found to be deficient in ventilated premature infants who develop BPD. If given endotracheally, it has been shown to result in decreased inflammatory markers in tracheal aspirate fluid. However, further studies are needed to evaluate recombinant CC10’s role in preventing BPD [78]. On the whole, exogenous antioxidants, proteinase inhibitors, and antiinflammatory proteins have not proven successful in reducing the incidence of BPD. Novel Insights into the Molecular Basis of BPD The initiation and continued use of mechanical ventilation in infants with birth weights of less than 1250 g is the strongest risk factor for the development of BPD. The surfactant-deficient alveoli of such premature infants undergo breath-by-breath collapse (derecruitment) and rerecruitment, resulting in atelectrauma, which sets in motion the molecular cascade resulting in the structural and morphologic changes characteristic of BPD. Experimental studies in preterm animals have clearly demonstrated that even a few large tidal volume breaths can result in acute lung injury that initiates the cascade of molecular events leading to BPD [79]. Given the central role of alveolar stretch in initiating the molecular injury leading to BPD, it is logical to pursue a stretch-sensitive gene(s) that may be expressed in the pulmonary alveolus and that is (are) essential for its structure and function. Parathyroid hormone-related protein (PTHrP) is one such gene. It is expressed in the developing endoderm and its receptor is present on the adepithelial mesoderm, and, most importantly, PTHrP knockout causes a stage-specific inhibition of fetal mouse lung development [80]. The lungs of these mice fail to transition from the pseudoglandular to the canalicular stage of lung development, i.e., failure of alveolarization, a hallmark of the ‘‘new’’ BPD. Based on cell/molecular mechanisms driven by PTHrP that are essential for normal physiologic lung development, we have taken a basic biological approach to elucidate the pathophysiology and molecular mechanism involved in the pathogenesis of BPD. 83 Physiologic Role of PTHrP in Alveolar Homeostasis Under the influence of sonic hedgehog, the developing endoderm expresses PTHrP and its receptor on the adjoining mesenchyme. PTHrP binding to its receptor on the mesenchyme activates the protein kinase A pathway, which actively downregulates the default Wingless/int (Wnt) pathway and upregulates the adipogenic pathway through a key nuclear transcription factor, peroxisome proliferator activated receptor c (PPARc), and its downstream regulatory genes adipocyte differentiation related protein (ADRP) and leptin (Fig. 1) [81]. ADRP is necessary for the transit of neutral lipid from the lipofibroblast to the alveolar type II (ATII) cell for surfactant phospholipid synthesis. Lipofibroblasts, in turn, secrete leptin, which acts on its receptor on ATII cells, stimulating both surfactant phospholipid and protein synthesis. Since PTHrP stimulates leptin production by lung fibroblasts, it provides a complete growth factor-mediated paracrine loop for the synthesis of pulmonary surfactant [82]. Overall, PTHrP signaling, by inhibiting Wingless/int signaling, inhibits the default myogenic phenotype and, by stimulating the PPARc signaling, induces the lipogenic phenotype, which is necessary for maintaining alveolar homeostasis through its paracrine effects on interstitial fibroblasts and ATII cells [13]. Specifically, the interstitial lipofibroblast phenotype provides protection against oxygen free radicals (i.e., protection against oxotrauma), trafficks neutral lipid substrate Fig. 1 Parathyroid hormone-related protein (PTHrP), secreted by the alveolar type II cell, binds to its receptor on the adjoining alveolar interstitial fibroblast, activating the protein kinase A pathway, which actively downregulates the default Wingless/int (Wnt) pathway and upregulates the adipogenic pathway through the key nuclear transcription factor peroxisome proliferator activated receptor c (PPARc) and its downstream regulatory genes adipocyte differentiation related protein (ADRP) and leptin. Lipofibroblasts in turn secrete leptin, which acts on its receptor on alveolar type II cell, stimulating surfactant synthesis 123 84 to ATII cells for surfactant phospholipid synthesis (i.e., protection against atelectrauma), and causes ATII cell proliferation (i.e., protection against any insult causing epithelial injury), thereby promoting alveolar growth, development, and injury repair [13, 83]. Physiologically, this would stabilize the alveolus, thus preventing its collapse, maintaining adequate gas exchange, and reducing the energy expenditure by decreasing the work of breathing. On the other hand, although myofibroblasts may also be important for normal lung development, these cells are the hallmark of all CLDs in both the neonate and the adult [84]. In the developing lung, myofibroblasts are fewer in number and localize to the periphery of the alveolar septa, where they very likely participate in the formation of new septa [85]. However, in CLDs, myofibroblasts not only increase in number but are also abnormally located in the center of the alveolar septum in great abundance. In summary, PTHrP signaling is critical in maintaining alveolar homeostasis. Under the influence of cyclic stretch, PTHrP is secreted by the ATII cell, which acts on its receptor on the adjacent lipofibroblast, which in turn secretes leptin that facilitates surfactant synthesis by the ATII cells, underlining the importance of PTHrP signaling in maintaining alveolar homeostasis. If normal PTHrP signaling is interrupted, e.g., by conditions such as prematurity, volutrauma, exposure to hyperoxia or inflammatory cytokines, all conditions that are known to be associated with BPD, normal alveolar homeostasis is altered and interstitial fibroblasts undergo molecular and phenotypic changes consistent with the development of BPD. Using a wide variety of pathophysiologic insults associated with BPD, i.e., barotrauma, oxotrauma, and infection, we have found that there are type II cell and/or fibroblast cell/molecular effects generated by these insults that can lead to the BPD phenotype. Furthermore, at least in animal models using PTHrP and PPARc agonists, we have effectively abrogated specific alveolar molecular changes following barotrauma, oxotrauma, and infection that are known to lead to BPD. Complementing these studies in animal models, as elaborated upon next, we have also examined the significance of PTHrP signaling in human BPD. PTHrP Signaling in Human BPD We hypothesized that PTHrP levels in the tracheal aspirates (TA) of ventilated very-low-birth-weight infants (VLBWI) would correlate with the development of BPD. We examined whether TA PTHrP content during the first week of life correlated with the later development of BPD. Forty VLBWIs [birth weight = 943 ± 302 g (mean ± SD); gestational age = 27 ± 2 weeks; 21 males and 19 females] who were ventilated for RDS were studied. The 123 Lung (2008) 186:75–89 TAs were collected once daily until the infants were extubated and the samples were assayed for PTHrP. The levels of TA PTHrP correlated with the later development of BPD. PTHrP in the TA during the first week of life was significantly lower in those infants who developed BPD (12/40) than among those who did not (28/40). The PTHrP levels also correlated with the duration of mechanical ventilation needed in these infants. PTHrP B 1.32 pg/mg protein predicted the subsequent development of BPD maximally [84.6% correct classifications (true positives + true negatives)], with a sensitivity of 76.9% and specificity of 88.5%. Using a TA PTHrP level of 1.32 pg/ mg protein as the cutoff, we constructed Kaplan-Meier curves to compare the duration of ventilation needed between the two groups, i.e., less than or equal to and greater than 1.32 pg/mg protein TA PTHrP level. Infants with TA PTHrP levels greater than 1.32 pg/mg protein were off ventilatory support significantly earlier. To determine how PTHrP levels compared with the other known predictors of BPD, such as birth weight (\1000 g), GA (\ 28 weeks), and gender (male), we performed multivariate logistic regression analysis for these four variables in predicting the development of BPD. Of these variables, PTHrP B 1.32 pg/mg protein was the strongest predictor of BPD and remained so after adjusting for the other three variables, i.e., birth weight less than 1000 g, GA less than 28 weeks, and male gender. From these data we concluded that lower TA PTHrP content during the first week of life in ventilated VLBWI inversely correlates with prolonged ventilation and the subsequent development of BPD. Scientific Rationale for Proposing PTHrP/PPARc Agonists and Other Novel Interventions for the Prevention of BPD It is clear from what is briefly outlined above that we have systematically demonstrated the central role of PTHrPdriven epithelial-mesenchymal signaling in maintaining alveolar homeostasis. The lipofibroblast expresses PPARc, ADRP, and leptin in response to PTHrP signaling from the ATII cell, resulting in direct protection of the mesoderm against oxidant injury, and protection against atelectasis by augmenting surfactant phospholipid and protein synthesis. Disruption of PTHrP signaling between ATII cells and fibroblasts downregulates the adipogenic signaling pathway and upregulates the myogenic signaling pathway, causing myofibroblast transdifferentiation [13, 86–91]. Unlike lipofibroblasts, myofibroblasts cannot promote ATII cell growth and differentiation [13], leading to the failed alveolarization characteristic of BPD. A variety of factors associated with failed alveolarization, i.e., barotrauma, oxotrauma, and infection, all cause disruption of PTHrP/ Lung (2008) 186:75–89 PPARc signaling and myofibroblast transdifferentiation in vitro [13, 86–88, 91] and in vivo [89, 90]. More importantly, we have shown that PTHrP/PPARc signaling pathway agonists such rosiglitazone (RGZ) can effectively prevent or rescue myofibroblast transdifferentiation, therefore, preventing the inhibition of alveolarization in the developing lung. Therefore, it makes a compelling case for using PTHrP/PPARc agonists to prevent BPD. However, as yet there are no human data to show that normalization of PTHrP levels is effective in preventing or treating BPD. Impaired Vascularization in BPD Abnormal vascular growth and the decreased alveolarization are the central hallmarks of the ‘‘new’’ BPD. However, the mechanism(s) that interferes with vascular growth is poorly understood. Of the many angiogenic factors, vascular endothelial growth factor (VEGF) has been shown to play a central role in pulmonary vascular development. It is a potent stimulant of angiogenesis, promotes vascular remodeling, and enhances endothelial survival. Furthermore, it has been shown that inhibition of vascular growth in and of itself directly impairs alveolarization. Recent studies have convincingly demonstrated reduced VEGF signaling in the lungs of infants with fatal BPD, suggesting that by upregulating VEGF signaling, it might be possible to maintain normal alveolarization in the face of insults that lead to BPD [92, 93]. Therefore, it is not surprising that novel approaches that protect the immature endothelium and enhance vascular growth are being actively pursued [94, 95]. It is important to note that PTHrP, in addition to its role in pulmonary surfactant homeostasis, also modulates alveolar capillary perfusion [96] and helps in ventilation/perfusion matching, suggesting that PTHrP signaling as discussed above is not only critical for ATII cell/fibroblast interactions, but also for alveolar/endothelial crosstalk. Stem Cell Research As the hallmark of the ‘‘new’’ BPD is arrest of alveolarization and abnormal vascular growth, the potential for stem cells to supplement natural repair and regeneration of lung tissue is of potential importance. Adult bone marrow (BM)-derived cells have been shown to have the capability of differentiating into nonhematopoietic lineages. However, due to the complexity of the lung, the use of BM cells for lung repair has been slower than for their use in other tissues [97]. Hennrick et al. [98] in 2007 show that tracheal aspirate fluid from premature infants on mechanical ventilators contains mesenchymal stem cells with 85 differentiation potential into mesenchymal cells of different lineages. The role of these cells is yet to be fully determined. However, better understanding of the cells involved in the pathogenesis of BPD will lend itself to potential repair/replacement with stem cell technology. In particular, knowledge of the intrinsic molecular mechanisms that allow for recruitment of stem cells for lung repair would allow engineering and targeting of such cells for treating BPD. Although the novel molecular insights into the pathogenesis of BPD reviewed above provide a rational framework to design innovative therapeutic molecular interventions against BPD, the safety of these interventions remains to be proven. In addition to the documentation of the efficacy, the absence of any major side effects will be of paramount significance in the development and clinical application of these proposed interventions. This is particularly true since the PTHrP/PPARc and VEGF signaling pathways are not lung-specific and are known to be expressed in almost all organ systems in the body. Therefore, until lung-specific targeting of these approaches can be ensured, the clinical application of the concepts outlined above would be challenging. This is further highlighted by the recent concerns about a significant increase in the risk of heart attack and a nonsignificant increase in the risk of cardiovascular death in patients with type 2 diabetes receiving PPARc agonist RGZ as compared to controls. Use of PPARc agonists will need to undergo further scrutiny before they could be used for reducing BPD [99, 100]. Conclusions Despite all the advances in neonatal care, BPD remains a major cause of morbidity and mortality among premature infants. As one can gather from this review, there is no single approach or agent that has been clearly shown to prevent or treat human BPD. There have been very few well-designed studies that have tested specific interventions to treat BPD as their endpoint. This is partly because most often the pulmonary changes in BPD improve with time due to the normal pulmonary repair process, making it hard to separate the effect of treatment from the normal repair process. Nevertheless, the affected infant remains at risk for repeated hospitalizations and growth failure, particularly during the first year of life, and ultimately neurodevelopmental delay, mandating long-term follow-up and close vigilance on the part of everyone involved in the child’s care. Although once the condition has developed there is no specific intervention to treat it, the following approach at least minimizes the chances of developing severe BPD: (1) Aim to prolong fetal gestation for as long as safely 123 86 Lung (2008) 186:75–89 possible. (2) When indicated, ensure administration of antenatal steroids. (3) If possible, avoid intubation altogether, but if intubation and ventilation for respiratory distress are necessary, administer pulmonary surfactant as early as possible. (4) Ventilate at the lowest acceptable ventilatory settings. (5) Accept lower blood oxygen saturations as well as higher PaCO2 values. (6) Intramuscular administration of vitamin A during the first month of life might reduce BPD. In addition, refinement of existing therapeutic approaches such as aerosolized delivery of the surfactant Surfaxin (AerosurfTM, Disocovery Laboratories) might confer additional benefit in further reducing the possibility of BPD. Furthermore, use of agents with known benefit in other pulmonary diseases, such as iNO, may find a place in the treatment of BPD. Currently, however, there is not adequate evidence to suggest that routine iNO administration in extremely preterm infants with respiratory failure prevents BPD. Novel therapeutic approaches such as stem cell infusion, PTHrP and VEGF supplementations, and administration of PPARc agonists that are based on our understanding of the molecular mechanisms involved in the pathogenesis of BPD hold promise for the future. However, it is critically important to further judiciously study these physiologically and developmentally relevant interventions with the intent of effectively and safely treating the developing neonate. Finally, another research focus should be to identify specific and sensitive biomarkers that could be followed, preferentially noninvasively, to predict the development of BPD and also to follow its clinical progression/regression. Acknowledgments This work was supported by grants from the National Institutes of Health (HL55268 and HL 75405), the American Heart Association (0265127Y), the Tobacco-Related Disease Research Program (14RT-0073 and 15IT-0250), and Philip Morris USA, Inc. and Philip Morris International (11108–02). References 1. Jobe AH, Bancalari E (2001) Bronchopulmonary dysplasia. Am J Respir Crit Care Med 163:1723–1729 2. Walsh MC, Szefler S, Davis J, Allen M, Van ML, Abman S, Blackmon L, Jobe A (2006) Summary proceedings from the bronchopulmonary dysplasia group. Pediatrics 117:S52-S56 3. Rojas MA, Gonzalez A, Bancalari E, Claure N, Poole C, SilvaNeto G (1995) Changing trends in the epidemiology and pathogenesis of neonatal chronic lung disease. J Pediatr 126:605–610 4. Chye JK, Gray PH (1995) Rehospitalization and growth of infants with bronchopulmonary dysplasia: a matched control study. J Paediatr Child Health 31:105–111 5. Gregoire MC, Lefebvre F, Glorieux J (1998) Health and developmental outcomes at 18 months in very preterm infants with bronchopulmonary dysplasia. Pediatrics 101:856–860 6. Short EJ, Klein NK, Lewis BA, Fulton S, Eisengart S, Kercsmar C, Baley J, Singer LT (2003) Cognitive and academic consequences of bronchopulmonary dysplasia and very low birth weight: 8-year-old outcomes. Pediatrics 112:e359 123 7. Northway WH Jr, Rosan RC, Porter DY (1967) Pulmonary disease following respirator therapy of hyaline-membrane disease. Bronchopulmonary dysplasia. N Engl J Med 276:357–368 8. Husain AN, Siddiqui NH, Stocker JT (1998) Pathology of arrested acinar development in postsurfactant bronchopulmonary dysplasia. Hum Pathol 29:710–717 9. Ambalavanan N, Tyson JE, Kennedy KA, Hansen NI, Vohr BR, Wright LL, Carlo WA (2005) Vitamin A supplementation for extremely low birth weight infants: outcome at 18 to 22 months. Pediatrics 115:e249–e254 10. Ballard RA, Truog WE, Cnaan A, Martin RJ, Ballard PL, Merrill JD, Walsh MC, Durand DJ, Mayock DE, Eichenwald EC, Null DR, Hudak ML, Puri AR, Golombek SG, Courtney SE, Stewart DL, Welty SE, Phibbs RH, Hibbs AM, Luan X, Wadlinger SR, Asselin JM, Coburn CE (2006) Inhaled nitric oxide in preterm infants undergoing mechanical ventilation. N Engl J Med 355:343–353 11. Barrington KJ, Finer NN (1998) Treatment of bronchopulmonary dysplasia. A review. Clin Perinatol 25:177–202 12. Doyle LW, Halliday HL, Ehrenkranz RA, Davis PG, Sinclair JC (2005) Impact of postnatal systemic corticosteroids on mortality and cerebral palsy in preterm infants: effect modification by risk for chronic lung disease. Pediatrics 115:655–661 13. Torday JS, Torres E, Rehan VK (2003) The role of fibroblast transdifferentiation in lung epithelial cell proliferation, differentiation, and repair in vitro. Pediatr Pathol Mol Med 22:189– 207 14. Avery ME, Tooley WH, Keller JB, Hurd SS, Bryan MH, Cotton RB, Epstein MF, Fitzhardinge PM, Hansen CB, Hansen TN (1987) Is chronic lung disease in low birth weight infants preventable? A survey of eight centers. Pediatrics 79:26–30 15. Van Marter LJ, Allred EN, Pagano M, Sanocka U, Parad R, Moore M, Susser M, Paneth N, Leviton A (2000) Do clinical markers of barotrauma and oxygen toxicity explain interhospital variation in rates of chronic lung disease? The Neonatology Committee for the Developmental Network. Pediatrics 105:1194–1201 16. Aly H, Massaro AN, Patel K, El-Mohandes AA (2005) Is it safer to intubate premature infants in the delivery room? Pediatrics 115:1660–1665 17. Aly H (2007) Is there a strategy for preventing bronchopulmonary dysplasia? Absence of evidence is not evidence of absence. Pediatrics 119:818–820 18. Kinsella JP, Greenough A, Abman SH (2006) Bronchopulmonary dysplasia. Lancet 367:1421–1431 19. Davis PG, Henderson-Smart DJ (2003) Nasal continuous positive airways pressure immediately after extubation for preventing morbidity in preterm infants. Cochrane Database Syst Rev CD000143 20. Davis PG, Lemyre B, De Paoli AG (2001) Nasal intermittent positive pressure ventilation (NIPPV) versus nasal continuous positive airway pressure (NCPAP) for preterm neonates after extubation. Cochrane Database Syst Rev CD003212 21. Kulkarni A, Ehrenkranz RA, Bhandari V (2006) Effect of introduction of synchronized nasal intermittent positive-pressure ventilation in a neonatal intensive care unit on bronchopulmonary dysplasia and growth in preterm infants. Am J Perinatol 23:233–240 22. Bhandari A, Bhandari V (2007) Bronchopulmonary dysplasia: an update. Indian J Pediatr 74:73–77 23. Kamlin CO, Davis PG (2004) Long versus short inspiratory times in neonates receiving mechanical ventilation. Cochrane Database Syst Rev CD004503 24. Miller JD, Carlo WA (2007) Safety and effectiveness of permissive hypercapnia in the preterm infant. Curr Opin Pediatr 19:142–144 Lung (2008) 186:75–89 25. Thome UH, Carroll W, Wu TJ, Johnson RB, Roane C, Young D, Carlo WA (2006) Outcome of extremely preterm infants randomized at birth to different PaCO2 targets during the first seven days of life. Biol Neonate 90:218–225 26. Greenough A, Sharma A (2007) What is new in ventilation strategies for the neonate? Eur J Pediatr 166:991–996 27. Courtney SE, Durand DJ, Asselin JM, Hudak ML, Aschner JL, Shoemaker CT (2002) High-frequency oscillatory ventilation versus conventional mechanical ventilation for very-low-birthweight infants. N Engl J Med 347:643–652 28. Johnson AH, Peacock JL, Greenough A, Marlow N, Limb ES, Marston L, Calvert SA (2002) High-frequency oscillatory ventilation for the prevention of chronic lung disease of prematurity. N Engl J Med 347:633–642 29. Moriette G, Paris-Llado J, Walti H, Escande B, Magny JF, Cambonie G, Thiriez G, Cantagrel S, Lacaze-Masmonteil T, Storme L, Blanc T, Liet JM, Andre C, Salanave B, Breart G (2001) Prospective randomized multicenter comparison of highfrequency oscillatory ventilation and conventional ventilation in preterm infants of less than 30 weeks with respiratory distress syndrome. Pediatrics 107:363–372 30. McCallion N, Davis PG, Morley CJ (2005) Volume-targeted versus pressure-limited ventilation in the neonate. Cochrane Database Syst Rev CD003666 31. Deulofeut R, Critz A, Adams-Chapman I, Sola A (2006) Avoiding hyperoxia in infants \ or = 1250 g is associated with improved short- and long-term outcomes. J Perinatol 26:700– 705 32. Askie LM, Henderson-Smart DJ, Irwig L, Simpson JM (2003) Oxygen-saturation targets and outcomes in extremely preterm infants. N Engl J Med 349:959–967 33. Greenspan JS, Goldsmith JP (2006) Oxygen therapy in preterm infants: hitting the target. Pediatrics 118:1740–1741 34. Soll RF, Blanco F (2001) Natural surfactant extract versus synthetic surfactant for neonatal respiratory distress syndrome. Cochrane Database Syst Rev CD000144 35. Suresh GK, Soll RF (2002) Lung surfactants for neonatal respiratory distress syndrome: animal-derived or synthetic agents? Paediatr Drugs 4:485–492 36. Sinha SK, Lacaze-Masmonteil T, Soler A, Wiswell TE, Gadzinowski J, Hajdu J, Bernstein G, Sanchez-Luna M, Segal R, Schaber CJ, Massaro J, d’Agostino R (2005) A multicenter, randomized, controlled trial of lucinactant versus poractant alfa among very premature infants at high risk for respiratory distress syndrome. Pediatrics 115:1030–1038 37. Moya FR, Gadzinowski J, Bancalari E, Salinas V, Kopelman B, Bancalari A, Kornacka MK, Merritt TA, Segal R, Schaber CJ, Tsai H, Massaro J, d’Agostino R (2005) A multicenter, randomized, masked, comparison trial of lucinactant, colfosceril palmitate, and beractant for the prevention of respiratory distress syndrome among very preterm infants. Pediatrics 115:1018– 1029 38. Moya F, Sinha S, Gadzinowski J, d’Agostino R, Segal R, Guardia C, Mazela J, Liu G (2007) One-year follow-up of very preterm infants who received lucinactant for prevention of respiratory distress syndrome: results from 2 multicenter randomized, controlled trials. Pediatrics 119:e1361–e1370 39. Pfister RH, Soll RF, Wiswell T (2007) Protein containing synthetic surfactant versus animal derived surfactant extract for the prevention and treatment of respiratory distress syndrome. Cochrane Database Syst Rev CD006069 40. Yost CC, Soll RF (2000) Early versus delayed selective surfactant treatment for neonatal respiratory distress syndrome. Cochrane Database Syst Rev CD001456 41. Kinsella JP, Cutter GR, Walsh WF, Gerstmann DR, Bose CL, Hart C, Sekar KC, Auten RL, Bhutani VK, Gerdes JS, George 87 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. TN, Southgate WM, Carriedo H, Couser RJ, Mammel MC, Hall DC, Pappagallo M, Sardesai S, Strain JD, Baier M, Abman SH (2006) Early inhaled nitric oxide therapy in premature newborns with respiratory failure. N Engl J Med 355:354–364 Kinsella JP, Abman SH (2007) Inhaled nitric oxide in the premature newborn. J Pediatr 151:10–15 Steinhorn RH, Kinsella JP (2007) Use of inhaled nitric oxide in the preterm infant. Neoreviews 8:e247–e252 Truog WE (2007) Inhaled nitric oxide for the prevention of bronchopulmonary dysplasia. Expert Opin Pharmacother 8:1505–1513 Schmidt B, Roberts RS, Davis P, Doyle LW, Barrington KJ, Ohlsson A, Solimano A, Tin W (2006) Caffeine therapy for apnea of prematurity. N Engl J Med 354:2112–2121 Crowley PA (1995) Antenatal corticosteroid therapy: a metaanalysis of the randomized trials, 1972 to 1994. Am J Obstet Gynecol 173:322–335 Roberts D, Dalziel S (2006) Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev 3:CD004454 Halliday HL, Ehrenkranz RA, Doyle LW (2003) Early postnatal (\96 hours) corticosteroids for preventing chronic lung disease in preterm infants. Cochrane Database Syst Rev CD001146 Watterberg KL, Gerdes JS, Cole CH, Aucott SW, Thilo EH, Mammel MC, Couser RJ, Garland JS, Rozycki HJ, Leach CL, Backstrom C, Shaffer ML (2004) Prophylaxis of early adrenal insufficiency to prevent bronchopulmonary dysplasia: a multicenter trial. Pediatrics 114:1649–1657 Halliday HL, Ehrenkranz RA, Doyle LW (2003) Moderately early (7–14 days) postnatal corticosteroids for preventing chronic lung disease in preterm infants. Cochrane Database Syst Rev CD001144 Halliday HL, Ehrenkranz RA, Doyle LW (2003) Delayed ([3 weeks) postnatal corticosteroids for chronic lung disease in preterm infants. Cochrane Database Syst Rev CD001145 Shah V, Ohlsson A, Halliday HL, Dunn MS (2007) Early administration of inhaled corticosteroids for preventing chronic lung disease in ventilated very low birth weight preterm neonates. Cochrane Database Syst Rev CD001969 Shah SS, Ohlsson A, Halliday H, Shah VS (2003) Inhaled versus systemic corticosteroids for preventing chronic lung disease in ventilated very low birth weight preterm neonates. Cochrane Database Syst Rev CD002058 Shah SS, Ohlsson A, Halliday H, Shah VS (2003) Inhaled versus systemic corticosteroids for the treatment of chronic lung disease in ventilated very low birth weight preterm infants. Cochrane Database Syst Rev CD002057 Watterberg KL, Demers LM, Scott SM, Murphy S (1996) Chorioamnionitis and early lung inflammation in infants in whom bronchopulmonary dysplasia develops. Pediatrics 97:210–215 Kallapur SG, Jobe AH (2006) Contribution of inflammation to lung injury and development. Arch Dis Child Fetal Neonatal Ed 91:F132–F135 Schelonka RL, Katz B, Waites KB, Benjamin DK Jr (2005) Critical appraisal of the role of Ureaplasma in the development of bronchopulmonary dysplasia with metaanalytic techniques. Pediatr Infect Dis J 24:1033–1039 Viscardi R, Manimtim W, He JR, Hasday JD, Sun CC, Joyce B, Pierce RA (2006) Disordered pulmonary myofibroblast distribution and elastin expression in preterm infants with Ureaplasma urealyticum pneumonitis. Pediatr Dev Pathol 9:143–151 Mabanta CG, Pryhuber GS, Weinberg GA, Phelps DL (2003) Erythromycin for the prevention of chronic lung disease in intubated preterm infants at risk for, or colonized or infected with 123 88 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. Lung (2008) 186:75–89 Ureaplasma urealyticum. Cochrane Database Syst Rev CD003744 Ballard HO, Anstead MI, Shook LA (2007) Azithromycin in the extremely low birth weight infant for the prevention of bronchopulmonary dysplasia: a pilot study. Respir Res 8:41 Brion LP, Primhak RA, Yong W (2006) Aerosolized diuretics for preterm infants with (or developing) chronic lung disease. Cochrane Database Syst Rev 3:CD001694 Brion LP, Primhak RA, Ambrosio-Perez I (2002) Diuretics acting on the distal renal tubule for preterm infants with (or developing) chronic lung disease. Cochrane Database Syst Rev CD001817 Brion LP, Primhak RA (2000) Intravenous or enteral loop diuretics for preterm infants with (or developing) chronic lung disease. Cochrane Database Syst Rev CD001453 Ng GY, da S, Ohlsson A (2001) Bronchodilators for the prevention and treatment of chronic lung disease in preterm infants. Cochrane Database Syst Rev CD003214 Lai NM, Rajadurai SV, Tan KH (2006) Increased energy intake for preterm infants with (or developing) bronchopulmonary dysplasia/ chronic lung disease. Cochrane Database Syst Rev 3:CD005093 Mataloun MM, Rebello CM, Mascaretti RS, Dohlnikoff M, Leone CR (2006) Pulmonary responses to nutritional restriction and hyperoxia in premature rabbits. J Pediatr (Rio J) 82:179–185 Biniwale MA, Ehrenkranz RA (2006) The role of nutrition in the prevention and management of bronchopulmonary dysplasia. Semin Perinatol 30:200–208 Tyson JE, Wright LL, Oh W, Kennedy KA, Mele L, Ehrenkranz RA, Stoll BJ, Lemons JA, Stevenson DK, Bauer CR, Korones SB, Fanaroff AA (1999) Vitamin A supplementation for extremely-low-birth-weight infants. National Institute of Child Health and Human Development Neonatal Research Network. N Engl J Med 340:1962–1968 Darlow BA, Graham PJ (2002) Vitamin A supplementation for preventing morbidity and mortality in very low birthweight infants. Cochrane Database Syst Rev CD000501 Hallman M, Arjomaa P, Hoppu K (1987) Inositol supplementation in respiratory distress syndrome: relationship between serum concentration, renal excretion, and lung effluent phospholipids. J Pediatr 110:604–610 Howlett A, Ohlsson A (2003) Inositol for respiratory distress syndrome in preterm infants. Cochrane Database Syst Rev CD000366 Davis JM (2002) Role of oxidant injury in the pathogenesis of neonatal lung disease. Acta Paediatr Suppl 91:23–25 Bolisetty S, Naidoo D, Lui K, Koh TH, Watson D, Whitehall J (2002) Antenatal supplementation of antioxidant vitamins to reduce the oxidative stress at delivery–a pilot study. Early Hum Dev 67:47–53 Chappell LC, Seed PT, Briley AL, Kelly FJ, Lee R, Hunt BJ, Parmar K, Bewley SJ, Shennan AH, Steer PJ, Poston L (1999) Effect of antioxidants on the occurrence of pre-eclampsia in women at increased risk: a randomised trial. Lancet 354:810– 816 Suresh GK, Davis JM, Soll RF (2001) Superoxide dismutase for preventing chronic lung disease in mechanically ventilated preterm infants. Cochrane Database Syst Rev CD001968 Shah P, Ohlsson A (2001) Alpha-1 proteinase inhibitor (a1PI) for preventing chronic lung disease in preterm infants. Cochrane Database Syst Rev CD002775 Stiskal JA, Dunn MS, Shennan AT, O’Brien KK, Kelly EN, Koppel RI, Cox DW, Ito S, Chappel SL, Rabinovitch M (1998) alpha1-Proteinase inhibitor therapy for the prevention of chronic lung disease of prematurity: a randomized, controlled trial. Pediatrics 101:89–94 123 78. Levine CR, Gewolb IH, Allen K, Welch RW, Melby JM, Pollack S, Shaffer T, Pilon AL, Davis JM (2005) The safety, pharmacokinetics, and anti-inflammatory effects of intratracheal recombinant human Clara cell protein in premature infants with respiratory distress syndrome. Pediatr Res 58:15–21 79. Bjorklund LJ, Ingimarsson J, Curstedt T, John J, Robertson B, Werner O, Vilstrup CT (1997) Manual ventilation with a few large breaths at birth compromises the therapeutic effect of subsequent surfactant replacement in immature lambs. Pediatr Res 42:348–355 80. Rubin LP, Kovacs CS, De Paepe ME, Tsai SW, Torday JS, Kronenberg HM (2004) Arrested pulmonary alveolar cytodifferentiation and defective surfactant synthesis in mice missing the gene for parathyroid hormone-related protein. Dev Dyn 230:278–289 81. Torday JS, Rehan VK (2004) Deconvoluting lung evolution using functional/comparative genomics. Am J Respir Cell Mol Biol 31:8–12 82. Torday JS, Rehan VK (2002) Stretch-stimulated surfactant synthesis is coordinated by the paracrine actions of PTHrP and leptin. Am J Physiol Lung Cell Mol Physiol 283:L130–L135 83. Torday JS, Torday DP, Gutnick J, Qin J, Rehan V (2001) Biologic role of fetal lung fibroblast triglycerides as antioxidants. Pediatr Res 49:843–849 84. Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB (1999) Myofibroblasts. I. Paracrine cells important in health and disease. Am J Physiol 277:C1–C9 85. Bostrom H, Willetts K, Pekny M, Leveen P, Lindahl P, Hedstrand H, Pekna M, Hellstrom M, Gebre-Medhin S, Schalling M, Nilsson M, Kurland S, Tornell J, Heath JK, Betsholtz C (1996) PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell 85:863–873 86. Boros LG, Torday JS, Paul Lee WN, Rehan VK (2002) Oxygeninduced metabolic changes and transdifferentiation in immature fetal rat lung lipofibroblasts. Mol Genet Metab 77:230–236 87. Rehan V, Torday J (2003) Hyperoxia augments pulmonary lipofibroblast-to-myofibroblast transdifferentiation. Cell Biochem Biophys 38:239–250 88. Rehan VK, Wang Y, Sugano S, Romero S, Chen X, Santos J, Khazanchi A, Torday JS (2005) Mechanism of nicotine-induced pulmonary fibroblast transdifferentiation. Am J Physiol Lung Cell Mol Physiol 289:L667–L676 89. Rehan VK, Wang Y, Patel S, Santos J, Torday JS (2006) Rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, prevents hyperoxia-induced neonatal rat lung injury in vivo. Pediatr Pulmonol 41:558–569 90. Rehan VK, Wang Y, Sugano S, Santos J, Patel S, Sakurai R, Boros LG, Lee WP, Torday JS (2007) In utero nicotine exposure alters fetal rat lung alveolar type II cell proliferation, differentiation, and metabolism. Am J Physiol Lung Cell Mol Physiol 292:L323–L333 91. Rehan VK, rgan-Batra SK, Wang Y, Cerny L, Sakurai R, Santos J, Beloosesky R, Gayle D, Torday JS (2007) A paradoxical temporal response of the PTHrP/PPARgamma signaling pathway to lipopolysaccharide in an in vitro model of the developing rat lung. Am J Physiol Lung Cell Mol Physiol 293:L182–L190 92. Bhatt AJ, Pryhuber GS, Huyck H, Watkins RH, Metlay LA, Maniscalco WM (2001) Disrupted pulmonary vasculature and decreased vascular endothelial growth factor, Flt-1, and TIE-2 in human infants dying with bronchopulmonary dysplasia. Am J Respir Crit Care Med 164:1971–1980 93. Stenmark KR, Balasubramaniam V (2005) Angiogenic therapy for bronchopulmonary dysplasia: rationale and promise. Circulation 112:2383–2385 94. Abman SH (2001) Bronchopulmonary dysplasia: ‘‘a vascular hypothesis’’. Am J Respir Crit Care Med 164:1755–1756 Lung (2008) 186:75–89 95. Thebaud B, Abman SH (2007) Bronchopulmonary dysplasia: where have all the vessels gone? Roles of angiogenic growth factors in chronic lung disease. Am J Respir Crit Care Med 175:978–985 96. Gao Y, Raj JU (2005) Parathyroid hormone-related proteinmediated responses in pulmonary arteries and veins of newborn lambs. Am J Physiol Lung Cell Mol Physiol 289:L60–L66 97. Van HT, Thebaud B (2006) Adult bone marrow-derived stem cells for the lung: implications for pediatric lung diseases. Pediatr Res 59:94R–99R 89 98. Hennrick KT, Keeton AG, Nanua S, Kijek TG, Goldsmith AM, Sajjan US, Bentley JK, Lama VN, Moore BB, Schumacher RE, Thannickal VJ, Hershenson MB (2007) Lung cells from neonates show a mesenchymal stem cell phenotype. Am J Respir Crit Care Med 175:1158–1164 99. Nissen SE, Wolski K (2007) Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 356:2457–2471 100. Psaty BM, Furberg CD (2007) The record on rosiglitazone and the risk of myocardial infarction. N Engl J Med 357:67–69 123